Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
INDOLINE COMPOUNDS AS GRANZYME B INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2014/153666
Kind Code:
A1
Abstract:
Indoline Granzyme B inhibitor compounds, compositions that include the compounds, and methods for inhibiting Granzyme B in a subject or for treating a disease, disorder or condition treatable by inhibiting Granzyme B using said compounds. The indoline compounds of the invention have advantageous water solubility and effectively inhibit Granzyme B.

Inventors:
CAMERON DALE R (CA)
Application Number:
PCT/CA2014/050317
Publication Date:
October 02, 2014
Filing Date:
March 28, 2014
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
VIDA THERAPEUTICS INC (CA)
International Classes:
C07K5/083; A61K38/06; A61P9/00; A61P17/02
Domestic Patent References:
WO2003065987A22003-08-14
WO2012076985A22012-06-14
Foreign References:
US20030148511A12003-08-07
Other References:
BUZZA, MS. ET AL.: "Extracellular matrix remodeling by human granzyme B via cleavage of vitronectin, fibronectin, and laminin.", THE JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 280, no. 25, 24 June 2005 (2005-06-24), pages 23549 - 23558
KAM, C-M. ET AL.: "Granzymes (lymphocyte serine proteases) : characterization with natural and synthetic substrates and inhibitors.", BIOCHIMICA ET BIOPHYSICA ACTA, vol. 1477, no. 1-2, 2000, pages 307 - 323
Attorney, Agent or Firm:
MBM INTELLECTUAL PROPERTY LAW LLP (14th FloorOttawa, Ontario K1P 5H9, CA)
Download PDF:
Claims:
CLAIMS

The embodiments of the invention in which an exclusive property or privilege is claimed are defined as follows:

1 . A compound having Formula (I):

Formula (I) its stereoisomers and pharmaceutically acceptable salts thereof, wherein:

Ria is a heteroaryl group selected from

(a) 1 ,2,3-triazolyl optionally substituted with C1-C2 alkyl,

(b) 1 ,2,3,4-tetrazolyl, and

(c) 1 ,2,3-thiadiazolyl;

Ri b and R| C are independently selected from hydrogen, methyl, and halogen; R2a and R2b are independently selected from hydrogen and methyl, or Ria and R2b taken together are oxo;

n is 1 or 2;

R3 is selected from hydrogen and C 1 -C4 alkyl optionally substituted with a carboxylic acid or a carboxylate group or an amine group;

Z is an acyl group selected from the group herein o is 1 or 2, and Rsa is C4-acyl substituted with

(c) wherein p is 1 or 2, and Rsa is C4-acyl substituted with a carboxylic acid

(d) , wherein Y is heteroaryl;

wherein

R4 is selected from

(i) C,-C12 alkyl,

(ii) Ci-Ce heteroalkyl optionally substituted with C1 -C6 alkyl,

(iii) C3-C6 cycloalkyl,

(iv) Ce-Cio aryl,

(v) heterocyclyl,

(vi) C3-C10 heteroaryl,

(vii) aralk l, and

(viii) heteroalkylaryl;

R5 is -C(=0)-Rio, wherein Rio is selected from

(i) C1-C12 alkyl optionally substituted with C6-C 10 aryl, optionally substituted C3-C10 heteroaryl, amino, or carboxylic acid,

(ii) C1-C10 heteroalkyl optionally substituted with C1-C6 alkyl or carboxylic acid,

(iii) C3-C6 cycloalkyl optionally substituted with Ci-Ce alkyl, optionally substituted C6-C10 aryl, optionally substituted C3-C10 heteroaryl, amino, or carboxylic acid, (iv) C6-C10 aryl optionally substituted with Q -Ce alkyl, optionally substituted C6-C10 aryl, optionally substituted C3-C10 heteroaryl, amino, or carboxylic acid,

heterocyclyl,

C3-C10 heteroaryl,

aralkyl, and

heteroalkylaryl; and

R6, R7, e, and R9 are independently selected from

(a) hydrogen,

(b) halogen,

(c) d -Ce alkyl,

(d) -XRi 1 , wherein X is selected from O, C(=0), S, S=0, or S(=0)2,

(e) -C(=0)N(R,2)(R13),

(f) -N(Rn) (R,2)(Ri 3),

(g) -N-C(=0)-RM , and

(h) -N-C(=0)0-R„,

wherein Rn , R12, and R13 are independently selected from the group consisting of hydrogen, C1-C6 alkyl, C1-C6 heteroalkyl, C2-C6 alkenyl, C -C]o aralkyl, and C3-C10 heteroaryl.

2. A compound having Formula (I):

Formula (I) its stereoisomers and pharmaceutically acceptable salts thereof, wherein:

Ria is a heteroaryl group selected from

(a) 1 ,2,3-triazolyl optionally substituted with C1 -C2 alkyl, and (b) 1 ,2,3,4-tetrazolyl;

Ri b and R| C are independently selected from hydrogen and methyl;

R2 and R2b are independently selected from hydrogen and methyl;

n is 1 ;

R3 is selected from hydrogen and C 1 -C4 alkyl optionally substituted with a carboxylic acid or a carboxylate group or an amine group;

Z is an acyl group selected from the group

, wherein o is 1 or 2, and Rsa is C4-acyl substituted with a carboxylic acid;

wherein p is 1 or 2, and Rsa is C4-acyl substituted with a carboxylic acid; and p

(d) R4 , wherein Y is heteroaryl;

wherein

R4 is selected from

(i) C1-C12 alkyl,

(ii) C3-C6 cycloalkyl,

(iii) C6-C10 aryl, and

(iv) C3-C10 heteroaryl;

R5 is -C(=0)-Rio, wherein Rio is selected from (i) C| -Ci2 alkyl optionally substituted with C6-C10 aryl, optionally substituted C3-C10 heteroaryl, amino, or carboxylic acid,

(ii) C 1-C10 heteroalkyl optionally substituted with Ci -Ce alkyl or carboxylic acid,

(iii) C3-C6 cycloalkyl optionally substituted with Q-Ce alkyl, optionally substituted C6-C10 aryl, optionally substituted C3-C10 heteroaryl, amino, or carboxylic acid,

(iv) Ci-Cio aryl optionally substituted with C -C alkyl, oplionally substituted Ce-Ci o aryl, optionally substituted C3-C10 heteroaryl, amino, or carboxylic acid, and

(v) C3-C10 heteroaryl; and

R.6, R7, e, and R9 are independently selected from

(a) hydrogen,

(b) halogen,

(c) Ci -C6 alkyl,

(d) -XR| 1 , wherein X is selected from O and C(=0),

(e) -N(R„)(R,2)(Ri3), and

(f) -N-C(=0)-R,„

wherein Rn , R12, and R^ are independently selected from the group consisting of hydrogen, C 1 -C6 alkyl, Ci-Ce heteroalkyl, C2-C6 alkenyl, Ce-Cio aryl, aralkyl, and C3-C10 heteroaryl.

3. The compound of Claims 1 or 2, wherein R| a is tetrazole or triazole; R^, and R | c are H; n is 1 ; R2a and R2b are H; R3 is H or C 1 -C4 alkyl substituted with a carboxylic acid or carboxylate group; R6-R9 are H; and Z is

4. The compound of Claims 1 or 2, wherein The compound of Claims 1 or 2, wherein Rla is tetrazole or triazole; R^, and R] C are H; n is 1 ; R2a and R2t, are H; R3 is H or C1-C4 alkyl substituted with a carboxylic acid or carboxylate group; R -R9 are H; and Z is

5. The compound of Claims 1 or 2, wherein Rla is tetrazole or triazole; Rj^, and Rlc are H; n is 1; R2a and R2t> are H; R3 is H or C1-C4 alkyl substituted with a carboxylic acid or carboxylate group; R -R9 are H; and Z is

6. The compound of Claims 1 or 2, wherein Rja is tetrazole or triazole; R l3 and Ric are H; n is 1; R2a and 2), are H; R3 is H or C1-C4 alkyl substituted with a carboxylic acid or carboxylate group; R5-R9 are H; and Z is

A com ound having Formula (11):

Formula (II) its stereoisomers and pharmaceutically acceptable salts thereof, wherein:

Rla is tetrazole or triazole;

Rjt, and R]c are H;

n is 1; R2a and R2b are H;

R3 is H or CI -C4 alkyl substituted with a carboxylic acid or carboxylate group; R6-R9 are H;

R4 is C3-C6 cycloalkyl or C1 -C6 alkyl optionally substituted with hydroxyl or C 1-C6 alkoxy; and

R5 is -C(=O)-R10, wherein R10 is C1 -C12 alkyl substituted with a carboxylic acid or carboxylate group, or C3-C6 cycloalkyl substituted with a carboxylic acid or carboxylate group.

8. A com ound having Formula (III):

Formula (III) its stereoisomers and pharmaceutically acceptable salts thereof, wherein:

R] a is tetrazole or triazole;

R3 is H or C 1 -C4 alkyl substituted with a carboxylic acid or carboxylate group; R4 is C3-C6 cycloalkyl or C 1 -C6 alkyl optionally substituted with hydroxyl or

C 1 -C6 alkoxy; and

Rj O is -(CH2)n-C02H, wherein n is 2, 3, 4, 5, or 6;

optionally wherein one or more single methylene carbons are substituted with a fluoro, hydroxy, amino, C 1 -C3 alkyl, or C6-C10 aryl group;

optionally wherein one or more single methylene carbons are substituted with two fluoro or C1-C3 alkyl groups;

optionally wherein one or more single methylene carbons are substituted with two alkyl groups that taken together with the carbon to which they are attached form a 3, 4, 5, or 6-membered carbocyclic ring; or

optionally wherein adjacent carbon atoms from an unsaturated carbon-carbon bond or taken together form a benzene ring. 9 A com ound having Formula (III):

Formula (III) its stereoisomers and pharmaceutically acceptable salts thereof, wherein:

RL A is tetrazole or triazole;

R3 is H or C 1 -C4 alkyl substituted with a carboxylic acid or carboxylate group; R4 is C3-C6 cycloalkyl or C1-C6 alkyl optionally substituted with hydroxyl or

C 1 -C6 alkoxy; and

wherein n is 1 , 2, 3, or 4; and

optionally, for n = 3 or 4, wherein adjacent carbon atoms from an unsaturated carbon-carbon bond.

10. A com ound having Formula (IV):

Formula (IV) its stereoisomers and pharmaceutically acceptable salts thereof, wherein:

RL A is tetrazole or triazole;

R3 is H or C 1 -C4 alkyl substituted with a carboxylic acid or carboxylate group; R4 is C3-C6 cycloalkyl or C 1 -C6 alkyl optionally substituted with hydroxyl or

C 1 -C6 alkoxy; and

RJ I is selected from: (a) optionally substituted C6-C10 aryl; and

(b) optionally substituted C3-C10 heteroaryl.

1 1. A compound as shown in Table 1.

12. A pharmaceutical composition, comprising a compound of any one of Claims 1 - 1 1 and a pharmaceutically acceptable carrier.

13. A method for inhibiting Granzyme B in a subject, comprising administering an effective amount of a compound of any one of Claims 1 - 1 1 or a pharmaceutical composition of Claim 12 to a subject in need thereof.

14. A method for treating a disease, disorder, or condition treatable by inhibiting Granzyme B, comprising administering a therapeutical ly effective amount of a compound of any one of Claims 1 - 1 1 or a pharmaceutical composition of Claim 12 to a subject in need thereof.

15. The method of Claim 14, wherein the disease, disorder, or condition treatable by inhibiting Granzyme B is selected from treating dissection, aneurysm, and atherosclerosis.

16. The method of Claim 14, wherein the condition treatable by inhibiting Granzyme B is a wound and administering the compound or composition promotes wound healing.

17. The method of any one of Claims 13-16, wherein administering the compound comprises topical administration, oral administration, and administration by injection.

Description:
INDOLINE COMPOUNDS AS GRANZYME B INHIBITORS

CROSS-REFERENCE TO RELATED APPLICATIONS This application claims the benefit of U.S. Patent Appl ication No. 61 /806,767, filed March 29, 2013, and U.S. Patent Application No. 61/941 ,358, filed February 1 8, 2014, each expressly incorporated herein by reference in its entirety.

FIELD OF THE INVENTION

The present invention disclosure relates generally to agents for treating diseases, disorders, and conditions treatable by inhibiting Granzyme B, and more specifically to substituted indoline compounds that are inhibitors of Granyzme B.

BACKGROUND OF THE INVENTION

Granzyme B is a pro-apoptotic serine protease found in the granules of cytotoxic lymphocytes (CTL) and natural killer (NK) cells. Granzyme B is released towards target cells, along with the pore-forming protein, perforin, resulting in its perforin-dependent internalization into the cytoplasm and subsequent induction of apoptosis (see, for e.g., Medema et al., Eur. J. Immunol. 27:3492-3498, 1997). However, during aging, inflammation and chronic disease, Granzyme B can also be expressed and secreted by other types of immune (e.g., mast cell, macrophage, neutrophil, and dendritic cells) or non-immune (keratinocyte, chondrocyte) cells and has been shown to possess extracellular matrix remodel ing activity (Choy et al., Arterioscler. Thromb. Vase. Biol. 24( 12):2245-2250, 2004 and Buzza et al., J. Biol. Chem. 280:23549-23558, 2005).

Inhibitors of Granzyme B in humans have been limited to (a) relatively weak, nonspecific inhibitors such as isocoumarins (Odake et al., (1991 ), Biochemistry, 30(8), 2217-2227); (b) biological inhibitors such as serpinB9 (Sun et al., (1996), J. Biol. Chem. , 271 (44), 27802-27809); (c) covalently coupled inhibitors such as aldehydes (Willoughby et al., (2002), Bioorg. Med. Chem. Lett., 12(16), 2197), halomethyl ketones (Kam et al., (2000), Biochim. Biophy. Acta, 1477(1-2), 307-323), and phosphonates (Mahrus and Craik, (2005), Chem. & Biol, 12, 567-77 and Kam et al., (2000)); and (d) tricyclic inhibitors (Willoughby et al., (2002)).

Nonspecific inhibitors (such as isocoumarins) are not sufficiently potent or specific to be effective treatments for Granzyme-B-related diseases, disorders, and conditions. Likewise, the use of biological inhibitors such as serpins is limited by the ability to deliver the inhibitor to the target mammal, the cost of manufacturing the biological agents, and other, off-target activities, such as inhibition of other serine proteases such as human neutrophil elastase (Dahlen et al., (1999), Biochim. Biophys. Acta, 14 1 (2-3), 233-41), Caspase-1 (Annaud et al., (1999), Biochem. J., Sep 15 ; 342 Pt3, 655-65 ; Krieg et al., (2001 ), Mol. Endocrinol, 15(1 1 ), 1971 -82; and Young et al., (2000), J. Exp. Med. , 191 (9), 1535- 1544); Caspase-4 and Caspase-8 (Annaud et al., (1999)).

The tricyclic inhibitors (Willoughby et al. (2001)) also suffer from synthetic complexity/high manufacturing cost due to the complex core and accompanying low water solubility.

Despite the advances in development of Granzyme B inhibitors, there exists a need for compounds that inhibit Granzyme B with selectivity, that are relatively simple to manufacture at low cost, that are highly water soluble, and that do not present drug delivery challenges. The present invention seeks to fulfil l this need and provides further related advantages.

SUMMARY OF THE INVENTION

The present invention provides Granzyme B inhibitor compounds, compositions that include the compounds, and methods for using the compounds.

In one aspect of the invention, the invention provides Granzyme B inhibitor compounds. In one embodiment, the invention provides the compounds having Formula (I):

its stereoisomers and pharmaceutically acceptable salts thereof, wherein:

Ri a is a heteroaryl group selected from

(a) 1 ,2,3-triazolyl optionally substituted with C1 -C2 alkyl,

(b) 1 ,2,3,4-tetrazolyl, and

(c) 1 ,2,3-thiadiazolyl;

Ri b and RiC are independently selected from hydrogen, methyl, and halogen; R 2 a and R 2 b are independently selected from hydrogen and methyl, or R2a and R2b taken together are oxo (i .e., =0, R2a and R 2 b together with the carbon to which they are attached form a carbonyl group);

n is 1 or 2;

R3 is selected from hydrogen and C 1 -C4 alkyl optionally substituted with a carboxylic acid or a carboxylate group (-C<¾H or -C0 2 ' ) or an amine group (-NH 2 );

Z is an acyl group selected from the group

wherein 0 is 1 or 2, and Rsa is C4-acyl substituted with carboxylic acid;

(c) wherein p is 1 or 2, and Rs a is C4-acyl substituted with a carboxylic acid; and

O

(d) R4 , wherein Y is heteroaryl;

wherein

R4 is selected from

(i) C 1 -C 1 2 alkyl,

(ii) C 1 -C6 heteroalkyl optionally substituted with C1 -C6 alkyl,

(iii) C3-C6 cycloalkyl,

(iv) C 6 -C | 0 aryl,

(v) heterocyclyl,

(vi) C3-C 1 0 heteroaryl,

(vii) aralkyl, and (viii) heteroalkylaryl;

Rs is -C(=0)-Ri o, wherein Rio is selected from

(i) C 1 -C 12 alkyl optionally substituted with Ce-Cio aryl, optionally substituted C3-C 10 heteroaryl, amino, or carboxyl ic acid,

(ii) C i-Cio heteroalkyl optionally substituted with C |-Q alkyl or carboxylic acid,

(iii) C3-C6 cycloalkyl optionally substituted with C 1 -C6 alkyl, optionally substituted C6-C 10 aryl, optionally substituted C3-C10 heteroaryl, amino, or carboxylic acid,

(iv) Ce-Cio aryl optionally substituted with C|-Ce alkyl, optionally substituted C6-C10 aryl, optionally substituted C3-C10 heteroaryl, amino, or carboxylic acid,

(v) heterocyclyl,

(vi) C3-C 10 heteroaryl,

(vii) aralkyl, and

(viii) heteroalkylaryl; and

R6, R7, Rs, and R9 are independently selected from

(a) hydrogen,

(b) halogen,

(c) C| -C 6 alkyl,

(d) -XR | I , wherein X is selected from O, C(=0), S, S=0, or S(=0) 2 ,

(e) -C(=0)N(R 12 )(R, 3),

(f) -N(R | , ) (R, 2 )( i 3),

(g) -N-C(=0)-R,„and

(h) -N-C(=0)0-R| , ,

wherein Rn, R12, and n are independently selected from the group consisting of hydrogen, C1 -C6 alkyl, C] -Ce heteroalkyl, C2-C6 alkenyl, Ce-Cio aryl, aralkyl, and C3-C 10 heteroaryl.

In another embodiment, the invention provides compounds having Formula (I), its stereoisomers and pharmaceutically acceptable salts thereof, wherein:

Ri a is a heteroaryl group selected from

(a) 1 ,2,3-triazolyl optionally substituted with C1 -C2 alkyl, and

(b) 1 ,2,3,4-tetrazolyl; Rib and Ric are independently selected from hydrogen and methyl;

R2a and R 2 b are independently selected from hydrogen and methyl;

n is 1 ;

R3 is selected from hydrogen and C1 -C4 alkyl optionally substituted with a carboxylic acid or a carboxylate group (-CO2H or -CO2 ' ) or an amine group (-NH2);

Z is an acyl group selected from the group

(b) , wherein 0 is 1 or 2, and Rs a is C4-acyl substituted with a carboxylic acid;

carboxylic acid; and

O

(d) R , wherein Y is heteroaryl;

R4 is selected from

(i) C, -C, 2 alkyl,

(ii) C3-C6 cycloalkyl,

(iii) C 6 -Cio aryl, and

(iv) C3-C 10 heteroaryl;

R5 is -C(=0)-R|o, wherein Ri o is selected from

(i) C1 -C12 alkyl optionally substituted with C6-C10 aryl, optionally substituted C3-C10 heteroaryl, amino, or carboxylic acid,

(ii) Ci -C i o heteroalkyl optionally substituted with Ci -C 6 alkyl or carboxylic acid, (iii) C3-C6 cycloalkyl optionally substituted with C \-C(, alkyl, optionally substituted Ce-C \ o aryl, optionally substituted C3-C 10 heteroaryl, amino, or carboxylic acid,

(iv) C6-C1 0 aryl optionally substituted with Q -C6 alkyl, optionally substituted C6-C1 0 aryl, optionally substituted C3-C10 heteroaryl, amino, or carboxylic acid,

(v) C3-C1 0 heteroaryl; and

R6, R7, Re, and R9 are independently selected from

(a) hydrogen,

(b) halogen,

(c) Ci -C 6 alkyl,

(d) -XR] 1 , wherein X is selected from O and C(=0),

(f) -N(R„)(R, 2 )(Ri3),

(g) -N-C(=0)-R, , , and

wherein Rn , R 12 , and R13 are independently selected from the group consisting of hydrogen, C| -C 6 alkyl, Ci-C 6 heteroalkyl, C 2 -C 6 alkenyl, C 6 -C,o aryl, aralkyl, and C3-C10 heteroaryl.

In further embodiments, the invention provides compounds having Formula (I), its stereoisomers and pharmaceutically acceptable salts thereof, wherein:

R l a is tetrazole or triazole; R ] | , and R c are H; n is 1 ; R 2a and R 2 b are H; R 3 is H or C1 -C4 alkyl substituted with a carboxylic acid or carboxyiate group; Rg-R 9 are H; and

Z is

H O R4 :

R ] a is tetrazole or triazole; R^, and R jc are H; n is 1 ; R 2a and R 2) , are H; R 3 is H or C 1 -C4 alkyl substituted with a carboxylic acid or carboxyiate group; R^-Rg are H; and Z is

R ] a is tetrazole or triazole; R^, and R l c are H; n is 1 ; R 2a and R 2( , are H; R 3 is H or C 1 -C4 alkyl substituted with a carboxylic acid or carboxylate group; R 6 -R 9 are H; and

Z is

R la is tetrazole or triazole; and R l c are H; n is 1 ; R 2a and R 2b are H; R 3 is H or C 1 -C4 alkyl substituted with a carboxylic acid or carboxylate group; R 6 -R 9 are H; and Z is

O i wherein R , R 5a , o, p, and Y are as described above.

In another embodiment, the invention provides compounds having Formula (II):

Formula (II) its stereoisomers and pharmaceutically acceptable salts thereof, wherein:

R ] a is tetrazole or triazole; R^ and R l c are H; n is 1 ; R 2a and R 2 b are H; R 3 is H or C1 -C4 alkyl substituted with a carboxylic acid or carboxylate group; Rg-R 9 are H;

R 4 is C3-C6 cycloalkyl or C1 -C6 alkyl optionally substituted with hydroxyl or C 1 -C6 alkoxy; and

R5 is -C(=0)-Rio, wherein Rio is selected from

(i) C 1 -C12 alkyl optionally substituted with Ce-Cio aryl, optionally substituted C3-C 10 heteroaryl, amino, or carboxylic acid, (ii) Ci-Cio heteroalkyl optionally substituted with C1 -C6 alkyl or carboxylic acid,

(iii) C3-C6 cycloalkyl optionally substituted with Ci -Ce alkyl, optionally substituted Ce-Ci o aryl, optionally substituted C3-C 1 0 heteroaryl, amino, or carboxylic acid,

(iv) -C\o aryl optionally substituted with C1 -C6 alkyl, optionally substituted C6-C 10 aryl, optionally substituted C3-C10 heteroaryl, amino, or carboxylic acid,

(v) C3-C 10 heteroaryl.

In a further embodiment, the invention provides compounds having Formula (III):

Formula (III) its stereoisomers and pharmaceutically acceptable salts thereof, wherein:

R ] A , R 3 , R 4 , and R 1 0 are as above for Formula (II);

wherein R ] 0 , when defined as CI -C I 2 alkyl substituted with a carboxylic acid or carboxylate group, is:

-(CH 2 ) n -C0 2 H, where n is 2, 3, 4, 5, or 6;

optionally wherein one or more single methylene carbons are substituted with a fluoro, hydroxy, amino, C 1 -C3 alkyl (e.g., methyl), or C6-C 10 aryl group;

optionally wherein one or more single methylene carbons are substituted with two fluoro (e.g., difluoro, perfiuoro) or C 1 -C3 alkyl (e.g., gem-dimethyl) groups;

optionally wherein one or more single methylene carbons are substituted with two alkyl groups that taken together with the carbon to which they are attached form a 3, 4, 5, or 6-membered carbocyclic ring (e.g., spiro groups such as cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl); and

optionally wherein adjacent carbon atoms from an unsaturated carbon-carbon bond (e.g., alkenyl such as -CH=CH-) or taken form a benzene ring (e.g., 1 ,2-, 1 ,3-, and 1 ,4-phenylene); or

wherein R J Q, when defined as C3-C6 cycloalkyl substituted with a carboxylic acid or carboxylate group, is: <CH 2 ) n

C0 _ H , wherein n is 1 , 2, 3, or 4; and optionally, for n = 3 or 4, wherein adjacent carbon atoms from an unsaturated carbon-carbon bond (e.g., cyclopentenyl or cyclohexenyl).

In another embodiment, the invention provides compounds having Formula (IV):

Formula (IV) its stereoisomers and pharmaceutically acceptable salts thereof, wherein :

Rl a , R3, and R 4 are as above for Formulae (II) and (III); and is selected from:

(a) optionally substituted C6-C 10 aryl (e.g., unsubstituted phenyl and substituted phenyl such as carboxyphenyl, aminophenyl, alkylaminophenyl, d ia!kylaminophenyl); and

(b) optionally substituted C3-C10 heteroaryl (e.g., optionally substituted pyridyl, optionally substituted thiazolyl, optionally substituted benzothiophenyl, optionally substituted tetrazolyl, and optionally substituted triazolyl (e.g., -NH 2 substituted).

In another aspect, the invention provides pharmaceutical compositions comprising a Granzyme B inhibitor compound of the invention and a pharmaceutically acceptable carrier.

In a further aspect of the invention, a method for inhibiting Granzyme B is provided. In one embodiment, the method comprises administering an effective amount of a Granzyme B inhibitor compound of the invention or a pharmaceutical composition of the invention to a subject in need thereof.

In a further aspect of the invention, a method for treating a disease, disorder, or condition treatable by inhibiting Granzyme B is provided. In one embodiment, the method comprises administering a therapeutically effective amount of a Granzyme B inhibitor compound of the invention or a pharmaceutical composition of the invention to a subject in need thereof. Representative routes of administration include topical administration, oral administration, and administration by injection.

BRIEF DESCRIPTION OF THE DRAWINGS FIGURE 1 is a schematic illustration of a representative synthetic pathway for the preparation of representative compounds of the invention P5-P4-P3-P2-P1 starting from P I .

FIGURE 2 is a schematic illustration of another representative synthetic pathway for the preparation of representative compounds of the invention P5-P4-P3-P2-P1 starting from P5.

FIGURE 3 is a schematic illustration of a further representative synthetic pathway for the preparation of representative compounds of the invention P5-P4-P3-P2-P1 starting from a component other than PI or P5.

DETAILED DESCRIPTION OF THE INVENTION

The present invention provides Granzyme B inhibitor compounds, compositions that include the compounds, and methods for using the compounds. The compounds of the invention have advantageous water solubility and effectively inhibit Granzyme B.

In one aspect of the invention, the invention provides Granzyme B inhibitor compounds. In one embodiment, the invention provides the compounds having Formula (I):

Formula (I)

its stereoisomers and pharmaceutically acceptable salts thereof, wherein:

Ria is a heteroaryl group selected from

(a) 1 ,2,3-triazolyl optionally substituted with C 1 -C2 alkyl,

(b) 1 ,2,3,4-tetrazolyl, and

(c) 1 ,2,3-thiadiazolyl;

Ri b and R] C are independently selected from hydrogen, methyl, and halog R.2a and R 2 b are independently selected from hydrogen and methyl, or R 2 a and Rib taken together are oxo (i.e., =0, R2a and R 2 b together with the carbon to which they are attached form a carbonyl group);

n is 1 or 2;

R3 is selected from hydrogen and C 1 -C4 alkyl optionally substituted with a carboxylic acid or a carboxylate group (-CO2H or -CC ) or an amine group (-NH2);

Z is an acyl group selected from the group

(b) , wherein 0 is 1 or 2, and Rs a is C4-acyl substituted with a carboxylic acid;

(c) wherein p is 1 or 2, and Rs a is C4-acyl substituted with a carboxylic acid; and

0

(d) R4 , wherein Y is heteroaryl;

wherein

R4 is selected from

(i) C 1 -C 12 alkyl,

(ii) C| -Ce heteroalkyl optionally substituted with C \ -C(, alkyl,

(iii) C3-C6 cycloalkyl,

(iv) C 6 -C | 0 aryl,

(v) heterocyclyl,

(vi) C3-C 10 heteroaryl,

(vii) aralkyl, and (viii) heteroalkylaryl;

R.5 is -C(=0)-Ri o, wherein Rio is selected from

(i) C 1-C12 alkyl optionally substituted with C6-C10 aryl, optionally substituted C3-C 10 heteroaryl, amino, or carboxylic acid,

(ii) C1-C10 heteroalkyl optionally substituted with Ci-C 6 alkyl or carboxylic acid,

(iii) C3-C6 cycloalkyl optionally substituted with C] -C alkyl, optionally substituted Ce-Cio aryl, optionally substituted C3-C10 heteroaryl, amino, or carboxylic acid,

(iv) C6-C10 aryl optionally substituted with Cj-Ce alkyl, optionally substituted C6-C10 aryl, optionally substituted C3-Q0 heteroaryl, amino, or carboxylic acid,

(v) heterocyclyl,

(vi) C3-C 10 heteroaryl,

(vii) aralkyl, and

(viii) heteroalkylaryl; and

R6, R7, Re, and R9 are independently selected from

(a) hydrogen,

(b) halogen,

(c) C | -C 6 alkyl,

(d) -XR, 1 , wherein X is selected from O, C(=0), S, S=0, or S(=0) 2 ,

(e) -C(=0)N(R, 2 )(R 13 ),

(f) -N(R„) (R,2)(Ri 3),

(g) -N-C(=0)-R„, and

(h) -N-C(=0)0-R, , ,

wherein Rn, R12, and R13 are independently selected from the group consisting of hydrogen, C1 -C6 alkyl, Ci -Ce heteroalkyl, C 2 -Ce alkenyl, Ce-Cio aryl, aralkyl, and C3-C 10 heteroaryl.

Representative compounds of the above embodiment include Examples A41 and

B2.

In another embodiment, the invention provides compounds having Formula (I), its stereoisomers and pharmaceutically acceptable salts thereof, wherein:

Ria is a heteroaryl group selected from (a) 1 ,2,3-triazolyl optionally substituted with C1 -C2 alkyl, and

(b) 1 ,2,3,4-tetrazol l;

Ri b and R| C are independently selected from hydrogen and methyl;

R2a and R2b are independently selected from hydrogen and methyl;

n is 1 ;

R3 is selected from hydrogen and C 1 -C4 alkyl optionally substituted with carboxylic acid or a carboxylate group (-C0 2 H or -C0 2 ' ) or an amine group (-NH 2 );

Z is an acyl group selected from the group

herein o is 1 or 2, and Rs a is C4-acyl substituted with

wherein p is 1 or 2, and Rs a is C4-acyl substituted with a carboxylic acid; and

O

(d) R - , wherein Y is heteroaryl;

R4 is selected from

(i) C -C 12 alkyl,

(ii) C3-C6 cycloalkyl,

(iii) C6-C10 aryl, and

(iv) C3-C 10 heteroaryl;

R5 is -C(=0)-Rio, wherein R10 is selected from

(i) C]-C\ 2 alkyl optionally substituted with Ce-C o aryl, optionally substituted

C3-C10 heteroaryl, amino, or carboxylic acid, (ii) Ci-Ci o heteroalkyl optionally substituted with Ci-Ce alkyl or carboxylic acid,

(iii) C3-C6 cycloalkyl optionally substituted with C1 -C6 alkyl, optionally substituted Ce-Ci o aryl, optionally substituted C3-C10 heteroaryl, amino, or carboxylic acid,

(iv) C -C10 aryl optionally substituted with C -Ce alkyl, optionally substituted C -C 10 aryl, optionally substituted C3-C10 heteroaryl, amino, or carboxylic acid,

(v) C3-C10 heteroaryl; and

R6, R7, Re, and R9 are independently selected from

(a) hydrogen,

(b) halogen,

(c) C, -C 6 alkyl,

(d) -XR| 1 , wherein X is selected from O and C(=0),

(f) -N(R„)(R, 2 )(Ri 3),

wherein Rn , R12, and R13 are independently selected from the group consisting of hydrogen, Ci -C 6 alkyl, C, -C 6 heteroalkyl, C 2 -C6 alkenyl, C 6 -Ci 0 aryl, aralkyl, and C3-C10 heteroaryl.

Representative compounds of the above embodiment include Examples C25, C26, C28, C29, C36, and C37.

Other representative compounds of the above embodiment include Examples C23, C24, and C38.

Other representative compounds of the above embodiment include Examples B3, B4, 1-5, A6, A7, A15, A29 and C47.

In further embodiments, the invention provides compounds having Formula (I), its stereoisomers and pharmaceutically acceptable salts thereof, wherein:

R l a is tetrazole or triazole; R^ and R j c are H; n is 1 ; R 2a and R 2 b are H; R 3 is H or C 1 -C4 alkyl substituted with a carboxylic acid or carboxylate group; R 6 -R 9 are H; and Z is R l a is tetrazole or triazole; and R j c are H; n is 1 ; R 2a and R 2j , are H; R 3 is H or C1 -C4 alkyl substituted with a carboxylic acid or carboxylate group; R -R 9 are H; and Z is

R la is tetrazole or triazole; R^ and Ri c are H; n is 1 ; R 2a and R 2( , are H; R 3 is H or C1-C4 alkyl substituted with a carboxylic acid or carboxylate group; R 6 -R 9 are H; and Z is

R ] a is tetrazole or triazole; R^ and R ] c are H; n is 1 ; R 2a and R 2 t, are H; R 3 is H or C 1 -C4 alkyl substituted with a carboxylic acid or carboxylate group; R 6 -R9 are H; and Z is

wherein R 4 , R 5a , o, p, and Y are as described above.

In another embodiment, the invention provides compounds having Formula (II):

Formula (II) its stereoisomers and pharmaceutically acceptable salts thereof, wherein:

Ri a is tetrazole or triazole; R j t, and R j c are H; n is 1 ; R 2a and R 2 t, are H; R 3 is H or C 1 -C4 alkyl substituted with a carboxylic acid or carboxylate group; R 6 -R 9 are H; R 4 is C3-C6 cycloalkyl or C 1 -C6 alkyl optionally substituted with hydroxy! or C 1 -C6 alkoxy; and

Rs is -C(=0)-Rio, wherein Rio is selected from

(i) C I -C 12 alkyl optionally substituted with Ce-Cio aryl, optionally substituted C3-C 10 heteroaryl, amino, or carboxylic acid,

(ii) C 1 -C10 heteroalkyl optionally substituted with C | -C 6 alkyl or carboxylic acid,

(iii) C3-C6 cycloalkyl optionally substituted with C \ -C alkyl, optionally substituted C6-C 10 aryl, optionally substituted C3-C10 heteroaryl, amino, or carboxylic acid,

(iv) C6-C10 aryl optionally substituted with Q-Ce alkyl, optionally substituted Ce-Cio aryl, optionally substituted C3-C10 heteroaryl, amino, or carboxylic acid,

(v) C3 -C 10 heteroary 1.

Representative compounds of the above embodiment include Examples B5, B6, B7, C 1 9, C20, C21 , C22, C27, C30, C3 1 , C32, C33, C34, and C35.

In a further embodiment, the invention provides compounds having Formula (III):

Formula (III) its stereoisomers and pharmaceutically acceptable salts thereof, wherein:

R l a , R3, R 4 , and R 10 are as above for Formula (II);

wherein R 10 , when defined as C 1 -C 12 alkyl substituted with a carboxylic acid or carboxylate group, is:

-(CH 2 ) n -C0 2 H, where n is 2, 3, 4, 5, or 6;

optionally wherein one or more single methylene carbons are substituted with a fluoro, hydroxy, amino, C 1 -C3 alkyl (e.g., methyl), or C6-C 10 aryl group;

optionally wherein one or more single methylene carbons are substituted with two fluoro (e.g., difluoro, perfluoro) or C1 -C3 alkyl (e.g., gem-dimethyl) groups;

optionally wherein one or more single methylene carbons are substituted with two alkyl groups that taken together with the carbon to which they are attached form a 3, 4, 5, or 6-membered carbocyclic ring (e.g., spiro groups such as cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl); and

optionally wherein adjacent carbon atoms from an unsaturated carbon-carbon bond (e.g., alkenyl such as -CH=CH-) or taken form a benzene ring (e.g., 1,2-, 1,3-, and 1,4-phenylene); or

wherein RJQ, when defined as C3-C6 cycloalkyi substituted with a carboxylic acid or carboxylate group, is: (CH 2 ) N

wherein n is 1, 2, 3, or 4; and optionally, for n = 3 or 4, wherein adjacent carbon atoms from an unsaturated carbon-carbon bond (e.g., cyclopentenyl or cyclohexenyl).

Representative compounds of the above embodiment include Examples A5, A13, A14, A16, A17-1, A17-2, A18, A19, A20-1, A20-2, A21-1, A21-2, A22-1, A22-2, A23-1, A23-2, A24, A25, A26-1, A26-2, A30-1, A30-2, A31, A32, A33, A36, A37, A38, A39, A40, A43, A44, A45, A46, A48, A51, A52, A53, A54-1, A54-2, A55, A56, A57-1, and A57-2.

In another embodiment, the invention provides compounds having Formula (IV):

Formula (IV) its stereoisomers and pharmaceutically acceptable salts thereof, wherein:

Rl a , R 3 , and R 4 are as above for Formulae (II) and (III); and R ] | is selected from:

(a) optionally substituted C6-C10 aryl (e.g., unsubstituted phenyl and substituted phenyl such as carboxyphenyl, aminophenyl, alkylaminophenyl, dialkylaminophenyl); and

(b) optionally substituted C3-C10 heteroaryl (e.g., optionally substituted pyridyl, optionally substituted thiazolyl, optionally substituted benzothiophenyl, optionally substituted tetrazolyl, and optionally substituted triazolyl (e.g., -NH 2 substituted). Representative compounds of the above embodiment include Examples A l , A2, A3, A4, A8, A9, A 10, Al l , A12, A27, A28, A34, A35, A42, A49, A50, B l , C I , C2, C3, C4, C5, C6, C7, C8, C9, C IO, C I 1 , C 12, C 13, C 14, C 1 5, C 16, C17, C18, and C39.

Each of the inhibitor compounds of the invention contain asymmetric carbon centers and give rise to stereoisomers (i.e., optical isomers such as diastereomers and enantiomers). It will be appreciated that the present invention includes such diastereomers as well as their racemic and resolved enantiomerically pure forms. It will also be appreciated that in certain configurations, the relative stereochemistry of certain groups may be depicted as "cis" or "trans" when absolute stereochemistry is not shown.

Some of the compounds described herein contain olefinic double bonds, and unless specified otherwise, are meant to include both E and Z geometric isomers.

When the compounds of the present invention are basic, salts may be prepared from pharmaceutically acceptable non-toxic acids, including inorganic and organic acids. Examples of such acids include acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, and p-toluenesulfonic acids.

The invention is described using the following definitions unless otherwise indicated.

As used herein, the term "alkyl" refers to a saturated or unsaturated, branched, straight-chain or cyclic monovalent hydrocarbon group derived by the removal of one hydrogen atom from a single carbon atom of a parent alkane, alkene, or alkyne. Representative alkyl groups include methyl; ethyls such as ethanyl, ethenyl, ethynyl; propyls such as propan-l -yl, propan-2-yl, cyclopropan-l -yl, prop-l-en- l -yl, prop-l -en-2- yl, prop-2-en-l -yl (allyl), cycloprop- l -en- l -yl; cycloprop-2-en- l -yl, prop-l -yn- l-yl, and prop-2-yn- l -yl; butyls such as butan-l-yl, butan-2-yl, 2-methyl-propan-1 -yl, 2-methyl- propan-2-yl, cyclobutan-l -yl, but-l -en-l-yl, but- l-en-2-yl, 2-methyl-prop-l -en-l -yl, but- 2-en- l -yl, but-2-en-2-yl, buta- l ,3-dien-l -yl, buta-l ,3-dien-2-yl, cyclobut-l -en-l -yl, cyclobut-l-en-3-yl, cyclobuta-l ,3-dien-l-yl, but-l -yn-l -yl, but-l -yn-3-yl, and but-3-yn- l - yl; and the like. Where a specific level of saturation is intended, the expressions "alkanyl," "alkenyl," and "alkynyl" are used. Alkyl groups include cycloalkyl groups. The term "cycloalkyl" refers to mono-, bi-, and tricyclic alkyl groups having the indicated number of carbon atoms. Representative cycloalkyl groups include cyclopropyl, cyclopentyl, cycloheptyl, adamantyl, cyclododecylmethyl, and 2-ethy 1-1- bicyclo[4.4.0]decyl groups. The alkyl group may be unsubstituted or substituted as described below.

"Alkanyl" refers to a saturated branched, straight-chain, or cyclic alkyl group.

Representative alkanyl groups include methanyl; ethanyl; propanyls such as propan-l-yl, propan-2-yl(isopropyl), and cyclopropan-l-yl; butanyls such as butan-l-yl, butan-2-yl (sec-butyl), 2-methyl-propan-l-yl(isobutyl), 2-methyl-propan-2-yl(t-butyl), and cyclobutan-l-yl; and the like. The alkanyl group may be substituted or unsubstituted. Representative alkanyl group substituents include

— R,4,— ORi4,— SR.4,— NR,4(Ri S ),

— ,— CX 3 ,— CN,— N0 2 ,

— C(=0)R,4,— C(=0)OR,4,— C(=0)NR, 4 (R l5 ),— C(=0)SR, 4 ,

— C(=S)Ri4,— C(=S)OR,4,— C(=S)NR, 4 (R, 5 ),— C(=S)SR, 4 ,

— NR| 4 C(=0)NR| 4 (Ri5),— NR 14 (=NR I4 )NR| 4 (R, 5 ),— NR, 4 C(=S)NR, 4 (R 15 ),

— S(=0) 2 Ri4,— S(=0) 2 OR, 4 ,— S(=0) 2 NR| 4 (Ri5),

— OC(=0)Ri4,— OC(=0)OR, 4 ,— OC(=0)NR| 4 (R| 5 ),— OC(=0)SR 14 ,

— OS(=0) 2 OR, 4 ,— OS(=0) 2 NR| 4 (R| 5 ), and

— OP(=0) 2 (OR 14 ),

wherein each X is independently a halogen; and R) 4 and R15 are independently hydrogen, C1-C6 alkyl, C6-C14 aryl, arylalkyi, C3-C10 heteroaryl, and heteroarylalkyl, as defined herein.

In certain embodiments, two hydrogen atoms on a single carbon atom can be replaced with =0, =NR] 2 , or =S.

"Alkenyl" refers to an unsaturated branched, straight-chain, cyclic alkyl group, or combinations thereof having at least one carbon-carbon double bond derived by the removal of one hydrogen atom from a single carbon atom of a parent alkene. The group may be in either the cis or trans conformation about the double bond(s). Representative alkenyl groups include ethenyl; propenyls such as prop-l-en-l-yl, prop-l-en-2-yl, prop-2- en-l-yl (allyl), prop-2-en-2-yl, and cycloprop-l-en-l-yl; cycloprop-2-en-l-yl; butenyls such as but-l-en-l-yl, but-l-en-2-yl, 2-methyl-prop-l-en-l-yl, but-2-en-l-yl, but-2-en-l- yl, but-2-en-2-yl, buta-l ,3-dien-l -yl, buta-l ,3-dien-2-yl, cyclobut-l -en-l -yl, cyclobut- 1 - en-3-yl, and cyclobuta- l ,3-dien-l -yl; and the like. The alkenyl group may be substituted or unsubstituted. Representative alkenyl group substituents include

— R| 4,

—X,— CX 3 ,— CN,

— C(=0)R|4,— C(=0)OR 14 ,— C(=0)NR, 4 (R 15 ),— C(=0)SR, 4 ,

— C(=NR| 4 )Ri 4,— C(=NR, 4 )OR,4,— C(=NR 14 )NRi 4 (R, s ),— C(=NR, 4 )SR, 4 , — C(=S)R 14) — C(=S)OR | 4 ,— C(=S)NR, 4 (R, s),— C(=S)SR, 4>

wherein each X is independently a halogen; and R| 4 and R 1 5 are independently hydrogen, C 1 -C6 alkyl, C6-C14 aryl, arylalkyl, C3-C10 heteroaryl, and heteroarylalkyl, as defined herein.

"Alkynyl" refers to an unsaturated branched, straight-chain, or cyclic alkyl group having at least one carbon-carbon triple bond derived by the removal of one hydrogen atom from a single carbon atom of a parent alkyne. Representative alkynyl groups include ethynyl; propynyls such as prop-l-yn-l-yl and prop-2-yn- l -yl; butynyls such as but-l -yn-l -yl, but-l-yn-3-yl, and but-3-yn- l -yl; and the like. The alkynyl group may be substituted or unsubstituted. Representative alkynyl group substituents include those as described above for alkenyl groups.

The term "haloalkyl" refers to an alkyl group as defined above having the one or more hydrogen atoms replaced by a halogen atom. Representative haloalkyl groups include halomethyl groups such as chloromethyl, fluoromethyl, and trifluoromethyl groups; and haloethyl groups such as chloroethyl, fluoroethyl, and perfluoroethyl groups. The term "heteroalkyl" refers to an alkyl group having the ind icated number of carbon atoms and where one or more of the carbon atoms is replaced with a heteroatom selected from O, N, or S. Where a specific level of saturation is intended, the expressions "heteroalkanyl," "heteroalkenyl," and "heteroalkynyl" are used. Representative heteroalkyl groups include ether, amine, and thioether groups. Heteroalkyl groups include heterocyclyl groups. The term "heterocyclyl " refers to a 5- to 1 0-membered non- aromatic mono- or bicyclic ring containing 1 -4 heteroatoms selected from O, S, and N. Representative heterocyclyl groups include pyrrolidinyl, piperidinyl, piperazinyl, tetrahydrofuranyl, tetrahydropuranyl, and morpholinyl groups. The heteroalkyl group may be substituted or unsubstituted. Representative heteroalkyl substituents include — Ri 4,— ORi 4,— SR, 4,— Ri4(Ri 5),

—X,— CX 3 ,— CN,— NO2,

— C(=0)R, 4,— C(=0)OR, 4,— C(=0)NR, 4 (Ris),— C(=0)SR 14>

— C(=NR, 4 )Ri 4,— C(=NR, 4 )OR 14 ,— C(=NR, 4 )NR, 4 (R, 5),— C(=NR, 4 )SR 14 ,

— C(=S)R,4,— C(=S)ORi4,— C(=S)NR, 4 (R| 5 ),— C(=S)SR, 4 ,

— NR, 4 C(=0)NR, 4 (R, s),— NR, 4 (=NR, 4 )NR, 4 (R, 5 ),— NR, 4 C(=S)NR , 4 (R, 5 ),

— S(=0) 2 R, 4 ,— S(=0) 2 OR 14 ,— S(=0) 2 NR 14 (R 15 ),

— OC(=0)R, 4,— OC(=0)OR,4,— OC(=0)NR, 4 (R, 5),— OC(=0)SR, 4 ,

— OS(=0) 2 OR M ,— OS(=0) 2 NR, 4 (R, 5 ), and

_ OP(=0) 2 (OR 14 ),

wherein each X is independently a halogen; and R| 4 and R15 are independently hydrogen, C 1 -C6 alkyl, C6-C 14 aryl, arylalkyi, C3-C 10 heteroaryl, and heteroarylalkyl, as defined herein.

In certain embodiments, two hydrogen atoms on a single carbon atom can be replaced with =0, =NR| 2 , or =S.

The term "alkoxy" refers to an alkyl group as described herein bonded to an oxygen atom. Representative C1 -C3 alkoxy groups include methoxy, ethoxy, propoxy, and isopropoxy groups.

The term "alkylamino" refers an alkyl group as described herein bonded to a nitrogen atom. The term "alkylamino" includes monoalkyl- and dialkylaminos groups. Representative C 1 -C6 alkylamino groups include methylamino, dimethylamino, ethylamino, methylethylamino, diethylamino, propylamino, and isopropylamino groups.

The term "alkylthio" refers an alkyl group as described herein bonded to a sulfur atom. Representative C 1 -C6 alkylthio groups include methylthio, propylthio, and isopropylthio groups.

The term "aryl" refers to a monovalent aromatic hydrocarbon group derived by the removal of one hydrogen atom from a single carbon atom of a parent aromatic ring system. Suitable aryl groups include groups derived from aceanthrylene, acenaphthylene, acephenanthrylene, anthracene, azulene, benzene, chrysene, coronene, fluoranthene, fluorene, hexacene, hexaphene, hexalene, as-indacene, s-indacene, indane, indene, naphthalene, octacene, octaphene, octalene, ovalene, penta-2,4-diene, pentacene, pentalene, pentaphene, perylene, phenalene, phenanthrene, picene, pleiadene, pyrene, pyranthrene, rubicene, triphenylene, trinaphthalene, and the like. In certain embodiments, the aryl group is a C5-C 14 aryl group. In other embodiments, the aryl group is a C5-C 10 aryl group. The number of carbon atoms specified refers to the number of carbon atoms in the aromatic ring system. Representative aryl groups are phenyl, naphthyl, and cyclopentadienyl . The aryl group may be substituted or un substituted. Representative aryl group substituents include

— Ri ,—OR, 4,— SR, 4,— NRH(RI S),

—X,— CX 3 ,— CN,— N0 2 ,

— C(=NR I 4 )R,4,— C(=NR, 4 )OR 14 ,— C(=NR M )NRi 4 (R, 5 ),— C(=NR, 4 )SR 14(

— C(=S)R, 4,— C(=S)OR, 4 ,— C(=S)NR 14 (R 15 ),— C(=S)SR, 4 ,

— NR 14 C(=0)NR M (Ri 5),— NRi 4 (=NR 15 )NR 14 (R, 5),— NR| 4 C(=S)NR 14 (R 15 ),

— S(=0) 2 Ri4,— S(=0) 2 OR 14 ,— S(=0) 2 NR, 4 (R, 5 ),

— OS(=0) 2 ORi4,— OS(=0) 2 NR, 4 (R, 5 ), and

-OP(=0) 2 (OR, 4 ),

wherein each X is independently a halogen; and R14 and R15 are independently hydrogen, C 1 -C6 alkyl, C6-C14 aryl, arylalkyl, C3-C10 heteroaryl, and heteroarylalkyl, as defined herein.

The term "aralkyl" refers to an alkyl group as defined herein with an aryl group, optionally substituted, as defined herein substituted for one of the alkyl group hydrogen atoms. Suitable aralkyl groups include benzyl, 2-phenylethan- l -yl, 2-phenylethen-l -yl, naphthylmethyl, 2-naphthylethan-l-yl, 2-naphthylethen- l-yl, naphthobenzyl, 2-naphthophenyIethan-l-yl, and the like. Where specific alkyl moieties are intended, the terms aralkanyl, aralkenyl, and aralkynyl are used. In certain embodiments, the aralkyl group is a C6-C20 aralkyl group, (e.g., the alkanyl, alkenyl, or alkynyl moiety of the aralkyl group is a C1-C6 group and the aryl moiety is a C5-C 14 group). In other embodiments, the aralkyl group is a C6-C13 aralkyl group (e.g., the alkanyl, alkenyl, or alkynyl moiety of the aralkyl group is a C 1 -C3 group and the aryl moiety is a C5-C 10 aryl group. In certain embodiments, the aralkyl group is a benzyl group.

The term "heteroaryl" refers to a monovalent heteroaromatic group derived by the removal of one hydrogen atom from a single atom of a parent heteroaromatic ring system, which may be monocyclic or fused ring (i.e., rings that share an adjacent pair of atoms). A "heteroaromatic" group is a 5- to 14-membered aromatic mono- or bicyclic ring containing 1 -4 heteroatoms selected from O, S, and N. Representative 5- or 6-membered aromatic monocyclic ring groups include pyridine, pyrimidine, pyridazine, furan, thiophene, thiazole, oxazole, and isooxazole, Representative 9- or 10-membered aromatic bicyclic ring groups include benzofuran, benzothiophene, indole, pyranopyrrole, benzopyran, quionoline, benzocyclohexyl, and naphthyridine. Suitable heteroaryl groups include groups derived from acridine, arsindole, carbazole, β-carbol ine, chromane, chromene, cinnoline, furan, imidazole, indazole, indole, indoline, indolizine, isobenzofuran, isochromene, isoindole, isoindoline, isoquinoline, isothiazole, isoxazole, naphthyridine, oxadiazole, oxazole, perimidine, phenanthridine, phenanthroline, phenazine, phthalazine, pteridine, purine, pyran, pyrazine, pyrazole, pyridazine, pyridine, pyrimidine, pyrrole, pyrrolizine, quinazoline, quinoline, quinolizine, quinoxaline, tetrazole, thiadiazole, thiazole, thiophene, triazole, xanthene, and the like. In certain embodiments, the heteroaryl group is a 5-14 membered heteroaryl group. In other embodiments, the heteroaryl group is a 5-10 membered heteroaryl group. Preferred heteroaryl groups are those derived from thiophene, pyrrole, benzothiophene, benzofuran, indole, pyridine, quinoline, imidazole, oxazole, and pyrazine. The heteroaryl group may be substituted or unsubstituted. Representative heteroaryl group substituents include those described above for aryl groups.

The term "heteroarylalkyl" refers to an alkyl group as defined herein with a heteroaryl group, optionally substituted, as defined herein substituted for one of the alkyl group hydrogen atoms. Where specific alkyl moieties are intended, the terms heteroarylalkanyl, heteroarylalkenyl, or heteroarylalkynyl are used. In certain embodiments, the heteroarylalkyl group is a 6-20 membered heteroarylalkyl (e.g., the alkanyl, alkenyl or alkynyl moiety of the heteroarylalkyl is a C I -C6 group and the heteroaryl moiety is a 5- 14-membered heteroaryl group. In other embodiments, the heteroarylalkyl group is a 6-13 membered heteroarylalkyl (e.g., the alkanyl, alkenyl or alkynyl moiety is C 1 -C3 group and the heteroaryl moiety is a 5-10-membered heteroaryl group). The term "acyl" group refers to the -C(=0)— R' group, where R' is selected from optionally substituted alkyl, optionally substituted aryl, and optionally substituted heteroaryl, as defined herein.

The term "halogen" or "halo" refers to fluoro, chloro, bromo, and iodo groups. The term "substituted" refers to a group in which one or more hydrogen atoms are each independently replaced with the same or different substituent(s).

Representative compounds of the invention and related intermediates were prepared from commercially available starting materials or starting materials prepared by conventional synthetic methodologies. Representative compounds of the invention were prepared according to Methods A to P as described below and illustrated in FIGURES 1 -3. The preparations of certain intermediates (1-1 to 1-15) useful in the preparation of compounds of the invention are described in the Synthetic Intermediate section below.

FIGURES 1 -3 present schematic illustrations of representative synthetic pathways for the preparation of representative compounds of the invention P5-P4-P3-P2-P1. As used herein, "P5-P4-P3-P2-P 1 " refers to compounds of the invention prepared from five (5) components: P I , P2, P3, P4, and P5. Protected version of the components useful in the preparation of the compounds of the invention are designated as, for example, "PG-P2," "PG-P2-P1 ," "PG-3," and "PG-P3-P2-P1 ," where "PG" is refers to a protecting group that allows for the coupling of, for example, PI to P2 or P3 to P 1 -P2, and that is ultimately removed to provide, for example, P1 -P2 or P1 -P2-P3.

FIGURE 1 is a schematic illustration of a representative synthetic pathway for the preparation of representative compounds of the invention P5-P4-P3-P2-P1 starting from PI . In this pathway, compound P5-P4-P3-P2-P1 is prepared in a stepwise manner starting with PI by sequential coupling steps, separated as appropriate by deprotection steps and other chemical modifications. As shown in FIGURE 1 , PI is coupled with PG-P2 to provide PG-P2-P1 , which is then deprotected to provide P2-P1 and ready for coupling with the next component, PG-P3. The process is continued with subsequent couplings PG-P4 with P3-P2-P1 and PG-P5 with P4-P3-P2-P1 to ultimately provide P5-P4-P3-P2-P 1 . Examples A1 -A57 were prepared by this pathway.

FIGURE 2 is a schematic illustration of another representative synthetic pathway for the preparation of representative compounds of the invention P5-P4-P3-P2-P1 starting from P5 , In this pathway, compound P5-P4-P3-P2-P1 is prepared in a stepwise manner starting with P5 by sequential coupling steps, separated as appropriate by deprotection steps and other chemical modifications. As shown in FIGURE 2, P5 is coupled with PG-P4 to provide P5-P4-PG, which is then deprotected to provide P5-P4 and ready for coupling with the next component, P3-PG. The process is continued with subsequent couplings PG-P2 with P5-P4-P3 and PG-P l with P5-P4-P3-P2 to ultimately provide P5-P4-P3-P2-P1. Examples B 1-B7 were prepared by this method.

FIGURE 3 is a schematic illustration of a further representative synthetic pathway for the preparation of representative compounds of the invention P5-P4-P3-P2-P1 starting from a component other than PI or P5. In this pathway, compound P5-P4-P3-P2-P1 is prepared in a stepwise manner starting with P2 by sequential coupling steps, separated as appropriate by deprotection steps and other chemical modifications. As shown in FIGURE 3, there are multiple pathways to P5-P4-P3-P2-P1. Pathways are shown involving representative Synthetic Intermediates (1-1 , 1-2, 1-3, 1-4, 1-6, 1-7, 1-9, 1-10, 1- 1 1 , and 1- 12). Examples C I -C39 were prepared by this method .

The preparation of representative compounds and their characterization are described in Examples A l to A57, B l to B7, and C I to C39. The structures of representative compounds are set forth in Table 1.

Table 1 . Representative Compounds.

A general kinetic enzyme assay useful for determining the inhibitory activity of the compounds of the invention is described in Example Dl .

Granzyme B enzymatic inhibition assays are described in Examples D2 and D6. The compounds of the invention identified in Table 1 exhibited Granzyme B inhibitory activity. In certain embodiments, select compounds exhibited IC50 <50,000 nM. In other embodiments, select compounds exhibited IC50 <10,000 nM. In further embodiments, select compounds exhibited IC50 <1 ,000 nM. In still further embodiments, select compounds exhibited IC50 <100 nM. In certain embodiments, select compounds exhibited IC50 from 10 nM to 100 nM, preferably from 1 nM to 10 nM, more preferably from 0.1 nM to 1 nM, and even more preferably from 0.01 nM to 0.1 nM.

Caspase enzymatic inhibition assays are described in Examples D3 and D7. None of the compounds of the invention tested demonstrated an ability to significantly inhibit any of the caspases evaluated at a concentration of 50 μΜ. In certain embodiments, the compounds exhibited less than 50% inhibition at 50 μΜ. In other embodiments, the compounds exhibited greater than 50% inhibition at 50 μΜ, but less than 10% inhibition at 25 μΜ. The results demonstrate that select compounds of the invention selectively inhibit Granzyme B without significantly inhibiting caspases.

A method for determining kinetic solubility is described in Example D4. The results demonstrate that select compounds of the invention have significantly greater solubil ity than Willoughby 20, a representative Granzyme B inhibitor known in the art.

Pharmaceutical Compositions

The pharmaceutical compositions of the present invention include an inhibitor compound of the invention (e.g., a compound of Formulae (I), (II), (III), or (IV)) as an active ingredient or a pharmaceutically acceptable salt thereof in combination with a pharmaceutically acceptable carrier, and optionally other therapeutic ingredients.

The term "pharmaceutically acceptable salts" refers to salts prepared from pharmaceutically acceptable bases including inorganic bases and organic bases. Representative salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic, manganous, ammonium, potassium, sodium, and zinc salts. Representative salts derived from pharmaceutically acceptable organic bases include salts of primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N,N- dibenzylethylenediamine, diethylamine, 2-diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethyl-morpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine, isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, and trimethamine.

Compositions can include one or more carriers acceptable for the mode of administration of the preparation, be it by topical administration, lavage, epidermal administration, sub-epidermal administration, dermal administration, subdermal administration, transdermal administration, subcutaneous administration, systemic administration, injection, inhalation, oral, or any other mode suitable for the selected treatment. Topical administration includes administration to external body surfaces (e.g., skin) as well as to internal body surfaces (e.g., mucus membranes for vaginal or rectal applications by, for example, suppositories). Suitable carriers are those known in the art for use in such modes of administration.

Suitable compositions can be formulated by means known in the art and their mode of administration and dose determined by a person of skill in the art. For parenteral administration, the compound can be dissolved in sterile water or saline or a pharmaceutically acceptable vehicle used for administration of non-water soluble compounds. For enteral administration, the compound can be administered in a tablet, capsule, or dissolved or suspended in liquid form. The tablet or capsule can be enteric coated, or in a formulation for sustained release. Many suitable formulations are known including, polymeric or protein microparticles encapsulating a compound to be released, ointments, pastes, gels, hydrogels, foams, creams, powders, lotions, oils, semi-solids, soaps, medicated soaps, shampoos, medicated shampoos, sprays, films, or solutions which can be used topically or locally to administer a compound. A sustained release patch or implant may be employed to provide release over a prolonged period of time. Many techniques known to one of skill in the art are described in Remington: the Science & Practice of Pharmacy by Alfonso Gennaro, 20th ed., Williams & Wilkins, (2000). Formulations can contain excipients, polyalkylene glycols such as polyethylene glycol, oils of vegetable origin, or hydrogenated naphthalenes. Biocompatible, biodegradable lactide polymer, lactide/glycolide copolymer, or polyoxyethylene-polyoxypropylene copolymers can be used to control the release of a compound. Other potentially useful delivery systems for a modulatory compound include ethylene-vinyl acetate copolymer particles, osmotic pumps, implantable infusion systems, and liposomes. Formulations can contain an excipient, for example, lactose, or may be aqueous solutions containing, for example, polyoxyethylene-9-lauryl ether, glycocholate, and deoxycholate, or can be an oily solution for administration in the form of drops, as a gel, or for other semi-solid formulation.

Compounds or pharmaceutical compositions in accordance with this invention or for use in the methods disclosed herein can be administered in combination with one or more other therapeutic agents as appropriate. Compounds or pharmaceutical compositions in accordance with this invention or for use in the methods d isclosed herein can be administered by means of a medical device or appliance such as an implant, graft, prosthesis, stents, and wound dressings. Also, implants can be devised that are intended to contain and release such compounds or compositions. An example would be an implant made of a polymeric material adapted to release the compound over a period of time.

One skilled in the art will appreciate that suitable methods of administering a Granzyme B inhibitor directly to the eye are available (i.e., invasive and noninvasive methods). Although more than one route can be used to administer the Granzyme B inhibitor, a particular route can provide a more immediate and more effective reaction than another route. The present use is not dependent on the mode of administering the agent to an animal, preferably a human, to achieve the desired effect, and the described routes of administration are exemplary. As such, any route of administration is appropriate so long as the agent contacts an ocular cell. Thus, the Granzyme B inhibitor can be appropriately formulated and administered in the form of an injection, eye lotion, ointment, and implant.

The Granzyme B inhibitor can be applied, for example, systemically, topically, intracamerally, subconjunctival^, intraocularly, retrobulbarly, pcriocularly (e.g., subtenon delivery), subretinally, or suprachoroidally. In certain cases, it can be appropriate to administer multiple applications and employ multiple routes to ensure sufficient exposure of ocular cells to the Granzyme B inhibitor (e.g., subretinal and intravitreous). Multiple applications of the Granzyme B inhibitor can also be required to achieve the desired effect.

Depending on the particular case, it may be desirable to non-invasively admin ister the Granzyme B inhibitor to a patient. For instance, if multiple surgeries have been performed, the patient displays low tolerance to anesthetic, or if other ocular-related disorders exist, topical administration of the Granzyme B inhibitor may be most appropriate. Topical formulations are well known to those of skill in the art. Such formulations are suitable in the context of the use described herein for application to the skin or to the surface of the eye. The use of patches, corneal shields (see, U.S. Patent No. 5, 185, 152), and ophthalmic solutions (see, e.g., U.S. Patent No. 5,710, 182) and ointments is within the skill in the art.

The Granzyme B inhibitor also can be present in or on a device that allows controlled or sustained release, such as an ocular sponge, meshwork, mechanical reservoir, or mechanical implant. Implants (see U .S. Patent Nos. 5,443,505, 4,853,224 and 4,997,652), devices (see U.S. Patent Nos. 5,554, 187, 4,863,457, 5,098,443 and 5,725,493), such as an implantable device (e.g., a mechanical reservoir, an intraocular device or an extraocular device with an intraocular conduit, or an implant or a device comprised of a polymeric composition are particu larly useful for ocular administration of the expression vector). The Granzyme B inhibitor also can be administered in the form of sustained-release formulations (see U.S. Patent No. 5,378,475) comprising, for example, gelatin, chondroitin sulfate, a polyphosphoester, such as iw-2-hydroxyethyl- terephthalate, or a polylactic-glycolic acid.

When used for treating an ocular disease the Granzyme B inhibitor is administered via an ophthalmologic instrument for del ivery to a specific region of an eye. Use of a specialized ophthalmologic instrument ensures precise administration while minimizing damage to adjacent ocular tissue. Delivery of the Granzyme B inhibitor to a specific region of the eye also limits exposure of unaffected cells to the Granzyme B inhibitor. A preferred ophthalmologic instrument is a combination of forceps and subretinal needle or sharp bent cannula. Alternatively, the Granzyme B inhibitor can be administered using invasive procedures, such as, for instance, intravitreal injection or subretinal injection, optionally preceded by a vitrectomy, or periocular (e.g., subtenon) delivery. The pharmaceutical composition of the invention can be injected into different compartments of the eye (e.g., the vitreal cavity or anterior chamber).

While intraocular injection is preferred, injectable compositions can also be administered intramuscularly, intravenously, intraarterially, and intraperitoneal ly. Pharmaceutically acceptable carriers for injectable compositions are well-known to those of ordinary skill in the art (see Pharmaceutics and Pharmacy Practice, J. B. Lippincott Co., Philadelphia, Pa., Banker and Chalmers, eds., pages 238-250 ( 1982), and ASHP Handbook on Injectable Drugs, Toissel, 4th ed., pages 622-630 ( 1 86)).

An "effective amount" of a Granzyme B inhibitor or a pharmaceutical composition of the invention as described herein includes a therapeutically effective amount or a prophylactically effective amount. A "therapeutically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired therapeutic result, such as reduced levels of Granzyme B activity. A therapeutically effective amount of a compound may vary according to factors such as the disease state, age, sex, and weight of the subject, and the ability of the compound to elicit a desired response in the subject. Dosage regimens can be adjusted to provide the optimum therapeutic response. A therapeutically effective amount is also one in which any toxic or detrimental effects of the compound are outweighed by the therapeutically beneficial effects. A "prophylactically effective amount" refers to an amount effective, at dosages and for periods of time necessary, to achieve the desired prophylactic result, such as Granzyme B activity. Typically, a prophylactic dose is used in subjects prior to or at an earlier stage of disease, so that a prophylactically effective amount may be less than a therapeutically effective amount.

It is to be noted that dosage values can vary with the severity of the condition to be alleviated. For any particular subject, specific dosage regimens can be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions. Dosage ranges set forth herein are exemplary only and do not limit the dosage ranges that can be selected by a medical practitioner. The amount of active compound(s) in the composition can vary according to factors such as the disease state, age, sex, and weight of the subject. Dosage regimens can be adjusted to provide the optimum therapeutic response. For example, a single bolus can be administered, several divided doses can be administered over time or the dose can be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It may be advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.

In general, compounds of the invention should be used without causing substantial toxicity. Toxicity of the compounds of the invention can be determined using standard techniques, for example, by testing in cell cultures or experimental animals and determining the therapeutic index (i.e., the ratio between the LD 50 , the dose lethal to 50% of the population, and the LD 10( the dose lethal to 1 00% of the population). In some circumstances however, such as in severe disease conditions, it may be necessary to administer substantial excesses of the composition.

Methods of Use

In a further aspect, the invention provides methods of using the compounds of the invention as Granzyme B inhibitors.

In one embodiment, the invention provides a method for inhibiting Granzyme B in a subject. In the method, an effective amount of a compound of the invention (e.g., a compound of Formulae (I), (II), (III), or (IV)) is administered to a subject in need thereof.

In another embodiment, the invention provides a method for treating a disease, disorder, or condition treatable by inhibiting Granzyme B. In the method, a therapeutically effective amount of a compound of the invention (e.g., a compound of Formulae (I), (II), (III), or (IV)) is administered to a subject in need thereof.

As used herein, the term "disease, disorder, or condition treatable by inhibiting

Granzyme B" refers to a disease, disorder, or condition in which Granzyme B is involved in the pathway related to for the disease, disorder, or condition, and that inhibiting Granzyme B results in the treatment or prevention of the disease, disorder, or condition.

Representative methods of treatment using the compounds of the invention include those described for Granzyme B inhibitors in WO 2007/101354 (Methods of Treating, Reducing, and Inhibiting the Appearance of Ageing in the Skin), WO 2009/0431 70 (Treatment of Dissection, Aneurysm, and Atherosclerosis Using Granzyme B Inhibitors), WO 2012/076985 (Granzyme B Inhibitor Compositions, Methods and Uses for Promoting Wound Healing), each expressly incorporated herein by reference in its entirety. The compounds of the invention are useful for treating, reducing, and inhibiting the appearance of aging of the skin; treating dissection, aneurysm, and atherosclerosis; and promoting wound healing.

Other disease and disorders described as treatable using the Granzyme B inhibitors are disclosed in WO 2003/065987 (Granzyme B Inhibitors), expressly incorporated herein by reference in its entirety. Disease and disorders described as treatable by Granzyme B inhibitors in this reference include autoimmune or chronic inflammatory diseases, such as systemic lupus erythematosis, chronic rheumatoid arthritis, type I diabetes mellitus, inflammatory bowel disease, biliary cirrhosis, uveitis, multiple sclerosis, Crohn's disease, ulcerative colitis, bullous pemphigoid, sarcoidosis, psoriasis, autoimmune myositis, Wegener's granulomatosis, ichthyosis, Graves ophthalmopathy, asthma, scleroderma and Sjogren's syndrome. The Granzyme B inhibitors described in the reference are noted as more particularly useful to treat or prevent diseases or disorders including diseases or disorders resulting from transplantation of organs or tissue, graft-versus-host diseases brought about by transplantation, autoimmune syndromes including rheumatoid arthritis, systemic lupus erythematosus, Hashimoto's thyroiditis, multiple sclerosis, myasthenia gravis, type I diabetes, uveitis, posterior uveitis, allergic encephalomyelitis, glomerulonephritis, postinfectious autoimmune diseases including rheumatic fever and post-infectious glomerulonephritis, inflammatory and hyperproliferative skin diseases, psoriasis, atopic dermatitis, contact dermatitis, eczematous dermatitis, seborrhoeic dermatitis, l ichen planus, pemphigus, bullous pemphigoid, epidermolysis bullosa, urticaria, angioedemas, vasculitis, erythema, cutaneous eosinophilia, lupus erythematosus, acne, alopecia areata, keratoconjunctivitis, vernal conjunctivitis, uveitis associated with Behcet's disease, keratitis, herpetic keratitis, conical cornea, dystrophia epithelialis corneae, corneal leukoma, ocular pemphigus, Mooren's ulcer, scleritis, Graves' opthalmopathy, Vogt- Koyanagi-Harada syndrome, sarcoidosis, pollen allergies, reversible obstructive airway disease, bronchial asthma, allergic asthma, intrinsic asthma, extrinsic asthma, dust asthma, chronic or inveterate asthma, late asthma and airway hyper-responsiveness, bronchitis, gastric ulcers, vascular damage caused by ischemic diseases and thrombosis, ischemic bowel diseases, inflammatory bowel diseases, necrotizing enterocolitis, intestinal lesions associated with thermal burns, coeliac diseases, proctitis, eosinophilic gastroenteritis, mastocytosis, Crohn's disease, ulcerative colitis, migraine, rhinitis, eczema, interstitial nephritis, Goodpasture's syndrome, hemolytic-uremic syndrome, diabetic nephropathy, multiple myositis, Guillain-Barre syndrome, Meniere's disease, polyneuritis, multiple neuritis, mononeuritis, radiculopathy, hyperthyroidism, Basedow's disease, pure red cell aplasia, aplastic anemia, hypoplastic anemia, idiopathic thrombocytopenic purpura, autoimmune hemolytic anemia, agranulocytosis, pernicious anemia, megaloblastic anemia, anerythroplasia, osteoporosis, sarcoidosis, fibroid lung, idiopathic interstitial pneumonia, dermatomyositis, leukoderma vulgaris, ichthyosis vu lgaris, photoallergic sensitivity, cutaneous T cell lymphoma, arteriosclerosis, atherosclerosis, aortitis syndrome, polyarteritis nodosa, myocardosis, scleroderma, Wegener's granuloma, Sjogren's syndrome, adiposis, eosinophilic fascitis, lesions of gingiva, periodontium, alveolar bone, substantia ossea dentis, glomerulonephritis, male pattern alopecia or alopecia senilis by preventing epilation or providing hair germination and/or promoting hair generation and hair growth, muscular dystrophy, pyoderma and Sezary's syndrome, Addison's disease, ischemia-reperfusion injury of organs which occurs upon preservation, transplantation or ischemic disease, endotoxin-shock, pseudomembranous colitis, colitis caused by drug or radiation, ischemic acute renal insufficiency, chronic renal insufficiency, toxinosis caused by lung-oxygen or drugs, lung cancer, pulmonary emphysema, cataracta, siderosis, retinitis pigmentosa, senile macular degeneration, vitreal scarring, corneal alkali burn, dermatitis erythema multiforme, linear IgA ballous dermatitis and cement dermatitis, gingivitis, periodontitis, sepsis, pancreatitis, diseases caused by environmental pollution, aging, carcinogenesis, metastasis of carcinoma and hypobaropathy, disease caused by histamine or leukotriene- C4 release, Behcet's disease, autoimmune hepatitis, primary biliary cirrhosis, sclerosing cholangitis, partial liver resection, acute liver necrosis, necrosis caused by toxin, viral hepatitis, shock, or anoxia, B-virus hepatitis, non-A/non-B hepatitis, cirrhosis, alcoholic cirrhosis, hepatic fai lure, fulminant hepatic failure, late-onset hepatic failure, "acute-on- chronic" liver failure, augmentation of chemotherapeutic effect, cytomegalovirus infection, HCMV infection, AIDS, cancer, senile dementia, trauma, and chronic bacterial infection. To the extent that the diseases and disorders noted in the reference are treatable by the Granzyme B inhibitors described in the reference, the Granzyme B inhibitors of the present invention are also useful in treating and/or ameliorating a symptom associated with these diseases and conditions.

Elevated Granzyme B levels have been identified in cells and tissues from subjects suffering from a variety of diseases and conditions including Rasmussen encephalitis, amyotrophic lateral sclerosis (ALS), chronic inflammation, Stevens-Johnson syndrome (SJS), toxic epidermal necrolysis (TEN), Kawasaki disease, idiopathic pulmonary fibrosis, chronic obstructive pulmonary disease (COPD), coronary artery disease (CAD), transplant vascular disease (TVD), restenosis, acute respiratory distress syndrome (ARDS), chronic obstructive sialadentis (associated with sialolithiasis), viti l igo, al lergic contact dermatitis (ACD), atopic dermatitis (AD), pityriasis rosea (PR), rheumatoid arthritis (RA), osteoarthritis (OA), vasculitic neuropathy, sensory perineuritis, ischemic stroke, spinal cord injury, myasthenia gravis (MG), lymphocytic gastritis, autoimmune cholangitis (AIC), nodular regenerative hyperplasia (NRH) of the l iver, achalasia, esophagitis, eosinophilic fasciitis, cryptorchidism, necrotizing lymphadenitis, Duchenne muscular dystrophy, facioscapulo humeral muscular dystrophy, and Higashi syndrome. Other diseases and conditions in which elevated Granzyme B levels have been identified include those described in WO 2009/043167 (Granzyme A and Granzyme B Diagnostics), expressly incorporated herein by reference in its entirety. The Granzyme B inhibitors of the invention may be useful for treating, alleviating or ameliorating a symptom of, diminishing the extent of, stabilizing, or ameliorating or palliating the diseases and conditions noted above in which elevated Granzyme B levels have been identified.

In the above methods, the administration of the Granzyme B inhibitor can be a systemic administration, a local administration (e.g., administration to the site, an inflamed microenvironment, an inflamed joint, an area of skin, a site of a myocardial infarct, an eye, a neovascularized tumor), or a topical administration to a site (e.g., a site of inflammation or a wound). The term "subject" or "patient" is intended to include mammalian organisms. Examples of subjects or patients include humans and non-human mammals, e.g., nonhuman primates, dogs, cows, horses, pigs, sheep, goats, cats, mice, rabbits, rats, and transgenic non-human animals. In specific embodiments of the invention, the subject is a human.

The term "administering" includes any method of delivery of a Granzyme B inhibitor or a pharmaceutical composition comprising a Granzyme B inhibitor into a subject's system or to a particular region in or on a subject. In certain embodiments, a moiety is administered topically, intravenously, intramuscularly, subcutaneously, intradermally, intranasally, orally, transcutaneously, intrathecal, intravitreally, intracerebral, or mucosally.

As used herein, the term "applying" refers to administration of a Granzyme B inhibitor that includes spreading, covering (at least in part), or laying on of the inhibitor. For example, a Granzyme B inhibitor may be applied to an area of inflammation on a subject or applied to, for example the eye or an area of inflammation by spreading or covering the surface of the eye with an inhibitor, by injection, oral or nasal administration.

As used herein, the term "contacting" includes contacting a cell or a subject with a Granzyme B inhibitor. Contacting also includes incubating the Granzyme B inhibitor and the cell together in vitro (e.g., adding the inhibitor to cells in culture) as well as administering the inhibitor to a subject such that the inhibitor and cells or tissues of the subject are contacted in vivo.

As used herein, the terms "treating" or "treatment" refer to a beneficial or desired result including, but not limited to, alleviation or amel ioration of one or more symptoms, dim inishing the extent of a disorder, stabilized (i.e., not worsening) state of a disorder, amelioration or palliation of the disorder, whether detectable or undetectable. "Treatment" can also mean prolonging survival as compared to expected survival in the absence of treatment.

Each reference cited is incorporated herein by reference in its entirety.

Abbreviations

As used herein, the following abbreviations have the indicated meanings. Ή NMR: proton nuclear magnetic resonance

19 F NMR: fluorine- 19 nuclear magnetic resonance

%lnh: Percent inhibition

Ac-IEPD-AMC: acetyl-isoleucyl-glutamyl-prolyl-aspartyl-(7-amino-4- methylcoumarin) substrate

ACN: acetonitrile

BHET: b s-2-hydroxyethyl-terephthalate

Boc: ier/-butoxycarbonyl

CHAPS: 3-[(3-cholamidopropyl)dimethylammonio]-l -propanesulfonate DCM: dichloromethane

DIPEA: diisopropylethylamine

DMAP: 4-dimethylaminopyridine

DMF: dimethylformamide

DMSO: dimethylsulfoxide

DMSO-i/6: dimethylsulfoxide-i¾

DTT: dithiothreitol

EDC: l -(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride EDTA: 2-({2-[5w(carboxymethyl)amino]ethyl} (carboxymethyl)amino)acetic acid

ESI: Electrospray ionization

EtOAc: ethyl acetate

eq.: equivalent(s)

GzmB: Granzyme B

HATU: 2-(7-aza-lH-benzotriazole-l-yl)- l , l , l , l-tetramethyluronium hexafluorophosphate

HC1: hydrochloric acid

HEPES: 4-(2-hydroxyethyl)-l -piperazineethanesulfonic acid

hGzmB: human Granzyme B

HPLC: high performance liquid chromatography

HOBt: 1 -hydroxy-benzotriazol

IC50: inhibitory concentration that provides 50% inhibition LC/MS: liquid chromatography / mass spectrometry

MeOH: methanol

mGzmB: murine Granzyme B

MS: mass spectrometry

m/z: mass to charge ratio.

Oxyma: ethyl 2-cyano-2-(hydroxyimino)acetate

PBS : phosphate buffered saline (pH 7.4)

RPM: revolution per minute

RT: room temperature

THF: tetrahydrofuran

TFA: trifluoroacetic acid

General Methods A-P

Representative compounds of the invention were prepared according to Methods A to P as described below and illustrated in FIGURES 1 -3.

The Willoughby 20 compound was synthesized by published procedures (see

Wil loughby et al ., Bioorg. Med. Chem. Lett. 12 (2002) 2197-2200 and WO 03/065987). It will be appreciated that in the following general methods and preparation of synthetic intermediates, reagent levels and relative amounts or reagents/intermediates can be changed to suit particular compounds to be synthesized, up or down by up to 50% without significant change in expected results.

Method A: General method for dcpmtection followed bv coupling reaction using

HC1 Solution in dioxane (4M, 5 ml) was added to respective carbamate compound (0.125 mmol) and stirred for 2 hrs at RT. The reaction mixture was concentrated to dryness under vacuum and swapped with MeOH (5 ml) three times, Resulting residue was dried well under vacuum and subjected to next reaction as it is. The residue obtained above, respective acid moiety (0. 125 mmol), EDC (0.19 mmol), HOBt (0.16 mmol) and DIPEA (0.5 mmol) were stirred in anhydrous DCM (5 ml) for 16 hrs. The reaction mixture was concentrated under vacuum to give the crude product which was purified on a C I 8 column using 10-50% MeOH in water to yield product as an off-white solid (35- 55%).

Method B: General method for de mlecl ion followed by reaction with anhydride.

HC1 Solution in d ioxane (4M, 5 ml) was added to a representative Boc-protected compound (0.125 mmol) and stirred for 2 hrs at RT. The reaction mixture was concentrated to dryness under vacuum and washed with MeOH (5 ml) three times.

Resulting residue was dried well under vacuum and subjected to next reaction as it is.

The residue obtained above, the respective anhydride moiety (0.125 mmol), and triethylamine (0.5 mmol) were added to anhydrous DCM (5 mL) and stirred for 16 hrs. The mixture was concentrated under vacuum to give the crude product which was purified on a C I 8 column using 10-50% MeOH in water to yield product as an off-white solid (40-60%). Method C: General method of coupling reaction using HATU/DIPEA.

The respective acid moiety (0.125 mmol), HATU (0.17 mmol), DIPEA (0.5 mmol) and respective amine moiety (0.125 mmol) were stirred in anhydrous DCM (5 ml) for 16 hrs. The reaction mixture was concentrated under vacuum to give the crude product which was purified on a C18 column using 10-50% MeOH in water (or sim i lar ratio as needed) to yield product as an off-white solid (35-55%).

Method D: General method of hydrolysis using LiOH.

To the stirring solution of the ester compound (0.08 mmol) in ethanol (1 ml) was added solution of lithium hydroxide monohydrate (0.4 mmol) in water (0.5 ml). After stirring the reaction mixture for 5 hrs at RT, the mixture was acidified using citric acid (saturated solution) and concentrated under vacuum to give the crude product which was purified on a C 18 column using 10-40% MeOH in water to yield product as an off-white solid (50-65%).

Method E: General method for Boc deprotection. HC1 Solution in dioxane (4 , 0.5 ml) was added to the respective carbamate compound (0.06 mmol) and stirred for 3 hrs at RT. The reaction mixture was concentrated under vacuum to give the crude product which was purified on a C I 8 column using 10-40% eOH in water to yield product as an off-white solid (50-60%).

Method F: General method for methanesulfonyl (mesyl) chloride mediated amino acid coupling.

A solution of an appropriate acid (1.0 mmol) in THF ( 10 mL) was cooled to -40 °C. To this solution was added DIPEA (1 .7 mmol) followed by CH 3 S0 2 C1 (1 .3 mmol). The reaction mixture was allowed to stir for 20 minutes before addition of an appropriate amine (such as (S)-methyl indol ine-2-carboxylate) ( 1 .1 mmol). After stirring for further 10 min at the same temperature the ice bath was removed and reaction mixture was allowed to warm up to room temperature. The reaction mixture was diluted with EtOAc and then washed with saturated aqueous solution of NaHC0 3 . The aqueous layer was then extracted with EtOAc (2x20 mL). Combined organic layer was washed with brine and dried over anhydrous Na 2 S04, filtered and concentrated in vacuo. Flash chromatography (MeOH: DCM 2-5%) afforded final compound.

Method G: EDC/Oxyma amino acid coupling.

To a mixture of an appropriate acid (0.05 mmol) in DMSO (0.5 mL) DIPEA (0.23 mmol), an appropriate acid (0.075 mmol), EDC (0.1 mmol), and oxyma (0.1 mmol) were added at RT. After stirring for 16 h the reaction mixture was purified by preparative HPLC (column: Ascentis C 18, 25 cm x 21.2 mm, Ι Ομιη, gradient 0%→100% methanol/water with 0.1 % TFA, 1 0 mL/min or similar) to furnish 1 1 mg of powder.

Method H: General method of acid chloride synthesis followed by amide formation.

Acid compound (1.5 mmol) and thionyl chloride (90 mmol) were stirred together for 1 hr. at room temperature. Thionyl chloride was removed by distillation under vacuum. The acid chloride was added to the stirring solution of L-isoleucine (1 .35 mmol) in NaOH (2N, 1 .8 ml) at 0 °C. The resulting reaction mixture was warmed to RT and stirred overnight. The reaction mixture was diluted with water (2 ml) and washed with diethyl ether (3 ml). Separated aqueous layer was acidified to pH 2 by adding mixture of 1 : 1 HCl-water. The precipitated solid was filtered and washed with water to get the product as a white to off-white solid (45-75%).

M ethod 1 : Genera! method Cor deprotecl ion fol lowed bv react ion with an hydride).

3) Acidify with formic acid

This method is an improved procedure for the method B. HC1 Solution in dioxane (4M, 5 ml) was added to a representative Boc-protected compound (0.125 mmol) and stirred for 2 hrs at RT. The reaction mixture was concentrated to dryness under vacuum and swapped with MeOH (5 ml) three times. The resulting residue was dried well under vacuum and subjected to next reaction as it was. The residue obtained above, the respective anhydride moiety (0.19 mmol, 1.5 eq.), and triethylamine (0.5 mmol, 4 eq.) were added to anhydrous DCM (5 mL) and stirred for 16 hrs. The mixture was acidified with formic acid and then concentrated under vacuum to give the crude product which was purified on a C I 8 column using 25-65% MeOH in water to yield product as an off- white solid (30-80%).

Method .1 : Genera l method lor deprotectkm fol lowed by reaction with a d i-acid.

OH O

HO

OH

O OH

3) Acidify with formic acid

HQ Solution in dioxane (4 , 5 ml) was added to a representative Boc-protected compound (0.125 mtnol) and stirred for 2 hrs at RT. The reaction mixture was concentrated to dryness under vacuum and swapped with MeOH (5 ml) three times. Resulting residue was dried well under vacuum and subjected to next reaction as it was. The residue obtained above, the respective di-acid moiety (0.62 mmol, 5 eq.), EDC (0.19 mmol), HOBt (0.15 mmol) and DIPEA (1.6 mmol, 13 eq.) were added to anhydrous DCM (5 mL) and stirred for 4 hrs. The mixture was acidified with formic acid and then concentrated under vacuum to give the crude product which was purified on a C 18 column using 25-65% MeOH in water to yield product as an off-white solid (30-80%).

Method : General method for Boc protection.

NaOH (0.5M),

di-fe/f-butyl

H,N dicarbonate

Dioxane

To respective amine compound (6.1 mmol) in dioxane (6 ml) and NaOH solution (0.5M, 12 ml) was added slowly solution of di-fer/-butyl dicarbonate (6.7 mmol) in dioxane (6 ml) at 0 °C. The reaction mixture was warmed to RT and stirred overnight. The reaction mixture was then washed with hexanes ( 10 m l). The separated water layer was acidified using saturated solution of citric acid and extracted with ethyl acetate (3 x 15 ml). The organic layer was washed with brine, separated, dried over sodium sulfate and concentrated to give Boc protected amine compound as off-white solid (65-90 %).

Method L: General method of hydrolysis followed by coupling reaction using EDC /

HOBt / D1PEA.

Η'Ή

To the stirring solution of the ester compound (0.1 1 mmol) in ethanol (4 ml) was added solution of lithium hydroxide monohydrate (0.23 mmol) in water (2 ml). After stirring the reaction mixture for 5 hrs at RT, the mixture was acidified using sat. citric acid solution and concentrated under vacuum to remove ethanol. The aqueous residue obtained was extracted with EtOAc (2 X 15 ml). Combined organic layer was dried over anhydrous Na2SC>4, fi ltered and concentrated in vacuo. Resulting residue was dried well under vacuum and subjected to next reaction as it was. The residue obtained above, the respective amine moiety (0.1 1 mmol), EDC (0.17 mmol), HOBT (0.15 mmol) and DIPEA (0.46 mmol) were stirred in DMF (4 ml) for 16 hrs. The mixture was concentrated under vacuum to give the crude product which was purified on a C I 8 column using 10-50% MeOH in water to yield product as an off-white solid (50-60%).

Method M: General method for EDC/HOBt/DIPEA coup ling of an intermediate existing as an HC1 salt uiid a free carboxylic acid.

RI / NH 2 HCI

O EDC, HOBt, DIPEA O

R OH DCM * R^ - R '

H

To an intermediate collected as an HC1 salt (0.125 mmol) were added the carboxylic acid (0.125 mmol), EDC (0.19 mmol), HOBt (0.1 6 mmol), and anhydrous DCM (5 ml). The flask was purged with N 2 , sonicated for 20 s and DIPEA (0.5 mmol) was added. The reaction was stirred at room temperature for 6 hrs then concentrated under reduced pressure. The residue was purified on a C I 8 column using 10-80% MeOH in water to yield the product as an off-white solid (40-90%).

Method N : General method for anhydride rinu opening by an intermediate exist inn as an HC1 salt.

R'-NH 2 HCI

To an intermediate collected as an HCI salt (0.1 1 mmol) were added the respective anhydride (0.13 mmol), and anhydrous DCM (6 mL). The flask was purged with N 2) sonicated for 20 s and triethylamine (0.44 mmol) was added. The reaction was stirred at room temperature for 5.5 hrs. The reaction was acidified to approximately pH 5 using citric acid (aqueous, saturated solution), and then concentrated under reduced pressure. The residue was purified on a C 18 column using 10-75% MeOH in water to yield the product as an off-white solid (40-67%).

Method O: Genera l ine hod for coupling ( 2H-Te ra/ol-5 -yl )mcthy lam i ne and a free carboxy lic acid.

To the carboxylic acid (0.18 mmol), were added the (2H-tetrazol-5- yl)methylamine (0.22 mmol), EDC (0.275 mmol), HOBt (0.22 mmol), and anhydrous DMF (15 ml). The flask was purged with N 2 , sonicated for 20 s and DIPEA (0.73 mmol) was added. The reaction was stirred at room temperature for 16 hrs. Analysis of the reaction by LC/MS showed approximately 75% conversion of the acid. An additional one half of the portion of the amine, EDC, HOBt, and DIPEA were added and the reaction was heated at 45 °C for another 6 hrs then concentrated under reduced pressure. The residue was purified on a C I 8 column using 10-70%» MeOH in water to yield the product as an off-white solid (40-95%). Method P; Genera! method for (a) preparation of un symmetric anhydride ing o ej linti prod ucts with prox imal substitution {via anhydride riiiti open in g with MeOH ) fo llowed by ( b) coup ling to an amine (ex. 1- 1 5 ) and c) subsequent hydrolysis.

R-NH 2 (ex. 1-15)

EDC, HOBt, DIPEA

DMF

To the respective anhydride (2.8 mmol) were added methanol (5.6 mmol), anhydrous DCM (28 mL) and triethylamine ( 1 1 mmol). The reaction was stirred at RT for 4 hrs then diluted with diethyl ether (30 mL) and HC1 (1 M, aqueous, 30 mL) and transferred to a separatory funnel. The organic layer was collected, washed with a NaCl solution (saturated, aqueous) and then dried over anhydrous Na2S0 4 . After filtration and concentration under reduced pressure, the crude ring-opened product was collected as a colourless oil and as a mixture of isomers. The ring-opened product mixture was used in the subsequent step without further purification. To the crude ring-opened product (0.1 1 mmol) were added the amine (ex. 1-15) (0.1 1 mmol) EDC (0.17 mmol), HOBt (0.13 mmol) and anhydrous DCM (6 mL). The flask was purged with N2, sonicated for 20 s and triethylamine (0.44 mmol) was added. The reaction was stirred at RT for 3 hrs. The reaction m ixture was concentrated under reduced pressure and the residue was purified on a C I 8 column using 10-70% MeOH in water to yield the methyl ester product as an off-white solid and as a mixture of isomers (49-65%). To the mixture of isomers (0. 12 mmol) were added LiOH H 2 0 (0.23 mmol), methanol (3 mL), and H 2 0 (4.5 mL). The reaction was stirred at room temperature in air for l Oh. The reaction was acidified to approximately pH 5 using citric acid (aqueous, saturated), and then concentrated under reduced pressure. The residue was purified on a CI 8 column using 10-75% MeOH in water to yield the product as an off-white solid ( 17-54%).

The following examples are provided for the purpose of illustrating, not limiting, the invention.

EXAMPLES

Synthetic Intermediates

The following is a description of synthetic intermediates (1-1 to 1-15) useful for making representative compounds of the invention.

Intermediate 1-1

1-1

tert-Butyl ( ^-Z-KZH-l^^^-tetrazol-S-ylmethy carbamoyll-l^-dihydro-lH- indole-l-carboxylate (1-1):

To (2S)-2,3-dihydro-lH-indole-2-carboxylic acid (1 g, 6.13 mmol) in dioxane (6 ml) and NaOH solution (0.5M, 12 ml) was added slowly di-/er/-butyl dicarbonate ( 1 .47 g, 6.74 mmol) in dioxane (6 ml) at 0 °C. The reaction mixture was warmed to RT and stirred overnight. The reaction mixture was then washed with hexanes ( 10 ml). The separated water layer was acidified using saturated solution of citric acid and extracted with ethyl acetate (3 x 15 ml). The organic layer was washed with brine, separated, dried over sodium sulfate and concentrated to give (2S)- \ -[(!ert- butoxy)carbonyl]-2,3-dihydro- lH-indole-2-carboxylic acid, as light orange colored solid ( 1 .45 g, 90 %), used further as described. ¾ NMR (400 MHz, DMSO-i 6) δ

I .41 (9H, s), 2.99-3.04 (I H, d, J=20 Hz), 3.48-3.55 ( I H, dd, J= 12, 16 Hz), 4.74-4.78 ( I H, dd, J= , 12 Hz), 6.91 -6.95 (IH, t, J=8 Hz), 7.14-7.18 (2H, m), 7.72-7.74 ( I H, d, J=8 Hz), 12.85 (I H, bs), MS (LC/MS) m/z observed 285.89, expected 286.1 1 [M+Na].

(2S)- l -[(ter -Butoxy)carbonyl]-2,3-dihydro- lH-indole-2-carboxyl ic acid (3 g,

I I .40 mmol), 2H-l ,2,3,4-tetrazol-5-ylmethanamine (1.16 g, 1 1.40 mmol), EDC (2.4 g, 12.53 mmol), HOBt ( 1.54 g, 1 1.40 mmol) and DIPEA ( 15.9 ml, 91.15 mmol) were stirred in anhydrous DMF (60 ml) for 1 6 hrs. The reaction mixture was concentrated under vacuum and re-dissolved in ethyl acetate (1 80 ml) and washed with citric acid (aqueous, saturated solution), water and brine. The organic layer was separated, dried over sodium sulfate and concentrated to give the crude product as an orange-red oil. The crude compound recrystallized in diethyl ether (80 ml) to yield ½r -butyl (25)-2-[(2H- l ,2,3,4-tetrazol-5-ylmethyl)carbamoyl]-2,3-dihydro-lH-indole- 1 -carboxylate (1-1) as an off-white solid (2.8 g, 71 %). Ή NMR (400 MHz, DMSO- d6) δ 1.32 (9H, s), 2.94-2.99 (I H, dd, J=A, 15 Hz), 3.41 -3.48 ( I H, dd, J=12, 20 Hz), 4.50-4.64 (2H, dq, J=4, 20 Hz), 4.73-4.77 (IH, dd, 7=4, 12 Hz), 6.89-6.83 (IH, t, J=% Hz ), 7.12-7.16 (2H, t, J=8 Hz), 7.71 (IH, bs), 7.86 ( I H, bs), MS (LC/MS) m/z observed 344.73, expected 345.17 [M+H].

(2S)-7V-(2H-l,2,3,4-Tetrazol-5-ylmethyl)-2,3-dihydro-lH-i ndole-2-carboxamide hydrochloride (1-2):

HCI Solution in dioxane (4M, 28 ml) was added to 1-1 (1.4 g, 4.07 mmol) and stirred for 2 hrs at RT. Precipitated solid was filtered under nitrogen and washed with diethyl ether (2 X 15 ml) to yield (2S)-N-(2H-l ,2,3,4-tetrazol-5-ylmethy])-2,3- dihydro-lH-indole-2-carboxamide hydrochloride(I-2) as a light pink colored solid (1 .1 g, 96%), Ή NMR (400 MHz, DMSO-</6) δ 2.93-2.99 (IH, dd, J=8, 16 Hz), 3.26-3.33 (IH, dd, J=12, 16 Hz), 4.24-4.29 (IH, t, J=8 Hz), 5.56-5.58 (2H, dd, J=\ .6,

8 Hz), 5.89 (I H, bs), 6.55-6.59 (2H, t, 7=8 Hz), 6.91-6.95 (IH, t, J=8 Hz), 6.99-7.01 ( 1 H, d, 7=8 Hz), 8.59-8.62 ( 1 H, t, 7=8 Hz), MS (LC/MS) m/z observed 244.97, expected 245.12[M+H].

Intermediate 1-3

dihydro-lH-indol-l-yl]ethyl}carbamate (1-3):

1-2 (0.5 g, 1.78 mmol), N-Boc-glycine (0.37g, 2.14 mmol), EDC (2.4 g, 12.5 mmol), HOBt (0.75 g, 3.92 mmol) and DIPEA (1 .24 ml, 7.125 mmol) were stirred in anhydrous DCM (20 ml) for 16 hrs. The reaction mixture was diluted using DCM ( 15 ml) and washed with citric acid (aqueous, saturated solution), followed by water and then brine. The organic layer was separated, dried over sodium sulfate and concentrated to give the crude product as a yellow-orange colored oil. The crude compound was purified on a CI 8 column using 10-50% MeOH in water to yield tert- butyl N-{2-oxo-2-[(2,S -2-[(277-l ,2,3,4-tetrazol-5-ylmethyl)carbamoyl]-2,3-dihydro- l /7-indol- l -yl]ethyl} carbamate (1-3) as an off-white solid (0.4 g, 56%), Ή NMR (400 MHz, DMSO-i 6) δ 1.39 (9H, s), 3.08-3.12 (2H, d, 7=16 Hz), 3.56-3.63( l H, dd, 7=12, 20 Hz), 3.92-3.97 (1H, dd, 7=4, 16 Hz), 4.56-4.62 (2H, m), 5.09-5.12 (1 H, d, 12 Hz), 6.96-7.00 (2H, t, 7=8 Hz), 7.13-7.17 ( 1 H, t, 7=8 Hz), 7.19-7.21 (1 H, d, 7=8 Hz), 8.00-8.02 (1 H, d, 7=8 Hz), 9.06 ( 1 H, s), MS (LC/MS) m/z observed 401 .82, expected 402.19 [M+H], and observed 424.04, expected 424.17 [M+Na]. Intermediate 1-4

1-3 , -4 (25 -l-(2-Aminoacetyl)-7V-(2H-l,2,3,4-tetrazol-5-ylmethyl)-2,3-d ihydro-lH- indole-2-carboxamide hydrochloride(I-4):

HC1 Solution in dioxane (4M, 5 ml) was added to 1-3 (0.24 g, 0.6 mmol) and stirred for 2 hrs at RT. The precipitated solid was filtered under nitrogen and washed with diethyl ether (2X5 ml) and dried to yield (2S)-l-(2-aminoacetyl)-N-(2H- 1,2,3,4- tetrazol-5-ylmethyl)-2,3-dihydro-lH-mdole-2-carboxamide hydrochloride (1-4) as an off-white solid (0.11 g, 54%), Ή NMR (400 MHz, DMSO-i6) δ 2.20-2.24 (2H, d, J=\6 Hz), 3.46-3.50(lH, d, J=16 Hz), 3.56-3.65 (IH, m), 4.05-4.09 (IH, d, 7=16 Hz), 5.09-5.12 (IH, d, 12 Hz), 4.60-4.61 (2H, d, J=4 Hz), 5.19-5.22 (IH, d, J=12 Hz), 7.04-7.08 (IH, t, J=8 Hz), 7.19-7.23 (IH, t, J=8 Hz), 7.25-7.27 (IH, d, J=8 Hz), 8.03- 8.05 (IH, d, J=8 Hz), 8.30 (2H, bs), 9.30 (2H, s), MS (LC/MS) m/z observed 301.98, expected 302.14 [M+H].

I iiLer mediate 1-5

tert-Butyl 7V-[(15',2^)-2-methyl-l-({2-oxo-2-[(2S)-2-[(2H-l,2,3,4-tetra zol-5- ylmethyl)carbamoyl]-2,3-dihydro-lH-indol-l- yl]ethyI}carbamoyl)butyl]carbamate (1-5):

1-4 (0.15 g, 0.44 mmol), N-Boc-Isoleucine (0.10 g, 0.44 mmol), EDC (0.13 g, 0.67 mmol), HOBt (0.078 g, 0.58 mmol) and D1PEA (0.31 ml, 1.78 mmol) were stirred in anhydrous DCM (15 ml) for 16 hrs. The reaction mixture was diluted using DCM (1 5 ml) and washed consecutively with citric acid (aqueous, saturated solution), water and brine. The organic layer was separated, dried over sodium sulfate and concentrated to give the crude product as a yellow-orange colored oil. The crude compound was purified on a C I 8 column using 10-50% MeOH in water to yield tert- butyl N-[(l S,2S)-2-methyl- l-({2-oxo-2-[(2S)-2-[(2H-l ,2,3,4-tetrazol-5- ylmethyl)carbamoyl]-2,3-dihydro- lH-indol- l -yl]ethyl}carbamoyl)butyl]carbamate (1-5) as an off-white solid (0.14 g, 60%). Ή NMR (400 MHz, DMSO-i/6) δ 0.80- 0.84 (3H, t, =8 Hz), 0.85-0.87 (3H, d, J=8 Hz), 1.39 (9H, s), 1.40- 1.46 ( 1 H, m), 1.69-1.75 (1 H, m), 3.02-3.12 (2H, m), 3.52-3.66 (3H, m), 3.89-3.93 (1H, t, J=8 Hz), 4.12-4. 16 ( 1 H, d, J= 16 Hz), 4.33-4.38 (1 H, dd, J=4, 16 Hz), 4.50-4.55 (1 H, dd, J=4, 16 Hz), 5.13-5.16 (1 H, d, J= 12 Hz), 6.73-6.75 (1H, d, J=8 Hz), 6.98-7.02 ( 1 H, t, J=8 Hz), 7.15-7.22 (2H, m), 8.04-8.06 (1H, d, J=8 Hz), 8.10 ) 1 H, s), 8.73 (1 H, s), MS (LC/MS) m/z observed 514.86, expected 515.27[M+H].

Intermediate 1-6

1-6

2-Phenylacetic acid (1-6) :

A solution of methyl 2-phenylacetate (10 g, 64 mmol) in methanol (60 ml) was treated with solution of sodium hydroxide (5.1 g, 127 mmol) in water (40 ml) at

70 °C for 3 hrs. The resulting mixture was concentrated under vacuum to remove the methanol. The residue was diluted with water (40 ml) and washed with diethyl ether (40 ml). The separated water layer was acidified to pH 2 using a mixture of water and HC1 (1 : 1 ) and extracted with DCM (3 X 80 ml). Combined organic extracts were washed with brine, 80 ml, separated, dried over sodium sulfate and concentrated to give 2-phenylacetic acid (1-6) as a white solid (9 g, 96%) used without further characterization. ] H NMR (400 MHz, CDC1 3 ) δ 3.64 (2H, s), 7.27-7.35 (5H, m), 1 1.5 (1 H, bs). Intermediate 1-7

(2-?,35)-3-Methyl-2-(2-phenylacetamido)pentanoic acid (1-7):

1-6 (2.0 g, 14.7 mmol) and thionyl chloride (6.6 ml, 90.3 mmol) were stirred together for 1 hr at RT. Thionyl chloride was removed by distillation under vacuum. The acid chloride was added to the stirring solution of L-isoleucine ( 1 .75 g, 13.37 mmol) in NaOH (2N, 17 ml) at 0 °C. The resulting mixture was warmed to RT and stirred overnight. The mixture was washed with diethyl ether (20 ml) and acidified to pH 4-5 by adding citric acid (aqueous, saturated solution). The precipitated solid was filtered, washed with diethyl ether and dried to yield (2S,3S)-3- methyl-2-(2-phenylacetamido)pentanoic acid (1-7) as a white solid ( 1.6 g, 44%). ! H NMR (400 MHz, DMSO-< 6) δ 0.78-0.82 (6H, t, J=8 Hz), 1.12-1.18 ( 1 H, m), 1.34- 1 .40 (1 H, m), 1.72-1.78 ( 1H, m), 3.41 -3.53 (2H, q, 7=16 Hz), 4.13-4.16 (1H, dd, J=4, 12 Hz), 7.15-7.19 ( 1H, m), 7.22-7.28 (1H, m), 8.19-8.21 ( 1 H, d, J=8 Hz), 12.54 ( 1 H, s), MS (LC MS) m/z observed 250.02, expected 250.14 [M+H].

Intermediate 1-8

2-[(2$,3S)-3-Methyl-2-(2-phenylacetamido)pentanamido]acetic acid (1-8):

To a suspension of 1-7 (24 g, 96.3 mmol) in DCM (400 mL) was added

DIPEA (87 mL, 500 mmol) at 0 °C. The reaction mixture was allowed to stir for 10 inin, upon which a homogeneous mixture was obtained. To this was added glycine ethyl ester hydrochloride (15.1 g, 108 mmol). After stirring a further 5 min at the same temperature, EDC (24.5 g, 1 16 mmol) and oxyma (1 8.8 g 1 16 mmol) were added. The reaction mixture was then allowed to stir overnight (approximately 15 hr) while allowed to slowly warm to RT. Another batch of glycine ethyl ester hydrochloride (2. 1 g) was added and the reaction mixture was allowed to stir a further 4 hr at ambient temperature. Reaction mixture was transferred to a separatory funnel and washed with NaHC0 3 (300 mL, aqueous, saturated), followed by HC1 (3M, 50 mL), and brine (200 mL). The organic layer was then dried over anhydrous sodium sulfate, filtered and concentrated in vacuo. Ethyl 2-[(2S,3S)-3-methyl-2-(2- phenylacetamido)pentanamido]acetate was obtained from flash column chromatography (MeOH: DCM 1 -5%) followed by recrystallization from acetonitrile ( 1 6 g, colourless crystals) and used further as described. Ή NMR (300 MHz, CD 3 OD) δ 7.32-7.21 (m, 5H), 4.34 (m, 1 H), 4.17 (q, 2H), 4.03-3.96 (m, 1H), 3.89- 3.81 (m, l H), 3.68-3.52 (m, 2H), 1.98- 1 .89 (m, 1 H), 1 .62-1 .35 (m, 1 H), 1 .26 (t, 3H), 1 .21 -1 .09 (m, 1 H), 0.96-0.86 (m, 6H), MS (ESI) m/z observed 357.29, expected

357.18 [M+Na] .

Intermediate 1-8 was obtained from hydrolysis of ethyl 2-[(2S,35)-3-methyl-2- (2-phenylacetamido)pentanamido]acetate (8.1 g, 24 mmol) using Method D (6 g, white powder). Ή NMR (300 MHz, CD 3 OD) δ 12.40 (br s, 1 H), 7.31 -7.1 6 (m, 5H), 4.23-4.1 8 (m, 1H), 3.76-3.70 (m, 2H), 3.57-3.40 (m, 2H), 1.83-1 .68 (m, 1H), 1.48-

1.35 (m, 1 H), 1.13-0.99 (m, 1 H), 0.83-0.75 (m, 6H), MS (ESI) m/z observed 305.20, expected 305.15 [M-H].

Intermediate 1-9

1-9

(2S)-l-{2-[(25 , ,3,S , )-3-Methyl-2-(2-phenylacetamido)pentanamido]acetyl}-2, 3- dihydro-lH-indole-2-carboxylic acid (1-9):

Intermediate 1-9 was obtained from 1-8 (170 mg, 0.37 mmol) using Method F followed by Method D (120 mg, white powder) and used further as is. Ή NMR (300 MHz, DMSO-i/6) δ 8.37-8.1 8 (m, I H), 8.06 (d, I H), 7.28-7.08 (m, 7H), 6.95- 6.90 (m, I H), 4.57-4.53 (m, I H), 4.44-4.24 (m, I H), 4.13-3.89 (m, 2H), 3.59-3.18 (m, 4H), 1 .92-1 .71 (m, I H), 1.47-1.22 (m, IH), 1.13-0.99 (m, I H), 0.84-0.75 (m, 6H).

Intermediate 1-10

1-10

Ethyl (25 -l-(2-{[(^r -butoxy)carbonyl]amino}acetyl)-2,3-dihydro-lH-indole-2- carboxylate (1-10) (2S)-2,3-dihydro-lH-indole-2-carboxylic acid (500 mg, 306 mmol) was suspended in EtOH (5 mL) at 0 °C and thionyl chloride (0.45 mL, 6.13 mmol, 2 eq.) was added. The resulting clear mixture was allowed to come to RT and stirred for 16 hours. The reaction mixture was then concentrated to dryness and swapped with EtOH (2 x 10 mL). The solid obtained was dried well under reduced pressure to give ethyl (2S)-2,3-dihydro-lH-indole-2-carboxylate hydrochloride as a light brown solid (0.58 g, quantitative). 1 !! NMR (400 MHz, DMSO-i d) 8 1.18 (3H, s), 3.10-3. 18 (1 H, m), 3.30-3.40 (1 H, m), 4.05-4.17 (2H, m), 4.55 ( 1 H, bs), 6.80 (2H, bs), 7.02-7.08 (2H, m), 7.7 (2H, bs). Compound was used further as described.

1-10 was prepared from ethyl (2S)-2,3-dihydro-lH-indole-2-carboxylate hydroch loride and 2-((terf-butoxycarbonyI)amino)acetic acid using method C however the purification was performed on normal phase using 0 % to 50 % ethyl acetate in hexanes as the eluent. Ή NMR (400 MHz, DMSO-i 6) 51.18 (3H, t, J=6Hz), 1 .38 (9H, s), 3.19 (1H, d, 16Hz), 3.48-3.62 (2H, m), 3.95-4.20 (3H, m), 5.35 (1 H, d, J=l l Hz), 7.00 (2H, t, J=8Hz), 7.15-7.25 (2H, m), 8.01 ( 1 H, d, J=SHz), MS (LC/MS) m/z 370.95 [M+Na]

Intermediate 1-11

Ethyl (25)-6-amino-2,3-dihydro-lH-indole-2-carboxylate (1-11)

(2S)-2,3-Dihydro- l H-indole-2-carboxyl ic acid (5 g, 30.640 mmol) was dissolved in su lfuric acid (concentrated, 40 ml) at 0 °C. The resulting reaction mixture was chilled to -20 °C and nitric acid (concentrated, 1 .4 ml, 33.1 mmol) was then added dropw ise. The reaction mixture was stirred at -20 °C for 1 hr and then allowed to warm to RT and stirred for 2 hrs. The reaction mixture was d iluted with cold water ( 100 ml), neutralized to pH 4 using NaOI I solution (10N) and extracted with EtOAc (3 X 200 ml). The combined organic layers were dried over sodium sulphate and concentrated to give an orange solid. This solid was dissolved in ethanol (200 niL) and treated with thionyl chloride (4.45 ml, 61 .3 mmol) at 0 °C and stirred overnight at RT. The reaction mixture was concentrated and purified by column chromatography on silica gel using 0 % to 50 % ethyl acetate in hexanes as the eluent to give ethyl (2S)-6-nitro-2,3-dihydro-lH-indole-2-carboxylate, as a yellow sol id ( 1 ,61 g, 22%). Ή NMR (400 MHz, CDC1 3 ) δ 1.30-1.34 (3H, t, J=8 Hz), 3.30-3.57 (2H, m), 4.21 -4.27 (2H, q, J=8 Hz), 4.47-4.52 ( I H, q, J=8 Hz), 4.70 ( I H, bs), 7.16-7.1 8 ( I H, d, J=8 Hz), 7.47.7.48 ( I H, d, J=4 Hz), 7.62-7.65 (I H, dd, .7=8, 4 Hz), MS (LC/MS) m observed 237.00, expected 237.08 [M+H].

Ethyl (2S)-6-nitro-2,3-dihydro- lH-indole-2-carboxylate (2 g, 8.474 mmol) and 10% Pd on activated carbon (0.4 g, 20% wt./wt.) in ethanol (60 ml) was stirred under hydrogen (balloon) for 3 hrs. The reaction mixture was filtered through a bed of CELITE ® and washed with ethanol (4 X 30 ml). Collected filtrate was concentrated and dried well under vacuum to yield 1-11 as pale yellow oil (1.4 g, 80%). 'Η NMR (400 MHz, CDCI 3 ) δ 1.14- 1.18 (3H, t, J=8 Hz), 2.94-3.01 (3H, m), 3.30 ( I H, s), 4.57-4.61 ( IH, t, J=8 Hz), 7.45-7.48 (IH, q, J=4 Hz), 8.03 (IH, s), 8.06-8.09 (IH, m), 8.41 -8.42 (IH, d, J=4 Hz), 8.96-8.97 (IH, d, J=4 Hz), MS (LC/MS) m/z observed 207.09, expected 207.1 1 [M+H].

(S)-Ethyl 6-((te/'i-butoxycarbonyl)amino)-l-(2-((2S',3 1 y)-3-methyl-2-(2- phenylacetamido)pentanamido)acetyl)indoline-2-carboxyIate (1-12)

1-11 (0.6 g, 2.91 mmol) and isoindoline- l ,3-dione (0.43 g, 2.91 mmol) in toluene was heated at 90 °C for 4 hrs. Resulting reaction mixture was concentrated and purified by column chromatography on silica gel using 0 % to 30 % ethyl acetate in hexanes as the eluent to give (S)-6-( l,3-dioxo- l ,3-dihydro-isoindolin-2-yl)-2,3- dihydro- l H-indole-2-carboxylic acid ethyl ester, as an off-white solid (0.7 g, 72%). Ή NMR (400 MHz, CDC1 3 ) δ 1.29-1.33 (3H, t, J=8 Hz), 3.38-3.44 (2H, m), 4.20- 4.26 (2H, m), 4.42-4.46 (IH, dd, J=12,8 Hz), 4.56 (IH, bs), 6.72 (I H, s), 6.75-6.78 ( IH, dd, .7=12,4 Hz), 7.17-7.19 (I H, d, .7=12 Hz), 7.76-7.78 (2H, m), 7.92-7.95 (2H, m), MS (LC/MS) m/z observed 337.07, expected 337.1 1 [M+H] ,

(S)- l -(2- er/-Butoxycarbonylamino-acetyl)-6-(l ,3-dioxo-l,3-dihydro-isoindol- 2-yl)-2,3-dihydro- lH-indole-2-carboxylic acid ethyl ester was prepared from (5)-6- (l ,3-dioxo-l ,3-dihydro-isoindolin-2-yl)-2,3-dihydro-lH-indole-2-carboxyl ic acid ethyl ester and Boc-glycine using method C and purified by column chromatography on silica gel using 0 % to 50 % ethyl acetate in hexanes as the eluent to give (S)- l -(2- feri-Butoxycarbonylamino-acetyl)-6-(l ,3-dioxo- l ,3-dihydro-isoindol-2-yl)-2,3- dihydro- lH-indole-2-carboxylic acid ethyl ester, as an off-white solid (42%). MS (LC/MS) m/z observed 493.87, expected 494.19[M+H] . Compound was confirmed using LC/MS and moved to next step as it was.

{S)- \ -(2-½rt-Butoxycarbonylamino-acetyl)-6-( l ,3-dioxo- l ,3-dihydro-isoindol- 2-yl)-2,3-dihydro- l H-indole-2-carboxylic acid ethyl ester and 1-7 were combined using method A to produce (S)-6-( l ,3-dioxo- l ,3-dihydro-isoindol-2-yl)- l -[2-(3- methyl-2-phenylacetylamino-pentanoylamino)-acetyl]-2,3-dihyd ro- lH-indole-2- carboxylic acid ethyl ester. MS (LC/MS) m/z observed 625.09, expected 625.26 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

(S)-6-(l,3-Dioxo- l ,3-dihydro-isoindol-2-yl)- l -[2-(3-methyl-2- phenylacetylamino-pentanoylamino)-acetyl]-2,3-dihydro- lH-indole-2-carboxylic acid ethyl ester (0.3 g, 0.48 mmol) was dissolved in 1 :4 mixture of DMF:dioxane ( 10 ml) and hydrazine monohydrate (0.026 ml, 0.53 mmol) was added. The reaction mixture was heated at 50 °C for 2 hr., concentrated and dried well under vacuum to give a yel low solid. This solid was dissolved in 1 :5 mixture of watendioxane (12 mL, v/v) and treated with DIPEA (0.67 ml, 3.85 mmol) and di-teri-butyl dicarbonate (0.42 g, 1 .92 mmol) and stirred overnight at RT. The reaction mixture was concentrated and purified by column chromatography on silica gel using 0 % to 50 % ethyl acetate in hexanes as the eluent to give 1-12, as a pale yellow solid (0.16 g, 56%). MS (LC/MS) m/z observed 595.09, expected 595.31 [M+H] used further as needed.

Intermediates 1-13 and 1-14

(5 -Ethyl 5-nitroindoline-2-carboxylate (1-12) and (5)-ethyl 7-nitroindoline-2- carboxylate (1-13)

(S)-2,3-Dihydro- lH-indole-2-carboxylic acid (20 g, 0.123 mol) was suspended in absolute ethanol (200 mL) and the suspension was cooled to 0 °C. SOCI2 (18 mL, 0.25 mol) was then added dropwise to the reaction mixture and it was allowed to warm to RT and was stirred for 16 hrs. The solvent was then evaporated and the residue was swapped twice with ethanol (100 mL) to get a brown solid. This solid was dissolved triethylamine (34.2 mL, 0.245 mol) and acetic anhydride (170 mL) and the mixture was stirred at RT for 2 hrs. Then it was diluted with EtOAc (500 mL) and washed with NaHC0 3 (2 x 200 mL, aqueous, saturated solution) and citric acid (2 x

200 mL, aqueous, saturated solution). The combined organic layers were dried over sodium sulphate and concentrated to give a brown solid. This solid was dissolved in acetic anhydride (200 mL) and a solution of nitric acid (1 1.1 mL) in acetic anhydride ( 1 61 mL) was added dropwise at 0 °C. The reaction mixture was left at 0 °C for 15 min then warm to RT and stirred for 3 hrs. Cold water (250 mL) was added to the reaction mixture and the product was extracted with EtOAC (3 x 450 mL). The combined organic layers were dried over sodium sulphate and concentrated. The two isomers obtained were separated by normal phase column chromatography using 10 % to 30 % ethyl acetate in hexanes as the eluent to give first, (S)- l -acetyl-5-nitro- 2,3-dihydro- lH-indole-2-carboxylic acid ethyl ester as a yellow solid ( 18.49 g, 54%) and second, (S)- l -acetyl-7-nitro-2,3-dihydro-lH-indole-2-carboxylic acid ethyl ester as an orange oil (8.21 g, 24 %).

(S)- l -Acetyl-5-nitro-2,3-dihydro-l H-indole-2-carboxylic acid ethyl ester: Ή NMR (400 MHz, CDC1 3 ) δ 1.30 (3H, t, J=7.0Hz), 2.22 (3H, bs), 3.34 ( I H, m), 3.65 ( IH, m), 4.19-4.34 (2H, m), 5.02 ( I H, bs), 8.03 (IH, s), 8.17 ( IH, dd, J=2, 9Hz), 8.34 (I H, bs), MS (LC/MS) m/z observed 279.01 , expected 279.10 [M+H].

(S)- l-Acetyl-7-nitro-2,3-dihydro-lH-indole-2-carboxylic acid ethyl ester: Ή NMR (400 MHz, CDC1 3 ) δ 1.28 (3H, t, =7.0Hz), 2.28 (3H, bs), 3.41 ( I H, d, J=16Hz), 3.62 ( I H, dd, 7=10, 17Hz), 4.19-4.30 (2H, m), 5.05 ( I H, dd, J=2, 10Hz), 7.16 (I H, t, J=8Hz), 7.42 ( I H, dd, J=2, 9Hz), 7.67 (I H, d, J=8Hz), MS (LC/MS) m/z observed 279.01 , expected 279.1 0 [M+H].

1-13: (S)- l -Acetyl-5-nitro-2,3-dihydro- lH-indole-2-carboxylic acid ethyl ester (1 8.49 g) was refluxed in a aqueous HCl solution ( I N, 500 mL) for 2 hrs until the compound dissolved completely. Then, the reaction mixture was concentrated to dryness and the residue was dissolved in ethanol (500 mL). SOCI2 (60 mL) was then added dropwise and the reaction was left at RT for 1 6 hrs. The solvent was then evaporated and the product was purified by normal phase column chromatography using 5 % to 20 % ethyl acetate in hexanes as the eluent to give intermediate 1-13 as a yellow solid ( 10.81 g, 69 %). Ή NMR (400 MHz, CDCI 3 ) δ 1.32 (3H, t, J=7.0Hz), 3.35-3.52 (2H, m), 4.22-4.30 (2H, q, J=7Hz), 4.57 (IH, dd, J=5, 10Hz), 5.01 ( I H, bs,), 6.63 (IH, d, J= 9Hz), 7.98 (IH, bs), 8.05 (I H, dd, J=2, 9Hz), MS (LC/MS) m/z observed 237.03, expected 237.09 [M+H].

1-14: (S)- l -Acetyl-7-nitro-2,3-dihydro- lH-indole-2-carboxylic acid ethyl ester ( 1 .6 g) was refluxed in a HCl solution (IN, aqueous, 80 mL) for 2 hrs until the compound dissolved completely. Then, the reaction mixture was concentrated to dryness and the residue was dissolved in ethanol (60 mL). SOCb (10 mL) was then added dropwise and the reaction was left at RT for 1 6 hrs. The solvent was then evaporated and the product was purified by normal phase column chromatography using 5 % to 20 % ethyl acetate in hexanes as the eiuent to give intermediate 1-14 as a yellow solid (0.97 g, 71 %). Ή NMR (400 MHz, CDC1 3 ) δ 1 .31 (3H, t, J=7.0Hz), 3.37-3.53 (2H, m), 4.19-4.30 (2H, q, J=7Hz), 4.69 (I H, dd, J=4, 10Hz), 6.66 (I H, dd, J=7, 8Hz), 7.03 ( I H, bs), 7.24 (I H, d, J= 7Hz), 7.83 ( I H, d, J=8Hz), MS (LC/MS) m/z observed 237.03, expected 237.09 [M+H].

Intermediate 1-15

(lS,2S)-2-Methyl-l-(2-oxo-2-{(2S)-2-[(2H-l,2,3,4-tetrazol-5- ylmethyl)- carbamoyl]-2,3-dihydro-indol-l-yl}-ethylcarbamoyl)-butyl-amm oniutn; chloride (1-15)

1-15 was collected as a faint pink solid from deprotection of 1-5 using method E. MS (LC/MS) m/z observed 415.09, expected 415.22 [M-Cl]. The compound was confirmed using LC/MS and moved to next step as it was.

Representative Granzyme B Inhibitor Compounds The following is a description of representative Granzyme B inhibitor compounds of the invention.

Examples A 1 -A57 were prepared by the representative synthetic pathway illustrated schematically in FIGURE 1. EXAMPLE A 1

(2S)-l-{2-[(25,3S)-3-METHYL-2-(2-PHENYLACETAMIDO)PENTANAMIDO ]ACETYL}-7V- (2H-l,2,3,4-TETRAZOL-5-YLMETHYL)-2,3-DIHYDRO-lH-INDOLE-2-CAR BOXAMIDE

Title compound Al was prepared from 1-5 and 1-6 using method A: Ή NMR (400 MHz, DMSCW6) δ 0.76-0.80 (3H, t, J=8 Hz), 0.82-0.84 (3H, d, J=8 Hz), 1.04-1.11 (IH, m), 1.39-1.45 (IH, m), 1.70-1.76 (IH, m), 3.10-3.14 (2H, d, J=16 Hz), 3.42-3.45 (IH, d, J=8 Hz), 3.53-3.56 (IH, d, J=8 Hz), 3.62-3.65 (IH, m), 3.66-3.69 (IH, m), 4.14- 4.16 (IH, t, J=4 Hz), 4.24-4.28 (IH, t, J=8 Hz), 4.47-4.51 (IH, d, J=16 Hz), 4.59-4.63 (IH, d, J=16 Hz), 5.14-5.16 (IH, d, .7=16 Hz), 6.97-7.01 (IH, t, J=8 Hz), 7.15-7.30 (7H, m), 8.08-8.11 (IH, d, J=12 Hz), 8.24(1H, bs), 8.99(1H, bs), MS (LC/MS) m/z observed 533.05, expected 533.26 [M+H] and observed 555.17, expected 555.24 [M+Na].

EXAMPLE A2

(2S)-l-{2-[(25',35)-3-METHYL-2-[2-(PYRiDiN-4-

YL)ACETAIVIIDO]PENTANAMIDO]ACETYL}-N-(2H-l,2,3,4-TETRAZOL -5-YLMETHYL)-2,3- DIHYDRO-1H-INDOLE-2-CARBOXAMIDE

Title compound A2 was prepared from 1-5 and 4-pyridineacetic acid using method A: l H NMR (400 MHz, DMSO-i6) δ 0.76-0.80 (3H, t, J=8 Hz), 0.82-0.84 (3H, d, J=8 Hz), 1.04-1.11 (IH, m), 1.38-1.46 (IH, m), 1.71-1.77 (IH, m), 3.07-3.11 (2H, t, J=8 Hz), 3.48-3.61 (2H, m), 4.11-4.15 (IH, d, J=16 Hz), 4.25-4.29 (IH, t, J=8 Hz), 4.39- 4.43 (IH, d, J=16 Hz), 4.52-56 (IH, d, .7=16 Hz), 5.12-5.15 (IH, d, J=12 Hz), 6.95-6.99 (IH, t, J=8 Hz), 7.13-7.22 (2H, m), 7.25-7.26 (2H, d, J=4 Hz), 7.25-7.28 (IH, d, J=8 Hz), 7.30 (IH, bs), 8.44-8.45(2H, d, J=4 Hz), 8.81(1H, bs), MS (LC/MS) m/z observed 534.11, expected 534.26 [M+H].

EXAMPLE A3

(2S)-1-{2-[(25',3 1 S)-3-METHYL-2-[2-(PYRIDIN-3-

YL)ACETAMIDO]PENTANAMIDO]ACETYL}-7V-(2H-l,2,3,4-TETRAZOL- 5-YLMETHYL)-2,3- DIHYDRO-1H-INDOLE-2-CARBOXAMIDE

Title compound A3 was prepared from 1-5 and 3-pyridineacetic acid using method A: Ή NMR (400 MHz, DMSO-<6) δ 0.76-0.80 (3H, t, 7=8 Hz), 0.82-0.84 (3H, d, J=8 Hz), 1.04-1.12 (IH, m), 1.37-1.46 (IH, m), 1.72-1.77 (IH, m), 3.06-3.12 (2H, m), 3.47-3.62 (2H, m), 4.12-4.16 (IH, d, J=16 Hz), 4.24-4.28 (IH, t, J=S Hz), 4.39-4.43 (IH, d, =16 Hz), 4.53-4.57 (1H, d, J=16 Hz), 5.13-5.15 (1H, d, J=8 Hz), 6.97-7.01 (1H, t, J=8 Hz), 7.14-7.18 (1H, t, J=8 Hz), 7.19-7.21 (1H, d, .7=8 Hz), 7.28-7.31 (1H, q, J=8 Hz), 7.65-7.67 (1H, d, J=8 Hz), 8.02-8.04(lH, d, 7=8 Hz), 8.23-8.26(lH, d, 7=12 Hz), 8.29 (1H, bs), 8.40-8.41 (1H, d, J=4 Hz), 8.44 (1H, s), 8.82 (1H, bs), MS (LC/MS) m/z observed 534.15, expected 534.26 [M+H].

EXAMPLE A4

(25 -l-{2-[(2S,35 -3-METHYL-2-[2-(PYRiDiN-2-

YL)ACETAMIDO]PENTANAMIDO]ACETYL}- J V-(2H-l,2,3,4-TETRAZOL-5-YLMETHYL)-2,3- DIHYDRO-1H-INDOLE-2-CARBOXAMIDE

Title compound A4 was prepared from 1-5 and 2-pyridineacetic acid using method A: Ή NMR (400 MHz, DMSO-rf6) δ 0.77-0.81 (3H, t, 7=8 Hz), 0.83-0.85 (3H, d,7=8 Hz), 1.06-1.13 (1H, m), 1.42-1.47 (1H, m), 1.74-1.81 (1H, m), 3.06-3.12 (2H, m), 3.53-3.62 (2H, m), 4.15-4.19 (1H, d, 7=16 Hz), 4.24-4.28 (1H, t, J=8 Hz), 4.25-4.29 (1H, q, 7=12 Hz), 4.44-4.47 (1H, d, 7=8 Hz), 4.59-4.63 (1H, d, J=16 Hz), 5.11-5.14 (1H, d, 7=12 Hz), 6.94-6.98 (1H, t, 7=8 Hz), 7.12-7.22 (3H, m), 7.68-7.72 (1H, td, 7=4, 8 Hz), 8.01-8.03 (1H, d, 7=8 Hz), 8.18-8.20(1H, d, 7=8 Hz), 8.34 (1H, bs), 8.43-8.45 (1H, d, .7=8 Hz), 8.87 (1H, bs), MS (LC/MS) m/z observed 534.12, expected 534.26 [M+H].

EXAMPLE A5

5-{[(lS,2S)-2-METHYL-l-({2-oxo-2-[(2.S -2-[(2H-l,2,3,4-TETRAZOL-5- YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l-

YL]ETHYL}CARBA OYL)BUTYL]CARBAMOYL}PENTANOIC ACID The starting compound (5-{[(lS,2S)-2-methyl-l-({2-oxo-2-[(2S)-2-[(2H-l,2,3,4- tetrazol-5-ylmethyl)carbamoyl]-2,3-dihydro-lH-indol-l- yl]ethyl}carbamoyl)butyl]carbamoyl}pentanoic acid, ethyl ester) was prepared from 1-5 and adipic acid, monoethyl ester using method A: MS (LC/MS) m/z observed 571.02, expected 571.29 [Μ+Η], Compound was confirmed using LCMS and moved to next step as it was.

Title compound A5 was prepared from 5-{[(lS,2S)-2-methyl-l-({2-oxo-2-[(2S)-2- [(2H-l,2,3,4-tetrazol-5-ylmethyl)carbamoyl]-2,3-dihydro-lH-i ndol-l- yl]ethyl}carbamoyl)butyl]carbamoyl}pentanoic acid, ethyl ester using method D: Ή NMR (400 MHz, DMSO-tf<5) δ 0.78-0.82 (3H, t, J=8 Hz), 0.84-0.86 (3H, d, J=8 Hz), 1.06-1.13 (IH, m), 1.41-1.50 (5H, m), 1.68-1.75 (IH, m), 2.08-2.20 (4H, m), 3.10-3.14 (IH, d, J=16 Hz), 3.50-3.62 (2H, m), 4.11-4.13 (IH, d, J=8 Hz), 4.22-4.26 (IH, m), 4.52- 4.56 (IH, d, J=16 Hz), 4.62-4.66 (IH, d, J=16 Hz), 5.14-5.17 (IH, d, J=U Hz), 6.97-7.01 (IH, t, J=S Hz), 7.13-7.17 (IH, t, J=8 Hz), 7.20-7.22 (IH, d, J=S Hz), 7.82-7.85 (IH, q, J=8 Hz), 8.00-8.02 (IH, d, J=8 Hz), 8.13 (IH, bs), 8.22-8.25 (IH, t, J=6 Hz) 9.09 (IH, bs), MS (LC/MS) m/z observed 542.99, expected 543.27 [M+H] and observed 565.09, expected 565.25 [M+Na]

EXAMPLE A6

(l^-l-i -^liS^-l-iS-AMI OPROPANAMIDO^-M rHYL ElNlAINAIVilDOlACETYLj-yV- (2H-l,2,3,4-TETRAZOL-5-YLMETHYL)-2,3-DIHYDRO-lH-INDOLE-2-CAR BOXAMIDE

(2S)-l-{2-[(2S,3S)-2-(3-(ier/-Butoxylcarbonyl)aminopropan amido)-3- methylpentanamido]acetyl}-N-(2H-l,2,3,4-tetrazol-5-ylrnethyl )-2,3-dihydro-lH-indole- 2-carboxamide was prepared from 1-5 and 3-((/ert-butoxycarbonyl)amino)propanoic acid using method A: ¾ NMR (400 MHz, DMSO-i6) δ 0.78-0.82 (3H, t, J=8 Hz), 0.84-0.86 (3H, d, J=8 Hz), 1.05-1.12 (IH, m), 1.35 (9H, s), 1.39-1.46 (IH, m), 1.70-1.75 (IH, m), 2.23-2.34 (2H, m), 3.06-3.12 (3H, m), 3.53-3.59 (2H, m), 4.10-4.14 (IH, d, J=16 Hz), 4.22-4.26 (IH, t, J=8 Hz), 4.37-4.42 (IH, dd, J=4, 16 Hz), 4.52-4.57 (IH, dd, 7=8, 16 Hz), 5.12-5.14 (IH, d, J=8 Hz), 6.96-7.00 (IH, t, J=8 Hz), 7.12-7.16 (IH, t, J=8 Hz), 7.18-7.20 (IH, d, J=8 Hz), 7.87-7.89 (IH, d, J=8 Hz), 8.01-8.03 (IH, d, J=8 Hz), 8.16 (IH, bs), 8.80 (IH, bs), MS (LC/MS) m/z observed 585.98, expected 586.31 [M+H] and observed 608.16, expected 608.29 [M+Na]. The compound was used further as described.

Title compound A6 was prepared from (2S)-l-{2-[(2S,35)-2-(3-(feri- butoxylcarbonyl)aminopropanamido)-3-methylpentanamido]acetyl }-N-(2H-l, 2,3,4- tetrazol-5-ylmethyl)-2,3-dihydro-lH-indole-2-carboxamide using method E: ! H NMR (400 MHz, DMSO-i/d) δ 0.80-0.84 (3H, t, J=8 Hz), 0.87-0.89 (3H, d, J=8 Hz), 1.07-1.15 (IH, m), 1.43-1.52 (IH, m), 1.72-1.78 (IH, m), 2.52-2.60 (2H, m), 2.97-3.09 (3H, m), 3.51-3.64 (2H, m), 4.09-4.12 (IH, d, J=12 Hz), 4.19-4.23 (IH, t, J=8 Hz), 4.33-4.41 (2H, m), 5.10-5.13 (IH, d, J=12 Hz), 6.96-7.00 (IH, t, J=8 Hz), 7.12-7.16 (IH, t, J=8 Hz), 7.18-7.20 (IH, d, J=8 Hz), 7.72 (2H, bs), 8.01-8.03 (IH, d, J=8 Hz), 8.22 (IH, s), 8.24 (IH, bs), 8.59 (IH, bs), MS (LC/MS) m/z observed 486.14, expected 486.26 [M+H]. EXAMPLE A7

(2S)-L-{2-[(25',35)-2-(4-AMINOBUTANAMIDO)-3-METHYLPENTANALVI IDO]ACETYL}-N- (2H-L,2,3,4-TETRAZOL-5-YLMETHYL)-2,3-DIHYDRO-LH-INDOLE-2-CAR BOXAMIDE

HYDROCHLORIDE

(2S)- l -{2-[(2S,3S)-2-(4-(/e^Butoxycarbonyl)am

methylpentanamido]acetyl}-N-(2H-l,2,3,4-tetrazol-5-ylmeth ^

2-carboxamide was prepared from 1-5 and 4-((ter/-butoxycarbonyl)amino)butanoic acid using method A: MS (LC/MS) m/z observed 599.97, expected 600.32 [Μ+Η], observed 622.15, expected 622.31 [M+Na]. Compound was confirmed using LC/MS and moved to next step as it was.

Title compound A7 was prepared from (25)-l-{2-[(2S,3S)-2-(4-(/eri- butoxycarbonyl)aminobutanamido)-3-methylpentanamido]acetyl}- N-(2H-l , 2,3,4- tetrazol-5-ylmethyl)-2,3-dihydro-lH-indole-2-carboxamide using method E: Ή NMR (400 MHz, DMSO-i 6) δ 0.80-0.84 (3H, t, 7=8 Hz), 0.88-0.90 (3H, d, J=8 Hz), 1.08-1.16 (IH, m), 1.47-1.53 (IH, m), 1.72- 1.84 (3H, m), 1.18-1.34 (2H, m), 2.75-2.79 (2H, t, J=6 Hz), 3.03-3.07 (IH, d, J=8 Hz), 3.51-3.64 (2H, m), 4.07-4. 1 1 (IH, m), 4.12-4.16 (IH, t, 7=12 Hz), 4.30-4.35 (I H, dd, J=6, 16 Hz), 4.41 -4.46 (IH, dd, J=6, 16 Hz), 5.08-5.1 1 (IH, d, J=12 Hz), 6.96-7.00 (IH, t, J=8 Hz), 7.12-7.16 (IH, t, J=8 Hz), 7.18-7.20 (I H, d, J=8 Hz), 7.92 (3H, bs), 8.00-8.02 (IH, d, J=8 Hz), 8.13-8.16 (IH, t, .7=6 Hz), 8.60 (I H, bs), MS (LC/MS) m/z observed 500.13, expected 500.27 [M+H],

EXAMPLE A 8

(25 l-{2-[(2S,3iS>2-[2-(4-AMINOPHENYL)ACETAMIDO]-3- METHYLPENTANAMIDO]ACETYL}-7V-(2H-l,2,3,4-TETRAZOL-5-YLMETHYL )-2,3- D1HYDRO-1H-INDOLE-2-CARBOXAMIDE HYDROCHLORIDE (25)- l -{2-[(2S,3S)-2-[2-(4-(ieri-Butoxycarbonyl)aminophenyl)acetam ido]-3- methylpentanamido]acetyl}-N-(2H-l ,2,3,4-tetrazol-5-ylmethyl)-2,3-dihydro- lH-indole- 2-carboxamide was prepared from 1-5 and 2-{<\-({tert- butoxycarbonyl)amino)phenyl)acetic acid using method A: MS (LC/MS) m/z observed 648.03, expected 648.31 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was. Title compound A8 was prepared from (2S)-l-{2-[(2S,3S)-2-[2-(4-(im- butoxycarbonyl)aminophenyl)acetamido]-3-methylpentanamido]ac etyl}-N-(2H-l, 2,3,4- tetrazol-5-ylmethyl)-2,3-dihydro-lH-indole-2-carboxamide using method E: Ή NMR (400 MHz, DMSO-</6) δ 0.75-0.79 (3H, t, .7=8 Hz), 0.81-0.83 (3H, d, 7=8 Hz), 1.01-1.06 (IH, m), 1.32-1.43 (IH, m), 1.62-1.74 (IH, m), 3.09-3.13 (2H, d, 7=16 Hz), 3.19-3.26 (2H, m), 3.55-3.62 (2H, m), 4.10-4.15 (IH, dd, 7=4, 16 Hz), 4.21-4.25 (IH, t, 7=8 Hz), 4.51-4.55 (IH, d, 7=16 Hz), 4.62-4.66 (IH, d, 7=16 Hz), 5.13-5.15 (IH, d, 7=8 Hz), 6.44- 6.46 (2H, d, 7=8 Hz), 6.88-6.90 (2H, d, 7=8 Hz), 6.96-7.00 (IH, t, 7=8 Hz), 7.14-7.18 (IH, t, 7=8 Hz), 7.19-7.21 (IH, d, 7=8 Hz), 7.79-7.81 (IH, d, 7=8 Hz), 8.01-8.03 (IH, d, 7=8 Hz), 8.20(1H, bs), 9.06(1H, bs), MS (LC/MS) m/z observed 548.06, expected 548.62 [M+H] and observed 570.17, expected 570.26 [M+Na].

EXAMPLE A9

(2S)- 1 - {2- [(2S,3S 2- [2-(3- AMINOPHENYL) ACETAMIDO] -3-

METHYLPENTANAMIDO]ACETYL}-/V-(2H-l,2,3,4-TETRAZOL-5-YLMET HYL)-2,3- DIHYDRO-1H-INDOLE-2-CARBOXAMIDE HYDROCHLORIDE

(2,S)-l-{2-[(2S,3S)-2-[2-(3-(iert-Butoxylcarbonyl)aminoph enyl)acetamido]-3- methylpentanamido]acetyl}-A ? -(2H-l,2,3,4-tetrazol-5-ylmethyl)-2,3-dihydro-l/J-indo le- 2-carboxamide was prepared from 1-5 and 2-(3-((tert- butoxycarbonyl)amino)phenyl)acetic acid using method A: MS (LC/MS) m/z observed 647.96, expected 648.33 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

Title compound A9 was prepared from (2S)-l-{2-[(2S,3S)-2-[2-(3-(/ert- butoxylcarbonyl)aminophenyl)acetamido]-3-methylpentanamido]a cetyl}-N-(2H-l, 2,3,4- tetrazol-5-ylmethyl)-2,3-dihydro-lH-indole-2-carboxamide using method E: Ή NMR (400 MHz, DMSO-i6) δ 0.76-0.80 (3H, t, 7=8 Hz), 0.82-0.84 (3H, d, 7=8 Hz), 1.03-1.11 (IH, m), 1.39-1.46 (IH, m), 1.68-1.74 (IH, m), 3.08-3.12 (2H, d, 7=16 Hz), 3.17-3.25 (2H, m), 3.54-3.61 (2H, m), 4.10-4.15 (IH, dd, 7=4, 16 Hz), 4.22-4.27 (IH, m), 4.42-4.48 (IH, m), 4.51-4.56 (IH, d, m), 5.12-5.14 (IH, d, 7=8 Hz), 6.36-6.40 (2H, t, 7=8 Hz), 6.43 (IH, s), 6.86-6.90 (IH, t, 7=8 Hz), 6.96-7.00 (IH, t, 7=8 Hz), 7.13-7.17 (IH, t, 7=8 Hz), 7.19-7.21 (IH, d, 7=8 Hz), 7.92-7.96 (IH, t, 7=8 Hz), 8.01-8.03(1H, d, 7=8 Hz), 8.23(1H, bs), 8.36(1 H, bs), 8.88(1 H, bs), MS (LC/MS) m/z observed, 548.03, expected 548.27 [M+H].

EXAMPLE A 10

(2S)-l-{2-[(2S,3,S -2-[2-(2-AMINO-l,3-TH]AZOL-4-YL)ACETAlVIIDO]-3- METHYLPENTANAMIDO]ACETYL}-yV-(2H-l,2,3,4-TETRAZOL-5-YLMETHYL )-2,3-

DiHYDRO-lH-iNDOLE-2-CARBOXAMiDE HYDROCHLORIDE

Thionyl chloride (0.460 mL, 6.321 mmol, 2 eq) was added to a stirred mixture of 2-(2-aminothiazol-4-yl)acetic acid (500 mg, 3.2 mmol) and ethanol (5 mL) at 0 °C. The resulting clear reaction mixture was slirred at RT fur 16 hr and then concentrated under vacuum to dryness and then swapped with ethanol twice. Ethyl 2-(2-aminothiazol-4- yl)acetate was obtained as a brown oil in a quantitative yield (588 mg). Ή NMR (400 MHz, DMSO-</6) δ 1.17 (3H, bs), 3.72 (2H, bs), 4.08 (2H, bs), 6,68 ( 1H, bs), 9.43 (2H, bs). The compound was used further as described.

Ethyl 2-(2-aminot iazol-4-yl)acetate (250 mg, 1.123 mmol) was treated with triethylamine (0.630 mL, 4.494 mmol, 4 eq.), N,N-dimethylaminopyridine (30 mg, 0.225 mmol, 0.2 eq.) and di-/eri-butyl dicarbonate (300 mg, 1.348 mmol, 1.2 eq.) in dichloromethane (10 mL) for 4 hr. The reaction mixture was diluted with dichloromethane (10 mL) and sequentially washed with water (1 x 10 mL), saturated citric acid solution (1 x 1 0 mL) and brine (1 x 10 mL). The organic layer was dried over sodium sulfate, filtered and concentrated to give a brown residue that was purified by column chromatography using a gradient hexanes/ethyl acetate (0% ethyl acetate to 50% ethyl acetate). Ethyl 2-(2-((½r/-butoxycarbonyl)amino)thiazol-4-yl)acetate was obtained as a yel low oil ( 130 mg, 34%). *H NMR (400 MHz, CDC1 3 ) δ 1.22 (3H, t, J=7Hz), 1 .55 (9H, s), 3.72 (2H, s), 4.1 1 (2H, q, J=7Hz), 6.75 ( 1 H, s), 9.55 ( 1 H, bs).

Ethyl 2-(2-((/ert-butoxycarbonyl)amino)thiazol-4-yl)acetate ( 100 mg,

0.350 mmol) was dissolved in ethanol (2 mL) and water (1 mL) and treated with l ithium hydroxide (44 mg, 1.05 mmol, 3 eq.) at RT for 2 hrs. The reaction mixture was acidified by adding a saturated solution of citric acid to pH 5 and then concentrated to remove all ethanol. The residue was then extracted with ethyl acetate (2 x 1 5 mL) and the combined organic layers were dried over sodium sulfate, filtered and concentrated to give 2-(2- ((?ert-butoxycarbonyl)amino)thiazol-4-yl)acetic acid as a yellow oil (80 mg, 89%). MS (LC/MS) m/z observed 258.74, expected 259.08 [M+H], Compound was confirmed using LC/MS and moved to next step as is.

(25)- 1 -{2-[(25,3S)-2-[2-(2-(ter/-Butoxycarbonyl)amino- 1 ,3-thiazol-4- yl)acetamido]-3-methylpentanamido]acetyI}-N-(2H-l,2,3,4-tetr azol-5-yImethyl)-2,3- dihydro-l//-indole-2-carboxamide was prepared from 2-(2-((tert- butoxycarbonyl)amino)thiazol-4-yl)acetic acid and 1-5 using method A: MS (LC/MS) m/z observed 654.97, expected 655.28 [M+H], The compound was confirmed using LC/MS and moved to next step as it was.

Title compound A10 was prepared from (2S)-\-{2-[(2S,3S)-2-[2-(2-(tert- butoxycarbonyl)amino-l,3-thiazol-4-yl)acetamido]-3-methylpen tanamido]acetyl}-N-(2H- l,2,3,4-tetrazol-5-ylmethyl)-2,3-dihydro-lH-indole-2-carboxa mide using method E: Ή NMR (400 MHz, DMSO-i/6) δ 0.77-0.92 (6H, m), 1.05-1.15 (IH, m), 1.40-1.52 (IH, m), 1.72-1.82 (IH, m), 3.00-3.65 (5H, m), 4.17 (IH, d, J=15Hz), 4.30 (IH, t, J=7Hz), 4.51- 4.72 (2H, m), 5.20 (IH, d, J=10Hz), 6.27 (IH, s), 6.85 (2H, m), 7.02 (IH, t, J=7Hz), 7.10-7.30 (2H, m), 7.88 (IH, d, J=9Hz), 8.10 (IH, d, J=8Hz), 8.25-8.35 (IH, m), 9.10 (IH, bs), MS (LC/MS) m/z observed 555.08, expected 555.23 [M+H].

EXAMPLE All

(25)-l-{2-[(2S)-2-CYCLOHEXYL-2-(2-PHENYLACETAMIDO)ACETAMIDO] ACETYL}-N- (2H-l,2,3,4-TETRAZOL-5-YLMETHYL)-2,3-DIHYDRO-lH-INDOLE-2-CAR BOXAIVlIDE /erf-Butyl ((S)-2-((2-((S)-2-(((2H-tetrazol-5-yl)methyl)carbamoyl)indol in-l-yl)-2- oxoethyl)amino)-l-cyclohexyl-2-oxoethyl)carbamate was prepared from 1-3 and S)-2- ((ieri-butoxycarbonyl)amino)-2-cyclohexylacetic acid using method A: MS (LC/MS) m/z observed 540.84, expected 541.29 [M+H], Compound was confirmed using LC/MS and moved to next step as it was.

Title compound All was prepared from teri-butyl ((S)-2-((2-((S)-2-(((2H- tetrazol-5-yl)methyl)carbamoyl)indolin-l-yl)-2-oxoethyl)amin o)-l-cyclohexyl-2- oxoethyl)carbamate and 1-6 using method A: 'Η NMR (400 MHz, DMSO-i/6) δ 0.93- 1.00- (2H, m), 1.05-1.14 (3H, m), 1.55-1.67 (6H, m), 3.05-3.11 (2H, q, = Hz), 3.52 (IH, s), 3.55-3.59 (2H, m), 4.11-4.15 (IH, d, =\6 Hz), 4.23-4.27 (IH, t, J=8 Hz), 4.37- 4.41 (IH, d, J=\6 Hz), 4.52-4.56 (IH, d, J=16 Hz), 5.12-5.14 (IH, d, .7=8 Hz), 6.96-7.00 (IH, t, J=8 Hz), 7.14-7.21 (3H, m), 7.24-7.29 (4H, m), 8.02-8.04 (IH, d, J=8 Hz), 8.06- 8.08 (IH, d, J=8 Hz), 8.27 (IH, bs), 8.78 (IH, bs), MS (LC/MS) m/z observed 559.01, expected 559.28 [M+H] and observed 581.13, expected 581.26 [M+Na].

EXAMPLE Al 2

(25)-l-{2-[(2S)-2-CYCLOPENTYL-2-(2-PHENYLACETAMIDO)ACETA IDO]ACETYL}-N- (2H-l,2,3,4-TETRAZOL-5-YL ETHYL)-2,3-DIHYDRO-lH-INDOLE-2-CARBOXAMIDE

/er-Butyl ((S)-2-((2-((S)-2-(((2H-tetrazol-5-yl)methyl)carbamoyl)indol in-l-yl)-2- oxoethyl)amino)-l-cyclopentyl-2-oxoethyl)carbamate was prepared from 1-3 and (<S)-2- ((fer/-butoxycarbonyl)amino)-2-cycIopentylacetic acid using method A: MS (LC/MS) m/z observed 526.81, expected 527.27 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

Title compound A12 was prepared from tert-butyl ((S)-2-((2-((S)-2-(((2H- tetrazol-5-yl)methyl)carbamoyl)indolin-l-yl)-2-oxoethyl)amin o)-l-cyclopentyl-2- oxoethyl)carbamate and 1-6 using method A: Ή NMR (400 MHz, DMSO-</6) 5 1.36- 1.44 (4H, m), 1.47-1.64 (4H, m), 2.12-2.18 (IH, m), 3.03-3.09 (2H, q, J=8 Hz), 3.39-3.43 (IH, d, J=16 Hz), 3.48-3.58 (2H, m), 4.09-4.17 (IH, t, J=16 Hz), 4.25-4.29 (IH, t, .7=8 Hz), 4.35-4.43 (IH, t, J=16 Hz), 4.47-4.55 (IH, t, J=16 Hz), 5.09-5.13 (IH, T, J=8 Hz), 6.96-7.00 (IH, t, J=8 Hz), 7.13-7.20 (3H, m), 7.23-7.28 (4H, m), 8.02-8.04 (IH, d, J=8 Hz), 8.18-8.21 (IH, d, J=12 Hz), 8.26 (IH, bs), 8.73 (IH, bs), MS (LC/MS) m/z observed 544.99, expected 545.26 [M+H].

EXAMPLE Al 3

3-{[(15 * ,2S)-2-METHYL-l-({2-OXO-2-[(2S)-2-[(2H-l,2,3,4-TETRAZO L-5- YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l- YL]ETHYL}CARBAM0YL)BUTYL]CARBAM0YL}PR0PAN0IC ACID Title compound A13 was prepared from 1-5 and succinic anhydride using method B: ! H NMR (400 MHz, DMSO-i/6) δ 0.79-0.83 (3H, t, J=8 Hz), 0.85-0.87 (3H, d, J=8 Hz), 1.10-1.16 (IH, m), 1.23-1.25 (4H, d, J=8 Hz), 1.43-1.52 (IH, m), 1.73-1.82 (IH, m), 3.05-3.09 (2H, d, J=16 Hz), 3.55-3.63 (2H, m), 4.13-4.22 (2H, m), 4.46-4.50 (2H, d, J=16 Hz), 5.11-5.13 (IH, d, J=8 Hz), 6.97-7.01 (IH, t, J=8 Hz), 7.15-7.19 (IH, d, J=16Hz), 7.94-7.96 (IH, d, J=8 Hz), 8.02 (IH, s), 8.15 (IH, bs), 8.86 (IH, bs), MS (LC/MS) m/z observed 515.07, expected 515.24 [M+H] and observed 537.11, expected 537.22 [M+Na]. EXAMPLE Al 4

4- { [(lS,2.y)-2-METHYL-l -( {2-OXO-2- [(2S)-2- [(2H-1 ,2,3,4-TETRAZOL-5-

YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l- YL] ETHYL}CARBAMOYL)BUTYL] CARBAMOYL} BUTANOIC ACID Title compound A14 was prepared from 1-5 and glutaric anhydride using method

B: Ή NMR (400 MHz, DMSO-d<5) δ 0.78-0.82 (3H, t, J=8 Hz), 0.84-0.86 (3H, d, 7=8 Hz), 1.06-1.13 (IH, m), 1.40-1.47 (IH, m), 1.66-1.75 (3H, m), 2.13-2.19 (4H, q, 7=8 Hz), 3.03-3.07 (IH, d, 7=16 Hz), 3.51-3.61 (2H, m), 4.09-4.11 (IH, d, 7=8 Hz), 4.21- 4.25 (IH, t, 7=8 Hz), 4.28-4.32 (IH, d, 7=12 Hz), 4.42-4.45 (IH, d, 7=12 Hz), 5.10-5.12 (IH, d, 7=8 Hz), 6.95-6.99 (IH, t, 7=8 Hz), 7.12-7.16 (IH, t, 7=8 Hz), 7.18-7.20 (IH, d, 7=8 Hz), 7.85-7.87 (IH, d, 7=8 Hz), 8.02-8.04 (IH, d, 7=8 Hz), 8.19 (IH, bs), 8.58 (IH, bs), MS (LC/MS) m/z observed 528.97, expected 529.24 [M+H] and observed 551.11, expected 551.23 [M+Na].

EXAMPLE Al 5

(2S)-l-{2-[(25 , ,3S)-2-ACETAMIDO-3-METHYLPENTANAMIDO]ACETYL}-N-(2H-l,2 ,3,4- TETRAZOL-5-YLMETHYL)-2,3-DIHYDRO-lH-INDOLE-2-CARBOXAMIDE

To 1-5 (0.12 mmol) was added HC1 (4M) solution in dioxane (4 ml) and the reaction mixture was stirred for 2 hr at RT, then concentrated to dryness under vacuum and swapped with MeOH (5 ml) three times. The resulting residue was dried well under vacuum and treated with a mixture of acetic anhydride/pyridinc (1:1, 1.5 mL) for 15 minutes at RT. Then the reaction mixture was concentrated and the residue was submitted to reverse phase C18 column using 10-50% MeOH in water to yield the title compound A15 as an off-white solid (5 mg): Ή NMR (400 MHz, DMSO-i/6) 50.77- 0.92 (6H, m), 1.05-1.15 (IH, m), 1.40-1.50 (IH, m), 1.68-1.77 (IH, m), 1.87 (3H, s), 3.08-3.15 (IH, m), 3.51-3.65 (2H, m), 4.10-4.18 (IH, m), 4.24 (IH, t, 7=9Hz), 4.48-4.68 (2H, m), 5.15 (IH, d, 7=10Hz), 6.95 (IH, t, 7=7Hz), 7.15-7.27 (2H, m), 7.90 (IH, d, 7=7Hz), 8.10 (IH, d, 7=8Hz), 8.18-8.33 (IH, m), 9.05 (IH, bs), MS (LC/MS) m/z observed 456.99, expected 457.23 [M+H]. EXAMPLE A16

3-{[(lS S)-2-METnYLA-({2-oxo-2-[(2S)-2-[(lH-l,2,3-TR\AZOL-4-

YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l- YL]ETHYL}CARBAM0YL)BUTYL]CARBAM0YL} PROPANOIC ACID (S)-/erf-Butyl 2-(((2H-l,2,3-triazol-4-yl)methyl)carbarnoyl)indoline-l-carb oxylate was prepared from (S)-l-(½ i-butoxycarbonyl)indoline-2-carboxylic acid, and (2/7-1,2,3- triazol-4-yl)methyl-amine following the same procedure for the preparation of 1-1. MS (LC/MS) m/z observed 365.99, expected 366.15 [M+Na]. Compound was confirmed using LC/MS and moved to next step as it was.

(S)-tert-Butyl (2-(2-(((2/7- 1 ,2,3-triazol-4-y l)methyl)carbamoyl)indol in- 1 -y l)-2- oxoethyl)carbamate was prepared from (S)-tert-buty\ 2-(((2H-l,2,3-triazol-4- yl)methyl)carbamoyl)indoline-l-carboxylate following the same procedure for the preparation of 1-2 followed by the same procedure for the preparation of 1-3. MS (LC/MS) m/z observed 400.83, expected 401.19 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

Title compound A16 was prepared from (S)-tert-buty\ (2-(2-(((2H-l,2,3-triazol-4- yl)methyl)carbamoyl)indolin-l-yl)-2-oxoethyl)carbamate and (2S,3S)-2-((tert- butoxycarbonyl)amino)-3-methylpentanoic acid using method A followed reaction with succinic anhydride using method B: ! H NMR (400 MHz, DMSO-tf6) δ 0.79-0.83 (3H, t, 7=8 Hz), 0.85-0.87 (3H,d, 7=8 Hz), 1.15-1.20 (IH, m), 1.40-1.50 (IH, m), 1.68-1.80 (IH, m), 2.31-2.45 (4H, m), 3.01-3.10 (IH, m), 3.50-3.63 (2H, m), 4.05-4.15 (IH, m), 4.25 (IH, t, 7= 7Hz), 4.30-4.45 (2H, m), 5.11 (IH, d, 7=9 Hz), 6.98 (IH, t, 7=8 Hz), 7.10-7.25 (2H, m), 7.90 (IH, d, 7=9 Hz), 8.02 (IH, d, 7=8 Hz), 8.13 (IH, bs), 8.86 (IH, bs), 12.02 (IH, bs), MS (LC/MS) m/z observed 514.02, expected 514.24 [M+H].

EXAMPLE Al 7

3-METHYL-4-{[(l 4 S , ,25)-2-METHYL-l-({2-OXO-2-[(25 -2-[(2H-l,2,3,4-TETRAZOL-5- YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l- YL]ETHYL}CARBAMOYL)BUTYL]CARBAMOYL}BUTANOIC ACID Title compound A17 was prepared from 1-5 and 3-methylglutaric anhydride using method I and was separated by chromatography, into two diastereomers, A17-1 and A17-2, each characterized as below: A17-1:

Ή NMR (400 MHz, DMSO-rfd) δ 0.82 (3H, t, J=7.4Hz), 0.84-0.90 (6H, m), 1.12 (IH, m), 1.45 (IH, m), 1.75 (IH, m), 2.05 (IH, m), 2.15-2.30 (4H, m), 3.10 (IH, m), 3.55-3.65 (2H, m), 4.15 (IH, dd, J=4, 18Hz), 4.28 (IH, t, J=15Hz), 4.55 (IH, dd, J=4, 16Hz), 4,65 (IH, dd, J=6, 16Hz), 5.16 (IH, d, J=llHz), 7.00 (IH, t, J=8Hz), 7.16 (IH, t, J=8Hz), 7.22 (IH, d, J=8Hz), 7.87 (IH, d, J=9Hz), 8.03 (IH, d, J=8Hz), 8.17 (IH, bs), 9.08 (IH, bs), MS (LC/MS) m/z observed 543.10, expected 543.27 [M+H].

A17-2

! H NMR (400 MHz, DMSO-i6) δ 0.82 (3H, t, J=7.4Hz), 0.84-0.90 (6H, m), 1.12 (IH, m), 1.45 (IH, m), 1.75 (IH, m), 2.05 (IH, m), 2.08-2.15 (2H, m), 2.20-2.30 (2H, m), 3.10 (IH, m), 3.55-3.65 (2H, m), 4.13 (IH, d, J=17Hz), 4.28 (IH, t, J=19Hz), 4.53 (IH, dd, J=4, 16Hz), 4,65 (IH, dd, J=6, 16Hz), 5.15 (IH, d, .7=1 lHz), 7.00 (IH, t, J=8Hz), 7.16 (IH, t, J=8Hz), 7.22 (IH, d, ,/=8Hz), 7.87 (IH, d, J=9Hz), 8.03 (IH, d,J=8¾), 8.17 (IH, bs), 9.08 (IH, bs), MS (LC/MS) m/z observed 543.07, expected 543.27 [M+H] .

EXAMPLE Al 8

3,3-DlMETHYL-4-{[(lS,2S)-2-METHYL-l-({2-OXO-2-[(2S)-2-[(2H-l ,2,3,4-TETRAZOL-5- YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l- YL]ETHYL}CARBAM0YL)BUTYL]CARBAM0YL}BU TAINOIC ACID Title compound A18 was prepared from 1-5 and 3,3-dimethylglutaric anhydride using method I without acidification with formic acid. The product was obtained as a triethylammonium salt: *H NMR (400 MHz, DMSO-</6) δ 0.82 (3H, t, =7.4Hz), 0.87 (3H, d, =7Hz), 1.02 (6H, s), 1.05-1.10 (10H, m), 1.45 (IH, m), 1.75 (IH, m), 2-22-2-29 (4H, m), 2.77-2.90 (6H, m), 3.05 (IH, d, J=17Hz), 3.55-3.65 (2H, m), 4.13 (IH, d, J=16Hz), 4.26-4.33 (2H, m), 4.53 (IH, m), 5.15 (IH, d, J=llHz), 6.98 (IH, t, J=8Hz), 7.12-7-24 (2H, m), 7.97 (IH, d, J=9Hz), 8.03 (IH, d, J=8Hz), 8.21 (IH, bs), 8.62 (IH, bs), MS (LCMS) m/z observed 557.14, expected 557.28 [M+H] EXAMPLE Al 9

2-[l-({[(lS,25)-2-METHYL-l-({2-oxo-2-[(25)-2-[(2H-l,2,3,4-TE TRAZOL-5- YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l- YL]ETHYL}CARBAM0YL)BUTYL] CARBAMOYL} METHYL)CYCLOPENTYL] ACETIC ACID Title compound A19 was prepared from 1-5 and 3,3-tetramethyleneglutaric anhydride using method I without acidification with formic acid. The product was obtained as a triethylammonium salt: Ή NMR (400 MHz, DMSO-</6) δ 0.82 (3H, t, J=7.4Hz), 0.87 (3H, d, J=7Hz), 1.07-1.13 (10H, m), 1.40-1.62 (9H, m), 1.75 (IH, m), 2-31-2-38 (3H, m), 2.45 (IH, d, J=15Hz), 2.86-2.93 (6H,m), 3.05 (IH, d, J-17Hz), 3.55- 3.65 (2H, m), 4.13 (IH, d, J=16Hz), 4.26-4.33 (2H, m), 4.49 (IH, dd, J=6, 15Hz), 5.15 (IH, d, J=\ lHz), 6.98 (IH, t, J=8Hz), 7.12-7-24 (2H, m), 7.97 (IH, d, J=9Hz), 8.03 (IH, d, ,/=8Hz), 8.23 (IH, bs), 8.65 (IH, bs), MS (LC/MS) m/z observed 583.11, expected 583.30 [M+H]

EXAMPLE A20

2-{[(15',25 , )-2-METHYL-l-({2-OXO-2-[(2S)-2-[(2H-l,2,3,4-TETRAZOL-5 - YLMETH YL) CARB A MO YL] -2 ,3-DIHYDRO- IH- INDOL- 1 - YL]ETHYL}CARBAMOYL)BUTYL]CARBAMOYL}-rft4Af5-CYCLOHEXANE-l-CA RBOXYLIC

ACID

Title compound A20 was prepared from 1-5 and /ram- 1,2- cyclohexanedicarboxylic anhydride using method I without acidification with formic acid and was separated by chromatography, into two diastereomers, A20-1 and A20-2, each characterized below as triethylammonium salts:

A20-1:

Ή NMR (400 MHz, OMSO-d6) δ 0.79 (3H, t, ./=7.4Hz), 0.85 (3H, d, ,/=7Hz), 1.06-1.26 (14H, m), 1.46 (IH, m), 1.65-1.77 (3H, m), 1.85 (IH, m), 1.95 (IH, m), 2.40- 2.45 (2H, m), 2.85-2.99 (3H, m), 3.05 (IH, d, ,/=17Hz), 3.32-3.47 (3H, m), 3.52-3.65 (2H, m), 4.08-4.24 (2H, m), 4.34 (IH, m), 4.47 (IH, m), 5.13 (IH, d, .7=11 Hz), 6.98 (IH, t, =8Hz), 7.12-7-24 (2H, m), 7.75 (IH, d, J=9Hz), 8.00-8-10 (2H, m), 8.65 (IH, bs), MS (LC/MS) m/z observed 569.11, expected 569.28 [M+H].

A20-2: Ή NMR (400 MHz, DMSO-i/<5) δ δ 0.80 (3H, t,

1.10-1-30 (14H, m), 1.50 (IH, m), 1.68-1.77 (3H, m), 1.91 (IH, m), 2.11 (IH, m), 2.43 (IH, m), 2.65 (IH, t, J=10.7Hz), 2.90-3.06 (4H, m), 3.32-3.50 (3H, m), 3.60 (IH, d, 7=11, 17Hz), 3.85 (IH, dd, J=5, 17Hz), 4.08 (IH, dd, J=5, 9Hz), 4.22 (IH, dd, J=6, 17Hz), 4.35 (IH, dd, J=6, 17Hz), 4.47 (IH, dd, J=6, 15Hz), 5.07 (IH, d, .7=1 IHz), 6.97 (IH, t, J=8Hz), 7.11-7-21 (2H, m), 7.95-8-06 (3H, m), 8.85 (IH, bs), MS (LC/MS) m/z observed 569.12, expected 569.28 [M+H] .

EXAMPLE A21

ό-ίΙίΙί',Ι^-Ι-ΜκτΗΥ^Ι-ίίΙ-ΟΧΟ-Ι-Κ ^- -Κ ^Ι,Ζ^^-ΤΕΤΚΑΖθί-δ- YLMETHYL)CARBAMOYL]-2,3-DIIIYDRO-lH-INDOL-l-

YL]ETHYL}CARBA OYL)BUTYL]CARBAMOYL}-C/IS-CYCLOHEX-3-ENE-1-CARBOXYLIC

ACID

Title compound A21 was prepared from 1-5 and cw-l,2,3,6-tetrahydrophtalic anhydride using method I and was separated by chromatography, into two diastereomers, A21-1 and A21-2, each characterized as below:

A21-1:

Ή NMR (400 MHz, DMSO-<½) δ 0.81 (3H, t, J=7.4Hz), 0.86 (3H, d, =7Hz),

1.10 (IH, m), 1.48 (IH, m), 1.75 (IH, m), 2.15-2.25 (2H, m), 2.33 (IH, m), 2.43 (IH, m), 2.82-2.95 (2H, m), 3.11 (IH, m), 3.54-3.66 (2H, m), 4.11 (IH, d, 17Hz), 4.23 (IH, t, J=8Hz), 4.54 (IH, dd, J=6, 17Hz), 4.65 (IH, dd, J=6, 15Hz), 5.16 (IH, d, J=l IHz), 5.60 (m, 2H), 7.00 (IH, t, J=8Hz), 7.11-7-26 (2H, m), 7.65 (IH, d, J=9Hz), 8.02 (IH, d, J=9Hz), 8.16 (IH, m), 9.07 (IH, bs), MS (LC/MS) m/z observed 567.11, expected 567.27 [M+H].

A21-2:

Ή NMR (400 MHz, DMSO-tftf) δ 0.81 (3H, t, y=7.4Hz), 0.86 (3H, d, J=7Uz),

1.11 (IH, m), 1.45 (IH, m), 1.81 (IH, m), 2.22 (IH, m), 2.30-2.36 (2H, m),2.46(IH, m), 2.82 (IH, m), 2.94 (IH, m), 3.11 (IH, d, ,/=17Hz), 3.54-3.66 (2H, m), 4.12-4.25 (2H, m), 4.50-4.65 (2H, m), 5.15 (IH, d, . l Hz), 5.62 (m, 2H), 7.00 (IH, t, J=8Hz), 7.11-7-26 (2H, m), 7.71 (IH, d, J=9Hz), 8.02 (IH, d, 7=9Hz), 8.16 (IH, bs), 9.05 (IH, bs), MS (LC/MS) m/z observed 567.11, expected 567.27 [M+H]. EXAMPLE A22

2-{[(15',25)-2-METHYL-l-({2-oxo-2-[(2 1 S)-2-[(2H-l,2,3,4-TETRAZOL-5-

YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l- YL]ETHYL}CARBAMOYL)BUTYL]CARBAMOYL}-C/5-CYCLOPENTANE-l-CARBO XYLIC

ACID

Title compound A22 was prepared from 1-5 and cs-l,2-cyclopentanedicarboxyIic anhydride using method I and was separated by chromatography, into two diastereomers, A22-1 and A22-2, each characterized as below:

A22-1:

Ή NMR (400 MHz, DMSO-i6) δ 0.81 (3H, t, J=7.4Hz), 0.86 (3H, d, J=7Hz),

1.11 (IH, m), 1.44-1.55 (2H, m), 1.66-1.85 (5H, m), 1.95 (IH, m), 2.82 (IH, m), 3.05 (IH, m), 3.11 (IH, m), 3.54-3.66 (2H, m), 4.10-4.23 (2H, m), 4.45-4.70 (2H, m), 5.16 (IH, d, J=llHz), 7.00 (IH, t, =8Hz), 7.14-7-26 (2H, m), 7.71 (IH, d, J=9Hz), 7.99-8.10 (2H, m), 9.08 (IH, bs), MS (LC/MS) m/z observed 555.10, expected 555.27 [M+H]. A22-2:

Ή NMR (400 MHz, DMSO-rf6) δ 0.81 (3H, t, J=7.4Hz), 0.85 (3H, d, J=7Hz), 1.14 (IH, m), 1.43 (IH, m), 1.52 (IH, m), 1.66-1.87 (5H, m), 1.96 (IH, m),2.82(lH, m), 3.08 (IH, m), 3.14 (IH, m), 3.56-3.69 (2H, m), 4.15-4.27 (2H, m), 4.52-4.68 (2H, m), 5.16 (IH, d, J=\ lHz), 7.00 (IH, t, J=8Hz), 7.14-7-26 (2H, m), 7.77 (IH, d, J=9Hz), 8.00- 8.08 (2H, m), 9.08 (IH, bs), MS (LCMS) m/z observed 555.09, expected 555.27 [M+H].

EXAMPLE A23

2-{[(15 , ,2^-2-METHYL-l-({2-OXO-2-[(2.S)-2-[(2H-l,2,3,4-TETRAZO L-5- YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l- YLlETHYLjCARBAMOY^BUTYLjCARBAMOYLj-CK-CYCLOHEXANE-l-CARBOXYL IC ACID Title compound A23 was prepared from 1-5 and cw-l,2-cyclohexanedicarboxylic anhydride using method 1 and was separated by chromatography, into two diastereomers, A23-1 and A23-2, each characterized as below:

A23-1:

Ή NMR (400 MHz, DMSO-<½) δ 0.79 (3H, t, J=7.4Hz), 0.83 (3H, d, 7=7Hz),

1.08 (IH, m), 1.20-1.32 (2H, m), 1.34-1.57 (4H, m), 1.63 (IH, m), 1.73 (IH, m), 1.87 (IH, m), 1.99 (IH, m), 2.55 (IH, m), 2.77 (IH, m), 3.10 (IH, m), 3.54-3.65 (2H, m), 4.10-4.23 (2H, m), 4.48-4.67 (2H, m), 5.14 (IH, d, 7=1 lHz), 6.99 (IH, t, J=8Hz), 7.12-7- 24 (2H, m), 7.56 (IH, d, =9Hz), 7.97-8.08 (2H, m), 9.07 (IH, bs), MS (LC/MS) m/z observed 569.08, expected 569.28 [M+H].

A23-2:

Ή NMR (400 MHz, DMSO-itf) δ 0.78 (3H, t, J=7.4Hz), 0.84 (3H, d, =7Hz), 1.11 (IH, m), 1.20-1.48 (4H, m), 1.50-1.69 (3H, m), 1.77 (IH, m), 1.90 (IH, m), 2.03 (IH, m), 2.53 (IH, m), 2.82 (IH, m), 3.10 (IH, m), 3.54-3.65 (2H, m), 4.12-4.23 (2H, m), 4.46-4.64 (2H, m), 5.13 (IH, d, J=llHz), 6.97 (IH, t, 7=8Hz), 7.12-7-24 (2H, m), 7.62 (IH, d, J=9Hz), 8.00 (IH, d, J=8Hz), 8.11 (IH, bs), 9.03 (IH, bs), MS (LC/MS) m/z observed 569.06, expected 569.28 [M+H] .

EXAMPLE A24

(2Z)-3-{[(15,25)-2-METHYL-l-({2-OXO-2-[(25')-2-[(2H-l,2,3,4- TETRAZOL-5- YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-LH-INDOL-L- YL]ETHYL}CARBAMOYL)BUTYL]CARBAMOYL}PROP-2-ENOIC ACID

Title compound A24 was prepared from 1-5 and maleic anhydride using method I without acidification with formic acid: Ή NMR (400 MHz, DMSCW6) δ 0.82 (3H, t, J=7.4Hz), 0.88 (3H, d, J=7Hz), 1.08 (IH, m), 1.12 (9H, t, J=7Hz), 1.47 (IH, m), 1.82 (IH, m), 2.96 (6H, q, 7=7Hz), 3.05 (IH, d, J=17Hz), 3.50-3.65 (2H, m), 4.13 (IH, d, J=16Hz), 4.26 (IH, t, J=7Hz), 4.38 (IH, m), 4.50 (IH, dd, J=6, 15Hz), 5.13 (IH, d, J=IlHz), 6.11-6.15 (2H, m), 6.98 (IH, t, J=8Hz), 7.12-7-24 (2H, m), 8.04 (IH, d, J=8Hz), 8.37 (IH, bs), 8.85 (IH, bs), MS (LC/MS) m/z observed 513.08, expected 513.22 [M+H]

EXAMPLE A25

2-{[(l 1 S,2 4 y)-2-METHYL-l-({2-OXO-2-[(25)-2-[(2H-l,2,3,4-TETRAZOL- 5- YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l-

YL]ETHYL}CARBAMOYL)BUTYL]CARBAMOYL}BENZOIC ACID Title compound A25 was prepared from 1-7 and phthalic anhydride using method I without acidification with formic acid: Ή NMR (400 MHz, DMSO-i5) δ 0.82 (3H, t, 7=7.4Hz), 0.93 (3H, d, J=7Hz), 1.10 (9H, t, J=7Hz), 1.25 (IH, m), 1.55 (1H, m), 1.97 (IH, m), 2.90 (6H, q, ,/=7Hz), 3.00 (IH, d, J=17Hz), 3.56 (IH, m), 3.73 (IH, m), 4.16 (IH, dd, 7=6, 16Hz), 4.25 (IH, dd, 7=6, 8Hz), 4.47-4.51 (2H, m), 5.18 (IH, d, 7=llHz), 6.98 (IH, t, 7=8Hz), 7.12-7-20 (2H, m), 7.28 (IH, d, J=8Hz), 7.42-7-50 (2H, m), 7.88 (IH, d, 7=9Hz), 8.03 (IH, d, J=8Hz), 8.62 (IH, d, 7=8Hz), 8.71 (IH, bs), 9.21 (IH, bs), MS (LC/MS) m/z observed 563.10, expected 563.24 [M+H]

EXAMPLE A26

2-{[(lS,25 -2-METHYL-l-({2-OXO-2-[(25')-2-[(2H-l,2,3,4-TETRAZOL-5- YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l- YL]ETHYL}CARBAMOYL)BUTYL]CARBA OYL}-C/£-CYCLOPROPANE-1-CARBOXYLIC

ACID

Title compound A26 was prepared from 1-5 and cw-3-oxabicyclo[3.1.0]hexane-

2,4-dione using method I and was separated by chromatography, into two diastereomers, A26-1 and A26-2, each characterized as below:

A26-1:

'H NMR (400 MHz, DMSO-i6) δ 0.81 (3H, t, 7=7.4Hz), 0.87 (3H, d, 7=7Hz), 1.04-1.14 (2H, m), 1.31 (IH, m), 1.47 (IH, m), 1.75 (IH, m), 1.92 (IH, m), 2.13 (IH, m), 3.11 (IH, m), 3.55-3.66 (2H, m), 4.15 (IH, dd, 7=6, 16Hz), 4.25 (IH, dd, 7=7, 9Hz), 4.54 (IH, m), 4.65 (IH, m), 5.17 (IH, d, 7=1 lHz), 7.01 (IH, t, 7=8Hz), 7.14-7-26 (2H, m), 8.03 (IH, d, 7=8Hz), 8.18-8.30 (2H, m), 9.10 (IH, bs), MS (LC/MS) m/z observed 527.07, expected 527.24 [M+H].

A26-2:

Ή NMR (400 MHz, DMSO-tf<5) δ δ 0.81 (3H, t, 7=7.4Hz), 0.87 (3H, d, 7=7Hz), 1.06-1.17 (2H, m), 1.31 (IH, m), 1.47 (IH, m), 1.76 (IH, tn), 1.91 (IH, m), 2.13 (IH, m), 3.11 (IH, m), 3.55-3.66 (2H, m), 4.16 (IH, dd, 7=6, 16Hz), 4.29 (IH, dd, 7=7, 9Hz), 4.54 (IH, m), 4.65 (IH, m), 5.17 (IH, d, 7=1 lHz), 7.01 (IH, t, 7=8Hz), 7.14-7-26 (2H, m), 8.03 (IH, d, 7=8Hz), 8.18-8.30 (2H, m), 9.10 (IH, bs), MS (LC/MS) m/z observed 527.06, expected 527.24 [M+H]. EXAMPLE A27

(l^-l-l -ti ^^-l-^-il-BENZOTHIOPHE -S-Y^ACETAMIDOl-S- ETHYLPENTANAiyHDO]ACETYL}-7V-(2H-l,2,3,4-TETRAZOL-5-YL!VlETH YL)-2,3- DIHYDRO-1H-INDOLE-2-CARBOXAMIDE

Title compound A27 was prepared from 1-5 and 2-(benzo[£>]thiophen-3-yl)acetic acid using method A except a 2: 1 ratio DCM/DMF as solvent, was used for the coupling reaction: ] H NMR (400 MHz, DMSO-</6) δ 0.80 (3H, t, J=7.4Hz), 0.86 (3H, d, J=7Hz), 1.10 (IH, m), 1.47 (IH, m), 1.76 (IH, m), 3.11 (IH, m), 3.55-3.66 (2H, m), 3.73 (I H, d, ,;=5Hz), 3.83 (IH, d, J=5Hz), 4.15 (IH, dd, J=6, 16Hz), 4.30 (I H, dd, J=7, 9Hz), 4.54 (IH, m), 4.65 ( IH, m), 5.17 (I H, d, .7=1 lHz), 7.01 (IH, t, J=8Hz), 7.14-7-26 (2H, m), 7.32-7.40 (2H, m), 7.52 (I H, s), 7.86 (IH, m), 7.95 (I H, m), 8.04 (IH, d, J=8Hz), 8.23- 8.32 (2H, m), 9.08 (I H, bs), MS (LC/MS) m/z observed 589.05, expected 589.23 [M+H]

EXAMPLE A28

(25")-l-{2-[(2S,35 -3-METHYL-2-[2-(2H-l,2,3,4-TETRAZOL-5- YL)ACETA IDO]PENTANAIVIIDO]ACETYL}-N-(2H-l,2,3,4-TETRAZOL-5-YLMETHYL) -2,3-

DIHYDRO-1H-INDOLE-2-CARBOXAMIDE

Title compound A28 was prepared from 1-5 and 2-(2H-tetrazol-5-yl)acetic acid using method A but with DMF as solvent for the coupling reaction: Ή NMR (400 MHz, DMSO-< 6) δ 0.82 (3H, t, J=7.4Hz), 0.88 (3H, d, J=7Hz), 1.12 (IH, m), 1.50 (IH, m), 1.77 (IH, m), 3.12 (IH, m), 3.56-3.67 (2H, m), 3.95-4.05 (2H, m), 4.16 (I H, dd, J=6, 16Hz), 4.31 (IH, t, J=8Hz), 4.56 (IH, m), 4.65 (IH, m), 5.17 (IH, d, J=l lHz), 7.01 (IH, t, J=8Hz), 7.14-7-26 (2H, m), 8.04 (IH, d, J=8Hz), 8.34 (IH, bs), 8.43 (IH, d, J=9Hz), 9.08 (IH, bs), MS (LC/MS) m/z observed 525.08, expected 525.24 [M+H]

EXAMPLE A29

METHYL (3fi)-3-METHYL-3-{[(lS,25 -2- ETHYL-l-({2-oxo-2-[(2S)-2-[(2H-l,2,3,4-

TETRAZOL-5-YL ETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l- YL]ETHYL}CARBAMOYL)BUTYL]CARBAMOYL}PROPANOATE

Title compound A29 was prepared from 1-5 and (R)-4-methoxy-2-methyl-4- oxobutanoic acid using method E, followed by method C: Ή NMR (400 MHz, DMSO- d6) δ 0.83 (3H, t, J=7.4Hz), 0.89 (3H, d, J=7Hz), 1.05 (3H, d, J= 7Hz), 1.14 (I H, m), 1.47 (I H, m), 1 .77 (I H, m), 2.33 (I H, dd, J=6, 16Hz), 2.57 (I H, dd, J=8, 16Hz), 2.87 (I H, m), 3.12 (I H, m), 3.56 (3H, s), 3.58-3.67 (2H, m), 4.16 ( I H, m), 4.25 (IH, t, ,/=8Hz), 4.56 (I H, m), 4.66 (I H, m), 5.17 ( I H, d, .7=1 l Hz), 7.01 (I H, t, J=8Hz), 7.14-7- 26 (2H, m), 7.91 (I H, d, J=9Hz), 8.02-8.12 (2H, m), 9.10 (IH, bs), MS (LC/MS) m/z observed 543.13, expected 543.27 [M+H]

EXAMPLE A30

3-METHYL-3-{[(lS,2.?)-2-IVIETHYL-l-({2-OXO-2-[(2^-2-[(2H-l,2 ,3,4-TETRAZOL-5-

YLMETHYL)CARBAMO YL] -2,3-DIHYDRO- LH-INDOL-1 - YL] ETHYL} CARBAMOYL)BTJTYL] CARBAMOYL} PROPANOIC ACID Title compound A30 was prepared from A29 using method D with 3 eq. υΓ LiOH l¾0 and was separated by chromatography, into two diastereomers, A30-1 and A30-2, each characterized as below:

A30-1:

Ή NMR (400 MHz, DMSO-</<i) δ 0.79 (3H, t, J=7.4Hz), 0.85 (3H, d, J=7Hz), 1.01 (3H, d, 7= 7Hz), 1.10 (IH, m), 1.45 (IH, m), 1 .74 (I H, m), 2.17 (IH, dd, J=6, 16Hz), 2.43 (IH, m), 2.79 ( IH, m), 3.08 (I H, m), 3.54-3.64 (2H, m), 4.12 (IH, m), 4.22 (I H, t, J=8Hz), 4.50 (IH, m), 4.62 ( IH, m), 5.14 (IH, d, J=l lHz), 6.98 (IH, t, J=8Hz), 7.1 1 -7-23 (2H, m), 7.82 (IH, d, J=9Hz), 7.98-8.08 (2H, m), 9.01 (IH, bs), MS (LC/MS) m/z observed 529.08, expected 529.25 [M+H].

A310-2:

Ή NMR (400 MHz, DMSO-ito) 0.79 (3H, t, J=7.4Hz), 0.85 (3H, d, J=7Hz),

1.03 (3H, d, J= 7Hz), 1.10 (IH, m), 1 .44 ( IH, m), 1.72 ( I H, m), 2.22 (IH, dd, J=l, 15Hz), 2.51 ( I H, m), 2.68 ( IH, m), 3.08 (IH, m), 3.54-3.64 (2H, m), 4.1 1 (IH, m), 4.22 (I H, t, ,/=8Hz), 4.48 (I H, m), 4.60 (IH, m), 5.14 (IH, d, J=l l Hz), 6.98 (IH, t, ,/=8Hz), 7.1 1 -7-23 (2H, m), 7.82 (I H, d, 7=9Hz), 8.02 (IH, m), 8.15 (I H, bs), 8.96 ( I H, bs), MS (LC/MS) m/z observed 529.10, expected 529.25 [M+H]

EXAMPLE A31

2-[3-({[(15 , ,2^-2-METHYL-l-({2-OXO-2-[(25 , )-2-[(2H-l,2,3,4-TETRAZOL-5- YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l-

YL]ETHYL}CARBAMOYL)BUTYL]CARBAMOYL}METHYL)PHENYL] ACETIC ACID Title compound A31 was prepared from 1-5 and l -cyclohexene-l ,2-dicarboxylic anhydride using method I without acidification with formic acid: Ή NMR (400 MHz, DMSO-i6) δ 0.78 (3H, t, J=7.4Hz), 0.84 (3H, d, J=7Hz), 1.08 (IH, m), 1.13 (9H, t, ,/=7Hz), 1.17-1.25 (2H, m), 1.43-1.63 (3H, m), 1.99 (IH, m), 2.17-2.25 (3H, m), 2.28 (IH, m), 2.97-3.05 (7H, m), 3.54-3.65 (2H, m), 4.07-4.18 (2H, m), 4.38-4.50 (2H, m), 5.12 (IH, d, J=l lHz), 6.95 (IH, t, J=8Hz), 7.12-7-20 (2H, m), 7.97 (IH, d, J=8Hz), 8.07 (IH, d, J=8Hz), 8.45 (IH, bs), 9.05 (IH, bs), MS (LC/MS) m/z observed 566.96, expected 567.27 [M+H]

EXAMPLE A32

(2Λ,3Λ)-2,3-ΟΙΗΥΟΚΟΧΥ-3-{[(15,25)-2- ΕΤΗΥ^1-({2-ΟΧΟ-2-[(2Λ)-2-[(2^1,2,3,4-

TETRAZOL-5-YLMETHYL)CARBAMOYL]-2,3-DlHYDRO-lH-INDOL-l- YL]ETHYL}CARBAM0YL)BUTYL]CARBAM0YL}PR0PAN0IC ACID

Title compound A32 was prepared from 1-5 and L-f+,)-tartaric acid using method J in DMF: Ή NMR (400 MHz, DMSO-<6) δ 0.83 (3H, t, J=7.4Hz), 0.87 (3H, d, J=7Hz), 1.08 (IH, m), 1.48 (IH, m), 1.80 (IH, m), 3.11 (IH, m), 3.56-3.68 (2H, m), 4.17 (IH, m), 4.26 (IH, s), 4.30-4.37 (2H, m), 4.55 (IH, m), 4.66 (IH, m), 5.16 (IH, d, J=llHz), 7.00 (IH, t, J=8Hz), 7.15-7-24 (2H, m), 7.57 (IH, d, J=9Hz), 8.07 (IH, d, J=8Hz), 8.33 (IH, bs), 9.10 (IH, bs), MS (LC/MS) m/z observed 547.06, expected 547.23 [M+H]

EXAMPLE A33

(25',35)-2,3-DiHYDROXY-3-{[(15',25)-2-METHYL-l-({2-OXO-2-[(2 1 y)-2-[(2/7-l,2,3,4- TETRAZOL-5-YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l- YL]ETHYL}CARBAM0YL)BUTYL]CARBAM0YL}PR0PAN0IC ACID

Title compound A33 was prepared from 1-5 and D-f- -tartaric acid using method J in DMF: Ή NMR (400 MHz, DMSO-i6) δ 0.83 (3H, t, J=7.4Hz), 0.87 (3H, d, J=7Hz), 1.06 (IH, m), 1.51 (IH, m), 1.73 (IH, m), 3.11 (IH, m), 3.55-3.67 (2H, m), 4.17 (IH, m), 4.22 (IH, s), 4.31 (IH, s), 4.37 (IH, dd, =7, 9Hz), 4.55 (IH, m), 4.63 (IH, m), 5.16 (IH, d, .7=11 Hz), 7.00 (IH, t, J=8Hz), 7.15-7-24 (2H, m), 7.44 (IH, d, ,/=9Hz), 8.02 (IH, d, J=8Hz), 8.47 (IH, bs), 9.10 (IH, bs), MS (LC/MS) m/z observed 546.92, expected 547.23 [M+H] EXAMPLE A34

2-[4-({[(lS,25 -2-METHYL-l-({2-oxo-2-[(2,S)-2-[(2H-l,2,3,4-TETRAZOL-5-

YLMETHYL)CARBA OYL]-2,3-DIHYDRO-lH-INDOL-l- YL] ETHYL} CARBAM0YL)BUTYL]CARBAM0YL}METHYL)PHENYL] ACETIC ACID Title compound A34 was prepared from 1-7 and 1,4-phenylenediacetic acid using method J: Ή NMR (400 MHz, DMSO-i6) δ 0.80 (3H, t, J=7.4Hz), 0.86 (3H, d, J=7Hz), 1.10 (IH, m), 1.44 (IH, m), 1.75 (IH, m), 3.11 (IH, m), 3.43 (2H, d, J=14Hz), 3.53 (2H, d, J=14Hz), 3.56-3.65 (2H, m), 4.15 (IH, m), 4.27 (IH, t, J=8Hz), 4.54 (IH, m), 4.65 (IH, m), 5.17 (IH, d, .7=1 IHz), 7.01 (IH, t, J=8Hz), 7.14-7-24 (6H, m), 8.04 (IH, d, ,/=8Hz), 8.10 (IH, d, J=9Hz), 8.25 (IH, bs), 9.08 (IH, bs), MS (LC/MS) m/z observed 591.15, expected 591.27 [M+H]

EXAMPLE A35

2-[3-({[(15',2S)-2-METHYL-l-({2-oxo-2-[(2S)-2-[(2H-l,2,3,4-T ETRAZOL-5-

YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l- YL]ETHYL}CARBAM0YL)BUTYL]CARBAM0YL}METHYL)PHENYL]ACETIC ACID

Title compound A35 was prepared from 1-7 and 1,3-phenylenediacetic acid using method J: Ή NMR (400 MHz, DMSO-</6) δ 0.80 (3H, t, J=7.4Hz), 0.86 (3H, d, J=7Hz), 1.10 (IH, m), 1.44 (IH, m), 1.75 (IH, m), 3.11 (IH, m), 3.43 (2H, d,J=14Hz), 3.55 (2H, d, J=14Hz), 3.57-3.66 (2H, m), 4.15 (IH, m), 4.27 (IH, t, J=8Hz), 4.55 (IH, m), 4.65 (IH, m), 5.17 (IH, d, =llHz), 7.01 (IH, t, ,/=8Hz), 7.08-7-24 (6H, m), 8.04 (IH, d, J=8Hz), 8.12 (IH, d, J=9Hz), 8.25 (IH, bs), 9.08 (IH, bs), MS (LC/MS) m/z observed 590.98, expected 591.27 [M+H]

EXAMPLE A36

(2£ -3-{[(15',25 , )-2-METHYL-l-({2-OXO-2-[(25 , )-2-[(2H-l,2,3,4-TETRAZOL-5- YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l-

YL]ETHYL}CARBAM0YL)BUTYL] CARBAMOYL} PROP-2-ENOIC ACID

Title compound A36 was prepared from 1-5 and fumaric acid using method J: Ή NMR (400 MHz, DMSO-ita) δ 0.83 (3H, t, J=7.4Hz), 0.90 (3H, d, J=7Hz), 1.13 (IH, m), 1.46 (IH, m), 1.78 (IH, m), 3.11 (IH, m), 3.57-3.66 (2H, m), 4.15 (IH, m), 4.37 (IH, t, J=8Hz), 4.55 (IH, m), 4.65 (IH, m), 5.17 (IH, d, J=llHz), 6.52 (IH, d, ,/=15Hz), 7.01 (1H, t, J=8Hz), 7.12-7-24 (3H, m), 8.04 (1H, d, J=8Hz), 8.35 (1H, bs), 8.60 (1H, d, J=9Hz), 9.08 (1H, bs), MS (LC/MS) m/z observed 513.46, expected 513.22 [M+H]

EXAMPLE A37

(3.S)-3-AMINO-3-{[(lS,2S)-2-lVIETHYL-l-({2-OXO-2-[(2S)-2-[(2 H-l,2,3,4-TETRAZOL-5- YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l-

YL] ETHYL} CARBAM0YL)BUTYL] CARBAMOYL} PROPANOIC ACID The intermediate compound ((S)-terf-Butyl 4-(((2S,35)-l-((2-(2-(((2H-tetrazol-5- yl)methyl)carbamoyl)indolin- l-yl)-2-oxoethyl)amino)-3 -methyl- 1 -oxopentan-2- yl)amino)-3-(S)-((te /-butoxycarbonyl)amino)-4-oxobutanoate) was prepared from 1-5 and Boc-l-aspartic acid-p-tert-butyl ester using method A. MS (LC/MS) m/z observed 685.99, expected 686.36 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

(S)-ier/-Butyl 4-(((2S,3S)-l-((2-(2-(((2H-tetrazol-5-yl)methyl)carbamoyl)in dolin- l-yl)-2-oxoethyl)amino)-3-methyl-l-oxopentan-2-yl)amino)-3-( .?)-((/eri- butoxycarbonyl)amino)-4-oxobutanoate (30mg) was dissolved in a 1:1 mixture TFA/DCM (4 mL) and left under stirring at RT for 2 h. The solvents were evaporated and the product was purified on a C18 column using 10-30% MeOH in water to give title compound A37 as an off-white solid: Ή NMR (400 MHz, DMSO-rftf) δ 0.83 (3H, t, J=7.4Hz), 0.90 (3H, d, J=7Hz), 1.15 (1H, m), 1.55 (1H, m), 1.82 (1H, m), 2.68-2.84 (2H, m), 3.17 (1H, d, J=16Hz), 3.57-3.67 (2H, m), 4.07-4.18 (311, m), 4.44 (1H, m), 4.56 (1H, m), 5.08 (1H, d, J=l 1Hz), 6.99 (1H, t, J=8Hz), 7.12-7-24 (2H, m), 8.04 (1H, d, J=8Hz), 8.32 (1H, bs), 8.45 (1H, m), 9.08 (1H, bs), MS (LC/MS) m/z observed 530.17, expected 530.25 [M+H]

EXAMPLE A38

(3R)-3-AMiNO-3-{[(15 , ,2S)-2-iViETHYL-l-({2-OXO-2-[(2S)-2-[(2H-l,2,3,4-TETRA ZOL-5- YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l- YL]ETHYL}CARBAMOYL)BUTYL]CARBAMOYL}PROPANOIC ACID (S)-/er/-Butyl 4-(((2S,3S)-l-((2-(2-(((2H-tetrazol-5-yl)methyl)carbamoyl)in dolin- 1 -yl)-2-oxoethyl)amino)-3-methyl- 1 -oxopentan-2-yl)amino)-3-(R)-((/er/- butoxycarbonyl)amino)-4-oxobutanoate was prepared from 1-7 and Boc-D-aspartic acid- β-ferZ-butyl ester dicyclohexylammonium salt using method A. MS (LC/MS) m/z observed 686.07, expected 686.36 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

(SJ-fert-Butyl 4-(((2S,3S)-l-((2-(2-(((2H-tetrazoI-5-yl)methyl)carbamoy!)in dolin- 1 -y l)-2-oxoethy l)am ino)-3-methy 1- 1 -oxopentan-2-yl)am mo)-3-(R)-((tert- butoxycarbonyl)amino)-4-oxobutanoate (30 mg) was dissolved in a 1:1 mixture tnfluoroacetic acid/DCM (4 mL) and left under stirring at RT for 2 h. The solvents were evaporated and the product was purified on a CI 8 column using 10-30% MeOH in water to give title compound A38 as an off-white solid: 'H NMR (400 MHz, DMSO-i6) δ 0.83 (3H, t, J=7.4Hz), 0.90 (3H, d, J=7Hz), 1.13 (1H, m), 1.46 (1H, m), 1.78 (1H, m), 2.55-2.72 (2H, m), 3.08 (1H, d, J=16Hz), 3.52-3.65 (2H, m), 4.05 (1H, m), 4.18 (1H, m), 4.27 (1H, m), 4.44-4.49 (2H, m), 5.14 (1H, d, J=llHz), 7.00 (1H, t, J=8Hz), 7.12-7-24 (2H, m), 8.03 (1H, d, =8Hz), 8.35 (1H, bs), 8.52 (1H, m), 8.87 (1H, bs), MS (LC/MS) m/z observed 530.16, expected 530.25 [M+H]

EXAMPLE A39

(2 1 S)-2-AMlNO-3-{[(/S,2S)-2-METHYL-l-({2-OXO-2-l(2S)-2-[( 2H-l,2,3,4-TETRAZOL-5- YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l- YL]ETHYL}CARBAMOYL)BUTYL]CARBAMOYL}PROPANOIC ACID (S)-tert-Butyl 4-((l-((2-(2-(S)-(((2H-tetrazol-5-yl)methyl)carbamoyl)indoli n-l- yl)-2-oxoethyl)amino)-3-(S)-methyl-l-oxopentan-2-yl)amino)-2 -(S)-((/eri- butoxycarbonyl)amino)-4-oxobutanoate was prepared from 1-5 and Boc-Z-aspartic acid- -tert-)\x\y\ ester using method A. MS (LC/MS) m/z observed 686.09, expected 686.36 [Μ+Η]. Compound was confirmed using LC/MS and moved to next step as it was.

(S)-iert-Butyl 4-((l-((2-(2-(S)-(((2H-tetrazol-5-yl)methyl)carbamoyl)indoli n-l- yl)-2-oxoethyl)amino)-3-(S)-methyl-l-oxopentan-2-yl)amino)-2 -(5)-((iert- butoxycarbonyl)amino)-4-oxobutanoate (30mg) was dissolved in a 1:1 mixture tnfluoroacetic acid/DCM (4 mL) and left under stirring at RT for 2 h. The solvents were evaporated and the product was purified on a CI 8 column using 10-30%) MeOH in water to give title compound A39 as an off-white solid: H NMR (400 MHz, DMSO-i/6) δ 0.83 (3H, t, J=7.4Hz), 0.88 (3H, d, J=7Hz), 1.20 (1H, m), 1.51 (1H, m), 1.86 (1H, m), 2.37 (1H, d, J=15Hz), 2.88 (1H, dd, J=10, 14Hz), 3.01 (1H, d, ,/=16Hz), 3.52-3.63 (2H, m), 3.86 (1H, m), 4.10-4.20 (2H, m), 4.49 (1H, m), 4.61 (1H, m), 5.11 (1H, d, J=llHz), 6.98 (I H, t, J=8Hz), 7.12-7-23 (2H, m), 8.03 (IH, d, J=8Hz), 8.35 (IH, d, J=8Hz), 8.52 (I H, m), 9.30 ( IH, bs), MS (LC/MS) m/z observed 530.10, expected 530.25 [M+H]

EXAMPLE A40

(l^- -AlVIINO -itCi^ ^^-lVIETHYL-l-iJ -OXO- -Iil^- -IilH-l^^^-TETRAZOL-S- YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l-

YL]ETHYL}CARBAMOYL)BUTYL]CARBAMOYL}PROPANOIC ACID

(S)-tert-Butyl 4-((l -((2-(2-(5)-(((2H-tetrazol-5-yl)methyl)carbamoyl)indolin-l- yl)-2-oxoethyl)amino)-3-(S)-methyl-l-oxopentan-2-yl)amino)-2 -(R)-((/err- butoxycarbonyl)amino)-4-oxobutanoate was prepared from 1-5 and Boc-D-aspartic acid- a-ferf-butyl ester using method A. MS (LC/MS) m/z observed 686.05, expected 686.36 [Μ+Η]. Compound was confirmed using LC/MS and moved to next step as it was.

(SHerf-Butyl 4-((l -((2-(2-(S)-(((2H-tetrazol-5-yl)methyl)carbamoyl)indolin-l- yl)-2-oxoethyl)amino)-3-(S)-methyl-l -oxopentan-2-yl)amino)-2-(R)-((feri- butoxycarbonyl)amino)-4-oxobutanoate (30mg) was dissolved in a 1 : 1 mixture trifluoroacetic acid/DCM (4 iiiL) and left under stirring at RT for 2 h. The solvents were evaporated and the product was purified on a CI 8 column using 10-30% MeOH in water to give title compound A40 as an off-white solid: Ή NMR (400 MHz, DMSO-rf6) δ 0.82 (3H, t, J=7.4Hz), 0.86 (3H, d, J=7Hz), 1.20 (IH, m), 1.50 (IH, m), 1.81 (IH, m), 2.50 (IH, m), 2.97-3.11 (2H, m), 3.55 (IH, m), 3.70 (IH, m), 3.82 (IH, m), 4.03 (IH, t, J= 8Hz), 4.18 (IH, dd, J=7, 16Hz), 4.49 (IH, dd, J=5, 6Hz), 4.66 (IH, dd, J=6, 17Hz), 5.08 (IH, d, J=l lHz), 6.98 (I H, t, ./=8Hz), 7.12-7-23 (2H, m), 8.03 (I H, d, y=8Hz), 8.35 (I H, m), 8.48 ( I H, d, J=8Hz), 9.34 (IH, bs), MS (LC/MS) m/z observed 530.13, expected 530.25 [M+H]

EXAMPLE A41

(2-?)-1-{2-[3-FLUORO-3-METHYL-2-(2-PHENYLACETAMIDO)BUTANAMID O]ACETYL}-/V- (2H-L,2,3,4-TETRAZOL-5-YLMETHYL)-2,3-DIHYDRO-LH-TNDOLE-2-CAR BOXAMIDE

3-Fluoro-3-methyl-2-(2-phenylacetamido)butanoic acid was prepared from 1-6 and 3-fluoro-DL-valine using method H: Ή NMR (400 MHz, DMSO-i 6) δ 1.33 (3H, d, J=219Hz), 1.39 (3H, d, J=19Hz), 3.52 ( I H, d, J=14Hz), 3.59 (IH, d, J=14Hz), 4.41 , 4.46 (I H, 2 x d, ,/=19Hz), 7.20 (IH, m), 7.22-7.29 (3H, m), 8.50 (IH, d, J=9Hz), 12.90 (IH, bs), MS (LC/MS) m/z observed 254.02, expected 254.12 [M+H]. Compound was used further as described.

Title compound A41 was prepared from 1-4 and 3-fluoro-3-mefhy]-2-(2- phenyIacetamido)butanoic acid using method C (as a mixture of diastereomers): MS (LC/MS) m/z observed 537.09, expected 537.24 [M+H].

EXAMPLE A42

(2S)-1 - {2-[(2»y,3R)-3-METHYL-2-(2-PHENYLACETAMIDO)PENTANAMIDO] ACETYL}-7V- (2H- 1 ,2,3,4-TETRAZOL-5-YLMETHYL)-2,3-DIHYDRO- 1H-INDOLE-2-CARBOXAMIDE

(2S,3R)-3-Methyl-2-(2-phenylacetamido)pentanoic acid was prepared from 1-6 and a//o-isoleucine using method H: ! H NMR (400 MHz, DMSO-i/6) δ 0.75-0.90 (6H, m), 1.10 (I H, m), 1.25 ( IH, m), 1 .85 (IH, m), 3.47 (I H, d, J=14Hz), 3.56 (I H, d, J=14Hz), 4.35 (IH, m), 7.20 (I H, m), 7.24-7.30 (3H, m), 8.15 (I H, d, J=9Hz), 12.57 (I H, bs), MS (LC/MS) m/z observed 250.04, expected 250.14 [M+H]. Compound was used further as described.

Title compound A42 was prepared from 1-3 and (2S,3R)-3-methyl-2-(2- phenylacetamido)pentanoic acid using method A: ! H NMR (400 MHz, MeOH-iW) δ 0.87-0.97 (6H, m), 1 .18 (I H, m), 1.45 (I H, m), 1.99 (I H, m), 3.21 (IH, d, 7=8Hz), 3.23 (I H, d, J=8Hz), 3.55-3.67 (2H, m), 3.69-3.76 (2H, m), 4.51 (I H, d, J=5Hz), 4.58-4.74 (2H, m), 5.16 ( I H, d, =l l Hz), 7.05 ( I H, t, 7=8Hz), 7.15-7-35 (7H, m), 8. 13 ( I H, bs), MS (LC/MS) m/z observed 533. 12, expected 533.26 [M+H] .

EXAMPLE A43

2,2,3,3-TETRAFLUORO-3-{[(lS,2S)-2-METHYL-l-({2-OXO-2-[(25')- 2-[(2H-l,2,3,4- TETRAZOL-5-YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-lNDOL-l- YL] ETHYL}CARBAMOYL)BUTYL]CARBA OYL}PROPANOIC ACID Title compound A43 was prepared from 1-5 and 2,2,3,3-tetrafluorosuccinic acid using method J in DMF: ] H NMR (400 MHz, DMSO-i 6) δ 0.80-0.84 (3H, t, J=& Hz), 0.88-0.90 (3H, d, J=8 Hz), 1.08- 1.15 (IH, m), 1.45- 1.5 1 (I H, m), 1.83-1.91 ( IH, m), 2.92-3.12 (2H, m), 3.56-3.64 (2H, m), 3.13-3.17 ( IH, d, J=16 Hz), 4.27-4.31 (I H, t, J=8 Hz), 4.42-4.46 ( I H, d, 7=8 Hz), 4.58-4.62 (IH, d, J=16 Hz), 5.13-5.15 (IH, d, J=8 Hz), 6.98-7.02 (IH, t, J=8 Hz), 7.15-7.22 (2H, m), 8.03-8.05 (I H, d, J=8 Hz), 8.39(1 H, s), 8.97(1H, bs), 9.90 ( IH, bs) 19 F NMR (376 MHz, DMSO-< 6) δ -1 14.28 , -115.00 , -115.62, -116.35, -116.56, -117.28 , -118.35, -119.06., MS (LC/MS) m/z observed 587.01, expected 587.19 [M+H].

EXAMPLE A44

2-[METHYL({[(lS , ,2-S)-2-METHYL-l-({2-OXO-2-[(2 1 S , )-2-[(2H-l,2,3,4-TETRAZOL- 5-YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l-

YL]ETHYL}CARBAMOYL)BUTYL]CARBAMOYL}METHYL)AMINO]ACETIC ACID

Title compound A44 was prepared from 1-5 and 4-methylmorpholine-2,6-dione using method I: Ή NMR (400 MHz, DMSO-</6) δ 0.82-0.86 (3H, t, J=8 Hz), 0.88-0.90 (3H, d, J=8 Hz), 1.04-1.11 (IH, m), 1.47-1.53 (IH, m), 1.77-1.84 (IH, m), 2.73 (lH,s), 2.89 (IH, s), 2.98-3.04 (4H, q, .7=8 Hz), 3.53-3.67 (2H, m), 4.13-4.17 (IH, d, J=\6 Hz), 4.30-4.34 (IH, t, 7=8 Hz), 4.36-4.40 (IH, dd, J=12, 4 Hz), 4.48-4.52 (IH, dd, J=12, 4 Hz), 5.10-5.13 (IH, d, J=12 Hz), 6.97-7.01 (IH, t, 7=8 Hz), 7.14-7.18 (IH, t, J=8 Hz), 7.20-7.22 (IH, d, J=8 Hz), 7.95 (IH, s), 7.98-8.04 (IH, m), 8.42(1H, bs), 8.79(1H, bs), MS (LC/MS) m/z observed 544.17, expected 544.26 [M+H].

EXAMPLE A45

3-{[(5)-CYCLOPENTYL({2-OXO-2-[(2S)-2-l(2H-l,2,3,4-TETRAZOL-5 - YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l- YL]ETHYL}CARBA OYL)METHYL]CARBAMOYL}PROPANOIC ACID terf-Butyl ((S)-2-((2-((S)-2-(((2H-tetrazol-5-yl)methyl)carbamoyl)indol in-l-yl)-2- oxoethyl)amino)-l-cyclopentyl-2-oxoethyl)carbamate was prepared from 1-3 and (S)-2- ((fcr/-butoxycarbonyl)amino)-2-cyclopentylacetic acid using general method A. MS (LC/MS) m/z observed 527.03, expected 527.27 [M+H], Compound was confirmed using LC/MS and moved to next step as is.

Title compound A45 was prepared from ier/-butyl ((S)-2-((2-((S)-2-(((2H- tetrazol-5-yl)methyl)carbamoyl)indolin-l-yl)-2-oxoethyl)amin o)-l-cyclopentyl-2- oxoethyl)carbamate and succinic anhydride using method I: Ή NMR (400 MHz, DMSO-i/6) δ 1.26-1.34 (2H, m), 1.41-1.48 (2H, m), 1.51-1.67 (4H, m), 2.14-2.20 (IH, m), 2.37-2.44 (3H, m), 3.54-3.67 (3H, m), 4.16-4.26 (3H, m), 4.34-4.38 (IH, d, .7=16 Hz), 4.44-4.48 (IH, d, J=16 Hz), 5.09-5.11 (IH, d, 7=8 Hz), 6.96-7.00 (IH, t, J=8 Hz), 7.13- 7.17 (IH, t, 7=8 Hz), 7.19-7.21 (IH, d, J=8 Hz), 8.02-8.09 (2H, dd, 7=20, 8 Hz), 8.16 (IH, bs), 8.72 (IH, bs), MS (LC/MS) m/z observed 527.10, expected 527.23 [M+H]. EXAMPLE A46

3- { [(15 -2-METHYL-1 -({2-OXO-2- [(2S)-2- [(2H- 1 ,2,3 ,4-TETRAZOL-5-

YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l- YL]ETHYL}CARBAMOYL)PROPYL]CARBAMOYL}PROPANOIC ACID tert-Butyl ((S)-l-((2-((5)-2-(((2H-tetrazol-5-yl)methyl)carbamoyl)indol in-l -yl)-2- oxocthyl)amino)-3-methyl-l -oxobutan-2-yl)carbamate was prepared from 1-3 and (S)-2- ((iert-butoxycarbonyl)amino)-3-methylbutanoic acid using general method A. MS (LC/MS) m/z observed 500.98, expected 501.25 [M+H]. Compound was confirmed using LC/MS and moved to next step as is.

Title compound A46 was prepared from /ert-butyl ((S)-l -((2-((5)-2-(((2H- tetrazol-5-yl)methyl)carbamoyl)indolin-l-yl)-2-oxoethyl)amin o)-3-methyl-l -oxobutan-2- yl)carbamate and succinic anhydride using method I: ! H NMR (400 MHz, DMSO-< 6) δ 0.90-0.96 (6H, m), 2.01-2.08 (IH, m), 2.44-2.50 (2H, m), 3.15-3.22 (2H, m), 3.57-3.66 (2H, m), 4.18-4.22 (IH, d, J=16 Hz), 4.26-4.30 (IH, t, =8 Hz), 4.57-4.61 (IH, d, J=16 Hz), 4.64-4.68 (IH, d, J=16 Hz), 5.20-5.23 (IH, d, J=12 Hz), 7.03-7.07 (I H, t, J=8 Hz), 7.18-7.22 (IH, t, J=8 Hz), 7.26-7.28 (I H, d, .7=8 Hz), 7.91 -7.95 (IH, dd, J=16, 4 Hz), 8.06-8.08 (I H, d, J=8 Hz), 8.21 (IH, bs), 8.27-8.31 (IH, t, .7=8 Hz), 9.14 (I H, bs), MS (LC/MS) m/z observed 501.07, expected 501.21 [M+H] and observed 523.19, expected 523.21 [M+Na].

EXAMPLE A47

2- [(2,2-DIMETHYLPROPANOYL)OXY] ETHYL 3-{[(15',25)-2-METHYL-1-({2-OXO-2-[(25)- 2-[(2H-L,2,3,4-TETRAZOL-5-YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-LH -INDOL-L- YL]ETHYL}CARBAMOYL)BUTYL]CARBAMOYL}PROPANOATE Triethylamine (4.13 mL, 29.63 mmol) was added to ethylene glycol (22.5 mL). Pivaloyl chloride (3.1 mL, 25.19 mL) was then added slowly to this mixture and it was left at RT for 2 hrs. The reaction mixture was diluted with water (25 mL) and the product was extracted with DCM (4 x 20 mL). The combined organic layers were dried over sodium sulphate and concentrated. The product was purified on normal phase using 0 % to 40 % ethyl acetate in hexanes as the eluent to give a 2-hydroxyethyl pivalate as a colorless oil (3.99 g, 97%). *H NMR (400 MHz, CDC1 3 ) δ 1.22-1.25 (9H, s), 2.02 (IH, bs), 3.80-3.85 (2H, m), 4.19-4.23 (2H, m). Title compound A47 (75 mg, 0.146 mmol), 2-hydroxyethyl pivalate (32 mg, 0.21 mmol), EDC (42 mg, 0.219 mmol), DMAP (3.6 mg, 0.0292 mmol) were dissolved in DMF (5 mL). DIPEA (59 μί, 0.584 mmol) was then added and the reaction was heated to 50 for 6 hrs. The reaction mixture was concentrated and the product was purified on a CI 8 column using 10-65% MeOH in water to yield A47 as an off-white solid (35 mg, 37%). Ή NMR (400 MHz, DMSO-tf<i) δ 0.81 (3H, t, J=7.4Hz), 0.87 (3H, d, J=7Hz), 1.06-1.16 (10H, m), 1.45 (IH, m), 1.75 (IH, m), 2.35-2.48 (4H, m), 3.1 1 (IH, m), 3.55-3.65 (2H, m), 4.1 1-4.28 (6H, m), 4.52 (IH, m), 4.65 (IH, m), 5.13 (IH, d, J=l lHz), 7.01 (IH, t, J=8Hz), 7.14-7-26 (2H, m), 7.92 (IH, d, J=9Hz), 8.03 (IH, d, J=8Hz), 8.18 (IH, bs), 9.08 (IH, bs), MS (LC/MS) m/z observed 643.15, expected 643.32 [M+H].

EXAMPLE A48

(l 1 S , ,2S)-2-{[(l 1 S , ,2.S)-2-METHYL-l-({2-oxo-2-[(25 -2-[(2H-l,2,3,4-TETRAZOL-5- YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-LH-INDOL-L- YL] ETHYL}CARBAMOYL)BUTYL]CARBAMOYL}CYCLOPROPANE-1-CARBOXYLIC ACID To the stirring mixture of (lS,2S)-diethyl cyclopropane- 1 ,2-dicarboxylate (0.5 gm, 2.69 mmol) in 1 : 1 mixture of watenTHF (2.7 ml) was added ammonium hydroxide (28%, 3.8 ml). The resulting reaction mixture was sealed in a flask and stirred at RT for 16hrs, The reaction mixture was concentrated to dryness under vacuum. The resulting residue of ( lS,2S)-cyclopropane-l ,2-dicarboxylic acid, was dried well under vacuum and subjected to next reaction as it is.

Title compound A48 was prepared from 1-5 and (lS,2S)-cyclopropane-l ,2- dicarboxylic acid using method J. MS (LC/MS) m/z observed 525.99, expected 527.55 [M+H]. Compound was confirmed using LCMS.

EXAMPLE A49

(25)- 1- {2- [(2S,3£)-3-METH YL-2- [(2Λ)-2- PHENYLPROPANAMIDO] PENTANAMIDO]ACETYL}-7V-(2H-l,2,3,4-TETRAZOL-5- YLMETHYL)-2,3-DIHYDRO-lH-INDOLE-2-CARBOXAMIDE Title compound A49 (7 mg, 0.01 mmol, 10%) was collected as an off white solid from intermediate 1-15 (60 mg, 0.13 mmol) using general method M via reaction with (R)-2-phenyl-propionic acid. Ή NMR (400 MHz, DMSO-</<5) δ 9.00 (IH, bs), 8.23 (IH, bs), 8.04 (IH, d, J=7Hz), 7.96 (IH, d, J=9Hz), 7.35 (2H, d, J=7Hz), 7.28 (2H, t, J=7Hz), 7.25-7.15 (3H, m), 7.01 (IH, t, J=7Hz), 5.17 (IH, d, J=9Hz), 4.64 (IH, d, J=16Hz), 4.50 (IH, d, J=16Hz), 4.23 (IH, t, J=8Hz), 4.10 (IH, dd, .7=10, 5Hz), 3.81 (IH, q, J=7Hz), 3.70-3.55 (2H, m), 3.16 (IH, d,7=5Hz), 1.68 (lH,m), 1.31 (3H, d, J=7Hz), 1.25 (IH, m), 0.97 (IH, m), 0.72 (3H, d, J=7Hz), 0.66 (3H, t, J=7Hz), MS (LC/MS) m/z observed 547.09, expected 547.28 [M+H].

EXAMPLE A50

(25)- 1-{2- [(25,350-3-METH YL-2- [(2S)-2- PHENYLPROPANAMIDO]PENTANAMIDO]ACETYL}-N-(2H-l,2,3,4- KTRAZOL-5- YLMETHYL)-2,3-DIHYDRO-lH-INDOLE-2-CARBOXAMIDE

Title compound A50 (10 mg, 0.01 mmol, 15%) was collected as an off white solid from intermediate 1-15 (60 mg, 0.13 mmol) using general method M via reaction with (S)-2-phenyl-propionic acid. Ή NMR (400 MHz, OMSO-d6) δ 9.06 (IH, bs), 8.15 (IH, bs), 8.00 (2H, d, J=9Hz), 7.32 (2H, d, J=7Hz), 7.27 (2H, t, J=7Hz), 7.23-7.13 (3H, m), 7.00 (IH, t, J=7Hz), 5.13 (IH, d, ,/=10Hz), 4.64 (IH, dd, J=16, 5Hz), 4.53 (IH, dd, J=16, 5Hz), 4.30 (IH, t, J=8Hz), 4.10 (IH, dd, J=16, 5Hz), 3.81 (IH, q, J=7Hz), 3.67-3.50 (2H, m), 3.13 (IH, d, J=16Hz), 1.77 (IH, m), 1.47 (IH, m), 1.33 (3H, d, J=7Hz), 1.18 (IH, m), 0.88 (3H, d, ,/=7Hz), 0.84 (3H, t, J=7Hz), MS (LC/MS) m/z observed 547.05, expected 547.28 [M+H],

EXAMPLE A51

4-{[(15',25)-2-METHYL-l-({2-OXO-2-[(2 1 S)-2-[(2H-l,2,3,4-TETRAZOL-5-

YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l- YL] ETHYL} CARBAMOYL)BUTYL]CARBAMOYL}-2-PHENYLBUTANOIC ACID

Title compound A51 (20 mg, 0.035 mmol, 49%) was collected as an off white solid from intermediate 1-15 (30 mg, 0.07 mmol) using general method N via reaction with 3-phenyl-dihydro-pyran-2,6-dione, as an inseparable mixture of diastereomers. Ή NMR (400 MHz, DMSO-<¾) δ 9.06 (IH, bs), 8.14 (IH, bs), 8.03 (IH, bs), 7.87 (IH, t, J=7Hz), 7.40-7.10 (7H, m), 7.01 (IH, t, J=7Hz), 5.16 (IH, m), 4.65 (IH, d, J=16Hz), 4.53 (IH, d, =16Hz), 4.25 (IH, t, J=7Hz), 4.14 (IH, m), 3.60 (2H, m), 3.50 (IH, t, J=7Hz), 3.13 (IH, d, J=12Hz), 2.25-2.00 (3H, m) ; 1.86 (IH, m), 1.73 (IH, m), 1.43 (IH, m), 1.10 (IH, m), 0.86 (3H, d, J=6Hz), 0.82 (3H, t, J=6Hz), (MS (LC/MS) m/z observed 605.06, expected 605.28 [M+H].

EXAMPLE A52

^-DlMETHYL^-ililS Si-Z-METHYL-l-dl-OXO-l-Ii^- -tiZH-l^^^-TETRAZOL-S- YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l-

YL]ETHYL}CARBAM0YL)BUTYL] CARBAMOYL} BUTANOIC ACID

Title compound A52 (24 mg, 0.04 mmol, 40%) was collected as an off white solid from intermediate 1-15 (50 mg, 0.11 mmol) using general method N via reaction with 3,3-dimethyl-dihydro-pyran-2,6-dione. Ή NMR (400 MHz, DMSO-i6) δ 9.05 (IH, bs), 8.15 (IH, bs), 8.03 (IH, d, J=8Hz), 7.89 (IH, d, J=9Hz), 7.22 (IH, d, =7Hz), 7.17 (lH,t, J=7Hz), 7.01 (IH, t,J=8Hz), 5.17 (IH, d, J=10Hz), 4.65 (IH, dd, J=16, 6Hz), 4.53 (IH, dd, J=16, 4Hz), 4.24 (IH, t, J=8Hz), 4.15 (IH, dd, ,7=16, 4Hz), 3.60 (2H, m), 3.14 (IH, d, J=16Hz), 2.14 (2H, m), 1.80-1.62 (3H, m), 1.45 (IH, m), 1.20-1.10 (7H, m), 0.86 (3H, d, ,/=7Hz), 0.81 (3H, t, J=8Hz), MS (LC/MS) m/z observed 557.16, expected 557.28 [M+H].

EXAMPLE A53

2.2- DlMETHYL-3-{[(lS,25)-2-METHYL-l-({2-OXO-2-[(25)-2-[(2H-l,2,3 ,4-TETRAZOL-5-

YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-lNDOL-l- YL] ETHYL} CARBAMOYL) BUTYL]CARBAMOYL}PROPANOIC ACID Title compound A53 (22 mg, 0.04 mmol, 62%) was collected as an off white solid from intermediate 1-15 (30 mg, 0.07 mmol) using general method N via reaction with

3.3- dimethyl-dihydro-furan-2,5-dione. *H NMR (400 MHz, DMSO-</6) δ 9.09 (IH, bs), 8.17 (IH, bs), 8.02 (IH, d, J=9Hz), 7.81 (IH, d, J=9Hz), 7.22 (IH, d, J=7Hz), 7.17 (IH, t, J=8Hz), 7.01 (IH, t, J=8Hz), 5.17 (IH, d, J=12Hz), 4.66 (IH, dd, J=16, 5Hz), 4.55 (IH, dd, 7=16, 4Hz), 4.26 (IH, t, J=8Hz), 4.14 (IH, dd, J=\6 t 5Hz), 3.60 (2H, m), 3.14 (IH, d, J=16Hz), 2.46 (IH, d, .7=1 lHz), 2.41 (IH, d, J=llHz), 1.74 (IH, m), 1.45 (IH, m) 1.19 (IH, m), 1.12 (3H, s), 1.11 (3H, s), 0.86 (3H, d, J=6Hz), 0.81 (3H, t, 7=7Hz), MS (LC/MS) m/z observed 543.04, expected 543.27 [M+H]. EXAMPLE A54

4- { [(15',25)-2-METHYL-l-({2-OXO-2-[(2iS)-2-[(2H-l,2,3,4-TETRAZO L-5-

YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l- YL]ETHYL}CARBAMOYL)BUTYL]CARBAMOYL}-4-PHENYLBUTANOIC ACID

Title compound A54 (25mg, 0.08 mmol, 37%) was prepared from 1-15 (lOOmg,

0.22 mmol) and 3-phenyl-dihydro-pyran-2,6-dione using method P, where the hydrolysis step was stopped after 8h. A54 was separated by chromatography, into two diastereomers, A54-1 (eluted at 63% MeOH in water) and A54-2 (eluted at 65% MeOH in water), each characterized as below:

A54-1:

Ή NMR (400 MHz, DMSO-rf6) δ 8.70 (IH, bs), 8.22-8.10 (2H, bs), 8.01 (IH, d, J=8Hz), 7.37-7.10 (7H, m), 6.99 (IH, t, J=8Hz), 5.08 (IH, d, J=9Hz), 4.51 (IH, d, J=14Hz), 4.40-4.20 (2H, m), 4.06 (IH, d,J=13Hz), 3.66 (IH, m), 3.51 (IH, m), 3.38 (IH, m), 3.06 (IH, d, J=18Hz), 2.20-2.05 (3H, m), 1.83 (IH, m), 1.77 (IH, m), 1.45 (IH, m), 1.12 (IH, m), 0.89 (3H, d, J=7Hz), 0.84 (3H, t, J=7Hz), MS (LC/MS) mz observed 605.09, expected 605.28 [M+H],

A54-2:

! H NMR (400 MHz, DMSO-i/6) δ 9.07 (IH, bs), 8.20 (IH, bs), 8.10-8.00 (2H, m), 7.40-7.10 (7H, m), 7.01 (IH, t, J=7Hz), 5.17 (IH, d, J=10Hz), 4.65 (IH, dd, J=16, 5Hz), 4.54 (IH, dd, J=16, 4Hz), 4.28-4.12 (2H, m), 3.68 (IH, t, J=6Hz), 3.61 (IH, m), 3.39 (IH, m), 3.14 (IH, d, J=16Hz), 2.25-2.00 (3H, m), 1.84 (IH, m), 1.70 (IH, m), 1.23 (IH, m), 0.89 (IH, m), 0.70 (3H, d, J=6Hz), 0.82 (3H, t, J=7Hz), MS (LC/MS) m/z observed 605.13, expected 605.28 [M+H],

EXAMPLE A55

4,4-DiMETHYL-4-{[(l 1 S',2S)-2-METHYL-l-({2-oxo-2-[(2S -2-[(2H-l,2,3,4-TETRAZOL-5-

YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l- YL]ETHYL}CARBAMOYL)BUTYL]CARBAMOYL}BUTANOIC ACID

Title compound A55 (9mg, 0.03 mmol, 30%) was prepared from 1-15 (51 mg, 0.11 mmol) and 3,3-dimethyl-dihydro-pyran-2,6-dione using method P, where the hydrolysis step was stopped after 20h. ! H NMR (400 MHz, DMSO-i/6) δ 9.06 (IH, bs), 8.15 (IH, bs), 8.05 (IH, d, J=8Hz), 7.30-7.15 (3H, m) 7.02 (IH, t, ,/=8Hz), 5.17 (IH, d, J=10Hz), 4.64 (IH, dd, J=16, 5Hz), 4.54 (IH, dd, J=\6, 5Hz), 4.24 (IH, t, J=8Hz), 4.19 (I H, m), 3.61 (2H, m), 3.39 (I H, m), 3.14 (IH, d, J=16Hz), 2.12 (2H, m), 1.86 ( I H, m), 1.75 (I H, m), 1.48 (] H, m), 1.20- 1.05 (7H, m), 0.89 (3H, d, J=7Hz), 0.83 (3H, t, J=7Hz), MS (LC/MS) m/z observed 557.10, expected 557.28 [M+H]

EXAMPLE A56

3,3-DlMETHYL-3-{[(l^,25)-2-METHYL-l-({2-OXO-2-[(2-?)-2-[(2H- l,2,3,4-TETRAZOL-5-

YLMETHYL)CARBAMOYL] -2,3-DIHYDRO- 1H-INDOL- 1 - YL] ETHYL}CARBAMOYL)BUTYL]CARBAMOYL}PROPANOIC ACID Title compound A56 (11 mg, 0.02 mmol, 11%) was prepared as an off-white solid, from 1-15 (80 mg, 0.18 mmol) and 3,3-dimethyl-dihydro-furan-2,5-dione using method P, where the hydrolysis step was stopped after 7h. ] H NMR (400 MHz, DMSO- d6) δ 9.04 (IH, bs), 8.17 (IH, bs), 8.02 (IH, d, J=8Hz), 7.82 ( I H, d, J=9Hz), 7.22 (I H, d, J=7Hz), 7.17 (IH, t, J=8Hz), 7.01 (IH, t, J=8Hz), 5.17 (I H, d, 7=10Hz), 4.63 (IH, dd, J=16, 5Hz), 4.53 (IH, dd, J=16, 4Hz), 4.25 (IH, t, J=8Hz), 4.14 (IH, dd, J=16, 4Hz), 3.62 (2H, m), 3.14 (I H, d, J=16Hz), 2.48 (I H, d, J=16Hz), 2.42 (I H, d, ,/=16Hz), 1 .76 (I H, m), 1.45 (IH, m), 1.25-1.05 (7H, m), 0.86 (3H, d, =6Hz), 0.82 (3H, t, J=7Hz), MS (LC/MS) m/z observed 543.10, expected 543.27 [M+H]

EXAMPLE A57

2-{[(l 1 S , ,2,y)-2-METHYL-l-({2-oxo-2-[(2 1 S -2-[(2H-l,2,3,4-TETRAZOL-5- YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-LH-INDOL-L-

YL]ETHYL}CARBAMOYL)BUTYL]CARBAMOYL}-TRANS-CYCLOPENTANE-1- CARBOXYLIC

ACID

Title compound A57 was prepared from 1-15 (71 mg, 0.16 mmol) and cis- cyclopentane-l,2-dicarboxylic acid using method J without HC1 treatment and was further separated by chromatography, into two diastereomers, A57-1 (9.9 mg, 0.04 mmol, 23%) and A57-2 (6.7 mg, 0.02 mmol, 15%), each characterized as below:

A57-1 :

Ή NMR (400 MHz, DMSO-rf6) δ 8.90 (IH, bs), 8.15 (IH, bs), 8.03 (IH, d, J=8Hz), 7.80 (IH, d, J=9Hz), 7.22 (IH, d, J=7Hz), 7.17 (IH, t, J=8Hz), 7.00 (IH, t, J=7Hz), 5.15 (IH, d, J=9Hz), 4.57 (IH, d, J=12Hz), 4.45 (IH, d, J=12Hz), 4.27 (IH, t, J=8Hz), 4.16 (IH, dd, J=9, 5Hz), 3.69 (IH, dd, J=8, 4Hz), 3.64 (I H, m), 3.1 1 (I H, d, J=14Hz), 3.06-2.85 (2H, m), 2.10-1.82 (2H, m), 1.80-1.66 (2H, m), 1.66-1.52 (3H, m), 1.46 (IH, m), 1.09 (IH, m), 0.86 (3H, d, J=6Hz), 0.81 (3H, t, J=7Hz), MS (LC/MS) m/z observed 554.94, expected 555.27 [M+H].

A57-2:

Ή NMR (400 MHz, DMSO-<6) δ 8.81 (IH, bs), 8.13 (IH, bs), 8.10-7.95 (2H, m), 7.21 (IH, d, J=7Hz), 7.16 (IH, t, J=8Hz), 6.99 (IH, t, J=8Hz), 5.15 (IH, d, J=10Hz), 4.49 (IH, dd, J=17, 6Hz), 4.45 (IH, d, J=17, 6Hz), 4.25 (IH, t, J=8Hz), 4.17 (IH, dd, J=]4, 5Hz), 3.69 (IH, dd, J=13, 4Hz), 3.58 (IH, dd, J=15, 8Hz), 3.05 (IH, d, J=16Hz), 2.97 (2H, m), 2.00-1.85 (2H, m), 1.85-153 (5H, m), 1.13 (IH, m), 0.87 (3H, d, ,/=6Hz), 0.82 (3H, t, ,/=7Hz), MS (LC/MS) m/z observed 554.98, expected 555.27 [M+H].

Examples B1-B7 were prepared by the representative synthetic pathway illustrated schematically in FIGURE 2.

EXAMPLE B 1

(25)-l-{2-[(2iS',3i9)-3-METHYL-2-(2-PHENYLACETAMIDO)PENTANAM IDO]ACETYL}-N- (lH-l,2,3-TRIAZOL-4-YLMETHYL)-2,3-DIHYDRO-lH-INDOLE-2-CARBOX AMIDE

Title compound Bl was prepared from 1-9 and (lH-[l,2,3]triazol-4- yl)methanamine using method G followed by purification by preparative HPLC (Column: ASCENTIS™ C18, 25 cm x 21.2 mm, ΙΟμηι, gradient 0%→100% Methanol/water with 0.1% TFA, 10 mL/min) (11 mg, cream-coloured powder). Ή NMR, (300 MHz, DMSO- d6) δ 8.91-8.83 (m, IH), 8.29-8.02 (m, 2H), 7.74-7.64 (m, IH), 7.30-7.15 (m, 7H), 7.03- 6.97 (m, IH), 5.15-5.04 (m, IH), 4.44-4.07 (m, 4H), 3.63-3.02 (m, 5H), 1.90-1.68 (m, IH), 1.50-1.21 (m, IH), 1.16-1.01 (m, IH), 0.85-0.76 (m, 6H). MS (ESI) m/z observed 532.33 observed, expected 532.27 [M+H],

EXAMPLE B2

(25)-l-{2-[(25,35)-3-METHYL-2-(2-PHENYLACETAMIDO)PENTANAIViI DO]ACETYL}-N- (l,2,3-THIADIAZOL-4-YLMETHYL)-2,3-DIHYDRO-lH-INDOLE-2-CARBOX AMIDE

Title compound B2 was prepared from 1-9 and ([l,2,3]thiadiazol-4- yl)methanamine using method G. Ή NMR, (300 MHz, DMSO-J6) δ 9.15-8.96 (m, IH), 8.26-7.98 (m, 2H), 7.31-7.15 (m, 8H), 7.04-6.94 (m, IH), 5.19-5.10 (m, IH), 4.87-4.68 (m, 2H), 4.31-4.22 (m, IH), 4.18-4.01 (m, IH), 3.66-3.43 (m, 4H), 3.12-3.02 (m, IH), 1.82-1.66 (m, IH), 1.51-1.37 (m, IH), 1.18-1.03 (m, IH), 0.89-0.76 (m, 6H). MS (ESI) m/z observed 583.20, expected 583.19 [M+Cl] and observed 549.20, expected 549.23 [M+H] and observed 571.20, expected 571.21 [M+Na].

EXAMPLE B3

(2S)-l-{2-[(2S,35 , )-3-METHYL-2-(2-PHENYLACETAMIDO)PENTANAMIDO]ACETYL}-V- [l- (lH-l,2,3-TRIAZOL-4-YL)ETHYL]-2,3-DIHYDRO-lH-INDOLE-2-CARBOX AMIDE

Title compound B3 was prepared from 1-9 and l-(2H-tetrazol-5-yl)ethanamine using method G as a mixture of diastereomers. Ή NMR, (300 MHz, CD 3 OD) δ 7.35- 7.21 (m, 8H), 7.09-7.03 (m, IH), 5.39-5.32 (m, IH), 5.17-5.09 (m, IH), 4.38-4.32 (m, 2H), 3.62-3.58 (m, 3H), 3.26-3.14 (m, 2H), 1.95-1.84 (m, IH), 1.68-1.61 (m, 3H), 1.59- 1.47 (m, IH), 1.22-1.09 (m, IH), 0.98-0.86 (m, 6H). MS (ESI) m/z observed 545.48, expected 545.26 [M-H],

EXAMPLE B4

(2 i y)-7V-[(4-METHYL-lH-l,2,3-TRIAZOL-5-YL)METHYL]-l-{2-[( 2S,3S)-3-METHYL-2-(2- PHENYLACETAMIDO)PENTANAMIDO]ACETYL}-2,3-DIHYDRO-lH-INDOLE-2- CARBOXAMIDE

Title compound B4 was prepared from 1-9 and (4-methyl-lH-[l,2,3]triazol-5- yl)methanamine using method C. 'H NMR, (300 MHz, DMSO-<6) δ 8.76 (br s, IH), 8.20 (br s, IH), 8.11-8.00 (m, IH), 7.31-7.15 (m, 8H), 7.03-6.97 (m, IH), 4.43-4.24 (m, 3H), 4.19-4.05 (m, IH), 3.61-3.34 (m, 5H), 3.04-2.98 (m, IH), 2.17 (s, 3H), 1.89-1.69 (m, IH), 1.48-1.19 (m, 2H), 0.85-0.77 (m, 6H). MS (ESI) m/z observed 544.52, expected 544.27 [M-H].

EXAMPLE B5

(25)-l-{2-[(25 , ,35')-3-METHYL-2-(2-PHENYLACETAMIDO)PENTANAMIDO]ACETYL }-N- (lH-l,2,3-TRIAZOL-4-YLMETHYL)-l,2,3,4-TETRAHYDROQUINOLINE-2- CARBOXAMIDE Title compound B5 was prepared from 1-8 and l,2,3,4-tetrahydroquinoline-2- carboxylic acid using method F followed by addition of l-(lH-[l,2,3]triazol-4- yl)methanamine using method C, as a mixture of diastereomers. Ή NMR, (300 MHz, acetone-i6) δ 7.83 (br s, IH), 7.61-7.17 (m, 9H), 5.18 (t, IH), 4.45-4.33 (m, 4H), 3.85- 3.76 (m, IH), 3.67-3.55 (m, 2H), 2.79-2.69 (m, IH), 2.62-2.45 (m, 2H), 1.96-1.78 (m, 2H), 1.53-1.40 (m, IH), 1.15-1.04 (m, IH), 0.88-0.78 (m, 6H). MS (ESI) m/z observed 546.60, expected 546.28 [M-H]. EXAMPLE B6

6-METHOXY-l-{2-[(2^,3S)-3-METHYL-2-(2- PHENYLACETAMIDO)PENTANAMIDO]ACETYL}- V-(lH-l ,2,3-TRIAZOL-5-YLMETHYL)- 2,3-DIHYDRO-lH-INDOLE-2-CARBOXAIVHDE

Title compound B6 was prepared from 1-8 and 5-mefhoxy-indoline-2-carboxylic acid methyl ester using method F followed by ester hydrolysis using method D and finally addition of l -(lH-[l ,2,3]triazol-4-yl)methanamine using method C. 'Η NMR, (300 MHz, DMSO-<¾) δ 8.87-8.78 (m, IH), 8.26-7.98 (m, 2H), 7.74-7.64 (m, IH), 7.30- 7.24 (m, 5H), 7.24-7.16 (m, I H), 7.12-7.06 (m, IH), 6.63-6.56 (m, IH), 5.15-5.06 (m, IH), 4.44-4.24 (m, 3H), 3.72 (s, 3H), 3.62-3.43 (m, 4H), 3.16 (s, I H), 3.01 -2.92 (m, IH), 1.90-1.70 (m, I H), 1 .49-1.26 (m, IH), 1.15-1.02 (m, I H), 0.85-0.77 (m, 6H). MS (ESI) m/z observed 560.59, expected 560.26 [M+H].

EXAMPLE B7

5-METHYL-1-{2-[(2S , ,35 -3-METHYL-2-(2- PHENYLACETAMIDO)PENTANA IDO]ACETYL}-N-(lH-l,2,3-TRIAZOL-4-YLMETHYL)-

2,3-DIHYDRO-lH-INDOLE-2-CARBOXAMIDE

Title compound B7 was prepared from 1-8 and 6-methoxy-indoline-2-carboxylic acid ethyl ester using method C followed by ester hydrolysis using method D and finally addition of l -( lH-[ l ,2,3]triazol-4-yl)methanamine using method C as a mixture of diastereomers. Ή NMR, (300 MHz, CD 3 OD) δ 8.86-8.62 (m, I H), 8.32-7.93 (m, 2H), 7.68 (m, I H), 7.37-6.95 (m, 8H), 5.18-5.02 (m, 2H), 4.55-4.44 (m, 3H), 4.38-4.29 (m, I H), 3.70-3.54 (m, 3H), 3.22-3.05 (m, IH), 2.30 (s, 3H), 2.03- 1 .84 (m, I H), 1.60- 1 .49 (m, I H), 1 .25-1 .10 (m, I H), 0.96-0.86 (m, 6H). MS (ESI) m/z observed 544.53, expected 544.27 [M-H].

Examples C 1 -C39 were prepared by the representative synthetic pathway illustrated schematically in FIGURE 3.

EXAMPLE C I

(2-y)-l-{2-[(2S)-3-METHYL-2-(2-PHENYLACETAMIDO)BUTANAMIDO]AC ETYL}-iV-(2H- l,2,3,4-TETRAZOL-5-YLMETHYL)-2,3-DIHYDRO-lH-INDOLE-2-CARBOXA MIDE 1-6 (0.5 g, 3.67 mmol) and thionyl chloride (1.6 ml, 22 mmol) were stirred together for 1 hr at room temperature. Thionyl chloride was removed by distillation under vacuum. The acid chloride was added to the stirring solution of L-valine (0.39 g, 3.305 mmol) in NaOH (2N, 4.2 ml) at 0 °C. The resulting reaction mixture was warmed to RT and stirred overnight. The mixture was washed with diethyl ether (5 ml) and acidified to pH 4-5 by adding citric acid (aqueous, saturated solution). The precipitated solid was filtered, washed with diethyl ether and dried to yield (S)-3-methyl-2-(2- phenylacetamido)butanoic acid as a white solid (0.64 g, 74%). Ή NMR (400 MHz, DMSO-i/6) δ 0.82-0.84 (3H, d, J=8 Hz), 0.85-0.87 (3H, d, = Hz), 1.99-2.06 ( I H, m), 3.44-3.55 (2H, q, =12 Hz), 4.10-4.14 (I H, dd, .7=8, 12 Hz), 7.16-7.21 (I H, m), 7.24-7.29 (4H, m), 7.19-7.21 (I H, d, J=8 Hz), 12.55 ( I H, s), MS (LC/MS) m/z observed 236.04, expected 236.13 [M+H] . The compound was used further as described.

Title compound CI was prepared from 1-3 and (S)-3-methyl-2-(2- phenylacetamido)butanoic acid using method A: Ή NMR (400 MHz, DMSO-rfd) δ 0.81 -0.85 (6H, m), 1.94-2.01 ( I H, m), 3.04-3.10 (2H, m), 3.42-3.45 ( I H, d, J=12 Hz), 3.53-3.58 (3H, m), 4.1 1 -4.15 ( IH, d, J=16 Hz), 4.22-4.26 (IH, q, J=12 Hz), 4.37-4.43 (I H, t, J=12 Hz), 4.47-4.54 (IH, m), 5.12-5.14 (IH, d, J=8 Hz), 6.96-7.00 (I H, t, J=S Hz), 7.13-7.21 (3H, m), 7.25-7.26 (4H, d, J=4 Hz), 8.02-8.03 (IH, d, J=4 Hz), 8.05- 8.13 (IH, m), 8.26(1 H, bs), 8.77(1H, bs), MS (LC/MS) m/z observed 519.01 , expected 5 19.25 [M+H] and observed 541 .1 1 , expected 541.23 [M+Na].

EXAMPLE C2

(2S)-l-{2-[(2,?,3i?)-3-HYDROXY-2-(2-PHENYLACETAMrDO)BUTANAMI DO]ACETYL}-N- (2H-l,2,3,4-TETRAZOL-5-YLMETHYL)-2,3-DIHYDRO-lH-INDOLE-2-CAR BOXAMIDE

1-6 (0.5 g, 3.67 mmol) and thionyl chloride ( 1.6 ml, 22 mmol) were stirred together for 1 hr at room temperature. Thionyl chloride was removed by distillation under vacuum. The acid chloride was added to the stirring solution of L-threonine (0.39 g, 3.305 mmol) in NaOH (2N, 4.2 ml) at 0 °C. The resulting reaction mixture was warmed to RT and stirred overnight. The reaction mixture was washed with diethyl ether (5 ml) and acidified to pH 4-5 by adding citric acid (aqueous, saturated solution). The precipitated solid was filtered, washed with diethyl ether and dried to yield (2S,3R)-3- hydroxy-2-(2-phenylacetamido)butanoic acid as a white solid (0.71 g, 82%). Ή NMR (400 MHz, DMSO-ito) δ 0.99- 1 .01 (3H, d, J=8 Hz), 3.49-3.60 (2H, q, 7=12 Hz), 4.08- 4.13 (I H, m), 4.17-4.20 ( I H, dd, J=4,8 Hz), 4.89 (l H,bs), 7.16-7.31 (5H, m), 7.97-7.99 (IH, d, J=8 Hz), 12.42 (IH, bs), MS (LCMS) m/z observed 238.00, expected 238.11 [M+H]. The compound was used further as described.

Title compound C2 was prepared from 1-3 and (2 L S',3R)-3-hydroxy-2-(2- phenylacetamido)butanoic acid using method A: 'I! NMR (400 MHz, DMSO-rf<5) δ 1.00-1.02 (3H, d, /=8 Hz), 1.66-1.74 (IH, m), 3.06-3.12 (2H, m), 3.49-3.53 (IH, t, J=8 Hz), 3.55-3.62 (2H, m), 3.98-4.02 (IH, t, .7=8 Hz), 4.14-4.18 (IH, d, .7=16 Hz), 4.24-4.27 (IH, dd, = , 8 Hz), 4.44-4.49 (IH, m), 4.54-4.62 (IH, m), 5.12-5.18 (IH, m), 6.96-7.02 (IH, m), 7.16-7.22(3H, m), 7.26-7.28 (4H, q, 7=4 Hz), 7.99-8.03 (IH, t, J=8 Hz), 8.08 (IH, bs), 8.99 (IH, bs), MS (LC/MS) /z observed 521.04, expected 521.23 [M+H] and observed 543.11, expected 543.21 [M+Na].

EXAMPLE C3

(2S)-l-{2-[(25',3S)-3-HYDROXY-2-(2-PHENYLACETAMIDO)BUTANA 1DO]ACETYL}-7V- (2H-l,2,3,4-TETRAZOL-5-YLMETHYL)-2,3-DIHYDRO-lH-INDOLE-2-CAR BOXAMIDE

(2S,3S)-3-Hydroxy-2-(2-phenylacetamido)butanoic acid was prepared from 1-6 and (2S,3S)-2-amino-4-hydroxybutanoic acid using method H: ] H NMR (400 MHz, DMSO-i/6) δ 1.05 (3H, d, J=6.4 Hz), 3.55 (2H, s), 3.85 (IH, m), 4.17 (IH, dd, J=4.0, 8.4 Hz), 4.89 (lH,bs), 7.13-7.30 (5H, m), 8.19 (IH, d, J=8.4 Hz), 12.35 (IH, bs), MS (LC/MS) m/z observed 237.86, expected 238.11 [M+H], Compound was used further as described.

Title compound C3 was prepared from (2S,3S)-3-hydroxy-2-(2- phenylacetamido)butanoic acid and 1-4 using method A: Ή NMR (400 MHz, DMSO- d6) δ 0.97-1.07 (3H, m), 2.93-3.15 (2H, m), 3.45-3.70 (3H, m), 3.82-3.88 (IH, m), 4.11- 4.20 (IH, m), 4.22-4.30 (IH, m), 4.32-4.42 (IH, m), 4.45-4.55 (IH, m), 5.15 (IH, d, J=9.5Hz), 6.97 (IH, t, J=7.4Hz), 7.13-7.30 (7H, m), 7.92-8.28 (3H, m), 8.75 (IH, bs), MS (LC/MS) m/z observed 521.00, expected 521.23 [M+H].

EXAMPLE C4

(25)-l-{2-[(25)-4-METHYL-2-(2-PHENYLACETAMIDO)PENTANAMIDO]AC ETYL}-iV-(2H- l,2,3,4-TETRAZOL-5-YLMETHYL)-2,3-DIHYDRO-lH-INDOLE-2-CARBOXA MIDE

(S)-4-Methyl-2-(2-phenylacetamido)pentanoic acid was prepared from 1-6 and (S)-2-amino-4-methylpentanoic acid using method H: Ή NMR (400 MHz, DMSO-id) δ 0.85 (3H, d, J=6.4 Hz), 1.05 (3H, d, .7=6.4 Hz), 1.45-1.53 (2H, m), 1.56-1.66 (IH, m), 3.40-3.50 (2H, m), 4.15-4.25 (I H, m), 7.13-7.30 (5H, m), 8.30 (I H, d, J=8.1 Hz), 12.50 (I H, bs), MS (LC/MS) m/z observed 249.98, expected 250.14 [M+H]. Compound was used further as described.

Title compound C4 was prepared from (S)-4-methyl-2-(2- phenylacetamido)pentanoic acid and 1-3 using method A: Ή NMR (400 MHz, DMSO- d6) δ 0.77 (3H, d, J=6.4 Hz), 0.88 (3H, d, .7=6.4 Hz), 1.45-1.53 (2H, m), 1.55- 1.66 (I H, m), 2.95-3.12 (2H, m), 3.40-3.65 (3H, m), 4.07-4.15 (I H, m), 4.3 1 -4.43 (2H, m), 4.45- 4.55 (IH, m), 5.12 (I H, m), 7.01 (IH, t, J=7Hz), 7.15-7.33 (7H, m), 7.97-8.10 (IH, d, J=8Hz), 8.20-8.38 (2H, m), 8.72 (IH, bs), MS (LC/MS) m/z observed 533.00, expected 533.26 [M+H].

EXAMPLE C5

(25)-l-{2-[(25)-2-(2-PHENYLACETAMIDO)PENTANAMIDO]ACETYL}-7V- (2H-l,2,3,4- TETRAZOL-5-YLMETHYL)-2,3-DIHYDRO-lH-INDOLE-2-CARBOXAMIDE

(S)-2-(2-Phenylacetamido)pentanoic acid was prepared from 1-6 and (<S)-2- aminopentanoic acid using method H: ! H NMR (400 MHz, DMSO-</6) δ 0.85 (3H, d, J=7 Hz), 1 .25- 1.35 (2H, m), 1.52-1.70 (2H, m), 3.42-3.50 (2H, m), 4.13-4.20 (I H, m), 7.15-7.32 (5H, m), 8.30 (I H, d, J=8 Hz), 12.50 (IH, bs), MS (LC/MS) m/z observed 235.97, expected 236.13 [M+H]. The compound was used further as described.

Title compound C5 was prepared from (S)-2-(2-phenylacetamido)pentanoic acid and 1-3 using method A: ] H NMR (400 MHz, DMSO-i/6) δ 0.83 (3H, d, .7=6.4 Hz), 1.25- 1 .35 (2H, m), 1 .45- 1 .57 ( I H, m), 1.61 -1.73 (I H, m), 2.95-3.12 (2H, m), 3.42-3.65 (3H, m), 4.07-4.18 (I H, m), 4.31 -4.43 (2H, m), 4.45-4.55 ( I H, m), 5.12 ( I H, m), 7.01 ( I H, t, J=7Hz), 7.15-7.33 (7H, m), 7.97-8.10 ( I H, d, 7=8Hz), 8.20-8.38 (2H, m), 8.75 (I H, bs), MS (LC/MS) m/z observed 518.99, expected 519.25 [M+H].

EXAMPLE C6

(2^)-l-{2-[(25)-3,3-DlMETHYL-2-(2-PHENYLACETAMIDO)BUTANAMIDO ]ACETYL}-7V- (2H-l,2,3,4-TETRAZOL-5-YLMETHYL)-2,3-DIHYDRO-lH-INDOLE-2-CAR BOXAMIDE

(S)-3,3-Dimethyl-2-(2-phenylacetamido)butanoic acid was prepared from 1-6 and (S)-2-amino-3,3-dimethylbutanoic acid using method H: Ή NMR (400 MHz, DMSO- d6) δ 0.93 (9H, s), 3.47 (IH, d, J=14Hz), 3.57 (IH, d, J=14Hz), 4.10 (IH, d, J=9Hz), 7.10-7.22 (IH, m), 7.24-7.32 (4H, m), 8.1 5 (I H, d, .7=9 Hz), 12.58 (I H, bs), MS (LC/MS) m/z observed 249.96, expected 250.14 [M+H]. The compound was used further as described.

Title compound C6 was prepared from (S)-3,3-dimethyl-2-(2- phenylacetamido)butanoic acid and 1-3 using method A: Ή NMR (400 MHz, DMSO- d6) δ 0.91 (9H, s), 2.95-3.12 (2H, m), 3.40-3.70 (3H, m), 4.07-4.18 ( IH, m), 4.28-4.40 (2H, m), 4.45-4.55 (I H, m), 5.12 (IH, m), 6.99 (I H, t, J=7Hz), 7.12-7.3 1 (7H, m), 7.95- 8.07 (I H, m), 8.30-8.50 (2H, m), 8.73 ( I H, bs), MS (LC/MS) m/z observed 533.03, expected 533.26 [M+H] .

EXAMPLE C7

(25)-l-{2-[(25',3/?)-3-METHOXY-2-(2-PHENYLACETAMIDO)BUTANAMI DO]ACETYL}- V- (2H-l,2,3,4-TETRAZOL-5-YLMETHYL)-2,3-DIHYDRO-lH-INDOLE-2-CAR BOXAMIDE

(2S,3R)-3-Methoxy-2-(2-phenylacetamido)butanoic acid was prepared from 1-6 and (2S,3R)-2-amino-3-methoxybutanoic acid using method H: Ή NMR (400 MHz, DMSO-rf6) δ 1.01 (3H, d, J=6Hz), 3.34 (3H, s), 3.51 (IH, d, J= 14Hz), 3.61 (I H, d, J=14Hz), 3.81 -3.85 ( I H, m), 4.35 (IH, dd, =3Hz, 9Hz), 7.16-7.22 (IH, m), 7.24-7.32 (4H, m), 8.17 (I H, d, J=9 Hz), 12.61 ( IH, bs), MS (LC/MS) m/z observed 252.01 , expected 252.12 [M+H]. The compound was used further as described.

Title compound C7 was prepared from (2S,3R)-3-methoxy-2-(2- phenylacetamido)butanoic acid and 1-4 using method C: *H NMR (400 MHz, DMSO- d6) δ 1 .04 (3H, m), 3.10-3.16 (2H, m), 3.24 (3H, s), 3.45-3.75 (4H, m), 4.1 1 -4.25 (I H, m), 4.37-4.42 (I H, m), 4.50-4.70 (2H, m), 5.15 ( IH, d, J=9.5Hz), 6.97 (I H, t, J=7.4Hz), 7. 13-7.30 (7H, m), 7.97-8.18 (3H, m), 9.12 ( IH, bs), MS (LC/MS) m/z observed 535.04, expected 535.24 [M+H].

EXAMPLE C8

(25 -l-{2-[(2S)-3-(7'£ir-BUTOXY)-2-(2-PHENYLACETAMIDO)PROPANAMI DO]ACETYL}- N-(2H-l,2,3,4-TETRAZOL-5-YLMETHYL)-2,3-DIHYDRO-lH-INDOLE-2-C ARBOXA IDE

(S)-3-(/ert-Butoxy)-2-(2-phenylacetamido)propanoic acid was prepared from 1-6 and (S)-2-amino-3-(/er?-butoxy)propanoic acid using method Η: Ή NMR (400 MHz, DMSO-dtf) δ 1 .06 (9H, s), 3.48 (I H, m), 3.49-3.57 (2H, m), 3.63 ( I H, dd, J=5Hz, 9Hz), 4.36 (I H, m), 7.16-7.22 ( I H, m), 7.24-7.32 (4H, m), 8.1 5 (I H, d, J=9 Hz), 12.61 (I H, bs), MS (LC/MS) m/z observed 279.86, expected 280.16 [M+H]. The compound was used further as described.

Title compound C8 was prepared from (S)-3-(ier/-butoxy)-2-(2- phenylacetamido)propanoic acid and 1-3 using method A: 1 H NMR (400 MHz, DMSO- d6) δ 1.08 (9H, ms), 2.93-3.15 (2H, m), 3.43-3.70 (5H, m), 4.11-4.20 (IH, m), 4.30-4.55 (3H, m), 5.15 (IH, m), 6.97 (IH, t, J=7.4Hz), 7.13-7.33 (7H, m), 7.95-8.25 (3H, m), 8.75 (IH, bs), MS (LC/MS) m/z observed 563.00, expected 563.27 [M+H].

EXAMPLE C9

(2£)-1-{2-[(25,3Λ)-3-(Γ£Λ7·-Βυτοχγ)-2-(2- PHENYLACETAMIDO)BUTANAMIDO]ACETYL}-yV-(2//-l,2,3,4-TETRAZOL- 5-YLMETHYL)-

2,3-DIHYDRO-lH-INDOLE-2-CARBOXAMIDE

(2S,3R)-3-(½rf-Butoxy)-2-(2-phenylacetamido)butanoic acid was prepared from 1-6 and (2S,3R)-2-amino-3-(/ert-butoxy)butanoic acid using method H: Ή NMR (400 MHz, DMSC 6) δ 1.06 (3H, d, J=6Hz), 1.09 (9H, s), 3.48 (IH, d, J=14Hz), 3.56 (IH, d, ,/=14Hz), 3.93 (IH, m), 4.33 (IH, dd, J=5Hz, 9Hz), 7.16-7.22 (IH, m), 7.24-7.32 (4H, m), 8.18 (IH, d, J=9 Hz), 12.61 (IH, bs), MS (LC/MS) m/z observed 293.85, expected 294.17 [M+H]. The compound was used further as described.

Title compound C9 was prepared from (25',3R)-3-(/er/-butoxy)-2-(2- phenylacetamido)butanoic acid and 1-4 using method C: J H NMR (400 MHz, DMSO- d6) δ 1.02 (3H, m), 1.13 (9H, s), 3.10-3.16 (2H, m), 3.50-3.70 (3H, m), 3.88-3.95 (IH, m), 4.11-4.25 (111, m), 4.30-4.37 (IH, m), 4.47-4.67 (2H, m), 5.15 (IH, d, J=9.5Hz), 6.97 (IH, t, J=7.4Hz), 7.13-7.30 (7H, m), 7.85 (IH, d, J=8Hz), 8.00-8.10 (2H, m), 9.12 (IH, bs), MS (LC/MS) m/z observed 577.11, expected 577.29 [M+H].

EXAMPLE CIO

(2 ι ϊ)-1-{2-[(25',3 ι ?)-3-(Γ£ ' Λ7--Βυτοχγ)-2-(2-

PHENYLACETAMlDO)BUTANAMIDO]ACETYL}-N-(2H-l,2,3,4-TETRAZOL -5-YL ETHYL)- 2,3-DIHYDRO- 1H-INDOLE-2-CARBOXA 1Y1IDE

(2S,3S)-3-(?er/-Butoxy)-2-(2-phenylacetamido)butanoic acid was prepared from 1-6 and (2S,3S)-2-amino-3-(/er/-butoxy)butanoic acid using method H: Ή NMR (400 MHz, DMSO-tftf) δ 1.03 (3H, d, J=6Hz), 1.09 (9H, s), 3.52 (IH, d, J=14Hz), 3.62 (IH, d, J=14Hz), 4.10 (IH, m), 4.27 (IH, dd, J=3Hz, 9Hz), 7.16-7.22 (IH, m), 7.24-7.32 (4H, m), 7.83 ( I H, d, J=9 Hz), 12.53 (IH, bs), MS (LC/MS) m/z observed 293.92, expected 294.17 [M+H]. The compound was used further as described.

Title compound CIO was prepared from (2S,3S)-3-(fer/-butoxy)-2-(2- phenylacetamido)butanoic acid and 1-4 using method C: MS (LC/MS) m/z observed 577.04, expected 577.29 [M+H].

EXAMPLE C I 1

(2S)-l-[(2S)-2-[(2S,3S)-3-METH\i-2-(2- PHENYLACETAMIDO)PENTANAMIDO] PROPANOYL] -N-(2H-1 ,2,3 ,4-TETRAZOL-5- YLMETHYL)-2,3-mHYDRO-lH-lNDOLE-2-CARBOXAMIDE fer/-Butyl ((S)-l-((S)-2-(((2H-tetrazol-5-yl)methyl)carbamoyl)indolin-l -yl)-l- oxopropan-2-yl)carbamate was prepared from 1-2 and (S)-2-((tert- butoxycarbonyl)amino)propanoic acid using method C: MS (LC/MS) m/z observed 415.76, expected 416.20 [M+H], and observed 438.01 , expected 438.19 [M+Na], Compound was confirmed using LC/MS and moved to next step as it was.

Title compound Cl l was prepared from ieri-butyl ((S)-l -((S)-2-(((2H-tetrazol-5- yl)methyl)carbamoyl)indolin- l -yl)- l -oxopropan-2-yl)carbamate and (2S,3S)-3-meth l-2- (2-phenylacetamido)pentanoic acid using method A: ! H NMR (400 MHz, CD 3 OD) δ 0.80-0.88 (6H, m), 1.06-1.13 (IH, m), 1.26- 1.28 (3H, d, J=8 Hz), 1.42- 1.48 ( I H, m), 1 .75- 1.83 (I H, m), 3.20-3.32 (2H, m), 3.48-3.64 (2H, m), 4.20-4.22 (I H, d, J=8 Hz), 4.37-4.41 (I H, d, J=16 Hz), 4.54-4.58 ( I H, d, J=16 Hz), 4.66-4.70 ( I H, d, J= 16 Hz), 5.49-5.5 1 ( I H, d, .7=8 Hz), 7.02-7.05 (IH, t, J=6 Hz), 7.15-7.27 (7H, m), 8.10-8.12 ( I H, d, J=8 Hz), MS (LC/MS) m/z observed 546.97, expected 547.28 [M+H] ,

EXAMPLE C12

(2S)- 1-[(2Λ)-2- [(2_ ,3S)-3-METH YL-2-(2- PHENYLACETAMIDO)PENTANAMIDO]PROPANOYL]-7V-(2H-l,2,3,4-TETRAZ OL-5- YLMETHYL)-2,3-DIHYDRO-lH-INDOLE-2-CARBOXAMIDE

/erf-Butyl ((R)- l -((S)-2-(((2H-tetrazol-5-yl)methyl)carbamoyl)indolin-l-yl)-l - oxopropan-2-yl)carbamate was prepared from 1-2 and {R)-2-({tert- butoxycarbonyl)amino)propanoic acid using method C: MS (LC/MS) m/z observed 415.79, expected 416.20 [Μ+Η], and observed 438.03, expected 438.19 [M+Na], Compound was confirmed using LC/MS and moved to next step as it was. Title compound C12 was prepared from tert-butyl ((R)-l-((S)-2-(((2H-tetrazol-5- yl)methyl)carbamoyl)indolin-l-yl)-l-oxopropan-2-yl)carbamate and (2S,3S)-3-methyl-2- (2-phenylacetamido)pentanoic acid using method A: Ή NMR (400 MHz, CD 3 OD) δ 0.83-0.91 (6H, m), 1.10-1.13 (IH, m), 1.34-1.36 (3H, d, .7=8 Hz), 1.48-1.56 (IH, m), 1.82-1.91 (IH, m), 3.18-3.28 (2H, m), 3.48-3.64 (2H, m), 4.20-4.22 (IH, d, .7=8 Hz), 4.36-4.40 (IH, d, J=16 Hz), 4.54-4.58 (IH, d, J=16 Hz), 4.68-4.72 (IH, d, J=16 Hz), 5.50-5.52 (IH, d, J=8 Hz), 7.02-7.06 (IH, t, J=8 Hz), 7.20-7.34 (7H, m), 8.11-8.13 (IH, d, J=8 Hz), MS (LC/MS) m/z observed, 546.99, expected 547.28 [M+H].

EXAMPLE C13

(4.?)-4-[(2 l S',3iy)-3-METHYL-2-(2-PHENYLACETAlVIIDO)PENTANAMIDO]-5 -OXO-5-[(2iS , )-2- [(2H-l,2,3,4-TETRAZOL-5-YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-li/- INDOL-l-

YL]PENTANOIC ACID

(S)-fcri-Butyl 5-((S)-2-(((2H-tetrazol-5-yl)methyl)carbamoyl)indolin-l-yl)- 4- ((ier?-butoxycarbonyl)amino)-5-oxopentanoate was prepared from 1-2 and (S-5-(tert- butoxy)-2-((/erf-butoxycarbonyl)amino)-5-oxopentanoic acid using method C: MS (LC/MS) m/z observed 529.83, expected 530.27 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

(S)-Ethyl 5-((S)-2-(((2H-tetrazol-5-yl)methyl)carbamoyl)indolin-l-yl)- 4-((2S,3S)- 3-methyl-2-(2-phenylacetamido)pentanamido)-5-oxopentanoate was prepared from (S)- /£T/-butyl 5-((S)-2-(((2H-tetrazol-5-yl)methyl)carbamoyl)indolin-l -y\)-A-{{tert- butoxycarbonyl)amino)-5-oxopentanoate and 1-7 using method F: MS (LC/MS) m/z observed 633.02, expected 633.32 [Μ+Η], Compound was confirmed using LC/MS and moved to next step as it was.

Title compound C13 was prepared from (5)-ethyl 5-((S)-2-(((2H-tetrazol-5- yl)methyl)carbamoyl)indolin-l-yl)-4-((2S,3S)-3-methyl-2-(2- phenylacetamido)pentanamido)-5-oxopentanoate using method D: Ή NMR (400 MHz, DMSO-i6) δ 0.74-0.81 (6H,m), 1.01-1.05 (IH, m), 1.20-1.27 (IH, m), 1.70-1.81 (2H, in), 2.25-2.31 (IH, m), 3.11-3.19 (IH, t, J=16 Hz), 3.36-3.61 (4H, m), 4.19-4.25 (IH, m), 4.32-4.38 (IH, m), 4.42-4.48 (IH, dd, J=8, 16 Hz), 4.54-4.60 (IH, dd, J=8, 16 Hz), 4.62- 4.68 (IH, dd, J=8, 16 Hz), 5.17-5.20 (IH, d, J=12 Hz) 6.99-7.03 (IH, t, J=8 Hz), 7.15- 7.26 (6H, m), 7.99-8.07(2H, m), 8.81 (IH, bs), 8.98 (IH, bs), MS (LC/MS) m/z observed 604.97, expected 605.28 [M+H].

EXAMPLE C14

(3S)-3-[(2-i,3iS -3 -2-(2 ]-4 -4-[(2 1 S r )-2- [(2H-l,2,3,4 -5 ]-2,3 -lH-lNDOL-l-

(S)-ter/-Butyl 4-((S)-2-(((2H-tetrazol-5-yl)methyl)carbamoyl)indolin-l-yl)- 3- ((/eri-butoxycarbonyl)amino)-4-oxobutanoate was prepared from 1-2 and (S)-4-(tert- butoxy)-2-((/eri-butoxycarbonyl)amino)-4-oxobulanoic acid using method C: MS (LC/MS) m/z observed 515.74, expected 516.26 [Μ+Η], Compound was confirmed using LC/MS and moved to next step as it was.

(S)-Ethyl 4-((S)-2-(((2H-tetrazol-5-yl)methyl)carbamoyl)indolin-l-yl)- 3-((2S,3S)- 3-methyl-2-(2-phenylacetamido)pentanamido)-4-oxobutanoate was prepared from (S)- /er/-butyl 4-((5)-2-(((2H-tetrazol-5-yl)methyl)carbamoyl)indolin-l-yl)- 3-((/e/-/- butoxycarbonyl)amino)-4-oxobutanoate and 1-7 using method F: MS (LC/MS) m/z observed 618.93, expected 619.30 [Μ+Η]. Compound was confirmed using LC/MS and moved to next step as it was.

Title compound C14 was prepared from (S)-ethyl 4-((S)-2-(((2H-tetrazol-5- yl)methyl)carbamoyl)indolin-l-yl)-3-((2S,35)-3-methyl-2-(2- phenylacetamido)pentanamido)-4-oxobutanoate using method D: NMR (400 MHz, DMSO-</6) δ 0.68-0.78 (6H,m), 0.95-1.04 (IH, m), 1.22-1.29 (IH, m), 1.66-1.73 (IH, m), 2.55-2.64 (2H, m), 3.15-3.19 (IH, d, J=16 Hz), 3.38-3.42 (IH, d, J=16 Hz), 3.48-3.56 (2H, t, 7=16 Hz), 4.29-4.31 (IH, t, J=16 Hz), 4.39-4.44 (IH, dd, J=4, 16 Hz), 4.57-4.66 (2H, dt, J=4, 16 Hz), 5.61-5.63 (IH, d, J=8 Hz), 7.00-7.04 (IH, t, J=8 Hz), 7.14-7.25 (6H, m), 7.96-7.98 (IH, d, .7=8 Hz), 8.04-8.06 (IH, d, J=8 Hz), 8.75 (IH, bs), 9.07 (IH, bs), MS (LC/MS) m/z observed 590.97, expected 591.27 [M+H] and observed 613.05, expected 613.25 [M+Na] .

EXAMPLE CI 5

(3^)-3-[(25 , ,35)-3- -2-(2- ]-4- -4-[(2S)-2- [(2H-L,2,3,4- -5- ]-2,3- H L- (R)-ierf-Butyl 4-((S)-2-(((2H-tetrazol-5-yl)methyl)carbamoyl)indolin- l -yl)-3- ((/ert-butoxycarbonyl)amino)-4-oxobutanoate was prepared from 1-2 and (R)-4-(tert- butoxy)-2-((/eri-butoxycarbonyl)amino)-4-oxobutanoic acid using method C: MS (LC/MS) m/z observed 515.77, expected 516.26 [Μ+Η]. Compound was confirmed using LC/MS and moved to next step as it was.

(R)-Ethyl 4-((S)-2-(((2H-tetrazol-5-yl)methyl)carbamoyl)indolin- l -yl)-3-((2S,3S)- 3-methyl-2-(2-phenylacetamido)pentanamido)-4-oxobutanoate was prepared from (R)- fert-butyl 4-((S)-2-(((2H-tetrazol-5-yl)methyl)carbamoyl)indolin-l-yl)- 3-((tert- butoxycarbonyl)amino)-4-oxobutanoate and 1-7 using method F: MS (LC/MS) m/z observed 618.94, expected 619.30 [Μ+Η]. Compound was confirmed using LC/MS and moved to next step as it was.

Title compound C15 was prepared from (R)-ethyl 4-((S)-2-(((2H-tetrazol-5- yl)methyl)carbamoyl)indolin- l -yl)-3-((2S,3S)-3-methyl-2-(2- phenylacetamido)pentanamido)-4-oxobutanoate using method D: ι Η NMR (400 MHz, DMSO-rftf) δ 0.66-0.76 (6H,m), 0.96-1.05 (IH, m), 1.32- 1.37 ( I H, m), 1.60- 1.66 ( I H, m), 2.57-2.63 (2H, m), 3.13-3.17 (I H, t, J=8 Hz), 3.37-3.40 (IH, d, J= 12 Hz), 3.46-3.52 (2H, t, J= 12 Hz), 4.16-4.20 (I H, t, J=8 Hz), 4.39-4.42 (I H, d, J=12 Hz), 4.60-4.65 (2H, m), 5.60-5.63 ( I H, d, J=12 Hz), 7.00-7.04 ( I H, t, J=8 Hz), 7.14-7.23 (6H, m), 8.04-8.08 ( I H, t, J=8 Hz), 8.04-8.06 (I H, d, J=8 Hz), 8.80 (I H, bs), 9.02 (I H, bs), MS (LC/MS) m/z observed 590.95, expected 591 .27 [M+H] and observed 61 3.08, expected 613.25 [M+Na].

EXAMPLE C 16

(4R)-4- [(25,35)-3-METHYL-2-(2-PHENYLACETAMIDO)PENTANAMIDO] -5-OXO-5- [(2S)-2-

[(2H-l,2,3,4-TETRAZOL-5-YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-l H-INDOL-l-

YL] PENTANOIC ACID

(R)-ieri-Butyl 5-((S)-2-(((2H-tetrazol-5-yl)methyl)carbamoyl)indolin-l -yl)-4-

(( er -butoxycarbonyl)amino)-5-oxopentanoate was prepared from 1-2 and (R)-5-(tert- butoxy)-2-((ter/-butoxycarbonyl)amino)-5-oxopentanoic acid using method C: MS (LC/MS) m/z observed 529.86, expected 530.27 [Μ+Η] . Compound was confirmed using LC/MS and moved to next step as it was.

(R)-Ethyl 5-((S)-2-(((2H-tetrazol-5-yl)methy])carbamoyl)indolin-l -yl)-4-((2S,3S)-

3-methyl-2-(2-phenylacetamido)pentanamido)-5-oxopentanoat e was prepared from (R)-teri-butyl 5-((S)-2-(((2H-tetrazol-5-yl)methyl)carbamoyl)indolin- l -yl)-4-((½ri- butoxycarbonyl)amino)-5-oxopentanoate and 1-7 using method F: MS (LC/MS) m/z observed 633.01 , expected 633.32 [Μ+Η]. Compound was confirmed using LC/MS and moved to next step as it was.

Title compound C16 was prepared from (R)-ethyl 5-((S)-2-(((2H-tetrazol-5- yl)methyl)carbamoyl)indolin- l-yl)-4-((2S,3S)-3-methyl-2-(2- phenylacetamido)pentanamido)-5-oxopentanoate using method D: Ή NMR (400 MHz, DMSO-<½) δ 0.75-0.82 (6H,m), 1.00-1.05 ( I H, m), 1 .21 -1.27 (IH, m), 1.73-1.83 (2H, m), 2.27-2.33 (IH, m), 3.1 2-3.20 (I H, t, .7=16 Hz), 3.36-3.61 (4H, m), 4.20-4.24 ( I H, t, J=8 Hz), 4.32-4.36 (IH, t, J=8 Hz), 4.42-4.48 (IH, dd, J=8, 16 Hz), 4.54-4.60 (I H, dd, J=8, 16 Hz), 4.62-4.68 ( IH, dd, J=8, 16 Hz), 5.17-5.20 ( I H, d, J=12 Hz) 6.99-7.03 ( I H, t, =8 Hz), 7.12-7.26 (6H, m), 7.99-8.07(2H, m), 8.80 (I H, bs), 9.02 (I H, bs), MS (LC/MS) m/z observed 604.95, expected 605.28 [M+H] and observed 627.08, expected 627.27 [M+Na]. EXAMPLE C 17

(5 i y)-5-[(25',3 l S)-3-METHYL-2-(2-PHENYLACETAMIDO)PENTANAMIDO]-6-OXO-6- [(25)-2- [(2H-l,2,3,4-TETRAZOL-5-YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-I NDOL-l-

YLjHEXANOIC ACID

(S)-tert- uty\ 6-((S)-2-(((2H-tetrazol-5-yl)methyl)carbamoyI)indolin- l -yl)-5-

((tert-butoxycarbonyl)amino)-6-oxohexanoate was prepared from 1-2 and (S)-6-(tert- butoxy)-2-((½rr-butoxycarbonyl)amino)-6-oxohexanoic acid using method C: MS (LC/MS) m/z observed 543.87, expected 544.29 [Μ+Η]. Compound was confirmed using LC/MS and moved to next step as it was.

(S)-Ethyl 6-((S)-2-(((2 -tetrazol-5-yl)methyl)carbamoyl)indolin- l -yl)-5-((2S,3S)-

3-methyl-2-(2-phenylacetamido)pentanamido)-6-oxohexanoate was prepared from (S)- terf-butyl 6-((S)-2-(((2H-tetrazol-5-yl)methyl)carbamoyl)indolin-l -yl)-5-((tert- butoxycarbonyl)amino)-6-oxohexanoate and 1-7 using method F: MS (LC/MS) m/z observed 647.01 , expected 647.33 [Μ+Η] . Compound was confirmed using LC/MS and moved to next step as it was.

Title compound C17 was prepared from (S)-ethyl 6-((S)-2-(((2H-tetrazol-5- yl)methyl)carbamoyl)indolin- 1 -yl)-5-((2S,3S)-3-methyl-2-(2- phenylacetamido)pentanamido)-6-oxohexanoate using method D: : H NMR (400 MHz, DMSO-rf6) δ 0.68-0.83 (6H, m), 0.95-1.08 ( IH, m), 1.16-1.25 (IH, m), 1.30- 1 .40 (I H, m), 1.45- 1 .80 (4H, m), 2.00-2.25 (2H, m), 3.10-3.55 (4H, m), 4.18 (IH, bs), 4.35 (IH, dd, J= 6Hz, 9Hz), 4.50 (I H, m), 4.65 (IH, dd, J=6Hz, 16Hz), 5.56 ( I H, dd, J=3Hz, 12 Hz), 6.93-7.02 (I H, m), 7.10-7.25 (7H, m), 7.90 (I H, d, J=9 Hz), 8.05 (IH, d, J=8 Hz), 8.52 (I H, d, J=7Hz), 9.10 ( I H, bs), MS (LC/MS) m/z observed 618.98, expected 619.30 [M+H].

EXAMPLE C I 8

(2-f)-l-[(2S)-6-AMlNO-2-[(2-?,35)-3-METHYL-2-(2-

PHENYLACETAMFDO)PENTANAMIDO]HEXANOYL]-N-(2H-l,2,3,4-TETRA ZOL-5- YLMETHYL)-2,3-DIHYDRO-lH-INDOLE-2-CARBOXAMIDE (S)-(5-Allyloxycarbonylamino-l -{(S)-2-[(2H-tetrazol-5-ylmethyl)-carbamoyl]- 2,3-dihydro-indole-l -carbonyl} -pentyl)-carbamic acid iert-butyl ester was prepared from 1-2 and (S)-6-(((allyloxy)carbonyl)amino)-2-((ter/-butoxycarbonyl)am ino)hexanoate dicyclohexylammonium salt using method C: MS (LC/MS) m/z observed 556.86, expected 557.28 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

(S)- l -[6-(l -Allyloxy-vinylamino)-((5)-2-((25,35)-3-tnethyl-2-(2- phenylacetamido)pentanamido)hexanoyl)]-indoline-2-carboxylic acid (2H-tetrazol-5- ylmethyl)-amide was prepared from (S)-(5-allyloxycarbonylamino- l -{(S)-2-[(2H- tetrazol-5-ylmethyl)-carbamoyl]-2,3-dihydro-indole- l -carbonyl} -pentyl)-carbamic acid tert-buXy\ ester and 1-7 using method A: MS (LC MS) m/z observed 688.04, expected 688.36 [Μ+Η] . Compound was confirmed using LC/MS and moved to next step as it was.

To a stirred solution of (S)- l -[6-(l-allyloxy-vinylamino)-((S)-2-((2S,3S)-3- methyl-2-(2-phenylacetamido)pentanamido)hexanoyl)]-indoline- 2-carboxylic acid (2H- tetrazol-5-ylmethyl)-amide (0.145 mmol) and palladium on charcoal ( 10%, 10 mg) in methanol (2 mL), was added triethylsilane (145 mmol, 10 equivalents) under nitrogen. The reaction mixture was stirred at RT for 1 hr then concentrated to give a residue that was purified by reverse phase column chromatography to give the title compound C18. ( 1.5 mg): MS (LC/MS) m/z observed 604.24, expected 604.34 [Μ+Η].

EXAMPLE C I

7 ' £/f7--BlJTYL /V-[(2-?)-l-{2-[(25',3 l S)-3-METHYL-2-(2- PHENYLACETAlVHDO)PENTANAMIDO]ACETYL}-2-[(2H-l,2,3,4-TETRAZOL -5-

YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-6-YL] CARBA MATE

Title compound C19 was prepared from 1-12 and (2H-tetrazol-5-yl)methyl-amine using general method L. ] H NMR (400 MHz, CD 3 OD) δ 0.88-0.98 (6H, m), 1.14- 1.22 (I H, m), 1.30- 1.41 (I H, m), 1 .52 (9H, s), 1.87-2.01 ( l h, m), 3.12-3.21 ( I H, m), 3.59-3.69 (2H, m), 3.84-3.92 (IH, t, J=16 Hz), 4.16-4.24 (I H, t, J=16 Hz), 4.34-4.36 (I H, d, 7=8 Hz), 4.51 -4.53 ( I H, d, J=8 Hz), 4.63-4.72 (2H, m), 5.15-5.17 ( I H, d, J=8 Hz), 7.06- 7.15 (2H, m), 7.18-7.22 (IH, m), 7.20-7.27 (I H, m), 7.29-7.34 (4H, m), 7.55 (I H, bs), 8.21 (IH, bs), 8.91 (IH, bs) MS (LC/MS) m/z observed 648.13, expected 648.32 [M+H] EXAMPLE C20

( ^-ό-Α ΙΝΟ-Ι-ΙΙ-ΙίΖ^^^ -ΜΕΤΗΥί- -ίΙ- PHENYLACETAMIDO)PENTANAMIDO]ACETYL}- V-(2H-l,2,3,4-TETRAZOL-5-YLMETHYL)- 2,3-DIHYDRO-lH-INDOLE-2-CARBOXAMIDE

Title compound C20 was prepared from C19 using general method E. 'El NMR

(400 MHz, CD 3 OD) δ 0.76-0.85 (6H, m), 1.05- 1.10 ( 1H, m), 1.30-1.42 ( 1H, m), 1.72- 1.86 (1 H, m), 3.13-3.17 (1H, d, 7=16 Hz), 3.39-3.47 (2H, m), 3.54-3.71 (3H, m), 4.10- 4.15 (1 H, dd, 7=16, 4 Hz), 4.19-4.26 (1H, m), 4.38-4.41 (1 H, dd, 7=8, 4 Hz), 4.53-4.69 (2H, m), 5.23-5.26 ( 1H, d, 7=12 Hz), 6.96-6.98 (1H, d, 7=8 Hz), 7.21 -7.29 (5H, m), 8.01 - 8.12 (2H, m), 8.27-8.33 (1 H, m), 9.17-9.22 (1 H, m), 10.01 (1H, bs) MS (LC/MS) mJz observed 548.12, expected 548.32 [M+H]

EXAMPLE C21

(2.y)-6-(BENZYLAMINO)-l-{2-[(2-S',3S -3-METHYL-2-(2- PHENYLACETAMIDO)PENTANAMIDO]ACETYL}-N-(2H-l,2,3,4-TETRAZOL-5 -YLMETHYL)- 2,3-DIHYDRO-lH-INDOLE-2-CARBOXAMIDE

A solution of compound C20 (0.025 g, 0.0385 mmol), DIPEA (0.032 ml, 0.23 mmol) and benzyl bromide (0.015 ml, 0.1 16 mol) in DCM (4 ml) was sealed in a microwave tube and heated to 70 °C for 10 hr. by microwave irradiation. The reaction mixture was concentrated to dryness and dried well under vacuum to give the crude product, which was purified on preparative HPLC using 0-80% MeOH in water to yield product as an off-white solid 0.0022 g (9%). MS (LC/MS) m/z observed 638.1 1, expected 638.31 [M+H]. Compound was confirmed using LC/MS and used as directed.

EXAMPLE C22

3-{[(2 1 S -1-{2-[(25',35)-3-METHYL-2-(2-PHENYLACETAMIDO)PENTANAIVIIDO] ACETYL}- 2-[(2H-L,2,3,4-TETRAZOL-5-YLMETHYL)CARBA OYL]-2,3-DIHYDRO-LH-INDOL-6-

YLJCARBAMOYL} PROPANOIC ACID

Title compound C22 was prepared from C19 and succinic anhydride using method B: Ή NMR (400 MHz, DMSO-J6) δ 0.78-0.87 (6H, m), 1 .06-1.13 (1H, m), 1 .30-1 .42 (1 H, m), 1.75-1.86 (1H, m), 3.03-3.07 ( 1H, d, 7=16 Hz), 3.44-3.61 (5H, m), 4.09-4.19 (1 H, m), 4.23-4.27 (1 H, t, 7=8 Hz), 4.40-4.44 (1 H, t, 7=8 Hz), 4.48-4.61 (2H, m), 5.14-5.17 ( 1H, d, 7=12 Hz), 7.09-7.1 1 (1 H, d, 7=8 Hz), 7.18-7.22 (1H, m), 7.25-7.3 1 (4H, m), 7.40-7.44 (I H, t, 7=8 Hz), 8.04-8.06 (IH, d, J=8 Hz), 8.12-8.14 ( I H, t, 7=8 Hz), 8.22 ( I H, bs), 8.28-8.32 (IH, t, 7=8 Hz), 8.95 (I H, bs), 9.96 (I H, s) 12.10 (IH, bs) MS (LC/MS) m/z observed 648.12, expected 648.28 [M+H]

EXAMPLE C23

3-{[(S)-({2-OXO-2-[(2S)-2-[(2H-l,2,3,4-TETRAZOL-5-YLMETHYL)C ARBAMOYL]-2,3- DIHYDRO-lH-INDOL-l-YL] ETHYL}CARBAIVIOYL)(TH10PHEN-3- YL)METHYL] CARBAMOYL} PROPANOIC ACID

(S)-2-((/er/-Butoxycarbonyl)amino)-2-(thiophen-3-yl)acetic acid was prepared from (S -2-amino-2-(thiophen-3-yl)acetic acid using general method K. MS (LC/MS) m z observed 279.92, expected 280.07 [M+Na], Compound was confirmed using LC/MS and moved to next step as it was.

(S)-Ethyl l -(2-((S)-2-((½ -butoxycarbonyl)amino)-2-(thiophen-3- yl)acetamido)acetyl)indoline-2-carboxylate was prepared from 1-10 and (R)-2-((tert- butoxycarbonyl)amino)-2-(thiophen-3-yl)acetic acid using general method A. The purification was performed by column chromatography on silica gel using 0 % to 50 % ethyl acetate in hexanes as the eluent. MS (LC/MS) m/z observed 487.84, expected 488.1 8 [M+H], observed 510.06, expected 510.18 [M+Na]. Compound was confirmed using LC/MS and moved to next step as it was.

tert-Butyl ((S)-2-((2-((5)-2-(((2H-tetrazol-5-yl)methyl)carbamoyl)indol in-l -yl)-2- oxoethyl)amino)-2-oxo- l -(thiophen-3-yl)ethyl)carbamate was prepared from (S)-ethyl 1 - (2-((R)-2-((ieri-butoxycarbonyl)amino)-2-(thiophen-3-yl)acet amido)acetyl)indoline-2- carboxylate and (2H-tetrazol-5-yl)methyl-amine using general method L. MS (LC/MS) m/z observed 541.02, expected 541.19 [Μ+Η] . Compound was confirmed using LC/MS and moved to next step as it was.

Title compound C23 was prepared from ZerZ-butyl ((S)-2-((2-((S)-2-(((2H- tetrazol-5-yl)methyl)carbamoyI)indolin-l -yl)-2-oxoethyl)amino)-2-oxo-l -(thiophen-3- yl)ethyl)carbamate and succinic anhydride using method I: Ή NMR (400 MHz, DMSO- d6) δ 2.39-2.48 (2H, m), 3.12-3.16 (2H, d, 7=16 Hz), 3.58-3.66 (3H, t, 7=16 Hz), 4.20- 4.24 ( I H, d, 7=16 Hz), 4.55-4.59 (IH, m), 4.62-4.69 ( I H, m), 5.15-5.18 (I H, d, 7=1 2 Hz), 5.64-5.68 (I H, t, 7=8 Hz), 7.00-7.04 (IH, t, 7=8 Hz), 7.1 8-7.22 (3H, m), 7.48-7.52 (2H, m), 8.04-8.06 ( 1 H, d, J=8 Hz), 8.53-8.55 (1H, d, J=S Hz), 8.61 -8.64 ( 1 H, m), 9.10 ( 1 H, bs), MS (LC/MS) m/z observed 541.03, expected 541 .15 [M+H]

EXAMPLE C24

3-{[(*)-({2-OXO-2-[(25)-2-[(2H-l,2,3,4-TETRAZOL-5-YLMETHYL)C A BAMOYL]-2,3- DIHYDRO-1 H- IN DOL- 1 -YL] ETHYL} CARBAMOYL)(THIOPHEN-2-

YL)METHYL]CARBAMOYL}PROPANOIC ACID

(R)-2-((½ri-Butoxycarbonyl)amino)-2-(thiophen-2-yl)acetic acid was prepared from (R)-2-amino-2-(thiophen-2-yl)acetic acid using general method K. MS (LC/MS) m/z observed 279.92, expected 280.07 [M+Na] . Compound was confirmed using LC/MS and moved to next step as it was.

(S)-Ethyl l -(2-((R)-2-((ieri-butoxycarbonyl)amino)-2-(thiophen-2- yl)acetamido)acetyl)indoline-2-carboxylate was prepared from 1-10 and (R)-2-((tert- butoxycarbonyl)amino)-2-(thiophen-2-yl)acetic acid using general method A. The purification was performed on normal phase using 0 % to 50 % ethyl acetate in hexanes as the eluent. MS (LC/MS) m/z observed 487.89, expected 488.18 [M+H], observed 51 0.06, expected 510.18 [M+Na], Compound was confirmed using LC/MS and moved to next step as it was.

fert-Butyl ((S)-2-((2-((S)-2-(((2H-tetrazol-5-yl)methyl)carbamoyl)indol in- l -yl)-2- oxoethyl)amino)-2-oxo-l -(thiophen-2-yl)ethyl)carbamate was prepared from (S)-ethyl 1 - (2-((R)-2-((ieri-butoxycarbonyl)amino)-2-(thiophen-2-yl)acet amido)acetyl)indoline-2- carboxylate and (2H-tetrazol-5-yl)methyl-amine using general method L. MS (LC/MS) m/z observed 541.04, expected 541.19 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

Title compound C24 was prepared from fert-butyl ((S)-2-((2-((S)-2-(((2H- tetrazol-5-yl)methyl)carbamoyl)indolin-l -yl)-2-oxoethyl)amino)-2-oxo- l -(thiophen-2- yl)ethyl)carbamate and succinic anhydride using method I: Ή NMR (400 MHz, DMSO- d6) δ 2.41-2.45 (2H, m), 3.10-3.14 (2H, d, J=16 Hz), 3.56-3.64 (3H, t, J=16 Hz), 4.19- 4.23 (1H, d, J=16 Hz), 4.52-4.62 (2H, m), 5.14-5.17 ( 1H, d, J=12 Hz), 5.80-5.84 (1 H, t, J=8 Hz), 6.95-6.98 ( 1 H, m), 7.18-7.22 (3H, m), 6.99-7.01 (lH, d, J=8 Hz), 7.13-7.18 (2H, m), 7.20-7.22 ( 1H, d, J=8 Hz), 7.40-7.42 (1H, d, J=8 Hz), 8.01 -8.03 ( 1 H, d, J=8 Hz), 8.62-8.64 (2H, d, J=8 Hz), 9.04 (I H, bs), MS (LC/MS) m/z observed 541.01 , expected 541.15 [M+H]

EXAMPLE C25

4-Oxo-4-[(25 -2-({2-OXO-2-[(2S)-2-[(2H-l,2,3,4-TETRAZOL-5- YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l- YL]ETHYL}CARBAMOYL)PIPERIDIN-l-YL] BUTANOIC ACID (S)- l -(½r?-Butoxycarbonyl)piperidine-2-carboxylic acid was prepared from (S)- piperidine-2-carboxylic acid using general method K. MS (LC/MS) m/z observed 25 1.99, expected 252.13 [M+Na]. Compound was confirmed using LC/MS and moved to next step as it was.

l -{2-[(l -/ert-Butoxycarbonyl-piperidine-(2S)-2-carbonyl)-amino]-acet yl} -(2S)- 2,3-dihydro-lH-indole-2-carboxylic acid ethyl ester was prepared from 1-10 and (S)-l - (/e^i-butoxycarbonyl)piperidine-2-carboxylic acid using general method A. The purification was performed by column chromatography on silica gel using 0 % to 50 % ethyl acetate in hexanes as the eluent. MS (LC/MS) m/z observed 459.96, expected 460.24 [M+H] observed 482.16, expected 482.24 [M+Na]. Compound was confirmed using LC/MS and moved to next step as it was.

(2S)-2-(2-Oxo-2-{(2S)-2-[(2H-tetrazol-5-ylmethyl)-carbamoyl] -2,3-dihydro- indol- l -yl }-ethylcarbamoyl)-piperidine-l -carboxylic acid /ert-butyl ester was prepared from l - {2-[(l -/e /-butoxycarbonyl-piperidine-(2S)-2-carbonyl)-amino]-acetyl} -(2S)-2,3- dihydro-l H-indole-2-carboxylic acid ethyl ester and (2H-tetrazol-5-yl)methyl-amine using general method L. MS (LC/MS) m/z observed 513.08, expected 513.25 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

Title compound C25 was prepared from (25)-2-(2-oxo-2-{(2S)-2-[(2H-tetrazol-5- ylmethyI)-carbamoyl]-2,3-dihydro-indol- 1 -yl} -ethylcarbamoyl)-piperidine- 1 -carboxylic acid /er/-butyl ester and succinic anhydride using method I: Ή NMR (400 MHz, DMSO-i 6) δ 1.32- 1.40 (2H, m), 1.55- 1.64 (2H, m), 3.56-3.64 (3H, t, J= 16 Hz), 2.19- 2.22 ( I H, d, J= 12 Hz), 2.39-2.47 (2H, m), 2.60-2.64 (2H, t, J=8 Hz), 3.12-3.16 ( I H, d, J=16 Hz), 3.79-3.83 (2H, d, J=16 Hz), 4.18-4.26 (I H, m), 4.32-4.35 (I H, d, J=12 Hz), 4.53-4.56 (2H, m), 5.10 ( I H, s), 5.16-5.19 (IH, d, J= 12 Hz), 6.99-7.03 ( I H, t, J=8 Hz), 7.15-7.19 (1H, t, 7=8 Hz), 7.22-7.24 (1H, d, J=8 Hz), 7.99-8.04 (2H, m), 9.09 ( 1H, bs), MS (LC/MS) m/z observed 512.99, expected 513.21 [M+H]

EXAMPLE C26

(2S)-l-{2-[(2 1 S , ,35)-3-METHYL-2-l(PYRIMIDIIV-2-YL)AMINO] PENTANAlVIIDO]ACETYL}-Af- (2H-l,2,3,4-TETRAZOL-5-YLMETHYL)-2,3-DIHYDRO-lH-INDOLE-2-CAR BOXAMIDE

(2S)-Ethyl l -(2-((2S,3S)-2-((rm-butoxycarbonyl)amino)-3- methylpentanamido)acetyl)indoline-2-carboxylate was prepared from 1-10 and Boc-L- isoleucine using method A. MS (LC/MS) m/z observed 461 .98, expected 462.26 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

(2S)- l -[2-((2 1 S , ,3S)-2-Amino-3-methyl-pentanoylamino)-acetyl]-2,3-dihy dro- l H- indole-2-carboxylic acid ethyl ester hydrochloride was prepared from (2S)-ethyl l -(2- ((2S,3S)-2-((fert-butoxycarbonyl)amino)-3-methylpentanamido) acetyl)indoline-2- carboxylate using method E. MS (LC/MS) m/z observed 362.08, expected 362.20 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

A solution of (2S)-l -[2-((2S,3S)-2-amino-3-methyl-pentanoylamino)-acetyl]-2,3- dihydro-lH-indole-2-carboxylic acid ethyl ester hydrochloride (0.3 g, 0.7537 mmol), DIPEA (0.46 ml, 2.638 mmol) and 2-chloropyrimidine (0.18 g, 1 .131 mol) in ACN (9 ml) was sealed in a microwave tube and heated to 130 °C for 16 hrs by microwave irradiation. The reaction mixture was concentrated to dryness and dried well under vacuum to give the crude product, which was purified by column chromatography on silica gel to get the pure product, (25)-l -{2-[(S)-3-Methyl-(S)-2-(pyrimidin-2-ylamino)-pentanoylamino ]- acetyl}-2,3-dihydro- lH-indole-2-carboxylic acid ethyl ester, as off-white solid, 0.07 g (21 %).. MS (LC/MS) m/z observed 440.10, expected 440.22 [Μ+Η]. Compound was confirmed using LC/MS and moved to next step as is.

Title compound C26 was prepared from (2S)- l -{2-[(S)-3-Methyl-(S)-2-

(pyrimidin-2-ylamino)-pentanoylamino]-acetyl}-2,3-dihydro - l H-indole-2-carboxylic acid ethyl ester and (2H-tetrazol-5-yl)methyl-amine using general method L. Ή NMR (400 MHz, DMSO-rf6) δ 0.82-0.86 (3H, t, 7=8 Hz), 0.92-0.94 (3H, d, 7=8 Hz), 1.16- 1.25 ( 1 H, m), 1 .50- 1.58 (1 H, m), 1.84-1.92 (1H, m), 3.1 1 -3.15 (2H, d, 7= 16 Hz), 3.52-3.60 (2H, m), 4.15-4.19 (1 H, d, 7=16 Hz), 4.37-4.41 ( 1 H, t, 7=8 Hz), 4.50-4.54 ( 1 H, d, 7=16 Hz), 4.62-4.66 (1 H, d, 7= 16 Hz), 5.14-5.17 ( 1H, d, 7=1 6 Hz), 6.60-6.62 (l h, t, 7=4 Hz), 6.92-6.94 ( I H, d, J=8 Hz), 6.98-7.02 ( I H, t, J=8 Hz), 7.14-7.18 ( I H, t, 7=8 Hz), 7.21 - 7.23 ( I H, d, J=8 Hz), 8.00-8.02 (IH, d, J=8 Hz), 8.14(1H, bs), 8.27-8.29(2H, d, J=8 Hz), 9.05 (IH, bs), MS (LC/MS) m/z observed 493.14, expected 493.23 [M+H].

EXAMPLE C27

( ^-ό-ΑθΕΤΑΜΙΟΟ-Ι-ίΖ-ΙίΖ^^^ - ΕΤΙΙΥί- -ίΙ-

PHENYLACETA IDO)PENTANAMIDO]ACETYL}-N-(2H-l,2,3,4-TET AZOL-5-YLMETHYL)- 2,3-DIHYDRO-lH-INDOLE-2-CARBOXAMIDE

Acetic acid (0.3 g, 4.854 mmol), EDC (1.02 g, 5.339 mmol), HOBt (0.74 g, 4.854 mmol), DIPEA (2.5 ml, 14.560 mmol) and I-ll (1 g, 4.845 mmol) were stirred in anhydrous DCM (100 ml) for 16 hrs. The reaction mixture was concentrated under vacuum to give the crude product, which was purified by column chromatography on silica gel using 0 % to 50 % ethyl acetate in hexanes as the eluent to give (S)-ethyl 6- acetamidoindoline-2-carboxylate as a pale yellow sol id (72%). J H NMR (400 MHz, CDC1 3 ) δ 1 .27-1.3 1 (3H, t, J=8 Hz), 2.12 (3H, s), 3.32-3.37 (2H, m), 4.17-4.23 (2H, q, J=8 Hz), 4.35-4.38 (I H, q, J=4 Hz), 4.51 ( I H, s), 6.71 -6.73 (I H, d, J=8 Hz), 6.97-6.99 ( I H, d, .7=8 Hz), 7.05 ( I H, s), 7.47 ( I H, s), MS (LC/MS) m/z observed 249.07, expected 249.12 [M+H].

(S)-Ethyl 6-acetamido- l -(2-((feri-butoxycarbonyl)amino)acetyl)indoline-2- carboxylate was prepared from (S)-ethyl 6-acetamidoindoline-2-carboxylate and Boc- glycine using method C and purified by column chromatography on silica gel using 0 % to 80 % ethyl acetate in hexanes as the eluent to give (S)-ethyl 6-acetamido- l -(2-((/e/Y- butoxycarbonyl)amino)acetyl)indoline-2-carboxylate, as an off-white solid (62%). MS (LC/MS) m/z observed 405.86, expected 406.19[M+H], observed 428.10, expected 428.1 [M+Na], Compound was confirmed using LC/MS and moved to next step as it was.

(S)-Ethyl 6-acetamido-l-(2-((2S,3S)-3-methyl-2-(2- phenylacetamido)pentanamido)acetyl)indoline-2-carboxylate was prepared from the intermediate (S)-ethyl 6-acetamido-l-(2-((ier/-butoxycarbonyl)amino)acetyl)indoline -2- carboxylate and 1-7 using method A. MS (LC/MS) m/z observed 537.03, expected 537.26 [M+H] . Compound was confirmed using LC/MS and moved to next step as it was. Title compound C27 was prepared from (S)-ethyl 6-acetamido-l-(2-((2S,3S)-3- methyl-2-(2-phenylacetamido)pentanamido)acetyl)indoline-2-ca rboxylate and (2H- tetrazol-5-yl)methyl-amine using general method L. 'Η NMR (400 MHz, DMSO-i6) δ 0.78-0.86 (6H, m), 1.06-1.13 (IH, m), 1.29-1.44 (IH, m), 1.75-1.86 (IH, m),2.01 (3H, s), 3.05-3.09 (2H, d, .7=16 Hz), 3.43-3.61 (4H, m), 4.09-4.13 (IH, d, J=16 Hz), 4.19-4.28 (IH, m), 4.38-4.44 (IH, m), 4.51-4.59 (2H, m), 5.15-5.18 (IH, d, J=8 Hz), 7.09-7.11 (IH, d, J=8 Hz), 7.18-7.22 (IH, m), 7.27-7.31 (3H, m), 7.40-7.44 (IH, t, J=8 Hz), 8.02-8.04 (IH, d, J≡i Hz), 8.15-8.23 (IH, m), 8.26-8.31 (IH, m), 9.04 (IH, bs), 9.93 (IH, bs), MS (LC/MS) m/z observed 590.15, expected 590.28 [M+H], m/z observed 612.28, expected 612.28 [M+Na].

EXAMPLE C28

4-Oxo-4-[(2S -2-({2-oxo-2-[(2S)-2-[(2H-l,2,3,4-TETRAZOL-5-

YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-l- YL]ETHYL}CARBAMOYL)PYRROLIDIN-l-YL]BUTANOIC CID l-{2-[(l-iei-i-Butoxycarbonyl-pyrrolidine-(2S)-2-carbonyl)-a mino]-acetyl}-(2S)- 2,3-dihydro-lH-indole-2-carboxylic acid ethyl ester was prepared from 1-10 and Boc-L- proline using method A. MS (LC/MS) m/z observed 445.94, expected 446.23 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

l-{2-[(l-terf-Butoxycarbonyl-pyrrolidine-(2S)-2-carbonyl)-am ino]-acetyl}-(2S)- 2,3-dihydro-lH-indole-2-carboxylic acid was prepared from l-{2-[(l-½rf- butoxycarbonyl-pyrrolidine-(2S)-2-carbonyl)-amino]-acetyl}-( 2S)-2,3-dihydro-lH- indole-2-carboxylic acid ethyl ester using method D with 2 eq. of L1OHH2O. MS (LC/MS) m/z observed 417.90, expected 418.20 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

(S)-tert-Butyl 2-((2-((S)-2-(((2H-tetrazol-5-yl)methyl)carbamoyl)indolin-l- yl)-2- oxoethyl)carbamoyl)pyrrolidine-l-carboxylate was prepared from l-{2-[(l-½r/- butoxycarbonyl-pyrrolidine-(2S)-2-carbonyl)-amino]-acetyl}-( 25)-2,3-dihydro-lH- indole-2-carboxylic acid and 2H-tetrazol-5-yl)methyl-amine using method A in DMF but without HC1 treatment. MS (LC/MS) m/z observed 498.94, expected 499.24 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was. Title compound C28 was prepared from (S)-tert-baly\ 2-((2-((S)-2-(((2H-tetrazoI- 5-yl)methyl)carbamoyl)indolin-l-yl)-2-oxoethyl)carbamoyl)pyr rolidine-l-carbox late and succinic anhydride using method I. 'H NMR (400 MHz, DMSO-i6) δ 1.86-1.91 (2H, m), 1.95-2.05 (2H, m), 2.40-2.65 (4H, m), 3.11 (IH, m), 3.36 (IH, m), 3.46 (IH, m), 3.63 (IH, m), 4.15 (IH, dd, J=6, 17 Hz), 4.32 (IH, d, J=7Hz), 4.48 (IH, dd, ,7=3, 9Hz), 4.56 (IH, m), 4.65 (IH, m), 5.17 (IH, d, ^=11 Hz), 7.01 (IH, t, =8Hz), 7.12-7-25 (2H, m), 8.00-8.10 (2H, m), 9.08 (IH, bs), MS (LC/MS) m/z observed 499.11, expected 499.21 [M+H]

EXAMPLE C29

3-{[l-({2-OXO-2-[(2S)-2-[(2H-l,2,3,4-TETRAZOL-5-YLMETHYL)CAR BAMOYL]-2,3-

DIHYDRO- lH-INDOL-1 - YL]ETHYL}CARBAM0YL)CYCL0PENTYL]CARBAM0YL} PROPANOIC ACID l-{2-[(l-/eri-Butoxycarbonylamino-cyclopentanecarbonyl)-amin o]-acetyl}-(2S)- 2,3-dihydro-lH-indole-2-carboxylic acid ethyl ester was prepared from 1-10 and Boc-I- cycloleucine using method A. MS (LC/MS) m/z observed 459.95, expected 460.24 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

l-{2-[(l-tert-Butoxycarbonylamino-cyclopentanecarbonyl)-amin o]-acetyl}-(2S)- 2,3-dihydro-lH-indole-2-carboxylic acid was prepared from l-{2-[(l-teri- Butoxycarbonylamino-cyclopentanecarbonyl)-amino]-acetyl}-(2S )-2,3-dihydro-l//- indole-2-carboxylic acid ethyl ester using method D with 2 eq. of L1OHH2O. MS (LC/MS) m/z observed 431.93, expected 432.21 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

(2S)-[l-(2-Oxo-2-{2-[(2H-tetrazol-5-ylmethyl)-carbamoyl]-2,3 -dihydro-indol-l- yl}-ethylcarbamoyl)-cyclopentyl]-carbamic acid /e -butyl ester was prepared from l-{2- [(l-½r/-butoxycarbonylamino-cyclopentanecarbonyl)-amino]-ac etyl}-(2S)-2,3-dihydro- lH-indole-2-carboxylic acid and (2H-tetrazol-5-yl)methyl-amine using method A in DMF but without HCl treatment. MS (LC/MS) m/z observed 512.90, expected 513.26 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

Title compound C29 was prepared from (2S)-[l-(2-Oxo-2-{2-[(2H-tetrazol-5- ylmethyl)-carbamoyl]-2,3-dihydro-indol-l-yl}-ethylcarbamoyl) -cyclopentyl]-carbamic acid fert-butyl ester and succinic anhydride using method I. Ή NMR (400 MHz, DMSO-i 6) δ 1.55- 1.67 (4H, m), 1 .83-1.93 (2H, m), 1.98-2.14 (2H, m), 2.34-2.39 (2H, m), 2.41 -2.46 (2H, m), 3.1 1 (1 H, m), 3.50-3.63 (2H, m), 4.05 (1 H, m), 4.52 (1H, dd, J=7, 16Hz), 4.68 (1H, dd, J=7, 16Hz), 5.17 (1H, d, J= l lHz), 7.01 ( 1 H, t, J=8Hz), 7.13-7-25 (2H, m), 7.65 (1 H, t, J=5Hz), 8.02 (1H, d, J=8Hz), 8.12 (1 H, bs), 9.10 (1H, bs), MS (LC/MS) m/z observed 513.06, expected 513.22 [M+H]

EXAMPLE C30

(2S)-7-ACETAMlDO- 1 - {2-[(25",3S)-3-METHYL-2-(2- PHENYLACETAMIDO)PENTANAMIDO]ACETYL}-7V-(2H-l,2,3,4-TETRAZOL- 5-YLMETHYL)- 2,3-DIHYDRO-lH-INDOLE-2-CARBOXAMIDE

Intermediate 1-14 (500 mg, 2.427 mmol) and palladium on charcoal ( 100 mg) were placed in a round bottom flask under nitrogen. Ethanol (10 mL) was then added and hydrogen was bubbled in the reaction mixture for 4 hrs. Then the reaction mixture was filtered over CELITE ® and the CELITE ® was washed with ethanol (3 x 1 5 mL). The filtrate and washings were concentrated to give a brown oil that was further dissolved in DCM (50 mL) and to this was added acetic acid (140 uL, 2.427 mmol) , HOBt (372 mg, 2.427 mmol), EDC (5 12 mg, 2.670 mmol) and DIPEA (845 uL, 4.854 mmol). The reaction was left at RT for 16 hrs and the solvent was evaporated. The product was purified by normal phase column chromatography using 25 % to 55 % ethyl acetate in hexanes as the eluent to give (S)-ethyl 7-acetamidoindoline-2-carboxylate as an off white solid ( 103 mg, 17 %). Ή NMR (400 MHz, CDC1 3 ) δ 1.32 (3H, t, J=7.0Hz), 2.19 (3H, s), 3.3 1 (1 H, dd, J=8, 16Hz), 3.45 (1 H, dd, J= l l , 16Hz), 4.19-4.30 (2H, qd, J=2, 7Hz), 4.51 (1 H, dd, J=7, 10Hz), 5.20 ( 1 H, bs), 6.70 ( 1H, t, J=8Hz), 6.85 (1H, d, J=8Hz), 6.96 ( 1 H, d, J= 8Hz), 7.40 (1 H, bs), MS (LC/MS) m/z observed 249.07, expected 249.12 [M+H].

(S)-Ethyl 7-acetamido-l -(2-((ier/-butoxycarbonyl)amino)acetyl)indoline-2- carboxylate was prepared from (S)-ethyl 7-acetamidoindoline-2-carboxylate and Boc- glycine using method C but purified by normal phase column chromatography using 25 % to 55 % ethyl acetate in hexanes as the eluent. MS (LC/MS) m/z observed 406.09, expected 406.20 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

(S)-Ethyl 7-acetamido-l -(2-((2S,3S)-3-methyl-2-(2- phenylacetamido)pentanamido)acetyl)indoline-2-carboxylate was prepared from (^-ethyl 7-acetamido- l -(2-((;m-butoxycarbonyl)amino)acetyl)indoline-2-carboxylate and 1-7 using method A but the product was purified by normal phase column chromatography using 20 % to 80 % ethyl acetate in hexanes as the eluent. MS (LC/MS) m/z observed 537.08, expected 537.27 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

The intermediate ((S)-7-Acetamido-l -(2-((25,3S)-3-methyl-2-(2- phenylacetamido)pentanamido)acetyl)indoline-2-carboxylic acid) was prepared from (S)- ethyl 7-acetamido-l -(2-((2S,3S)-3-methyl-2-(2- phenylacetam ido)pentanamido)acetyl)indol ine-2-carboxylate using method D with 2 eq. of LiOH H 2 0. MS (LC/MS) m/z observed 509.08, expected 509.24 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

Title compound C30 was prepared from (S)-7-acetamido-l -(2-((2S,3S)-3-methyl- 2-(2-phenylacetamido)pentanamido)acetyl)indoline-2-carboxyli c acid and (2H-tetrazol-5- yl)methyl-amine using method A in DMF but without HC1 treatment. Ή NMR (400 MHz, CD 3 OD) δ 0.88-0.92 (3H, t, J= 7Hz), 0.99 (3H, d, J=7Hz), 1.15 (I H, m), 1.45 (I H, m), 1.92 ( I H, m), 2.07 (3H, s), 3.25 (IH, d, J=\ 6Rz), 3.53-3.70 (3H, m), 4.05 ( I H, m), 4.21 -4.35 (2H, m), 4.5 1 ( I H, dd, J=6, 12Hz), 4.80 (IH, m), 5.28 ( IH, t, J=9 Hz), 7.12-7.18 (3H, m), 7.23 (IH, m), 7.26-7.34 (4H, m), MS (LC MS) m/z observed 590.17, expected 590.28 [M+H].

EXAMPLE C31

7-^7'-BUTYL /V-[(25}-1-{2-[(2 1 S,35 , )-3-METHYL-2-(2-

PHENYLACETAMIDO)PENTANAMIDO]ACETYL}-2-[(2H-l,2,3,4-TETRAZ OL-5- YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-5-YL]CARBAMATE

Intermediate 1-13 (1 g, 4.850 mmol) and palladium on charcoal (200 mg) were placed in a round bottom flask under nitrogen. Ethanol (20 mL) was then added and hydrogen was bubbled in the reaction mixture for 4 hrs. Then the reaction mixture was filtered over CELITE ® and the CELITE® was washed with ethanol (3 x 25 mL). The filtrate and washings were concentrated to give a brown oil. This brown oil was the dissolved in toluene (50 mL) and phtalic anhydride (718 mg, 4.85 mmol) was added. The reaction was heated to 80 °C for 7 hrs and the solvent was evaporated. The product was purified by normal phase column chromatography using 5 % to 20 % ethyl acetate in hexanes as the eluent to give (S)-Ethyl 5-(l ,3-dioxoisoindolin-2-yl)indoline-2-carboxylate as an off white solid (750 mg, 46 %). l U NMR (400 MHz, CDC1 3 ) δ 1.3 1 (3H, t, J=7.0Hz), 3.38 ( I H, dd, 7=6, 17Hz), 3.45 (I H, dd, .7=10, 17Hz), 4.19-4.27 (2H, qd, 7=2, 7Hz), 4.43 ( I H, dd, 7=7, 10Hz), 4.58 (IH, bs), 6.78 (IH, d, 7=8Hz), 7.05 ( I H, dd, 7=2, 8Hz), 7.08 ( I H, bs), 7.76-7.78 (2H, m), 7.91 -7.95 (2H, m), MS (LC/MS) m/z observed 337. 10, expected 337.12 [M+H],

(S)-Ethyl l -(2-((½ /-butoxycarbonyl)amino)acetyl)-5-( l ,3-dioxoisoindolin-2- yl)indol ine-2-carboxylate was prepared from (S^-ethyl 5-( l ,3-dioxoisoindol in-2- yl)indol ine-2-carboxylate and Boc-glycine using method C. MS (LC/MS) m/z observed 493.87, expected 494.19[M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

(S)-Ethyl 5-(l ,3-dioxoisoindolin-2-yl)-l -(2-((2S,3S)-3-methyl-2-(2- phenylacetamido)pentanamido)acetyl)indoline-2-carboxylate was prepared from (S)-ethyl l-(2-((ieri-butoxycarbonyl)amino)acetyl)-5-(l ,3-dioxoisoindolin-2-yl)indoline-2- carboxylate and 1-7 using method A but the product was purified by filtration of the reaction mixture. The solid obtained was the pure compound. MS (LC/MS) m/z observed 625.07, expected 625.27 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

(5)-Ethyl 5-(l ,3-dioxoisoindolin-2-yl)-l -(2-((2S,3S)-3-methyl phenylacetamido)pentanamido)acetyl)indoline-2-carboxylate (420 mg, 0.672 mmol) was dissolved in a 2. mixture DMF/ EtOH (90 mL) and hydrazine hydrate 60% solution in water was added (75uL 1.01 mmol). The reaction was heated to 40 °C for 1 hr and then to 80 °C for 5 hrs. LC/MS showed that the starting material was converted to the desired free amine. The solvents were evaporated and the residue was dissolved in EtOH (25 mL) and lithium hydroxide monohydrate ( 141 mg, 3.360 mmol) dissolved in water (25 mL) was added. The reaction was left at RT for 4 hrs and then acidified to pH 4 with a saturated solution of citric acid. The mixture was concentrated to dryness and the residue was suspended in water (25 mL) and the solid in suspension was filtered and washed with water (3 x 10 mL). (S)-5-amino- l-(2-((2S,3S)-3-methyl-2-(2- phenylacetamido)pentanamido)acetyl)indoline-2-carboxylic acid hydrochloride was obtained as a brown solid (325 mg, 96 %). MS (LC/MS) m/z observed 466.99, expected 467.23 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

(S)-5-Amino- 1 -(2-((2S,3S)-3-methyl-2-(2- phenylacetamido)pentanamido)acetyl)indoline-2-carboxylic acid hydrochloride (325 mg, 0.646 mmol) was dissolved in dioxane (20 mL) and NaOH (1M, 2 mL, 1.938 mmol) was added, followed by a solution of B0C 2 O (1.41 g, 6.461 mmol) in dioxane (5 mL). The reaction was left at RT for 16 hrs and then acidified to pH 4 with citric acid (aqueous, saturated solution). The mixture was concentrated and the product was purified on a CI 8 column using 10-55% MeOH in water to yield (S)-5-((tert-butoxycarbonyl)amino)-l-(2- ((2S,3S)-3-methyl-2-(2-phenylacetamido)pentanamido)acetyl)in doline-2-carboxylic acid as an off-white solid (255 mg, 70%). MS (LC/MS) m/z observed 567.10, expected 567.28 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

Title compound C31 was prepared from (S)-5-((fe /-butoxycarbonyl)amino)-l-(2- ((2S,3S)-3-methyl-2-(2-phenylacetamido)pentanamido)acetyl)in doline-2-carboxylic acid and (2H-tetrazol-5-yl)methyl-amine using method A in DMF but without HC1 treatment. Ή NMR (400 MHz, DMSO-<½) δ 0.65-0.87 (6H, m), 1.10 (IH, m), 1.43 (IH, m), 1.75 (IH, m), 3.00 (IH, d, J=16Hz), 3.42-3.63 (4H, m), 4.10 (IH, m), 4.25 (IH, m), 4.45-4.67 (2H, m), 5.05 (IH, d, J=\ 1 Hz), 6.35 (IH, d, J=8 Hz), 6.44 (IH, m), 7.23 (IH, m), 7.26- 7.32 (4H, m), 7.72 (IH, d, J=9 Hz), 8.00-8.25 (2H, m), 8.95 (IH, bs), MS (LC/MS) m/z observed 648.10, expected 648.33 [M+H].

EXAMPLE C32

(2_?)-5-AMINO-l-{2-[(2_?,35 -3-METHYL-2-(2- PHENYLACETAMIDO)PENTANAMIDO]ACETYL}-N-(2H-l,2,3,4-TETRAZOL-5 -YLIVIETHYL)- 2,3-DIHYDRO-l//-INDOLE-2-CARBOXAMIDE HYDROCHLORIDE

Title compound C32 was prepared from C31 using method E. l U NMR (400 MHz, DMSO-i/<i) δ 0.75-0.87 (6H, m), 1.08 (IH, m), 1.40-1.50 (1 OH, m), 1.74 (IH, m), 3.10 (IH, d, J=16Hz), 3.42-3.63 (4H, m), 4.10 (IH, m), 4.25 (IH, t, .7=11 Hz), 4.40- 4.67 (2H, m), 5.13 (IH, d, J=ll Hz), 7.14-7.21 (2H, m), 7.24-7.30 (4H, m), 7.36 (IH, m), 7.88 (IH, d, J=9 Hz), 8.11 (IH, d, J=9Hz), 8.24 (IH, m), 9.00 (IH, bs), 9.27 (IH, bs), MS (LC/MS) m/z observed 548.05, expected 548.27 [M+H]. EXAMPLE C33

(2-?)-5-(7'£:«r-BuTYLAiviiNO)-l-{2-[(25',3S)-3-METHYL-2-(2 -

PHENYLACETAMIDO)PENTANAM[DO]ACETYL}-A / -(2H-l,2,3,4-TETRAZOL-5-YLMETHYL)- 2,3-DIHYDRO-lH-INDOLE-2-CARBOXAMlDE

Title compound C32 (25 mg, 0.0428 mmol) was dissolved in DMF (5mL) in a microwaved vial. Triethylamine (2 niL) and 2-bromo-2-methyl bromide (500uL) were then added. The reaction was then microwaved at 100 °C for 15 minutes. Additional triethylamine (3 mL) and 2-bromo-2-methyl bromide ( l mL) were added and the reaction was microwaved for an additional 50 minutes at 100 °C. Additional triethylamine (2mL) 2-bromo-2-methyl bromide ( 1 mL) were added and the reaction was microwaved for an additional 30 minutes at 100 °C. The reaction mixture was then filtered to remove salts and the filtrate was concentrated. The product was purified on a C I 8 column using 10-55% MeOH in water to yield title compound C33 as a brown solid ( 1 .2 mg, 5%). MS (LC/MS) m/z observed 604.20, expected 604.34 [M+H].

EXAMPLE C34

(2S)-7V ^V-TRIMETHYL-l-{2-[(25',3S)-3-METHYL-2-(2- PHENYLACETAMIDO)PENTANAMIDO]ACETYL}-2-[(2H-l,2,3,4-TETRAZOL- 5- YLMETHYL)CARBAMOYL]-2,3-DIHYDRO-lH-INDOL-5-AMINIUM IODIDE C32 (25 mg, 0.0428 mmol) was dissolved in DMF (8mL) in a microwavable vial. Triethylamine (500 uL) and methyl iodide (150uL) were then added. The reaction was then microwaved at 75 °C for 35 minutes. The reaction mixture was then concentrated and the product was purified on a C I 8 column using 10-55% MeOH in water to yield title compound C34 as an off-white solid (4 mg, 13%). Ή NMR (400 MHz, DMSO-< <5) δ 0.75-0.87 (6H, m), 1 .08 (1 H, m), 1.33 ( 1H, m), 1 .74 ( 1 H, m), 2.80-2.86 (9H, m), 3.03 ( 1 H, d, J=16Hz), 3.42-3.62 (4H, m), 4.1 0 ( 1 H, m), 4.40 ( 1 H, m), 4.54 ( 1 H, m), 4.63 ( 1 H, m), 5.1 0 ( 1 H, m), 6.54 ( 1 H, m), 6.64 ( 1 H, bs), 7.20 ( 1 H, m), 7.25-7.32 (4H, m), 7.86 ( 1 H, m), 8.03 (1 H, m), 8.09-8.1 8 (2H, m), 9.07 (1 H, bs), MS (LC/MS) m/z observed 590.1 1 , expected 590.32[M]. EXAMPLE C35

(2S)-5-(BENZYLAMINO)-l-{2-[(25 , ,3.S)-3-METHYL-2-(2- PHENYLACETAMIDO)PENTANAMIDO]ACETYL}-yV-(2H-l,2,3,4-TETRAZOL- 5-YLMETHYL)- 2,3-DIHYDRO-lH-INDOLE-2-CARBOXAMIDE

C32 (36 mg, 0.0616 mmol) was dissolved in DMF (5mL) in a microwaved vial .

Triethylamine ( 1 mL) and benzyl bromide (8 uL, 0.0678 mmol) were then added. The reaction was then microwaved at 50 °C for 35 minutes. Additional benzyl bromide (15 uL, 0.126 mmol) was added and the reaction was then microwaved at 50 °C for 30 additional minutes. The reaclion mixture was then concentrated and the product was purified on a C I 8 column using 10-55% MeOH in water to yield title compound C35 as an off-white solid (5 mg, 13%). Ή NMR (400 MHz, DMSO-</6) δ 0.73-0.85 (6H, m), 1 .08 (IH, m), 1 .44 (IH, m), 1.75 (I H, m), 2.90 (IH, d, J=16Hz), 3.42-3.62 (6H, m), 4.08 (I H, m), 4.41 (IH, m), 4.56 ( IH, m), 4.68 (IH, m), 5.05 (I H, d, .7= 1 l Hz), 6.35 (IH, m), 6.45 (IH, m), 7.19 (I H, m), 7.21 -7.32 (6H, m), 7.33-7.39 (3H, m), 7.78 (IH, m), 8.03 (I H, m), 8.09-8.18 (2H, m), 9.10 (I H, bs), MS (LC/MS) m/z observed 637.99, expected 638.32 [M+H].

EXAMPLE C36

(25)-1-{2-[(25 , ,35 -2-[(DIMETHOXY-1,3,5-TRIAZIN-2-YL)AMINO]-3-

METHYLPENTANAMrDO]ACETYL}-7V-(2H-l,2,3,4-TETRAZOL-5-YLMET HYL)-2,3- DIHYDRO- 1H-INDOLE-2-CARBOXAMIDE

A solution of (2S)- l -[2-((2S,3S)-2-amino-3-methyl-pentanoylamino)-acetyl]-2,3- dihydro-lH-indole-2-carboxylic acid ethyl ester hydrochloride (0.1 gm, 0.1941 mmol) that was prepared as in Example C26, D1PEA (0.085 ml, 0.291 mmol) and 2-chloro-4,6- dimethoxy- l ,3,5-triazine (0.05 1 gm, 0.291 mmol) in ACN (3 ml) was sealed in a microwave tube and heated to 100 °C for 10 min by microwave irradiation. The reaction mixture was concentrated to dryness and dried well under vacuum to give the crude product, which was purified by column chromatography on silica gel to get the pure product (S)-l -(2-((2S,3S)-2-((4,6-dimethoxy-l ,3,5-triazin-2-yl)amino)-3- methylpentanamido)acetyl)indoline-2-carboxylic acid ethyl ester as an off-white solid, 0.1 gm (80%). MS (LC/MS) m/z observed 501.15, expected 501.24 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was. Title compound C36 was prepared from (S)- l -(2-((2S,3S)-2-((4,6-dimethoxy- l ,3,5-triazin-2-yl)amino)-3-methylpentanamido)acetyl)indoline -2-carboxylic acid ethyl ester and (2H-tetrazol-5-yl)methyl-amine using general method L. l H NMR (400 MHz, DMSO-i 6) δ 0.82-0.86 (3H, t, 7=8 Hz), 0.90-0.92 (3H, d, 7=8 Hz), 1.16- 1.24 ( 1 H, m), 1.46-1 .53 (1 H, m), 1.80-1 .87 (1 H, m), 3.1 1 -3.15 (2H, d, 7=16 Hz), 3.52-3.60 (2H, m), 3.84 (6H, s), 4.16-4.20 (1H, t, 7=8 Hz), 4.35-4.42 ( 1 H, m), 4.56-4.62 ( 1H, d, 7=16 Hz), 5.16 (1H, s), 5.14-5.17 ( 1 H, d, 7=16 Hz), 6.60-6.62 (l h, t, 7=4 Hz), 6.99-7.03 ( 1 H, t, 7=8 Hz), 7.17-7.23 (2H, m), 7.81 -7.85 (1H, t, 7=8 Hz), 8.02 (1H, s), 8.20 (1 H, bs), 8.28 ( 1 H, bs), 9.06 (1 H, bs) MS (LC/MS) m/z observed 554.18, expected 554.25 [M+H].

EXAMPLE C37

(2 i y)-l-{2-[(2iS)-2-[(DlMETHOXY-l,3,5-TRIAZIN-2-YL)AMINO] -3- METHYLBllTANAIVIIDO]ACETYL}-7V-(2H-l,2,3,4-TETRAZOL-5-YLlVIE THYL)-2,3- DIHYDRO-1H-INDOLE-2-CARBOX AMIDE

A solution of 2-amino-3-methylbutanoate hydrochloride (0.5 g, 2.8 mmol), DIPEA (1.16 ml, 6.7 mmol) and 2-chloro-4,6-dimethoxy- l ,3,5-triazine (0.4 g, 1 .9 mmol) in ACN ( 10 ml) was sealed in a microwave tube and heated to 100 °C for 10 min by microwave irradiation. The reaction mixture was concentrated to dryness and dried well under vacuum to give the crude product, which was purified by column chromatography on silica gel to get the pure product (S)-/er/-butyl 2-((4,6-dimethoxy- l ,3,5-triazin-2-yl)amino)-3-methylbutanoate as an off-white solid, 0.45 g (76%). Ή NMR (400 MHz, CDC1 3 ) δ 0.96- 1.00 (6H, t, , 7=8 Hz), 1.45 (9H, s), 2.19-2.27 (1 H, m), 3.94 (3H, s), 3.95 (3H, s), 4.58-4.61 (1H, q, 7=4H), 5.76-5.78 (1 H, d, 7=8 Hz). MS (LC/MS) m/z observed 313.06, expected 313.18 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

(S)-lert-Buty\ 2-((4,6-dimethoxy-l ,3,5-tnazin-2-yl)amino)-3-methylbutanoate

(0.1 g, 0.32 mmol) was stirred in 1 : 1 mixture of DCM:TFA (4 ml) for 2 hrs at RT. The reaction mixture was concentrated to dryness under vacuum and swapped with DCM (4 ml) three times. Resulting residue was dried well under vacuum and subjected to next reaction as it is. The residue obtained above, 1-4 (0.1 1 g, 0.32 mmol), EDC (0.092 g, 0.48 mmol), HOBt (0.064 g, 0.42 mmol) and DIPEA (0.22 ml, 1 .28 mmol) were stirred in anhydrous DCM (5 ml) for 16 hrs. The reaction mixture was concentrated under vacuum to give the crude product which was purified on a CI 8 column using 10-50% MeOH in water to yield the title compound C37 as an off-white solid (45%). Ή NMR (400 MHz, DMSO-rf6) δ 0.92-0.98 (6H, m), 2.08-2.06 (1H, m), 1.80-1.87 (1H, m), 2.98-3.11 (2H, m), 3.52-3.58 (2H, m), 3.83 (6H, s), 4.13-4.17 (1H, t, J=8 Hz), 4.32-4.40 (2H, m), 4.47- 4.52 (1H, m), 5.13-5.16 (1H, d, J=\2 Hz), 6.98-7.02 (1H, t, J=8 Hz), 7.14-7.22 (2H, m), 7.77-7.81 (1H, t, J=8 Hz), 8.03-8.05 (1H, d, J=8 Hz), 8.23 (1H, bs), 8.34 (1H, bs), 8.72 (1H, bs) MS (LC/MS) m/z observed 540.17, expected 540.24 [M+H].

EXAMPLE C38

3-{[(5)-({2-OXO-2-[(2S)-2-[(2H-l,2,3,4-l lRAZOL-5-YLIV 1 t;rHYL)CARBAlVIOYL]-2,3- DIHYDRO-lH-INDOL-1-

YL]ETHYL}CARBAMOYL)(PHENYL) ETHYL]CARBAMOYL} PROPANOIC ACID

(2S)-2-{[(iert-Butoxy)carbonyl]amino}-2-phenylacetic acid (3.0 g, 12.1 mmol, 92%) was collected as a colorless oil from (5)-phenylglycine (2.0 g, 13.2 mmol) using general method . Ή NMR (400 MHz, CDCh) δ 7.98 (1H, bs), 7.44 (2H, d, ,/=7Hz), 7.34 (3H, m), 5.14 (1H, d, =7Hz), 1.23 (9H, s), MS (LC/MS) m/z observed 273.96, expected 274.10 [M+Na]

(2S)-Ethyl l-{2-[(2S)-2-{[(rm-butoxy)carbonyl]amino}-2- phenylacetamido]acetyl}-2,3-dihydro-lH-indole-2-carboxylate (131 mg, 0.27 mmol, 96%) was collected as an off white solid from the coupling of 1-10 (80 mg, 0.28 mmol) with (25)-2-{[(/er/-butoxy)carbonyl]amino}-2-phenylacetic acid (71 mg 0.28 mmol) using general method M. MS (LC/MS) m/z observed 481.88, expected 482.23 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

{IS)- 1 - {2-[(2S)-2- { [(ierf-Butoxy)carbonyl]amino} -2-pheny lacetamidojacety 1 } - 2,3-dihydro-lH-indole-2-carboxylic acid (130 mg, 0.28 mmol, 98%) was collected as an off white solid from (2S)-ethyl l-{2-[(2S)-2-{[(/eri-butoxy)carbonyl]amino}-2- phenylacetamido]acetyl}-2,3-dihydro-lH-indole-2-carboxylate (137 mg, 0.285 mmol) using general method D. In this example, the reaction was stopped after 30 min and a gradient of 10-70%o MeOH in H 2 0 was used during purification. MS (LC/MS) m/z observed 453.89, expected 454.20 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

/er/-Butyl N-[(S)-({2-oxo-2-[(2S)-2-[(2H-l,2,3,4-tetrazol-5-ylmethyl)ca rbamoyl]- 2,3-dihydro- lH-indol- l -yl]ethyl}carbamoyl)(phenyl)methyl]carbamate (96 mg, 0.18 mmol, 63%) was collected as an off white solid from (2S)-l - {2-[(2S)-2-{ [(tert- butoxy)carbonyl]amino}-2-phenylacetamido]acetyl}-2,3-dihydro - lH-indole-2-carboxyl ic acid (130 mg, 0.28 mmol) using general method O. MS (LC/MS) m/z observed 534.91 , expected 535.24 [Μ+Η], Compound was confirmed using LC/MS and moved to next step as it was.

Title compound C38 (41 mg, 0.08 mmol, 43%) was collected as an off white sol id from tert-butyl N-[(S)-({2-oxo-2-[(2S)-2-[(2H- l ,2,3,4-tetrazol-5-ylmethyl)carbamoyl]- 2,3-dihydro-lH-indol-l -yl]ethyl}carbamoyl)(phenyl)methyl]carbamate (96 mg, 0.18 mmol) using general method I. Ή NMR (400 MHz, DMSO-</6) δ 9.08 (I H, bs), 8.65 (I H, bs), 8.56 (I H, d, J=8Hz), 8.05 (I H, d, J=8Hz), 7.47 (2Η, d, J=7Hz), 7.34 (2H, t, =7Hz), 7.28 ( IH, d, J=7Hz), 7.26-7.15 (2H, m), 7.01 (IH, d, J=7Hz), 5.60 (IH, d, J=6Hz), 5.17 ( IH, d, J=10Hz), 4.65 (I H, dd, J=16, 5Hz), 4.53 (I H, dd, J=16, 4Hz), 4.22 (I H, dd, J=16, 5Hz), 3.68-3.50 (2H, m), 3.14 ( I H, d, J=16Hz), 2.48-2.39 (4H, m), MS (LC/MS) m/z observed 534.97, expected 535.20 [M+H],

EXAMPLE C39

(25)-l-{2-[(2S,3S)-3-METHYL-2-[2-(2H-l,2,3,4-TETRAZOL-5- YL)ACETAMIDO]PENTANAMIDO]ACETYL}-N-(lH-l,2,3-TRIAZOL-4-YLMET HYL)-2,3- D1HYDRO-1H-INDOLE-2-CARBOXAMIDE

(2S)- 1 -[2-((2S,3S)-2-½; -Butoxycarbonylamino-3-methyl-pentanoylamino)-acetyl]-2,3- dihydro-lH-indole-2-carboxylic acid ethyl ester was prepared from 1-10 and Boc-L- isoleucine using method A. MS (LCMS) m/z observed 461.98, expected 462.26 [Μ+Η]. Compound was confirmed using LC/MS and moved to next step as it was.

(2S)-l-[2-((2S,3S)-2-ferf-Butoxycarbonylamino-3-methyl-penta noylaminu)- acetyl]-2,3-dihydro-lH-indole-2-carboxylic acid was prepared from (2S)-l-[2-((2S,3S)-2-er/-butoxycarbonylamino-3-methyl-pentan oylamino)-acetyl]-2,3-dihydro-lH-indole-2- carboxylic acid ethyl ester using method D with 2 eq. of L1OHH 2 O. MS (LC/MS) m/z observed 433.96, expected 434.23 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

N[(lS,2S)-2-Methyl-l-({2-oxo-2-[(2S)-2-[(l -l,2,3-triazol-4- ylmethyl)carbamoyl]-2,3-dihydro-lH-indol-l-yl]ethyl}carbamoy l)butyl]carbamic acid /er/-butyl ester was prepared from (2S)-l-[2-((2S,3S)-2-½ri-Butoxycarbonylamino-3- methyl-pentanoylamino)-acetyl]-2,3-dihydro-lH-indole-2-carbo xylic acid and (2H-1,2,3- triazol-4-yl)methyl-amine) using method A in DMF but without HC1 treatment. MS (LC/MS) m/z observed 513.97, expected 514.28 [M+H]. Compound was confirmed using LC/MS and moved to next step as it was.

Title compound C39 was prepared from N-[(lS,2S)-2-methyl-l-({2-oxo-2-[(2S)- 2-[(lH-l,2,3-triazol-4-ylmethyl)carbamoyl]-2,3-dihydro-lH-in dol-l- yl]ethyl}carbamoyl)butyl]carbamic acid ieri-butyl ester and 2-(2H-tetrazol-5-yl)acetic acid using method A but with DMF as solvent for the coupling reaction: 'I! NMR (400 MHz, DMSO-i/6) δ 0.82 (3H, t, 1.12 (IH, m), 1.50 (IH, m), 1.78 (IH, m), 3.05 (IH, m), 3.56-3.67 (2H, m), 3.92-4.02 (2H, m), 4.13 (IH, m), 4.31 (IH, t, J=8Hz), 4.37-4.43 (2H, m), 5.13 (IH, d, J=8Hz), 7.01 (IH, t, J=8Hz), 7.14-7- 28 (2H, m), 7.70 (IH, s), 8.04 (IH, d, J=8Hz), 8.34 (IH, bs), 8.47 (IH, d, J=9Hz), 8.88 (IH, bs), MS (LC/MS) m/z observed 524.62, expected 524.25 [M+H]. EXAMPLE Dl

GENERAL KINETIC ENZYME ASSAY PROTOCOL

A specific 2X assay buffer was prepared for the enzyme to be tested (see Table 2 for final I X assay buffer compositions). If the assay buffer included DTT, it was added immediately prior to running the assay. A 2X enzyme mix was prepared (see Table 3 for enzyme assay conditions) at 80 uL per well. Compounds were screened at one or two appropriate concentrations (to determine the percent inhibition at those concentrations) and/or a full dose response curve (typically 8 points, to identify the IC50) in duplicate, triplicate, or higher replicates as needed. An appropriate control was also assessed in full dose response, in duplicate for each assay/plate. Background control wells consisted of I X assay buffer, DMSO (5% v/v) and substrate. Positive control wells consisted of enzyme, DMSO (5% v/v) and substrate. Test compounds and control compounds were diluted in DMSO to 40X the final desired concentration. For example, a test compound may be tested in dose response, in serial, tripling dilution condition starting at 20uM and ending at 9.1 nM (or any appropriate concentration range and dilution scheme). Control compounds were prepared similarly. Diluted compounds were prepared in a dilution plate and transferred to the reaction plate (96-well medium binding plate (Greiner Bio- One FLUOTRAC™)) to allow for the desired final concentrations when added to the enzyme with AB. After mixing, the reaction plate was placed on a shaker (at 300 RPM) for 5 min, followed by incubation (covered) on the bench, for 20 min. Plates were warmed to reaction temperature (see Table 3) for a total incubation time of 30 min. Plates so prepared were ready for addition of substrate and the subsequent reaction,

An appropriate substrate for each assay was prepared in advance at 2X the final desired concentration (see Table 2) in DMSO. The appropriate substrate mix was added to each appropriate well on the reaction plate, and the plate was read immediately in the TECAN plate reader (TECAN INFINITE® Ml 000 Pro), set to the correct wavelength as needed for each assay (see Table 3) using 25 cycles, kinetic interval of l min, number of reads per well of 20 with shaking set to I s, double orbital, 2mm amplitude. For fluorescent assays the gain was set to optimal (50%).

Table 2. Assay Buffer Composition. Assay Buffer Composition

Enzyme

50 mM HEPES pH 7.2

50 mM NaCl

Caspase 1 , 3, 4, 5, 7, 8*, 9 & 10/a

0. 1 % (w/v) CHAPS

(General caspase assay buffer)

10 mM EDTA

5% (v/v) Glycerol

l O mM DTT

50 mM HEPES pH 7.5

10% (w/v) sucrose

GzmB & Caspase 8

0.2% (w/v) CHAPS

5 mM DTT

*Can also use GzmB assay buffer for the Caspase-8 assay; Assay buffer components were sourced as follows: HEPES, DTT, Glycerol and sucrose: Sigma- Aldrich, St. Louis, MO, USA, NaCl and EDTA: Fisher Scientific, Pittsburgh, PA, USA, CHAPS: Calbiochem, Billerica, MA, USA.

Table 3. Enzyme assay conditions.

* Ex/Em λ is the excitation and emission wavelengths at which to measure fluorescence. Enzyme and substrate concentrations are the final concentrations in the well. Note that most protocols require preparing 2X enzyme and substrate mixes, as they are diluted 2-fold in the well.

Enzymes were sourced as follows: hGzmB, Froelich Lab, Northshore University

Health Systems Research Institute, Evanston, IL, USA; Caspases, Biovision Inc., Milpitas, CA, USA. Substrates were sourced as follows: Ac-IEPD-AMC, California Peptide Research Inc., Napa, CA, USA; YVAD-AFC, Biovision Inc., Milpitas, CA, USA; Ac-DEVD-AMC, LEHD-AFC, AC-WEHD-AFC and Ac-IETD-AMC, Enzu Life Sciences Inc, Farmingdale, NY, USA. Control inhibitors were sourced as follows: Ac- IEPD-CHO, Ac-WEHD-FM and Q-LEHD-Oph, Biovision Inc., Milpitas, CA, USA; Z-VAD-FMK, R&D Systems, Minneapolis, MN, USA; and Ac-AEVD-CHO, Enzo Life Sciences Inc, Farmingdale, NY, USA.

EXAMPLE D2

HUMAN GRANZYME B ENZYMATIC INHIBITION ASSAY

An in vitro fluorogenic detection assay for assessing the IC50 and/or percent inhibition at a given concentration of inhibitors against human Granzyme B (hGzmB) enzyme was performed as described in Example D l . When appropriate, percent inhibition data was collected and fitted to generate IC50 data using GraphPad Prism 5 (GraphPad Software, La Jolla California USA, www.graphpad.com) and its non-linear regression analysis tools or other equivalent tools.

Select compounds of Examples A1-A57, B 1 -B7 and C1-C39 exhibited inhibitory activity against hGzmB. Each of the compounds of the invention identified in Table 1 exhibited Granzyme B inhibitory activity.

In certain embodiments, select compounds exhibited IC50 <50,000 nM. In other embodiments, select compounds exhibited IC50 <10,000 nM. In further embodiments, select compounds exhibited IC50 <1 ,000 nM. In still further embodiments, select compounds exhibited IC50 <100 nM. In certain embodiments, select compounds exhibited IC50 from 10 nM to 100 nM, preferably from 1 nM to 10 nM, more preferably from 0.1 nM to 1 nM, and even more preferably from 0.01 nM to 0.1 nM. EXAMPLE D3

HUMAN CASPASE ENZYMATIC INHIBITION ASSAY

In vitro fluorogenic detection assays for assessing the IC50 and/or percent inh ibition at a given concentration of inhibitors, against a set of human Caspase enzymes, was performed as described in Example D l . Representative compounds of the invention do not significantly inhibit any caspase enzyme tested at a concentration of 50 μΜ.

In certain embodiments, the compounds exhibited less than 50% inhibition at 50 μΜ. In other embodiments, the compounds exhibited greater than 50% inhibition at 50 μΜ, but less than 10% inhibition at 25 μΜ.

EXAMPLE D4

KINETIC SOLUBILITY DETERMINATION

Kinetic solubility buffer (phosphate buffered saline (PBS, I X) at pH 7.4) was prepared from PBS ( 10X) solution, (Fisher Scientific, Pittsburgh, PA, USA, 10X), by adding PBS (50mL, 10X) to approximately water (450mL HPLC grade). The volume of the solution was then adjusted to 500mL for a total dilution factor of 1 : 10 and a final PBS concentration of I X. The pH of the final solution (PBS (I X)) was measured and found to be 7.4.

A DMSO stock solution (typically l OmM or greater) was used for each compound to be tested. A final DMSO concentration of 2.0% and maximum theoretical compound concentration of typically 200 μΜ (or greater) was achieved by diluting an al iquot (6 μΐ) of each stock with kinetic solubility buffer (294 μΐ, PBS ( l x) at pH 7.4) using a liquid hand ling system (Hamilton STARlet, Hamilton Robotics, Inc., Reno, NV, USA) and incubated directly in a solubility filter plate (Millipore, Billerica, MA, USA). Following 24 hour incubation at ambient temperature (20.5-21 .7 °C), each sample was vacuum filtered. The filtrate was injected into the chemiluminescent nitrogen detector for quantification (Automated Discovery Workstation, Analiza, Inc., Cleveland, OH, USA). Each result was reported both in μΜ and μg/mL units. Three separate on-board performance indicating standards were assayed in triplicate with each test compound to ensure test results were within the acceptable range. The equimolar nitrogen response of the detector was calibrated using standards which spanned the dynamic range of the instrument from 0.08 to 4500 μg/ml nitrogen. Each filtrate was quantified with respect to this calibration curve. Each calculated solubility value was corrected for background nitrogen present in the DMSO, and buffer used to prepare each sample. All reported values for samples containing adjacent n itrogen atoms in a ring structure were corrected for the expected non-equimolar response. Each calculated solubility result assumed that each sample was free of nitrogen containing impurities and was stable under the assay conditions. Analyses were performed at Analiza Inc. (Cleveland, OH, USA) by published rnelhods (Bhaltaehar, S.N., et al, J. Pharma. BioMedAnal. 41 : 152-7 2006).

Table 4. Kinetic solubility in PBS.

EXAMPLE D5

GENERAL KINETIC ENZYME ASSAY PROTOCOL (384 WELL)

A specific 2X assay buffer was prepared for the enzyme to be tested (see Table 5 for final I X assay buffer compositions). If the assay buffer included DTT, it was added immediately prior to running the assay. A 2X enzyme mix was prepared (see Table 3 for enzyme assay conditions) at 26 uL per well. Compounds were screened at one or two appropriate concentrations (to determine the percent inhibition at those concentrations) and/or a full dose response curve (typically 12 points, to identify the IC5 0 ) in duplicate, triplicate, or higher replicates as needed. An appropriate control was also assessed in full dose response, in duplicate for each assay/plate. Background control wells consisted of IX assay buffer and substrate. Positive control wells consisted of enzyme (no DMSO) and substrate. Test compounds and control compounds were diluted in I X Assay Buffer to 1 5X the final desired concentration. For example, a test compound may be tested in dose response, in serial, tripling dilution condition starting at 20uM and ending at 0.1 nM (or any appropriate concentration range and dilution scheme). Control compounds were prepared similarly. Diluted compounds were prepared in a dilution plate and transferred to the reaction plate (384-well medium binding plate (Greiner Bio-One FLUOTRAC™)) to allow for the desired final concentrations when added to the enzyme with AB. After mixing, the reaction plate was placed on a shaker (at 300 RPM) for 5 min, fol lowed by incubation (covered) on the bench, for 20 min. Plates were warmed to reaction temperature (see Table 6) for 5 mins for a total incubation time of 30 min. Plates so prepared were ready for addition of substrate and the subsequent reaction.

An appropriate substrate for each assay was prepared in advance at 2X the final desired concentration (see Table 5) in assay buffer. 30uL of the appropriate substrate mix was added to each appropriate well on the reaction plate, and the plate was read immediately in the TECAN plate reader (TECAN INFINITE ® M l 000 Pro), set to the correct wavelength as needed for each assay (see Table 6) using 15 cycles, kinetic interval of l min, number of reads per well of 20 with shaking set to Is, double orbital, 2mm amplitude. For fluorescent assays the gain was set to optimal (100% with gain regulation) for all assays except human GzmB which waswefe set to 85 (with the z set at 23000 urn).

Table 5. Assay Buffer Composition.

*Can also use GzmB assay buffer for the Caspase-8 assay; Assay buffer components were sourced as follows: HEPES, DTT, Glycerol and sucrose: Sigma- Aldrich, St. Louis, MO, USA, NaCl and EDTA: Fisher Scientific, Pittsburgh, PA, USA, CHAPS: Calbiochem, Billerica, MA, USA.

Table 6. Enzyme assay conditions.

* Ex/Em λ is the excitation and emission wavelengths at which to measure fluorescence. Enzyme and substrate concentrations are the final concentrations in the well. Note that most protocols require preparing 2X enzyme and substrate mixes, as they are diluted 2-fold in the well.

Enzymes were sourced as follows: hGzmB, Froelich Lab, Northshore University Health Systems Research Institute, Evanston, IL, USA; Caspases and Elastase, Biovision Inc., Milpitas, CA, USA; Cathepsin G, Athens Research and Technologies, Athens, GA, USA. Substrates were sourced as follows: Ac-IEPD-AMC, California Peptide Research Inc., Napa, CA, USA; YVAD-AFC and MeOSuc-AAPF-AFC Biovision Inc., Miipitas, CA, USA; LEHD-AFC and Suc-AAPF-pNA Millipore, Bi llerica MA, USA. Ac-DEVD- AMC, AC- WEHD-AFC and Ac-IETD-AMC, Enzo Life Sciences Inc, Farmingdale, NY, USA. Control inhibitors were sourced as follows: Ac-IEPD-CHO, Ac-WEHD-FMK, Q- LEHD-Oph and CatG inhibitor Biovision Inc., Miipitas, CA, USA; Z-VAD-FMK, R&D Systems, Minneapolis, MN, USA; and Ac-AEVD-CHO, Enzo Life Sciences Inc, Farmingdale, NY, USA. Sivelestat, Tocris Bioscience, Bristol, UK.

EXAMPLE D6

HUMAN GRANZYME B ENZYMATIC INHIBITION ASSAY

An in vitro fluorogenic detection assay for assessing the IC50 and/or percent inhibition at a given concentration of inhibitors against human Granzyme B (hGzmB) enzyme was performed as described in Example D5. When appropriate, percent inhibition data was collected and fitted to generate IC50 data using GraphPad Prism 5 (GraphPad Software, La Jolla California USA, www.graphpad.com) and its non-linear regression analysis tools or other equivalent tools.

Select compounds of Examples A1 -A57, B 1 -B7 and C 1 -C39 exhibited inhibitory activity against hGzmB. Each of the compounds of the invention identified in Table 1 exhibited Granzyme B inhibitory activity.

In certain embodiments, select compounds exhibited IC50 <50,000 nM. In other embodiments, select compounds exhibited IC50 <10,000 nM. In further embodiments, select compounds exhibited IC50 <1 ,000 nM. In still further embodiments, select compounds exhibited ICso <100 nM. In certain embodiments, select compounds exhibited IC 50 from 10 nM to 100 nM, preferably from 1 nM to 10 nM, more preferably from 0.1 nM to 1 nM, and even more preferably from 0.01 nM to 0.1 nM.

EXAMPLE D7

HUMAN CASPASE ENZYMATIC INHIBITION ASSAY

In vitro fluorogenic detection assays for assessing the IC50 and/or percent inhibition at a given concentration of inhibitors, against a set of human Caspase enzymes, was performed as described in Example D5 , Representative compounds of the invention do not significantly inhibit any caspase enzyme tested at a concentration of 50 μΜ.

In certain embodiments, the compounds exhibited less than 50% inhibition at 50 μΜ. In other embodiments, the compounds exhibited greater than 50% inhibition at 50 μΜ, but less than 10% inhibition at 25 μΜ.