Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
3-SUBSTITUTED-1,2,4-OXADIAZOLE AND THIADIAZOLE COMPOUNDS AS IMMUNOMODULATORS
Document Type and Number:
WIPO Patent Application WO/2016/142886
Kind Code:
A2
Abstract:
The present invention relates to 3-substituted-1,2,4-oxadiazole and thiadiazole compounds of formula (I) or formula (II) and their use to inhibit the programmed cell death 1 (PD-1) signaling pathway and/or for treatment of disorders by inhibiting an immunosuppressive signal induced by PD-1, PD-L1 or PD-L2.

Inventors:
SASIKUMAR POTTAYIL GOVINDAN NAIR (IN)
RAMACHANDRA MURALIDHARA (IN)
PRASAD APPUKKUTTAN (IN)
NAREMADDEPALLI SEETHARAMAIAH SETTY SUDARSHAN (IN)
Application Number:
PCT/IB2016/051343
Publication Date:
September 15, 2016
Filing Date:
March 09, 2016
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
AURIGENE DISCOVERY TECH LTD (IN)
International Classes:
A61K31/00
Other References:
See references of EP 3267985A4
Download PDF:
Claims:
We Claim:

1. A compound of formula (I):

or a pharmaceutically acceptable salt thereof or a stereoisomer thereof;

wherein,

X is O or S;

each dotted line [— ] independently represents an optional bond;

Rj is hydrogen or -CO-Aaa;

Aaa represents an amino acid residue;

R2 is side chain of an amino acid, hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, heterocyclylalkyl heteroaralkyl, aralkyl, heteroaryl or aryl, each optionally substituted by one or more substituents selected from carboxylate, carboxylic acid, carboxylic acid ester, thiocarboxylate, thio acid, amido, amino, heterocyclyl, hydroxyl, cycloalkyl, aryl, aryl-COOH, heteroaryl, guanidino, amidino, -NH, -N(alkyl), -SH and - S(alkyl), optionally wherein two or three carbon atoms of the alkyl, alkenyl or alkynyl form part of a 3-7-membered carbocyclic or heterocyclic ring which is optionally substituted with 1 to 4 substituents, each independently selected from alkyl, alkoxy, carboxylic acid, carboxylate and hydroxyl;

R3 is aryl, heteroaryl, heterocyclyl or cycloalkyl; wherein the said aryl, heteroaryl, heterocyclyl or cycloalkyl is optionally substituted by 1 to 4 occurrences of

Ra, independently for each occurrence, is alkyl, alkoxy, halo, hydroxyl, amino, - C(0)OH, aralkyl, aryl, alkoxy, heteroaralkyl, heteroaryl, cycloalkyl, (cycloalkyl)alkyl, hydroxyalkyl, alkoxyalkyl or acyl; or any two Ra groups attached to the same carbon atom together represent an oxo (=0) or thioxo (=S);

each of R4 and R5 independently is hydrogen or absent; and

R6 is hydrogen or alkyl.

2. The compound of claim 1 , wherein the compound is of formula (IA):

or a pharmaceutically acceptable salt thereof or a stereoisomer thereof;

wherein,

R2 is (C1-C6)alkyl, cycloalkyl, (C2-C6)alkynyl or heteroarylalkyl; wherein (Q- Ce)alkyl or heteroarylalkyl is substituted with one or more substituents selected from carboxylic acid, carboxylate, thiocarboxylate, thioacid, amido, amino, hydroxyl, cycloalkyl, aryl, aryl-COOH, heterocyclyl, heteroaryl, guanidino, amidino, -SH and - S(alkyl); optionally wherein two or three carbon atoms of the alkyl, alkenyl or alkynyl form part of a 3-7-membered carbocyclic or heterocyclic ring optionally substituted with 1 to 4 substituents independently selected from alkyl, alkoxy, carboxylic acid, carboxylate and hydroxyl; and

Ri, R3 and R6 are same as defined in claim 1.

3. The compound of claim 1 or 2, wherein the compound is of formula (IB):

(IB)

wherein,

R2, R3, R6 and Aaa are same as defined in claim 1.

4. The compound of claim 3, wherein Aaa is a natural amino acid residue.

5. The compound of claim 3 or 4, wherein Aaa is Ser, Met, Thr, Gly, Lys, Phe or Ala.

6. The compound of any one of claims 3 to 5, wherein Aaa is Ser or Thr.

7. The compound of any one of claims 1 to 6, wherein R3 is aryl, cycloalkyl, heterocyclyl or heteroaryl optionally substituted by 1 to 4 occurrences of Ra; wherein Ra is as defined in claim 1.

8. The compound of any one of claims 1 to 7, wherein Ra is hydrogen, alkyl, halo, hydroxyl, amino, -C(0)OH, aralkyl, aryl, alkoxy, heteroaralkyl, heteroaryl, cycloalkyl,

(cycloalkyl)alkyl, hydroxyalkyl, alkoxyalkyl or acyl.

The compound of any one of claims 1 to 7, wherein any two Ra groups attached same carbon atom together represent an oxo (=0) or thioxo (=S).

11. The compound of any one of claims 1 to 10, wherein R2 is (Ci-Ce)alkyl, (C2- Ce)alkenyl or (C2-Ce)alkynyl substituted by one or more substituents selected from carboxylic acid, carboxylate, thiocarboxylate, thioacid, amido, amino and amidino.

12. The compound of any one of claims 1 to 11 wherein R2 is (Ci-Ce)alkyl substituted by carboxylic acid, amido or amidino.

13. The compound of any one of claims 1 to 12, wherein R2 is (Ci-C6)alkyl substituted by -C(0)OH, -C(0)NH2 or -C(=NH)NH2.

14. The compound of any one of claims 1 to 13, wherein R2 is -(CH2)COOH, - (CH2)2COOH, -(CH2)CONH2, -(CH2)2CONH2 or -(CH2)C(=NH)NH2.

15. The compound of any one of claims 1 to 14, wherein R2 is -(CH2)COOH or - (CH2)CONH2.

16. The compound of any one of claims 1 to 15, wherein R6 is hydrogen.

17. A compound of formula (II),

or a pharmaceutically acceptable salt thereof or a stereoisomer thereof;

wherein,

Y and Z are each independently -CRaRb-, -NRC-, O or S;

X is O or S;

each dotted line [— ] independently represents an optional bond;

Ra and R are each independently hydrogen or a substituent such as alkyl, acyl, hydroxyl, amino, halo, aralkyl, aryl, heteroaralkyl, heteroaryl, cycloalkyl, aminoalkyl, alkoxy hydroxyalkyl, alkoxyalkyl or (cycloalkyl)alkyl; preferably hydroxyl, amino, lower alkyl, lower acyl or lower aralkyl;

Rc is hydrogen or a substituent, such as alkyl, acyl, aralkyl, aryl, heteroaralkyl, heteroaryl, cycloalkyl or (cycloalkyl)alkyl; preferably lower alkyl, lower acyl or lower aralkyl;

Rj is hydrogen or -CO-Aaa;

Aaa represents an amino acid residue;

R2 is side chain of an amino acid, hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, heterocyclylalkyl, heteroaralkyl, aralkyl, heteroaryl or aryl, each optionally substituted by one or more substituents selected from carboxylate, carboxylic acid, carboxylic acid ester, thiocarboxylate, thio acid, amido, amino, heterocyclyl, hydroxyl, cycloalkyl, aryl, aryl-COOH, heteroaryl, guanidino, amidino, -SH and -S(alkyl), optionally wherein two or three carbon atoms of the alkyl, alkenyl or alkynyl form part of a 3-7-membered carbocyclic or heterocyclic ring (such as a cyclobutyl or oxirane ring) which is optionally substituted with 1 to 4 substituents, each independently selected from alkyl, alkoxy, carboxylic acid, carboxylate and hydroxyl;

each of R4 and R5 independently is hydrogen or absent; and

R7 represents 0-4 substituents on the ring to which it is attached, wherein each substituent is independently selected from alkyl, aralkyl, aryl, alkoxy, heteroaralkyl, heteroaryl, halo, cycloalkyl, (cycloalkyl)alkyl, amino, -C(0)OH, hydroxyl, hydroxyalkyl, alkoxyalkyl or acyl; preferably lower alkyl, lower acyl or lower aralkyl; or two R7 groups attached to the same carbon atom together represent an oxo (=0) or thioxo (=S);

18. The compound of claim 17, wherein X is O and each dotted line [— ] represents a bond.

19. The compound of claim 17 or 18, wherein Rj is -CO-Aaa.

20. The compound of any one of claims 17 to 19, wherein Aaa represents an amino acid residue wherein the amino acid residue comprises a side chain that includes a -OH, - O-acyl, -SH, -NH2, -NH(alkyl) or -S(alkyl) moiety. 21. The compound of any one of claims 17 to 20, wherein R2 represents (Ci-Ce)alkyl, (C2-Ce)alkenyl or (C2-Ce)alkynyl substituted by one or more substituents selected from carboxylate, carboxylic acid, carboxylic acid ester, thiocarboxylate, thio acid, amido, amino and heterocyclyl, preferably from lower alkyl, lower acyl or lower aralkyl, optionally wherein two or three carbon atoms of the (Ci-Ce)alkyl, (C2-C6)alkenyl or (C2- Ce)alkynyl form part of a 3-7-membered carbocyclic or heterocyclic ring (such as a cyclobutyl or oxirane ring).

22. The compound of any one of claims 17 to 21, wherein R7 represents 0-4 substituents on the ring to which it is attached, wherein each substituent is independently selected from alkyl, aralkyl, aryl, alkoxy, heteroaralkyl, heteroaryl, halo, cycloalkyl, (cycloalkyl)alkyl, amino, hydroxyl, hydroxyalkyl, alkoxyalkyl or acyl; preferably lower alkyl, lower acyl or lower aralkyl.

23. A compound selected from the group consisting of:

or a pharmaceutically acceptable salt thereof or a stereoisomer thereof.

24. A pharmaceutical composition comprising a compound of any one of claims 1 to 23 and a pharmaceutically acceptable carrier.

25. A use of a compound of any one of claims 1 to 23 in the manufacture of a medicament for the treatment of cancer.

26. The use of claim 25, wherein the cancer is selected from lung cancer, breast cancer, colon cancer, renal cancer, bladder cancer, thyroid cancer, prostate cancer, osteosarcoma and Hodgkin's lymphoma.

27. A method of treating cancer, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of any one of claims 1 to 23.

28. The method of claim 27, wherein the cancer is selected from lung cancer, breast cancer, colon cancer, renal cancer, bladder cancer, thyroid cancer, prostate cancer, osteosarcoma and Hodgkin's lymphoma.

29. The method of claim 27 or 28, wherein the subject is a mammal, e.g., a human. 30. The method of any one of claims 27 to 28, further comprising conjointly administering to the subject a second chemotherapeutic agent.

31. The method of any one of claims 27 to 29, further comprising conjointly administering to the subject one or more non-chemical cancer treatments, e.g., radiation therapy, surgery, thermo ablation, focused ultrasound therapy or cryotherapy.

32. A method for inhibiting the PD-1 pathway (e.g., PD-1 , PD-L1 or PD-L2) in a subject, comprising administering to the subject a therapeutically effective amount of a compound of any one of claims 1 to 23.

33. A method for treating a bacterial, viral or fungal infection or an immunological condition, comprising administering to a subject in need thereof a therapeutically effective amount of a compound of any one of claims 1 to 23.

34. A use of a compound of any one of claims 1 to 23 in the manufacture of a medicament for the treatment of bacterial, viral or fungal infection or an immunological condition.

35. A use of a compound of any one of claims 1 to 23 in inhibiting the PD-1 pathway (e.g., PD-1 , PD-L1 or PD-L2).

36. Compound according to any one of claims 1 to 23 for use as a medicament.

37. Compound according to any one of claims 1 to 23 for use in the treatment of cancer.

38. Compound according to any one of claims 1 to 23 for use in the treatment of bacterial, viral or fungal infection or an immunological condition.

Description:
3-SUBSTITUTED-l,2,4-OXADIAZOLE AND THIADIAZOLE COMPOUNDS AS

IMMUNOMODULATORS

This application claims the benefit of Indian provisional application number 1174/CHE/2015, filed on March 10, 2015 and Indian provisional application number 1176/CHE/2015, filed on March 10, 2015; the specifications of which are hereby incorporated by reference herein in their entirety.

TECHNICAL FIELD

The present invention relates to 3-substituted-l,2,4-oxadiazole and thiadiazole compounds and their derivatives therapeutically useful as immune modulators. The invention also relates to pharmaceutical compositions comprising 3-substituted 1,2,4- oxadiazole and thiadiazole compounds and their derivatives as therapeutic agents.

BACKGROUND OF THE INVENTION

Immune system in mammals sustains the ability to control the homeostasis between the activation and inactivation of lymphocytes through various regulatory mechanisms during and after an immune response. Among these mechanisms, there are mechanisms that specifically modulate the immune response as and when required. Mechanism via PD-1 pathway relates to almost every aspect of immune responses including autoimmunity, tumour immunity, infectious immunity, transplantation immunity, allergy and immunological privilege. PD-1 (or Programmed Cell Death 1 or PDCD1) is a ~55kD type I membrane glycoprotein and is a receptor of the CD28 superfamily that negatively regulates T cell antigen receptor signalling by interacting with the specific ligands and is suggested to play significant role in the maintenance of self-tolerance.

The PD-1 protein's structure comprises of an extracellular IgV domain followed by a trans-membrane region and an intracellular tail. The intracellular tail contains two phosphorylation sites located in an immunoreceptor tyrosine-based inhibitory motif and an immunoreceptor tyrosine-based switch motif, which suggests that PD-1 negatively regulates TCR signals. Also, PD-1 is expressed on the surface of activated T cells, B cells and macrophages, (Y. Agata et al., Int Immunol, May 1996, 8, 765) suggesting that compared to CTLA-4 [(Cytotoxic T-Lymphocyte Antigen 4), also known as CD 152 (Cluster of differentiation 152), a protein that also plays an important regulatory role in the immune system], PD-1 more broadly negatively regulates immune responses.

Indeed, functional "exhaustion" (immune dysfunction) among T and B cell subsets is a well-described feature of chronic viral infections, such as hepatitis B and C and HIV viruses. T cell exhaustion was initially described for CD8 T cells in mice chronically infected with lymphocytic choriomeningitis virus clone 13. In the lymphocytic choriomeningitis virus mouse model, repeated antigen stimulation through the T cell antigen receptor drives the sustained expression of T cell inhibitory receptors, including programmed cell death- 1 (PD-1) and lymphocyte activationgene-3 (LAG-3), on virus-specific CD8 T cells (Joseph Illingworth et al., Journal of Immunology (2013), 190(3), 1038-1047).

Blockade of PD-1, an inhibitory receptor expressed by T cells, can overcome immune resistance. PD-1 is a key immune check point receptor expressed by activated T cells and it mediates immune suppression. PD-1 functions primarily in peripheral tissues, where T cells may encounter the immune suppressive PD-1 ligands; PD-L1 (B7-H1) and PD-L2 (B7-DC), which are expressed by tumor cells, stromal cells or both. Inhibition of the interaction between PD-1 and PD-L1 can enhance T-cell responses in vitro and mediate preclinical antitumor activity (Suzanne L. Topalian et al., N Engl J Med. 2012, 366(26): 2443-2454).

PD-1 plays critical roles in the regulation of the immune response to cancer, allergy and chronic viral infection (Julie R. Brahmer et al., N Engl J Med. 2012, 366(26): 2455-2465).

Tumour cells and virus (including HCV and HIV) infected cells are known to exploit the PD-1 signalling pathway (to create Immunosuppression) in order to escape immune surveillance by host T cells. It has been reported that the PD-1 gene is one of genes responsible for autoimmune diseases like systemic lupus erythematosus (Prokunina et al, Nature Genetics, 2002, Vol. 32, No. 4, 666-669.).

International applications, WO2011161699 and WO2012168944 report peptides and their derivatives derived from PD-1 ectodomain capable of inhibiting the programmed cell death 1 (PD-1) signalling pathway. Further, WO2013144704 and WO2013132317 report cyclic peptides and peptidomimetic compounds as therapeutic agents capable of inhibiting the programmed cell death 1 (PD-1) respectively. Furthermore, International applications, WO2011161699, WO2012168944, WO2013144704 and WO2013132317 report peptides or peptidomimetic compounds which are capable of suppressing and/or inhibiting the programmed cell death 1 (PD-1) signaling pathway.

Still there is a need for more potent, better and/or selective immune modulators of

PD-1 pathway.

SUMMARY OF INVENTION

The present invention relates to 3-substituted 1,2,4-oxadiazole and thiadiazole compounds or a stereoisomer thereof or a pharmaceutically acceptable salt thereof. The compounds of present invention are capable of suppressing and/or inhibiting the programmed cell death 1 (PD- 1) signalling pathway.

In one aspect, the presen nd of formula (I):

wherein,

X is O or S;

each dotted line [— ] independently represents an optional bond;

Rj is hydrogen or -CO-Aaa;

Aaa represents an amino acid residue;

P2 is side chain of an amino acid, hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, heterocyclylalkyl heteroaralkyl, aralkyl, heteroaryl or aryl, each optionally substituted by one or more substituents selected from carboxylate, carboxylic acid, carboxylic acid ester, thiocarboxylate, thio acid, amido, amino, heterocyclyl, hydroxyl, cycloalkyl, aryl, aryl-COOH, heteroaryl, guanidino, amidino, -NH, -N(alkyl), -SH and - S(alkyl), optionally wherein two or three carbon atoms of the alkyl, alkenyl or alkynyl form part of a 3-7-membered carbocyclic or heterocyclic ring which is optionally substituted with 1 to 4 substituents, each independently selected from alkyl, alkoxy, carboxylic acid, carboxylate and hydroxyl; R3 is aryl, heteroaryl, heterocyclyl or cycloalkyl; wherein the said aryl, heteroaryl, heterocyclyl or cycloalkyl is optionally substituted by 1 to 4 occurrences of

R a , independently for each occurrence, is alkyl, alkoxy, halo, hydroxyl, amino, - C(0)OH, aralkyl, aryl, alkoxy, heteroaralkyl, heteroaryl, cycloalkyl, (cycloalkyl)alkyl, hydroxyalkyl, alkoxyalkyl or acyl; or any two R a groups attached to the same carbon atom together represent an oxo (=0) or thioxo (=S);

each of R4 and R5 independently is hydrogen or absent;

R6 is hydrogen or alkyl;

or a pharmaceutically acceptable salt thereof or a stereoisomer thereof.

In another aspect, the present invention provides compounds of formula (II):

wherein,

Y and Z are each independently -CR a R b -, -NR C -, O or S;

X is O or S;

each dotted line [— ] independently represents an optional bond;

R a and R are each independently hydrogen or a substituent such as alkyl, acyl, hydroxyl, amino, halo, aralkyl, aryl, heteroaralkyl, heteroaryl, cycloalkyl, aminoalkyl, alkoxy hydroxyalkyl, alkoxyalkyl or (cycloalkyl)alkyl; preferably hydroxyl, amino, lower alkyl, lower acyl or lower aralkyl;

R c is hydrogen or a substituent, such as alkyl, acyl, aralkyl, aryl, heteroaralkyl, heteroaryl, cycloalkyl or (cycloalkyl)alkyl; preferably lower alkyl, lower acyl or lower aralkyl;

Ri is hydrogen or -CO-Aaa;

Aaa represents an amino acid residue; R2 is hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, heterocyclylalkyl heteroaralkyl, aralkyl, heteroaryl or aryl, each optionally substituted by one or more substituents selected from carboxylate, carboxylic acid, carboxylic acid ester, thiocarboxylate, thio acid, amido, amino, heterocyclyl, hydroxyl, cycloalkyl, aryl, aryl- COOH, heteroaryl, guanidino, amidino, -SH and -S(alkyl), optionally wherein two or three carbon atoms of the alkyl, alkenyl or alkynyl form part of a 3-7-membered carbocyclic or heterocyclic ring (such as a cyclobutyl or oxirane ring) which is optionally substituted with 1 to 4 substituents, each independently selected from alkyl, alkoxy, carboxylic acid, carboxylate and hydroxyl;

each of R4 and R5 independently is hydrogen or absent; and

R 7 represents 0-4 substituents on the ring to which it is attached, wherein each substituent is independently selected from alkyl, aralkyl, aryl, alkoxy, heteroaralkyl, heteroaryl, halo, cycloalkyl, (cycloalkyl)alkyl, amino, -C(0)OH, hydroxyl, hydroxyalkyl, alkoxyalkyl or acyl; preferably lower alkyl, lower acyl or lower aralkyl; or two R 7 groups attached to the same carbon atom together represent an oxo (=0) or thioxo (=S);

or a pharmaceutically acceptable salt thereof or a stereoisomer thereof.

In another aspect, the present invention relates to a process for preparation of compounds of formula (I) or formula (II) or a pharmaceutically acceptable salt or a stereoisomer thereof.

In a further aspect of the present invention, it relates to the pharmaceutical composition comprising a compound of formula (I) or formula (II) or a pharmaceutically acceptable salt or a stereoisomer and processes for preparing such compositions.

In yet another aspect of the present invention, it provides use of 3-substituted 1,2,4-oxadiazole and thiadiazole compounds and derivatives of formula (I) or formula (II), salts and stereoisomers thereof, which are capable of suppressing and/or inhibiting the programmed cell death 1 (PD-1) signaling pathway. For example, these compounds can be used to treat one or more diseases characterized by aberrant or undesired activity of the PD- 1 signaling pathway.

DETAILED DESCRIPTION OF THE INVENTION

The present invention provides 3-substituted 1,2,4-oxadiazole and thiadiazole compounds and their derivatives as therapeutic agents useful for treatment of disorders via immunopotentiation comprising inhibition of immunosuppressive signal induced due to PD-1 , PD-L1 or PD-L2 and therapies using them.

Each embodiment is provided by way of explanation of the invention and not by way of limitation of the invention. In fact, it will be apparent to those skilled in the art that various modification and variations can be made in the present invention without departing from the scope or spirit of the invention. For instance, features illustrated or described as part of one embodiment can be used on another embodiment to yield a still further embodiment. Thus it is intended that the present invention cover such modifications and variations as come within the scope of the appended claims and their equivalents. Other objects, features and aspects of the present invention are disclosed in or are obvious from, the following detailed description. It is to be understood by one of ordinary skill in the art that the present discussion is a description of exemplary embodiments only and is not to be construed as limiting the broader aspects of the present invention.

In certain embodiments, the present invention provides compounds of formula

(I):

or a pharmaceutically acceptable salt thereof or a stereoisomer thereof;

wherein,

X is O or S;

each dotted line [— ] independently represents an optional bond;

Ri is hydrogen or -CO-Aaa;

Aaa represents an amino acid residue;

P2 is side chain of an amino acid, hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, heterocyclylalkyl heteroaralkyl, aralkyl, heteroaryl or aryl, each optionally substituted by one or more substituents selected from carboxylate, carboxylic acid, carboxylic acid ester, thiocarboxylate, thio acid, amido, amino, heterocyclyl, hydroxyl, cycloalkyl, aryl, aryl-COOH, heteroaryl, guanidino, amidino, -NH, -N(alkyl), -SH and - S(alkyl), optionally wherein two or three carbon atoms of the alkyl, alkenyl or alkynyl form part of a 3-7-membered carbocyclic or heterocyclic ring which is optionally substituted with 1 to 4 substituents, each independently selected from alkyl, alkoxy, carboxylic acid, carboxylate and hydroxyl;

R3 is aryl, heteroaryl, heterocyclyl or cycloalkyl; wherein the said aryl, heteroaryl, heterocyclyl or cycloalkyl is optionally substituted by 1 to 4 occurrences of

R a , independently for each occurrence, is alkyl, alkoxy, halo, hydroxyl, amino, - C(0)OH, aralkyl, aryl, alkoxy, heteroaralkyl, heteroaryl, cycloalkyl, (cycloalkyl)alkyl, hydroxyalkyl, alkoxyalkyl or acyl; or any two R a groups attached to the same carbon atom together represent an oxo (=0) or thioxo (=S);

each of R4 and R5 independently is hydrogen or absent; and

R6 is hydrogen or alkyl.

In certain embodiments, the compound has a structure of formula (I),

wherein,

X is O or S;

each dotted line [— ] independently represents an optional bond;

Rj is hydrogen or -CO-Aaa;

Aaa represents an amino acid residue wherein the amino acid residue comprises a side chain that includes a -OH, -O-acyl, -SH, -N¾ or -NH(alkyl) moiety;

R2 is (Ci-Ce)alkyl, (C2-C6)alkenyl or (C2-Ce)alkynyl substituted by one or more substituents selected from carboxylate, carboxylic acid, carboxylic acid ester, thiocarboxylate, thio acid, amido, amino or heterocyclyl, wherein two or three carbon atoms of the alkyl, alkenyl or alkynyl optionally form part of a 3-7-membered carbocyclic or heterocyclic ring; R3 is aryl, heteroaryl, N-linked heterocycloalkyl or 4-5 membered C-linked heterocycloalkyl; wherein the said aryl, heteroaryl and heterocycloalkyl is optionally substituted by 1 to 4 occurrences of R a ; and

R a , at each occurrence, is alkyl, alkoxy, halo, hydroxyl, amino, -C(0)OH, aralkyl, aryl, alkoxy, heteroaralkyl, heteroaryl, cycloalkyl, (cycloalkyl)alkyl, hydroxyalkyl, alkoxyalkyl or acyl; or two R a groups together represent an oxo (=0) or thioxo (=S); each of R4 and R5 independently is hydrogen or absent;

or a pharmaceutically acceptable salt thereof or a pharmaceutically acceptable stereoisomer thereof.

In certain preferred embodiments of formula (I), X is O. In certain such embodiments, the ring containing X is 1,2,4-oxadiazole ring.

In certain preferred embodiments,— represents a bond;

In certain embodiments of formula (I), Rj is hydrogen or -CO-Aaa.

In certain embodiments, Aaa represents an amino acid residue wherein the amino acid residue comprises a side chain that includes a -OH, -O-acyl, -SH, -S(alkyl), -N¾ or -NH(alkyl) moiety.

In certain embodiments of formula (I), Rj is hydrogen.

In certain embodiments of the compound of formula (I), Rj is -CO-Aaa, wherein Aaa is same as defined in compound of formula (I).

In certain embodiments of the compound of formula (I), Aaa is a natural amino acid residue.

In alternative embodiments of the compound of formula (I), Aaa represents an amino acid residue, wherein the amino acid residue comprises a side chain that does not include a -OH, -O-acyl, -SH, -NH 2 or -NH(alkyl) moiety.

In certain embodiments of the compound of formula (I), Aaa is Ser, Thr, Gly, Lys, Met, Phe or Ala.

In certain embodiments of the compound of formula (I), Aaa is Ser or Thr.

In certain embodiments of the compound of formula (I), Aaa is Thr.

In certain embodiments of the compound of formula (I), Aaa is Met. In certain embodiments of the compound of formula (I), Aaa is Ser.

In certain embodiments of the compound of formula (I), Aaa is Lys.

In certain embodiments of the compound of formula (I), R 2 is (Ci-C6)alkyl, (C 2 - Ce)alkenyl or (C 2 -Ce)alkynyl substituted by one or more substituents selected from carboxylate, carboxylic acid, carboxylic acid ester, thiocarboxylate, thio acid, amido, amino or heterocyclyl, wherein two or three carbon atoms of the alkyl, alkenyl or alkynyl optionally form part of a 3-7-membered carbocyclic or heterocyclic ring.

In alternative embodiments of the compound of formula (I), R 2 is hydrogen or (C7-Cio)alkyl, (C7-Cio)alkenyl, (C7-Cio)alkynyl, cycloalkyl, heterocyclyl, heterocyclylalkyl, heteroaralkyl, aralkyl, heteroaryl or aryl, each optionally substituted by one or more substituents selected from carboxylate, carboxylic acid, carboxylic acid ester, thiocarboxylate, thio acid, amido, amino, heterocyclyl, hydroxyl, cycloalkyl, aryl, aryl-COOH, heteroaryl, guanidino, amidino, -NH, -N(alkyl), -SH and -S(alkyl), optionally wherein two or three carbon atoms of the alkyl, alkenyl or alkynyl form part of a 3-7-membered carbocyclic or heterocyclic ring which is optionally substituted with 1 to 4 substituents, each independently selected from alkyl, alkoxy, carboxylic acid, carboxylate and hydroxyl.

In further alternative embodiments of the compound of formula (I), R 2 is (Ci- Ce)alkyl, (C 2 -Ce)alkenyl or (C 2 -Ce)alkynyl, each substituted by one or more substituents selected from hydroxyl, cycloalkyl, aryl, aryl-COOH, heteroaryl, guanidino, amidino, - NH, -N(alkyl), -SH and -S(alkyl).

In yet further alternative embodiments of the compound of formula (I), R 2 is (Ci- Ce)alkyl, (C 2 -Ce)alkenyl or (C 2 -Ce)alkynyl, wherein two or three carbon atoms of the alkyl, alkenyl or alkynyl form part of a 3-7-membered carbocyclic or heterocyclic ring substituted with 1 to 4 substituents, each independently selected from alkyl, alkoxy, carboxylic acid, carboxylate and hydroxyl.

In certain embodiments, R 2 is (Ci-Ce)alkyl, cycloalkyl, (C 2 -Ce)alkynyl or heteroarylalkyl; wherein (Ci-Ce)alkyl or heteroarylalkyl is substituted with one or more substituents selected from carboxylic acid, carboxylate, thiocarboxylate, thioacid, amido, amino, hydroxyl, cycloalkyl, aryl, aryl-COOH, heterocyclyl, heteroaryl, guanidino, amidino, -NH, -N(alkyl), -SH and -S(alkyl); optionally wherein two or three carbon atoms of the alkyl, alkenyl or alkynyl form part of a 3-7-membered carbocyclic or heterocyclic ring which are optionally substituted with 1 to 4 same or different substituents independently selected from alkyl, alkoxy, carboxylic acid, carboxylate and hydroxyl;

In certain embodiments of the compound of formula (I), R 3 is aryl, heteroaryl, NT- linked heterocycloalkyl or 4-5 membered C-linked heterocycloalkyl; wherein the aryl, heteroaryl and heterocycloalkyl is optionally substituted by 1 to 4 occurrences of R a ; and

R a , at each occurrence, is alkyl, alkoxy, halo, hydroxyl, amino, -C(0)OH, aralkyl, aryl, alkoxy, heteroaralkyl, heteroaryl, cycloalkyl, (cycloalkyl)alkyl, hydroxyalkyl, alkoxy alkyl or acyl;

or any two R a groups attached to the same carbon together represent an oxo (=0) or thioxo (=S).

In alternative embodiments of the compound of formula (I), R 3 is aryl, cycloalkyl or heterocyclyl, optionally substituted by 1 to 4 occurrences of R a , wherein the heterocyclyl is not linked at N atom and is not a 4-5 membered C-linked heterocycloalkyl.

In certain embodiments of the compound of formula (I), R6 is hydrogen.

Alternatively, R6 may be alkyl.

In certain embodiments, the present invention provides compounds of formula

(II):

wherein,

Y and Z are each independently -CR a R b -, -NR C -, O or S;

X is O or S;

each dotted line [— ] independently represents an optional bond;

R a and R are each independently hydrogen or a substituent such as alkyl, acyl, hydroxyl, amino, halo, aralkyl, aryl, heteroaralkyl, heteroaryl, cycloalkyl, aminoalkyl, alkoxy hydroxyalkyl, alkoxyalkyl or (cycloalkyl)alkyl; preferably hydroxyl, amino, lower alkyl, lower acyl or lower aralkyl; R c is hydrogen or a substituent, such as alkyl, acyl, aralkyl, aryl, heteroaralkyl, heteroaryl, cycloalkyl or (cycloalkyl)alkyl; preferably lower alkyl, lower acyl or lower aralkyl;

Rj is hydrogen or -CO-Aaa;

Aaa represents an amino acid residue;

R2 is side chain of an amino acid, hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, heterocyclylalkyl, heteroaralkyl, aralkyl, heteroaryl or aryl, each optionally substituted by one or more substituents selected from carboxylate, carboxylic acid, carboxylic acid ester, thiocarboxylate, thio acid, amido, amino, heterocyclyl, hydroxyl, cycloalkyl, aryl, aryl-COOH, heteroaryl, guanidino, amidino, -NH, -N(alkyl), -SH and - S(alkyl), optionally wherein two or three carbon atoms of the alkyl, alkenyl or alkynyl form part of a 3-7-membered carbocyclic or heterocyclic ring (such as a cyclobutyl or oxirane ring) optionally substituted with 1 to 4 substituents, each independently selected from alkyl, alkoxy, carboxylic acid, carboxylate and hydroxyl;

each of R4 and R5 independently is hydrogen or absent; and

R 7 represents 0-4 substituents on the ring to which it is attached, wherein each substituent is independently selected from alkyl, aralkyl, aryl, alkoxy, heteroaralkyl, heteroaryl, halo, cycloalkyl, (cycloalkyl)alkyl, amino, -C(0)OH, hydroxyl, hydroxyalkyl, alkoxyalkyl or acyl; preferably lower alkyl, lower acyl or lower aralkyl;

two R 7 groups attached to the same carbon atom together represent an oxo (=0) or thioxo (=S);

or a pharmaceutically acceptable salt thereof or a stereoisomer thereof.

In certain embodiments, the invention provides a compound of formula (II),

wherein,

Y and Z are each independently -CR a R b -, -NR C -, O or S;

X is O or S; each dotted line [— ] independently represents an optional bond;

R a and R b are each independently hydrogen or a substituent such as alkyl, acyl, hydroxyl, amino, halo, aralkyl, aryl, heteroaralkyl, heteroaryl, cycloalkyl, aminoalkyl, alkoxy hydroxyalkyl, alkoxyalkyl or (cycloalkyl)alkyl; preferably hydroxyl, amino, lower alkyl, lower acyl or lower aralkyl;

R c is hydrogen or a substituent, such as alkyl, acyl, aralkyl, aryl, heteroaralkyl, heteroaryl, cycloalkyl or (cycloalkyl)alkyl; preferably lower alkyl, lower acyl or lower aralkyl;

Rj is hydrogen or -CO-Aaa;

Aaa represents an amino acid residue wherein the amino acid residue comprises a side chain that includes a -OH, -O-acyl, -SH, N¾, NH(alkyl) or -S(alkyl) moiety;

R2 is alkyl, alkenyl or alkynyl substituted by one or more substituents selected from carboxylate, carboxylic acid, carboxylic acid ester, thiocarboxylate, thio acid, amido, amino or heterocyclyl, optionally wherein two or three carbon atoms of the alkyl, alkenyl or alkynyl form part of a 3-7-membered carbocyclic or heterocyclic ring (such as a cyclobutyl or oxirane ring);

each of R4 and R5 independently is hydrogen or absent; and

R 7 represents 0-4 substituents on the ring to which it is attached, wherein each substituent is independently selected from alkyl, aralkyl, aryl, alkoxy, heteroaralkyl, heteroaryl, halo, cycloalkyl, (cycloalkyl)alkyl, amino, hydroxyl, hydroxyalkyl, alkoxyalkyl or acyl; preferably lower alkyl, lower acyl or lower aralkyl;

or a pharmaceutically acceptable salt thereof.

In certain preferred embodiments of formula (II), X is O. In certain such embodiments, the ring containing X is a 1,2,4-oxadiazole ring.

In certain embodiments of the compound of formula (II), Rj is hydrogen or -CO-

Aaa. In certain such embodiments, [Aaa] represents an amino acid residue wherein the amino acid residue comprises a side chain that includes a -OH, -O-acyl, -SH, -N¾, - NH(alkyl) or -S(alkyl) moiety;.

In certain embodiments of the compound of formula (II), Rj is hydrogen. In certain embodiments of the compound of formula (II), Rj is -CO-Aaa, wherein Aaa is same as defined in compound of formula (II).

In certain embodiments of the compound of formula (II), Aaa is a natural amino acid residue.

In certain embodiments of the compound of formula (II), Aaa represents an amino acid residue wherein the amino acid residue comprises a side chain that includes a -OH, -O-acyl, -SH, -NH 2 , -NH(alkyl) or -S(alkyl) moiety.

In alternative embodiments of the compound of formula (II), Aaa represents an amino acid residue, wherein the amino acid residue comprises a side chain that does not include a -OH, -O-acyl, -SH, -NH 2 , -NH(alkyl) or -S(alkyl) moiety.

In certain embodiments of the compound of formula (II), Aaa is Ser, Thr, Gly, Lys, Met, Phe or Ala.

In certain embodiments of the compound of formula (II), Aaa is Ser or Thr.

In certain embodiments of the compound of formula (II), Aaa is Thr.

In certain embodiments of the compound of formula (II), Aaa is Met.

In certain embodiments of the compound of formula (II), Aaa is Ser.

In certain embodiments of the compound of formula (II), Aaa is Lys.

In certain embodiments of formula (II), at least one of Y and Z is -NR C -, O or S. Preferably, at least one of Y and Z is -NR C -, for example -NH-.

In certain embodiments of the compound of formula (II), R 2 represents (Ci-

Ce)alkyl, (C2-C6)alkenyl or (C2-C6)alkynyl substituted by one or more substituents selected from carboxylate, carboxylic acid, carboxylic acid ester, thiocarboxylate, thio acid, amido, amino and heterocyclyl, preferably from lower alkyl, lower acyl or lower aralkyl, optionally wherein two or three carbon atoms of the (Ci-Ce)alkyl, (C2-Ce)alkenyl or (C2-Ce)alkynyl form part of a 3-7-membered carbocyclic or heterocyclic ring (such as a cyclobutyl or oxirane ring).

Alternatively, in certain embodiments of the compound of formula (II), R2 is hydrogen or cycloalkyl, heterocyclyl, heterocyclylalkyl heteroaralkyl, aralkyl, heteroaryl or aryl, each optionally substituted by one or more substituents selected from carboxylate, carboxylic acid, carboxylic acid ester, thiocarboxylate, thio acid, amido, amino, heterocyclyl, hydroxyl, cycloalkyl, aryl, aryl-COOH, heteroaryl, guanidino, amidino, -NH, -N(alkyl), -SH and -S(alkyl), optionally wherein two or three carbon atoms of the alkyl, alkenyl or alkynyl form part of a 3-7-membered carbocyclic or heterocyclic ring (such as a cyclobutyl or oxirane ring) optionally substituted with 1 to 4 substituents, each independently selected from alkyl, alkoxy, carboxylic acid, carboxylate and hydroxyl.

In further alternative embodiments of the compound of formula (II), I¾ is alkyl, alkenyl or alkynyl, substituted by one or more substituents selected from hydroxyl, cycloalkyl, aryl, aryl-COOH, heteroaryl, guanidino, amidino, -SH and -S(alkyl).

In further alternative embodiments of the compound of formula (II), I¾ is alkyl, alkenyl or alkynyl, wherein two or three carbon atoms of the alkyl, alkenyl or alkynyl form part of a 3-7-membered carbocyclic or heterocyclic ring (such as a cyclobutyl or oxirane ring) substituted with 1 to 4 substituents, each independently selected from alkyl, alkoxy, carboxylic acid, carboxylate and hydroxyl.

In certain embodiments of the compound of formula (II), R 7 represents 0 substituents on the ring to which it is attached.

In certain embodiments of the compound of formula (II), R7 represents 0-4 substituents on the ring to which it is attached, wherein each substituent is independently selected from alkyl, aralkyl, aryl, alkoxy, heteroaralkyl, heteroaryl, halo, cycloalkyl, (cycloalkyl)alkyl, amino, hydroxyl, hydroxyalkyl, alkoxyalkyl or acyl; preferably lower alkyl, lower acyl or lower aralkyl.

In alternative embodiments of the compound of formula (II), at least one occurrence of R 7 represents -C(0)OH.

In further alternative embodiments of the compound of formula (II), two R 7 groups attached to the same carbon atom together represent an oxo (=0) or thioxo (=S).

In certain embodiments, the present invention provides compounds of formula

(IA):

or a pharmaceutically acceptable salt thereof or a stereoisomer thereof; wherein, Rj, R2, R 3 and R 6 are as defined in formula (I).

In certain embodiments, the present invention provides compounds of formula

(IB)

or a pharmaceutically acceptable salt thereof or a stereoisomer thereof;

wherein,

R2 , R 3 and Aaa are same as defined in formula (I).

In certain embodiments of formula (IB), Aaa is a natural amino acid residue.

In certain embodiments, Aaa is Ser, Thr, Gly, Lys, Phe, Met or Ala.

In certain embodiments, Aaa is Ser or Thr.

In certain embodiments, Aaa is Thr.

In certain embodiments, R 3 is aryl, cycloalkyl, heterocyclyl or heteroaryl optionally substituted by 1 to 4 occurrences of R a . In certain embodiments, R a is alkyl, alkoxy, halo, hydroxyl, amino, -C(0)OH, aralkyl, aryl, alkoxy, heteroaralkyl, heteroaryl, cycloalkyl, (cycloalkyl)alkyl, hydroxyalkyl, alkoxyalkyl or acyl;

In certain embodiments, R a is alkyl, halo, hydroxyl, amino, -C(0)OH, aralkyl, aryl, alkoxy, heteroaralkyl, heteroaryl, cycloalkyl, (cycloalkyl)alkyl, hydroxyalkyl, alkoxyalkyl or acyl.

In certain embodiments, any two R a groups attached to the same carbon together represent an oxo (=0) or thioxo (=S).

In certain embodiments, R2 is (Ci-C6)alkyl or (C2-C6)alkynyl substituted by one or more substituents selected from carboxylic acid, carboxylate, thiocarboxylate, thioacid, amido, amino and amidino.

In certain embodiments, R2 is (Ci-C6)alkyl substituted by carboxylic acid, amido or amidino.

In certain embodiments, R 2 is (Ci-C 6 )alkyl substituted by -C(0)OH, -C(0)NH 2 or -C(=NH)NH 2 .

In certain embodiments, R 2 is -(CH 2 )COOH, -(CH 2 ) 2 COOH, -(CH 2 )CONH 2 , - (CH 2 ) 2 CONH 2 or -(CH 2 )C(=NH)NH 2 .

In certain embodiments, R 2 is -(CH 2 )COOH or -(CH 2 )CONH 2 .

In certain embodiments, R2 IS (C2-C6)alkynyl; Particularly R2 is 1-propynyl.

In certain embodiments, R 6 is hydrogen. An amino acid residue is understood in the art to mean a carboxylic acid, substituted at the alpha, beta or gamma carbon by an amino (-NH 2 ) group. In the group - CO-Aaa, the amino acid residue Aaa is connected to the carbonyl group CO via a covalent bond between the carbonyl carbon and the amino group of the amino acid residue. In preferred embodiments, the amino acid is an alpha-amino acid and the amino acid residue Aaa is connected to the carbonyl group CO via a covalent bond between the carbonyl carbon and the alpha-amino group of the amino acid residue.

In certain embodiments, R and R c may be combined together with the atoms to which they attached to form a 5-7 membered ring optionally substituted with one or more groups independently selected from hydroxyl, halo, amino, cyano and alkyl;

In accordance with any of the foregoing embodiments, in certain embodiments, one, more or all amino acid residues are D amino acid residues.

In certain embodiments, one, more than one or all amino acid residues are L amino acid residues.

In certain embodiments, the present invention provides a compound or a pharmaceutically acceptable salt thereof or a stereoisomer thereof, selected from:

In certain embodiments, compounds of the invention may be prodrugs of the compounds of formula (I), e.g., wherein a hydroxyl in the parent compound is presented as an ester or a carbonate or carboxylic acid present in the parent compound is presented as an ester. In a further embodiment, the prodrug is metabolized to the active parent compound in vivo (e.g., the ester is hydrolyzed to the corresponding hydroxyl or carboxylic acid).

In certain embodiments, the compounds of the present invention can also contain unnatural proportions of atomic isotopes at one or more of the atoms that constitute such compounds. For example, the present invention also embraces isotopically-labeled variants of the present invention which are identical to those recited herein, but for the fact that one or more atoms of the compound are replaced by an atom having the atomic mass or mass number different from the predominant atomic mass or mass number usually found in nature for the atom. All isotopes of any particular atom or element as specified are contemplated within the scope of the compounds of the invention and their uses. Exemplary isotopes that can be incorporated in to compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulfur, fluorine, chlorine and iodine, such as 2 H ("D"), ¾ n C, 13 C, 14 C, 13 N, 15 N, 15 0, 17 0, 18 0, 35 S, 18 F,

36 Cl, 123 I and 125 I. Isotopically labeled compounds of the present inventions can generally be prepared by following procedures analogous to those disclosed in the schemes and/or in the examples herein below, by substituting an isotopically labeled reagent for a non- isotopically labeled reagent.

Pharmaceutical Compositions

In certain embodiments, the present invention provides a pharmaceutical composition comprising a compound as disclosed herein, optionally admixed with a pharmaceutically acceptable carrier or diluent.

The present invention also provides methods for formulating the disclosed compounds for pharmaceutical administration.

The compositions and methods of the present invention may be utilized to treat an individual in need thereof. In certain embodiments, the individual is a mammal such as a human or a non-human mammal. When administered to an animal, such as a human, the composition or the compound is preferably administered as a pharmaceutical composition comprising, for example, a compound of the invention and a pharmaceutically acceptable carrier. Pharmaceutically acceptable carriers are well known in the art and include, for example, aqueous solutions such as water or physiologically buffered saline or other solvents or vehicles such as glycols, glycerol, oils such as olive oil or injectable organic esters. In a preferred embodiment, when such pharmaceutical compositions are for human administration, particularly for invasive routes of administration (i.e., routes, such as injection or implantation, that circumvent transport or diffusion through an epithelial barrier), the aqueous solution is pyrogen-free or substantially pyrogen-free. The excipients can be chosen, for example, to effect delayed release of an agent or to selectively target one or more cells, tissues or organs. The pharmaceutical composition can be in dosage unit form such as tablet, capsule (including sprinkle capsule and gelatin capsule), granule, lyophile for reconstitution, powder, solution, syrup, suppository, injection or the like. The composition can also be present in a transdermal delivery system, e.g., a skin patch. The composition can also be present in a solution suitable for topical administration, such as an eye drop.

A pharmaceutically acceptable carrier can contain physiologically acceptable agents that act, for example, to stabilize, increase solubility or to increase the absorption of a compound such as a compound of the invention. Such physiologically acceptable agents include, for example, carbohydrates, such as glucose, sucrose or dextrans, antioxidants, such as ascorbic acid or glutathione, chelating agents, low molecular weight proteins or other stabilizers or excipients. The choice of a pharmaceutically acceptable carrier, including a physiologically acceptable agent, depends, for example, on the route of administration of the composition. The preparation of pharmaceutical composition can be a self-emulsifying drug delivery system or a self-microemulsifying drug delivery system. The pharmaceutical composition (preparation) also can be a liposome or other polymer matrix, which can have incorporated therein, for example, a compound of the invention. Liposomes, for example, which comprise phospholipids or other lipids, are nontoxic, physiologically acceptable and metabolizable carriers that are relatively simple to make and administer.

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

The phrase "pharmaceutically acceptable carrier" as used herein means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation and not injurious to the patient. Some examples of materials which can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and (21) other non-toxic compatible substances employed in pharmaceutical formulations.

A pharmaceutical composition (preparation) can be administered to a subject by any of a number of routes of administration including, for example orally (for example, drenches as in aqueous or non-aqueous solutions or suspensions, tablets, capsules (including sprinkle capsules and gelatin capsules), boluses, powders, granules, pastes for application to the tongue); absorption through the oral mucosa (e.g., sublingually); anally, rectally or vaginally (for example, as a pessary, cream or foam); parenterally (including intramuscularly, intravenously, subcutaneously or intrathecally as, for example, a sterile solution or suspension); nasally; intraperitoneally; subcutaneously; transdermally (for example as a patch applied to the skin); and topically (for example, as a cream, ointment or spray applied to the skin or as an eye drop). The compound may also be formulated for inhalation. In certain embodiments, a compound may be simply dissolved or suspended in sterile water. Details of appropriate routes of administration and compositions suitable for same can be found in, for example, U.S. Pat. Nos. 6,110,973, 5,763,493, 5,731,000, 5,541,231, 5,427,798, 5,358,970 and 4,172,896, as well as in patents cited therein.

The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration. The amount of active ingredient that can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect. Generally, out of one hundred percent, this amount will range from about 1 percent to about ninety-nine percent of active ingredient, preferably from about 5 percent to about 70 percent, most preferably from about 10 percent to about 30 percent.

Methods of preparing these formulations or compositions include the step of bringing into association an active compound, such as a compound of the invention, with the carrier and, optionally, one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association a compound of the present invention with liquid carriers or finely divided solid carriers or both and then, if necessary, shaping the product.

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

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

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

The tablets and other solid dosage forms of the pharmaceutical compositions, such as dragees, capsules (including sprinkle capsules and gelatin capsules), pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be sterilized by, for example, filtration through a bacteria- retaining filter or by incorporating sterilizing agents in the form of sterile solid compositions that can be dissolved in sterile water or some other sterile injectable medium immediately before use. These compositions may also optionally contain opacifying agents and may be of a composition that they release the active ingredient(s) only or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. The active ingredient can also be in microencapsulated form, if appropriate, with one or more of the above-described excipients. Liquid dosage forms useful for oral administration include pharmaceutically acceptable emulsions, lyophiles for reconstitution, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as, for example, water or other solvents, cyclodextrins and derivatives thereof, solubilizing agents and emulsifiers, such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan and mixtures thereof.

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

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

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

Formulations of the pharmaceutical compositions for administration to the mouth may be presented as a mouthwash or an oral spray or an oral ointment.

Alternatively or additionally, compositions can be formulated for delivery via a catheter, stent, wire or other intraluminal device. Delivery via such devices may be especially useful for delivery to the bladder, urethra, ureter, rectum or intestine.

Formulations which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate. Dosage forms for the topical or transdermal administration include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches and inhalants. The active compound may be mixed under sterile conditions with a pharmaceutically acceptable carrier and with any preservatives, buffers or propellants that may be required.

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

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

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

Ophthalmic formulations, eye ointments, powders, solutions and the like, are also contemplated as being within the scope of this invention. Exemplary ophthalmic formulations are described in U.S. Publication Nos. 2005/0080056, 2005/0059744, 2005/0031697 and 2005/004074 and U.S. Pat. No. 6,583,124, the contents of which are incorporated herein by reference. If desired, liquid ophthalmic formulations have properties similar to that of lacrimal fluids, aqueous humor or vitreous humor or are compatable with such fluids. A preferred route of administration is local administration (e.g., topical administration, such as eye drops or administration via an implant).

The phrases "parenteral administration" and "administered parenterally" as used herein means modes of administration other than enteral and topical administration, usually by injection and includes, without limitation, intravenous, intramuscular, intraarterial, intrathecal, intracapsular, intraorbital, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal and intrasternal injection and infusion.

Pharmaceutical compositions suitable for parenteral administration comprise one or more active compounds in combination with one or more pharmaceutically acceptable sterile isotonic aqueous or nonaqueous solutions, dispersions, suspensions or emulsions or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, bacteriostats, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.

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

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

In some cases, in order to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution, which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered drug form is accomplished by dissolving or suspending the drug in an oil vehicle. Injectable depot forms are made by forming microencapsulated matrices of the subject compounds in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions that are compatible with body tissue.

For use in the methods of this invention, active compounds can be given per se or as a pharmaceutical composition containing, for example, 0.1 to 99.5% (more preferably, 0.5 to 90%) of active ingredient in combination with a pharmaceutically acceptable carrier.

Methods of introduction may also be provided by rechargeable or biodegradable devices. Various slow release polymeric devices have been developed and tested in vivo in recent years for the controlled delivery of drugs, including proteinaceous biopharmaceuticals. A variety of biocompatible polymers (including hydrogels), including both biodegradable and non-degradable polymers, can be used to form an implant for the sustained release of a compound at a particular target site.

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

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

A physician or veterinarian having ordinary skill in the art can readily determine and prescribe the therapeutically effective amount of the pharmaceutical composition required. For example, the physician or veterinarian could start doses of the pharmaceutical composition or compound at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved. By "therapeutically effective amount" is meant the concentration of a compound that is sufficient to elicit the desired therapeutic effect. It is generally understood that the effective amount of the compound will vary according to the weight, sex, age and medical history of the subject. Other factors which influence the effective amount may include, but are not limited to, the severity of the patient's condition, the disorder being treated, the stability of the compound and, if desired, another type of therapeutic agent being administered with the compound of the invention. A larger total dose can be delivered by multiple administrations of the agent. Methods to determine efficacy and dosage are known to those skilled in the art (Isselbacher et al. (1996) Harrison's Principles of Internal Medicine 13 ed., 1814-1882, herein incorporated by reference).

In general, a suitable daily dose of an active compound used in the compositions and methods of the invention will be that amount of the compound that is the lowest dose effective to produce a therapeutic effect. Such an effective dose will generally depend upon the factors described above.

If desired, the effective daily dose of the active compound may be administered as one, two, three, four, five, six or more sub-doses administered separately at appropriate intervals throughout the day, optionally, in unit dosage forms. In certain embodiments of the present invention, the active compound may be administered two or three times daily. In preferred embodiments, the active compound will be administered once daily.

The patient receiving this treatment is any animal in need, including primates, in particular humans and other mammals such as equines, cattle, swine and sheep; and poultry and pets in general.

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

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

Methods of Treatment

The programmed cell death protein 1 pathway (PD-1) pathway has been implicated in a number of diseases and conditions and the pathway is known to regulate various immune responses. Numerous studies have sought to activate immune response by targeting the PD-1 pathway, thereby providing a therapy for certain conditions, such as cancers. In fact, studies indicate that blockade of the PD-1 pathway, for example by inhibiting an immunosuppressive signal induced by PD-1, PD-LI or PD-L2, leads to antitumor activity in various cancers, including lung, breast, colon, renal, bladder, thyroid, prostate, osteosarcoma and Hodgkin's lymphoma.

Furthermore, PD-1 activity has also been associated with autoimmune conditions, such as lupus erythematosus, juvenile idiopathic arthritis and allergic encephalomyelitis.

In certain embodiments, the present invention provides uses of a compound of the present invention for the preparation of a medicament, e.g., for the treatment of cancer.

In certain embodiments, the present invention provides methods for treating cancer, wherein the method comprises administration of a therapeutically effective amount of a compound of the present invention to the subject in need thereof.

In certain embodiments, the present invention provides methods for inhibiting growth of tumour cells and/or metastasis by administering a therapeutically effective amount of compounds of the present invention to the subject in need thereof.

In certain embodiments, the present invention provides methods for inhibiting growth of tumour cells and/or metastasis by administering a therapeutically effective amount of compound of formula (I) or formula (II) to the subject in need thereof.

In certain embodiments, the present invention provides methods for treating cancer, by administering a therapeutically effective amount of compound of formula (I) or formula (II) to the subject in need thereof.

Representative tumour cells include cells of a cancer such as but are not limited to melanoma, renal cancer, prostate cancer, breast cancer, colon cancer and lung cancer, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular malignant melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, testicular cancer, carcinoma of the fallopian tubes, carcinoma of the endometrium, carcinoma of the cervix, carcinoma of the vagina, carcinoma of the vulva, Hodgkin's Disease, non-Hodgkin's lymphoma, cancer of the esophagus, cancer of the small intestine, cancer of the endocrine system, cancer of the thyroid gland, cancer of the parathyroid gland, cancer of the adrenal gland, sarcoma of soft tissue, cancer of the urethra, cancer of the penis, chronic or acute leukemias including acute myeloid leukemia, chronic myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, solid tumours of childhood, lymphocytic lymphoma, cancer of the bladder, cancer of the kidney or ureter, carcinoma of the renal pelvis, neoplasm of the central nervous system (CNS), non-small cell lung cancer (NSCLC), primary CNS lymphoma, tumour angiogenesis, spinal axis tumour, brain stem glioma, pituitary adenoma, Kaposi's sarcoma, epidermoid cancer, squamous cell cancer, T-cell lymphoma, environmentally induced cancers including those induced by asbestos and combinations of said cancers.

In certain embodiments, the present invention provides methods for treating cancer, wherein the cancer is selected from lung cancer, breast cancer, colon cancer, renal cancer, bladder cancer, thyroid cancer, prostate cancer, osteosarcoma and Hodgkin's lymphoma.

In certain embodiments, the present invention provides methods for treating bacterial, viral or fungal infection or an immunological condition, by administering a therapeutically effective amount of compound of formula (I) or formula (II) or a pharmaceutically acceptable salt thereof and a stereoisomer thereof to the subject in need thereof.

In certain embodiments, the present invention provides uses of a compound of the present invention for the preparation of a medicament for the treatment of bacterial, viral and fungal infection, as well as methods of administering a therapeutically effective amount of a compound of the present invention for the treatment of a bacterial, viral or fungal infection.

In certain embodiments, the present invention provides uses of a compound of formula (I) or formula (II) for the preparation of a medicament for the treatment of bacterial, viral and fungal infection, as well as methods of administering a therapeutically effective amount of compound of formula (I) or a pharmaceutically acceptable salt thereof and a stereoisomer thereof for the treatment of a bacterial, viral or fungal infection.

Still yet other embodiments of the present invention provides a method of treatment of infection by blockade of the PD-1 pathway, for example inhibiting an immunosuppressive signal induced by PD-1, PD-Ll or PD-L2, wherein the method comprises administration of a therapeutically effective amount of a compound of the present invention to the subject in need thereof.

In certain embodiments, the invention provides uses of a compound of the present invention in inhibiting the PD-1 pathway (e.g., PD-1, PD-Ll or PD-L2).

In certain embodiments, the present invention provides methods for treating infectious disease in a subject comprising administering a therapeutically effective amount of a compound of the present invention for the treatment of the infectious disease.

In certain embodiments, the present invention provides compound of formula (I) or formula (II) or a pharmaceutically acceptable salt thereof and a stereoisomer thereof for use as a medicament.

In certain embodiments, the present invention provides compound of formula (I) or formula (II) or a pharmaceutically acceptable salt thereof and a stereoisomer thereof for use in the treatment of cancer.

In certain embodiments, the present invention provides compound of formula (I) or formula (II) or a pharmaceutically acceptable salt thereof and a stereoisomer thereof for use in the treatment of lung cancer, breast cancer, colon cancer, renal cancer, bladder cancer, thyroid cancer, prostate cancer, osteosarcoma and Hodgkin's lymphoma.

In certain embodiments, the present invention provides compound of formula (I) or formula (II) or a pharmaceutically acceptable salt thereof and a stereoisomer thereof for use in the treatment of bacterial, viral or fungal infection or an immunological condition.

Representative infectious disease include but are not limited to HIV, Influenza,

Herpes, Giardia, Malaria, Leishmania, the pathogenic infection by the virus Hepatitis (A, B, & C), herpes virus (e.g., VZV, HSV-I, HAV-6, HSV-II and CMV, Epstein Ban- virus), adenovirus, influenza virus, flaviviruses, echovirus, rhinovirus, coxsackie virus, cornovirus, respiratory syncytial virus, mumps virus, rotavirus, measles virus, rubella virus, parvovirus, vaccinia virus, HTLV virus, dengue virus, papillomavirus, molluscum virus, poliovirus, rabies virus, JC virus and arboviral encephalitis virus, pathogenic infection by the bacteria chlamydia, rickettsial bacteria, mycobacteria, staphylococci, streptococci, pneumonococci, meningococci and conococci, klebsiella, proteus, serratia, pseudomonas, E. coli, legionella, diphtheria, salmonella, bacilli, cholera, tetanus, botulism, anthrax, plague, leptospirosis and Lyme's disease bacteria, pathogenic infection by the fungi Candida (albicans, krusei, glabrata, tropicalis, etc.), Cryptococcus neoformans, Aspergillus (fumigatus, niger, etc.), Genus Mucorales (mucor, absidia, rhizophus), Sporothrix schenkii, Blastomyces dermatitidis, Paracoccidioides brasiliensis, Coccidioides immitis and Histoplasma capsulatum and pathogenic infection by the parasites Entamoeba histolytica, Balantidium coli, Naegleriafowleri, Acanthamoeba sp., Giardia lambia, Cryptosporidium sp., Pneumocystis carinii, Plasmodium vivax, Babesia microti, Trypanosoma brucei, Trypanosoma cruzi, Leishmania donovani, Toxoplasma gondi, Nippostrongylus brasiliensis.

The compounds of the present invention may be used as single drugs (monotherapy) or conjointly with one or more other agents (conjoint therapy). The compounds may be used by themselves or, preferably, in a pharmaceutical composition in which the compound is mixed with one or more pharmaceutically acceptable materials.

The pharmaceutical composition may be administered by oral or inhalation routes or by parenteral administration route. For example, compositions can be administered orally, by intravenous infusion, topically, intraperitoneally, intravesically or intrathecally. Examples of parenteral administration includes but not limited to intraarticular (in the joints), intravenous, intramuscular, intradermal, intraperitoneal and subcutaneous routes. Suitable liquid compositions may be aqueous or non-aqueous, isotonic sterile injection solutions and may contain antioxidants, buffers, bacteriostats and solutes that render the formulation isotonic with the blood of the intended recipient and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers and preservatives. Oral administration, parenteral administration, subcutaneous administration and intravenous administration are preferred methods of administration.

The dosage of the compounds of the present invention varies depending on a patient's age, weight or symptoms, as well as the compound's potency or therapeutic efficacy, the dosing regimen and/or treatment time. Generally, suitable routes of administration may, for example, include oral, eyedrop, rectal, transmucosal, topical or intestinal administration; parenteral delivery, including intramuscular, subcutaneous, intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, intraperitoneal, intranasal or intraocular injections. The compounds of the invention may be administered in an amount of 0.5 mg or 1 mg up to 500 mg, 1 g or 2 g per dosage regimen. The dosage may be administered once per week, once per three days, once per two days, once per day, twice per day, three times per day or more often. In alternative embodiments, in certain adults the compound can be continuously administered by intravenous administration for a period of time designated by a physician. Since the dosage is affected by various conditions, an amount less than or greater than the dosage ranges contemplated about may be implemented in certain cases. A physician can readily determine the appropriate dosage for a patient undergoing therapeutic treatment.

The compounds of the present invention may be administered in combination with one or more other drugs (1) to complement and/or enhance effect of the compound of the present invention, (2) to modulate pharmacodynamics, improve absorption or reduce dosage of the compound of the present invention and/or (3) to reduce or ameliorate the side effects of the compound of the present invention. As used herein, the phrase "conjoint administration" refers to any form of administration of two or more different therapeutic compounds such that the second compound is administered while the previously administered therapeutic compound is still effective in the body (e.g., the two compounds are simultaneously effective in the patient, which may include synergistic effects of the two compounds). For example, the different therapeutic compounds can be administered either in the same formulation or in a separate formulation, either concomitantly or sequentially. In certain embodiments, the different therapeutic compounds can be administered within one hour, 12 hours, 24 hours, 36 hours, 48 hours, 72 hours or a week of one another. Thus, an individual who receives such treatment can benefit from a combined effect of different therapeutic compounds. The respective compounds may be administered by the same or different route and the same or different method.

The dosage of the other drug can be a dosage that has been clinically used or may be a reduced dosage that is effective when administered in combination with a compound of the present invention. The ratio of the compound of the present invention and the other drug can vary according to age and weight of a subject to be administered, administration method, administration time, disorder to be treated, symptom and combination thereof. For example, the other drug may be used in an amount of 0.01 to 100 parts by mass, based on 1 part by mass of the compound of the present invention.

Conjoint therapy can be employed to treat any diseases discussed herein. For example, in the methods of the invention directed to the treatment of cancer, the compound of the present invention can be used with an existing chemotherapeutic conjointly using a single pharmaceutical composition or a combination of different pharmaceutical compositions. Examples of the chemotherapeutic include an alkylation agent, nitrosourea agent, antimetabolite, anticancer antibiotics, vegetable-origin alkaloid, topoisomerase inhibitor, hormone drug, hormone antagonist, aromatase inhibitor, P- glycoprotein inhibitor, platinum complex derivative, other immunotherapeutic drugs and other anticancer drugs. Further, a compound of the invention can be administered conjointly with a cancer treatment adjunct, such as a leucopenia (neutropenia) treatment drug, thrombocytopenia treatment drug, antiemetic and cancer pain intervention drug, concomitantly or in a mixture form. Chemotherapeutic agents that may be conjointly administered with compounds of the invention include: aminoglutethimide, amsacrine, anastrozole, asparaginase, beg, bicalutamide, bleomycin, bortezomib, buserelin, busulfan, campothecin, capecitabine, carboplatin, carfilzomib, carmustine, chlorambucil, chloroquine, cisplatin, cladribine, clodronate, colchicine, cyclophosphamide, cyproterone, cytarabine, dacarbazine, dactinomycin, daunorubicin, demethoxyviridin, dexamethasone, dichloroacetate, dienestrol, diethylstilbestrol, docetaxel, doxorubicin, epirubicin, estradiol, estramustine, etoposide, everolimus, exemestane, filgrastim, fludarabine, fludrocortisone, fluorouracil, fluoxymesterone, flutamide, gemcitabine, genistein, goserelin, hydroxyurea, idarubicin, ifosfamide, imatinib, interferon, irinotecan, ironotecan, lenalidomide, letrozole, leucovorin, leuprolide, levamisole, lomustine, lonidamine, mechlorethamine, medroxyprogesterone, megestrol, melphalan, mercaptopurine, mesna, metformin, methotrexate, mitomycin, mitotane, mitoxantrone, nilutamide, nocodazole, octreotide, oxaliplatin, paclitaxel, pamidronate, pentostatin, perifosine, plicamycin, pomalidomide, porfimer, procarbazine, raltitrexed, rituximab, sorafenib, streptozocin, sunitinib, suramin, tamoxifen, temozolomide, temsirolimus, teniposide, testosterone, thalidomide, thioguanine, thiotepa, titanocene dichloride, topotecan, trastuzumab, tretinoin, vinblastine, vincristine, vindesine and vinorelbine.

In certain embodiments, a compound of the invention may be conjointly administered with non-chemical methods of cancer treatment. In a further embodiment, a compound of the invention may be conjointly administered with radiation therapy. In a further embodiment, a compound of the invention may be conjointly administered with surgery, with thermoablation, with focused ultrasound therapy, with cryotherapy or with any combination of these.

In certain embodiments, different compounds of the invention may be conjointly administered with one or more other compounds of the invention. Moreover, such combinations may be conjointly administered with other therapeutic agents, such as other agents suitable for the treatment of cancer, immunological or neurological diseases, such as the agents identified above. In certain embodiments, conjointly administering one or more additional chemotherapeutic agents with a compound of the invention provides a synergistic effect. In certain embodiments, conjointly administering one or more additional chemotherapeutics agents provides an additive effect.

The compound of the present invention can be used with one or more other immunomodulators and/or potentiating agents conjointly using a single pharmaceutical composition or a combination of different pharmaceutical compositions. Suitable immunomodulators include various cytokines, vaccines and adjuvants. Examples of cytokines, vaccines and adjuvants that stimulate immune responses include GM-CSF, M- CSF, G-CSF, interferon-a, β or γ, IL-1, IL-2, IL-3, IL-12, Poly(I:C) and C P G.

In certain embodiments, the potentiating agents includes cyclophosphamide and analogs of cyclophosphamide, anti-TGF and Imatinib (Gleevec), a mitosis inhibitor, such as paclitaxel, Sunitinib (Sutent) or other antiangiogenic agents, an aromatase inhibitor, such as letrozole, an A2a adenosine receptor (A2AR) antagonist, an angiogenesis inhibitor, anthracyclines, oxaliplatin, doxorubicin, TLR4 antagonists and IL-18 antagonists.

Definitions and Abbreviations: Unless defined otherwise, all technical and scientific terms used herein have the same meaning and the meaning of such terms is independent at each occurrence thereof and is as commonly understood by one of skill in art to which the subject matter herein belongs. That notwithstanding and except where stated otherwise, the following definitions apply throughout the specification and claims. Chemical names, common names and chemical structures may be used interchangeably to describe the same structure. If a chemical compound is referred to using both a chemical structure and a chemical name and an ambiguity exists between the structure and the name, the structure predominates. These definitions apply regardless of whether a term is used by itself or in combination with other terms, unless otherwise indicated. Hence, the definition of "alkyl" applies to "alkyl" as well as the "alkyl" portions of "hydroxyalkyl," "haloalkyl," "-O-alkyl," etc.

The term "compound(s) of the present invention", unless otherwise specifically stated, comprises compounds of formula (I) or formula (II) or formula (IA) or formula (IB) or a pharmaceutical acceptable salt thereof and stereoisomers thereof.

The term "acyl" is art-recognized and refers to a group represented by the general formula hydrocarbylC(O)— , preferably alkylC(O)— . Acyl groups include -C(0)CH 3 , - C(0)CH 2 CH 3 and the like.

The term "acylamino" refers to an amino group substituted with acyl. Acylamino groups include -N(H)C(0)CH 3 , -N(H)C(0)CH 2 CH 3 and the like.

The term "alkoxy" refers to an alkyl group, preferably a lower alkyl group, having oxygen attached thereto. Representative alkoxy groups include methoxy, ethoxy, propoxy, tert-butoxy and the like.

The term "alkoxyalkyl", as used herein, refers to an alkyl group substituted with an amino group. Alkoxyalkyl groups include -CH2OCH 3 , -CH 2 0CH 2 CH 3, _ CH2CH2OCH3 and the like.

The term "alkenyl", as used herein, refers to an aliphatic group containing at least one double bond and is intended to include both "unsubstituted alkenyls" and

"substituted alkenyls", the latter of which refers to alkenyl moieties having substituents replacing a hydrogen on one or more carbons of the alkenyl group. Such substituents may occur on one or more carbons that are included or not included in one or more double bonds. Moreover, such substituents include all those contemplated for alkyl groups, as discussed below, except where stability is prohibitive. For example, substitution of alkenyl groups by one or more alkyl, carbocyclyl, aryl, heterocyclyl or heteroaryl groups is contemplated.

An "alkyl" group or "alkane" is a straight chained or branched non-aromatic hydrocarbon which is completely saturated. Typically, a straight chained or branched alkyl group has from 1 to about 20 carbon atoms, preferably from 1 to about 10 unless otherwise defined. Examples of straight chained and branched alkyl groups include methyl, ethyl, n-propyl, iso-propyl, n-butyl, sec-butyl, tert-butyl, pentyl, hexyl, pentyl and octyl. A C \ -Ce straight chained or branched alkyl group is also referred to as a "lower alkyl" group. An alkyl group may be optionally substituted at one or more positions as permitted by valence. Such optional substituents include, for example, halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, alkoxyl, amino, nitro, sulfhydryl, imino, amido, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, sulfonamido, ketone, aldehyde, ester, heterocyclyl, aromatic or heteroaromatic moieties, -CF 3 , -CN or the like.

The term "alkylamino", as used herein, refers to an amino group substituted with at least one alkyl group.

The term "alkylthio", as used herein, refers to a thiol group substituted with an alkyl group and may be represented by the general formula alkylS— .

The term "alkynyl", as used herein, refers to an aliphatic group containing at least one triple bond and is intended to include both "unsubstituted alkynyls" and "substituted alkynyls", the latter of which refers to alkynyl moieties having substituents replacing a hydrogen on one or more carbons of the alkynyl group. Such substituents may occur on one or more carbons that are included or not included in one or more triple bonds. Moreover, such substituents include all those contemplated for alkyl groups, as discussed above, except where stability is prohibitive. For example, substitution of alkynyl groups by one or more alkyl, carbocyclyl, aryl, heterocyclyl or heteroaryl groups is contemplated.

The term "amide" or "amido" as used herein, refers to a group

wherein each Rn independently represent a hydrogen or hydrocarbyl group or two Rn are taken together with the N atom to which they are attached complete a heterocycle having from 4 to 8 atoms in the ring structure.

The term "amino" as used herein, refers to -NH 2 .

The term "aminoalkyl", as used herein, refers to an alkyl group substituted with an amino group. Aminoalkyl groups include -CH2NH2, -(CH2)2NH2, -(CH2) 3 NH2, - (CH 2 ) 4 NH 2 and the like.

The term "aminoaryl", as used herein, refers to an aryl group substituted with an amino group. Aminoaryl groups include aniline and the like.

The terms "aralkyl" and "arylalkyl", as used herein, refers to an alkyl group substituted with an aryl group. Arylalkyl groups include benzyl and the like.

The term "aryl" as used herein include substituted or unsubstituted single -ring aromatic groups in which each atom of the ring is carbon. Preferably the ring is a 5- to 7- membered ring, more preferably a 6-membered ring. The term "aryl" also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is aromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls and/or heterocyclyls. Aryl groups include benzene, naphthalene, phenanthrene, phenol, aniline and the like.

A "cycloalkyl" group is a cyclic hydrocarbon which is completely saturated. "Cycloalkyl" includes monocyclic and bicyclic rings. Typically, a monocyclic cycloalkyl group has from 3 to about 10 carbon atoms, more typically 3 to 8 carbon atoms unless otherwise defined. The second ring of a bicyclic cycloalkyl may be selected from saturated, unsaturated and aromatic rings. Cycloalkyl includes bicyclic molecules in which one, two or three or more atoms are shared between the two rings. The term "fused cycloalkyl" refers to a bicyclic cycloalkyl in which each of the rings shares two adjacent atoms with the other ring. The second ring of a fused bicyclic cycloalkyl may be selected from saturated, unsaturated and aromatic rings. A "cycloalkenyl" group is a cyclic hydrocarbon containing one or more double bonds. A cycloalkyl group may be substituted at one or more positions, as permitted by valence, with any optional substituents described herein. Cycloalkyl groups include but are not limited to cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.

The term "cyano" refers to -CN group.

The term "carboxy" or "carboxylic acid", as used herein, refers to a group represented by the formula — C(¾H. The term "carboxylate" refers to a group represented by the formula -(C0 2 ) ~ .

The term "amidino", as used herein, refers to -C(=NH)NH2 group.

The term "ester", as used herein, refers to a group -C(0)ORn wherein

Rii represents a hydrocarbyl group.

The term "guanidino", as used herein, refers to -NH-C(=NH)-NH2 group.

The terms "halo" and "halogen" as used herein means halogen and includes chloro, fluoro, bromo and iodo.

The term "haloalkyl", as used herein, refers to an alkyl group substituted with a halogen group.

As used herein, the term "carbocycle", "carbocyclic" or "carbocyclyl" is intended to mean any stable 3-, 4-, 5-, 6- or 7-membered monocyclic or bicyclic or 7-, 8-, 9-, 10-, 11-, 12- or 13-membered bicyclic or tricyclic hydrocarbon ring, any of which may be saturated, partially unsaturated, unsaturated or aromatic. Examples of carbocycles include, but are not limited to, cyclopropyl, cyclobutyl, cyclobutenyl, cyclopentyl, cyclopentenyl, cyclohexyl, cycloheptenyl, cycloheptyl, cycloheptenyl, adamantyl, cyclooctyl, cyclooctenyl, cyclooctadienyl, [3.3.0]bicyclooctane, [4.3.0]bicyclononane, [4.4.0]bicyclodecane, [2.2.2]bicyclooctane, fluorenyl, phenyl, naphthyl, indanyl, adamantyl, anthracenyl and tetrahydronaphthyl (tetralin). As shown above, bridged rings are also included in the definition of carbocycle (e.g., [2.2.2]bicyclooctane). Preferred carbocycles, unless otherwise specified, are cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, phenyl and indanyl. When the term "carbocycle" or "carbocyclyl" is used, it is intended to include "aryl". A bridged ring occurs when one or more carbon atoms link two non-adjacent carbon atoms. Preferred bridges are one or two carbon atoms. It is noted that a bridge always converts a monocyclic ring into a tricyclic ring. When a ring is bridged, the substituents recited for the ring may also be present on the bridge.

The terms "hetaralkyl", "heteroaralkyl" and "heteroarylalkyl" as used herein, refers to an alkyl group substituted with a hetaryl group.

The term "heteroalkyl", as used herein, refers to a saturated or unsaturated chain of carbon atoms and at least one heteroatom, wherein no two heteroatoms are adjacent.

The terms "heteroaryl" and "hetaryl" include substituted or unsubstituted aromatic single ring structures, preferably 5- to 7-membered rings, more preferably 5- to 6-membered rings, whose ring structures include at least one heteroatom, preferably one to four heteroatoms, more preferably one or two heteroatoms. The terms "heteroaryl" and "hetaryl" also include polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is heteroaromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls and/or heterocyclyls. Heteroaryl groups include, for example, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, pyrazole, pyridine, pyrazine, pyridazine, indole, 1 ,2,4-oxadiazole, 1,3,4-oxadiazole, 1,3,4-thiadiazole, benzimidazole, pyrimidine and the like. A heteroaryl group may be substituted at one or more positions, as permitted by valence, with any optional substituents described herein.

The term "heteroatom" as used herein means an atom of any element other than carbon or hydrogen. Preferred heteroatoms are nitrogen, oxygen and sulfur.

The terms "heterocyclyl", "heterocycle", "heterocyclic" or "heterocycloalkyl" refer to substituted or unsubstituted non-aromatic ring structures, preferably 3- to 10- membered rings, more preferably 3- to 7-membered rings, whose ring structures include at least one heteroatom, preferably one to four heteroatoms, more preferably one or two heteroatoms. The terms "heterocyclyl" and "heterocyclic" also include polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is heterocyclic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls and/or heterocyclyls. Heterocyclyl groups include, for example, piperidine, piperazine, pyrrolidine, morpholine, azepane, azetidine, 2,3-dihydrobenzo[b][l,4]dioxine, tetrahydro-2H-pyran, lactones, lactams and the like. Heterocyclyl groups may be optionally substituted as permitted by valence. The term "heterocyclylalkyl", as used herein, refers to an alkyl group substituted with a heterocycle group.

The term "hydroxyalkyl", as used herein, refers to an alkyl group substituted with a hydroxy group.

As used herein, the term "hydroxy" or "hydroxyl" refers to -OH group.

As used herein, the term "nitro" refers to -N(¾ group.

The term "lower" when used in conjunction with a chemical moiety, such as, acyl, acyloxy, alkyl, alkenyl, alkynyl or alkoxy is meant to include groups where there are ten or fewer non-hydrogen atoms in the substituent, preferably six or fewer. A "lower alkyl", for example, refers to an alkyl group that contains ten or fewer carbon atoms, preferably six or fewer. In certain embodiments, acyl, acyloxy, alkyl, alkenyl, alkynyl or alkoxy substituents defined herein are respectively lower acyl, lower acyloxy, lower alkyl, lower alkenyl, lower alkynyl or lower alkoxy, whether they appear alone or in combination with other substituents, such as in the recitations hydroxyalkyl and aralkyl (in which case, for example, the atoms within the aryl group are not counted when counting the carbon atoms in the alkyl substituent).

The term "substituted" refers to moieties having substituents replacing a hydrogen on one or more carbons of the backbone. It will be understood that "substitution" or "substituted with" includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. As used herein, the term "substituted" is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds. The permissible substituents can be one or more and the same or different for appropriate organic compounds. For purposes of this invention, the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. Substituents can include any substituents described herein, for example, a halogen, a hydroxyl, a carbonyl (such as a carboxyl, an alkoxycarbonyl, a formyl or an acyl), a thiocarbonyl (such as a thioester, a thioacetate or a thioformate), an alkoxyl, a phosphoryl, a phosphate, a phosphonate, a phosphinate, an amino, an amido, an amidine, an imine, a cyano, a nitro, an azido, a sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a sulfonamido, a sulfonyl, a heterocyclyl, an aralkyl or an aromatic or heteroaromatic moiety. It will be understood by those skilled in the art that substituents can themselves be substituted, if appropriate. Unless specifically stated as "unsubstituted," references to chemical moieties herein are understood to include substituted variants. For example, reference to an "aryl" group or moiety implicitly includes both substituted and unsubstituted variants.

The term "thioacid", "thiocarboxy" or "thiocarboxylic acid", as used herein, refers to a group represented by the formula -C(0)SH. The term "thiocarboxylate" refers to a group represented by the formula -(C(0)S) ~ .

As used herein, a therapeutic that "prevents" a disorder or condition refers to a compound that, in a statistical sample, reduces the occurrence of the disorder or condition in the treated sample relative to an untreated control sample or delays the onset or reduces the severity of one or more symptoms of the disorder or condition relative to the untreated control sample.

The term "treating" includes prophylactic and/or therapeutic treatments. The term "prophylactic or therapeutic" treatment is art-recognized and includes administration to the host of one or more of the subject compositions. If it is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the host animal) then the treatment is prophylactic (i.e., it protects the host against developing the unwanted condition), whereas if it is administered after manifestation of the unwanted condition, the treatment is therapeutic, (i.e., it is intended to diminish, ameliorate or stabilize the existing unwanted condition or side effects thereof).

The term "prodrug" is intended to encompass compounds which, under physiologic conditions, are converted into the therapeutically active agents of the present invention (e.g., a compound of formula (I)). A common method for making a prodrug is to include one or more selected moieties which are hydrolyzed under physiologic conditions to reveal the desired molecule. In other embodiments, the prodrug is converted by an enzymatic activity of the host animal. For example, esters or carbonates (e.g., esters or carbonates of alcohols or carboxylic acids) are preferred prodrugs of the present invention. In certain embodiments, some or all of the compounds of formula (I) in a formulation represented above can be replaced with the corresponding suitable prodrug, e.g., wherein a hydroxyl in the parent compound is presented as an ester or a carbonate or carboxylic acid present in the parent compound is presented as an ester.

As used herein, the term "comprise" or "comprising" is generally used in the sense of include, that is to say permitting the presence of one or more additional (unspecified) features or components.

As used herein, the term "including" as well as other forms, such as "include", "includes," and "included," is not limiting.

As used herein, the term "amino acid" means a molecule containing both an amino group and a carboxyl group and includes its salts, esters, combinations of its various salts, as well as tautomeric forms. In solution, at neutral pH, amino and acid groups of an amino acid can exchange a proton to form a doubly ionized, through overall neutral, entity identified as a zwitterion. In some embodiments, the amino acids are α-, β- , γ- or δ-amino acids, including their stereoisomers and racemates. As used herein, the term "L-amino acid" denotes an a-amino acid having the levorotatory configuration around the a-carbon, that is, a carboxylic acid of general formula CH(COOH)(NH 2 )- (side chain), having the L-configuration. The term "D-amino acid" similarly denotes a carboxylic acid of general formula CH(COOH)(NH2)-(side chain), having the dextrorotatory-configuration around the a-carbon. Side chains of L-amino acids can include naturally occurring and non-naturally occurring moieties. Non-naturally occurring (i.e., unnatural) amino acid side chains are moieties that are used in place of naturally occurring amino acid side chains in, for example, amino acid analogs.

An "amino acid residue" as used herein, means a moiety sharing structural similarity to the parent amino acid. An amino acid residue may be covalently bonded to another chemical moiety via the amino group of the residue or the carboxylate group of the residue (i.e., a hydrogen atom of -NH2 or -OH is replaced by a bond to another chemical moiety).

As used herein, the phrase "side chain of amino acid" means a moiety that is covalently attached to D or L-amino acid structure and can be represented as CH(COOH)(NH 2 )-R. For example, in case of alanine CH(COOH)(NH 2 )(CH 3 ), side chain of amino acid (R) is -CH 3 . Examples of "side chain of amino acid" include, but are not limited to, (Ci-Ce)alkyl, (C2-Ce)alkenyl or (C2-Ce)alkynyl. The side chain of amino acid may be substituted by one or more, same or different substituents selected from, but are not limited to, amino, amido, alkylamino, acylamino, carboxylic acid, carboxylate, thiocarboxylate, thioacid, hydroxyl, cycloalkyl, (cycloalkyl)alkyl, aryl, heterocyclyl, heteroaryl, guanidino, -SH, -NH(alkyl), -S(alkyl); optionally wherein cycloalkyl, aryl, heterocyclyl and heteroaryl are further substituted by one or more substituents such as hydroxy, alkoxy, halo, amino, nitro, cyano or alkyl.

Amino acids include the twenty standard amino acids used by most biological organisms in protein synthesis. Unnatural amino acid residues may be selected from, but are not limited to, alpha and alpha-disubstituted amino acids, N-alkyl amino acids and natural amino acids substituted with lower alkyl, aralkyl, hydroxyl, aryl, aryloxy, heteroarylalkyl or acyl.

For example, lysine can be substituted to form an unnatural amino acid, e.g., at a carbon atom of its side chain or alternatively by mono- or dialkylation of its terminal N¾ group (e.g., wherein the amino group of the lysine side chain is taken together with its substituents to form a heterocyclic ring such as piperidine or pyrrolidine). In another example, the terminal amino group of the lysine sidechain can form a ring with the amino acid backbone, as in capreomycidine. Further unnatural derivatives of lysine include homolysine and norlysine. The sidechain of lysine can alternatively be substituted by a second amino group. In another example, the alkyl portion of the lysine side chain can be incorporated into a carbocyclic ring structure to form a semirigid analog, such as, e.g., cyclohexyl or cyclopentyl.

Throughout this specification and claims, the 'L-threonine residue' and/or 'side chain of L-threonine' mentioned in compound of formula (I) or compounds of the present invention and/or preparation thereof can be represented by any one of the following formulae.

L-threonine L-threonine L-threonine L-threonine

In certain embodiments, the unnatural amino acid can be a derivative of a amino acid having one or more double bonds. In other example embodiments, in threonine, the beta-methyl group can be replaced with an ethyl, phenyl or other higher alkyl group. In histidine, the imidazole moiety can be substituted or alternatively, the alkylene backbone of the side chain can be substituted

Further examples of unnatural amino acids include homoserine and homologs of natural amino acids.

In further example embodiments, an unnatural amino acid can be alkylated (e.g., methylated) at the alpha position.

Further examples of unnatural amino acids include alpha,beta- and beta,gamma- dehydroamino amino acid analogs.

Further exemplary amino acids include penicillamine and betamethoxyvaline.

Further examples of unnatural amino acids include the amino acids wherein the side chain comprises amino, alkylamino, acylamino, -COO-alkyl, cycloalkyl, heterocyclyl, heteroaryl, guanidino, (cycloalkyl)alkyl, (heterocyclyl)alkyl and (heteroaryl)alkyl.

"Modified N-terminal amino group" and "modified C-terminal carboxyl group" mean that the amino group or carboxyl group is altered.

Modification of the N-terminal amino group is preferably with the general formula -NR x R y ; wherein R x is hydrogen or alkyl and R y is alkyl, alkenyl, -C(=NH)NH2, alkynyl or acyl.

Examples of N-terminal modifications include, but are not limited to, are acetylated, formylated or guanylated N-termini.

Modification of the C-terminal carboxyl group is preferably with the general formula COR z (R z replaces the hydroxyl group of the last amino acid); wherein R z is - NR R c, alkoxy, amino or an imide; wherein R and R c are independently are hydrogen, (Ci-Ce)alkyl, aryl or heterocyclyl; wherein (Ci-C6)alkyl, aryl and heterocyclyl are optionally substituted by one or more substituents selected from halogen, hydroxyl, amino, nitro, cyano, cycloalkyl, heterocyclyl, heteroaryl, aryl, guanidino, (cycloalkyl)alkyl, (heterocyclyl)alkyl and (heteroaryl)alkyl. This invention includes pharmaceutically acceptable salts of compounds of the invention and their use in the compositions and methods of the present invention. In certain embodiments, contemplated salts of the invention include, but are not limited to, alkyl, dialkyl, trialkyl or tetra-alkyl ammonium salts. In certain embodiments, contemplated salts of the invention include, but are not limited to, L-arginine, benenthamine, benzathine, betaine, calcium hydroxide, choline, deanol, diethanolamine, diethylamine, 2-(diethylamino)ethanol, ethanolamine, ethylenediamine, N- methylglucamine, hydrabamine, lH-imidazole, lithium, L-lysine, magnesium, 4-(2- hydroxyethyl)morpholine, piperazine, potassium, l-(2-hydroxyethyl)pyrrolidine, sodium, triethanolamine, tromethamine and zinc salts. In certain embodiments, contemplated salts of the invention include, but are not limited to, Na, Ca, K, Mg, Zn or other metal salts.

The pharmaceutically acceptable acid addition salts can also exist as various solvates, such as with water, methanol, ethanol, dimethylformamide and the like. Mixtures of such solvates can also be prepared. The source of such solvate can be from the solvent of crystallization, inherent in the solvent of preparation or crystallization or adventitious to such solvent.

"Pharmaceutically acceptable" means that which is useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable and includes that which is acceptable for veterinary as well as human pharmaceutical use.

The term "stereoisomers" refers to any enantiomers, diastereoisomers or geometrical isomers, such as of the compounds of the invention. When compounds of the invention are chiral, they can exist in racemic or in optically active form. Since the pharmaceutical activity of the racemates or stereoisomers of the compounds according to the invention may differ, it may be desirable to use compounds that are enriched in one of the enantiomers. In these cases, the end product or even the intermediates can be separated into enantiomeric compounds by chemical or physical measures known to the person skilled in the art or even employed as such in the synthesis. In the case of racemic amines, diastereomers are formed from the mixture by reaction with an optically active resolving agent. Examples of suitable resolving agents are optically active acids such as the R and S forms of tartaric acid, diacetyltartaric acid, dibenzoyltartaric acid, mandelic acid, malic acid, lactic acid, suitable N-protected amino acids (for example N- benzoylproline or N-benzenesulfonylproline) or the various optically active camphorsulfonic acids. Also advantageous is chromatographic enantiomer resolution with the aid of an optically active resolving agent (for example dinitrobenzoylphenylglycine, cellulose triacetate or other derivatives of carbohydrates or chirally derivatised methacrylate polymers immobilised on silica gel).

In certain embodiments, compounds of the invention may be racemic. In certain embodiments, compounds of the invention may be enriched in one enantiomer. For example, a compound of the invention may have greater than 30% ee, 40% ee, 50% ee, 60% ee, 70% ee, 80% ee, 90% ee or even 95% or greater ee. In certain embodiments, compounds of the invention may have more than one stereocenter. In certain such embodiments, compounds of the invention may be enriched in one or more diastereomer. For example, a compound of the invention may have greater than 30% de, 40% de, 50% de, 60% de, 70% de, 80% de, 90% de or even 95% or greater de.

The term "subject" includes mammals (especially humans) and other animals, such as domestic animals (e.g., household pets including cats and dogs) and non- domestic animals (such as wildlife).

Naturally-occurring amino acids are identified throughout the description and claims by the conventional three-letter abbreviations indicated in the below table.

Table (Amino acid codes)

The abbreviations used in the entire specification may be summarized herein below with their particular meaning.

°C (degree Celsius); % (percentage); brine (NaCl solution); CH 2 C1 2 /DCM (Dichloromethane); Boc (Tert-butyloxycarbonyl); Bzl (Benzyloxy-carbonyl); CS2CO 3 (Caesium carbonate); DIC: Ν,Ν'-Diisopropylcarbodiimide; DIPEA (N,N- Diisopropylethylamine); DMF (Dimethyl formamide); EtOH (Ethanol); Et2NH (Diethylamine); Fmoc (9-Fluorenylmethyloxycarbonyl); g or gr (gram); HOBt (1- Hydroxy benzotriazole); h or hr (Hours); HPLC (High-performance liquid chromatography); K2CO 3 (Potassium carbonate); LCMS (Liquid chromatography mass spectroscopy); Liq.N¾ (Liquid ammonia); mmol (Millimoles); M (Molar); μΐ (Microlitre); mL (Millilitre); mg (Milligram); MS (ES) (Mass spectroscopy-electro spray); min (Minutes); Na (Sodium); NaHC0 3 (Sodium bicarbonate); NH 2 NH 2 .H 2 0 (Hydrazine hydrate); NMM (N-Methylmorpholine); Na 2 S0 4 (Sodium sulphate); NH 2 OH.HCl (Hydroxylamine hydrochloride); PD-l/PD-1 (Programmed cell death 1); PD-L1 (Programmed death-ligand 1); PD-L2 (Programmed cell death 1 ligand 2); prep- HPLC/preparative HPLC (Preparative High-performance liquid chromatography); TEA/Et 3 N (Trie thy lamine); TFAA: Tifluoroacetic anhydride; TLC (Thin Layer Chromatography); THF (Tetrahydrofuran); TIPS (Triisopropylsilane); TFA (Trifluoroacetic acid); t R (Retention time); Trt (Trityl or Triphenylmethyl), etc.

EXPERIMENTAL

An embodiment of the present invention provides the preparation of compounds of formula (I) according to the procedures of the following example(s), using appropriate materials. Those skilled in the art will understand that known variations of the conditions and processes of the following preparative procedures can be used to prepare these compounds. Moreover, by utilizing the procedures described in detail, one of ordinary skill in the art can prepare additional compounds of the present invention.

The starting materials are generally available from commercial sources such as Sigma-Aldrich, USA or Germany; Chem-Impex USA; G.L. Biochem, China and Spectrochem, India.

LCMS Conditions:

Method A: LC-MSD (Agilent 1100 series with Single Quad, Dual Mode mass spectrometer/ API 2000, Triple Quad, ESI/ APCI SHIMADZU LCMS-2020 WITH SINGLE QUAD)

Column Mercury MS Synergi 2μ, 20X4.0mm; Gradient: A- 0.1% formic acid in water/B-MeCN; 0-0.5min 70A-30B; 1.5-2.4min 5A-95B; 2.5-3.0min 70A-30B; Flow 2.0mL/min; Column temperature 30 °C.

Method B:

LC-MSD (Agilent 1100 series with Single Quad, Dual Mode mass spectrometer/ API 2000, Triple Quad, ESI / APCI SHIMADZU LCMS-2020 WITH SINGLE QUAD) Column Mercury MS Synergi 2μ, 20X4.0mm; Gradient: A- 0.1% formic acid in water/B-MeCN; 0-0.5min 30A-70B; 1.5-2.4min 100B-0A; 2.5-3.0min 30A-70B; Flow 2.0mL/min; Column temperature 30 °C.

X H-NMR Instrument: Varian Mercury 300 MHz and Varian 400 MHz (MR-400)

Examples

The present invention is explained in detail in the following by referring to compounds of Experimental Examples. However, the examples do not limit the present invention and the present invention can be modified within the scope of the present invention.

Example 1

ompound 1

Step 1: Synthesis of compound lb

1a 1 b

Potassium carbonate (18.8 g, 136.3 mmol) was added to a stirred solution of 1- cyanopiperidine (compound la) (5.0 g, 45.4 mmol) in ethanol (40.0 mL) . To the resulting reaction mixture hydroxylamine hydrochloride (6.30 g, 90.9 mmol) in water (20.0 mL) was added and the solution was stirred at 80 °C for 16h. After completion of 16 h, the reaction mixture was diluted with water (50.0 mL) and was extracted with EtOAc (3 x 100 mL). The combined organic layers were washed once with brine (ca. 100 mL), dried over Na2SC>4 and the solvents were removed under reduced pressure to yield compound lb (4.1 g). LCMS: 144.3 (M+H) + .

Step 2: Synthesis of compound Id

HOBT (1.15 g, 7.54 mmol) and DIC (0.95 g, 7.54 mmol) were added to a stirred solution of the compound lc (3.0 g, 5.03 mmol) in DMF (3.0 mL) under argon atmosphere at room temperature and the reaction mixture was stirred at room temperature for 15 min. Compound lb (0.72 g, 5.03 mmol) was added to the above reaction mass and the resultant mixture was stirred at room temperature for 16 h, before pouring the reaction contents into ice-cold water. The precipitate thus generated was collected by filtration, re-dissolved in EtOAc, dried over Na2SC>4 and the solvents were removed under reduced pressure to obtain compound Id (3.1 g). LCMS: 722.3 (M+H) + . Step 3: Synthesis of compound le

1d 1e

Acetic acid (3.1 mL) was added to a stirred solution of compound Id (3.1 g, 4.30 mmol) in MeCN (30.0 mL) under inert atmosphere and the resultant mixture was stirred at 70 °C for 16h, until the completion of the reaction. The reaction mixture was then cooled down to room temperature, poured onto crushed ice. The resultant precipitate was collected by vacuum filtration, washed with water and was dried under vacuum. The crude product was further purified by MPLC (CombiFlash, Gradient: 3:2 Hex-EtOAc) to obtain the compound le (2.20 g).

J H-NMR (DMSO-De, 400 MHz,) δ = 8.81 (s, 1H), 8.17-8.12 (m, 1H), 7.92-7.86 (m, 2H), 7.74-7.68 (m, 2H), 7.44-7.34 (m, 2H), 7.32-7.10 (m, 16H), 5.05-4.98 (m, 1H), 4.45-4.19 (m, 3H), 3.39-3.28 (m, 4H), 3.01-2.94 (m, 1H), 2.84-2.77 (m, 1H), 1.61-1.49 (m, 6H). Step 4: Synthesis of compound If

1e 1f

Piperidine (2.5 mL) was added to a stirred solution of le (2.20 g, 3.13 mmol) in dry DMF (9.0 mL) under inert atmosphere and the resultant mixture was stirred at room temperature for 3h, until the completion of the reaction. The reaction mixture was poured onto crushed ice. The precipitate generated was collected by vacuum filtration, washed with water and dried in vacuum. The crude product was then repeatedly agitated with pentane and the solvent was removed by filtration (3 x 20 mL) to obtain the title compound If (1.10 g).

^-NMR (DMSO-D 6 , 300 MHz) δ = 9.06 (s, 1H), 7.30-7.12 (m, 15H), 4.24-4.38 (m, 1H), 3.39-3.28 (m, 4H), 2.75-2.70 (m, 2H), 2.30-2.22 (m, 2H), 1.60-1.51 (m, 6H).

Step 5: Synthesis of Com ound 1

Compound 1

TFA (2.0 mL) was added to the compound If (0.20 g, 0.42 mmol) followed by triisopropylsilane (0.30 mL) and the resultant mixture was stirred at room temperature for 3 h. The solvents were removed under reduced pressure and the crude product thus obtained was agitated with diethyl ether (20.0 mL), before collecting the solid product by vacuum filtration. The solid was repeatedly washed with diethyl ether (2 x 10 mL) to obtain the Compound 1. LCMS: 240.3 (M+H) + .

The below compounds were prepared according to the procedure described in Example- 1 (compound 1) with appropriate variations in reactants, quantities of reagents, solvents and reaction conditions. The characterization data of the compounds are summarized herein below table.

Example 2

Compound 15

Step 1: Synthesis of compound

H-ThrC'BiO-O'Bu (compound 2a) (5.0 g, 21.61 mmol), TEA (6.2 mL, 43.22 mmol) in CH2CI2 (25 mL) was slowly added to a solution of 4-nitrophenylchloroformate (4.79 g, 23.77 mmol) in DCM (25.0 mL) at 0 °C and allowed to stir for 30 min. The completion of the reaction was confirmed by TLC analysis. After completion of reaction it was diluted with DCM and washed with 1.0 M of citric acid followed by l.OM sodium carbonate solution. The organic layer was dried over Na2SC>4 and evaporated under reduced pressure to afford crude material, which was further purified by silica gel column chromatography (eluent: 0-5% ethyl acetate in hexane) to obtain the title compound 2b (3.0 g).

1H-NMR (CDCI 3 , 400 MHz): δ= 1.17 (s, 9H), 1.28 (d, 3H), 1.50 (s, 9H), 4.11 (m, 1H), 4.28 (m, 1H), 5.89 (d, 1H), 7.37 (d, 2H), 8.26 (d, 2H).

Triethylamine (0.13 g, 0.62 mmol) was added to a stirred solution of compound If (0.30 g, 0.62 mmol) in dry DMF (3.0 mL) under inert atmosphere at 0 °C. was added Compound 2b (0.25 g, 0.62 mmol) was further added to the above mixture and the resultant mixture was stirred at 0 °C for 2h until the completion of the reaction. The reaction mixture was poured onto crushed ice, the precipitated material was collected by vacuum filtration, washed with ice cold water and was dried under vacuum. The crude product thus obtained was dissolved in DCM (5.0 mL) to which was added n-pentane (10.0 mL) and the resultant precipitate was collected by vacuum filtration to obtain the title compound 2c (0.30 g). LCMS: 739.2 (M+H) + .

Step 3:

Compound 15

TFA (3.0 mL) was added to the compound 2c (0.30 g, 0.41 mmol) followed by triisopropylsilane (0.90 mL) and the resultant mixture was stirred at room temperature for 2 h. The solvents were removed under reduced pressure and the crude product thus obtained was precipitated using diethyl ether (20.0 mL), before collecting the solid product by vacuum filtration. The solid was repeatedly washed with diethyl ether (2 x 10 mL) to obtain the title compound 15. HPLC: 98.9 % [t R = 5.55 min], LCMS: 385.2 (M+H) + .

1H-NMR (DMSO-D 6 , 400 MHz): δ = 12.45 (bs, 1H), 7.48 (s, 1H), 7.03-6.98 (m, 2H), 6.40 (d, 1H), 5.09 (q, 1H), 4.12-4.10 (m, 1H), 4.04 (dd, 1H), 3.30 (m, 4H), 2.66 (d, 2H), 1.54 (m, 6H), 1.04 (d, 3H).

The below compounds were prepared by procedure similar to the one described in Example 2 (compound 15) with appropriate variations in reactants, quantities of reagents, solvents and reaction conditions. The characterization data of the compounds are summarized herein below table.

Comp. LCMS

Structure 1 H-NMR

No (M+H) +

7.54 (s, 1H), 7.17 (d, 1H), 7.06 (d,

0 1H), 7.03 (s, 1H), 6.49 (d, 1H), 6.36

26 382.0 (dd, 1H), 5.28 (q, 1H), 4.11 (dd, 1H),

A H O H H OH 4.04 (d, 1H), 2.82-2.74 (m, 2H), 2.38

(s, 3H), 1.05 (d, 3H)

The below compounds were prepared by procedure similar to the one described in Example 2 (compound 15) with appropriate variations in reactants, quantities of reagents, solvents and reaction conditions. The characterization data of the compounds are summarized herein below table.

Example- 3

Compound 51

Step 1: Synthesis of compound 3b

3a 3b

HOBt (4.60 g, 34.01 mmol) and EDC.HC1 (6.52 g, 34.01 mmol) were added to a solution of compound 3a (8.14 g, 28.34 mmol) in DCM (100.0 mL) and the resultant mixture was stirred at 0 °C for 30 min under inert atmosphere. Triethylamine (12.0 mL, 85.02 mmol) and liq. ammonia (5.5 mL, 141.7 mmol) was added to the above reaction mixture and the solution was stirred at 0-5 °C for 14 h, when TLC-analysis has indicated the completion of the reaction. The reaction mixture was partitioned between water and DCM and the organic layer was washed with saturated NaHCC>3 solution, brine and water and was dried over Na2SO/t. The solvent was removed under reduced pressure to obtain the crude amide, which was further purified by column chromatography (Silica gel, 0-50% EtOAc in Hexane) to yield compound 3b (5.30 g). LCMS: 287.2 (M+H) + . Step 2: Synthesis of com ound 3c

3b 3c

Trifluroacetic anhydride (4.0 mL, 27.75 mmol) was added to a solution of compound 3b (5.30 g, 18.5 mmol) in pyridine (25.3 mL) and the mixture was stirred at room temperature for 3 h. The completeness of the reaction was confirmed by TLC analysis. The reaction mixture was then diluted with water and the resulting solid was collected by vacuum filtration and was dried under vacuum to obtain compound 3c (4.12 g). LCMS: 269.2 (M+H) + . Step 3: Synthesis of compound 51

The compound 51 was prepared according to the procedure described in steps 2 - 5 of Example - 1 (Compound 1) with appropriate variations in reactants, quantities of reagents, solvents and reaction conditions. LCMS (M+H) + : 387.4.

Compound 52

The compound 52 was prepared according to the procedure described in Example - 3 with appropriate variations in reactants, quantities of reagents, solvents and reaction conditions. LCMS (M+H) + : 355.1

Although the present application has been illustrated by certain of the preceding examples, it is not to be construed as being limited thereby; but rather, the present application encompasses the generic area as hereinbefore disclosed. Various modifications and embodiments can be made without departing from the spirit and scope thereof. For example, the following compounds which can be prepared by following similar procedure as described above with suitable modification known to the one ordinar skilled in the art are also included in the scope of the present application:

Example 4: Rescue of mouse splenocyte proliferation in the presence of recombinant PD-L1

Recombinant mouse PD-L1 (rm-PDL-1, cat no: 1019-B7-100; R&D Systems) were used as the source of PD-L1.

Requirement:

Mouse splenocytes harvested from 6-8 weeks old C57 BL6 mice; RPMI 1640 (GIBCO, Cat # 11875); DMEM with high glucose (GIBCO, Cat # D6429); Fetal Bovine Serum [Hyclone, Cat # SH30071.03]; Penicillin (10000unit/mL)- Streptomycin(10,000μg/mL) Liquid (GIBCO, Cat # 15140-122); MEM Sodium Pyruvate solution lOOmM (lOOx), Liquid (GIBCO, Cat # 11360); Nonessential amino acid (GIBCO, Cat # 11140); L-Glutamine (GIBCO, Cat # 25030); Anti-CD3 antibody (eBiosciences - 16-0032); Anti-CD28 antibody (eBiosciences - 16-0281); ACK lysis buffer (lmL) (GIBCO, Cat # -A10492); Histopaque (density- 1.083 gm/mL) (SIGMA 10831); Trypan blue solution (SIGMA-T8154); 2 mL Norm Ject Luer Lock syringe - (Sigma 2014-12); 40μπι nylon cell strainer (BD FALCON 35230); Hemacytometer (Bright line-SIGMA Z359629); FACS Buffer (PBS/0.1% BSA): Phosphate Buffered Saline (PBS) pH 7.2 (Hi-Media TS1006) with 0.1% Bovine Serum Albumin (BSA) (SIGMA A7050) and sodium azide (SIGMA 08591); 5 mM stock solution of CFSE: CFSE stock solution was prepared by diluting lyophilized CFSE with 180 μL· of Dimethyl sulfoxide (DMSO C 2 H 6 SO, SIGMA-D-5879) and aliquoted in to tubes for further use. Working concentrations were titrated from 10 μΜ to 1 μΜ. (eBioscience- 650850-85); 0.05% Trypsin and 0.02% EDTA (SIGMA 59417C); 96-well format ELISA plates (Corning CLS3390); BD FACS caliber (E6016); Recombinant mouse B7- H1/PDL1 Fc Chimera, (rm-PD-Ll cat no: 1019-B7-100).

Splenocyte preparation and culturing:

Splenocytes harvested in a 50 mL falcon tube by mashing mouse spleen in a 40 μπι cell strainer were further treated with 1 mL ACK lysis buffer for 5 min at room temperature. After washing with 9 mL of RPMI complete media, cells were re- suspended in 3 mL of lxPBS in a 15 mL tube. 3 mL of Histopaque was added carefully to the bottom of the tube without disturbing overlaying splenocyte suspension. After centrifuging at 800xg for 20 min at room temperature, the opaque layer of splenocytes was collected carefully without disturbing / mixing the layers. Splenocytes were washed twice with cold lxPBS followed by total cell counting using Trypan Blue exclusion method and used further for cell based assays.

Splenocytes were cultured in RPMI complete media (RPMI + 10% fetal bovine serum + ImM sodium pyruvate + 10,000units/mL penicillin and 10,000μg/mL streptomycin) and maintained in a C(¾ incubator with 5% C(¾ at 37 °C.

CFSE Proliferation assay:

CFSE is a dye that passively diffuses into cells and binds to intracellular proteins. lxlO 6 cells/mL of harvested splenocytes were treated with 5 μΜ of CFSE in pre-warmed lxPBS/0.1% BSA solution for 10 min at 37 °C. Excess CFSE was quenched using 5 volumes of ice-cold culture media to the cells and incubated on ice for 5 min. CFSE labelled splenocytes were further given three washes with ice cold complete RPMI media. CFSE labelled lxlO 5 splenocytes added to wells containing either MDA-MB231 cells (1x10 s cells cultured in high glucose DMEM medium) or recombinant human PDL- 1 (100 ng/mL) and compounds of the present invention. Splenocytes were stimulated with anti-mouse CD3 and anti- mouse CD28 antibody (lμg/mL each) and the culture was further incubated for 72 h at 37 °C with 5% C(¾. Cells were harvested and washed thrice with ice cold FACS buffer and % proliferation was analysed by flow cytometry with 488 nm excitation and 521 nm emission filters.

Data compilation, processing and inference:

Percent splenocyte proliferation was analysed using cell quest FACS program and percent rescue of splenocyte proliferation by compound was estimated after deduction of % background proliferation value and normalising to % stimulated splenocyte proliferation (positive control) as 100%. The results are given in Table I.

Stimulated splenocytes: Splenocytes + anti-CD3/CD28 stimulation

Background proliferation: Splenocytes + anti-CD3/CD28 + PD-L1

Compound proliferation: Splenocytes + anti-CD3/CD28 + PD-L1 + Compound

Compound effect is examined by adding required cone, of compound to anti-CD3/CD28 stimulated splenocytes in presence of ligand (PDL-1).

Table I: Percent rescue of splenocyte proliferation data of compounds of invention