Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
NOVEL SUBSTITUTED [1.1.1] BICYCLO COMPOUNDS AS INDOLEAMINE 2,3-DIOXYGENASE INHIBITORS
Document Type and Number:
WIPO Patent Application WO/2019/231870
Kind Code:
A1
Abstract:
Disclosed herein are compounds of formula (I) which are inhibitors of an IDO enzyme: (I). Also disclosed herein are uses of the compounds in the potential treatment or prevention of an IDO-associated disease or disorder. Also disclosed herein are compositions comprising these compounds. Further disclosed herein are uses of the compositions in the potential treatment or prevention of an IDO-associated disease or disorder.

Inventors:
HE SHUWEN (US)
CLAUSEN DANE (US)
GUO LIANGQIN (US)
HAN YONGXIN (US)
HUANG XIANHAI (US)
PASTERNAK ALEXANDER (US)
PU QINGLIN (US)
XIAO DONG (US)
XIAO LI (US)
ZHANG HONGJUN (US)
YE FENG
Application Number:
PCT/US2019/034088
Publication Date:
December 05, 2019
Filing Date:
May 28, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
MERCK SHARP & DOHME (US)
HE SHUWEN (US)
CLAUSEN DANE (US)
GUO LIANGQIN (US)
HAN YONGXIN (US)
HUANG XIANHAI (US)
PASTERNAK ALEXANDER (US)
PU QINGLIN (US)
XIAO DONG (US)
XIAO LI (US)
ZHANG HONGJUN (US)
International Classes:
C07C13/08; A61K31/166; C07C233/65
Domestic Patent References:
WO2019204180A12019-10-24
WO2019231871A12019-12-05
WO2016165613A12016-10-20
WO2001002369A22001-01-11
WO2002068470A22002-09-06
WO2002010192A22002-02-07
WO2004004771A12004-01-15
WO2004072286A12004-08-26
WO2004056875A12004-07-08
WO2013019906A12013-02-07
WO2010077634A12010-07-08
WO2010027827A22010-03-11
WO2011066342A22011-06-03
Foreign References:
US20160075654A12016-03-17
US7488802B22009-02-10
US7521051B22009-04-21
US8008449B22011-08-30
US8354509B22013-01-15
US8168757B22012-05-01
US20110271358A12011-11-03
US8383796B22013-02-26
Other References:
DATABASE PubChem 29 January 2018 (2018-01-29), Database accession no. 132090779
NELP ET AL.: "Immune-modulating enzyme indoleamine-2,3-dioxygenase is effectively inhibited by targeting its apo-form", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 115, no. 13, 27 March 2018 (2018-03-27), pages 3249 - 3254, XP055643566, ISSN: 0027-8424, DOI: 10.1073/pnas.1719190115
DAUBENER ET AL., ADV. EXP. MED. BIOL, vol. 467, 1999, pages 517 - 24
TAYLOR ET AL., FASEB J., vol. 5, 1991, pages 2516 - 22
LOGAN ET AL., IMMUNOLOGY, vol. 105, 2002, pages 478 - 87
BROWN ET AL., ADV. EXP. MED. BIOL, vol. 294, 1991, pages 425 - 35
PORTULA ET AL., BLOOD, vol. 106, 2005, pages 2382 - 90
MEDAWAR, SYMP. SOC. EXP. BIOL., vol. 7, 1953, pages 320 - 38
MOAN ET AL., SCIENCE, vol. 281, 1998, pages 1191 - 3
UYTTENHOVE ET AL., NATURE MED., vol. 9, 2003, pages 1269 - 74
MULLER ET AL., NATURE MED., vol. 11, 2005, pages 312 - 9
MUNN ET AL., SCIENCE, vol. 297, 2002, pages 1867 - 70
MUNN ET AL., J. CLIN. INVEST, vol. 114, no. 2, 2004, pages 280 - 90
GROHMANN ET AL., TRENDS IMMUNOL, vol. 24, 2003, pages 242 - 8
WIRLEITNER ET AL., CURR. MED. CHEM., vol. 10, 2003, pages 1581 - 91
JAE EUN CHEONG ET AL.: "A patent review of IDO 1 inhibitors for cancer", EXPERT OPINION ON THERAPEUTIC PATENTS, 23 February 2018 (2018-02-23)
the American Pharmaceutical Association and the Pharmaceutical Press; "NCBI", Database accession no. NP_079515
T. W. GREENEP. G. M. WUTS: "Protective Groups in Organic Synthesis", 1999, WILEY
Attorney, Agent or Firm:
MERCK SHARP & DOHME CORP. (US)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A compound of formula (I), or a pharmaceutically acceptable salt thereof:

wherein:

X is selected from a bond and -CH(Ra)-; where Ra is selected from hydrogen and Ci-6 alkyl;

Rb is selected from: (i) hydrogen and (ii) Ci-6 alkyl; each occurrence of R1 and R2 is independently selected from:(i) hydrogen and (ii) Ci-6 alkyl; and each occurrence of R3 and R4 is independently selected from:

(i) aryl, and

(ii) heterocyclyl;

wherein each of the aryl of (i) and heterocyclyl of (ii) is optionally substituted with one to four substituents independently selected from:

(a) halogen,

(b) Ci-6 alkyl, optionally substituted with one to four halogens, and

(c) -CN.

2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein each occurrence of R1 and R2 is independently selected from: (i) hydrogen, (ii) methyl and (iii) ethyl.

3. The compound of claim 2, or a pharmaceutically acceptable salt thereof, wherein R3 is selected from:

(i) phenyl, and

(ii) pyridinyl;

wherein each of the phenyl of (i) and pyridinyl of (ii) is optionally substituted with one to four substituents independently selected from: (a) halogen,

(b) Ci-6 alkyl, optionally substituted with one to three halogens, and

(c) -CN.

4. The compound of claim 2, or a pharmaceutically acceptable salt thereof, wherein R3 is phenyl, optionally substituted with a halogen.

5. The compound of claim 2, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from:

(i) phenyl, and

(ii) a heterocyclyl selected from furanyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxazolyl, naphthyridinyl, phthalazinyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridinyl, pyrimidinyl, quinazolinyl and a fused bicyclic ring moiety wherein a 6-membered heterocyclic ring comprising 2 nitrogen atoms and a 5- membered carbocyclic ring are connected through two atoms;

wherein each of the phenyl of (i) and heterocyclyl of (ii) is optionally substituted with one to four substituents independently selected from:

(a) halogen,

(b) Ci-6 alkyl, optionally substituted with one to four halogens, and

(c) -CN.

6. The compound of claim 5, or a pharmaceutically acceptable salt thereof, wherein R4 is selected from:

(i) phenyl, and

(ii) a heterocyclyl selected from naphthyridinyl, phthalazinyl, pyridinyl, pyrimidinyl, quinazolinyl and a fused bicyclic ring moiety wherein a 6-membered aromatic heterocyclic ring comprising 2 nitrogen atoms and a 5-membered carbocyclic ring are connected through two carbon atoms

wherein each of the phenyl of (i) and heterocyclyl of (ii) is optionally substituted with one to four substituents independently selected from:

(a) halogen,

(b) Ci-6 alkyl, optionally substituted with one to four halogens, and

(c) -CN.

7. The compound of claim 2, or a pharmaceutically acceptable salt thereof, wherein:

R1 is hydrogen;

R2 is selected from:

(i) hydrogen,

(ii) methyl, and

(iii) ethyl;

R3 is selected from:

(i) phenyl, and

(ii) pyridinyl;

wherein each of the phenyl of (i) and pyridinyl of (ii) is optionally substituted with one to four substituents independently selected from:

(a) halogen,

(b) Ci-6 alkyl, optionally substituted with one to four halogens, and

(c) -CN; and

R4 is selected from:

(i) phenyl, and

(ii) a heterocyclyl selected from naphthyridinyl, phthalazinyl, pyridinyl, pyrimidinyl, quinazolinyl and a fused bicyclic ring moiety wherein a 6-membered aromatic heterocyclic ring comprising 2 nitrogen atoms and a 5-membered carbocyclic ring are connected through two carbon atoms;

wherein each of the phenyl of (i) and heterocyclyl of (ii) is optionally substituted with one to four substituents independently selected from:

(a) halogen,

(b) Ci-6 alkyl, optionally substituted with one to four halogens, and

(c) -CN.

8. The compound of claim 7, or a pharmaceutically acceptable salt thereof, wherein: R1 is hydrogen;

R2 is selected from:

(i) methyl, and

(ii) ethyl;

R3 is selected from:

(i) phenyl, and

(ii) pyridinyl;

wherein each of the phenyl of (i) and pyridinyl of (ii) is optionally substituted with one to four substituents independently selected from:

(a) halogen, and

(b) Ci-6 alkyl, optionally substituted with one to four halogens; and

R4 is selected from:

(i) phenyl, and

(ii) a heterocyclyl selected from 6,7-dihydro-5H-cyclopenta[d]pyrimidinyl, 1,7- naphthyridinyl, phthalazinyl, pyridinyl, pyrimidinyl and quinazolinyl;

wherein each of the phenyl of (i) and heterocyclyl of (ii) is optionally substituted with one to four substituents independently selected from:

(a) halogen,

(b) Ci-4 alkyl, optionally substituted with one to four halogens, and

(c) -CN.

9. The compound of claim 1 of formula (la), or a pharmaceutically acceptable salt thereof:

wherein:

Rb is selected from (i) hydrogen and (ii) methyl;

R3 is selected from: (i) phenyl, and

(ii) pyridinyl;

wherein each of the phenyl of (i) and pyridinyl of (ii) is optionally substituted with one to three substituents independently selected from:

(a) halogen, and

(b) Ci-6 alkyl, optionally substituted with one to three halogens; and

R4 is selected from:

(i) phenyl, and

(ii) a heterocyclyl selected from naphthyridinyl, phthalazinyl, pyridinyl, pyrimidinyl, quinazolinyl and a fused bicyclic ring moiety wherein a 6-membered aromatic heterocyclic ring comprising 2 nitrogen atoms and a 5-membered carbocyclic ring are connected through two carbon atoms;

wherein each of the phenyl of (i) and heterocyclyl of (ii) is optionally substituted with one to three substituents independently selected from:

(a) halogen,

(b) Ci-6 alkyl, optionally substituted with one to three halogens, and

(c) -CN.

10. The compound of claim 9, or a pharmaceutically acceptable salt thereof: wherein:

Rb is hydrogen;

R3 is phenyl, optionally substituted with one to three substituents independently selected from:

(a) halogen, and

(b) Ci-4 alkyl, optionally substituted with one to three halogens; and

R4 is phenyl, optionally substituted with one to three substituents independently selected from:

(a) halogen, and

(b) Ci-4 alkyl, optionally substituted with one to three halogens.

11. The compound of claim 1 of formula (lb), or a pharmaceutically acceptable salt thereof:

wherein:

Rb is selected from:

(i) hydrogen,

(ii) methyl, and

(iii) ethyl;

R3 is selected from:

(i) phenyl, and

(ii) pyridinyl;

wherein each of the phenyl of (i) and pyridinyl of (ii) is optionally substituted with one to four substituents independently selected from:

(a) halogen,

(b) Ci-6 alkyl, optionally substituted with one to four halogens, and

(c) -CN; and

R4 is selected from:

(i) phenyl, and

(ii) a heterocyclyl selected from naphthyridinyl, phthalazinyl, pyridinyl, pyrimidinyl, quinazolinyl and a fused bicyclic ring moiety wherein a 6-membered aromatic heterocyclic ring comprising 2 nitrogen atoms and a 5-membered carbocyclic ring are connected through two carbon atoms;

wherein each of the phenyl of (i) and heterocyclyl of (ii) is optionally substituted with one to four substituents independently selected from:

(a) halogen,

(b) Ci-6 alkyl, optionally substituted with one to four halogens, and

(c) -CN.

12. The compound of claim 11, or a pharmaceutically acceptable salt thereof: wherein: Rb is hydrogen;

R3 is selected from:

(i) phenyl, and

(ii) pyridinyl;

wherein each of the phenyl of (i) and pyridinyl of (ii) is optionally substituted with one to three substituents independently selected from:

(a) halogen, and

(b) Ci-4 alkyl, optionally substituted with one to three halogens; and

R4 is selected from:

(i) phenyl, and

(ii) a heterocyclyl selected from 6,7-dihydro-5H-cyclopenta[d]pyrimidinyl, 1,7- naphthyridinyl, phthalazinyl, pyridinyl, pyrimidinyl and quinazolinyl;

wherein each of the phenyl of (i) and heterocyclyl of (ii) is optionally substituted with one to three substituents independently selected from:

(a) halogen,

(b) Ci-4 alkyl, optionally substituted with one to three halogens, and

(c) -CN.

13. The compound of Claim 1 selected from the group consisting of:

N-(4-chlorophenyl)-2-(3-((2-methylpyrimidin-4-yl)amino)bicyclo[l .1. l]pentan-l- yl)propanamide,

N-(4-fluorophenyl)-2-(3-(quinazolin-4-ylamino)bicyclo[l .1. l]pentan-l-yl)propanamide, 2-(3-((6-fluoro-2-methylquinazolin-4-yl)amino)bicyclo[l .1. l]pentan-l-yl)-N-(4- fluorophenyl)prop anami de,

2-(3-((2-chloro-6,7-dihydro-5H-cyclopenta[d]pyrimidin-4-yl)amino)bicyclo[l .1. l]pentan-l-yl)- N -(4-fluorophenyl)prop anami de,

N-(4-fluorophenyl)-2-(3-((6-fluoroquinazolin-4-yl)amino)bicyclo[l .1. l]pentan-l- yl)propanamide,

N-(4-fluorophenyl)-2- { 3 -[(1 ,7-naphthyridin-8-yl)amino]bicyclo[ 1.1.1 Jpentan- 1 -yl | propanamide, N-(4-fhlorophenyl)-2-(3-{[3-fluoro-5-(trifluoromethyl)pyridin-2-yl]amino}bicyclo[l . l . l]pentan- 1 -yl)propanamide, N-(4-fluorophenyl)-2-(3-{[6-methyl-5-(trifluoromethyl)pyridin-2- yl]amino}bicyclo[ 1.1.1 Jpentan- 1 -yl)propanamide,

N-(4-fluorophenyl)-2-(3-{[5-(trifluoromethyl)pyridin-2-yl]amino}bicyclo[l . l . l]pentan-l- yl)propanamide,

N-(4-fluorophenyl)-2-(3-{[6-(trifluoromethyl)pyrimidin-4-yl]amino}bicyclo[l . l . l]pentan-l- yl)propanamide,

2-(3 -{ [3 -cyano-4-(trifluorom ethyl )pyridin-2-yl]amino}bicyclo[ 1.1.1 Jpentan- 1 -yl)-N-(4- fluorophenyl)prop anami de,

N-(4-fluorophenyl)-2-(3 - { [4-(trifluorom ethyl )pyri din-2 -yl]amino}bicyclo[ 1.1.1 Jpentan- 1 - yl)propanamide,

N-(4-fluorophenyl)-2-(3-{[2-fluoro-4-(trifluoromethyl)pyridin-3-yl]amino}bicyclo[l .1.1 Jpentan- 1 -yl)propanamide,

N-(4-fluorophenyl)-2-(3-{[3-(trifluoromethyl)pyridin-2-yl]amino}bicyclo[l . l . l]pentan-l- yl)propanamide,

N-(4-fluorophenyl)-2-(3 - { [3 -(trifluoromethyl)pyrazin-2-yl]amino}bicyclo[ 1.1.1 Jpentan- 1 - yl)propanamide,

N-(4-fluorophenyl)-2-(3-{[3-fluoro-5-(trifluoromethyl)pyridin-2-yl]amino}bicyclo[l .1.1 Jpentan- 1 -yl)propanamide,

N-(4-fluorophenyl)-2- { 3 -[(phthalazin- 1 -yl)amino]bicyclo[ 1.1.1 Jpentan- 1 -yl | propanamide, N-(4-fluorophenyl)-2-(3 - { [2-(trifluorom ethyl )pyridin-4-yl]amino}bicyclo[ 1.1.1 Jpentan- 1 - yl)propanamide,

N-(4-fluorophenyl)-2-(3-{[2-(trifluoromethyl)pyrimidin-4-yl]amino}bicyclo[l . l . l]pentan-l- yl)propanamide,

N-(4-fluorophenyl)-2-(3 - { [6-(trifluorom ethyl )pyri din-2 -yl]amino}bicyclo[ 1.1.1 Jpentan- 1 - yl)propanamide,

(S)-N-(4-fluorophenyl)-2-(3-((2-(trifluoromethyl)pyridin-4-yl)amino)bicyclo[l .1.1 Jpentan- 1- yl)propanamide,

(S)-2-(3-((3-chlorobenzyl)amino)bicyclo[l .1.1 Jpentan- l-yl)-N-(4-fluorophenyl)propanamide, (S)-2-(3-((3-chlorobenzyl)(methyl)amino)bicyclo[l .1.1 Jpentan- l-yl)-N-(4- fluorophenyl)propanamide,

(S)-N-(4-fluorophenyl)-2-(3-((3-(trifluoromethyl)benzyl)amino)bicyclo[l .1.1 Jpentan- 1- yl)propanamide, (S)-N-(4-fluorophenyl)-2-(3 -(methyl (3 -(trifluorom ethyl )benzyl)amino)bicy clo[ 1.1. l]pentan-l- yljpropanamide,

(S)-2-(3-((3,4-dichlorobenzyl)amino)bicyclo[l .1. l]pentan-l-yl)-N-(4- fluorophenyljpropanamide, and

(S)-2-(3 -((3 ,4-dichlorobenzyl)(methyl)amino)bicyclo[ 1.1.1 jpentan- 1 -yl)-N-(4- fluorophenyljprop anami de ;

or a pharmaceutically acceptable salt thereof.

14. A composition which comprises an inert carrier and a compound of any of claims 1-13 or a pharmaceutically acceptable salt thereof.

15. Use of a compound of any of claims 1-13 or a pharmaceutically acceptable salt thereof for the manufacture of a medicament for the treatment or prevention of an IDO- associated disease or disorder.

16. A method for treating or preventing an IDO-associated disease or disorder in a mammalian subject which comprises administering to the subject an effective amount of a compound of any of claims 1-13 or a pharmaceutically acceptable salt thereof.

17. A method for treating an IDO-associated disease or disorder in a mammalian subject which comprises administering to the subject an effective amount of a compound of any of claims 1-14 or a pharmaceutically acceptable salt thereof in combination with another anti cancer agent.

18. A method for treating an IDO-associated disease or disorder in a mammalian subject which comprises administering to the subject an effective amount of a compound of any of claims 1-13 or a pharmaceutically acceptable salt thereof in combination with two additional anti-cancer agents.

19. The method of any of claims 16-18, wherein the IDO-associated disease or disorder is a cancer, viral infection, HCV infection, depression, neurodegenerative disorders, trauma, age-related cataracts, organ transplantation, and autoimmune diseases.

20. The method of claim 19, wherein the cancer is selected from colon cancer, pancreas cancer, breast cancer, prostate cancer, lung cancer, brain cancer, ovarian cancer, cervical cancer, testes cancer, renal cancer, head and neck cancer, lymphoma, leukemia and melanoma.

Description:
TITLE OF THE INVENTION

NOVEL SUBSTITUTED [1.1.1] BICYCLO COMPOUNDS AS INDOLEAMINE 2,3- DIOXYGENASE INHIBITORS

BACKGROUND OF THE INVENTION

Tryptophan (Trp) is an essential amino acid required for the biosynthesis of proteins, niacin and the neurotransmitter 5-hydroxytryptamine (serotonin). The enzyme indoleamine 2,3 -di oxygenase (IDO) catalyzes the first and rate limiting step in the degradation of L-tryptophan to N-formyl-kynurenine. In human cells, a depletion of Trp resulting from IDO activity is a prominent gamma interferon (IFN-g) -inducible antimicrobial effector mechanism. IFN-g stimulation induces activation of IDO, which leads to a depletion of Trp, thereby arresting the growth of Trp-dependent intracellular pathogens such as Toxoplasma gondii and Chlamydia trachomatis. IDO activity also has an antiproliferative effect on many tumor cells, and IDO induction has been observed in vivo during rejection of allogeneic tumors, indicating a possible role for this enzyme in the tumor rejection process (Daubener, et al, 1999, Adv. Exp. Med. Biol, 467: 517-24; Taylor, et al, 1991, FASEB L, 5: 2516-22).

It has been observed that HeLa cells co-cultured with peripheral blood

lymphocytes (PBLs) acquire an immuno-inhibitory phenotype through up-regulation of IDO activity. A reduction in PBL proliferation upon treatment with interleukin-2 (IL2) was believed to result from IDO released by the tumor cells in response to IFN-g secretion by the PBLs. This effect was reversed by treatment with l-methyl- tryptophan (l-MT), a specific IDO inhibitor. It was proposed that IDO activity in tumor cells may serve to impair antitumor responses (Logan, et al, 2002, Immunology, 105: 478-87).

Several lines of evidence suggest that IDO is involved in induction of immune tolerance. Studies of mammalian pregnancy, tumor resistance, chronic infections and

autoimmune diseases have shown that cells expressing IDO can suppress T-cell responses and promote tolerance. Accelerated Trp catabolism has been observed in diseases and disorders associated with cellular immune activation, such as infection, malignancy, autoimmune diseases and AIDS, as well as during pregnancy. For example, increased levels of IFNs and elevated levels of urinary Trp metabolites have been observed in autoimmune diseases; it has been postulated that systemic or local depletion of Trp occurring in autoimmune diseases may relate to the degeneration and wasting symptoms of these diseases. In support of this hypothesis, high levels of IDO were observed in cells isolated from the synovia of arthritic joints. IFNs are also elevated in human immunodeficiency virus (HIV) patients, and increasing IFN levels are associated with a worsening prognosis. Thus, it was proposed that IDO is induced chronically by HIV infection, and is further increased by opportunistic infections, and that the chronic loss of Trp initiates mechanisms responsible for cachexia, dementia and diarrhea and possibly immunosuppression of AIDS patients (Brown, et al., 1991, Adv. Exp. Med. Biol, 294: 425-35). To this end, it has recently been shown that IDO inhibition can enhance the levels of virus- specific T cells and, concomitantly, reduce the number of virally-infected macrophages in a mouse model of HIV (Portula et al., 2005, Blood, 106: 2382-90).

IDO is believed to play a role in the immunosuppressive processes that prevent fetal rejection in utero. More than 40 years ago, it was observed that, during pregnancy, the genetically disparate mammalian conceptus survives in spite of what would be predicted by tissue transplantation immunology (Medawar, 1953, Symp. Soc. Exp. Biol. 7: 320-38).

Anatomic separation of mother and fetus and antigenic immaturity of the fetus cannot fully explain fetal allograft survival. Recent attention has focused on immunologic tolerance of the mother. Because IDO is expressed by human syncytiotrophoblast cells and systemic tryptophan concentration falls during normal pregnancy, it was hypothesized that IDO expression at the maternal-fetal interface is necessary to prevent immunologic rejection of the fetal allografts. To test this hypothesis, pregnant mice (carrying syngeneic or allogeneic fetuses) were exposed to l-MT, and a rapid, T cell-induced rejection of all allogeneic conception was observed. Thus, by catabolizing tryptophan, the mammalian conceptus appears to suppress T- cell activity and defends itself against rejection, and blocking tryptophan catabolism during murine pregnancy allows maternal T cells to provoke fetal allograft rejection (Moan, et al., 1998, Science, 281 : 1191-3).

Further evidence for a tumoral immune resistance mechanism based on tryptophan degradation by IDO comes from the observation that most human tumors

constitutively express IDO, and that expression of IDO by immunogenic mouse tumor cells prevents their rejection by preimmunized mice. This effect is accompanied by a lack of accumulation of specific T cells at the tumor site and can be partly reverted by systemic treatment of mice with an inhibitor of IDO, in the absence of noticeable toxicity. Thus, it was suggested that the efficacy of therapeutic vaccination of cancer patients might be improved by concomitant administration of an IDO inhibitor (Uyttenhove et al., 2003, Nature Med., 9: 1269- 74). It has also been shown that the IDO inhibitor, l-MT, can synergize with chemotherapeutic agents to reduce tumor growth in mice, suggesting that IDO inhibition may also enhance the anti-tumor activity of conventional cytotoxic therapies (Muller et al, 2005, Nature Med., 11 : 312-9).

One mechanism contributing to immunologic unresponsiveness toward tumors may be presentation of tumor antigens by tolerogenic host APCs. A subset of human IDO- expressing antigen-presenting cells (APCs) that coexpressed CD 123 (IL3RA) and CCR6 and inhibited T-cell proliferation have also been described. Both mature and immature CD123- positive dendritic cells suppressed T-cell activity, and this IDO suppressive activity was blocked by l-MT (Munn, et al, 2002, Science, 297: 1867-70). It has also been demonstrated that mouse tumor-draining lymph nodes (TDLNs) contain a subset of plasmacytoid dendritic cells (pDCs) that constitutively express immunosuppressive levels of IDO. Despite comprising only 0.5% of lymph node cells, in vitro, these pDCs potently suppressed T cell responses to antigens presented by the pDCs themselves and also, in a dominant fashion, suppressed T cell responses to third- party antigens presented by nonsuppressive APCs. Within the population of pDCs, the majority of the functional IDO-mediated suppressor activity segregated with a novel subset of pDCs coexpressing the B-lineage marker CD19. Thus, it was hypothesized that IDO-mediated suppression by pDCs in TDLNs creates a local microenvironment that is potently suppressive of host antitumor T cell responses (Munn, et al. , 2004, J. Clin. Invest, 114(2): 280-90).

IDO degrades the indole moiety of tryptophan, serotonin and melatonin, and initiates the production of neuroactive and immunoregulatory metabolites, collectively known as kynurenines. By locally depleting tryptophan and increasing proapoptotic kynurenines, IDO expressed by dendritic cells (DCs) can greatly affect T-cell proliferation and survival. IDO induction in DCs could be a common mechanism of deletional tolerance driven by regulatory T cells. Because such tolerogenic responses can be expected to operate in a variety of

physiopathological conditions, tryptophan metabolism and kynurenine production might represent a crucial interface between the immune and nervous systems (Grohmann, et al, 2003, Trends Immunol, 24: 242-8). In states of persistent immune activation, availability of free serum Trp is diminished and, as a consequence of reduced serotonin production, serotonergic functions may also be affected (Wirleitner, et al., 2003, Curr. Med. Chem., 10: 1581-91).

In light of the experimental data indicating a role for IDO in immunosuppression, tumor resistance and/or rejection, chronic infections, HIV-infection, AIDS (including its manifestations such as cachexia, dementia and diarrhea), autoimmune diseases or disorders (such as rheumatoid arthritis), and immunologic tolerance and prevention of fetal rejection in utero, therapeutic agents aimed at suppression of tryptophan degradation by inhibiting IDO activity are desirable. Inhibitors of IDO can be used to activate T cells and therefore enhance T cell activation when the T cells are suppressed by pregnancy, malignancy or a virus such as HIV. Inhibition of IDO may also be an important treatment strategy for patients with neurological or neuropsychiatric diseases or disorders such as depression. [1.1.1] Bicyclo compounds disclosed herein are useful in the potential treatment or prevention of IDO-related diseases.

SUMMARY OF THE INVENTION

Disclosed herein are novel [1.1.1] bicyclo compounds of formula (I) which are inhibitors of the IDO enzyme. Also disclosed herein are uses of these compounds in the potential treatment or prevention of an IDO-associated disease or disorder. Also disclosed herein are compositions comprising one or more of the compounds. Further disclosed herein are uses of these compositions in the potential prevention or treatment of an IDO-associated disease or disorder.

DETAILED DESCRIPTION OF THE INVENTION

Disclosed herein are compounds of formula (I), or a pharmaceutically acceptable salt thereof:

wherein:

X is selected from a bond and -CH(R a )-; where R a is selected from hydrogen and Ci -6 alkyl;

R b is selected from: (i) hydrogen and (ii) Ci -6 alkyl;

each occurrence of R 1 and R 2 is independently selected from:(i) hydrogen and (ii) Ci -6 alkyl; and each occurrence of R 3 and R 4 is independently selected from:

(i) aryl, and

(ii) heterocyclyl;

wherein each of the aryl of (i) and heterocyclyl of (ii) is optionally substituted with one to four substituents independently selected from:

(a) halogen,

(b) Ci- 6 alkyl, optionally substituted with one to four halogens, and

(c) -CN. In one embodiment of the compounds of formula (I), or a pharmaceutically acceptable salt thereof, each occurrence of R 1 and R 2 is independently selected from: (i) hydrogen, (ii) methyl and (iii) ethyl.

In one embodiment of the compounds of formula (I), or a pharmaceutically acceptable salt thereof:

R 3 is selected from:

(i) phenyl, and

(ii) pyridinyl;

wherein each of the phenyl of (i) and pyridinyl of (ii) is optionally substituted with one to four substituents independently selected from:

(a) halogen,

(b) Ci- 6 alkyl, optionally substituted with one to three halogens, and

(c) -CN.

In one embodiment of the compounds of formula (I), or a pharmaceutically acceptable salt thereof: R 3 is phenyl, optionally substituted with a halogen.

In one embodiment of the compounds of formula (I), or a pharmaceutically acceptable salt thereof:

R 4 is selected from:

(i) phenyl, and

(ii) a heterocyclyl selected from furanyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxazolyl, naphthyridinyl, phthalazinyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridinyl, pyrimidinyl, quinazolinyl and a fused bicyclic ring moiety wherein a 6-membered heterocyclic ring comprising 2 nitrogen atoms and a 5- membered carbocyclic ring are connected through two atoms;

wherein each of the phenyl of (i) and heterocyclyl of (ii) is optionally substituted with one to four substituents independently selected from:

(a) halogen,

(b) Ci- 6 alkyl, optionally substituted with one to four halogens, and

(c) -CN.

In one embodiment of the compounds of formula (I), or a pharmaceutically acceptable salt thereof:

R 4 is selected from:

(i) phenyl, and (ii) a heterocyclyl selected from naphthyridinyl, phthalazinyl, pyridinyl, pyrimidinyl, quinazolinyl and a fused bicyclic ring moiety wherein a 6-membered aromatic heterocyclic ring comprising 2 nitrogen atoms and a 5-membered carbocyclic ring are connected through two carbon atoms;

wherein each of the phenyl of (i) and heterocyclyl of (ii) is optionally substituted with one to four substituents independently selected from:

(a) halogen,

(b) Ci- 6 alkyl, optionally substituted with one to four halogens, and

(c) -CN.

In one embodiment of the compounds of formula (I), or a pharmaceutically acceptable salt thereof:

R 1 is hydrogen;

R 2 is selected from:

(i) hydrogen,

(ii) methyl, and

(iii) ethyl;

R 3 is selected from:

(i) phenyl, and

(ii) pyridinyl;

wherein each of the phenyl of (i) and pyridinyl of (ii) is optionally substituted with one to four substituents independently selected from:

(a) halogen,

(b) Ci- 6 alkyl, optionally substituted with one to four halogens, and

(c) -CN; and

R 4 is selected from:

(i) phenyl, and

(ii) a heterocyclyl selected from 6,7-dihydro-5H-cyclopenta[d]pyrimidinyl, furanyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxazolyl, l,7-naphthyridinyl, phthalazinyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridinyl, pyrimidinyl and quinazolinyl;

wherein each of the phenyl of (i) and heterocyclyl of (ii) is optionally substituted with one to four substituents independently selected from:

(a) halogen, (b) Ci- 6 alkyl, optionally substituted with one to four halogens, and

(c) -CN.

In one embodiment of the compounds of formula (I), or a pharmaceutically acceptable salt thereof:

R 1 is hydrogen;

R 2 is selected from:

(i) methyl, and

(ii) ethyl;

R 3 is selected from:

(i) phenyl, and

(ii) pyridinyl;

wherein each of the phenyl of (i) and pyridinyl of (ii) is optionally substituted with one to four substituents independently selected from:

(a) halogen, and

(b) Ci- 6 alkyl, optionally substituted with one to four halogens; and R 4 is selected from:

(i) phenyl, and

(ii) a heterocyclyl selected from 6,7-dihydro-5H-cyclopenta[d]pyrimidinyl, 1,7- naphthyridinyl, phthalazinyl, pyridinyl, pyrimidinyl and quinazolinyl;

wherein each of the phenyl of (i) and heterocyclyl of (ii) is optionally substituted with one to four substituents independently selected from:

(a) halogen,

(b) Ci- 4 alkyl, optionally substituted with one to four halogens, and

(c) -CN.

In one embodiment of the compounds of formula (I), or a pharmaceutically acceptable salt thereof, the compound is of formula (la):

wherein:

R b is selected from (i) hydrogen and (ii) methyl;

R 3 is selected from:

(i) phenyl, and (ii) pyridinyl;

wherein each of the phenyl of (i) and pyridinyl of (ii) is optionally substituted with one to three substituents independently selected from:

(a) halogen, and

(b) Ci- 6 alkyl, optionally substituted with one to three halogens; and

R 4 is selected from:

(i) phenyl, and

(ii) a heterocyclyl selected from naphthyridinyl, phthalazinyl, pyridinyl, pyrimidinyl, quinazolinyl and a fused bicyclic ring moiety wherein a 6-membered aromatic heterocyclic ring comprising 2 nitrogen atoms and a 5-membered carbocyclic ring are connected through two carbon atoms;

wherein each of the phenyl of (i) and heterocyclyl of (ii) is optionally substituted with one to three substituents independently selected from:

(a) halogen,

(b) Ci- 6 alkyl, optionally substituted with one to three halogens, and

(c) -CN.

In one embodiment of the compounds of formula (la), or a pharmaceutically acceptable salt thereof:

R b is hydrogen;

R 3 is phenyl, optionally substituted with one to three substituents independently selected from:

(a) halogen, and

(b) Ci- 4 alkyl, optionally substituted with one to three halogens; and

R 4 is phenyl, optionally substituted with one to three substituents independently selected from:

(a) halogen, and

(b) Ci- 4 alkyl, optionally substituted with one to three halogens.

In one embodiment of the compounds of formula (I), or a pharmaceutically acceptable salt thereof, the compound is of formula (lb):

wherein:

R b is selected from: (i) hydrogen,

(ii) methyl, and

(iii) ethyl;

R 3 is selected from:

(i) phenyl, and

(ii) pyridinyl;

wherein each of the phenyl of (i) and pyridinyl of (ii) is optionally substituted with one to four substituents independently selected from:

(a) halogen,

(b) Ci- 6 alkyl, optionally substituted with one to four halogens, and

(c) -CN; and

R 4 is selected from:

(i) phenyl, and

(ii) a heterocyclyl selected from naphthyridinyl, phthalazinyl, pyridinyl, pyrimidinyl, quinazolinyl and a fused bicyclic ring moiety wherein a 6-membered aromatic heterocyclic ring comprising 2 nitrogen atoms and a 5-membered carbocyclic ring are connected through two carbon atoms;

wherein each of the phenyl of (i) and heterocyclyl of (ii) is optionally substituted with one to four substituents independently selected from:

(a) halogen,

(b) Ci- 6 alkyl, optionally substituted with one to four halogens, and

(c) -CN.

In one embodiment of the compounds of formula (lb), or a pharmaceutically acceptable salt thereof:

R b is hydrogen;

R 3 is selected from:

(i) phenyl, and

(ii) pyridinyl;

wherein each of the phenyl of (i) and pyridinyl of (ii) is optionally substituted with one to three substituents independently selected from:

(a) halogen, and

(b) Ci- 4 alkyl, optionally substituted with one to three halogens; and

R 4 is selected from: (i) phenyl, and

(ii) a heterocyclyl selected from 6,7-dihydro-5H-cyclopenta[d]pyrimidinyl, 1,7- naphthyridinyl, phthalazinyl, pyridinyl, pyrimidinyl and quinazolinyl;

wherein each of the phenyl of (i) and heterocyclyl of (ii) is optionally substituted with one to three substituents independently selected from:

(a) halogen,

(b) Ci- 4 alkyl, optionally substituted with one to three halogens, and

(c) -CN.

In one embodiment, the compound disclosed herein is selected from the group consisting of the compounds exemplified herein, for example, in Examples 1 - 33, or a pharmaceutically acceptable salt thereof.

Also disclosed herein is a pharmaceutical composition comprising a compound of formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable carrier.

Also disclosed herein is a method of inhibiting activity of indoleamine 2,3- dioxygenase (IDO) comprising contacting IDO with a compound of formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof.

Also disclosed herein is a method of inhibiting immunosuppression in a patient comprising administering to said patient an effective amount of a compound of formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof.

Also disclosed herein is a method of treating cancer, viral infection, depression, a neurodegenerative disorder, trauma, age-related cataracts, organ transplant rejection, or an autoimmune disease in a patient comprising administering to said patient an effective amount of a compound of formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof.

Also disclosed herein is a method of treating melanoma in a patient comprising administering to said patient an effective amount of a compound of formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof.

Further disclosed herein is a compound of formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof, for use in therapy. In one embodiment, disclosed herein is the use of a compound of formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof, for the preparation of a medicament for use in therapy.

As used herein, "alkyl" refers to both branched- and straight-chain saturated aliphatic hydrocarbon groups of 1 to 18 carbon atoms, or more specifically, 1 to 12 carbon atoms. Examples of such groups include, but are not limited to, methyl (Me), ethyl (Et), n- propyl (Pr), n-butyl (Bu), n-pentyl, n-hexyl, and the isomers thereof such as isopropyl (i-Pr), isobutyl (i-Bu), sec-butyl (s-Bu), /cvV-butyl (t-Bu), isopentyl, and isohexyl. Alkyl groups may be optionally substituted with one or more substituents as defined herein. “Ci- 6 alkyl" refers to an alkyl group as defined herein having 1 to 6 carbon atoms.

"Aryl" refers to an aromatic monocyclic or multicyclic ring moiety comprising 6 to 14 ring carbon atoms, or more specifically, 6 to 10 ring carbon atoms. Monocyclic aryl rings include, but are not limited to, phenyl. Multicyclic rings include, but are not limited to, naphthyl and bicyclic rings wherein phenyl is fused to a Cs-vcycloalkyl or Cs-vcycloalkenyl ring. Aryl groups may be optionally substituted with one or more substituents as defined herein. Bonding can be through any of the carbon atoms of any ring.

“Cycloalkyl” refers to a monocyclic saturated carbocyclic ring having the specified number of carbon atoms. For example, C3-6 cycloalkyl refers to a cycloalkyl group as defined herein having 3 to 6 carbon atoms. Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl. Cycloalkyl groups may be optionally substituted with one or more substituents as defined herein.

“Halo” or“halogen” refers to fluoro, chloro, bromo or iodo, unless otherwise noted.

"Heterocycle" or "heterocyclyl" refers to a saturated, partially unsaturated or aromatic ring moiety having at least one ring heteroatom and at least one ring carbon atom. In one embodiment, the heteroatom is oxygen, sulfur, or nitrogen. A heterocycle containing more than one heteroatom may contain different heteroatoms. Heterocyclyl moieties include both monocyclic and multicyclic (e.g., bicyclic) ring moieties. Bicyclic ring moieties include fused, spirocyclic and bridged bicyclic rings and may comprise one or more heteroatoms in either of the rings. The ring attached to the remainder of the molecule may or may not contain a heteroatom. Either ring of a bicyclic heterocycle may be saturated, partially unsaturated or aromatic. The heterocycle may be attached to the rest of the molecule via a ring carbon atom, a ring oxygen atom or a ring nitrogen atom. Non-limiting examples of heterocycles are described below.

In one embodiment, a heterocyclyl is a fused bicyclic ring moiety wherein a 4-6 membered heterocyclic ring comprising 1-4 heteroatoms selected from oxygen, sulfur and nitrogen and a 4-6 membered heterocyclic ring comprising 0-4 heteroatoms selected from oxygen, sulfur and nitrogen are connected through two atoms. Exemplary heterocycles of this type include, but are not limited to, azaindolyl, dihydronaphthyridinyl, imidazopyridinyl, indolinyl, indolizinyl, isoquinolinyl, naphthyridinyl, pteridinyl, purinyl, pyrazolo[l,5-a]pyridinyl, quinolinyl, quinolizinyl, tetrahydroindolizinyl, tetrahydronaphthyridinyl, tetrahydroquinolizinyl,

4.5.6.7-tetrahydro-l,2-benzoxazolyl and triazolo[l,5-a]pyridinyl. In one embodiment, the heterocyclyl is selected from imidazo[l,5-a]pyridinyl, indolinyl, indolizinyl, isoquinolinyl, pyrazolo[l,5-a]pyridinyl, quinolinyl, 4,5,6,7-tetrahydro-l,2-benzoxazolyl and triazolo[l,5- ajpyridinyl.

Such fused ring may be attached to the rest of the molecule via a carbon atom or a nitrogen atom on either ring. A heterocycle of this type includes a bicyclic ring comprising only one nitrogen as the sole heteroatom when the nitrogen is located at the bridgehead.

Heterocyclic groups may be optionally substituted with one or more substituents as defined herein.

In one embodiment, a heterocyclyl is selected from furanyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxazolyl, naphthyridinyl, phthalazinyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridinyl, pyrimidinyl, quinazolinyl and a fused bicyclic ring moiety wherein a 6-membered heterocyclic ring comprising 2 nitrogen atoms and a 5-membered carbocyclic ring are connected through two atoms.

In one embodiment, a heterocyclyl is selected from naphthyridinyl, phthalazinyl, pyridinyl, pyrimidinyl, quinazolinyl and a fused bicyclic ring moiety wherein a 6-membered heterocyclic ring comprising 2 nitrogen atoms and a 5-membered saturated carbocyclic ring are connected through two carbon atoms.

In one embodiment, a heterocyclyl is selected from 6,7-dihydro-5H- cyclopenta[d]pyrimidinyl, furanyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxazolyl,

1.7-naphthyridinyl, phthalazinyl, pyranyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridinyl, pyrimidinyl and quinazolinyl.

“Optionally substituted” refers to“unsubstituted or substituted,” and therefore, the generic structural formulas described herein encompass compounds containing the specified optional substituent(s) as well as compounds that do not contain the optional substituent(s).

Each substituent is independently defined each time it occurs within the generic structural formula definitions.

Polymorphism

A compound of formula (I), (la) or (lb), including a salt or solvate thereof, may exist in crystalline form, non-crystalline form, or a mixture thereof. A compound or a salt or solvate thereof may also exhibit polymorphism, i.e. the capacity of occurring in different crystalline forms. These different crystalline forms are typically known as "polymorphs".

Polymorphs have the same chemical composition but differ in packing, geometrical arrangement, and other descriptive properties of crystalline solid state. Polymorphs, therefore, may have different physical properties such as shape, density, hardness, deformability, stability, and dissolution properties. Polymorphs typically exhibit different melting points, IR spectra, and X- ray powder diffraction patterns, all of which may be used for identification. One of ordinary skill in the art will appreciate that different polymorphs may be produced, for example, by changing or adjusting the conditions used in crystallizing/recrystallizing a compound of formula (I), (la) or (lb),.

Optical Isomers - Diastereomers - Geometric Isomers - Tautomers

Included herein are various isomers of the compounds of formula (I), (la) or (lb),. The term "isomers" refers to compounds that have the same composition and molecular weight but differ in physical and/or chemical properties. The structural difference may be in

constitution (geometric isomers) or in the ability to rotate the plane of polarized light

(stereosiomers).

With regard to stereoisomers, a compound of formula (I), (la) or (lb) may have one or more asymmetric carbon atom and may occur as a racemic mixture or as individual enantiomers or diastereomers. All such isomeric forms are included herein, including mixtures thereof. If a compound of formula (I), (la) or (lb) contains a double bond, the substituent may be in the E or Z configuration. If a compound of formula (I), (la) or (lb) contains a disubstituted cycloalkyl, the cycloalkyl substituent may have a cis- or trans- configuration. All tautomeric forms are also intended to be included.

Any asymmetric atom (e.g., carbon) of a compound of formula (I), (la) or (lb) can be present in racemic mixture or enantiomerically enriched, for example the (R)-, (S)- or (R,S)- configuration. In certain embodiments, each asymmetric atom has at least 50 % enantiomeric excess, at least 60 % enantiomeric excess, at least 70 % enantiomeric excess, at least 80 % enantiomeric excess, at least 90 % enantiomeric excess, at least 95 % enantiomeric excess, or at least 99 % enantiomeric excess in the (R)- or (S)- configuration. Substituents at atoms with unsaturated double bonds may, if possible, be present in cis- (Z)- or trans- (E)- form.

A compound of formula (I), (la) or (lb) can be in the form of one of the possible isomers, rotamers, atropisomers, tautomers or mixtures thereof, for example, as substantially pure geometric (cis or trans) isomers, diastereomers, optical isomers (antipodes), racemates or mixtures thereof.

Any resulting mixtures of isomers can be separated on the basis of the physicochemical differences of the constituents, into the pure or substantially pure geometric or optical isomers, diastereomers, racemates, for example, by chromatography and/or fractional crystallization.

Any resulting racemates of the final compounds of the examples or intermediates can be resolved into the optical antipodes by known methods, e.g., by separation of the diastereomeric salts thereof, obtained with an optically active acid or base, and liberating the optically active acidic or basic compound. In particular, a basic moiety may thus be employed to resolve the compounds of the present invention into their optical antipodes, e.g., by fractional crystallization of a salt formed with an optically active acid, e.g., tartaric acid, dibenzoyl tartaric acid, diacetyl tartaric acid, di-0,0'-p-toluoyl tartaric acid, mandelic acid, malic acid or camphor- lO-sulfonic acid. Racemic compounds can also be resolved by chiral chromatography, e.g., high pressure liquid chromatography (HPLC) using a chiral adsorbent.

Some of the compounds described herein may exist with different points of attachment of hydrogen, referred to as tautomers. For example, compounds including carbonyl -CFbC(O)- groups (keto forms) may undergo tautomerism to form hydroxyl - CH=C(OH)- groups (enol forms). Both keto and enol forms, individually as well as mixtures thereof, are included within the scope of the present invention.

Isotopic Variations

Compounds of formula (I), (la) or (lb) include unlabeled forms, as well as isotopically labeled forms. Isotopically labeled compounds have structures depicted by the formulas given herein except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Examples of isotopes that can be incorporated into compounds disclosed herein include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulfur, fluorine, iodine and chlorine, such as 2 H (i.e., Deuterium or“D”), ¾, U C, 13 C, 14 C, 13 N, 15 N, 15 0, 17 0, 18 0, 32 P, 35 S, 18 F, 123 I, 125 I and 36 Cl. The invention includes various isotopically labeled compounds as defined herein, for example those into which radioactive isotopes, such as ¾ and 14 C, or those into which non-radioactive isotopes, such as 2 H and 13 C are present. Such isotopically labelled compounds are useful in metabolic studies (with 14 C), reaction kinetic studies (with, for example 2 H or ¾), detection or imaging techniques, such as positron emission tomography (PET) or single-photon emission computed tomography (SPECT) including drug or substrate tissue distribution assays, or in radioactive treatment of patients. In particular, substitution with positron emitting isotopes, such as U C, 18 F, 15 0 and 13 N, may be particularly desirable for PET or SPECT studies.

Isotopically-labeled compounds of formula (I), (la) or (lb) can generally be prepared by conventional techniques known to those skilled in the art. Furthermore, substitution with heavier isotopes, particularly deuterium (i.e., 2 H or D) may afford certain therapeutic advantages resulting from greater metabolic stability, for example increased in vivo half-life or reduced dosage requirements or an improvement in therapeutic index.

Pharmaceutically Acceptable Salts

The term "pharmaceutically acceptable salt" refers to a salt prepared from a pharmaceutically acceptable non-toxic base or acid, including inorganic or organic base and inorganic or organic acid. Salts derived from inorganic bases include aluminum, ammonium, calcium, copper, ferric, ferrous, lithium, magnesium, manganic salts, manganous, potassium, sodium, zinc, and the like. Particular embodiments include ammonium, calcium, magnesium, potassium, and sodium salts. Salts in the solid form may exist in more than one crystal structure, and may also be in the form of hydrates. Salts derived from pharmaceutically acceptable organic non-toxic bases include salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, and basic ion exchange resins, such as arginine, betaine, caffeine, choline, N,N'-dibenzylethylene-diamine, diethylamine, 2- diethylaminoethanol, 2-dimethylaminoethanol, ethanolamine, ethylenediamine, N-ethyl- morpholine, N-ethylpiperidine, glucamine, glucosamine, histidine, hydrabamine,

isopropylamine, lysine, methylglucamine, morpholine, piperazine, piperidine, polyamine resins, procaine, purines, theobromine, triethylamine, trimethylamine, tripropylamine, tromethamine, and the like.

When a compound of formula (I), (la) or (lb) is basic, a salt may be prepared from a pharmaceutically acceptable non-toxic acid, including an inorganic and organic acid. Such acids include acetic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethanesulfonic, fumaric, gluconic, glutamic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phosphoric, succinic, sulfuric, tartaric, p- toluenesulfonic acid, trifluoroacetic acid (TFA) and the like. Particular embodiments include the citric, hydrobromic, hydrochloric, maleic, phosphoric, sulfuric, fumaric, tartaric and trifluoroacetic acids. It will be understood that, as used herein, references to the compounds disclosed herein are meant to also include pharmaceutically acceptable salts thereof.

Methods of Use

Compounds disclosed herein can inhibit activity of the enzyme indoleamine-2,3- dioxygenase (IDO). For example, the compounds disclosed herein can potentially be used to inhibit activity of IDO in cell or in an individual in need of modulation of the enzyme by administering an effective amount of a compound. Further disclosed herein are methods of inhibiting the degradation of tryptophan in a system containing cells expressing IDO such as a tissue, living organism, or cell culture. In some embodiments, the present invention provides methods of altering (e.g., increasing) extracellular tryptophan levels in a mammal by

administering an effective amount of a compound or composition provided herein. Methods of measuring tryptophan levels and tryptophan degradation are routine in the art.

Also disclosed herein are methods of inhibiting immunosuppression such as IDO- mediated immunosuppression in a patient by administering to the patient an effective amount of a compound or composition recited herein. IDO-mediated immunosuppression has been associated with, for example, cancers, tumor growth, metastasis, viral infection, viral replication, etc.

Also disclosed herein are methods of treating diseases associated with activity or expression, including abnormal activity and/or overexpression, of IDO in an individual (e.g., patient) by administering to the individual in need of such treatment an effective amount or dose of a compound disclosed herein or a pharmaceutical composition thereof. Example diseases can include any disease, disorder or condition that may be directly or indirectly linked to expression or activity of the IDO enzyme, such as over expression or abnormal activity. An IDO-associated disease can also include any disease, disorder or condition that may be prevented, ameliorated, or cured by modulating enzyme activity. Examples of IDO-associated diseases include cancer, viral infection such as HIV and HCV, depression, neurodegenerative disorders such as

Alzheimer's disease and Huntington's disease, trauma, age-related cataracts, organ

transplantation (e.g., organ transplant rejection), and autoimmune diseases including asthma, rheumatoid arthritis, multiple sclerosis, allergic inflammation, inflammatory bowel disease, psoriasis and systemic lupus erythematosusor. Example cancers potentially treatable by the methods herein include cancer of the colon, pancreas, breast, prostate, lung, brain, ovary, cervix, testes, renal, head and neck, lymphoma, leukemia, melanoma, and the like. The compounds of the invention may also be useful in the treatment of obesity and ischemia. As used herein, the term "cell" is meant to refer to a cell that is in vitro, ex vivo or in vivo. In some embodiments, an ex vivo cell can be part of a tissue sample excised from an organism such as a mammal. In some embodiments, an in vitro cell can be a cell in a cell culture. In some embodiments, an in vivo cell is a cell living in an organism such as a mammal.

As used herein, the term "contacting" refers to the bringing together of indicated moieties in an in vitro system or an in vivo system. For example, "contacting" the IDO enzyme with a compound disclosed herein includes the administration of a compound of the present invention to an individual or patient, such as a human, as well as, for example, introducing a compound of the invention into a sample containing a cellular or purified preparation containing the IDO enzyme.

A subject administered with a compound disclosed herein, or a pharmaceutically acceptable salt thereof, is generally a mammal, such as a human being, male or female. A subject also refers to cows, sheep, goats, horses, dogs, cats, rabbits, rats, mice, fish, and birds. In one embodiment, the subject is a human.

As used herein, the terms "treatment" and "treating" refer to all processes wherein there may be a slowing, interrupting, arresting, controlling, or stopping of the progression of a disease or disorder that may be associated with IDO enzyme activity. The terms do not necessarily indicate a total elimination of all disease or disorder symptoms. The terms also include the potential prophylactic therapy of the mentioned conditions, particularly in a subject that is predisposed to such disease or disorder.

The terms "administration of' and or "administering a" compound should be understood to include providing a compound described herein, or a pharmaceutically acceptable salt thereof, and compositions of the foregoing to a subject.

The amount of a compound administered to a subject is an amount sufficient to inhibit IDO enzyme activity in the subject. In an embodiment, the amount of a compound can be an“effective amount”, wherein the subject compound is administered in an amount that will elicit a biological or medical response of a tissue, system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinician. An effective amount does not necessarily include considerations of toxicity and safety related to the administration of a compound. It is recognized that one skilled in the art may affect physiological disorders associated with an IDO enzyme activity by treating a subject presently afflicted with the disorders, or by prophylactically treating a subject likely to be afflicted with the disorders, with an effective amount of a compound disclosed herein, or a pharmaceutically acceptable salt thereof.

An effective amount of a compound will vary with the particular compound chosen (e.g. considering the potency, efficacy, and/or half-life of the compound); the route of administration chosen; the condition being treated; the severity of the condition being treated; the age, size, weight, and physical condition of the subject being treated; the medical history of the subject being treated; the duration of the treatment; the nature of a concurrent therapy; the desired therapeutic effect; and like factors and can be routinely determined by the skilled artisan.

The compounds disclosed herein may be administered by any suitable route including oral and parenteral administration. Parenteral administration is typically by injection or infusion and includes intravenous, intramuscular, and subcutaneous injection or infusion.

The compounds disclosed herein may be administered once or according to a dosing regimen wherein a number of doses are administered at varying intervals of time for a given period of time. For example, doses may be administered one, two, three, or four times per day. Doses may be administered until the desired therapeutic effect is achieved or indefinitely to maintain the desired therapeutic effect. Suitable dosing regimens for a compound disclosed herein depend on the pharmacokinetic properties of that compound, such as absorption, distribution and half-life which can be determined by a skilled artisan. In addition, suitable dosing regimens, including the duration such regimens are administered, for a compound disclosed herein depend on the disease or condition being treated, the severity of the disease or condition, the age and physical condition of the subject being treated, the medical history of the subject being treated, the nature of concurrent therapy, the desired therapeutic effect, and like factors within the knowledge and expertise of the skilled artisan. It will be further understood by such skilled artisans that suitable dosing regimens may require adjustment given an individual subject's response to the dosing regimen or over time as the individual subject needs change. Typical daily dosages may vary depending upon the particular route of administration chosen. Typical daily dosages for oral administration, to a human weighing approximately 70 kg would range from about 0.1 mg to about 2 grams, or more specifically, 0.1 mg to 500 mg, or even more specifically, 0.2 mg to 100 mg, of a compound of formula (I), (la) or (lb).

One embodiment of the present invention provides for a method of treating a disease or disorder associated with IDO enzyme activity comprising administration of an effective amount of a compound of formula (I), (la) or (lb) to a subject in need of treatment thereof. In one embodiment, the disease or disorder associated with an IDO enzyme is a cell proliferation disorder.

In one embodiment, disclosed herein is the use of a compound of formula (I), (la) or (lb) in a therapy. The compound may be useful in a method of inhibiting IDO enzyme activity in a subject, such as a mammal in need of such inhibition, comprising administering an effective amount of the compound to the subject.

In one embodiment, disclosed herein is a pharmaceutical composition comprising a compound of formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof, for use in potential treatment of a disorder or disease related to IDO enzyme activity.

Compositions

The term "composition" as used herein is intended to encompass a dosage form comprising a specified compound in a specified amount, as well as any dosage form which results, directly or indirectly, from combination of a specified compound in a specified amount. Such term is intended to encompass a dosage form comprising a compound of formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers or excipients. Accordingly, the compositions of the present invention encompass any composition made by admixing a compound of the present invention and one or more pharmaceutically acceptable carrier or excipients. By "pharmaceutically acceptable" it is meant the carriers or excipients are compatible with the compound disclosed herein and with other ingredients of the composition.

In one embodiment, disclosed herein is a composition comprising a compound of formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers or excipients. The composition may be prepared and packaged in bulk form wherein an effective amount of a compound of the invention can be extracted and then given to a subject, such as with powders or syrups. Alternatively, the composition may be prepared and packaged in unit dosage form wherein each physically discrete unit contains an effective amount of a compound of formula (I), (la) or (lb). When prepared in unit dosage form, the composition of the invention typically contains from about 0.1 mg to 2 grams, or more specifically, 0.1 mg to 500 mg, or even more specifically, 0.2 mg to 100 mg, of a compound of formula (I), (la) or (lb), or a pharmaceutically acceptable salt thereof.

A compound disclosed herein and a pharmaceutically acceptable carrier or excipient(s) will typically be formulated into a dosage form adapted for administration to a subject by a desired route of administration. For example, dosage forms include those adapted for (1) oral administration, such as tablets, capsules, caplets, pills, troches, powders, syrups, elixirs, suspensions, solutions, emulsions, sachets, and cachets; and (2) parenteral administration, such as sterile solutions, suspensions, and powders for reconstitution. Suitable pharmaceutically acceptable carriers or excipients will vary depending upon the particular dosage form chosen. In addition, suitable pharmaceutically acceptable carriers or excipients may be chosen for a particular function that they may serve in the composition. For example, certain

pharmaceutically acceptable carriers or excipients may be chosen for their ability to facilitate the production of uniform dosage forms. Certain pharmaceutically acceptable carriers or excipients may be chosen for their ability to facilitate the production of stable dosage forms. Certain pharmaceutically acceptable carriers or excipients may be chosen for their ability to facilitate the carrying or transporting of a compound disclosed herein, once administered to the subject, from one organ or portion of the body to another organ or another portion of the body. Certain pharmaceutically acceptable carriers or excipients may be chosen for their ability to enhance patient compliance.

Suitable pharmaceutically acceptable excipients include the following types of excipients: diluents, lubricants, binders, disintegrants, fillers, glidants, granulating agents, coating agents, wetting agents, solvents, co-solvents, suspending agents, emulsifiers, sweeteners, flavoring agents, flavor masking agents, coloring agents, anti -caking agents, hemectants, chelating agents, plasticizers, viscosity increasing agents, antioxidants, preservatives, stabilizers, surfactants, and buffering agents.

A skilled artisan possesses the knowledge and skill in the art to select suitable pharmaceutically acceptable carriers and excipients in appropriate amounts for the use in the invention. In addition, there are a number of resources available to the skilled artisan, which describe pharmaceutically acceptable carriers and excipients and may be useful in selecting suitable pharmaceutically acceptable carriers and excipients. Examples include Remington's Pharmaceutical Sciences (Mack Publishing Company), The Handbook of Pharmaceutical Additives (Gower Publishing Limited), and The Handbook of Pharmaceutical Excipients (the American Pharmaceutical Association and the Pharmaceutical Press).

The compositions of the invention are prepared using techniques and methods known to those skilled in the art. Some methods commonly used in the art are described in Remington's Pharmaceutical Sciences (Mack Publishing Company). In one embodiment, the invention is directed to a solid oral dosage form such as a tablet or capsule comprising an effective amount of a compound of the invention and a diluent or filler. Suitable diluents and fillers include lactose, sucrose, dextrose, mannitol, sorbitol, starch (e.g. corn starch, potato starch, and pre-gelatinized starch), cellulose and its derivatives, (e.g. microcrystalline cellulose), calcium sulfate, and dibasic calcium phosphate. The oral solid dosage form may further comprise a binder. Suitable binders include starch (e.g. com starch, potato starch, and pre-gelatinized starch) gelatin, acacia, sodium alginate, alginic acid, tragacanth, guar gum, povidone, and cellulose and its derivatives (e.g. microcrystalline cellulose). The oral solid dosage form may further comprise a disintegrant. Suitable

disintegrants include crospovidone, sodium starch glycolate, croscarmelose, alginic acid, and sodium carboxymethyl cellulose. The oral solid dosage form may further comprise a lubricant. Suitable lubricants include stearic acid, magnesium stearate, calcium stearate, and talc.

Where appropriate, dosage unit formulations for oral administration can be microencapsulated. The composition can also be prepared to prolong or sustain the release as, for example, by coating or embedding particulate material in polymers, wax, or the like.

The compounds disclosed herein may also be coupled with soluble polymers as targetable drug carriers. Such polymers can include polyvinylpyrrolidone, pyrancopolymer, polyhydroxypropylmethacrylamidephenol, polyhydroxyethylaspartamidephenol, or

polyethyleneoxidepolylysine substituted with palmitoyl residues. Furthermore, the compounds of the invention may be coupled to a class of biodegradable polymers useful in achieving controlled release of a drug, for example polylactic acid, polepsilon caprolactone, polyhydroxy butyric acid, polyorthoesters, polyacetals, polydihydropyrans, polycyanacrylates and cross- linked or amphipathic block copolymers of hydrogels.

In one embodiment, the invention is directed to a liquid oral dosage form. Oral liquids such as solution, syrups and elixirs can be prepared in dosage unit form so that a given quantity contains a predetermined amount of a compound disclosed herein. Syrups can be prepared by dissolving the compound of the invention in a suitably flavored aqueous solution; while elixirs are prepared through the use of a non-toxic alcoholic vehicle. Suspensions can be formulated by dispersing a compound disclosed herein in a non-toxic vehicle. Solubilizers and emulsifiers such as ethoxylated isostearyl alcohols and polyoxy ethylene sorbitol ethers, preservatives, flavor additives such as peppermint oil or other natural sweeteners or saccharin or other artificial sweeteners and the like can also be added. In one embodiment, the invention is directed to compositions for parenteral administration. Compositions adapted for parenteral administration include aqueous and non- aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The compositions may be presented in unit-dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze dried (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets.

Combinations

A compound disclosed herein may be used in combination with one or more other active agents, including but not limited to, other anti-cancer agents, that are used in the prevention, treatment, control, amelioration, or reduction of risk of a particular disease or condition (e.g., cell proliferation disorders). In one embodiment, a compound disclosed herein is combined with one or more other anti-cancer agents for use in the prevention, treatment, control amelioration, or reduction of risk of a particular disease or condition for which the compounds disclosed herein are useful. Such other active agents may be administered, by a route and in an amount commonly used therefor, contemporaneously or sequentially with a compound of the present invention.

When a compound disclosed herein is used contemporaneously with one or more other active agents, a composition containing such other active agents in addition to the compound disclosed herein is contemplated. Accordingly, the compositions of the present invention include those that also contain one or more other active ingredients, in addition to a compound disclosed herein. A compound disclosed herein may be administered either simultaneously with, or before or after, one or more other therapeutic agent(s). A compound disclosed herein may be administered separately, by the same or different route of

administration, or together in the same pharmaceutical composition as the other agent(s).

Products provided as a combined preparation include a composition comprising a compound of formula (I), (la) or (lb) and one or more other active agent(s) together in the same pharmaceutical composition, or a compound of formula (I), (la) or (lb) and one or more other therapeutic agent(s) in separate form, e.g. in the form of a kit. The weight ratio of a compound disclosed herein to a second active agent may be varied and will depend upon the effective dose of each agent. Generally, an effective dose of each will be used. Thus, for example, when a compound disclosed herein is combined with another agent, the weight ratio of the compound disclosed herein to the other agent will generally range from about 1000: 1 to about 1 :1000, such as about 200: 1 to about 1 :200. Combinations of a compound disclosed herein and other active agents will generally also be within the

aforementioned range, but in each case, an effective dose of each active agent should be used. In such combinations, the compound disclosed herein and other active agents may be administered separately or in conjunction. In addition, the administration of one element may be prior to, concurrent to, or subsequent to the administration of other agent(s).

In one embodiment, the invention provides a composition comprising a compound of formula (I), (la) or (lb) and at least one other therapeutic agent as a combined preparation for simultaneous, separate or sequential use in therapy. In one embodiment, the therapy is the treatment of a disease or disorder associated with IDO enzyme activity.

In one embodiment, the invention provides a kit comprising two or more separate pharmaceutical compositions, at least one of which contains a compound of formula (I), (la) or (lb). In one embodiment, the kit comprises means for separately retaining said compositions, such as a container, divided bottle, or divided foil packet. An example of such a kit is a blister pack, as typically used for the packaging of tablets, capsules and the like.

A kit disclosed herein may be used for administering different dosage forms, for example, oral and parenteral, for administering the separate compositions at different dosage intervals, or for titrating the separate compositions against one another. To assist with compliance, a kit of the invention typically comprises directions for administration.

Disclosed herein is a use of a compound of formula (I), (la) or (lb) for treating a disease or disorder associated with IDO enzyme activity, wherein the medicament is prepared for administration with another active agent. The invention also provides the use of another active agent for treating a disease or disorder associated with an IDO enzyme, wherein the medicament is administered with a compound of formula (I), (la) or (lb).

The invention also provides the use of a compound of formula (I), (la) or (lb) for treating a disease or disorder associated with IDO enzyme activity, wherein the patient has previously (e.g. within 24 hours) been treated with another active agent. The invention also provides the use of another therapeutic agent for treating a disease or disorder associated with IDO enzyme activity, wherein the patient has previously (e.g. within 24 hours) been treated with a compound of formula (I), (la) or (lb). The second agent may be applied a week, several weeks, a month, or several months after the administration of a compound disclosed herein.

In one embodiment, the other active agent is selected from the group consisting of vascular endothelial growth factor (VEGF) receptor inhibitors, topoisomerase II inhibitors, smoothen inhibitors, alkylating agents, anti-tumor antibiotics, anti-metabolites, retinoids, immunomodulatory agents including but not limited to anti-cancer vaccines, CTLA-4, LAG-3 and PD- 1 antagonists.

Examples of vascular endothelial growth factor (VEGF) receptor inhibitors include, but are not limited to, bevacizumab (sold under the trademark AVASTIN by

Genentech/Roche), axitinib, (N-methyl-2-[[3 -[([pound])-2-pyri din-2 -ylethenyl]-l H-indazol-6- yl]sulfanyl]benzamide, also known as AG013736, and described in PCT Publication No. WO 01 /002369), Brivanib Alaninate ((S)-((R)-l-(4-(4-Fluoro-2-methyl-lH-indol-5-yloxy)-5- methylpyrrolo[2,l-f][l,2,4]triazin-6-yloxy)propan-2-yl)2-ami nopropanoate, also known as BMS- 582664), motesanib (N-(2,3-dihydro-3,3-dimethyl-l H-indoi-6-yl)-2-[(4- pyridinyimethyj)amino]-3-pyfidinecarboxamide. and described in PCT Publication No. WO 02/068470), pasireotide (also known as SO 230, and described in PCT Publication No. WO 02/010192), and sorafenib (sold under the tradename NEXAVAR).

Examples of topoisomerase II inhibitors, include but are not limited to, etoposide (also known as VP-16 and Etoposide phosphate, sold under the tradenames TOPOSAR,

VEPESID and ETOPOPHOS), and teniposide (also known as VM-26, sold under the tradename VUMON).

Examples of alkylating agents, include but are not limited to, 5-azacytidine (sold under the trade name VIDAZA), decitabine (sold under the trade name of DECOGEN), temozolomide (sold under the trade names TEMODAR and TEMODAL by Schering- Plough/Merck), dactinomycin (also known as actinomycin-D and sold under the tradename COSMEGEN), melphalan (also known as L-PAM, L-sarcolysin, and phenylalanine mustard, sold under the tradename ALKERAN), altretamine (also known as hexamethylmelamine (HMM), sold under the tradename HEXALEN), carmustine (sold under the tradename BCNU), bendamustine (sold under the tradename TREANDA), busulfan (sold under the tradenames BETSLiLFEX and MYLERAN), carboplatin (sold under the tradename PARAPLATIN), lomustine (also known as CCNU, sold under the tradename CeeNU), cisplatin (also known as CDDP, sold under the tradenames PLATINOL and PLATINOL-AQ), chlorambucil (sold under the tradename LELTKERAN), cyclophosphamide (sold under the tradenames CYTOXAN and NEOSAR), dacarbazine (also known as DTIC, DIC and imidazole carboxamide, sold under the tradename DTIC -DOME), altretamine (also known as hexamethylmelamine (HMM) sold under the tradename HEXALEN), ifosfamide (sold under the tradename IFEX), procarbazine (sold under the tradename MATUEANE), mechlorethamine (also known as nitrogen mustard, mustine and mechloroethamine hydrochloride, sold under the tradename METSTARGEN), streptozocin (sold under the tradename ZANOSAR), thiotepa (also known as thiophosphoamide, TESPA and TSPA, and sold under the tradename THIOPLEX).

Examples of anti-tumor antibiotics include, but are not limited to, doxorubicin (sold under the tradenames ADRIAMYCIN and REIBEX), bleomycin (sold under the tradename LENOXANE), daunorubicin (also known as dauorubicin hydrochloride, daunomycin, and rubidomycin hydrochloride, sold under the tradename CEREIBIDINE), daunorubicin liposomal (daunorubicin citrate liposome, sold under the tradename DAUNOXOME), mitoxantrone (also known as DHAD, sold under the tradename NOVANTRONE), epirubicin (sold under the tradename ELLENCE), idarubicin (sold under the tradenames IDAMYCIN, IDAMYCIN PFS), and mitomycin C (sold under the tradename METTAMYCIN).

Examples of anti-metabolites include, but are not limited to, claribine (2- chlorodeoxyadenosine, sold under the tradename LEETSTATIN), 5-fluorouracil (sold under the tradename ADRETCIL), 6-thioguanine (sold under the tradename PERINETHOL), pemetrexed (sold under the tradename ALIMTA), cytarabine (also known as arabinosyl cytosine (Ara-C), sold under the tradename CYTOSAR-U), cytarabine liposomal (also known as Liposomal Ara- C, sold under the tradename DEPOCYT), decitabine (sold under the tradename DACOGEN), hydroxyurea (sold under the tradenames HYDREA, DROXIA and MYLOCEL), fludarabine (sold under the tradename FLETDARA), floxuridine (sold under the tradename FETDR), cladribine (also known as 2-chlorodeoxyadenosine (2-CdA) sold under the tradename

LEETSTATIN), methotrexate (also known as amethopterin, methotrexate sodium (MTX), sold under the tradenames RHEETMATREX and TREXALL), and pentostatin (sold under the tradename NIPENT).

Examples of retinoids include, but are not limited to, alitretinoin (sold under the tradename PANRETIN), tretinoin (all-trans retinoic acid, also known as ATRA, sold under the tradename VESANOID), Isotretinoin (l3-c/s-retinoic acid, sold under the tradenames

ACCUTANE, AMNESTEEM, CLARAVIS, CLARUS, DECUTAN, ISOTANE, IZOTECH, ORATANE, ISOTRET, and SOTRET), and bexarotene (sold under the tradename

TARGRETIN). "PD-l antagonist" means any chemical compound or biological molecule that blocks binding of PD-L1 expressed on a cancer cell to PD-l expressed on an immune cell (T cell, B cell or NKT cell) and preferably also blocks binding of PD-L2 expressed on a cancer cell to the immune-cell expressed PD-l. Alternative names or synonyms for PD-l and its ligands include: PDCD1, PD1, CD279 and SLEB2 for PD-l; PDCD1L1, PDL1, B7H1, B7-4, CD274 and B7-H for PD-L1; and PDCD1L2, PDL2, B7-DC, Btdc and CD273 for PD-L2. In any of the treatment method, medicaments and uses of the present invention in which a human individual is being treated, the PD-l antagonist blocks binding of human PD-L1 to human PD-l, and preferably blocks binding of both human PD-L1 and PD-L2 to human PD-l. Human PD-l amino acid sequences can be found in NCBI Locus No.: NP 005009. Human PD-L1 and PD-L2 amino acid sequences can be found in NCBI Locus No.: NP_054862 and NP_0795l5, respectively.

PD-l antagonists useful in any of the treatment method, medicaments and uses of the present invention include a monoclonal antibody (mAh), or antigen binding fragment thereof, which specifically binds to PD-l or PD-L1, and preferably specifically binds to human PD-l or human PD-L1. The mAh may be a human antibody, a humanized antibody or a chimeric antibody, and may include a human constant region. In some embodiments the human constant region is selected from the group consisting of IgGl, IgG2, IgG3 and IgG4 constant regions, and in preferred embodiments, the human constant region is an IgGl or IgG4 constant region. In some embodiments, the antigen binding fragment is selected from the group consisting of Fab, Fab'-SH, F(ab') 2 , scFv and Fv fragments. Examples of PD-l antagonists include, but are not limited to, pembrolizumab (sold under the tradename KEYTRUDA) and nivolumab (sold under the tradename OPDIVO).

Examples of mAbs that bind to human PD-l, and useful in the treatment method, medicaments and uses of the present invention, are described in ETS7488802, US7521051, US8008449, US8354509, US8168757, W02004/004771, W02004/072286, W02004/056875, and US2011/0271358.

Examples of mAbs that bind to human PD-L1, and useful in the treatment method, medicaments and uses of the present invention, are described in W02013/019906, W02010/077634 Al and ETS8383796. Specific anti-human PD-L1 mAbs useful as the PD-l antagonist in the treatment method, medicaments and uses of the present invention include MPDL3280A, BMS-936559, MEDI4736, MSB0010718C and an antibody which comprises the heavy chain and light chain variable regions of SEQ ID NO:24 and SEQ ID NO:2l, respectively, of WO2013/019906.

Other PD-l antagonists useful in any of the treatment method, medicaments and uses of the present invention include an immunoadhesin that specifically binds to PD-l or PD- Ll, and preferably specifically binds to human PD-l or human PD-L1, e.g., a fusion protein containing the extracellular or PD-l binding portion of PD-L1 or PD-L2 fused to a constant region such as an Fc region of an immunoglobulin molecule. Examples of immunoadhesion molecules that specifically bind to PD-l are described in W02010/027827 and WO2011/066342. Specific fusion proteins useful as the PD-l antagonist in the treatment method, medicaments and uses of the present invention include AMP-224 (also known as B7-DCIg), which is a PD-L2-FC fusion protein and binds to human PD-l.

Examples of other cytotoxic agents include, but are not limited to, arsenic trioxide (sold under the tradename TRISENOX), asparaginase (also known as L-asparaginase, and Erwinia L-asparaginase, sold under the tradenames ELSPAR and KIDROLASE).

EXPERIMENTAL

The following examples are intended to be illustrative only and not limiting in any way. Abbreviations used are those conventional in the art or the following.

ACN acetonitrile

Boc tert-butyloxy carbonyl

°C degree Celsius

DCM dichloromethane

DIEA diisopropylethylamine

DMF N,N-dimethylformamide

EMEM Eagle's minimal essential medium

EtOAc ethyl acetate

EtOH ethanol

g gram

h hour(s)

HATU (l-[Bis(dimethylamino)methylene]-lH-l,2,3-triazolo[4,5-b]pyr idinium 3- oxide hexafluorophosphate)

HPLC high pressure liquid chromatography kg kilogram

L liter

LC liquid chromatography

LC-MS liquid chromatography and mass spectrometry

MeOH methanol

MS mass spectrometry

MTBE methyl /er/-butyl ether

min minutes

mL or ml milliliter(s)

m/z mass to charge ratio

nm nanometer

nM nanomolar

NMP N-methyl-2-pyrrolidone

N normal

RPMI medium Roswell Park Memorial Institute medium

RT or rt room temperature

sat. saturated

SFC supercritical fluid chromatography

TEA triethylamine

TFA trifluoroacetic acid

THF tetrahydrofuran

GENERAL SYNTHETIC SCHEMES

The compounds of formula (I), (la) or (lb) may be prepared by methods known in the art of organic synthesis as set forth in part by the following synthetic schemes and synthetic procedures and conditions for the illustrative intermediates and examples.

In the schemes described below, it is well understood that protecting groups for sensitive or reactive groups are employed where necessary in accordance with general principles of chemistry. Protecting groups are manipulated according to standard methods of organic synthesis (T. W. Greene and P. G. M. Wuts, "Protective Groups in Organic Synthesis", 3rd edition, Wiley, NY 1999). These groups are removed at a convenient stage of the compound synthesis using methods that are readily apparent to those skilled in the art. The compounds described herein may be made from commercially available starting materials or synthesized using known organic, inorganic, and/or enzymatic processes.

Scheme 1

Compounds of formula I-Ia, I-Ib and I-Ic may be prepared according to Scheme 1. Commercially available alcohol 1-1 can be converted to the corresponding nitrile 1-2 in a variety of ways, for example, by step-wise treatment with methanesulfonyl chloride and trimethylamine to make the mesylate, followed by reaction of the mesylate with sodium cyanide in a solvent such as DMF. Alkylation to the nitrile group of 1-2 can be accomplished by initial treatment with two equivalents of a base, such as lithium bis(trimethylsilyl) amide, followed by the addition of an alkyl halide, such as iodomethane to afford compounds 1-3. Hydrolysis of the nitrile group of intermediate 1-3 can be achieved in a variety of ways, including by treatment with aqueous potassium hydroxide in a solvent such as ethanol.

The resulting carboxylic acids 1-4 can be converted to amides 1-5 using amines and any of a number of amide coupling reagents, for example l-[bis(dimethylamino)methylene]- lH-l,2,3-triazolo[4,5-b]pyridinium 3-oxide hexafluorophosphate in the presence of a base such as diisopropylethylamine. The tert-butoxy carbonyl protective group present in amides 1-5 may be removed with an acid such as hydrogen chloride or trifluoroacetic acid to afford amines 1-6. Amines 1-6, in turn, may be converted to aryl or heteroaryl amines or alkyl amines. For example, heteroarylamines I-Ia may be prepared by nucleophilic aromatic substitution by reaction with a heteroaryl halide in the presence of a base such as trimethylamine or sodium bicarbonate in a solvent such as THF or DMF at elevated temperatures. Alternatively, alkylamines I-Ia can be prepared by reaction of amines 1-6 with an aldehyde in the presence of an appropriate reducing agent such as sodium triacetoxy borohydride. Racemic compounds of the formula I-Ia may be separated to two single enantiomers I-Ib and I-Ic by preparative chiral chromatography.

Scheme 2

In Scheme 2, compounds of the formula I-Ib and I-Ic can be alternatively prepared in chiral form by initial preparative chiral HPLC separation of the enantiomers of intermediate 1-5 to afford single enantiomers 1-5-1 (first eluting peak) and 1-5-2 (second eluting peak). These can be carried out in the same fashion as described in Scheme 1 to give compounds I-Ib and I-Ic directly without the need for any further separation.

EXAMPLES

Example 1: N-(4-chlorophenv0-2-(3-(Y2-methylpyrimidin-4-v0amino)bicvclo r 1.1. llpentan-l- vDpropanamide (racemic)

Step 1 : butyl (3-(cvanomethvQ bicvclo G1.1.11 pentan-l-vO carbamate

To a solution of /c/7-butyl (3 -(hydroxymethyl )bicyclo[ 1.1.1 ]pentan-l- yl)carbamate (1.1 g, 5.16 mmol) in DCM (17.2 ml) at 0° C was added triethyl amine (0.86 ml, 6.2 mmol) and methane sulfonyl chloride (0.42 ml, 5.42 mmol). The mixture was stirred at 0° C for 1 h, then diluted with DCM and sat. NaHCO, solution. The organic layer was separated, washed with brine, dried over MgS0 4 , and concentrated. The residue was taken up in 15 ml of DMF followed by the addition of NaCN (1.01 g, 20.6 mmol). The mixture was stirred at 60° C for 4 h. The reaction mixture was cooled down, diluted with sat. NaHCO, aqueous solution, and extracted with EtOAc. The organic layer was separated, washed with brine, dried over MgS0 4 , and concentrated. The residue was purified by flash chromatography (40 g silica column, 0-50% ethyl acetate/hexanes) to afford the title compound as a solid.

Step 2: Tert- butyl (3-P -cyanoethyl ) bicycle G1.1. P pentan-l-yl) carbamate

To a solution of /ert-butyl (3-(cyanomethyl) bicyclo [1.1.1] pentan-l-yl) carbamate (1.12 g, 5.04 mmol) in THF (6 ml) at -30 °C was added lithium bis (trimethyl silyl) amide (11.1 ml, 11.1 mmol). The mixture was stirred at -30 °C for 20 min, and then the solution of methyl iodide (0.32 ml, 5.0 mmol) in 1 ml of THF was added dropwise. After addition, the reaction mixture was stirred under around -20°C for 30 min. The reaction mixture was diluted with sat. NaHCO,, and extracted with EtOAc. The organic layer was separated, washed with brine, dried over MgSOq, and concentrated. The residue was purified by flash chromatography

(0-50% EtO Ac/hexanes) to afford the title compound as a solid.

Step 3: 2-(3 butoxycarbonvO amino) bicvclo G1.1.11 pentan-l-yl) propanoic

To a microwave reaction vial was added KOH (950 mg, 16.9 mmol), H 2 0 (2.1 mL), followed by a solution of tert- butyl (3 -(1 -cyanoethyl) bicyclo [1.1.1] pentan-l-yl) carbamate (100 mg, 0.42 mmol) in EtOH (2.1 mL). The resultant solution was heated at 100 °C for 18 h. The mixture was cooled down, neutralized with 1N HC1 to pH 3~4. The mixture was extracted with 25% IPA in chloroform twice. The combined organics were washed with brine, dried over MgS0 4 , and concentrated to afford the title compound as a solid. This material was used directly for next step. Step 4: Tert- butyl (3-(T-(Y4-chlorophenvD amino)- l-oxopropan-2-nP bicyclo G1.1.11 pentan-l- vD carbamate

To a solution of 2-(3-((/er/-butoxycarbonyl) amino) bicyclo [1.1.1] pentan-l-yl) propanoic acid (108 mg, 0.423 mmol) in DMF (1.5 ml) was added 4-chloroaniline (108 mg, 0.846 mmol), HATU (209 mg, 0.55 mmol) and DIEA (0.30 ml, 1.69 mmol). The mixture was stirred at RT overnight. The reaction mixture was diluted with sat. NaHC03, and extracted with EtOAc. The organic layer was separated, washed with water, brine, dried over MgSC>4, and concentrated. The residue was purified by flash chromatography (24g silica column, 0-50% EtO Ac/hexanes) to afford the title compound as a solid.

Step 5: 2-(3-Aminobicvclo G1.1.11 pentan- 1 -yl )-N-(4-chlorophenyl ) propanamide hydrochloride

To a solution of /c/V-butyl (3-(l-((4-chlorophenyl) amino)- l-oxopropan-2-yl) bicyclo [1.1.1] pentan-l-yl) carbamate (25 mg, 0.069 mmol) in dioxane (0.5 ml) was added HC1 (4.0M in dioxane, 1.0 ml). The mixture was stirred at RT overnight. Then the resulting mixture was concentrated under reduced pressure to afford the title compound as a solid. The resulting product was used for next step without further purification. Step 6: N-(4-chlorophenv0-2-(3-(Y2-methylpyrimidin-4-v0amino)bicvclo [ 1.1. llpentan-l- vQpropanamide

To a solution of 2-(3-aminobicy clo[ 1.1.1 ]pentan-l-yl)-N-(4- chlorophenyl)propanamide hydrochloride (25 mg, 0.083 mmol) in THF (1 ml) was added triethylamine (0.046 ml, 0.332 mmol) and 4-bromo-2-methylpyrimidine (21.54 mg, 0.124 mmol). The mixture was stirred at 80 °C overnight The reaction mixture was diluted with sat. NaHCO, solution and extracted with EtOAc. The organic layer was separated, washed with brine, dried over MgSOzp and concentrated. The residue was purified by flash chromatography (12 g silica column, 0-50% 3 : 1 EtOAc: ethanol//hexanes) to afford racemic N-(4-chlorophenyl)- 2-(3-((2-methylpyrimidin-4-yl)amino)bicyclo[l . l . l]pentan-l-yl)propanamide as a solid.

Examples 2 and 3: N-(4-chlorophenvO-2-(3-(Y2-methylpyrimidin-4- vDamino)bicvclor 1.1.1 Ipentan- 1 -vQpropanamide (enantiomers)

The racemic compound from Step 6 was submitted for chiral SFC separation (Column OJ-H, 2lX 250, Condition MeOH+O.25% DMEA) to afford two chiral isomers as solids.

Example 2 (isomer 1, peak 1): LCMS: 357 0 [M+H] + ; ¾ NMR (600 MHz, DMSO-^e) d 9.93 (s, 1H), 7.97 (s, 1H), 7.80 (s, 1H), 7.63 (d, j = 8.9 Hz, 2H), 7.35 (d, J = 8.8 Hz, 2H), 6.29 (s, 1H),

2.74 (q, J= 6.8 Hz, 1H), 2.31 (s, 3H), 2.04 - 1.83 (m, 6H), 1.07 (d, J = 6.8 Hz, 3H).

Example 3 (isomer 2, peak 2): LCMS: 357.0 [M+H] + ; ¾ NMR (600 MHz, DMSO-^e) d 9.93 (s, 1H), 7.97 (s, 1H), 7.80 (s, 1H), 7.63 (d, J = 8.9 Hz, 2H), 7.35 (d, J = 8.8 Hz, 2H), 6.29 (s, 1H), 2.74 (q, J= 6.8 Hz, 1H), 2.31 (s, 3H), 2.04 - 1.83 (m, 6H), 1.07 (d, J = 6.8 Hz, 3H).

Intermediate 1 : 2-(3-Aminobicvclor 1.1.1 Ipentan- l-v0-N-(4-fluorophenv0propanamide hydrochloride

HCI

Step 1. Tert- butyl (3 -(1 -(Y4-fluorophenvDamino)- 1 -oxopropan-2-vDbicvclor 1 1.1 Ipentan- 1 - vDcarbamate

To a stirred solution of 2-(3-((/er/-butoxy carbonyl)amino)bicyclo[ 1.1.1 ]pentan-l- yl)propanoic acid (12 g, 47.0 mmol) and HATH (20.55 g, 54.1 mmol) in dry DMF (100 ml) were added DIEA ( (24.6 ml, 141 mmol) and 4-fluoroaniline (Aldrich) (5.80 ml, 61.1 mmol). The mixture was stirred at RT overnight. LCMS showed the desired product as the major product. The mixture was partitioned between EtOAc (200 ml) and water (100 ml). The aqueous was extracted with EtOAc for three times. Organic phases were separated, combined and dried over Na 2 S0 4 , then filtered and concentrated. The crude residue was purified by Teledyne Isco system, using 120 g silica combiflash gold column and 0-100% EtOAc in hexane as eluting solvent to give the title compound as a solid. LC-Mass (M+H) calc. = 349.18 ; found title compound M+H = 349; M+H-56 (-tBu) = 293.22

Step 2. 2-(3-Aminobicvclor 1.1. npentan-l-vO-N-(4-fluorophenvOpropanamide hydrochloride

The title compound was prepared in an analogous fashion as described in

Example 1, Step 5. LC-Mass (M+H) calc. = 249 ; found M+H = 249

ETsing analogous chemistry to that shown in Example 1, Step 6, the compounds in the Table 1 were prepared starting from 2-(3-aminobicyclo[ 1.1.1 ]pentan-l -yl)-N-(4- fluorophenyl)propanamide hydrochloride (Intermediate 1).

Table 1

Synthesis of the compounds shown in Table 2 is describe below.

To each monomer (Ar-Y, 0. l2lmmol; Y represents halide) was added lml NMP solution of 2-(3-aminobicyclo[l .1. l]pentan-l-yl)-N-(4-fluorophenyl)propanamide hydrochloride (30mg, 0. l2lmmol). To each reaction was added NaHCCb (50.7mg, 0.604mmol). The reactions were then sealed and heated at 130 °C for 16 h. They were then filtered and submitted for HPLC purification.

Table 2

Intermediates 2 and 3. (R)-2-(3 - Aminobicvclor 1.1.1 Ipentan- 1 -yl)-N-(4- fluorophenyl )propanamide hydrochloride and (S)-2-(3 -aminobicvclor 1.1.1 Ipentan-l -yl)-N-(4- fluorophenyl )propanamide hydrochloride

Intermediate 2 Intermediate 3

Step 1. Tert- butyl (3-(T-(Y4-fluorophenvOamino)-l-oxopropan-2-vObicvclor 1.1. ljpentan-l- vDcarbamate

To the stirred solution of 2-(3-((/er/-butoxycarbonyl)amino)bicyclo[ 1.1.1 jpentan- l-yl)propanoic acid (12 g, 47.0 mmol) and HATU (20.55 g, 54.1 mmol) in dry DMF (100 ml) were added DIEA ( (24.6 ml, 141 mmol) and 4-fluoroaniline (Aldrich) (5.80 ml, 61.1 mmol). The mixture was stirred at RT overnight. LCMS showed the desired product as the major product. The mixture was partitioned between EtOAc (200 ml) and water (100 ml). The aqueous was extracted with EtOAc for three times. Organic phases were separated, combined and derived over Na 2 S0 4 , then filtered and concentrated. The crude residue was purified by Teledyne Isco system, using 120 g silica combiflash gold column and 0-100% EtOAc in hexane as eluting solvent to give the title compound as a solid. LC-Mass (M+H) calc. = 349.18 ; found M+H = 349; M+H-56 (-tBu) = 293.22 Step 2. Tert- butyl (R)-(3-(T-(Y4-fluorophenvOamino)-l-oxopropan-2-vObicvclor 1.1. llpentan-l- vDcarbamate and tert- butyl (SV(3-(T-(Y4-fluorophenvOamino)-l-oxopropan-2- vDbicvclor 1.1.1 Ipentan- 1 -vOcarbamate

The racemic compound from Step 1 was resolved by chiral SFC using an OJ- H, 50 x 250mm column to give both enantiomers as solids. The absolute stereochemistry of each enantiomer was independently determined by Vibrational Circular Dichroism.

The absolute configuration of peak 1 was assigned to be ( R ) using Vibrational Circular Dichroism (VCD) spectroscopy. Analysis was done comparing experimental data to the calculated VCD and IR spectra of the (R) configuration. The experimental VCD spectrum of peak 1 matched with the calculated (R) spectrum over the region from 1000-1850 cm 1 .

The absolute configuration of peak 2 was assigned to be ( S) using Vibrational Circular Dichroism (VCD) spectroscopy. Analysis was done comparing experimental data to the calculated VCD and IR spectra of the ( R ) configuration. The experimental VCD spectrum of peak 2 matched with the mirror image of the calculated (R) spectrum over the region from 1000- 1850 cm 1 .

Peak 1 : LC-Mass (M+H) calc. = 349.18 ; found isomer 1 : M+H = 349; M+H-56 (-tBu) = 293.21 Peak 2: LC-Mass (M+H) calc. = 349.18 ; found isomer 2: M+H = 349; M+H-56 (-tBu) = 293.21 Step 3A. (R)-2-(3-Aminobicvclor 1.1.1 Ipentan- l-vD-N-(4-fluorophenvDpropanamide

hydrochloride

Tert- butyl (R)-(3 -( 1 -((4-fluorophenyl)amino)- 1 -oxopropan-2- yl)bicyclo[l. l.l]pentan-l-yl)carbamate (2130 mg, 6.11 mmol) was dissolved in HC1 (4.0 M in dioxane) (38.2 ml, 153 mmol). The mixture was stirred at RT for about 2 h. LCMS showed the reaction was complete, and the desired product as the major product. The mixture was concentrated in vacuo to afford the title compound as a solid. LC-MS (M+H) calc. = 249.13; found: M+H = 249.13 Step 3B. (S)-2-(3-Aminobicvclor 1.1. l1pentan-l-vD-N-(4-fluorophenvDpiOpanamide hydrochloride

Tert- butyl (S)-(3 -( 1 -((4-fluorophenyl)amino)- 1 -oxopropan-2- yl)bicyclo[l . l . l]pentan-l-yl)carbamate (2130 mg, 6.11 mmol) was dissolved in HC1 (4.0 M in dioxane) (38.2 ml, 153 mmol). The mixture was stirred at RT for about 2 h. LCMS showed the reaction was complete, and the desired product as the major product. The mixture was concentrated in vacuo to afford the title compound as a solid. LC-MS (M+H) calc. = 249.13; found: M+H = 249.13

Example 27. (S)-N-(4-fluorophenvn-2-(3-((2-(trifluoromethvnpyridin-4- vDamino)bicvclor 1.1.1 Ipentan- 1 -vOpropanamide

To a microwave with (S)-2-(3-aminobicyclo[ 1.1.1 ]pentan-l -yl)-N-(4- fhiorophenyl)propanamide, HC1 (150 mg, 0.527 mmol), sodium bicarbonate (133 mg, 1.580 mmol) and 4-fluoro-2-(triflouromethyl)pyridine (MBBC) (104 mg, 0.632 mmol) was added NMP (2634 mΐ). The vial was capped and stirred at 1 l0°C overnight. LCMS showed reaction completed, and desired product as the major product. The mixture was filtered, and was purified by mass directed reverse HPLC purification (Reverse HPLC on a 19x100 mm, Waters CSH Cl 8 column, 5m particle size, flow rate 25ml/min, linear gradient, 10% ACN/H20 to 45 %

ACN/H 2 O, total run time 12 min, buffering with 0.16% TFA) to afford the desired product as a solid after lyophilization. LCMS m/z (M+H) calc’d: 394.15; found (M+H) : 394.27. 1 H NMR (500 MHz, CD 3 OD): d 8.19 (d, J = 6.6 Hz,lH); 7.56 (dd, J = 4.85, 9.10 Hz, ,2 H); 7.11-7.04 (m, 4H); 2.82 (q, j = 7.5 Hz, lH); 2.67 (S, 1 H); 2.16 (dd, j = 9.45, 15.45 Hz, 6H); 1.21 (d, 7 = 6.9 Hz, 3 H)

Compounds in Table 3 were prepared in a synthetic route as shown below: F

rt, 4h rt, 1-2h

Step 1. Representative procedure:

To the stirred solution of (S)-2-(3-aminobicyclo[ 1.1.1 ]pentan-l -yl)-N-(4- fluorophenyl)propanamide, HC1 (35 mg, 0.123 mmol) in DCM (1229 mΐ) were added DIEA (Aldrich) (42.9 mΐ, 0.246 mmol) and (3-chloro-phenyl)-acetaldehyde (15.39 mΐ, 0.123 mmol) . The mixture was stirred at RT for 2 min, then was added sodium triacetoxyborohydride (Aldrich) (104 mg, 0.492 mmol). The mixture was stirred at RT for additional 4 h, LC-Mass showed desired product as the major product. The reaction was quenched with addition of TFA(Aldrich) (18.94 mΐ, 0.246 mmol) and MeOH (~ lml), then concentrated. The residue was re-dissolved in DMSO (- 1.8 ml), filtered and purified by the mass-directed reverse phase HPLC purification using the following condition: Reverse HPLC on a 19x100 mm, Waters CSH C18 column, 5m particle size, flow rate 25ml/min, linear gradient, 15% ACN/H2O to 50 % ACN/H2O, total run time 8 min, buffering with 0.16% TFA, followed by Genevac drying to afford the desired product (S)-2-(3-((3-chlorobenzyl)amino)bicyclo[l .1. l]pentan-l-yl)-N-(4- fluorophenyl)propanamide, TFA as a TFA salt. LCMS m/z (M+H) calc’d: 373.14; found (M+H) : 372.98.

Step 2. Representative procedure:

To a stirred solution of (S)-2-(3-aminobicyclo[l. l. l]pentan-l-yl)-N-(4- fluorophenyl)propanamide, HC1 (35 mg, 0.123 mmol) in DCM (1229 mΐ) were added DIEA (Aldrich) (42.9 mΐ, 0.246 mmol) and (3-chloro-phenyl)-acetaldehyde (15.39 mΐ, 0.123 mmol).

The mixture was stirred at RT for 2 min, then was added sodium triacetoxyborohydride (Aldrich) (104 mg, 0.492 mmol). The mixture was stirred at RT for additional 4 h, LC-Mass showed desired NH product (product from step 1) as the major product. To this mixture was added 4 A molecular sieve (Aldrich, activated) (~ 150 mg), followed by addition of formaldehyde (37% in water, 73.2 mΐ, 0.983 mmol), and additional sodium triacetoxyborohydride (Aldrich) (104 mg, 0.492 mmol). The mixture was stirred at RT for additional 1 h. LCMS check showed the desired N-Me product as the desired product.

The reaction was quenched with the addition of TFA (Aldrich) (18.94 mΐ, 0.246 mmol) and MeOH (~ 2 ml). The mixture was mixed well and then filtered, and the filtrate was concentrated. The residue was re-dissolved in DMSO (~ 1.8 ml), filtered and purified by the mass-directed reverse phase HPLC purification using the following condition: Reverse HPLC on a 19x100 mm, Waters CSH C18 column, 5m particle size, flow rate 25ml/min, linear gradient, 15% ACN/H2O to 50 % ACN/H2O, total run time 8 min, buffering with 0.16% TFA, followed by Genevac drying of collected fractions to afford the N-Me product (S)-2-(3-((3- chlorobenzyl)(methyl)amino)bicyclo[ 1.1.1 Jpentan- 1 -yl)-N-(4-fluorophenyl)propanamide, TFA as a TFA salt. LCMS m/z (M+H) calc’d: 387.16; found (M+H): 387.01.

Table 3

Biological Assays

IDOl Cellular Assay in Hela Cells Stimulated with IFNy

Hela cells were cultured in complete Hela culture medium (90% EMEM, 10% heat-inactivated fetal bovine serum) and expanded to about lxlO 9 cells. The cells were then collected and frozen down at lxlO 7 cells/vial in 1 mL frozen medium (90% complete Hela culture medium, lO% DMSO)

Compounds to be tested were serially diluted in ten 3 -fold steps in DMSO starting from 10 mM DMSO stocks in Echo low volume plate(s). Compound dilutions or DMSO alone were then dispensed from the dilution plate(s) into Greiner black 384-well assay plate(s) (catalog #781086, 50 nL/well) using an Echo 550 acoustic liquid handler (Labcyte).

Frozen Hela cells were thawed and transferred into Hela assay medium (99% complete Hela culture medium, 1% Pen/Strep) with 20 mL medium/vial of cells. The cells were spun down at 250g in a table top centrifuge for 5 min and suspended in same volume of Hela assay medium. The cells were then counted and adjusted to a density of 2 x 10 5 cells/ml in Hela assay medium. Sterile L-tryptophan were added to the cells with final concentration of 300 uM L-tryptophan. A small aliquot (2 mL/plate) of Hela cells were set aside and were not treated with åFNy, to serve as the Max-E control. The rest of Hela cells were added with sterile IFNy (Cat# 285-IF, R & D systems) with a final concentration of 100 ng/mL.

Hela cells with and without IFNy were dispensed to the respective wells of 384- well assay plates containing the compounds. The plates were incubated for about 48 hours at a 37 °C, 5% C0 2 incubator. Afterwards, 12 pL of 0.5 M methyl isonipecotate in dimethyl sulfoxide were added into each well and the plates were sealed and incubated at 37 °C without CO2 overnight. The plates were centrifuged for 1 min at 200xg. The resulting fluorescence was measured in a Spectramax plate reader (Molecular Devices) with a 400 nm excitation filter and a 510 nm emission filter.

The fluorescence intensity of each well was corrected for the background observed in wells with non-IFNy-treated cells and was expressed as a fraction of the intensity observed in wells of rFNy-treated cells and DMSO only. Potencies were calculated by linear least squares fit to the four parameter logistic IC50 equation.

The biological activity data using the IDOl cellular assay described above are summarized in the table below. Compounds disclosed herein generally have IC50 of about 1 nM to about 1,000 nM, or more specifically, about 2 nM to about 800 nM, or more specifically, about 5 nM to about 600 nM, or still more specifically, about 10 nM to about 500 nM. Such a result is indicative of the intrinsic activity of the compounds in use as an inhibitor of an IDO enzyme. Specific IC50 activity data for the exemplified compounds disclosed herein is provided in the following table.

IDOl Human whole blood Assay

Compounds to be tested were serially diluted in ten 3 -fold steps in DMSO starting from 10 mM. 3 pL of compound dilutions or DMSO alone were then dispensed from the dilution plate into a polypropylene 96-well assay plate containing 97 pL of RPMI medium using an Echo 555 acoustic liquid handler (Labcyte). LPS and IFNy was prepared in RPMI medium to a 10X of final cone. (1000 ng/mL), final concentration is 100 ng/mL. Human whole blood was drawn in sodium heparin coated tubes from healthy internal donors. Two hundred forty pL of blood was transferred to each of the wells of a v- bottom 96 well plate. Thirty pL of compound was transferred from intermediate dilution plate, and incubated for 15 min. Thirty pL from stimulants was then transferred to blood and mixed thoroughly. Plate was covered with breathable membrane and incubated at 37 °C for overnight (18 h).

On day 2 isotope labeled standard of kynurenine and tryptophan was made in water at lOx concentration and 30 pL was added to the blood at 3 pM final concentration. The assay plates were centrifuged at 300xG for 10 min with no brake to separate plasma from red blood cells. Sixty pL of plasma samples was removed without disturbing red blood cells. Plasma was diluted with RPMI in 1 : 1 ratio and proteins were precipitated out with two volume of acetonitrile. The plates were centrifuged at 4000xG for 60 min. Twenty pL of supernatant was carefully transferred to a 384 well plate containing 40 pL of 0.1% formic acid in water and analyzed by LC/MS/MS.

LC/MS/MS analyses were performed using Thermo Fisher's LX4-TSQ Quantum Ultra system. This system consists of four Agilent binary high-performance liquid

chromatography (HPLC) pumps and a TSQ Quantum Ultra triple quadruple MS/MS instrument. For each sample, 5 pL were injected onto an Atlantis T3 column (2.1 mm x 150 mm, 3 pm particle size) from Waters. The mobile phase gradient pumped at 0.8 mL/min was used to elute the analytes from the column at 25 °C. The elution started at 0% B increasing linearly to 25% B at 6.5 min, holding at 25% for 1 min, re-equilibrating to 10 min. Mobile phase A consisted of 0.1% formic acid in water. Mobile phase B consisted of 0.1% of formic acid in acetonitrile.

Data was acquired in positive mode using a HESI interface. The operational parameters for the TSQ Quantum Ultra instrument were a spray voltage of 4000 V, capillary temperature of 380 °C, vaporizer temperature 400 °C, shealth gas 60 arbitrary units, Aux gas 20 arbitrary units, tube lens 85 and collision gas 1.2 mTorr. SRM chromatograms of kynurenine (Ql : 209.2>Q3:94.0) and internal standard (Ql : 215.3>Q3 :98.2) were collected for 90 sec. The peak area was integrated by Xcalibur Quan software. The ratios between the kynurenine generated in the reaction and 2D6-Kynurenine spiked-in internal standard were used to generate percentage inhibition and IC50 values. Compounds were titrated and ICso’s were calculated by 4 parameter sigmoidal curve fitting formula.

The biological activity data of selective compounds using the IDOl human whole blood assay described above are summarized in the table below.

While the invention has been described and illustrated with reference to certain particular embodiments thereof, those skilled in the art will appreciate that various adaptations, changes, modifications, substitutions, deletions, or additions of procedures and protocols may be made without departing from the spirit and scope of the invention.