Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
MACROCYCLIC COMPOUNDS FOR THE INHIBITION OF INDOLEAMINE-2,3-DIOXYGENASE ACTIVITY AND USE THEREOF
Document Type and Number:
WIPO Patent Application WO/2018/053302
Kind Code:
A1
Abstract:
The present invention relates to compounds, and pharmaceutically acceptable compositions thereof, useful as antagonists of indoleamine-2,3-dioxygenase (IDO) activity, and methods of treating IDO-related disorders.

Inventors:
BRIGGS TIMOTHY F (US)
ALEXANDER SETHU L (US)
CIPRIANI TYLER J (US)
DAVIS HEATHER (US)
DUVALL JEREMY R (US)
HALE STEPHEN P (US)
JIANG JUN (US)
LEE MAURICE D IV (US)
MEHOVIC REVONDA M (US)
MUNCIPINTO GIOVANNI (US)
OALMANN CHRISTOPHER J (US)
SHORTSLEEVES KELLEY C (US)
TERRETT NICHOLAS K (US)
Application Number:
PCT/US2017/051833
Publication Date:
March 22, 2018
Filing Date:
September 15, 2017
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
ENSEMBLE THERAPEUTICS CORP (US)
International Classes:
C07D498/18; A61K31/407; A61K31/424; A61P35/00; C07D498/04
Domestic Patent References:
WO2014074658A12014-05-15
WO2010022249A22010-02-25
WO2017049383A12017-03-30
WO2014150677A12014-09-25
Foreign References:
US20070021331A12007-01-25
Other References:
SUN, H. ET AL.: "Cyclopeptide Smac mimetics as antagonists of IAP proteins", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 20, no. 10, 2010, pages 3043 - 3046, XP027036786
Attorney, Agent or Firm:
BRODOWSKI, Michael, H. et al. (US)
Download PDF:
Claims:
CLAIMS

1. A com ound of Formula I:

(I), or a pharmaceutically acceptable salt thereof, wherein: ring A is selected from phenylene, pyridinylene, or pyrrolidinylene, wherein ring A is connected through adjacent ring atoms to moiety X and to the adjacent carbonyl and is optionally substituted;

X is -O- or -CH2-;

Y is a bond or -C(Rlla)(Rl lb);

R1 is selected from *-N(R10)-C(O , *-N(R10)-CH2-, *-N(R10)-C(O)-CH2-, -CH2-CH2- or nitrogen-containing heteroarylene, wherein "*" represents a portion of R1 bound to R2;

R2 is selected from -(C5-C10 alkylene)-†, -(C5-C10 alkenylene)-†, -(C1-C3 alkylene)- phenylene-0-(Ci-C3 alkylene)-†, -(C2-C3 alkylene)-0-(C2-C5 alkylene)-†, -(C2-C3 alkylene)-0- (C2-C3 alkylene)-0-(C2-C3 alkylene)-†, -(C2-C3 alkylene)-N(R7e)-(C2-C3 alkylene)-†, -(C2-C3 alkylene)-heteroarylene-(Ci-C3 alkylene)-†, or -(C2-C3 alkylene)-heterocyclylene-(Ci-C3 alkylene)-†, wherein "†" represents a portion of R2 bound to R3; and wherein each alkylene, alkenylene, phenylene, heteroarylene, and heterocyclylene portion of R2 is optionally substituted;

R3 is selected from -CH2-, **-CH2-N(R10)-, **-C(0)N(R7d)-, **-(CH2)0-2-NR7dC(O)-, -NR7d-, **-C(0)0-, heterocyclylene, heteroarylene, or phenylene, wherein "**" represents a portion of R3 bound to C(R4a)(R4b)†; and wherein said heterocyclylene, heteroarylene, and phenylene are optionally substituted;

each of R4a and R4b is independently selected from hydrogen, deuterium, halogen, or optionally substituted C1-C4 alkyl;

R5a is selected from hydrogen, deuterium or optionally substituted C1-C4 alkyl;

R5b is selected from hydrogen, deuterium, Ci-C6 alkyl, -O-C1-C6 alkyl, -CH2-heteroaryl, -C]¾-aryl, -(CH2)o-i-carbocyclyl, or -(CH2)o-i-heterocyclyl, wherein any alkyl, -CH2-heteroaryl, -CH2-aryl, -(CH2)o-i-carbocyclyl, or -(CH2)o-i-heterocyclyl portion of R5b is optionally substituted;

R6 is selected from hydrogen, Ci-Cio alkyl, CH2-C3-C6 cycloalkyl, or benzyl, wherein R6 is optionally substituted;

R7a is selected from hydrogen, C1-C4 alkyl, C2-C4 alkenyl, C2-C4 alkynyl, -CH2-aryl, -CH2 heteroaryl, C3-C6 cycloalkyl, or heterocyclyl, wherein R7a is optionally substituted;

each of R7b, R7c, R7d and R7e is independently selected from hydrogen or C1-C4 alkyl;

R8 is selected from hydrogen, deuterium or optionally substituted C1-C4 alkyl; or R7b and R8, when taken together with atoms to which they are bound, form a

heterocyclylene comprising 0-1 additional heteroatoms selected from N, O or S, wherein said heterocyclylene is optionally substituted;

R9 is hydrogen, deuterium or methyl;

each R10 is independently selected from hydrogen, -(C1-C4 alkyl), -C(0)-0-(Ci-C5 alkyl), -C(0)-0-(C2-C4 alkenyl), or C3-C6 cycloalkyl; and

each of Rlla and Rllb is independently selected from hydrogen, halogen, or optionally substituted C1-C4 alkyl; and

represents a bond between two adjacent ring atoms in ring A.

2. The compound of claim 1, wherein R1 is *-N(R10)-C(O)- or *-N(R10)-CH2-.

3. The compound of claim 1, wherein R1 is selected from *-NH-C(0 , *-N(CH3)-C(0)-, *-N(CH3)-CH2-, *-N(CH(CH3)2)- -, *-NH-CH2-, *-N(cyclopropyl)-CH2-,

*N(C(0)OCH2CH=CH2)-C(0)-,

4. The compound of any one of claims 1-3, wherein R2 is -(C5-C10 alkylene)-†.

5. The compound of any one of claims 1-3, wherein R2 is selected from -(012)5-9-, - CH(CH3)-(CH2)7-†, -CH(CH(CH3)2)-(CH2)7-†, -CH(CH3)-CH=CH-(CH2)5-†, -CH2-CHOH- (CH2)6-†, -(CH2)2-0-(CH2)5-†, -(CH2)2-0-(CH2)2-0-(CH2)2-†, -(CH2)3-0-(CH2)3-†,

6. The compound of any one of claims 1-5, wherein R3 is **-C(0)N(R7d)-, **-(CH2)o-2- NR7dC(0)-, or heteroarylene optionally substituted with by 1 or 2 substituents independently selected from the group consisting of halogen, hydroxyl, and Ci-C6 alkyl.

7. The compound of any one of claims 1-5, wherein R3 is selected from -CH2-, **-C(0)- N(CH3 , **-C(0)-NH-, **-(CH2)2-NH-C(0)-, **-CH2-NH-C(0 , -NH-, **-C(0)0-,

**-CH2-N(C(0)0-CH2-CH=CH2)-,

8. The compound of any one of claims 1-7, wherein one of R a or R is hydrogen; and the other is selected from hydrogen and -CH3.

9. The compound of any one of claims 1-8, wherein R5ais hydrogen or -CH3, and R5b is hydrogen, Ci-C6 alkyl, -CH2-heteroaryl or -CH2-aryl, wherein said aryl or heteroaryl are optionally substituted with 1, 2, or 3 substituents independently selected from the group consisting of halogen, hydroxyl, Ci-C6 alkyl, and C3-C6 cycloalkyl.

10. The compound of any one of claims 1-8, wherein R5a is selected from hydrogen or -CH3; and

R5b is selected from hydrogen, -CH3, -CH2CH(CH3)2, -CH(CH3)CH2CH3, -CH2-0- C(CH3)3, -CH(OH)CH3, -CH(CH3)2, -CH(CH3)-0-C(CH3)3, -CH2C(CH3)3, cyclohexyl, cyclopropylmethyl, cyclobutylmethyl, benzyl, pyridin-2-ylmethyl, pyridin-3-ylmethyl, pyridin- 2-ylmethyl, 5-fluororpyridin-3-ylmethyl, 2,4-difluoropyridin-3-ylmethyl, 4-methylpyridin-3- ylmethyl, 2-methylpyridin-3-ylmethyl, 2-hydroxypyridin-3-ylmethyl, phenylethan-l-yl, 1- oxopyridin-3-ylmethyl, 3,4-difluorophenylmethyl, 4-hydroxyphenylmethyl, 2- fluorophenylmethyl, 3-fluorophenylmethyl, 4-fluorophenylmethyl, 4-cyanophenylmethyl, 4- trifluoromethylphenylmethyl, 4-bromophenylmethyl, 4-chlorophenylmethyl, 4- methoxyphenylmethyl, pyrazin-2-ylmethyl, thiazol-4-ylmethyl, thiophen-3 -ylmethyl, pyrimidin- 4-ylmethyl, benzo[b]thiophen-3-ylmethyl, l-methyl-lH-indol-3-ylmethyl, or 1- butoxycarbonylpiperidin-4-ylmethyl.

11. The compound of any one of claims 1-10, wherein R6 is C1-C5 alkyl.

12. The compound of any one of claims 1-10, wherein R6 is selected from -CH(CH3)2, -CH2CH(CH3)2, -C(CH3)3, cyclopropyl, cyclobutyl, cyclohexyl, or phenyl.

Rx

13. The compound of any one of claims 1-10, wherein R . is - ^ , wherein Rx is selected from C1-C9 alkyl, C3-C6 cycloalkyl, or phenyl, wherein Rx is optionally substituted with one, two, or three occurrences of Ci-C6 alkyl, C3-C6 carbocyclyl, -OH, -N(R10)2, or halogen. 14. The compound of any one of claims 1-13, wherein R7a is C1-C4 alkyl.

15. The compound of any one of claims 1-13, wherein R7a is selected from -CH3,

-CH2C≡CH, cyclopropyl, cyclohexyl, benzyl, 4-hydroxyphenylmethyl, furan-2-ylmethyl, or 1- tert-butyloxycarbonylpiperidin-4-yl.

16. The compound of any one of claims 1-14, wherein R7b and R8 are taken together with atoms to which they are bound to form a 5-6 membered, saturated heterocyclylene optionally substituted with 1, 2, or 3 substituents independently selected from the group consisting of halogen, hydroxyl, Ci-C6 alkyl, and C3-C6 cycloalkyl.

17. The compound of any one of claims 1-14, wherein R 7b is -C¾; and R 8 is selected from -CH3, CH2OH, or -CH2OC(CH3)3; or

R 7b and R 8 , when taken together with atoms to which they are bound, form a saturated heterocyclylene ring selected from pyrrolidin-l,2-diyl, piperidin-l,2-diyl, piperazin-l,2-diyl, morpholin-3,4-diyl, or thiomorpholin-3,4-diyl, wherein said saturated heterocyclylene ring is optionally substituted with one, two, or three occurrences of Ci-C6 alkyl, C3-C6 carbocyclyl, halogen, haloalky, oxo, -C(0)-0-(Ci-C5 alkyl), -C(0)-0-(C2-C4 alkenyl), -OH, -N(R10)2, heteroaryl, aryl, or heteroalkyl. 18. The compound of claim 17, wherein R7b and R8, when taken together with atoms to which they are bound, form pyrrolidin-l,2-diyl, 4-hydroxypyrrolidin-l,2-diyl, 4,4- difluoropyrrolidin-l,2-diyl, 4-oxopyrrolidin-l,2-diyl, 4-hydroxypyrrolidin-l,2-diyl, 4- dimethylaminopyrrolidin- 1 ,2-diyl, 4-methylaminopyrrolidin- 1 ,2-diyl, 4-fluoropyrrolidin- 1 ,2- diyl, piperidin-l,2-diyl, piperazin-l,2-diyl, morpholin-3,4-diyl, l-oxothiomorpholin-3,4-diyl, l,l-dioxothiomorpholin-3,4-diyl, or 4-tert-butyloxycarbonylpiperazin-l,2-diyl.

19. The compound of any one of claims 1-18, wherein R7c is independently selected from hydrogen or -CH3. 20. The compound of any one of claims 1-19, wherein R9 is selected from hydrogen or -CH3.

21. The compound of any one of claims 1-20, wherein ring A is selected from:

wherein "1" represents a portion of ring A bound to moiety X.

22. The compound of any one of claims 1-20, wherein ring A is

wherein "1" represents a portion of ring A bound to moiety X.

23. The compound of any one of claims 1-22, wherein Y is selected from a bond or -C¾-.

24. The compound of any one of claims 1-22, wherein Y is a bond.

25. The compound of any one of claims 1-24, wherein X is O.

26. The compound of any one of claims 1-25, wherein the compound is represented by Formula I-A:

(I-A)

or a pharmaceutically acceptable salt thereof, wherein:

R1 is *-N(R10)-C(O or *-N(R10)-CH2-, where"*" represents a portion of R1 bound to R2; R2 is -(C5-C10 alkylene)-;

R3 is **-C(0)N(R7d)- or heteroarylene optionally substituted by 1 or 2 substituents independently selected from the group consisting of halogen, hydroxyl, and Ci-C6 alkyl, wherein "**" represents a portion of R3 bound to C(R4a)(R4b)†; each of R a and R are independently hydrogen, halogen, or C1-C4 alkyl;

R5a is hydro gen or C1-C4 alkyl;

R5b is hydrogen, Ci-C6 alkyl, -CH2-heteroaryl, or -C]¾-aryl, wherein said heteroaryl and aryl are optionally substituted by 1 or 2 substituents independently selected from the group consisting of halogen, hydroxyl, and Ci-C6 alkyl;

R6 is C1-C10 alkyl;

R7a is C1-C4 alkyl;

R7d is hydrogen or C1-C4 alkyl;

R7b and R8 are taken together with atoms to which they are bound, form a 5-7 membered, saturated heterocyclylene optionally substituted by 1 or 2 substituents independently selected from the group consisting of halogen, hydroxyl, and Ci-C6 alkyl;

R9 is hydrogen or methyl;

R10 is hydrogen or C1-C4 alkyl; and

R12 represents independently for each occurrence halogen, hydroxyl, or Ci-C6 alkyl; and n is 0, 1, or 2.

27. A compound of Formula (II) :

(II), or a pharmaceutically acceptable salt thereof, wherein:

ring A is selected from phenylene, pyridinylene, or pyrrolidinylene, wherein ring A is connected through adjacent ring atoms to moiety X and to the adjacent carbonyl and is optionally substituted;

X is -O- or -CH2-; Y is a bond or -C(Rlla)(Rl lb);

Z is a heteroarylene or a phenylene, wherein Z is optionally substituted;

R1 is selected from *-N(R10)-C(O , *-N(R10)-CH2-, *-N(R10)-C(O)-CH2-, -CH2-CH2-, *- C(O)-N(R10)-CH2-, or a nitrogen-containing heteroarylene, wherein "*" represents a portion of R1 bound to R2;

R2 is selected from -(C5-C10 alkylene)-†, -(C5-C10 alkenylene)-†, -(C1-C3 alkylene)- phenylene-0-(Ci-C3 alkylene)-†, -(C2-C3 alkylene)-0-(C2-C5 alkylene)-†, -(C2-C3 alkylene)-0- (C2-C3 alkylene)-0-(C2-C3 alkylene)-†, -(C2-C3 alkylene)-N(R7e)-(C2-C3 alkylene)-†, -(C2-C3 alkylene)-heteroarylene-(Ci-C3 alkylene)-†, and -(C2-C3 alkylene)-heterocyclylene-(Ci-C3 alkylene)-†, wherein "†" represents a portion of R2 bound to R3; and wherein each alkylene, alkenylene, phenylene, heteroarylene, or heterocyclylene portion of R2 is optionally substituted;

R3 is selected from -CH2-, **-CH2-N(R10)-, **-C(0)N(R7d)-, **-(CH2)0.2-NR7dC(O)-, -NR7d-, **-C(0)0-, heterocyclylene, heteroarylene, or phenylene, wherein "**" represents a portion of R3 bound to C(R4a)(R4b)†; and wherein each heterocyclylene, heteroarylene, or phenylene is optionally substituted;

each of R4a and R4b is independently selected from hydrogen, deuterium, halogen, or optionally substituted C1-C4 alkyl;

R6 is selected from C1-C9 alkyl, C3-C6 cycloalkyl, or phenyl, wherein R6 is optionally substituted;

each of R7b, R7c, R7d and R7e is independently selected from hydrogen or C1-C4 alkyl;

R8 is selected from hydrogen, deuterium or optionally substituted C1-C4 alkyl; or

R7b and R8, when taken together with atoms to which they are bound, form optionally substituted heterocyclylene comprising 0-1 additional heteroatoms selected from N, O or S; wherein said heterocyclylene is optionally substituted;

R9 is hydrogen, deuterium or methyl;

R10 is selected from hydrogen, -(d-C4 alkyl), -C(0)-0-(C2-C4 alkenyl), -C(0)-0-(Ci-C5 alkyl), or C3-C6 cycloalkyl;

each of Rlla and Rllb is independently selected from hydrogen, halogen, or optionally substituted C1-C4 alkyl; and

"— " represents a bond between two adjacent ring atoms in ring A.

28. The compound of claim 27, wherein Z is selected from or phen-l,3-diyl, wherein "††" represents a portion of Z bound to R1.

29. A compound in Table 1 herein, or a pharmaceutically acceptable salt thereof.

30. A pharmaceutical composition comprising a compound of any one of claims 1-29 and a pharmaceutically acceptable carrier.

31. A method of treating a cancer in a subject in need thereof, the method comprising the step of administering to the subject an effective amount of a compound of any one of claims 1-

29, or a composition of claim 30.

32. The method of claim 31, wherein cells associated with the cancer are characterized as having an elevated level of IDO activity and/or expression when compared to non-cancerous cells of the same cell type.

33. The method of claim 31 or 32, wherein the subject is characterized as having an elevated plasma concentration of kynurenine when compared to a subject without the cancer. 34. A method of treating a cancer in a subject in need thereof, the method comprising:

administering to the subject an effective amount of a compound of any one of claims 1- 29 or a composition of claim 30, wherein the cancer is characterized as having an elevated level of IDO gene expression relative to average IDO gene expression in cells of the same cell type without the cancer.

35. A method of treating a cancer in a subject in need thereof, the method comprising:

administering to the subject an effective amount of a compound of any one of claims 1- 29 or a composition of claim 30 wherein the subject has a kynurenine level in a body fluid that is elevated relative to the average kynurenine level in the same type of body fluid in subjects without the cancer.

36. A method of treating a cancer in a subject in need thereof, the method comprising:

a. obtaining a cancer cell sample from the subject;

b. determining the level of IDO gene expression in the sample; c. comparing the IDO gene expression level in the sample to a threshold value in a normal cell to determine if IDO gene expression is elevated in the cancer sample;

d. administering to the subject having an elevated level of IDO gene expression an effective amount of a compound of any one of claims 1-29, or a composition of claim 30.

37. A method of treating a cancer in a subject in need thereof, the method comprising:

a. obtaining a body fluid sample from the subject;

b. determining the concentration of kynurenine in the fluid sample;

c. comparing the kynurenine concentration in the fluid sample to a threshold value of kynurenine present the same type of body fluid of subjects without the cancer thereby to determine if kynurenine is elevated in the fluid sample of the subject;

d. administering to the subject having an elevated concentration of kynurenine an effective amount of a compound of any one of claims 1-29, or a composition of claim 30.

38. The method of any one of claims 31-37, wherein the cancer is selected from cervical, renal, endometrial, squamous cell, colorectal, prostate, head and neck, lung, large B cell lymphoma, or acute myeloid leukemia.

39. The method of any one of claims 31 - 38, further comprising administering to the subject a cancer immunotherapy agent.

40. The method of claim 39, wherein the cancer immunotherapy agent is a PD-1 inhibitor.

41. The method of claim 40, wherein the PD-1 inhibitor is pembrolizumab.

42. A method of reducing IDO activity in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound of any one of claims 1-29, or a composition of claim 30.

Description:
MACROCYCLIC COMPOUNDS FOR THE INHIBITION OF

INDOLEAMINE-2,3-DIOXYGENASE ACTIVITY AND USE THEREOF

CROSS-REFERENCE TO RELATED APPLICATION

[0001] This application claims the benefit of and priority to U.S. Provisional Patent

Application No. 62/396,531, filed September 19, 2016, which is hereby incorporated by reference herein in its entirety.

FIELD OF THE INVENTION

[0002] The present invention relates to macrocyclic compounds useful as antagonists of indoleamine-2,3-dioxygenase (IDO) activity. The invention also provides pharmaceutically acceptable compositions comprising such macrocyclic compounds and methods of using such macrocyclic compounds and compositions in the treatment of IDO-related disorders.

BACKGROUND OF THE INVENTION

[0003] The oxygenase indoleamine-2,3-dioxygenase (IDO, also known as IDO 1 or IDO-1), is a heme-containing cytosolic enzyme that catalyzes the extra-hepatic metabolism of the essential amino acid, tryptophan (Trp). The heme pocket of IDO 1 binds both molecular oxygen and Trp, catalyzing oxidative cleavage of the indole ring of Trp. The product of this first and rate- limiting step of Trp metabolism is N-formyl kynurenine, which is a precursor of a variety of bioactive molecules known as kynurenines that have immunomodulatory properties (Shwarcz et al. (2012) NAT. REV. NEUROSCI. 13(7):465-77).

[0004] IDO is an inducible enzyme that has a primary role in immune cell modulation. Given that Trp is required for immune T cell activation, the reduction of Trp levels and/or an increase in the pool of kynurenines can prevent the activation of effector immune cells and promote adaptive immune suppression.

[0005] It has been discovered that certain cancers evade tumor directed immune responses by over expressing IDO, thereby down regulating the anti-tumor T cell response through tryptophan depletion. Comparative studies on the levels of IDO gene expression in human cancer versus normal tissues indicate a significant role in various cancers including cervical, renal, endometrial and squamous cell carcinomas and large B cell lymphoma. Another study using patient plasma levels of kynurenine, in comparison to normal values, as an indicator of IDO disease activity implicated the following cancers: acute myeloid leukemia (AML), colorectal, prostate, head and neck cancer, and lung cancer. [0006] Exemplary IDO inhibitors are described in U.S. Patent Nos. 8,034,953, 8,088,803, 8,232,313, 8,389,568 and PCT Publication No. WO2014/150677, and include the small molecules INCB024360, also known as Epacadostat (Incyte Corporation), Indoximod (NewLink Genetics), NLG919 (NewLink Genetics), and F001287 (Flexus Biosciences). Certain compounds that specifically inhibit IDO activity have been shown to enhance an anti-tumor response and are undergoing clinical evaluation to assess their safety and efficacy (Moon et al. (2015) J. IMMUNOTHER. CANCER 3:51). For example, clinical trials have been testing

Epacadostat, a hydroxyamidine small molecule IDO inhibitor, either alone or in combination with other agents, in the treatment of a variety of cancers including melanoma, ovarian cancer, myelodysplastic syndrome, and non-small cell lung cancer.

[0007] Despite these efforts to date, there is still a need for other more inhibitors of IDO 1 for use in treating various medical disorders.

SUMMARY

[0008] The present invention provides macrocyclic compounds, methods of inhibiting IDO 1 activity, and methods of treating various medical conditions implicated or otherwise associated with elevated levels of IDO enzyme activity, for example, cancer, using such compounds.

[0009] In one aspect, the invention provides a compound of Formula I:

or a pharmaceutically acceptable salt thereof, wherein the variables are described in the detailed description. The invention also provides pharmaceutical compositions comprising a compound of Formula I.

[0010] In another aspect, the invention provides a compound of Formula (II):

(Π)

[0011] or a pharmaceutically acceptable salt thereof, wherein the variables are described in the detailed description. The invention also provides pharmaceutical compositions comprising a compound of Formula II.

[0012] In another aspect, the invention provides a method of reducing IDO activity in a subject in need thereof. The method comprises administering to the subject an effective amount of a compound of Formula I (including Formula I- A, as discussed below) or II, or a pharmaceutical composition comprising a compound of Formula I (including Formula I- A) or II.

[0013] The invention also provides methods of treating a subject suffering from or susceptible to a medical condition that is implicated or otherwise associated with elevated levels of IDO enzyme activity, for example, cancer, such as cervical, renal, endometrial, squamous cell, colorectal, prostate, head and neck, lung, large B cell lymphoma, or acute myeloid leukemia.

[0014] Accordingly, in another aspect, the invention provides a method of treating a cancer in a subject in need thereof, the method comprising administering to the subject an effective amount of a compound of Formula I (including Formula I- A) or II, or a pharmaceutical composition comprising a compound of Formula I (including Formula I- A) or II. The cancer may be characterized as having an elevated level of IDO activity and/or expression when compared to non-cancerous cells or tissue. Alternatively or in addition, the subject may be characterized as having an elevated concentration of kynurenine in a body fluid, such as whole blood or a blood product, when compared to similar body samples harvested from subjects without the cancer.

[0015] In another aspect, the invention provides a method of treating a cancer in a subject in need thereof, the method comprising: administering to the subject an effective amount of a compound of Formula I (including Formula I- A) or II, or a pharmaceutical composition comprising a compound of Formula I (including Formula I- A) or II, wherein cells associated with the cancer are characterized as having an elevated level of IDO gene expression relative to average IDO gene expression levels in cells of the same cell type without the cancer.

[0016] In another aspect, the invention provides a method of treating a cancer in a subject in need thereof, the method comprising: administering to the subject an effective amount of a compound compound of Formula I (including Formula I- A) or II, or a pharmaceutical composition comprising a compound of Formula I (including Formula I- A) or II, wherein the subject has a kynurenine level in a body fluid, for example, whole blood or a blood product, that is elevated relative to the average serum kynurenine levels in similar body fluids of subjects without the cancer.

[0017] In each of the foregoing methods, it is contemplated that method can also include the administration of an additional therapeutic agent, for example, a cancer immunotherapy agent, such as a PD-1 inhibitor.

[0018] The foregoing and other aspects and embodiments of the invention may be more fully understood by reference to the following detailed description and claims.

BRIEF DESCRIPTION OF THE DRAWINGS

[0019] Figure 1 is a graph showing the in vivo effect of compound 163 on tumor volume reduction over time when compared to control.

DETAILED DESCRIPTION OF THE INVENTION

I. Definitions

[0020] To facilitate an understanding of the present invention, a number of terms and phrases are defined below.

[0021] The terms "a" and "an" as used herein mean "one or more" and include the plural unless the context is inappropriate.

[0022] The term "alkyl" as used herein refers to a saturated straight or branched hydrocarbon, such as a straight or branched group of 1-12, 1-10, or 1-6 carbon atoms, referred to herein as C1-C12 alkyl, C1-C1 0 alkyl, and Ci-C 6 alkyl, respectively. Exemplary alkyl groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, 2-methyl-l -propyl, 2-methyl-2-propyl, 2-methyl-l -butyl, 3-methyl-l-butyl, 2-methyl-3-butyl, 2,2-dimethyl-l -propyl,

2-methyl-l-pentyl, 3 -methyl- 1-pentyl, 4-methyl-l-pentyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 2,2-dimethyl-l -butyl, 3,3-dimethyl-l-butyl, 2-ethyl-l -butyl, butyl, isobutyl, t-butyl, pentyl, isopentyl, neopentyl, hexyl, heptyl, octyl, etc. [0023] The term "heteroalkyl" as used herein refers to alkyl groups in which one or more carbon atoms are replaced with a heteroatom (e.g., oxygen, nitrogen, sulfur, or the like).

[0024] The terms "alkenyl" and "alkynyl" as used herein refer to unsaturated hydrocarbon groups analogous in length and possible substitution to the alkyls described above, but that contain at least one double or triple bond, respectively.

[0025] The term "cycloalkyl" as used herein refers to a monovalent fully saturated cyclic, bicyclic, or bridged cyclic (e.g., adamantyl) hydrocarbon group of 3-10, 3-8, 4-8, or 4-6 carbons, referred to herein, e.g., as "C4-8 cycloalkyl," derived from a cycloalkane. Exemplary cycloalkyl groups include, but are not limited to, cyclohexane, cyclopentane, cyclobutane, and

cyclopropane.

[0026] The term "alkylene" refers to the diradical of an alkyl group.

[0027] The term "Co alkylene" as used herein refers to a bond. Thus, a moiety defined herein as "-(Co-C 6 alkylene)-aryl" includes both -aryl (i.e., Co alkylene-aryl) and -(Ci-C 6 alkylene)-aryl.

[0028] The terms "alkenylene" and "alkynylene" refer to the diradicals of an alkenyl and an alkynyl group, respectively.

[0029] The term "methylene" as used herein refers to a divalent -C¾- group present in an alkyl or alkylene moiety.

[0030] The term "haloalkyl" as used herein refers to an alkyl group that is substituted with at least one halogen. For example, -C¾F, -CHF 2 , -CF 3 , -CH 2 CF 3 , -CF 2 CF 3 , and the like.

[0031] The term "carbocyclyl", as used herein, means a monocyclic, bicyclic or polycyclic hydrocarbon ring system, wherein each ring is either completely saturated or contains one or more units of unsaturation, but where no ring is aromatic. Representative carbocyclyl groups include cycloalkyl groups (e.g., cyclopentyl, cyclobutyl, cyclopentyl, cyclohexyl and the like), and cycloalkenyl groups (e.g., cyclopentenyl, cyclohexenyl, cyclopentadienyl, and the like).

[0032] The term "aryl" as used herein refers to a monocyclic, bicyclic or polycyclic hydrocarbon ring system, wherein at least one ring is aromatic. Representative aryl groups include fully aromatic ring systems, such as phenyl, naphthyl, and anthracenyl, and ring systems where an aromatic carbon ring is fused to one or more non-aromatic carbon rings, such as indanyl, phthalimidyl, naphthimidyl, or tetrahydronaphthyl, and the like. In certain

embodiments, the aromatic ring may be substituted at one or more ring positions or may not be substituted, i.e., it is unsubstituted. [0033] The term "aralkyl" refers to an alkyl group substituted with an aryl group.

[0034] The term "arylene" refers to the diradical of an aryl group.

[0035] The term "phenylene" refers to a diradical of a phenyl group.

[0036] The term "heteroaryl" as used herein refers to monocyclic, bicyclic or polycyclic ring system wherein at least one ring is both aromatic and comprises a heteroatom; and wherein no other rings are heterocyclyl (as defined below). In certain instances, a ring which is aromatic and comprises a heteroatom contains 1, 2, 3, or 4 ring heteroatoms in such a ring.

Representative heteroaryl groups include ring systems where (i) each ring comprises a heteroatom and is aromatic, e.g., imidazolyl, oxazolyl, thiazolyl, triazolyl, pyrrolyl, furanyl, thiophenyl pyrazolyl, pyridinyl, pyrazinyl, pyridazinyl, pyrimidinyl, indolizinyl, purinyl, naphthyridinyl, and pteridinyl; (ii) each ring is aromatic or carbocyclyl, at least one aromatic ring comprises a heteroatom and at least one other ring is a hydrocarbon ring or e.g., indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl,

benzothiazolyl, quinolyl, isoquinolyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl,

pyrido[2,3-b]-l,4-oxazin-3(4H)-one, 5,6,7,8-tetrahydroquinolinyl and

5,6,7,8-tetrahydroisoquinolinyl; and (iii) each ring is aromatic or carbocyclyl, and at least one aromatic ring shares a bridgehead heteroatom with another aromatic ring, e.g., 4H-quinolizinyl. In certain embodiments, the aromatic ring or may be substituted at one or more ring positions may not be substituted, i.e., it is unsubstituted.

[0037] The term "heteroarylene" refers to the diradical of a heteroaryl group.

[0038] The term "pyridinylene" refers to the diradical of pyridine.

[0039] The term "pyrrolidinylene" refers to the diradical of pyrrolidine.

[0040] The term "heterocyclyl" refers to monocyclic, bicyclic and polycyclic ring systems where at least one ring is saturated or partially unsaturated (but not aromatic) and comprises at least one heteroatom. A heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure and any of the ring atoms can be optionally substituted. Representative heterocyclyls include ring systems in which (i) every ring is non-aromatic and at least one ring comprises a heteroatom, e.g., tetrahydrofuranyl,

tetrahydrothienyl, pyrrolidinyl, pyrrolidonyl, piperidinyl, pyrrolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, morpholinyl, and quinuclidinyl; (ii) at least one ring is non-aromatic and comprises a heteroatom and at least one other ring is an aromatic carbon ring, e.g., 1,2,3,4-tetrahydroquinolinyl, 1,2,3,4-tetrahydroisoquinolinyl; and (iii) at least one ring is non-aromatic and comprises a heteroatom and at least one other ring is aromatic and comprises a heteroatom, e.g.,

3,4-dihydro-lH-pyrano[4,3-c]pyridine, and l,2,3,4-tetrahydro-2,6-naphthyridine. In certain embodiments, the aromatic ring may be substituted at one or more ring positions or may not be substituted, i.e., it is unsubstituted.

[0041] The term "saturated heterocyclyl" refers to a heterocyclyl wherein every ring is saturated, e.g., tetrahydrofuran, tetrahydro-2H-pyran, pyrrolidine, piperidine and piperazine.

[0042] The term "heterocyclylene" refers to the diradical of a heterocyclyl group.

[0043] The terms "amine" and "amino" refer to both unsubstituted and substituted amines, e.g., a moiety represented by the general formula -N(R 50 )(R 51 ), wherein R 50 and R 51 each independently represent hydrogen, alkyl, cycloalkyl, heterocyclyl, alkenyl, aryl, aralkyl, or -(CH 2 ) m -R 61 ; or R 50 and R 51 , taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure; R 61 represents an aryl, a cycloalkyl, a cycloalkenyl, a heterocycle or a polycycle; and m is zero or an integer in the range of 1 to 8. In certain embodiments, R 50 and R 51 each independently represent hydrogen, alkyl, alkenyl, or - (CH 2 ) m -R 61 .

[0044] The terms "alkoxyl" or "alkoxy" refer to an alkyl group, as defined above, having an oxygen radical attached thereto. Representative alkoxyl groups include methoxy, ethoxy, propyloxy, tert-butoxy and the like. An "ether" is two hydrocarbons covalently linked by an oxygen.

[0045] In general, the term "substituted," whether preceded by the term "optionally" or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent. Unless otherwise indicated, an "optionally substituted" group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at each position. Combinations of substituents envisioned under this invention are preferably those that result in the formation of stable or chemically feasible compounds.

[0046] Suitable monovalent substituents on a substitutable carbon atom of an "optionally substituted" group (such as an alkyl, alkenyl, alkynyl, alkylene, alkenylene, alkynylene or the carbon atom of a carbocyclyl, aryl, heterocyclyl or heteroaryl) are independently, for example, halogen; -(CH 2 ) 0 - 4 R o ; -(CH 2 ) 0 - 4 OR°; -O-(CH 2 ) 0 - 4 C(O)OR°; -(CH 2 ) 0 - 4 CH(OR°) 2 ; -(CH 2 ) 0 - 4 SR°; -(CH2)o-4Ph, which may be substituted with R°; -(CH 2 )o- 4 0(CH 2 )o-iPh which may be substituted with R°; -CH=CHPh, which may be substituted with R°; -N0 2 ; -CN; -N 3 ; -(CH 2 )o-4N(R 0 ) 2 ; -(CH 2 )o- 4 N(R 0 )C(0)R 0 ; -N(R°)C(S)R°; -(CH 2 ) 0 - 4 N(R°)C(O)NR° 2 ; -N(R°)C(S)NR° 2 ;

-(CH 2 ) 0 . 4 N(R o )C(O)OR°; -N(R°)N(R°)C(0)R°; -N(R°)N(R°)C(0)NR° 2 ; -N(R°)N(R°)C(0)OR°; -(CH 2 )o- 4 C(0)R°; -C(S)R°; -(CH 2 ) 0 ^C(O)OR°; -(CH 2 ) 0 - 4 C(O)SR°; -(CH 2 ) 0 - 4 C(O)OSiR° 3 ;

-(CH 2 )o_ 4 OC(0)R°; -OC(O)(CH 2 ) 0 . 4 SR°-, SC(S)SR°; -(CH 2 ) 0 _ 4 SC(O)R°; -(CH 2 ) 0 ^C(O)NR° 2 ; -C(S)NR° 2 ; -C(S)SR°; -(CH 2 ) 0 ^OC(O)NR° 2 ; -C(0)N(OR°)R°; -C(0)C(0)R°;

-C(0)CH 2 C(0)R°; -C(NOR°)R°; -(CH 2 ) 0 - 4 SSR°; -(CH 2 ) 0 - 4 S(O) 2 R°; -(CH 2 ) 0 - 4 S(O) 2 OR°;

-(CH 2 )o- 4 OS(0) 2 R°; -S(0) 2 NR° 2 ; -(CH 2 ) 0 - 4 S(O)R°; -N(R°)S(0) 2 NR° 2 ; -N(R°)S(0) 2 R°;

-N(OR°)R°; -C(NH)NR° 2 ; -P(0) 2 R°; -P(0)R° 2 ; -OP(0)R° 2 ; -OP(0)(OR°) 2 ; -SiR° 3 ; -(C 1-4 straight or branched alkylene)0-N(R°) 2 ; or -(Ci_ 4 straight or branched alkylene)C(0)0-N(R°) 2 , wherein each R° may be substituted as defined below and is independently hydrogen, Ci_6 alkyl, Ci-6 alkenyl, Ci_6 alkynyl, -CH 2 Ph, -0(CH 2 )o-iPh, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R°, taken together with their intervening atom(s), form a 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, which may be substituted as defined below.

[0047] Suitable monovalent substituents on R° (or the ring formed by taking two independent occurrences of R° together with their intervening atoms), are independently, for example, deuterium, halogen, -(CH 2 ) 0 - 2 R*, -(haloR*), -(CH 2 ) 0 - 2 OH, -(CH 2 ) 0 - 2 OR*, -(CH^CHCOR*),; -O(haloR'), -CN, -N 3 , -(CH^CCC R*, -(CH 2 ) 0 _ 2 C(O)OH, -(CH^CCC OR*, -(CH 2 ) 0 . 2 SR e , -(CH 2 )o- 2 SH, -(CH 2 )o- 2 NH 2 , -(CH 2 )o- 2 NHR*, -(CH^NR*,, -N0 2 , -SiR* 3 , -OSiR* 3 , -C(0)SR* -(Ci_ 4 straight or branched alkylene)C(0)OR*, or -SSR* wherein each R* is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently selected from Ci_ 4 alkyl, Ci_ 4 alkenyl, Ci_ 4 alkynyl, -CH 2 Ph, -0(CH 2 )o-iPh, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents on a saturated carbon atom of R° include =0 and =S.

[0048] Suitable divalent substituents on a saturated carbon atom of an "optionally substituted" group include the following: =0, =S, =NNR * 2 , =NNHC(0)R * , =NNHC(0)OR * , =NNHS(0) 2 R * , =NR * , =NOR * , -0(C(R * 2 )) 2 _ 3 0-, or -S(C(R * 2 )) 2 _ 3 S-, wherein each independent occurrence of R * is selected from hydrogen, Ci_6 alkyl, Ci_6 alkenyl, Ci_6 alkynyl, which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur. Suitable divalent substituents that are bound to vicinal substitutable carbons of an "optionally substituted" group include: -0(C(R*)2)2- 3 0-, wherein each independent occurrence of R * is selected from hydrogen, Ci_6 alkyl, Ci_6 alkenyl, Ci_6 alkynyl, which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.

[0049] Suitable substituents on the alkyl, alkenyl, alkynyl, or carbocyclyl group of R * include halogen, -R*, -(haloR*), -OH, -OR*, -O(haloR'), -CN, -C(0)OH, -C(0)OR*, -NH 2 , -NHR*, -NR*2, or -NO2, wherein each R* is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently Ci_6 alkyl, Ci_6 alkenyl, Ci_6 alkynyl, -CH2PI1, -0(CH 2 )o-iPh, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.

[0050] Suitable substituents on a substitutable nitrogen of an "optionally substituted" group include -R , -NR 2 , -C(0)R , -C(0)OR , -C(0)C(0)R , -C(0)CH 2 C(0)R , -S(0) 2 R ,

-S(0) 2 NR 2 , -C(S)NR 2 , -C(NH)NR 2 , or -N(R )S(0) 2 R ; wherein each R is independently hydrogen, Ci_6 alkyl, Ci_6 alkenyl, Ci_6 alkynyl, which may be substituted as defined below, unsubstituted -OPh, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R , taken together with their intervening atom(s) form an unsubstituted 3-12-membered saturated, partially unsaturated, or aryl mono- or bicyclic ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.

[0051] Suitable substituents on the alkyl, alkenyl, alkynyl, or carbocyclyl group of R are independently, for example, halogen, -R*, -(haloR*), -OH, -OR*, -O(haloR'), -CN, -C(0)OH, -C(0)OR*, -NH 2 , -NHR*, -NR* 2 , or -N0 2 , wherein each R* is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently Ci_6 alkyl, Ci-6 alkenyl, Ci_6 alkynyl, -CH2PI1, -0(CH 2 )o-iPh, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.

[0052] Any hydrogen atom present in a compound may be substituted with deuterium isotope, unless otherwise noted.

[0053] The term "oxo" refers to the group =0. [0054] The term "moiety" refers to a portion of a compound or molecule.

[0055] Certain compounds of the present invention may exist in particular geometric or stereoisomeric forms. The present invention contemplates all such compounds, including cis- and trans-isomers, R- and S-enantiomers, diastereomers, (D)-isomers, (L)-isomers, the racemic mixtures thereof, and other mixtures thereof, as falling within the scope of the invention.

Additional asymmetric carbon atoms may be present in a substituent such as an alkyl group. All such isomers, as well as mixtures thereof, are intended to be included in this invention.

[0056] If, for instance, a particular enantiomer of a compound of the present invention is desired, it may be prepared by asymmetric synthesis, or by derivatizing with a chiral auxiliary, where the resulting diastereomeric mixture is separated and the auxiliary group cleaved to provide the pure desired enantiomers. Alternatively, where the molecule contains a basic functional group, such as amino, or an acidic functional group, such as carboxyl, diastereomeric salts are formed with an appropriate optically-active acid or base, followed by resolution of the diastereomers thus formed by fractional crystallization or chromatographic means well known in the art, and subsequent recovery of the pure enantiomers.

[0057] Unless otherwise indicated, when a disclosed compound is named or depicted by a structure without specifying the stereochemistry and has one or more chiral centers, it is understood to represent all possible stereoisomers of the compound, as well as enantiomeric mixtures thereof.

[0058] As used herein, the terms "subject" and "patient" refer to organisms to be treated by the methods of the present invention. Such organisms preferably include, but are not limited to, mammals (e.g. , murines, simians, equines, bovines, porcines, canines, felines, and the like), and more preferably include humans.

[0059] As used herein, the term "effective amount" refers to the amount of a compound (e.g. , a compound of the present invention) sufficient to effect beneficial or desired results. An effective amount can be administered in one or more administrations, applications or dosages and is not intended to be limited to a particular formulation or administration route. As used herein, the term "treating" includes any effect, e.g., lessening, reducing, modulating, ameliorating or eliminating, that results in the improvement of the condition, disease, disorder, and the like, or ameliorating a symptom thereof.

[0060] As used herein, the term "pharmaceutical composition" refers to the combination of an active agent with a carrier, inert or active, making the composition especially suitable for diagnostic or therapeutic use in vivo or ex vivo. [0061] As used herein, the term "pharmaceutically acceptable salt" refers to any

pharmaceutically acceptable salt (e.g. , acid or base) of a compound of the present invention which, upon administration to a subject, is capable of providing a compound of this invention or an active metabolite or residue thereof. As is known to those of skill in the art, "salts" of the compounds of the present invention may be derived from inorganic or organic acids and bases. Examples of acids include, but are not limited to, hydrochloric, hydrobromic, sulfuric, nitric, perchloric, fumaric, maleic, phosphoric, glycolic, lactic, salicylic, succinic, toluene-p-sulfonic, tartaric, acetic, citric, methanesulfonic, ethanesulfonic, formic, benzoic, malonic,

naphthalene-2-sulfonic, benzenesulfonic acid, and the like. Other acids, such as oxalic, while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable acid addition salts.

[0062] Examples of bases include, but are not limited to, alkali metals (e.g. , sodium) hydroxides, alkaline earth metals (e.g. , magnesium) hydroxides, ammonia, and compounds of formula NW 4 + hydroxide, wherein W is Ci_ 4 alkyl, and the like.

[0063] Examples of salts include, but are not limited to: acetate, adipate, alginate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, fumarate,

glucoheptanoate, glycerophosphate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, methanesulfonate,

2-naphthalenesulfonate, nicotinate, oxalate, palmoate, pectinate, persulfate, phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, thiocyanate, tosylate, undecanoate, and the like. Other examples of salts include anions of the compounds of the present invention compounded with a suitable cation such as Na + , NH 4 + , and NW 4 + (wherein W is a Ci_ 4 alkyl group), and the like.

[0064] For therapeutic use, salts of the compounds of the present invention are contemplated as being pharmaceutically acceptable. However, salts of acids and bases that are

non-pharmaceutically acceptable may also find use, for example, in the preparation or purification of a pharmaceutically acceptable compound.

[0065] The term "pharmaceutically acceptable" as used herein refers to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.

[0066] The term "pharmaceutically acceptable carrier" as used herein refers to any of the standard pharmaceutical carriers, such as a phosphate buffered saline solution, water, emulsions (e.g. , such as an oil/water or water/oil emulsions), and various types of wetting agents. The compositions also can include stabilizers and preservatives. For examples of carriers, stabilizers and adjuvants, see, e.g., Martin, Remington's Pharmaceutical Sciences, 15th Ed., Mack Publ. Co., Easton, PA [1975].

[0067] The terms "inhibitor" or "antagonist" refer to a molecule, for example, a small molecule, that reduces or inhibits the activity of a target molecule, for example, an enzyme such as IDO. Similarly, the terms "inhibit" or "antagonize" refer to the reduction or inhibition of activity of a target molecule, for example, an enzyme such as IDO.

[0068] Throughout the description, where compositions and kits are described as having, including, or comprising specific components, or where processes and methods are described as having, including, or comprising specific steps, it is contemplated that, additionally, there are compositions and kits of the present invention that consist essentially of, or consist of, the recited components, and that there are processes and methods according to the present invention that consist essentially of, or consist of, the recited processing steps.

[0069] As a general matter, compositions specifying a percentage are by weight unless otherwise specified. Further, if a variable is not accompanied by a definition, then the previous definition of the variable controls.

II. Macrocyclic Compounds

[0070] In one aspect, the invention provides a compound of Formula I:

or a pharmaceutically acceptable salt thereof, wherein: ring A is selected from phenylene, pyridinylene, or pyrrolidinylene, wherein ring A is connected through adjacent ring atoms to moiety X and to the adjacent carbonyl and is optionally substituted with, for example, is optionally substituted with one or more, for example, one, two, or three occurrences of, halogen, deuterium, hydroxyl, haloalkyl, heteroalkyl, Ci-C 6 alkyl, -CN, -N(R 10 ) 2 , C 3 -C 6 carbocyclyl, -C(0)-0-(Ci-C 5 alkyl), -C(0)-0-(C 2 -C 4 alkenyl), aryl, or heteroaryl;

X is -O- or -CH 2 -;

Y is a bond or -C(R l la )(R l lb );

R 1 is selected from *-N(R 10 )-C(O , *-N(R 10 )-CH 2 -, *-N(R 10 )-C(O)-CH 2 -, -CH 2 -CH 2 - or nitrogen-containing heteroarylene, wherein "*" represents a portion of R 1 bound to R 2 ;

R 2 is selected from -(C5-C1 0 alkylene)-†, -(C5-C1 0 alkenylene)-†, -(C1-C3 alkylene)- phenylene-0-(C C 3 alkylene)-†, -(C 2 -C 3 alkylene)-0-(C 2 -C 5 alkylene)-†, -(C 2 -C 3 alkylene)-0- (C 2 -C 3 alkylene)-0-(C 2 -C 3 alkylene)-†, -(C 2 -C 3 alkylene)-N(R 7e )-(C 2 -C 3 alkylene)-†, -(C 2 -C 3 alkylene)-heteroarylene-(Ci-C 3 alkylene)-†, or -(C 2 -C 3 alkylene)-heterocyclylene-(Ci-C 3 alkylene)-†, wherein "†" represents a portion of R 2 bound to R 3 ; and wherein each alkylene, alkenylene, phenylene, heteroarylene, and heterocyclylene portion of R 2 is optionally substituted with, for example, is optionally substituted with one or more, for example, one, two, or three occurrences of, Ci-C 6 alkyl, C 3 -C 6 carbocyclyl, halogen, -OH, -N(R 10 ) 2 , aryl, heteroaryl, heterocyclyl, heteroalkyl, -COOH, -C(0)0-(Ci-C 5 alkyl), -C(0)-0-(C 2 -C 4 alkenyl), -C(0)-NR 10 - (C1-C5 alkyl), -C(O)-NR 10 -(Ci-C 5 alkylene)-0-(Ci-C 5 alkyl), -0-(d-C 5 alkyl), -(C1-C5 alkyl)- OH, or haloalkyl;

R 3 is selected from -CH 2 -, **-CH 2 -N(R 10 )-, **-C(0)N(R 7d )-, **-(CH 2 ) 0 - 2 -NR 7d C(O)-, -NR 7d -, **-C(0)0-, heterocyclylene, heteroarylene, or phenylene, wherein "**" represents a portion of R 3 bound to C(R 4a )(R 4b )†; and wherein each heterocyclylene, heteroarylene, or phenylene is optionally substituted with, for example, are optionally substituted with, one or more, for example, one, two, or three occurrences of, Ci-C 6 alkyl, C 3 -C 6 carbocyclyl, hydroxyl, - N(R 10 ) 2 , or halogen;

each of R 4a and R 4b is independently selected from hydrogen, deuterium, halogen or Ci- C 4 alkyl optionally substituted with, for example, optionally substituted with one or more, for example, one, two, or three occurrences of, Ci-C 4 alkyl, C 3 -C 6 carbocyclyl, or halogen;

R 5a is selected from hydrogen, deuterium or Ci-C 4 alkyl optionally substituted with, for example, optionally substituted with one or more, for example, one, two, or three occurrences of, Ci-C 6 alkyl, C 3 -C 6 carbocyclyl, hydroxyl, -N(R ) 2 , or halogen;

R 5b is selected from hydrogen, deuterium, Ci-C 6 alkyl, -O-C1-C 6 alkyl, -CH2-heteroaryl, -CH 2 -aryl, -(CH 2 )o-i-carbocyclyl, or -(CH 2 )o-i-heterocyclyl, wherein any alkyl, -CH 2 -heteroaryl, -CH 2 -aryl, -(CH 2 )o-i-carbocyclyl, or -(CH 2 )o-i-heterocyclyl portion of R 5b is optionally substituted with, for example, is optionally substituted with one or more, for example, one, two, or three occurrences of, Ci-C 6 alkyl, C 3 -C 6 carbocyclyl, halogen, haloalkyl, -C(0)-0-(Ci-C5 alkyl), -C(0)-0-(C 2 -C 4 alkenyl), -0-(Ci-C 5 alkyl), -CN, -OH, -N(R 10 ) 2 , heteroaryl, aryl, or heteroalkyl;

R 6 is selected from hydrogen, C1-C1 0 alkyl, CH 2 -C 3 -C 6 cycloalkyl, or benzyl, wherein R 6 is optionally substituted with, for example, is optionally substituted with one or more, for example, one, two, or three occurrences of, Ci-C 6 alkyl, C 3 -C 6 carbocyclyl, hydroxyl, -N(R 10 ) 2 , or halogen;

R 7a is selected from hydrogen, C1-C4 alkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, -CH 2 -aryl, -CH 2 heteroaryl, C 3 -C 6 cycloalkyl, or heterocyclyl, wherein R 7a is optionally substituted with, for example, is optionally substituted with one or more, for example, one, two, or three occurrences of, Ci-C 6 alkyl, C 3 -C 6 carbocyclyl, -C(0)-0-(Ci-C 5 alkyl), -C(0)-0-(C 2 -C 4 alkenyl), hydroxyl, - N(R 10 ) 2 , or halogen;

each of R 7b , R 7c , R 7d and R 7e is independently selected from hydrogen or Ci-C 4 alkyl;

R 8 is selected from hydrogen, deuterium or Ci-C 4 alkyl optionally substituted with, for example, optionally substituted with one or more, for example, one, two, or three occurrences of, Ci-C 6 alkyl, C 3 -C 6 carbocyclyl, halogen, -OH, -N(R 10 ) 2 , heteroaryl, aryl, or heteroalkyl; or

R 7b and R 8 , when taken together with atoms to which they are bound, form a

heterocyclylene comprising 0-1 additional heteroatoms selected from N, O or S, wherein said heterocyclylene is optionally substituted with, for example, is optionally substituted with one or more, for example, one, two, or three occurrences of, Ci-C 6 alkyl, C 3 -C 6 carbocyclyl, halogen, oxo, -OH, -N(R 10 ) 2 , -C(0)-0-(Ci-C 5 alkyl), -C(0)-0-(C 2 -C 4 alkenyl), heteroaryl, aryl, haloalkyl, or heteroalkyl;

R 9 is hydrogen, deuterium or methyl;

each R 10 is independently selected from hydrogen, -(Ci-C 4 alkyl), -C(0)-0-(C 2 -C 4 alkenyl), -C(0)-0-(d-C 5 alkyl), or C 3 -C 6 cycloalkyl; and

each of R lla and R llb is independently selected from hydrogen, halogen, or Ci-C 4 alkyl optionally substituted with, for example, optionally substituted with one or more, for example, one, two, or three occurrences of, Ci-C 6 alkyl, C3-C6 carbocyclyl, hydroxyl, -N(R ) 2 , or halogen; and

represents a bond between two adjacent ring atoms in ring A.

[0071] Definitions of the variables in Formula I above encompass multiple chemical groups. The application contemplates embodiments where, for example, (i) the definition of a variable is a single chemical group selected from those chemical groups set forth above, (ii) the definition is a collection of two or more of the chemical groups selected from those set forth above, and (iii) the compound is defined by a combination of variables in which the variables are defined by (i) or (ii), e.g., such as where A is optionally substituted phenylene, X is -0-, Y is a bond, R 1 is *-N(R 10 )-C(O or *-N(R 10 )-CH 2 -, R 2 is -(C 5 -C 10 alkylene)-†, R 3 is **-C(0)N(R 7d or heteroarylene, R 5b is hydrogen, Ci-C 6 alkyl, -CH 2 -aryl, or -CH2-heteroaryl, R 6 is C1-C1 0 alkyl, R 7a is C C 4 alkyl, R 7b and R 8 are taken together to form an optionally substituted

heterocyclylene, and each R 10 is hydrogen or -Ci-C 4 alkyl.

[0072] In certain embodiments, R 1 is *-N(R 10 )-C(O)- or *-N(R 10 )-CH 2 -.

[0073] In certain embodiments, R 1 is selected from *-NH-C(0 , *-N(CH 3 )-C(0)-,

*-N(CH 3 )-CH 2 -, *-N(CH(CH 3 ) 2 )- -, *-NH-CH 2 -, *-N(cyclopropyl)-CH 2 -,

*N(C(0)OCH 2 CH=CH 2 )-C(0)-,

[0074] In certain embodiments, R 2 is selected from -(C5-C1 0 alkylene)-†, -(C5-C1 0

alkenylene)-†, -(C1-C3 alkylene)-phenylene-0-(Ci-C3 alkylene)-†, -(C 2 -C 3 alkylene)-0-(C 2 -C5 alkylene)-†, -(C 2 -C 3 alkylene)-0-(C 2 -C 3 alkylene)-0-(C 2 -C 3 alkylene)-†, -(C 2 -C 3 alkylene)- N(R 7e )-(C 2 -C 3 alkylene)-†, -(C 2 -C 3 alkylene)-heteroarylene-(Ci-C 3 alkylene)-†, or -(C 2 -C 3 alkylene)-heterocyclylene-(Ci-C 3 alkylene)-†, wherein "†" represents a portion of R 2 bound to

[0075] In certain embodiments, R 2 is -(C 3 -Cio alkylene)-†.

[0076] In certain embodiments of Formula I, R 2 is selected from -(CH 2 ) 5 _ 9 -, -CH(CH 3 )-(CH 2 ) 7 -

†, -CH(CH(CH 3 ) 2 )-(CH 2 ) 7 -†, -CH(CH 3 )-CH=CH-(CH 2 ) 5 -†, -CH 2 -CHOH-(CH 2 ) 6 -†, -(CH 2 ) 2 -0- (CH 2 ) 5 -†, -(CH 2 ) 2 -0-(CH 2 ) 2 -0-(CH 2 ) 2 -†, -(CH 2 ) 3 -0-(CH 2 ) 3 -†, -(CH 2 ) 3 -N(CH 3 )-(CH 2 ) 3 -†, -

[0077] In certain embodiments of Formula I, R 3 is **-C(0)N(R 7d , **-(CH 2 )o- 2 -NR 7d C(0)-, or heteroarylene optionally substituted with by 1 or 2 substituents independently selected from the group consisting of halogen, hydroxyl, and Ci-C 6 alkyl.

[0078] In certain embodiments of Formula I, R 3 is selected from -CH 2 -, **-C(0)-N(CH 3 )-, **- -NH-, **-(CH 2 ) 2 -NH-C(0)- **-CH 2 -NH-C(0)-, -NH-, **-C(0)0-,

[0079] In certain embodiments of Formula I, R a or R is hydrogen; and the other is selected from hydrogen and -CH 3 . [0080] In certain embodiments of Formula I, R 5a is hydrogen or -CH 3 , and R 5b is hydrogen, Ci- C 6 alkyl, -CH2-heteroaryl or -CH2-aryl, wherein said aryl or heteroaryl are optionally substituted with 1, 2, or 3 substituents independently selected from the group consisting of halogen, hydroxyl, Ci-C 6 alkyl, and C 3 -C6 cycloalkyl.

[0081] In certain embodiments of Formula I, R 5a is selected from hydrogen or -CH 3 ; and R 5b is selected from hydrogen, -CH 3 , -CH 2 CH(CH 3 ) 2 , -CH(CH 3 )CH 2 CH 3 , -CH 2 -0-C(CH 3 ) 3 , -CH(OH)CH 3 , -CH(CH 3 ) 2 , -CH(CH 3 )-0-C(CH 3 ) 3 , -CH 2 C(CH 3 ) 3 , cyclohexyl,

cyclopropylmethyl, cyclobutylmethyl, benzyl, pyridin-2-ylmethyl, pyridin-3-ylmethyl, pyridin- 2-ylmethyl, 5-fluororpyridin-3-ylmethyl, 2,4-difluoropyridin-3-ylmethyl, 4-methylpyridin-3- ylmethyl, 2-methylpyridin-3-ylmethyl, 2-hydroxypyridin-3-ylmethyl, phenylethan-l-yl, 1- oxopyridin-3-ylmethyl, 3,4-difluorophenylmethyl, 4-hydroxyphenylmethyl, 2- fluorophenylmethyl, 3-fluorophenylmethyl, 4-fluorophenylmethyl, 4-cyanophenylmethyl, 4- trifluoromethylphenylmethyl, 4-bromophenylmethyl, 4-chlorophenylmethyl, 4- methoxyphenylmethyl, pyrazin-2-ylmethyl, thiazol-4-ylmethyl, thiophen-3 -ylmethyl, pyrimidin- 4-ylmethyl, benzo[b]thiophen-3-ylmethyl, l-methyl-lH-indol-3-ylmethyl, or 1- butoxycarbonylpiperidin-4-ylmethyl.

[0082] In certain embodiments of Formula I, R 6 is C1-C5 alkyl.

[0083] In certain embodiments of Formula I, R 6 is selected from -CH(CH 3 ) 2 , -CH 2 CH(CH 3 ) 2 , - C(CH 3 ) 3 , cyclopropyl, cyclobutyl, cyclohexyl, or phenyl.

R x

[0084] In certain embodiments of Formula I, R is ^ , wherein R x is selected from C1-C9 alkyl, C 3 -C 6 cycloalkyl, or phenyl, wherein R x is optionally substituted with, for example, is optionally substituted with one or more, for example, one, two, or three occurrences of Ci-C 6 alkyl, C 3 -C 6 carbocyclyl, -OH, -N(R 10 ) 2 , or halogen.

R x

[0085] In certain embodiments of Formula I, R is ^ , wherein R x is selected from C1-C4 alkyl, C 3 -C 6 cycloalkyl, or phenyl.

[0086] In certain embodiments of Formula I, R 7a is C1-C4 alkyl.

[0087] In certain embodiments of Formula I, R 7a is selected from -CH 3 , -CH 2 C≡CH, cyclopropyl, cyclohexyl, benzyl, 4-hydroxyphenylmethyl, furan-2-ylmethyl, or 1-tert- butyloxycarbonylpiperidin-4-yl. [0088] In certain embodiments of Formula I, R 7b and R 8 are taken together with atoms to which they are bound to form a 5-6 membered, saturated heterocyclylene optionally substituted with 1, 2, or 3 substituents independently selected from the group consisting of halogen, hydroxyl, Ci-C 6 alkyl, and C 3 -C 6 cycloalkyl.

[0089] In certain embodiments of Formula I, R 7b is -CH 3 ; and R 8 is selected from -CH 3 , CH 2 OH, or -CH 2 OC(CH 3 ) 3 ; or R 7b and R 8 , when taken together with atoms to which they are bound, form a saturated heterocyclylene ring selected from pyrrolidin-l,2-diyl, piperidin-1,2- diyl, piperazin-l,2-diyl, morpholin-3,4-diyl, or thiomorpholin-3,4-diyl, wherein said saturated heterocyclylene ring is optionally substituted with, for example, is optionally substituted with one or more, for example, one, two, or three occurrences of Ci-C 6 alkyl, C 3 -C 6 carbocyclyl, halogen, -OH, -N(R 10 )2, heteroaryl, aryl, heteroalkyl, haloalkyl, oxo, -C(0)-0-(Ci-C5 alkyl), or - C(0)-0-(C 2 -C 4 alkenyl).

[0090] In certain embodiments of Formula I, R 7b and R 8 , when taken together with atoms to which they are bound, form pyrrolidin-l,2-diyl, 4-hydroxypyrrolidin-l,2-diyl, 4,4- difluoropyrrolidin-l,2-diyl, 4-oxopyrrolidin-l,2-diyl, 4-hydroxypyrrolidin-l,2-diyl, 4- dimethylaminopyrrolidin- 1 ,2-diyl, 4-methylaminopyrrolidin- 1 ,2-diyl, 4-fluoropyrrolidin- 1 ,2- diyl, piperidin-l,2-diyl, piperazin-l,2-diyl, morpholin-3,4-diyl, l-oxothiomorpholin-3,4-diyl, l, l-dioxothiomorpholin-3,4-diyl, or 4-tert-butyloxycarbonylpiperazin-l,2-diyl.

[0091] In certain embodiments of Formula I, R 7c is independently selected from hydrogen or

-CH 3 .

[0092] In certain embodiments of Formula I, R 9 is selected from hydrogen or -CH 3 .

[0093] In certain embodiments of Formula I, ring A is selected from:

wherein "1" represents a portion of ring A bound to moiety X.

[0094] In certain embodiments of Formula I, ring A is or , wherein

"1" represents a portion of ring A bound to moiety X.

[0095] In certain embodiments of Formula I, Y is selected from a bond or -CH2-.

[0096] In certain embodiments of Formula I, Y is a bond.

[0097] In certain embodiments of Formula I, X is O.

[0098] It is understood that one or more of the variables of Formula I as discussed hereinabove in each of the embodiments (e.g., A, X, R 1 , R 2 , R 3 , R 4a ,R 4b , R 5a , R 5b , R 6 , R 7a , R 7b , R 7c , R 8 , and R 9 ) may be combined with one another, in any combination.

[0099] In certain embodiments, the compound is represented by Formula I-A:

(I-A)

or a pharmaceutically acceptable salt thereof, wherein:

R 1 is *-N(R 10 )-C(O or *-N(R 10 )-CH 2 -, where"*" represents a portion of R 1 bound to R 2 ; R 2 is -(C5-C1 0 alkylene)-;

R 3 is **-C(0)N(R 7d )- or heteroarylene optionally substituted by 1 or 2 substituents independently selected from the group consisting of halogen, hydroxyl, and Ci-C 6 alkyl, wherein "**" represents a portion of R 3 bound to C(R 4a )(R 4b )†;

each of R 4a and R 4b are independently hydrogen, halogen, or C1-C4 alkyl; R 5a is hydro gen or C 1 -C 4 alkyl;

R 5b is hydrogen, Ci-C 6 alkyl, -CH 2 -heteroaryl, or -CH 2 -aryl, wherein said heteroaryl and aryl are optionally substituted by 1 or 2 substituents independently selected from the group consisting of halogen, hydroxyl, and Ci-C 6 alkyl;

R 6 is C1-C10 alkyl;

R 7a is C 1 -C 4 alkyl;

R 7d is hydrogen or C 1 -C 4 alkyl;

7b 8

R' u and R° are taken together with atoms to which they are bound, form a 5-7 membered, saturated heterocyclylene optionally substituted by 1 or 2 substituents independently selected from the group consisting of halogen, hydroxyl, and Ci-C 6 alkyl;

R 9 is hydrogen or methyl;

R 10 is hydrogen or C 1 -C 4 alkyl; and

R 12 represents independently for each occurrence halogen, hydroxyl, or Ci-C 6 alkyl; and n is 0, 1, or 2.

[00100] It is understood that each of the variables of Formula I-A may be selected from any one or more the embodiments noted above for Formula I-A.

[00101] In another aspect, the invention provides a compound of Formula (II):

(Π),

or a pharmaceutically acceptable salt thereof, wherein:

ring A is selected from phenylene, pyridinylene, or pyrrolidinylene, wherein ring A is connected through adjacent ring atoms to moiety X and to the adjacent carbonyl and is optionally substituted with, for example, is optionally substituted with one or more, for example one, two, or three occurrences of, halogen, deuterium, hydroxyl, haloalkyl, heteroalkyl, Ci-C 6 alkyl, -CN, -N(R 1U ) 2 , C 3 -C 6 carbocyclyl, -C(0)-0-(Ci-C 5 alkyl), -C(0)-0-(C 2 -C 4 alkenyl), aryl, or heteroaryl;

X is -O- or -CH 2 -;

Y is a bond or -C(R l la )(R l lb );

Z is a heteroarylene or a phenylene, wherein Z is optionally substituted with, for example, is optionally substituted with one or more, for example, one, two, or three occurrences of, Ci-C 6 alkyl, C3-C6 carbocyclyl, halogen, -OH, -N(R 10 )2, heteroaryl, aryl, heteroalkyl, or haloalkyl;

R 1 is selected from *-N(R 10 )-C(O , *-N(R 10 )-CH 2 -, *-N(R 10 )-C(O)-CH 2 -, -CH 2 -CH 2 -, *- C(O)-N(R 10 )-CH 2 -, or a nitrogen-containing heteroarylene, wherein "*" represents a portion of R 1 bound to R 2 ;

R 2 is selected from -(C5-C1 0 alkylene)-†, -(C5-C1 0 alkenylene)-†, -(C1-C3 alkylene)- phenylene-0-(Ci-C 3 alkylene)-†, -(C 2 -C 3 alkylene)-0-(C 2 -C 5 alkylene)-†, -(C 2 -C 3 alkylene)-0- (C 2 -C 3 alkylene)-0-(C 2 -C 3 alkylene)-†, -(C 2 -C 3 alkylene)-N(R 7e )-(C 2 -C 3 alkylene)-†, -(C 2 -C 3 alkylene)-heteroarylene-(Ci-C 3 alkylene)-†, and -(C 2 -C 3 alkylene)-heterocyclylene-(Ci-C 3 alkylene)-†, wherein "†" represents a portion of R 2 bound to R 3 ; and wherein each alkylene, alkenylene, phenylene, heteroarylene, or heterocyclylene portion of R 2 is optionally substituted with, for example, is optionally substituted with one or more, for example, one, two, or three occurrences of, Ci-C 6 alkyl, C 3 -C 6 carbocyclyl, halogen, -OH, -N(R 10 ) 2 , aryl, heteroaryl, heterocyclyl, heteroalkyl, -COOH, -C(0)0-(d-C 5 alkyl), -C(0)-0-(C 2 -C 4 alkenyl), -C(0)-NR 10 - (C1-C5 alkyl), -C(O)-NR 10 -(Ci-C 5 alkylene)-0-(Ci-C 5 alkyl), -0-(d-C 5 alkyl), or (C1-C5 alkyl)- OH;

R 3 is selected from -CH 2 -, **-CH 2 -N(R 10 )-, **-C(0)N(R 7d )-, **-(CH 2 ) 0 - 2 -NR 7d C(O)-, -NR 7d -, **-C(0)0-, heterocyclylene, heteroarylene, or phenylene, wherein "**" represents a portion of R 3 bound to C(R 4a )(R 4b )†; and wherein said heterocyclylene, heteroarylene, and phenylene are optionally substituted with, for example, are optionally substituted with one or more, for example, one, two, or three occurrences of, Ci-C 6 alkyl, C 3 -C 6 carbocyclyl, hydroxyl, - N(R 10 ) 2 , or halogen;

each of R 4a and R 4b is independently selected from hydrogen, deuterium, halogen, or Ci- C 4 alkyl optionally substituted with, for example, optionally substituted with one or more, for example, one, two, or three occurrences of, Ci-C 4 alkyl, C 3 -C 6 carbocyclyl, or halogen; R 6 is selected from C1-C9 alkyl, C 3 -C 6 cycloalkyl, or phenyl, wherein R 6 is optionally substituted with, for example, is optionally substituted with one or more, for example, one, two, or three occurrences of, Ci-C 6 alkyl, C 3 -C 6 carbocyclyl, hydroxyl, -N(R 10 )2, or halogen;

each of R 7b , R 7c , R 7d and R 7e is independently selected from hydrogen or C1-C4 alkyl; R 8 is selected from hydrogen, deuterium or C1-C4 alkyl optionally substituted with, for example, optionally substituted with one or more, for example, one, two, or three occurrences of, Ci-C 6 alkyl, C 3 -C 6 carbocyclyl, halogen, -OH, -N(R 10 )2, heteroaryl, aryl, or heteroalkyl; or

7b 8

R and R , when taken together with atoms to which they are bound, form optionally substituted heterocyclylene comprising 0-1 additional heteroatoms selected from N, O or S, wherein said heterocyclylene is optionally substituted with, for example, is optionally substituted with one or more, for example, one, two, or three occurrences of, Ci-C 6 alkyl, C 3 -C 6 carbocyclyl, halogen, oxo, -OH, -N(R 10 ) 2 , -C(0)-0-(Ci-C 5 alkyl), -C(0)-0-(C 2 -C 4 alkenyl), heteroaryl, aryl, haloalkyl, or heteroalkyl;

R 9 is hydrogen, deuterium or methyl;

R 10 is selected from hydrogen, -(C C 4 alkyl), -C(0)-0-(C 2 -C 4 alkenyl), -C(0)-0-(d-C 5 alkyl), or C 3 -C 6 cycloalkyl;

each of R l la and R l lb is independently selected from hydrogen, halogen, or Ci-C 4 alkyl optionally substituted with, for example, optionally substituted with one or more, for example, one, two, or three occurrences of, Ci-C 6 alkyl, C 3 -C 6 carbocyclyl, hydroxyl, -N(R 10 )2, or halogen; and

"— " represents a bond between two adjacent ring atoms in ring A.

[00102] Definitions of the variables in Formula II above encompass multiple chemical groups. The application contemplates embodiments where, for example, (i) the definition of a variable is a single chemical group selected from those chemical groups set forth above, (ii) the definition is a collection of two or more of the chemical group selected from those set forth above, and (iii) the compound is defined by a combination of variables in which the variables are defined by (i) or (ii), e.g., such as where A is optionally substituted phenylene, X is -0-, Y is a bond, Z is an optionally substituted heteroarylene or a phenylene, R 1 is *-N(R 10 )-C(O)- or *-C(0)-N(R 10 )- CH 2 -, R 2 is -(C5-C1 0 alkylene)-†, R 3 is **-C(0)N(R 7d or heteroarylene, R 6 is C1-C9 alkyl, R 7b is Ci-C 4 alkyl and R 8 is hydrogen or C1-C4 alkyl. [00103] In certain embodiments of Formula II, Z is selected from or phen-1,3- diyl, wherein "††" represents a portion of Z bound to R 1 .

[00104] In certain embodiments, the invention provides a compound set forth in Table 1 or a salt thereof, or a pharmaceutical composition comprising a compound set forth in Table 1 or a salt thereof.

III. Methods of Synthesizing Compounds of the Invention

[00105] The compounds of the present invention can be prepared using an iterative peptide coupling procedure as illustrated in following synthetic schemes. Exemplary general synthetic protocols are presented in Schemes 1 through 4. The schemes and accompanying description of synthetic procedures are given for the purpose of illustrating the invention, and should not be construed as limiting the scope or spirit of the invention.

[00106] Abbreviations as used herein include

0-(7-azabenzotriazol-l-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HATU);

diisopropylethylamine (DIPEA); dimethylformamide (DMF); 9-fluorenylmethoxycarbonyl (Fmoc); methanol (MeOH); methylene chloride (DCM); tert-butoxycarbonyl (Boc); tert-butyl (tBu); tetrahydrofuran (THF); trifluoroacetic acid (TFA); 1,8-diazobicyclo [5.4.0]-undec-7-ene (DBU); N-methylmorpholine (NMM); l-hydroxy-7-azabenzotriazole (HOAt); phenyl (Ph); trifluoroacetic acid (TFA); triethylamine (Et 3 N); petroleum ether (PE); ethyl acetate (EA); acetic acid (AcOH); diethyl ether (Et 2 0); Boc anhydride ((Boc) 2 0); dimethylsulfoxide (DMSO); diisopropylethylamine (DIEA); N-bromosuccinimide (NBS); trityl chloride (TrtCl); triphenyl phosphine (PPh 3 ); (9H-fluoren-9-yl)methyl (2,5-dioxopyrrolidin-l-yl) carbonate (Fmoc-Osu); diisopropyl azodicarboxylate (DIAD); methanesulfonyl chloride (MsCl); room temperature (r.t. or RT); and thin-layer chromatography (TLC).

[00107] Scheme 1 depicts a general synthesis method for the compound 5a, an exemplary intermediate to compounds of Formula I, wherein R 1 is *-N(R 10 )-C(O)- or *-N(R 10 )-C(O)-CH 2 -, as defined for Formula I. A 2-chloro-trityl chloride resin 1 is combined with an appropriate protected alkylamino acetic acid 2 in DCM to form resin 3. Resin 3 is then deprotected with DBU and piperidine in DMF and then coupled to a protected amino acid 4 using HATU and NMM to produce resin 5a, which is further coupled according to Scheme 3, below. The preparation of exemplary intermediate compounds 2 are described, for example, in EXAMPLES 1, 3, 4, and 5.

5b

R =*N(R 0 )-CH 2 -, -CH 2 -CH 2 -, or a nitrogen- containing heteroarylene

[00108] Scheme 2 depicts a general synthesis method for the compound 5b, an intermediate to compounds of Formula I, wherein R 1 is *-N(R 10 )-CH 2 -, -CH 2 -CH 2 -, or a nitrogen-containing heteroarylene, as defined for Formula I. A 2-chloro-trityl chloride resin 1 is combined with an appropriate protected alkylamino acetic acid 6 in DCM containing DIEA to form resin 5b, which is further coupled according to Scheme 3, below. The preparation of exemplary compounds of 6 are described, for example, in EXAMPLES 6 and 7.

[00109] Scheme 3 depicts a general synthesis for compounds of Formula I, starting with intermediate 5a (Scheme 1) or 5b (Scheme 2). Intermediate 5a or 5b is deprotected with DBU and piperdine in DMF and then coupled to the appropriate amino acid 7. The Fmoc group of 8 is removed again with DBU and piperidine in DMF and the resulting deprotected resin is coupled to amino acid 9. The deprotection of 10 and treatment with TFA provides the salt 11, which is cyclized using HATU, HO At, DIEA and DMF to form a compound of Formula I.

[00110] Scheme 4 depicts a general synthesis method for the compound 13a, an exemplary intermediate to compounds of Formula II, wherein R 1 is *-N(R 10 )-C(O)- or *-N(R 10 )-C(O)-CH 2 -, as defined for Formula II. Resin 3 (see SCHEME 1) is deprotected with DBU and piperidine in DMF and then coupled to a protected amino acid 12 using HATU and NMM to produce resin 13a, which is further coupled according to Scheme 6, below. The preparation of exemplary intermediate compound 12 is described, for example, in EXAMPLE 8.

R 1 = * N(R 10 )-CH 2 -, -CH 2 -CH 2 -,

or a nitrogen-containing

heteroarylene

[00111] Scheme 5 depicts a general synthesis method for the compound 13b, an intermediate to compounds of Formula I, wherein R 1 is *-N(R 10 )-CH 2 -, -CH 2 -CH 2 -, or a nitrogen-containing heteroarylene, as defined for Formula I. A 2-chloro-trityl chloride resin 1 is combined with an appropriate protected alkylamino acetic acid 14 in DCM containing DIEA to form resin 13b, which is further coupled according to Scheme 6, below.

[00112] Scheme 6 depicts a general synthesis for compounds of Formula II, starting with intermediate 13a (Scheme 4) or 13b (Scheme 5). Intermediate 13a or 13b is deprotected with DBU and piperdine in DMF and then coupled to the appropriate amino acid 15. The deprotection of 16 and treatment with TFA provides the salt 17, which is cyclized using HATU, HO At, DIE A and DMF to form a compound of Formula II.

[00113] Combinations of substituents and variables contemplated by the present invention are only those that result in the formation of compounds which possess stability sufficient to allow for their manufacture and which maintain the integrity of the compound for a sufficient period of time to be useful for the purposes detailed herein (e.g., formulation into therapeutic products, intermediates for use in production of therapeutic compounds, isolatable or storable intermediate compounds, treating a disease or condition responsive to therapeutic agents).

IV. Therapeutic Applications

[00114] The invention provides compounds for use in a method of antagonizing of inhibiting IDO activity in a subject or in a biological sample comprising the step of administering to the subject or contacting the biological sample with a compound of the invention, namely a compound of Formula I (including Formula 1 A) or Formula II. [00115] The compounds of the invention can be used to treat a disease, condition, or disorder where elevated IDO enzyme activity is implicated in the disease state. For example, the compounds of the invention can be used to treat a disease, condition, or disorder where a compound of the invention reduces IDO enzyme activity, for example, upon binding to IDO. The invention provides a method of inhibiting IDO activity in a subject by administering to the subject an effective amount of a compound of the invention, for example, a compound of Formula I (including Formula 1A) or Formula II. In certain embodiments, the method is used to treat or prevent a condition selected from a proliferative or hyperproliferative disease, for example, cancer.

[00116] In particular, the invention provides for the treatment of diseases, disorders, and conditions characterized by excessive or abnormal cell proliferation. Such diseases include a proliferative or hyperproliferative disease. Examples of proliferative and hyperproliferative diseases include cancer and myeloproliferative disorders.

[00117] The invention provides a method of treating cancer in a subject in need thereof. The method comprises administering to the subject an effective amount of a compound of the invention, namely a compound of Formula I (including Formula 1 A) or Formula II, or a pharmaceutical composition comprising a compound of the invention, namely a compound of Formula I (including Formula 1A) or Formula II. In certain embodiments, cells associated with the cancer are characterized as having an elevated level of IDO activity and/or expression when compared to cells associated with non-cancerous tissue. Alternatively or in addition, the subject is characterized as having an elevated concentration of kynurenine in a body fluid, such as blood, serum or plasma, when compared to a similar body of a subject without the cancer.

[00118] In particular, it is contemplated that the compounds of the invention can be used to treat one or more of the following cancers: Oral: head and neck, including buccal cavity, lip, tongue, mouth, pharynx; Cardiac: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; Lung: Non-small cell lung carcinoma including adenocarcinoma, bronchioalveolar, squamous cell carcinoma

(basaloid, clear cell, papillary, small cell), large cell carcinoma, large cell neuroendocrine carcinoma (LCNEC); small cell lung cancer including small cell (oat cell) carcinoma, combined small cell; adenoid cystic carcinoma; hamartoma; lymphoma; neuroendocrine/carcinoid;

sarcoma; Gastrointestinal: esophagus (squamous cell carcinoma, larynx, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel or small intestines (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel or large intestines (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma), colon, colon-rectum, colorectal, rectum; Genitourinary tract: kidney (adenocarcinoma, Wilm's tumor, lymphoma, leukemia), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); Liver: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma, biliary passages; Bone: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma,

osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors; Nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma,

meningiosarcoma, gliomatosis), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinoma, glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, meningioma, glioma, sarcoma);

Female/Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian carcinoma, granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), fallopian tubes (carcinoma), breast; Hematologic: blood (myeloid leukemia [acute and chronic], acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome), Hodgkin's disease, non-Hodgkin's lymphoma, hairy cell; lymphoid disorders; Skin: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Karposi's sarcoma, keratoacanthoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis, Thyroid gland: papillary thyroid carcinoma, follicular thyroid carcinoma,

undifferentiated thyroid cancer, medullary thyroid carcinoma, multiple endocrine neoplasia type 2A, multiple endocrine neoplasia type 2B, familial medullary thyroid cancer,

pheochromocytoma, paraganglioma; and Adrenal glands: neuroblastoma.

[00119] In certain embodiments, the cancer is selected from head and neck, ovarian, melanoma cervical, endometrial, esophageal, or breast cancer. [00120] In certain embodiments, the cancer is a hematopoietic disorder, for example, acute- myelogenous leukemia (AML), chronic-myelogenous leukemia (CML), acute-promyelocytic leukemia (APL), and acute lymphocytic leukemia (ALL).

[00121] In certain embodiments, the cancer is selected from cervical, renal, endometrial, squamous cell, colorectal, prostate, head and neck, lung, large B cell lymphoma, or AML.

[00122] The invention provides a method of treating a cancer in a subject in need thereof. The method comprises administering to the subject an effective amount of a compound of the invention, namely a compound of Formula I (including Formula 1 A) or Formula II, or a pharmaceutical composition comprising a compound of the invention, namely a compound of Formula I (including Formula 1A) or Formula II, wherein cells associated with the cancer are characterized as having an elevated level of IDO gene expression relative to average IDO gene expression in cells of the same cell type without the cancer.

[00123] In such a method, a subject suitable for treatment can be identified by obtaining a cancer cell sample from the subject; determining the level of IDO gene expression in the sample; comparing the IDO gene expression level in the sample to a threshold value typically present in normal cells thereby to determine if IDO gene expression is elevated in the cancer sample. Once an elevated level of IDO gene expression has been identified in the subject, the subject can then be treated by administering an effective amount of the compound of the invention, namely a compound of Formula I (including Formula 1 A) or Formula II, or a pharmaceutical composition comprising a compound of the invention, namely a compound or Formula I (including Formula 1A) or Formula II. Exemplary gene expression assays are described, for example, in WO 2013/095793.

[00124] The expression levels of IDO can be determined using a variety of approaches known in the art. For example, a tissue or cell sample can be obtained by using conventional tumor biopsy instruments and procedures. Endoscopic biopsy, excisional biopsy, incisional biopsy, fine needle biopsy, punch biopsy, shave biopsy and skin biopsy are examples of recognized medical procedures that can be used by one of skill in the art to obtain tumor samples for use in practicing the invention. The tumor tissue sample should be large enough to provide sufficient RNA for measuring individual gene expression levels.

[00125] The tumor tissue sample can be in any form that allows quantitative analysis of gene expression or transcript abundance. In some embodiments, RNA is isolated from the tissue sample prior to quantitative analysis. Some methods of RNA analysis, however, do not require RNA extraction, e.g. , the qNPA™ technology commercially available from High Throughput Genomics, Inc. (Tucson, AZ). Accordingly, the tissue sample can be fresh, preserved through suitable cryogenic techniques, or preserved through non-cryogenic techniques. Tissue samples used in the invention can be clinical biopsy specimens, which often are fixed in formalin and then embedded in paraffin. Samples in this form are commonly known as formalin-fixed, paraffin-embedded (FFPE) tissue. Techniques of tissue preparation and tissue preservation suitable for use in the present invention are well-known to those skilled in the art.

[00126] Gene expression levels (transcript abundance) can be determined by any suitable method. Exemplary methods for measuring individual gene expression levels include quantitative RT-PCR (qRT-PCR) and qNPA™.

[00127] qRT-PCR typically is performed on RNA isolated from a sample of interest, for example, a tissue sample. Methods for rapid and efficient extraction of eukaryotic mRNA (poly(a) RNA), from tissue samples are well-established and known to those of skill in the art. See, e.g. , Ausubel et al. , 1997, Current Protocols of Molecular Biology, John Wiley & Sons. The tissue sample can be fresh, frozen or fixed paraffin-embedded (FFPE) clinical study tumor specimens. In general, RNA isolated from fresh or frozen tissue samples tends to be less fragmented than RNA from FFPE samples. FFPE samples of tumor material, however, are more readily available, and FFPE samples are suitable sources of RNA for use in methods of the present invention. For a discussion of FFPE samples as sources of RNA for gene expression profiling by RT-PCR, see, e.g., Clark-Langone et al., 2007, BMC Genomics 8:279. Also see, De Andres et al., 1995, Biotechniques 18:42044; and Baker et al., U.S. Patent Application

Publication No. 2005/0095634. The use of commercially available kits with vendor's instructions for RNA extraction and preparation is widespread and common. Commercial vendors of various RNA isolation products and complete kits include Qiagen (Valencia, CA), Invitrogen (Carlsbad, CA), Ambion (Austin, TX) and Exiqon (Woburn, MA).

[00128] Certain advantages of qRT-PCR include sensitivity, flexibility, quantitative accuracy, and ability to discriminate between closely related mRNAs. Guidance concerning the processing of tissue samples for quantitative PCR is available from various sources, including

manufacturers and vendors of commercial products for qRT-PCR (e.g., Qiagen (Valencia, CA) and Ambion (Austin, TX)). Instrument systems for automated performance of qRT-PCR are commercially available and used routinely in many laboratories. An example of a well-known commercial system is the Applied Biosystems 7900HT Fast Real-Time PCR System (Applied Biosystems, Foster City, CA). [00129] Once isolated mRNA is in hand, the first step in RT-PCR is the reverse transcription of the mRNA template into cDNA, which is then exponentially amplified in a PCR reaction. Two commonly used reverse transcriptases are avilo myeloblastosis virus reverse transcriptase (AMV-RT) and Moloney murine leukemia virus reverse transcriptase (MMLV-RT). The reverse transcription reaction typically is primed with specific primers, random hexamers, or oligo(dT) primers. Suitable primers are commercially available, e.g., GeneAmp ® RNA PCR kit (Perkin Elmer, Waltham, MA). The resulting cDNA product can be used as a template in the subsequent polymerase chain reaction.

[00130] The PCR step is carried out using a thermostable DNA-dependent DNA polymerase. The polymerase most commonly used in PCR systems is a Thermus aquaticus (Taq) polymerase. The selectivity of PCR results from the use of primers that are complementary to the DNA region targeted for amplification, i.e., regions of the cDNAs reverse transcribed from the genes of the Transcription Cluster. Therefore, when qRT-PCR is employed in the present invention, primers specific to each gene in a given Transcription Cluster are based on the cDNA sequence of the gene. Commercial technologies such as SYBR ® green or TaqMan ® (Applied Biosystems, Foster City, CA) can be used in accordance with the vendor's instructions. Messenger RNA levels can be normalized for differences in loading among samples by comparing the levels of housekeeping genes such as beta-actin or GAPDH. The level of mRNA expression can be expressed relative to any single control sample such as mRNA from normal, non-tumor tissue or cells. Alternatively, it can be expressed relative to mRNA from a pool of tumor samples, or tumor cell lines, or from a commercially available set of control mRNA.

[00131] PCR primer sets for practicing the disclosed methods can be purchased from commercial sources, e.g., Applied Biosystems. PCR primers preferably are about 17 to 25 nucleotides in length. Primers can be designed to have a particular melting temperature (Tm), using conventional algorithms for Tm estimation. Software for primer design and Tm estimation are available commercially, e.g., Primer Express™ (Applied Biosystems). By applying established principles of PCR primer design, a large number of different primers can be used to measure the expression level of any given gene.

[00132] In addition, an example of a method for determining expression levels of a gene of interest without performing an RNA extraction step is the quantitative nuclease protection assay (qNPA™), which is commercially available from High Throughput Genomics, Inc. (aka "HTG"; Tucson, AZ). In the qNPA method, samples are treated in a 96-well plate with a proprietary Lysis Buffer (HTG), which releases total RNA into solution. Gene-specific DNA oligonucleotides, i.e. , specific for each gene of interest, are added directly to the Lysis Buffer solution, and they hybridize to the RNA present in the Lysis Buffer solution. The DNA oligonucleotides are added in excess, to ensure that all RNA molecules complementary to the DNA oligonucleotides are hybridized. After the hybridization step, S 1 nuclease is added to the mixture. The S 1 nuclease digests the non-hybridized portion of the target RNA, all of the non- target RNA, and excess DNA oligonucleotides. Then the SI nuclease enzyme is inactivated. The RNA:: DNA heteroduplexes are treated to remove the RNA portion of the duplex, leaving only the previously protected oligonucleotide probes. The surviving DNA oligonucleotides are a stoichiometrically representative library of the original RNA sample. The qNPA oligonucleotide library can be quantified using the ArrayPlate Detection System (HTG).

[00133] It is contemplated that the expression level of IDO can be determined using a variety of accurate and reliable systems, including protocols, reagents and instrumentation that are commercially available. Selection and use of a suitable system for generating gene expression data for use in the methods described herein is a design choice, and can be accomplished by a person of skill in the art, without undue experimentation.

[00134] In addition, the invention provides a method of treating a cancer in a subject in need thereof. The method comprises administering to the subject an effective amount of a compound of the invention, namely a compound of Formula I (including Formula I- A) or Formula II, or a pharmaceutical composition comprising a compound of the invention, namely a compound or Formula I (Formula I- A) or Formula II, wherein the subject has a kynurenine level in a body fluid that is elevated relative the average serum kynurenine level in a similar body fluid harvested from subjects without the cancer. Examplary body fluids include, for example, whole blood or blood products such as plasma or serum.

[00135] In such a method, a subject suitable for treatment can be identified as having an elevated concentration of kynurenine in a body fluid. This can be accomplished by obtaining a body fluid sample from the subject; determining the concentration of kynurenine in the fluid sample; and comparing the kynurenine concentration in the fluid sample against a threshold value representive of the average concentration of kynurenine present in the same type of body fluid of subjects without the cancer so as to determine if the kynurenine is elevated in the fluid sample of the subject being tested. Once an elevated concentration of kynurenine has been identified in the subject, the subject may then be treated by administering to the subject an effective amount of a compound of Formula I (including Formula I- A) or Formula II, or a pharmaceutical composition comprising a compound of the invention, namely a compound or Formula I (including Formula I- A) or Formula II.

[00136] It is contemplated that the kynurenine concentration (as well as tryptophan

concentration) can be determined in a sample (tissue or body fluid) of interest using standard methodologies known in the art, for example, by high pressure liquid chromatography (HPLC) (see, Widner et al. (1997) CLINICAL CHEM., vol. 43, no. 2: 2424-2426; Laich et al. (2002)

CLINICAL CHEM., vol. 48, no. 3: 579-581), or HPLC-Tandem mass spectometry (see, de Jong et al. (2009) J. CHEMOTHERAPY B, vol. 877: 603-609), spectroscopy (see, Tokikawa et al. (1988) J. BIOL. CHEM., 263: 2041-2048; U.S. Publication No. 2016/0120857).

[00137] The invention provides compounds that are useful for the treatment of other diseases, disorders, and conditions, for example, viral disease, sepsis, pneumonia, bacteremia, trauma, tuberculosis, parasitic disease, neuroinflammation, schizophrenia, depression, neurodegenerative disease, and pain. In certain embodiments, the neurodegenerative disease is selected from Alzheimer's disease, Parkinson's disease, Amyotrophic Lateral Sclerosis (ALS), Dementia, multiple Sclerosis, and Huntington's disease. In certain embodiments, the viral disease is selected from Human Immunodeficiency Virus (HIV), Hepatitis A-D, Human Papilloma Virus (HPV), and Herpes, including Herpes Simplex I and II, as well as the Epstein Barr Virus.

[00138] Additionally, it is contemplated that one or more compounds of the invention can be used in the manufacture of a medicament for the treatment of at least one of the aforementioned disorders.

[00139] The invention also provides for combination therapies using a macrocyclic compound described herein and a second therapeutic agent. Combination therapy (or co-therapy) includes the administration of a macrocyclic compound described herein and at least a second agent as part of a specific treatment regimen intended to provide the beneficial effect from the co-action of these therapeutic agents. The beneficial effect of the combination includes, but is not limited to, pharmacokinetic or pharmacodynamic co-action resulting from the combination of therapeutic agents. Administration of these therapeutic agents in combination typically is carried out over a defined time period (usually minutes, hours, days or weeks depending upon the combination selected).

[00140] Combination therapy is intended to embrace administration of these therapeutic agents in a sequential manner, that is, wherein each therapeutic agent is administered at a different time, as well as administration of these therapeutic agents, or at least two of the therapeutic agents, in a substantially simultaneous manner. Substantially simultaneous administration can be accomplished, for example, by administering to the subject a single dosage form having a fixed ratio of each therapeutic agent or in multiple, single dosage forms for each of the therapeutic agents. Sequential or substantially simultaneous administration of each therapeutic agent can be effected by any appropriate route including, but not limited to, oral routes, intravenous routes, intramuscular routes, and direct absorption through mucous membrane tissues. The therapeutic agents can be administered by the same route or by different routes. For example, a first therapeutic agent of the combination selected may be administered by intravenous injection while the other therapeutic agents of the combination may be administered orally. Alternatively, for example, all therapeutic agents may be administered orally or all therapeutic agents may be administered by intravenous injection. Combination therapy also can embrace the

administration of the therapeutic agents as described above in further combination with other biologically active ingredients and non-drug therapies (e.g., surgery or radiation treatment.) Where the combination therapy further comprises a non-drug treatment, the non-drug treatment may be conducted at any suitable time so long as a beneficial effect from the co-action of the combination of the therapeutic agents and non-drug treatment is achieved. For example, in appropriate cases, the beneficial effect is still achieved when the non-drug treatment is temporally removed from the administration of the therapeutic agents, perhaps by days or even weeks.

[00141] Accordingly, the invention provides a method treatment, as described above, further comprising an additional step of administering to the subject an additional therapeutic agent selected from a chemotherapeutic or anti-proliferative agent, an anti-inflammatory agent, an immunomodulatory or immunosuppressive agent, a neurotrophic factor, an agent for treating cardiovascular disease, an agent for treating destructive bone disorders, an anti- viral agent, an agent for treating blood disorders, or an agent for treating immunodeficiency disorders, wherein the additional therapeutic agent is appropriate for the disease being treated.

[00142] In certain embodiments, the additional therapeutic agent is administered together with the compound of Formula I (including Formula I- A) or II or a composition containing the compound of Formula I (including Formula I- A) or II as a single dosage form. In certain embodiments, the additional therapeutic agent is administered separately from the

compound/composition of Formula I (including Formula I- A) or Formula II as part of a multiple dosage form.

[00143] In certain embodiments, the invention is directed towards a method of treating cancer in a subject in need thereof, comprising administration of a compound of Formula I (including Formula I- A) or Formula II and an additional therapeutic agent. In certain embodiments, the additional therapeutic agent is administered together with the compound of Formula I (including Formula I- A) or Formula II as a single dosage form. In certain embodiments, the additional therapeutic agent is administered separately from the compound of Formula I (including Formula I- A) or Formula II as part of a multiple dosage form.

[00144] In certain embodiments, the additional therapeutic agent is an anti-cancer agent, an anti-proliferative agent, or a chemotherapeutic agent.

[00145] In certain embodiments, the additional therapeutic agent is selected from cisplatin (Platino ® ), carboplatin (Paraplatin ® ), oxaliplatin (Eloxatin ® ), daunomycin (Daunorubicin ® , DanuoXome ® , Cerubidine ® ), doxorubicin (Adriamycin ® , Rubex ® ), epirubicin (Ellence ® ), idarubicin (Idamycin ® ), valrubicin (Valstar ® ), mitoxantrone (Novantrone ® ), paclitaxel (Taxol ® ), docetaxel (Taxotere ® ) and cyclophosphamide (Cytoxan ® ).

[00146] In other embodiments, the additional therapeutic agent is selected from anti-cancer antibody or immunoglobulin therapies or agents including, but not limited to, ipilimumab (Yervoy ® ), tremelimumab, antibodies or agents that target programmed death receptor 1 [PD-1] or programmed death ligand 1 [PD-L1], e.g., CT-011 (Curetech), BMS-936558 (Bristol-Myers Squibb), BMS-936559 (Bristol-Myers Squibb), AMP-224 (Amplimmune/Glaxo-Smithkline), pembrolizumab (Merck & Co.), MPDL3280A (Roche), MGA-271 (Macrogenics), dacarbazine, Lambrolizumab (MK-3475), MSB0010718C (MerckSerono), or MEDI-4736 (Medlmmune). In certain embodiments, the PD-1 inhibitor is pembrolizumab.

[00147] In other embodiments, the additional therapeutic agent is selected from a CTLA4 agent (e.g., ipilimumab (BMS)); GITR agent (e.g., MK-4166 (MSD)); vaccines (e.g., Nanovacc (MerckSerono), Stimuvax (MerckSerono), Sipuleucel-T (Dendron); or a SOC agent (e.g., radiation, docetaxel, Temozolomide (MSD), Gemcitibine, or Paclitaxel). In other embodiments, the additional therapeutic agent is an immune enhancer such as a vaccine, immune-stimulating antibody, immunoglobulin, agent or adjuvant including, but not limited to, sipuleucel-t

(Provenge ® ), BMS-663513 (Bristol-Myers Squibb), CP-870893 (Pfizer/VLST), anti-OX40 (AgonOX), or CDX-1127 (CellDex).

[00148] In certain embodiments, the additional therapeutic agent is an anti-PD-1 or anti-PD-Ll agent and is administered together with the compound Formula I or Formula II as a single dosage form. In certain embodiments, the additional therapeutic agent is an anti-PD- 1 or anti- PD-Ll agent and is administered separately from the compound of Formula I or Formula II as part of a multiple dosage form. In certain embodiments, the anti-PD-1 or anti-PD-Ll is administered as an intravenous infusion.

[00149] In certain embodiments, more than one additional therapeutic agents are used and are administered together with the compound of Formula I or Formula II as a single dosage form. In certain embodiments, more than one additional therapeutic agents are used and are administered separately from the compound of Formula I or Formula II as part of a multiple dosage form. In certain embodiments, the more than one additional therapeutic agents are anti-PD-1 or anti-PD- Ll agents. In certain embodiments, the anti-PD-1 or anti-PD-Ll agents are administered as an intravenous infusion.

[00150] Other cancer therapies or anticancer agents that may be used in combination with the inventive agents of the present invention include surgery, radiotherapy (e.g., gamma-radiation, neutron beam radiotherapy, electron beam radiotherapy, proton therapy, brachytherapy, low- dose radiotherapy, and systemic radioactive isotopes), immune response modifiers such as chemokine receptor antagonists, chemokines and cytokines (e.g., interferons, interleukins, tumour necrosis factor (TNF), and GM-CSF)), hyperthermia and cryotherapy, agents to attenuate any adverse effects (e.g. antimetics, steroids, anti-inflammatory agents), and other approved chemotherapeutic drugs.

[00151] A compound of the invention may also be useful for treating cancer in combination with or in addition to any of the following standard of care (SOC) therapeutic agents: abarelix (Plenaxis Depot ® ); aldesleukin (Prokine ® ); Aldesleukin (Proleukin ® ); Alemtuzumab

(Campath ® ); alitretinoin (Panretin ® ); allopurinol (Zyloprim ® ); altretamine (Hexalen ® );

amifostine (Ethyol ® ); anastrozole (Arimidex ® ); arsenic trioxide (Trisenox ® ); asparaginase

(Elspar ® ); azacitidine (Vidaza ® ); bevacuzimab (Avastin ® ); bexarotene capsules (Targretin ® ); bexarotene gel (Targretin ® ); bleomycin (Blenoxane ® ); bortezomib (Velcade ® ); busulfan intravenous (Busulfex ® ); busulfan oral (Myleran ® ); calusterone (Methosarb ® ); capecitabine

(Xeloda ® ); carboplatin (Paraplatin ® ); carmustine (BCNU ® , BiCNU ® ); carmustine (Gliadel ® ); carmustine with Polifeprosan 20 Implant (Gliadel Wafer ® ); celecoxib (Celebrex ® ); cetuximab

(Erbitux ® ); chlorambucil (Leukeran ® ); cisplatin (Platinol ® ); cladribine (Leustatin ® , 2-CdA ® ); clofarabine (Clolar ® ); cyclophosphamide (Cytoxan ® , Neosar ® ); cyclophosphamide (Cytoxan Injection ® ); cyclophosphamide (Cytoxan Tablet ® ); cytarabine (Cytosar-U ® ); cytarabine liposomal (DepoCyt ® ); dacarbazine (DTIC-Dome ® ); dactinomycin, actinomycin D

(Cosmegen ® ); Darbepoetin alfa (Aranesp ® ); daunorubicin liposomal (DanuoXome ® );

daunorubicin, daunomycin (Daunorubicin ® ); daunorubicin, daunomycin (Cerubidine ® ); Denileukin diftitox (Ontak"); dexrazoxane (Zinecard"); docetaxel (Taxotere"); doxorubicin (Adriamycin PFS ® ); doxorubicin (Adriamycin ® , Rubex ® ); doxorubicin (Adriamycin PFS Injection ® ); doxorubicin liposomal (Doxil ® ); dromostanolone propionate (Dromostanolone); dromostanolone propionate (masterone Injection ® ); Elliott's B Solution (Elliott's B Solution ® ); epirubicin (Ellence ® ); Epoetin alfa (Epogen ® ); erlotinib (Tarceva ® ); estramustine (Emcyt ® ); etoposide phosphate (Etopophos ® ); etoposide, VP- 16 (Vepesid ® ); exemestane (Aromasin ® ); Filgrastim (Neupogen ® ); floxuridine (intraarterial) (FUDR ® ); fludarabine (Fludara ® );

fluorouracil, 5-FU (Adrucil ® ); fulvestrant (Faslodex ® ); gefitinib (Iressa ® ); gemcitabine

(Gemzar ® ); gemtuzumab ozogamicin (Mylotarg ® ); goserelin acetate (Zoladex Implant ® );

goserelin acetate (Zoladex ® ); histrelin acetate (Histrelin Implant ® ); hydroxyurea (Hydrea ® ); Ibritumomab Tiuxetan (Zevalin ® ); idarubicin (Idamycin ® ); if osf amide (IFEX ® ); imatinib mesylate (Gleevec ® ); interferon alfa 2a (Roferon A ® ); Interferon alfa-2b (Intron A ® ); irinotecan (Camptosar ® ); lenalidomide (Revlimid ® ); letrozole (Femara ® ); leucovorin (Wellcovorin ® , Leucovorin ® ); Leuprolide Acetate (Eligard ® ); levamisole (Ergamisol ® ); lomustine, CCNU (CeeBU ® ); meclorethamine, nitrogen mustard (Mustargen ® ); megestrol acetate (Megace ® ); melphalan, L-PAM (Alkeran ® ); mercaptopurine, 6-MP (Purinethol ® ); mesna (Mesnex ® ); mesna (Mesnex Tabs ® ); methotrexate (Methotrexate ® ); methoxsalen (Uvadex ® ); mitomycin C

(Mutamycin ® ); mitotane (Lysodren ® ); mitoxantrone (Novantrone ® ); nandrolone phenpropionate (Durabolin-50 ® ); nelarabine (Arranon); Nofetumomab (Verluma ® ); Oprelvekin (Neumega ® ); oxaliplatin (Eloxatin ® ); paclitaxel (Paxene ® ); paclitaxel (Taxol ® ); paclitaxel protein-bound particles (Abraxane ® ); palifermin (Kepivance ® ); pamidronate (Aredia ® ); pegademase (Adagen (Pegademase Bovine) ® ); pegaspargase (Oncaspar ® ); Pegfilgrastim (Neulasta ® ); pemetrexed disodium (Alimta ® ); pentostatin (Nipent ® ); pipobroman (Vercyte ® ); plicamycin, mithramycin (Mithracin ® ); porfimer sodium (Photofrin ® ); procarbazine (Matulane ® ); quinacrine (Atabrine ® ); Rasburicase (Elitek ® ); Rituximab (Rituxan ® ); sargramostim (Leukine ® ); Sargramostim

(Prokine ® ); sorafenib (Nexavar ® ); streptozocin (Zanosar ® ); sunitinib maleate (Sutent ® ); talc (Sclerosol ® ); tamoxifen (Nolvadex ® ); temozolomide (Temodar ® ); teniposide, VM-26 (Vumon ® ); testolactone (Teslac ® ); thioguanine, 6-TG (Thioguanine ® ); thiotepa (Thioplex ® ); topotecan (Hycamtin ® ); toremifene (Fareston ® ); Tositumomab (Bexxar ® ); Tositumomab/I-131

tositumomab (Bexxar); Trastuzumab (Herceptin ® ); tretinoin, ATRA (Vesanoid ® ); Uracil

Mustard (Uracil Mustard Capsules ® ); valrubicin (Valstar ® ); vinblastine (Velban ® ); vincristine (Oncovin ® ); vinorelbine (Navelbine ® ); zoledronate (Zometa ® ) and vorinostat (Zolinza ® ). [00152] In certain embodiments and dependining upon the indication to be treated, the additional therapeutic agent is selected from an antibiotic, a vasopressor, a steroid, an inotrope, an anti-thrombotic agent, a sedative, opioids, or an anesthetic.

[00153] The invention provides compounds of the invention for use as a pharmaceutical especially in the treatment or prevention of the diseases noted above. Also provided herein is the use of the present compounds in the manufacture of a medicament for the treatment or prevention of one of the diseases noted above. The present invention also provides the use of a compound of the invention or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment of a disease noted above.

V. Pharmaceutical Compositions and Dosing

[00154] In another aspect, the invention provides a pharmaceutical composition comprising a compound of the invention, namely a compound of Formula I or Formula II, and a

pharmaceutically acceptable carrier. For example, the invention also provides pharmaceutically acceptable compositions which comprise a therapeutically effective amount of one or more of the macrocyclic compounds of Formula I (including Formula I- A) or Formula II, formulated together with one or more pharmaceutically acceptable carriers (additives) and/or diluents, and optionally, one or more additional therapeutic agents described above.

[00155] As described in detail below, the pharmaceutical compositions of the present invention may be specially formulated for administration in solid or liquid form, including those adapted for the following: (1) oral administration, for example, drenches (aqueous or non-aqueous solutions or suspensions), tablets, e.g., those targeted for buccal, sublingual, and systemic absorption, boluses, powders, granules, pastes for application to the tongue; (2) parenteral administration, for example, by subcutaneous, intramuscular, intravenous or epidural injection as, for example, a sterile solution or suspension, or sustained-release formulation; (3) topical application, for example, as a cream, ointment, or a controlled-release patch or spray applied to the skin; (4) intravaginally or intrarectally, for example, as a pessary, cream or foam; (5) sublingually; (6) ocularly; (7) transdermally; or (8) nasally.

[00156] Exemplary materials which can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) pH buffered solutions; (21) polyesters, polycarbonates and/or polyanhydrides; and (22) other non- toxic compatible substances employed in pharmaceutical formulations.

[00157] Wetting agents, emulsifiers and lubricants, such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, release agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the compositions.

[00158] Examples of pharmaceutically acceptable antioxidants include: (1) water soluble antioxidants, such as ascorbic acid, cysteine hydrochloride, sodium bisulfate, sodium metabisulfite, sodium sulfite and the like; (2) oil-soluble antioxidants, such as ascorbyl palmitate, butylated hydroxyanisole (BHA), butylated hydroxytoluene (BHT), lecithin, propyl gallate, alpha-tocopherol, and the like; and (3) metal chelating agents, such as citric acid, ethylenediamine tetraacetic acid (EDTA), sorbitol, tartaric acid, phosphoric acid, and the like.

[00159] Formulations of the present invention include those suitable for oral, nasal, topical (including buccal and sublingual), rectal, vaginal and/or parenteral administration. The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect. Generally, out of one hundred percent, this amount will range from about 0.1 percent to about ninety-nine percent of active ingredient, preferably from about 5 percent to about 70 percent, most preferably from about 10 percent to about 30 percent.

[00160] In certain embodiments, a formulation of the present invention comprises an excipient selected from the group consisting of cyclodextrins, celluloses, liposomes, micelle forming agents, e.g., bile acids, and polymeric carriers, e.g., polyesters and polyanhydrides; and a compound of the present invention. In certain embodiments, an aforementioned formulation renders orally bioavailable a compound of the present invention. [00161] Methods of preparing these formulations or compositions include the step of bringing into association a compound of the present invention with the carrier and, optionally, one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association a compound of the present invention with liquid carriers, or finely divided solid carriers, or both, and then, if necessary, shaping the product.

[00162] Formulations of the invention suitable for oral administration may be in the form of capsules, cachets, pills, tablets, lozenges (using a flavored basis, usually sucrose and acacia or tragacanth), powders, granules, or as a solution or a suspension in an aqueous or non-aqueous liquid, or as an oil-in- water or water-in-oil liquid emulsion, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia) and/or as mouth washes and the like, each containing a predetermined amount of a compound of the present invention as an active ingredient. A compound of the present invention may also be administered as a bolus, electuary or paste.

[00163] In solid dosage forms of the invention for oral administration (capsules, tablets, pills, dragees, powders, granules, troches and the like), the active ingredient is mixed with one or more pharmaceutically-acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds and surfactants, such as poloxamer and sodium lauryl sulfate; (7) wetting agents, such as, for example, cetyl alcohol, glycerol monostearate, and non-ionic surfactants; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, zinc stearate, sodium stearate, stearic acid, and mixtures thereof; (10) coloring agents; and (11) controlled release agents such as crospovidone or ethyl cellulose. In the case of capsules, tablets and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may also be employed as fillers in soft and hard-shelled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.

[00164] A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.

[00165] The tablets, and other solid dosage forms of the pharmaceutical compositions of the present invention, such as dragees, capsules, pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be formulated for rapid release, e.g., freeze-dried. They may be sterilized by, for example, filtration through a bacteria-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved in sterile water, or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. The active ingredient can also be in micro-encapsulated form, if appropriate, with one or more of the above-described excipients.

[00166] Liquid dosage forms for oral administration of the compounds of the invention include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.

[00167] Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming and preservative agents.

[00168] Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.

[00169] Formulations of the pharmaceutical compositions of the invention for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compounds of the invention with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or a salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vaginal cavity and release the active compound.

[00170] Formulations of the present invention which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.

[00171] Dosage forms for the topical or transdermal administration of a compound of this invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active compound may be mixed under sterile conditions with a

pharmaceutically-acceptable carrier, and with any preservatives, buffers, or propellants which may be required.

[00172] The ointments, pastes, creams and gels may contain, in addition to an active compound of this invention, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.

[00173] Powders and sprays can contain, in addition to a compound of this invention, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder, or mixtures of these substances. Sprays can additionally contain customary propellants, such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.

[00174] Transdermal patches have the added advantage of providing controlled delivery of a compound of the present invention to the body. Such dosage forms can be made by dissolving or dispersing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate of such flux can be controlled by either providing a rate controlling membrane or dispersing the compound in a polymer matrix or gel.

[00175] Ophthalmic formulations, eye ointments, powders, solutions and the like, are also contemplated as being within the scope of this invention. [00176] Pharmaceutical compositions of this invention suitable for parenteral administration comprise one or more compounds of the invention in combination with one or more

pharmaceutically-acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain sugars, alcohols, antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.

[00177] Examples of suitable aqueous and nonaqueous carriers which may be employed in the pharmaceutical compositions of the invention include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.

[00178] These compositions may also contain adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents. Prevention of the action of microorganisms upon the subject compounds may be ensured by the inclusion of various antibacterial and antifungal agents, for example, paraben, chlorobutanol, phenol sorbic acid, and the like. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like into the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.

[00179] In some cases, in order to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally-administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle.

[00180] Injectable depot forms are made by forming microencapsulated matrices of the subject compounds in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue.

[00181] When the compounds of the present invention are administered as pharmaceuticals, to humans and animals, they can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99% (more preferably, 10 to 30%) of active ingredient in combination with a pharmaceutically acceptable carrier.

[00182] Regardless of the route of administration selected, the compounds of the present invention, which may be used in a suitable hydrated form, and/or the pharmaceutical compositions of the present invention, are formulated into pharmaceutically-acceptable dosage forms by conventional methods known to those of skill in the art.

[00183] Actual dosage levels of the active ingredients in the pharmaceutical compositions of this invention may be varied so as to obtain an amount of the active ingredient which is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.

[00184] The selected dosage level will depend upon a variety of factors including the activity of the particular compound of the present invention employed, or the ester, salt or amide thereof, the route of administration, the time of administration, the rate of excretion or metabolism of the particular compound being employed, the rate and extent of absorption, the duration of the treatment, other drugs, compounds and/or materials used in combination with the particular compound employed, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well known in the medical arts.

[00185] A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the compounds of the invention employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.

[00186] In general, a suitable daily dose of a compound of the invention will be that amount of the compound which is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above. Generally, oral, intravenous, intracerebroventricular and subcutaneous doses of the compounds of this invention for a patient will range from about 0.01 to about 50 mg per kilogram of body weight per day. [00187] If desired, the effective daily dose of the active compound may be administered as two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms. In certain aspects of the invention, dosing is one administration per day.

[00188] While it is possible for a compound of the present invention to be administered alone, it is preferable to administer the compound as a pharmaceutical composition.

[00189] The description above describes multiple aspects and embodiments of the invention. The patent application specifically contemplates all combinations and permutations of the aspects and embodiments.

EXAMPLES

[00190] The invention now being generally described, will be more readily understood by reference to the following examples, which are included merely for purposes of illustration of certain aspects and embodiments of the present invention, and are not intended to limit the invention.

EXAMPLE 1 - Synthesis of Intermediate 2-((5-(8-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)octyl)-l,2,4-oxadiazol-3-yl)methox y)benzoic acid (24)

18 19 20

[00191] To a solution of methyl-2-hydroxybenzoate (18, 200 mg, 1.28 mmol, 1.0 equiv) and 2- bromoacetonitrile (19, 0.175 mL, 2.56 mmol, 2.0 equiv) in DMF (6.5 mL) at room temperature was added potassium carbonate (530 mg, 3.8 mmol, 3.0 equiv). The reaction was stirred for 18 hours, and then diluted with water extracted with DCM (40 mL). The combined organic extracts were dried over MgS0 4 , filtered and concentrated. The crude residue 20 was used in the next step without further purification.

20 21 [00192] To a solution of the crude material from the previous step 20 (250 mg, 1.28 mmol, 1.0 equiv) in MeOH (10 mL) at room temperature was added hydroxylamine (50 wt% sol'n in water, 0.169 mL, 2.56 mmol, 2.0 equiv). The reaction was stirred for 2 hours, until the reaction was complete. The reaction mixture was partitioned between DCM (50 mL) and water (50 mL). The organic layer was collected, dried over MgS0 4 , filtered and concentrated. The crude

[00193] A solution of 9-((tert-butoxycarbonyl)amino)nonanoic acid (22, 0.308 g, 1.128 mmol) and di(lH-imidazol-l-yl)methanone (0.183 g, 1.128 mmol) in DMF (5.0 ml) was heated to 50 °C for 30 minutes. Then the crude material from the previous step 21 (0.234 g, 1.025 mmol) was added, and the reaction was heated to 115 °C for 18 hours. The reaction mixture was concentrated, and the crude residue was purified via flash chromatography on silica gel (gradient 0-50% EtOAc in hexanes), which provided pure methyl 2-((5-(8-((tert- butoxycarbonyl)amino)octyl)-l,2,4-oxadiazol-3-yl)methoxy)ben zoate (23, 307 mg, 64% over 3 steps).

[00194] To a solution of methyl 2-((5-(8-((tert-butoxycarbonyl)amino)octyl)-l,2,4-oxadiazol- 3- yl)methoxy)benzoate (23, 307 mg, 650 mmol, 1.0 equiv) in THF (6.5 mL) at room temperature was added lithium hydroxide (2.0 M in water, 6.5 mL, 13.1 mmol, 20 equiv). The reaction was stirred for 20 hours. The reaction mixture was quenched with 1 M HC1 solution, and extracted with EtOAc (2 x 20 mL). The organic layer was dried (MgS0 4 ), filtered and concentrated. The crude material was used in the next step without further purification.

[00195] To a solution of the crude material from the previous step in DCM (6.6 mL) at room temperature was added 2,2,2-trifluoroacetic acid (0.50 mL, 6.60 mmol, 10.0 equiv). The reaction was stirred for 18 hours, concentrated, and then co-evaporated with toluene (3 x 15 mL) to remove excess TFA. The crude residue was used in the next step without further purification. [00196] To a solution of the crude material from the previous step and potassium carbonate (274 mg, 1.98 mmol, 3.0 equiv) in water (3.3 mL) at 0°C was added a solution of (9H-fluoren-9- yl)methyl (2,5-dioxopyrrolidin-l-yl) carbonate (245 mg, 7.26 mmol, 1.1 equiv) in MeCN (3.3 mL). The reaction was stirred for 1 hour and then partitioned between 1.0 M HC1 and diethyl ether (15 mL each). The organic layer was dried (MgS0 4 ), filtered and concentrated, which provided the pure product 24 (204 mg, 54% yield over 3 steps). - Synthesis of Compound 418 (Table I)

[00197] 2-Chloro-trityl chloride resin 1 (3.51 g, 5.62 mmol) was swelled in anhydrous DCM (15 mL) for 15 minutes and then filtered and washed with DCM (2 x 15 mL). The intermediate 24 (3.2 g, 5.62 mmol) and N-ethyl-N-isopropylpropan-2-amine (2.94 mL, 16.9 mmol) were dissolved in anhydrous DCM (15 mL). The resulting solution was added to the swelled resin and agitated for 2 hours. The resin was then washed with 85: 10:5 DCM:MeOH:DIEA (15 mL x 2); DCM (15 mL x 3). After flushing with argon and dried under vacuum, resin 25 (5.94 g) was obtained.

[00198] Resin 25 (0.075 mmol, 100 mg) was suspended in DMF (5 mL x 10 minutes) and mixed with a stream of N2 every 30 seconds. The Fmoc group was removed from the resin- supported building block by mixing the resin twice with a solution of 2% DBU, 2% piperidine in DMF (5 mL x 5 minutes) while agitating with a stream of N2 every 30 seconds. The resin was washed four times with DMF (5 mL x 30 seconds). N-Fmoc-N-Methyl-L-valine (26, 0.1 M solution in DMF, 1.65 mL, 2.2 equiv, 0.165 mmol), followed by HATU (0.2 M solution in DMF, 0.8 mL, 2.1 equiv, 0.16 mmol) and N-methyl morpholine (1.0 M in DMF, 0.45 mL, 6.0 equiv, 0.45 mmol) were added to the resin. The reaction mixture was agitated by a stream of nitrogen for 120 minutes. The reagents were drained from the reaction vessel, and the resin 27 was

[00199] Resin 27 (0.075 mmol, 100 mg) was used directly from the previous step. The Fmoc group was removed from the resin-supported building block by mixing the resin twice with a solution of 2% DBU, 2% piperidine in DMF (5 mL x 5 minutes) while agitating with a stream of N2 every 30 seconds. The resin was washed four times with DMF (5 mL x 30 seconds). N- Fmoc-D-proline (28, 0.1 M solution in DMF, 1.65 mL, 2.2 equiv, 0.165 mmol), followed by HATU (0.2 M solution in DMF, 0.8 mL, 2.1 equiv, 0.16 mmol) and N-methyl morpholine (1.0 M in DMF, 0.45 mL, 6.0 equiv, 0.45 mmol) were added to the resin and the reaction mixture was agitated by a stream of nitrogen for 120 minutes. The reagents were drained from the vessel and then the coupling step was repeated once more. The resin 29 was then washed six times

[00200] Resin 29 (0.075 mmol, 100 mg) used directly from the previous step. The Fmoc group was removed from the resin-supported building block by mixing the resin twice with a solution of 2% DBU, 2% piperidine in DMF (5 mL x 5 minutes) while agitating with a stream of Ν2 every 30 seconds. The resin was washed four times with DMF (5 mL x 30 seconds). N-Fmoc-D- leucine (30, 0.1 M solution in DMF, 1.65 mL, 2.2 equiv, 0.165 mmol), followed by HATU (0.2 M solution in DMF, 0.8 mL, 2.1 equiv, 0.16 mmol) and N-methyl morpholine (1.0 M in DMF, 0.45 mL, 6.0 equiv, 0.45 mmol) were added to the resin. The reaction mixture was agitated by a stream of nitrogen for 120 minutes. The reagents were drained from the reaction vessel, and the resin 31 was washed four times with DMF (5 mL x 30 seconds) and then used directly in the next reaction.

[00201] Resin 31 (0.075 mmol, 100 mg) used directly from the previous step. The Fmoc group was removed from the resin-supported building block by mixing the resin twice with a solution of 2% DBU, 2% piperidine in DMF (5 mL x 5 minutes) while agitating with a stream of N2 every 30 seconds. The resin was washed four times with DMF (5 mL x 30 seconds) and then three times with DCM (5 mL x 30 seconds). The resin was treated with 5% TFA in DCM (5 mL x 5 minutes) then washed with DCM (3 mL), collecting both the cleavage solution and the wash. This was repeated once more. The volatiles were removed by evaporation using a Genevac

.

[00202] Crude reaction product containing 32 (0.075 mmol) and DIEA (0.13 mL, 10 equiv) was dissolved in DMF (4 mL). This solution was added dropwise to a solution containing HATU (34 mg, 0.090 mmol, 1.2 equiv) and HO At (12 mg, 0.090 mmol, 1.2 equiv) dissolved in DMF (11 mL). After 10 minutes the volatiles were evaporated under reduced pressure at 60 °C. The resultant crude mixture was dissolved in DMSO and purified on a Waters HPLC. Evaporation of volatiles followed by lyophilization resulted in final product 418 (11 mg, 0.0.016 mmol, 22% yield) as a white powder.

[00203] Compounds 339, 340, 380, 383, 384, 388, 391, 393, 394, 400, 401, 407, 408, 409, 411, 413, 416, 417, 419, 420, 421, 422, 423, 424, 425, 426, 427, 428, 429, 431, 438, 439, 441, 442, 444, 445, 446, 447, 450, 451, 452, 453, 454, 455, 456, 467, 468, 475, 476, 477, 479, 482, 500,

507 of Table I were produced by a similar procedure. EXAMPLE 3 - Synthesis of Intermediate 2-(2-((8-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)octyl)amino)-2-oxoethoxy)benzoic acid (39)

33 34 35

[00204] To a suspension of tert-butyl-2-hydroxybenzoate (33, 2.0 g, 10.3 mmol, 1.0 equiv) and potassium carbonate (7.12 g, 51.5 mmol, 5.0 equiv) in DMF (20 mL) at room temperature was added methyl-2-bromoacetate (34, 3.15 g, 20.6 mmol, 2.0 equiv). The reaction was stirred for 18 hours, and then diluted with water (100 mL) and extracted with EtOAc (100 mL). The organic layer was washed with brine, dried (MgS0 4 ), filtered and concentrated. The crude residue purified by flash chromatography on silica gel (gradient 0-20% EtOAc in hexanes), which provided pure product tert-butyl 2-(2-methoxy-2-oxoethoxy)benzoate (35, 2.3 g, 84% yield).

35 36

[00205] Lithium hydroxide (2.0 M in water, 21.5 mL, 43.2 mmol, 5.0 equiv) was added to a solution of tert-butyl 2-(2-methoxy-2-oxoethoxy)benzoate (35, 2.3 g, 8.64 mmol, 1.0 equiv) in THF (75 mL) at room temperature. The reaction was stirred for 4 hours. The reaction mixture was adjusted to pH 3 with 1 M HC1 solution, and extracted with EtOAc (2 x 75 mL). The organic layer was washed with brine, dried (MgS0 4 ), filtered and concentrated. The crude material 36 was used in the next step without further purification.

36 37 38

[00206] HATU (1.96 g, 5.15 mmol) was added to a solution of 2-(2-(tert- butoxycarbonyl)phenoxy)acetic acid (36, 1.00 g, 3.96 mmol) and N-ethyl-N-isopropylpropan-2- amine (2.77 ml, 15.8 mmol) in DMF (20 ml) at room temperature. The mixture was stirred for 10 minutes at room temperature, and then tert-butyl (8-aminooctyl)carbamate (37, 1.45 g, 5.95 mmol) was added. The reaction was stirred for 1 hour and then concentrated. The crude residue was purified by flash chromatography on silica gel (gradient 0-100% EtOAc in hexanes), which provided pure product 38 (1.89 g, 99% yield).

[00207] Then 2,2,2-trifluoroacetic acid (9.15 ml, 119 mmol, 30.0 equiv) was added to a solution of tert-butyl 2-(2-((8-((tert-butoxycarbonyl)amino)octyl)amino)-2-oxoethox y)benzoate (38, 1.89 g, 3.96 mmol, 1.0 equiv) in DCM (40 ml) at room temperature. The reaction was stirred for 18 hours, and then concentrated. The crude residue was co-evaporated with toluene (3 x 15 mL) to remove excess TFA, and then used in the next step without further purification.

[00208] To a solution of the crude material from the previous step (3.96 mmol, 1.0 equiv) and potassium carbonate (1.64 g, 11.9 mmol, 3.0 equiv) in water (40 ml) at 0 °C was added a solution of (9H-fluoren-9-yl)methyl (2,5-dioxopyrrolidin-l-yl) carbonate (1.34 g, 3.96 mmol, 1.0 equiv) in MeCN (40 ml) dropwise over 5 minutes. The reaction was stirred for 90 minutes and then was quenched with 1 M HC1 solution (100 mL) and extracted with diethyl ether (100 mL). The organic layer was concentrated, and the crude residue was purified by flash chromatography on silica gel (gradient 0-50% of a [90:5:5 DCM/MeOH/AcOH solution] in DCM, which provided pure 2-(2-((8-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)octyl)am ino)-2- oxoethoxy)benzoic acid (39, 1.41 g, 65% yield over two steps).

[00209] Compounds 183, 239, 273, 334, 335, 472, 473, 474, 478, 492, 493, 494, and 502 were made by a procedure similar to that described for the synthesis of compound 418 in Example 2, however, intermediate 24 was replaced with intermediate 39.

EXAMPLE 4 - Synthesis of Intermediate 2-((l-(8-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)octyl)-lH-l,2,3-triazol-4-yl)metho xy)benzoic acid (44)

40 41 42

[00210] To a suspension of crude (9H-fluoren-9-yl)methyl (8-aminooctyl)carbamate (40, 9.88 g, 20.57 mmol), copper(II) sulfate pentahydrate (0.514 g, 2.057 mmol), and potassium carbonate (7.82 g, 56.6 mmol) in MeOH (54.5 ml) was added lH-imidazole-l-sulfonyl azide (41, 5.39 g, 25.7 mmol). The reaction mixture was stirred at room temperature overnight then diluted with EtOAc. Adjusted pH to 2 with 1M HC1. Separated layers, washed organic layer with brine, dried (MgS0 4 ), and concentrated. Purified by flash chromatography on silica gel (gradient 0-20% EtOAc in hexanes) to give (9H-fluoren-9-yl)methyl (8-azidooctyl)carbamate (42, 5.7 g, 70.6% yield).

42 43 44 [00211] To a suspension of (9H-fluoren-9-yl)methyl (8-azidooctyl)carbamate (42, 2g, 5.10 mmol) and 2-(prop-2-yn-l-yloxy)benzoic acid (43, 0.898g, 5.10 mmol) in THF (2.83 mL), tert- butanol (2.83 mL), and water (2.83 mL) was added copper sulfate pentahydrate (0.064 g, 0.255 mmol) and sodium ascorbate (0.151 g, 0.764 mmol). The reaction was stirred at room temperature for 2 hours, diluted with DCM (5 mL) then washed with 1M HCl (5 mL) and brine (5 mL). The resulting organic layer was dried over MgS0 4 , filtered, and concentrated. The resulting material was purified by flash chromatography on silica gel (gradient 0-50% of a [90:5:5 DCM/MeOH/AcOH solution] in DCM, to give 2-((l-(8-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)octyl)-lH-l,2,3-triazol-4-yl)metho xy)benzoic acid (44, 2.72g, 94% yield) as a white solid. H NMR (400 MHz, DMSO-d 6 ) δ 12.53 (s, 1H), 8.15 (s, 1H), 7.87 (d, / = 7.5 Hz, 2H), 7.67 (d, / = 7.5 Hz, 2H), 7.61 (dd, / = 7.6, 1.9 Hz, 1H), 7.48 (ddd, / = 8.9, 7.4, 1.9 Hz, 1H), 7.40 (t, 7 = 7.5 Hz, 2H), 7.31 (ddd, J = 9.2, 7.6, 2.0 Hz, 3H), 7.26 - 7.21 (m, 1H), 7.00 (t, / = 7.5 Hz, 1H), 5.20 (s, 2H), 4.34 (t, / = 7.1 Hz, 2H), 4.28 (d, / = 6.9 Hz, 2H), 4.21 - 4.16 (m, 1H), 2.94 (q, / = 6.6 Hz, 2H), 1.79 (t, / = 7.2 Hz, 2H), 1.40 - 1.30 (m, 2H), 1.28 - 1.00 (m, 8H).

[00212] Compounds 163, 164, 194, 195, 197, 221, 302, 325, 328, and 329 were made by a procedure similar to that described for the synthesis of compound 418 in Example 2, however, intermediate 24 was replaced with intermediate 44.

EXAMPLE 5 - Synthesis of Intermediate 2-((ll-((((9H-fluoren-9- yl)methoxy)carbonyl)amino)undecyl)oxy)benzoic acid (51)

45 46 [00213] Isobutyl chloroformate (0.56 mL, 4.31 mmol) was added to a 0°C solution of l l-((tert- butoxycarbonyl)amino)undecanoic acid (45, 1 g, 3.32 mmol) and triethylamine (0.56 ml, 3.98 mmol) in THF (12 mL). The reaction was stirred for 45 minutes at 0 °C then sodium

borohydride (0.38 g, 9.95 mmol) was added. MeOH (23 ml) was then added slowly over 15 minutes and the reaction mixture was allowed to warm to room temperature. The reaction mixture was concentrated and the residue was dissolved in EtOAc, washed with 10% citric acid, 5% NaHCC^, water then brine, dried and concentrated. Purified on MPLC eluting with hexanes/EtOAc to give tert-butyl (l l-hydroxyundecyl)carbamate (46, 762 mg, 2.65 mmol, 80 % yield).

46 47 48

[00214] DIAD (0.26 mL, 1.31 mmol) was added dropwise over 30 minutes to a 0 °C solution of tert-butyl (l l-hydroxyundecyl)carbamate (46, 300 mg, 1.04 mmol), methyl 2-hydroxybenzoate (47, 0.14 ml, 1.04 mmol) and triphenylphosphine (342 mg, 1.31 mmol) in THF (10 ml). The reaction was allowed to warm to room temp and stirred for 16 hours. The mixture was concentrated onto silica gel and purified by MPLC eluting with hexanes/EtOAc to give methyl 2-((l l-((tert-butoxycarbonyl)amino)undecyl)oxy)benzoate (48, 303 mg, 0.72 mmol, 68.9 % yield).

[00215] 2N lithium hydroxide (1.90 ml, 3.81 mmol) was added to a solution of methyl 2-((l 1- ((tert-butoxycarbonyl)amino)undecyl)oxy)benzoate (48, 303 mg, 0.72 mmol) in THF (10 ml). The mixture was stirred at room temperature for 16 hours then diluted with EtOAc, washed with IN HC1, brine, dried and concentrated. The crude residue 49 was used without further purification in the next step.

[00216] 2-((l l-((tert-butoxycarbonyl)amino)undecyl)oxy)benzoic acid (49, 293 mg, 0.72 mmol) was dissolved in DCM (5 ml) and 2,2,2-trifluoroacetic acid (1.46 ml, 19.04 mmol) was added. The reaction mixture was stirred for 3 hours, concentrated to dryness and chased with toluene (3x). The crude residue 50 was used without further purification in the next step.

[00217] A solution of 2-((ll-aminoundecyl)oxy)benzoic acid 2,2,2-trifluoroacetic acid salt (50, 303 mg, 0.72 mmol) in a mixture of acetonitrile (10 mL) and water (5 mL) was cooled to 0 °C. Sodium carbonate (323 mg, 3.05 mmol) was added followed by the portionwise addition of (9H- fluoren-9-yl)methyl (2,5-dioxopyrrolidin-l-yl) carbonate (308 mg, 0.91 mmol). The reaction mixture was stirred for 4 hours slowly warming to room temperature. The pH was adjusted to 2 with IN HC1 and the mixture was diluted with EtOAc, washed with brine, dried and

concentrated. Purified on MPLC eluting with DCM/5% AcOH, 5% MeOH, DCM to give 2- ((ll-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)undecyl)oxy) benzoic acid (51, 343 mg, 0.65 mmol, 85 % yield).

[00218] Compounds 387, 480, and 487 were made by a procedure similar to that described for the synthesis of compound 418 in Example 2, however, intermediate 24 was replaced with intermediate 51.

EXAMPLE 6 - Synthesis of 2-((5-(8-((2-((((9H-fluoren-9- yl)methoxy)carbonyl)(methyl)amino)ethyl)(methyl)amino)octyl) -l,2,4-oxadiazol-3- yl)methoxy)benzoic acid (55)

[00219] To a solution of starting material 52 (0.17 g, 0.47 mmol) in DCE (5 mL) was added (9H-fluoren-9-yl)methyl-methyl(2-oxoethyl)carbamate (53, 0.14g, 0.35 mmol) followed by sodium triacetoxyborohydrate (75mg, 0.35 mmol) and the reaction mixture was stirred at room temperature for 1 hour. Then excessive formaldehyde solution and additional sodium triacetoxyborohydrate (0.5 mmol each) was added into the mixture and reaction continued for 2 hours. The mixture was diluted with DCM (50 mL) and washed with NaHCC>3 solution (50 mL). The resulting organic layer was dried, concentrated, and purified by HPLC to afford product 54 (87 mg).

54 55

[00220] To a solution of starting material 54 (87 mg) in dioxane (4 mL) was added aqueous 4N HCl (4 mL) and the mixture was heated at 85°C for 48 hours. The volatiles were removed and diluted with water (4 mL) and freeze dried to afford product 55 as HCl salt (90 mg).

[00221] Compounds 458 and 497 were made by a procedure similar to that described for the synthesis of compound 418 in Example 2, however, intermediate 24 was replaced with intermediate 55, and fewer amide coupling reactions were required to make the foregoing compounds.

EXAMPLE 7 - Synthesis of 2-((l-(9H-fluoren-9-yl)-4,7-dimethyl-3,17-dioxo-2-oxa-4,7,16 - triazaoctadecan-18-yl)oxy)benzoic acid (63) [00222] 2-chloroacetyl chloride (57, 0.35 ml, 4.43 mmol) was added to a solution of 8- aminooctan-l-ol (56, 1.9 g, 13.28 mmol) and DIPEA (2.3 ml, 13.28 mmol) in DCM (44 mL) at 0°C. After 30 minutes, complete consumption of the starting material was observed. The reaction mixture was quenched with an aqueous 1M HCl solution and extracted with DCM. The combined organic extracts were dried over MgS0 4 , filtered and concentrated. Purification via silica gel chromatography gave 2-chloro-N-(8-hydroxyoctyl)acetamide (58, 500 mg, 51% yield).

[00223] Potassium carbonate (1.5 g, 10.84 mmol), 2-chloro-N-(8-hydroxyoctyl)acetamide (58, 481 mg, 2.169 mmol) and methyl 2-hydroxybenzoate (47, 280 μί, 2.169 mmol) were stirred in DMF (21 mL). The reaction was stirred at 60°C for 18 hours before the mixture was cooled and quenched with an aqueous 1M HC1 solution. The mixture was extracted with DCM. The combined organic extracts were dried over MgS0 4 , filtered and concentrated. Purification via silica gel chromatography gave methyl 2-(2-((8-hydroxyoctyl)amino)-2-oxoethoxy)benzoate (59, 470 mg, 64% yield).

[00224] Mesyl-Cl (217 μΐ,, 2.79 mmol) was added to a solution of methyl 2-(2-((8- hydroxyoctyl)amino)-2-oxoethoxy)benzoate (59, 470 mg, 1.393 mmol) and triethylamine (777 μΐ, 5.57 mmol) in DCM (1.4 mL) at 0 °C. After 30 minutes, complete consumption of the starting material was observed. The reaction was quenched with a saturated NH 4 C1 solution and extracted with DCM. The combined organic extracts were dried over MgS0 4 and filtered.

Removal of solvent by rotary evaporation gave methyl 2-(2-((8- ((methylsulfonyl)oxy)octyl)amino)-2-oxoethoxy)benzoate (60), which was used without further purification.

H NMR (400 MHz, DMSO-d 6 ) δ 7.99 (t, J = 5.7 Hz, 1H), 7.78 (dd, J = 7.7, 1.8 Hz, 1H), 7.57 (ddd, J = 8.8, 7.4, 1.8 Hz, 1H), 7.14 (d, / = 8.4 Hz, 1H), 7.08 (t, / = 7.6 Hz, 1H), 4.60 (s, 2H), 4.17 (t, / = 6.4 Hz, 2H), 3.83 (s, 3H), 3.23 - 3.16 (m, 2H), 3.15 (s, 3H), 1.69 - 1.59 (m, 2H), 1.50 - 1.42 (m, 2H), 1.42 - 1.38 (m, 1H), 1.36 - 1.21 (m, 7H).

[00225] Tert-butyl methyl(2-(methylamino)ethyl)carbamate (61, 0.525 g, 2.79 mmol) was added to a solution of methyl 2-(2-((8-((methylsulfonyl)oxy)octyl)amino)-2-oxoethoxy)benzo ate (60, 0.579 g, 1.393 mmol) in DMF (14 mL). The reaction was stirred at 60-70°C for 48 hours before the solvent was removed by rotary evoparation. Purification by reverse-phase

chromatography gave methyl 2-((2,2,5,8-tetramethyl-4,18-dioxo-3-oxa-5,8,17-triazanonade can- 19-yl)oxy)benzoate (62, 650 mg, 92% yield).

62 63

[00226] Aqueous LiOH (1M, 5 mL, excess) was added to a solution of methyl 2-((2,2,5,8- tetramethyl-4,18-dioxo-3-oxa-5,8,17-triazanonadecan-19-yl)ox y)benzoate (62, 650 mg, 1.280 mmol) in sufficient THF for dissolution. The reaction was stirred at 35°C until complete conversion to the desired product was observed (12 hours). The reaction was cooled and concentrated. The residue was taken up in 33% TFA in DCM and stirred at room temperature for 30 minutes to permit Boc deprotection. The reaction product was concentrated and the residue was subsequently azeotroped with toluene to remove any residual TFA. The residue was then dissolved in 12 mL of THF before DIPEA (1.1 mL, 6.40 mmol) and Fmoc-OSu (648 mg, 1.92 mmol) were added. The reaction was stirred at room temperature until complete conversion to the desired product was observed (20 minutes). The reaction product was concentrated. Purification by reverse phase chromatography gave 2-((l-(9H-fluoren-9-yl)-4,7-dimethyl-3,17- dioxo-2-oxa-4,7,16-triazaoctadecan-18-yl)oxy)benzoic acid (63, 310 mg, 40% yield) as the TFA salt.

1H NMR (400 MHz, DMSO-d6) δ 13.07 (s, 1H), 9.10 (s, 1H), 8.21 (t, J = 5.7 Hz, 1H), 7.90 (d, J = 7.5 Hz, 2H), 7.78 (dd, J = 7.7, 1.8 Hz, 1H), 7.64 (d, J = 7.5 Hz, 2H), 7.55 (td, J = 7.7, 1.8 Hz, 1H), 7.42 (t, J = 7.4 Hz, 2H), 7.34 (t, J = 7.3 Hz, 2H), 7.14 (d, J = 8.4 Hz, 1H), 7.07 (t, J = 7.5 Hz, 1H), 4.61 (s, 2H), 4.58 - 4.53 (m, 1H), 4.43 - 4.34 (m, 1H), 4.29 (d, J = 6.3 Hz, 1H), 3.29 - 3.09 (m, 6H), 3.07 - 2.90 (m, 2H), 2.85 - 2.71 (m, 6H), 1.69 - 1.38 (m, 4H), 1.33 - 1.13 (m,

8H).

[00227] Compound 501 was made by a procedure similar to that described for the synthesis of compound 418 in Example 2, however, intermediate 24 was replaced with intermediate 63, and fewer amide coupling reactions were required to make compound 501.

EXAMPLE 8 - Synthesis of intermediate 68

63 64 65 [00228] To a solution of (((9H-fluoren-9-yl)methoxy)carbonyl)-D-proline (63, 0.70 g, 2.08 mmol, 1 equiv) in THF (2.08 ml) cooled to -15 °C (salt water ice bath) was added N- methylmorpholine (0.25 ml, 2.28 mmol, 1.1 equiv) and isopropyl carbonochloridate (64, 1M solution in toluene, 2.28 ml, 2.282 mmol, 1.1 equiv). The mixture was stirred for 20 min, and then ammonia (0.5 M sol'n in dioxane, 4.56 ml, 2.282 mmol) was added. The reaction was brought to room temperature and stirred for 16 hours. The mixture was quenched with 1M HCl and extracted with DCM (2 x 15 mL). The organic layer was dried over MgS0 4 , filtered and concentrated under reduced pressure, and the crude material 65 (691 mg, 99% yield) was used in the next step without further purification.

65 66

[00229] A suspension of the crude product from above 65 (0.473 g, 1.41 mmol, 1.0 equiv) and Lawesson's Reagent (0.341 g, 0.84 mmol, 0.6 equiv) were stirred in benzene (6.1 ml) at reflux under inert atmosphere. The solution became homogeneous after several hours. The reaction was cooled to room temperature, and let stand overnight. The mixture was purified directly by flash chromatography on silica gel (gradient 0-60% methanol in DCM), which provided pure product 66 (472 mg, 95% yield).

66 67 68

[00230] A solution of pure product from above 66 (0.472 g, 1.34 mmol, 1.0 equiv), 3-bromo-2- oxopropanoic acid (67, 0.313 g, 1.875 mmol, 1.4 equiv) and cesium carbonate (0.375 g, 3.75 mmol, 2.8 equiv) was stirred in ethanol (10.3 ml) at room temperature under inert atmosphere. The reaction was stirred for 24 hours until LCMS showed complete conversion to the final product. The reaction mixture was diluted with EtOAc (25 mL), and washed with 1M HCl solution and brine. The organic layer was dried (MgS0 4 ), filtered and concentrated under reduced pressure. The crude residue was purified by flash chromatography on silica gel (gradient 0-35% of a [90:5:5 DCM/MeOH/ Ac OH] solution in DCM), which provided the pure product 68 (203 mg, 36% yield). [00231] Compound 496 was made by a procedure similar to that described for the synthesis of compound 418 in Example 2, however, intermediate 24 was replaced with intermediate 68, and fewer amide coupling reactions were required to make compound 496.

EXAMPLE 9 - Synthesis of Compound 508 (Table I)

69 70 71

[00232] di(lH-imidazol-l-yl)methanone (448 mg, 2.76 mmol) was added to a solution of ((allyloxy)carbonyl)-D-proline (69, 500 mg, 2.51 mmol) in DMF (15 ml) and heated at 50 °C for 30 min. tert-butyl (Z)-(2-amino-2-(hydroxyimino)ethyl)carbamate (70, 570 mg, 3.01 mmol) was added and heating continued at 115 °C for 16 hours. The reaction mixture was cooled to room temperature, diluted with EtOAc and washed with water and then brine, dried and concentrated. The crude mixture was purified on MPLC eluting with hexanes/EtOAc to give allyl (R)-2-(3-(((tert-butoxycarbonyl)amino)methyl)-l,2,4-oxadiazo l-5-yl)pyrrolidine-l- carboxylate (71, 530 mg, 1.5 mmol, 60 % yield). LRMS [M+H+] found m/z 353.34, calcd for

[00233] allyl (R)-2-(3-(((tert-butoxycarbonyl)amino)methyl)-l,2,4-oxadiazo l-5-yl)pyrrolidine- 1-carboxylate (71, 265 mg, 0.75 mmol) was dissolved in DCM (5 ml) and 2,2,2-trifluoroacetic acid (1 ml, 13.03 mmol) was added. The reaction mixture was stirred for 3 hours, concentrated to dryness and chased with toluene (3 X) to give allyl (R)-2-(3-(aminomethyl)-l,2,4-oxadiazol- 5-yl)pyrrolidine-l-carboxylate 2,2,2-trifluoroacetate (72, 274 mg, 0.75 mmol, 99 % yield). The crude residue was used without further purification in the next step. LRMS [M+H+] found m/z 253.21, calcd for CnH 17 N 4 0 3 253.12.

[00234] N-ethyl-N-isopropylpropan-2-amine (0.533 ml, 2.99 mmol) was added to a solution of 9-((tert-butoxycarbonyl)amino)nonanoic acid (73, 245 mg, 0.90 mmol) and 2-(3H- [l,2 ]triazolo[4,5-b]pyridin-3-yl)-l,l,3,3-tetramethylisouronium hexafluorophosphate(V) (341 mg, 0.90 mmol) in DMF (5 ml) and stirred for 15 minutes. This solution was then added to a flask containing allyl (R)-2-(3-(aminomethyl)-l,2,4-oxadiazol-5-yl)pyrrolidine-l-ca rboxylate 2,2,2-trifluoroacetate (72, 274 mg, 0.75 mmol) and the mixture was stirred for 16 hours. Diluted with EtOAc, then washed with IN HC1, sat. aq NaHCC>3, brine then dried and concentrated. Purified on MPLC eluting with hexanes/EtOAc to give allyl (R)-2-(3-((9-((tert- butoxycarbonyl)amino)nonanamido)methyl)-l,2,4-oxadiazol-5-yl )pyrrolidine-l-carboxylate (74, 348 mg, 0.69 mmol, 92 % yield). LRMS [M+H+] found m/z 508.59, calcd for C25H42N5O 6 508.31.

[00235] allyl (R)-2-(3-((9-((tert-butoxycarbonyl)amino)nonanamido)methyl)- l,2,4-oxadiazol-5- yl)pyrrolidine-l-carboxylate (74, 185 mg, 0.36 mmol) was dissolved in DCM (5 ml) and 2,2,2- trifluoroacetic acid (1 ml, 13.03 mmol) was added. The reaction mixture was stirred for 3 hours, concentrated to dryness and chased with toluene (3 X) to give allyl (R)-2-(3-((9- aminononanamido)methyl)- 1 ,2,4-oxadiazol-5-yl)pyrrolidine- 1 -carboxylate 2,2,2-trifluoroacetate (75, 190 mg, 0.36 mmol, 100 % yield). The crude residue was used without further purification in the next step. LRMS [M+H+] found m/z 408.77, calcd for C2 0 H 3 4N5O4 408.25.

[00236] N-ethyl-N-isopropylpropan-2-amine (0.26 ml, 1.46 mmol) was added to a solution of 2-(2-(methoxycarbonyl)phenoxy)acetic acid (76, 96 mg, 0.46 mmol) and 2-(3H-

[l,2,3]triazolo[4,5-b]pyridin-3-yl)-l,l,3,3-tetramethylis ouronium hexafluorophosphate(V) (173 mg, 0.46 mmol) in DMF (5 ml) and stirred for 15 minutes. This solution was then added to a flask containing allyl (R)-2-(3-((9-aminononanamido)methyl)-l,2,4-oxadiazol-5-yl)py rrolidine- 1-carboxylate 2,2,2-trifluoroacetate (75, 190 mg, 0.364 mmol) and the mixture was stirred for 16 hours. Diluted with EtOAc, then washed with IN HC1, sat. aq NaHCC>3, then brine, dried and concentrated. Purified on MPLC eluting with hexanes/EtOAc to give allyl (R)-2-(3-((9-(2-(2 - (methoxycarbonyl)phenoxy)acetamido)nonanamido)methyl)-l,2,4- oxadiazol-5-yl)pyrrolidine-l- carboxylate (77, 166 mg, 0.28 mmol, 76 % yield). LRMS [M+H+] found m/z 600.08, calcd for

[00237] A mixture of l,3-dimethylpyrimidine-2,4,6(lH,3H,5H)-trione (65 mg, 0.42 mmol), Pd(P(Ph) 3 ) 4 (32 mg, 0.03 mmol) and allyl (R)-2-(3-((9-(2-(2-

(methoxycarbonyl)phenoxy)acetamido)nonanamido)methyl)-l,2 ,4-oxadiazol-5-yl)pyrrolidine-l- carboxylate (77, 166 mg, 0.28 mmol) were stirred at room temperature in DMF (3 ml) for 16 hours. Purified directly on reverse phase MPLC eluting with CH 3 CN/H2O containing 0.1% TFA to give methyl (R)-2-(2-oxo-2-((9-oxo-9-(((5-(pyrrolidin-2-yl)-l,2,4-oxadia zol-3- yl)methyl)amino)nonyl)amino)ethoxy)benzoate 2,2,2-trifluoroacetate (78, 113 mg, 0.18 mmol, 65 % yield). LRMS [M+H+] found m/z 516.55, calcd for C2 6 H 38 N5O 6 516.27.

[00238] N-ethyl-N-isopropylpropan-2-amine (0.13 ml, 0.71 mmol) was added to a solution of (tert-butoxycarbonyl)-D-leucine hydrate (79, 55 mg, 0.22 mmol), 2-(3H-[l,2,3]triazolo[4,5- b]pyridin-3-yl)-l, l,3,3-tetramethylisouronium hexafluorophosphate (V) (84 mg, 0.22 mmol) and 3H-[l,2,3]triazolo[4,5-b]pyridin-3-ol (30 mg, 0.22 mmol) in DMF (3 ml) and stirred for 15 minutes, methyl (R)-2-(2-oxo-2-((9-oxo-9-(((5-(pyrrolidin-2-yl)-l,2,4-oxadia zol-3- yl)methyl)amino)nonyl)amino)ethoxy)benzoate 2,2,2-trifluoroacetate (78, 111 mg, 0.18 mmol) was added and the reaction mixture was stirred for 16 hr. Diluted with EtOAc and washed with IN HCl, sat aq NaHCC>3 and brine then dried and concentrated. Purified on MPLC eluting with DCM/MeOH to give methyl 2-(2-((9-(((5-((R)-l-((tert-butoxycarbonyl)-D-leucyl)pyrroli din-2- yl)-l,2,4-oxadiazol-3-yl)methyl)amino)-9-oxononyl)amino)-2-o xoethoxy)benzoate (80, 111 mg, 0.15 mmol, 86 % yield). LRMS [M+H+] found m/z 730.40, calcd for C 3 7H57N 6 O9 729.41.

[00239] 2N lithium hydroxide (0.15 ml, 0.31 mmol) was added to a solution of methyl 2-(2-((9- (((5-((R)-l-((tert-butoxycarbonyl)-D-leucyl)pyrrolidin-2-yl) -l,2,4-oxadiazol-3- yl)methyl)amino)-9-oxononyl)amino)-2-oxoethoxy)benzoate (80, 56 mg, 0.08 mmol) in THF (2 ml) and stirred for 4 hours. The mixture was concentrated to dryness then 4 N hydrogen chloride in dioxane (1.5 ml, 6.00 mmol) was added and stirring continued for 2 hours.

Concentrated and chased with toluene (3X) to give 2-(2-((9-(((5-((R)-l-(D-leucyl)pyrrolidin-2- yl)-l,2,4-oxadiazol-3-yl)methyl)amino)-9-oxononyl)amino)-2-o xoethoxy)benzoic acid hydrochloride (81, 50 mg, 0.077 mmol, 100 % yield). Used without further purification. LRMS [M+H+] found m/z 616.24, calcd for C 3 1H47N 6 O7 615.34.

81 508

[00240] A mixture of 2-(3H-[l,2,3]triazolo[4,5-b]pyridin-3-yl)-l, l,3,3-tetramethylisouronium hexafluorophosphate(V) (32 mg, 0.08 mmol), 3H-[l,2,3]triazolo[4,5-b]pyridin-3-ol (13 mg, 0.09 mmol) and N-ethyl-N-isopropylpropan-2-amine (0.06 ml, 0.31 mmol) in DMF (2.5 ml) was added dropwise to a solution of 2-(2-((9-(((5-((R)-l-(D-leucyl)pyrrolidin-2-yl)-l,2,4-oxadia zol- 3-yl)methyl)amino)-9-oxononyl)amino)-2-oxoethoxy)benzoic acid hydrochloride (81, 50 mg, 0.08 mmol) in DMF (2.5 ml) over 20 minutes. The mixture was stirred for 3 hours then purified directly on prep. HPLC eluting with CH3CN/H20 containing 0.1% TFA to give (22R,4R,Z)-4- isobutyl-8-oxa-5,l l,21-triaza-l(5,3)-oxadiazola-2(2,l)-pyrrolidina-7(l,2)- benzenacyclodocosaphane-3,6,10,20-tetraone (508, 18 mg, 0.030 mmol, 39.3 % yield). 1H NMR (400 MHz, D6-DMSO): δ 8.72 (1 H, d, J = 7.0 Hz), 8.42 (1 H, t, J = 6.0 Hz), 8.26 (1 H, t, J = 5.9 Hz), 7.57 (1 H, dd, J = 7.6, 1.8 Hz), 7.41 (1 H, td, J = 7.8, 1.9 Hz), 7.04-6.99 (2 H, m), 5.23 (1 H, dd, J = 8.3. 3.2 Hz), 4.69-4.64 (1 H, m), 4.52 (2 H, d, J = 3.6 Hz), 4.29 (2 H, dd, J = 5.9, 1.9 Hz), 3.85-3.79 (1 H, m), 3.70-3.64 (1 H, m), 3.18-3.10 (1 H, m), 2.99-2.91 (1 H, m), 2.29-2.23 (1 H, m), 2.09-1.93 (5 H, comp), 1.73-1.59 (2 H, comp), 1.59-1.35 (5 H, comp), 1.25-1.13 (8 H, comp), 0.89 (6 H, dd, J = 6.6, 3.7 Hz). LRMS [M+H+] found m/z 597.66, calcd for C 3 1H45N 6 O 6 597.33.

EXAMPLE 10 - Characterization of Exemplary Compounds of Formulae I and II

[00241] Other compounds of Formulae I and II were made by procedures similar to those described above with the appropriate substitution for one or more of the reagents. Mass

Spectrometry values and/or 1H NMR data for compounds of the invention are set forth in Table 1.

TABLE 1 - Characterization of Exemplary Compounds

2.52 (dd, J = 13.7, 10.1 Hz, IH), 1.74 - 1.66 (m, IH), 1.63 - 1.52 (m, IH), 1.26 (dt, J = 9.4, 4.9 Hz, 2H), 1.19 (dt, J = 12.4, 5.7 Hz, IH), 0.79 (dd, J = 6.6, 2.4 Hz, 6H). LCMS - Calculated: 812.36; Found: 812.65

IH NMR (400 MHz, DMSO-d6) δ 8.65 (d, J = 8.0 Hz, IH), 8.25 (s, IH), 8.13 (dd, J = 11.8, 8.1 Hz, 2H), 7.90 (t, J = 8.0 Hz, 2H), 7.85 (dd, J = 7.8, 1.9 Hz, IH), 7.49 (ddd, J = 8.8, 7.2, 1.9 Hz, IH), 7.42 - 7.28 (m, 4H), 7.14 (d, J = 8.6 Hz, 2H), 7.04 (t, J = 7.5 Hz, IH), 6.82 (d, J = 8.7 Hz, 2H), 5.34 (d, J = 11.7 Hz, IH), 5.20 (d, J = 11.7 Hz, IH), 4.93 (d, J = 4.0 Hz, IH), 4.75 (td, J = 8.4, 6.6, 4.2 Hz, 3H),

125 4.55 (q, J = 6.9 Hz, IH), 4.42 - 4.33 (m,

2H), 4.33 - 4.24 (m, 2H), 4.07 - 3.99 (m, IH), 3.65 (s, 3H), 3.58 - 3.43 (m, 2H), 3.13 - 3.01 (m, 2H), 2.92 (dd, J = 14.7, 10.3 Hz, IH), 2.77 (dd, J = 14.2, 11.3 Hz,

IH), 1.75 - 1.61 (m, 2H), 1.42 (dt, J = 13.5, 6.6 Hz, IH), 1.23 - 1.11 (m, 2H), 0.71 - 0.61 (m, IH), 0.21 (d, J = 7.7 Hz, 2H), 0.06 - -0.02 (m, IH), -0.03 - -0.12 (m, IH). LCMS - Calculated: 850.32; Found: 850.7

IH NMR (400 MHz, DMSO-d6) δ 8.63 (d, J = 8.1 Hz, IH), 8.22 (s, IH), 8.10 (d, J = 7.8 Hz, 2H), 7.93 - 7.88 (m, 2H), 7.85 (dd, J = 7.8, 1.9 Hz, IH), 7.48 (ddd, J = 8.9, 7.3, 1.9 Hz, IH), 7.42 - 7.29 (m, 4H), 7.13 (d, J = 8.7 Hz, 2H), 7.04 (t, J = 7.5 Hz, IH), 6.82 (d, J = 8.7 Hz, 2H), 5.34 (d, J = 11.8 Hz, IH), 5.22 (d, J = 11.9 Hz,

JL X J .- H IH), 4.92 (d, J = 4.0 Hz, IH), 4.77 (td, J =

9.4, 4.5 Hz, IH), 4.72 (t, J = 5.1 Hz, 2H),

126 4.49 (q, J = 7.5 Hz, IH), 4.44 - 4.30 (m,

3H), 4.27 (t, J = 7.5 Hz, IH), 4.03 (d, J =

/ N=N V 4.2 Hz, IH), 3.65 (s, 3H), 3.52 - 3.46 (m,

IH), 3.40 (dd, J = 10.3, 4.7 Hz, IH), 3.15

U - 3.03 (m, 2H), 2.91 (dd, J = 14.6, 9.8 Hz,

IH), 2.76 (dd, J = 14.1, 11.2 Hz, IH), 1.74 - 1.64 (m, IH), 1.53 - 1.26 (m, 4H), 1.21 (dt, J = 12.5, 5.6 Hz, IH), 1.16 - 1.02 (m, 2H), 0.75 (dd, J = 6.6, 4.3 Hz, 6H). LCMS - Calculated: 866.35; Found: 866.73

LCMS - Calculated: 747.4; Found:

331

747.58

F

LCMS - Calculated: 775.43; Found:

332

775.62

F

LCMS - Calculated: 752.39; Found:

333

752.6

LCMS - Calculated: 710.39; Found:

334

710.58

LCMS - Calculated: 713.38; Found:

335

713.54

LCMS - Calculated: 721.43; Found: 721.78

- Calculated: 696.38; Found:

- Calculated: 695.39; Found:

- Calculated: 695.39; Found:

- Calculated: 695.39; Found:

EXAMPLE 11 - EVALUATION OF BIOLOGICAL ACTIVITY

[00242] Exemplary compounds of Formula I and Formula II were tested for their ability to inhibit IDO activity in the assay described below, which used HeLa Cells.

[00243] Briefly, HeLa cells were treated for 48 hours with 50 ng/mL human interferon gamma (IFNy) and 50 μΜ L-tryptophan, with and without compound in assay media. Tryptophan conversion to N-formlkynurenine was determined by using NFKgreen reagent (Netherlands Translational Research Center, Oss, The Netherlands) added directly onto the cells. Then 8000 HeLa cells in 35 μΐ. complete medium (RPMI1640/10%FBS) were seeded into the wells (8000 cells/well) of a 384 well black clear bottom plate (Greiner). The plate was incubated overnight at 37 °C. Serial dilutions of the compounds to be tested (5 μί) were added to end of the wells of the plate and the plate was incubated at 37°C for 1 hour.

[00244] A solution containing 5X the final concentration of IFNy (250 ng/mL) and Trp (250 μΜ) was prepared in complete medium. 10 of the IFN/Trp solution was added to each well and the plate was incubated at 37°C for 45-48 hours. Controls for background level of Trp oxidation were derived from wells receiving Trp but no IFNy.

[00245] Then, 5 μί NFK green reagent was added to end well and the plate was sealed and incubated at 37°C for 4 hours. The level of fluorescence from each well of the plate was determined. The IC 50 value for the inhibition of the fluorescence due to the oxidation of Trp was determined from a four parameter fit of the dose response data.

[00246] A separate cell line, SW48, was used to assess the ability of compounds to inhibit TDO (tryptophan-2,3-dioxygenase) activity. SW48 cells were found to exhibit an endogenous level of TDO but no IDO is determined by Western Blotting. None of the compounds tested exhibited a significant level of inhibition of TDO over the range of concentrations tested.

[00247] The resulting IC 50 values of IDO inhibition are provided in Table 2, where "A" indicates an IC 50 value less than 100 nM; "B" indicates an IC 50 value in the range of 100 nM up to 1 μΜ; "C" indicates an IC 50 value in the range of 1 μΜ up to 10 μΜ; "D" indicates an IC 50 value 10 μΜ or greater, and "NT" indicates that the compound has not been tested.

TABLE 2 - Assay Results of Exemplary Compounds of Formulae I and II

IDO Inhibitor IDO Inhibitor IDO Inhibitor if if

Activity Activity Activity

137 A 189 A 243 A

138 C 190 B 244 B

139 B 191 A 245 A

140 B 192 A 246 A

141 A 193 C 247 A

142 A 194 B 248 A

143 A 195 A 249 A

144 A 196 C 250 A

145 A 197 A 251 A

146 B 198 B 252 A

147 A 199 B 253 A

148 A 200 A 254 A

149 A 201 A 255 A

150 B 202 B 256 A

151 C 203 A 257 A

152 B 204 A 258 A

153 C 205 A 259 B

154 C 206 A 260 A

155 B 207 A 261 A

156 A 208 A 262 A

157 A 209 A 263 A

158 C 210 A 264 A

159 A 211 B 265 B

160 A 212 A 266 A

161 A 213 A 267 A

162 A 214 A 268 A

163 A 215 A 269 B

164 A 216 A 270 B

165 A 217 A 271 B

166 D 218 A 272 B

167 C 219 A 273 B

168 A 220 A 274 B

169 A 221 A 275 A

170 A 222 A 276 B

171 A 223 A 277 A

172 B 224 A 278 A

173 B 225 A 279 C

174 A 226 A 280 B

175 A 227 A 281 A

176 A 228 A 282 B

177 A 231 B 283 B

178 B 232 A 284 A

179 A 233 A 285 B

180 A 234 A 286 D

181 A 235 B 287 A

182 A 236 A 288 A

183 A 237 A 289 A

184 C 238 A 290 B

185 D 239 A 291 A

186 C 240 A 292 A

187 B 241 B 293 B

188 A 242 A 294 D IDO Inhibitor IDO Inhibitor IDO Inhibitor if if

Activity Activity Activity

295 A 347 A 399 A

296 B 348 B 400 A

297 B 349 B 401 A

298 A 350 A 402 A

299 A 351 B 403 A

300 A 352 B 404 A

301 A 353 A 405 C

302 A 354 A 406 B

303 A 355 C 407 A

304 A 356 C 408 A

305 A 357 A 409 A

306 C 358 NT 410 A

307 A 359 A 411 A

308 A 360 A 412 A

309 A 361 A 413 A

310 B 362 B 414 A

311 B 363 C 415 A

312 A 364 B 416 A

313 A 365 A 417 A

314 A 366 D 418 A

315 A 367 D 419 A

316 A 368 A 420 A

317 A 369 A 421 A

318 A 370 A 422 B

319 A 371 A 423 B

320 A 372 A 424 NT

321 A 373 A 425 A

322 B 374 B 426 A

323 A 375 B 427 NT

324 D 376 C 428 NT

325 NT 377 A 429 C

326 C 378 A 430 D

327 A 379 A 431 C

328 A 380 A 432 A

329 B 381 NT 433 B

330 A 382 A 434 NT

331 A 383 A 435 C

332 A 384 A 436 B

333 NT 385 C 437 NT

334 NT 386 A 438 B

335 A 387 A 439 B

336 C 388 A 440 C

337 B 389 C 441 C

338 B 390 A 442 C

339 A 391 A 443 C

340 A 392 C 444 A

341 B 393 A 445 A

342 A 394 A 446 NT

343 C 395 A 447 A

344 A 396 A 448 C

345 A 397 A 449 C

346 A 398 A 450 NT IDO Inhibitor IDO Inhibitor if if

Activity Activity

451 B 503 B

452 A 504 A

453 B 505 A

454 D 506 A

455 NT 507 C

456 NT 508 B

457 NT

458 A

459 D

460 C

461 A

462 NT

463 D

464 C

465 B

466 A

467 B

468 NT

469 A

470 A

471 A

472 A

473 A

474 NT

475 NT

476 C

477 B

478 B

479 B

480 A

481 A

482 B

483 A

484 A

485 A

486 A

487 A

488 NT

489 B

490 NT

491 NT

492 NT

493 NT

494 A

495 A

496 B

497 NT

498 B

499 B

500 A

501 A

502 C EXAMPLE 12 - Anti-tumor Activity of Examplary Compound 163 In Vivo

[00248] This example demonstrates the in vivo efficacy of compound 163 (Table 1) against growth of mouse pancreatic adenocarcinoma cells subcutaneously inoculated into female C57BL/6 mice.

[00249] Forty-eight female C57BL/6 mice were inoculated subcutaneously with 5xl0 6 Pan02 cells. Seven days post-inoculation (Study Day 0), twenty-four of the animals were randomized by tumor size into two groups of 12 and treatment commenced. Animals in each group received either 10 mL/kg Vehicle Control (33% Solutol, 67% Water) or 100 mg/kg of compound 163 dissolved in vehicle ((33% Solutol, 67% Water) administered twice daily, eight hours apart for 27 consecutive days. Clinical observations, animal weights and tumor size measurements were recorded two times weekly for the duration of the study. All groups demonstrated a significant increase in body weight during the study. None of the animals were euthanized due to adverse clinical signs and all animals are sacrificed at study termination.

[00250] As shown in Figure 1, there was a significant (p < 0.05) decrease in tumor volume following treatment with compound 163 compared with Vehicle Control on Study Day 26. These results show that compound 163 and related compounds may be helpful as part of a combination therapy with other therapeutic (e.g., anti-cancer) agents for the treatment of disorders associated with elevated levels of IDO activity. INCORPORATION BY REFERENCE

[00251] The entire disclosure of each of the patent documents and scientific articles referred to herein is incorporated by reference for all purposes.

EQUIVALENTS

[00252] The invention may be embodied in other specific forms without departing from the spirit or essential characteristics thereof. The foregoing embodiments are therefore to be considered in all respects illustrative rather than limiting the invention described herein. Scope of the invention is thus indicated by the appended claims rather than by the foregoing description, and all changes that come within the meaning and range of equivalency of the claims are intended to be embraced therein.