Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
1,2,4-OXADIAZOLE DERIVATIVES AS IMMUNOMODULATORS
Document Type and Number:
WIPO Patent Application WO/2015/033299
Kind Code:
A1
Abstract:
The present invention relates to 1,2,4-oxadiazole and 1,2,4-thiadiazole compounds as therapeutic agents capable of inhibiting the programmed cell death 1 (PD1) signaling pathway. The invention also refers to derivatives of the therapeutic agents. The invention also encompasses the use of the said therapeutic agents and 5 derivatives 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.

Inventors:
SASIKUMAR POTTAYIL GOVINDAN NAIR (IN)
RAMACHANDRA MURALIDHARA (IN)
NAREMADDEPALLI SEETHARAMAIAH SETTY SUDARSHAN (IN)
Application Number:
PCT/IB2014/064279
Publication Date:
March 12, 2015
Filing Date:
September 05, 2014
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
AURIGENE DISCOVERY TECH LTD (IN)
International Classes:
C07D271/06; A61K31/4245; A61K31/433; C07D285/08
Domestic Patent References:
WO2012168944A12012-12-13
WO2001014557A12001-03-01
WO2002079499A12002-10-10
WO2002086083A22002-10-31
WO2003042402A22003-05-22
WO2004004771A12004-01-15
WO2004056875A12004-07-08
WO2006121168A12006-11-16
WO2008156712A12008-12-24
WO2010077634A12010-07-08
WO2011066389A12011-06-03
WO2014055897A22014-04-10
WO2014059173A22014-04-17
WO2014100079A12014-06-26
WO2011161699A22011-12-29
WO2012168944A12012-12-13
WO2013144704A12013-10-03
WO2013132317A12013-09-12
Foreign References:
US8735553B12014-05-27
Other References:
SUSSAN K. ARDESTANI, FATEMAH POORRAJAB, SEPIDEH RAZMI, ALIREZA FOROUMADI, SOHEILA AJDARY, BEHNAZ GHAREGOZLOU ET AL.: "Cell death features induced in Leishmania major by 1, 3, 4-thiadiazole derivatives", EXPERIMENTAL PARASITOLOGY, vol. 132, 14 August 2012 (2012-08-14), pages 116 - 122, XP002732914
ARIEL PEDOEEM ET AL., CURR TOP MICROBIOL IMMUNOL., vol. 350, 2011, pages 17 - 37
LI SHI ET AL., JOURNAL OF HEMATOLOGY & ONCOLOGY, vol. 6, 2013, pages 74
RD HARVEY, CLINICAL PHARMACOLOGY & THERAPEUTICS, vol. 96, no. 2, 2014, pages 214 - 223
HYUN-TAK JIN ET AL., CLINICAL IMMUNOLOGY (AMSTERDAM, NETHERLANDS, vol. 153, no. 1, 2014, pages 145 - 152
Download PDF:
Claims:
A compound of formula (I)

wherein,

Q is S or O;

Ri is side chain of an amino acid Ser or Thr, optionally substituted with alkyl or acyl;

R2 is hydrogen or -CO- Aaa;

Aaa is an amino acid residue Thr or Ser; wherein a C-terminus thereof is a free terminus, is amidated or is esterified;

R3 is side chain of an amino acid selected from Asn, Asp, Gin or Glu;

is an optional bond;

R4 and R5 independently are hydrogen or absent;

R6 is selected from hydrogen, alkyl or acyl;

or a pharmaceutically acceptable salt or a stereoisomer thereof.

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

or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein,

Ri is side chain of an amino acid Ser or Thr, optionally substituted with alkyl or acyl;

R3 is side chain of an amino acid selected from Asn, Asp, Gin or Glu;

Aaa is an amino acid residue Thr or Ser; wherein a C-terminus thereof is a free terminus, is amidated or is esterified.

3. The compound according to claim 1, wherein the compound of formula (I) is a compound of formula (IB):

(IB)

or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein,

Ri is side chain of an amino acid Ser or Thr, optionally substituted with alkyl or acyl;

R3 is side chain of an amino acid selected from Asn, Asp, Gin or Glu.

4. The compound according to claim 1 , wherein

Ri is side chain of Ser or Thr;

R2 is -CO- Aaa;

Aaa is an amino acid residue Thr or Ser; wherein the C-terminus is free;

R3 is side chain of Asn or Glu.

5. The compound according to claim 1 , wherein Q is O.

6. The compound according to claim 1 , wherein R6 is -C(0)CH3, -C(0)CH2CH3j C(0)(CH2)2CH3, -C(0)(CH2)3CH3, -C(0)(CH2)4CH3 or -C(0)(CH2)5CH3.

7. A compound according to claim 1 is selected from the group consisting of

or a p armaceut ca y accepta e sa t or a stereo somer t ereo .

8. A pharmaceutical composition comprising at least one compound according to any one of claims 1 to 7 or a pharmaceutically acceptable salt or a stereoisomer thereof, and a pharmaceutically acceptable carrier or excipient.

9. The pharmaceutical composition according to claim 8 further comprising at least one of an anticancer agent, chemotherapy agent, or antiproliferative compound.

10. A compound according to any one of claims 1 to 7, or a pharmaceutically acceptable salt or a stereoisomer thereof, for use as a medicament.

11. A compound according to any one of claims 1 to 7, or a pharmaceutically acceptable salt or a stereoisomer thereof, for use as a medicament for the treatment of cancer or infectious disease.

12. The compound for use according to claim 1 1, wherein the cancer is selected from the group consisting of breast cancer, colon cancer, lung cancer, melanoma, prostate cancer and renal cancer, bone cancer, cancer of the head or neck, pancreatic cancer, skin cancer, cutaneous or intraocular malignant melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, testicular cancer, uterine 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), 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.

13. The compound for use according to claim 11, wherein the cancer is breast cancer, colon cancer, lung cancer, melanoma, prostate cancer and renal cancer.

14. The compound for use according to claim 1 1, wherein the infectious disease is a bacterial infectious disease, a viral infectious disease or a fungal infectious disease.

Description:
1,2,4-OXADIAZOLE DERIVATIVES AS IMMUNOMODULATORS

This application claims the benefit of Indian provisional application number 401 l/CHE/2013, filed on September 06, 2013; which hereby incorporated by reference. TECHNICAL FIELD

The present invention relates to 1,2,4-oxadiazole and 1 ,2,4-thiadiazole compounds and their derivatives therapeutically useful as immune modulators. The invention also relates to pharmaceutical compositions comprising the said 1,2,4- oxadiazole and 1,2,4-thiadiazole compounds and their derivatives as therapeutic agents. BACKGROUND OF THE INVENTION

Programmed cell death- 1 (PD-1) is a member of the CD28 superfamily that delivers negative signals upon interaction with its two ligands, PD-L1 or PD-L2. PD-1 and its ligands are broadly expressed and exert a wider range of immunoregulatory roles in T cells activation and tolerance compared with other CD28 members. PD-1 and its ligands are involved in attenuating infectious immunity and tumor immunity, and facilitating chronic infection and tumor progression. The biological significance of PD- 1 and its ligand suggests the therapeutic potential of manipulation of PD- 1 pathway against various human diseases (Ariel Pedoeem et al., Curr Top Microbiol Immunol. (2011); 350:17-37).

T-cell activation and dysfunction relies on direct and modulated receptors. Based on their functional outcome, co-signaling molecules can be divided as co-stimulators and co-inhibitors, which positively and negatively control the priming, growth, differentiation and functional maturation of a T-cell response (Li Shi, et al., Journal of Hematology & Oncology 2013, 6:74).

Therapeutic antibodies that block the programmed cell death protein-1 (PD-1) immune checkpoint pathway prevent T-cell down regulation and promote immune responses against cancer. Several PD-1 pathway inhibitors have shown robust activity in various phases of on-going clinical trials (RD Harvey, Clinical Pharmacology & Therapeutics (2014); 96 2, 214-223).

Programmed death-1 (PD-1) is a co-receptor that is expressed predominantly by T cells. The binding of PD-1 to its ligands, PD-L1 or PD-L2, is vital for the physiological regulation of the immune system. A major functional role of the PD-1 signaling pathway is the inhibition of self-reactive T cells, which serve to protect against autoimmune diseases. Elimination of the PD-1 pathway can therefore result in the breakdown of immune tolerance that can ultimately lead to the development of pathogenic autoimmunity. Conversely, tumor cells can at times co-opt the PD-1 pathway to escape from immunosurveillance mechanisms. Therefore, blockade of the PD-1 pathway has become an attractive target in cancer therapy. Current approaches include six agents that are either PD-1 and PD-L1 targeted neutralizing antibodies or fusion proteins. More than forty clinical trials are underway to better define the role of PD-1 blockade in variety of tumor types (Hyun-Tak Jin et al., Clinical Immunology (Amsterdam, Netherlands) (2014), 153(1), 145-152).

International applications WO 01/14557, WO 02/079499, WO 2002/086083, WO 03/042402, WO 2004/004771 , WO 2004/056875, WO2006121168, WO2008156712, WO2010077634, WO2011066389, WO2014055897, WO2014059173, WO2014100079 and US patent US08735553 report PD-1 or PD-L1 inhibitory antibodies or fusion proteins.

Further, International applications, WO2011161699, WO2012/168944, WO2013144704 and WO2013132317 report peptides or peptidomimetic compounds which are capable of suppressing and/or inhibiting the programmed cell death 1 (PD1) signaling pathway.

Still there is a need for more potent, better and/or selective immune modulators of PD-1 pathway. The present invention provides 1 ,2,4-oxadiazole and 1 ,2,4-thiadiazole compounds which are capable of suppressing and/or inhibiting the programmed cell death 1 (PD1) signaling pathway.

SUMMARY OF INVENTION

In accordance with the present invention, 1 ,2,4-oxadiazole and 1 ,2,4-thiadiazole compounds or a pharmaceutically acceptable salt or a stereoisomer thereof, provided which are capable of suppressing and/or inhibiting the programmed cell death 1 (PD1) signaling pathway.

In one aspect, the present invention provides a 1,2,4-oxadiazole and 1 ,2,4- thiadiazole compounds of formula (I):

wherein,

Q is S or O;

Ri is side chain of an amino acid Ser or Thr, optionally substituted with alkyl or acyl;

R 2 is hydrogen or -CO-Aaa;

Aaa is an amino acid residue Thr or Ser; wherein a C-terminus thereof is a free terminus, is amidated or is esterified;

R 3 is side chain of an amino acid selected from Asn, Asp, Gin or Glu;

is an optional bond;

R4 and R5 independently are hydrogen or absent;

R6 is selected from hydrogen, alkyl or acyl;

or a pharmaceutically acceptable salt or a stereoisomer thereof.

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

In yet another aspect of the present invention, it provides use of 1 ,2,4-oxadiazole and 1 ,2,4-thiadiazole compounds of formula (I) or a pharmaceutically acceptable salt or a stereoisomer thereof, which are capable of suppressing and/or inhibiting the programmed cell death 1 (PD1) signaling pathway.

DETAILED DESCRIPTION OF THE INVENTION

The present invention provides 1 ,2,4-oxadiazole and 1 ,2,4-thiadiazole compounds 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 one embodiment, the present invention relates to compounds of formula (I)

wherein,

Q is S or O;

Ri is side chain of an amino acid Ser or Thr, optionally substituted with alkyl or acyl;

R 2 is hydrogen or -CO- Aaa;

Aaa is an amino acid residue Thr or Ser; wherein a C-terminus thereof is a free terminus, is amidated or is esterified;

R 3 is side chain of an amino acid selected from Asn, Asp, Gin or Glu;

is an optional bond;

R4 and R5 independently are hydrogen or absent;

R6 is selected from hydrogen, alkyl or acyl;

or a pharmaceutically acceptable salt or a stereoisomer thereof.

In yet another embodiment, the present invention provides compounds of formula (IA)

or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein, Ri, R3 and Aaa are same as defined in formula (I).

In yet another further embodiment, the present invention provides compounds formula (IB)

(IB)

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

According to yet another embodiment, the present invention provides compounds of the formula (I) in which

Ri is side chain of Ser or Thr;

R 2 is -CO-Aaa;

Aaa is an amino acid residue Thr or Ser; wherein the C-terminus is free;

R 3 is side chain of Asn or Glu.

In yet another further embodiment, the compound according to formula (I) in which Q is S.

In yet another further embodiment, the compound according to formula (I) in which R4 is hydrogen.

In yet another further embodiment, the compound according to formula (I) in which R 5 is hydrogen.

The embodiments below are illustrative of the present invention and are not intended to limit the claims to the specific embodiments exemplified.

According to one embodiment, specifically provided are compounds of the formula (I), (IA) and (IB) in which Ri is side chain of Ser.

According to another embodiment, specifically provided are compounds of the formula (I), (IA) and (IB) in which Ri is side chain of Thr.

According to yet another embodiment, specifically provided are compounds of the formula (I) in which R 2 is hydrogen.

According to yet another embodiment, specifically provided are compounds of the formula (I) in which R 2 is -CO-Aaa.

According to yet another embodiment, specifically provided are compounds of the formula (I) and (IA) in which Aaa is Thr.

According to yet another embodiment, specifically provided are compounds of the formula (I) and (IA) in which Aaa is Ser. According to yet another embodiment, specifically provided are compounds of the formula (I), (IA) and (IB) in which R 3 is side chain of Asn.

According to yet another embodiment, specifically provided are compounds of the formula (I) and (IA) in which R3 is side chain of Asp.

According to yet another embodiment, specifically provided are compounds of the formula (I) and (IA) in which R 3 is side chain of Gin.

According to yet another embodiment, specifically provided are compounds of the formula (I) and (IA) in which R 3 is side chain of Glu.

According to yet another embodiment, specifically provided are compounds of the formula (I) in which Q is O.

According to yet another embodiment, specifically provided are compounds of the formula (I) in which Ri is side chain of Ser, optionally substituted with Ci_5 alkyl such as methyl.

According to yet another embodiment, specifically provided are compounds of the formula (I) in which R6 is hydrogen.

According to yet another embodiment, specifically provided are compounds of the formula (I) in which R 6 is acyl such as -C(0)CH 3 , -C(0)CH 2 CH 3 , -C(0)(CH 2 ) 2 CH 3 , - C(0)(CH 2 ) 3 CH 3 , -C(0)(CH 2 ) 4 CH 3 or -C(0)(CH 2 ) 5 CH 3 .

According to yet another embodiment, specifically provided are compounds of the formula (I) in which R 6 is butyryl.

According to yet another embodiment, specifically provided are compounds of the formula (I) in which R4 and R5 are absent.

According to yet another embodiment, specifically provided are compounds of the formula (I) in which C-terminus of Aaa is free (e.g. -C0 2 H form).

According to yet another embodiment, specifically provided are compounds of the formula (I) in which in which C-terminus of Aaa is esterified (e.g. -C0 2 Me form).

According to yet another embodiment, specifically provided are compounds of the formula (I) in which C-terminus of Aaa is amidated (e.g. -CONH 2 form).

According to yet another embodiment, specifically provided are compounds of the formula (I), (IA) and (IB) in which one, more or all amino acid/s is/are D amino acid/s.

In an embodiment, specific compounds of formula (I) without any limitation are enumerated in table (1): Table 1

or a pharmaceutically acceptable sa t or a stereoisomer thereof.

In one embodiment, the present invention provides a pharmaceutical composition comprising the compound as disclosed, and a pharmaceutically acceptable carrier or diluent.

In another embodiment, the said pharmaceutical composition further comprising at least one of an anticancer agent, chemotherapy agent, or antiproliferative compound.

The compounds as disclosed in the present invention are formulated for pharmaceutical administration.

In one embodiment, the present invention provides compounds as disclosed in the present invention for use in the preparation of a medicament.

In another embodiment, the present invention provides compounds as disclosed in the present invention for use in the preparation of a medicament for the treatment of cancer or infectious disease.

In one embodiment, the present invention provides compounds as disclosed in the present invention for use in the preparation of a medicament for the treatment of bacterial infectious disease, a viral infectious disease or a fungal infectious disease. In one embodiment, the present invention provides compounds for use in the preparation of a medicament for the treatment of cancer wherein the cancer is selected from the group consisting of breast cancer, colon cancer, lung cancer, melanoma, prostate cancer and renal cancer, bone cancer, cancer of the head or neck, pancreatic cancer, skin cancer, cutaneous or intraocular malignant melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, testicular cancer, uterine 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), 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 one embodiment, the present invention provides the compounds as disclosed in the present invention for use in the treatment of cancer.

In one embodiment, the present invention provides the compounds as disclosed in the present invention for use in the treatment of bacterial infectious disease, a viral infectious disease or a fungal infectious disease.

In one embodiment, the present invention provides a method of treatment of cancer, wherein the method comprises administration of an effective amount of the compound of the present invention to the subject in need thereof.

In another embodiment the present invention provides a method of modulating an immune response mediated by PD-1 signaling pathway in a subject, comprising administering to the subject a therapeutically effective amount of the compound of the present invention such that the immune response in the subject is modulated.

In yet another embodiment the present invention provides a method of inhibiting growth of tumour cells and/or metastasis in a subject, comprising administering to the subject a therapeutically effective amount of compound of the present invention capable of inhibiting the programmed cell death 1 (PD1) signaling pathway.

The said tumour cells include cancer such as but not limited to bone cancer, cancer of the head or neck, pancreatic cancer, skin cancer, cutaneous or intraocular malignant melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, testicular cancer, uterine 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), 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 yet another further embodiment the present invention provides a method of treating an infectious disease in a subject comprising administering to the subject a therapeutically effective amount of the compound of the present invention capable of inhibiting the programmed cell death 1 (PD1) signaling pathway such that the subject is treated for the infectious disease.

Still yet another embodiment of the present invention provides a method of treating bacterial, viral and fungal infections in a subject comprising administering to the subject a therapeutically effective amount of the compound of the present invention capable of inhibiting the programmed cell death 1 (PD1) signaling pathway such that the subject is treated for the bacterial, viral and fungal infections.

The infectious disease includes but 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 Barr 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 or as a pharmaceutical composition in which the compound is mixed with various pharmacologically acceptable materials.

The pharmaceutical composition is usually administered by oral or inhalation routes, but can be administered by parenteral administration route. In the practice of this invention, compositions can be administered, for example, by orally, intravenous infusion, topically, intraperitoneally, intravesically or intrathecally. Examples of the parenteral administration includes but not limited to intraarticular (in the joints), intravenous, intramuscular, intradermal, intraperitoneal, and subcutaneous routes, include aqueous and non-aqueous, isotonic sterile injection solutions, which can 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 the preferred methods of administration.

The dosage of the compounds of the present invention varies depending on age, weight, symptom, therapeutic efficacy, dosing regimen and/or treatment time. Generally, they may be administered by oral or inhalation routes, in an amount of 1 mg to 100 mg per time, from once a couple of days, once 3 days, once 2 days, once a day to a couple of times a day, in the case of an adult, or continuously administered by oral or inhalation routes from 1 to 24 hours a day. Since the dosage is affected by various conditions, an amount less than the above dosage may sometimes work well enough, or higher dosage may be required in some cases.

The compounds of the present invention may be administered in combination with other drugs for (1) complementation and/or enhancement of prevention and/or therapeutic efficacy of the preventive and/or therapeutic drug of the present invention, (2) dynamics, absorption improvement, dosage reduction of the preventive and/or therapeutic drug of the present invention, and/or (3) reduction of the side effects of the preventive and/or therapeutic drug of the present invention.

A concomitant medicine comprising the compounds of the present invention and other drug may be administered as a combination preparation in which both components are contained in a single formulation, or administered as separate formulations. The administration by separate formulations includes simultaneous administration and administration with some time intervals. In the case of the administration with some time intervals, the compound of the present invention can be administered first, followed by another drug or another drug can be administered first, followed by the compound of the present invention. The administration method of the respective drugs may be the same or different.

The dosage of the other drug can be properly selected, based on a dosage that has been clinically used. The compounding ratio of the compound of the present invention and the other drug can be properly selected 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. The other drug may be a combination of two or more kind of arbitrary drugs in a proper proportion. The other drug that complements and/or enhances the preventive and/or therapeutic efficacy of the compound of the present invention includes not only those that have already been discovered, but those that will be discovered in future, based on the above mechanism.

Diseases on which this concomitant use exerts a preventive and/or therapeutic effect are not particularly limited. The concomitant medicine can be used for any diseases, as long as it complements and/or enhances the preventive and/or therapeutic efficacy of the compound of the present invention.

The compound of the present invention can be used with an existing chemotherapeutic concomitantly or in a mixture form. 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, it can be used 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.

In one embodiment, the compound(s) of the present invention can be used with other immunomodulators and/or a potentiating agent concomitantly or in a mixture form.

Examples of the immunomodulator include various cytokines, vaccines and adjuvants. Examples of these cytokines, vaccines and adjuvants that stimulates immune responses include but not limited to 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 another embodiment, the potentiating agents includes cyclophosphamide and analogs of cyclophosphamide, anti-TGF and Imatinib (Gleevac), 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.

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in art to which the subject matter herein belongs. As used herein, the following definitions are supplied in order to facilitate the understanding of the present invention.

As used herein, the term 'compound(s)' refers to the compounds disclosed in the present invention.

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 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 "optionally substituted" refers to the replacement of one or more hydrogen radicals in a given structure with the radical of a specified substituent including, but not limited to: alkyl, alkoxy, acyl, halo, and hydroxyl. It is understood that the substituent may be further substituted.

As used herein the term "alkyl" refers to a hydrocarbon chain radical that includes solely carbon and hydrogen atoms in the backbone, containing no unsaturation, having from one to twenty carbon atoms (i.e., Ci- 20 alkyl) or one to ten carbon atoms (i.e. , Ci-io alkyl) or one to five carbon atoms (i.e., C 1 -5 alkyl) and which is attached to the rest of the molecule by a single bond, e.g., including but not limited to methyl, ethyl, propyl, butyl, isobutyl, sec-butyl, tert-butyl, isopentyl or neopentyl. Unless set forth or recited to the contrary, all alkyl groups described or claimed herein may be straight chain or branched, substituted or unsubstituted.

As used herein, the term "acyl" refers to RC(O)-, wherein R is alkyl as defined above. Examples of acyl group include, but are not limited to acetyl, -C(0)(CH 2 ) 4 CH 3, - C(0)(CH 2 ) 6 CH 3 and -C(0)(CH 2 ) 8 CH 3 .

As used herein, the term "amidated C-terminus" refers to that the C-terminal of the amino acid in amide form.

As used herein, the term "amide form" refers to primary, secondary and/or tertiary amides and may be represented by the formula -C(0)NR x R y , wherein each of R x and R y independently represents hydrogen or alkyl.

As used herein, the term "amino" refers to -NH 2 group. Unless set forth or recited to the contrary, all amino groups described or claimed herein may be substituted or unsubstituted.

As used herein, the term "amino acid" refers to amino acids having L or D stereochemistry at the alpha carbon. Optional substituent on amino acid means replacement of one or more hydrogen radicals in a given structure with the radical of a specified substituent, in case of amino acid containing hydroxyl group such as Serine or Threonine, the hydroxyl group can be substituted with the specified substituent.

As used herein, the term "aryl" refers to C4-C 10 carbocyclic aromatic system containing one or two rings wherein such rings may be fused. Examples of aryl groups include, but are not limited to phenyl and naphthyl. As used herein, the term "arylalkyl" refers to an aryl group as defined above directly bonded to an alkyl group (e.g., benzyl and the like).

As used herein, the term "carboxylic acid" refers to -COOH group.

As used herein, the term "coupling agent" means a compound that reacts with the hydroxyl moiety of a carboxy moiety thereby rendering it susceptible to nucleophilic attack. Coupling agents of this type are known in the art and include, but are not limited to, EDCI, HATU, HOBt, DIC and DCC.

As used herein the term "ester" refers to (Q-Ce) linear or branched alkyl, (C 4 - Cio)aryl, (C 4 -Cio)heteroaryl or arylalkyl esters.

As used herein the term "esterified C-terminus" refers to that the C-terminal of the amino acid in ester form.

As used herein the term "free C-terminus' refers to that the C-terminal of the amino acid in -CO 2 H form.

As used herein, the terms "halogen" or "halo" includes fluorine, chlorine, bromine or iodine.

As used herein the term "Hydroxy" or "Hydroxyl" refers to -OH group.

"Pharmaceutically acceptable salt" is taken to mean an active ingredient, which comprises a compound of the formula (I) in the form of one of its salts, in particular if this salt form imparts improved pharmacokinetic properties on the active ingredient compared with the free form of the active ingredient or any other salt form of the active ingredient used earlier. The pharmaceutically acceptable salt form of the active ingredient can also provide this active ingredient for the first time with a desired pharmacokinetic property which it did not have earlier and can even have a positive influence on the pharmacodynamics of this active ingredient with respect to its therapeutic efficacy in the body.

"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 "stereoisomer/stereoisomers" refers to any enantiomers, diastereoisomers, or geometrical isomers of the compounds of formula (I), wherever they are chiral or when they bear one or more double bond. When the compounds of the formula (I) and related formulae 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 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).

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).

The phrase "therapeutically effective amount" or "efficient amount" refers to sufficient amount of the compound(s) of the present invention that (i) treats or prevents the particular disease, disorder or syndrome (ii) attenuates, ameliorates or eliminates one or more symptoms of the particular disease, disorder or syndrome or (iii) prevents or delays the onset of one or more symptoms of the particular disease, disorder or syndrome described herein. In the case of cancer, the therapeutically effective amount of the drug may decrease the number of cancer cells; decrease the cancer size; inhibit (i.e., slow to some extent and alternatively stop) cancer cell infiltration into peripheral organs; suppress (i.e., slow to some extent and alternatively stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer. In the case of infectious disease states, the therapeutic effective amount is an amount sufficient to decrease or alleviate an infectious diseases, the symptoms of an infections caused by bacterial, viral and fungal.

Naturally-occurring amino acids are identified throughout the specification by the conventional three-letter abbreviations indicated in the below table 2:

Table 2 (Amino acid codes) Name 3-letter code Name 3-letter code

Asparagine Asn Glutamine Gin

Aspartic acid Asp Serine Ser

Glutamic acid Glu Threonine Thr

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

°C (degree Celsius); δ (delta); % (percentage); brine (NaCl solution); CDI CH 2 C1 2 /DCM (Dichloromethane); DMF (Dimethyl formamide); DMSO (Dimethyl sulphoxide); DCC (Dicyclohexylcarbodiimide); DIC (N,N'-diisopropylcarbodiimide); DMSO-de (Deuterated DMSO); EDC.HC1/EDCI (l-(3-Dimethyl aminopropyl)-3- carbodiimide hydrochloride); Et 2 NH (Diethyl amine); EtOH (Ethanol); EtOAc (Ethyl acetate); ECF (ethylchloro formate); Fmoc (Fluorenylmethyloxycarbonyl chloride); g or gr (gram); H or H 2 (Hydrogen); H 2 0 (Water); HOBt/HOBT (1 -Hydroxy benzotriazole); HC1 (Hydrochloric acid); h or hr (Hours); HATU (2-(lH-7-Azabenzotriazol-l-yl)- 1,1 ,3,3-tetramethyl uranium hexafluoro phosphate methanaminium); Hz (Hertz); HPLC (High-performance liquid chromatography); K 2 CO 3 (Potassium carbonate); LiOH (Lithium hydroxide); LCMS (Liquid chromatography mass spectroscopy); mmol (Millimoles); M (Molar); μΐ (Micro litre); mL (Millilitre); mg (Milligram); m (Multiplet); MHz (Megahertz); MS (ES) (Mass spectroscopy-electro spray); min. (Minutes); Na (Sodium); NaHCOs (Sodium bicarbonate); NaOAc (Sodium acetate); NMM (N-methyl morpholine); Na 2 S0 4 (Sodium sulphate); N 2 (Nitrogen); NMR (Nuclear magnetic resonance spectroscopy); NH 3 (Ammonia); NH 2 OH.HCI (Hydroxylamine hydrochloride; PD-L1 (Programmed death-ligand 1); PD-L2 (Programmed cell death 1 ligand 2); prep-HPLC/preparative HPLC (Preparative High-performance liquid chromatography); S (Singlet); 'Bu (tertiary butyl); TEA/Et 3 N (Triethyl amine); TFA (Trifluoroaceticacid); TFA A (Trifluroacetic anhydride); TLC (Thin Layer Chromatography); THF (Tetrahydrofuran); TIPS (Triisopropylsilane); TFA/CF 3 COOH (Trifluoroacetic acid); t (Triplet); tR = (Retention time); Trt (Triphenyl methane); etc. EXPERIMENTAL

An embodiment of the present invention provides the preparation of compounds of formula (I) according to the procedures of the following examples, 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.

Purification and characterization of compounds

Analytical HPLC method: Analytical HPLC was performed using on ZIC

HILIC 200 A° column (4.6 mm x 250 mm, 5 μπι), Flow rate: 1.0 mL / min. The elution conditions used are: Buffer A: 5 mmol ammonium acetate, Buffer B: Acetonitrile,

Equilibration of the column with 90 % buffer B and elution by a gradient of 90 % to 40

% buffer B during 30 min.

Preparative HPLC Method: Preparative HPLC was performed using on

SeQuant ZIC HILIC 200 A° column (10 mm x 250 mm, 5 μιη), Flow rate: 5.0 mL/min. The elution conditions used are: Buffer A: 5 mmol ammonium acetate (adjust to pH-4 with Acetic Acid), Buffer B: Acetonitrile, Equilibration of the column with 90 % buffer

B and elution by a gradient of 90 % to 40 % buffer B during 20 min..

LCMS was performed on API 2000 LC/MS/MS triple quad (Applied bio systems) with Agilent 1100 series HPLC with G1315 B DAD, using Mercury MS column or using Agilent LC/MSD VL single quad with Agilent 1100 series HPLC with

G1315 B DAD, using Mercury MS column or using Shimadzu LCMS 2020 single quad with Prominence UFLC system with SPD-20 A DAD.

Example 1: Synthesis of Compound 1

Step la:

Ethylchloroformate (1.5 g, 13.78 mniol) and N-Methylmorpholine ( 1.4 g, 13.78 mmol) were added to a solution of compound la (3 g, 11.48 mmol) in THE (30 mL) arid stirred at -20 °C. After 20 min. Liquid ammonia (0.77 g, 45.92 mmol) was added to the active mixed anhydride formed in- situ and stirred at 0-5 °C for 20 min. The completeness of the reaction was confirmed by TLC analysis. The reaction mixture was evaporated under reduced pressure and partitioned between water and ethyl acetate. Organic layer was washed with NaHCOs, citric acid, brine solution, dried over Na 2 S0 4 and evaporated under reduced pressure to get 2.9 g of compound lb (Yield: 96.3%). LCMS: 261.0 ( Vi+H ; .

Step lb:

1 b 1c

Trifluroacetic anhydride (9.7 g, 46.0 mmol) was added to a solution of compound lb (8 g, 30.7 mmol) in pyridine (24.3 g, 307.0 mmol) and stirred at room temperature for 3 h. The completeness of the reaction was confirmed by TLC analysis. The reaction mixture was evaporated under reduced pressure and partitioned between water and ethyl acetate. Organic layer was washed with NaHCO ? ,, citric acid, brine solution, dried over Na 2 -S04 and evaporated under reduced pressure to afford 7 g of compound lc (Yield: 94.0%). LCMS: 187.2 (M-¾u ) + .

Step lc:

1 c 1d

Hydroxylamine hydrochloride (3 g, 43.37 mmol) and potassium carbonate (6 g, 43.37 mmol) were added to a solution of compound lc (7 g, 28.91 mmol) in EtOH (70 m L) and stirred at 90 °C for 2 h. The completeness of the reaction was confirmed by TLC analysis. The reaction mixture was evaporated under reduced pressure and partitioned between water and ethyl acetate. Organic layer was washed with brine solution, dried over Na 2 S0 4 and evaporated under reduced pressure. The crude compound was purified by silica gel column chromatography (Eluent: 0-5% ethyl acetate in hexane) to get 4.2 g of compound Id (Yield: 52.8%). LCMS: 276.4 (M+H) + .

Step Id:

Deoxo-Fluor® (1.83 g, 8.3 mmol) was added to a solution of Fmoc-Asn(Trt)-OH (4.5 g, 7.5 mmol) in CH 2 Q 2 (50 mL) and stirred at 0 °C for 3 h. Then CH 2 CI 2 was evaporated and triturated with hexane, decanted and evaporated under vacuum to get the corresponding acid fluoride. NMM (1.17 g, 1 1.6 mmol) and compound Id (1.6 g, 5.8 mmol) in THF were added to the acid fluoride and stirred at room temperature for 12 h. Then THF was evaporated and sodium acetate (0.72 g, 8.7 mmol) was added followed by EtOH (50 mL). The reaction mixture was stirred at 90 °C for 2 h. The completeness of the reaction was confirmed by TLC analysis. The reaction mixture was evaporated under reduced pressure and partitioned between water and ethyl acetate. Organic layer was washed with NaHCOa, citric acid, brine solution, dried over Na 2 S0 4 and evaporated under reduced pressure, which was further purified by silica gel column chromatography (Eluent: 0-5% ethyl acetate in hexane) to afford 2.8 g of compound le (Yield: 44.4%). LCMS: 836.4 (M+Hf .

Step le:

Ph 3

To compound le (2.3 g, 2.7 mmol) in CH 2 CI 2 (10 mL) diethyiarnine (10 mL) was added and the reaction mixture was stirred at room temperature for 30 min. The resulting solution was concentrated in vacuum to get gummy residue. The crude compound was purified by neutral alumina column chromatography (Eluent: 0-50% ethyl acetate in hexane then 0-5% methanol in chloroform) to get 1.4 g of If (Yield: 90 %). LCMS: 636.5 (M+Na) + .

1f 1

To a solution of compound If (0.45 g) in CH 2 CI 2 (5 mL), trifluoroacetic acid (5 mL) and catalytic amount of triisopropylsilane were added and stirred for 3 h at room temperature to remove the acid sensitive protecting groups. The resulting solution was concentrated in vacuum to afford 0.29 g of crude compound 1 which was purified using prep-HPLC method described under experimental conditions. \H NMR (DMSQ-d 6 , 400 MHz): δ 2.58 (m, 2H), 3.53 (m, 3H), 3.91 (t, 1H), 4.36 (t, 1H), 6.91 (s, 1H), 7.45 (s, 1H); 1 C NMR (DMSO-de, 400 MHz): δ 20.85, 45.71 , 50.23, 65.55, 171.03, 171 .41, 181.66. LCMS: 216.2 (Μ+ΗΓ; HPLC: t R = 13.1 min.

Example 2: Synthesis of Co

Step 2a:

1f

2a

The urea linkage was carried out by the coupling compound If (2.7 g, 4.39 mmoi) in THF (30 mL) at room temperature with compound 2b (1.67 g, 4.39 mmoi). The coupling was initiated by the addition of TEA (0.9 g, 8.78 mmoi) in THF (10 m L) and the resultant mixture was stirred at room temperature. After completion of 20 h, THF was evaporated from the reaction mass, and partitioned between water and ethyl acetate. Organic layer was washed with water, brine, dried over Na 2 S0 4 and evaporated under reduced pressure to get compound 2a, which was further purified by silica gel column chromatography (Fluent: 0-50% ethyl acetate in hexane) to afford 3.46 g of compound 2a (Yield: 92.10%). LCMS 857.4 (M+H) + .

2a

To a solution of compound 2a (0.22 g, 0.25 mmol) in 0¾ί¾ (5 m L), trifluoroaeetic acid (5 mL) and catalytic amount of triisopropyisilane were added and stirred for 3h at room, temperature. The resulting solution was concentrated under reduced pressure to obtain 0.35 g of crude compound. The crude solid material was purified using preparative- HPLC method described under experimental conditions. LCMS: 347.1 (M+H) + ; HPLC: t R = 12.9 min.

Synthesis of

2b

To the compound H-Ser( t Bu)-O i Bu (2 g, 9.2 mmol) in C I I ■(. ' {■ (20 mL), triethylamine (1.39 g, 13.8 mmol) was added and the solution was stirred at room temperature for 5-10 min. To this mixture, solution of 4-Nitrophenyl chioro formate (2.22 g, 11.04 mmol) in CH 2 CI 2 was added and the resultant mixture was stirred at room temperature for 30 min. The completion of the reaction was confirmed by TLC analysis. After completion of reaction, reaction mixture was diluted with CH 2 CI 2 and washed with water and 5.0 M citric acid solution, dried over Na 2 SC> 4 and evaporated under reduced pressure to get crude compound 2b, which was further purified by silica gel column chromatography (Eiuent: 0-20% ethyl acetate in hexane) to yield 2.1 g (58.9%) of 2b.

Example 3: Synthesis of Compound 3

The compound was synthesised using similar procedure as depicted in Example 1 (compound 1) and D-amino acids are linked up in reverse order. Boc-D-Thr('Bu)-OH was used in place of Boc-Ser('Bu)-OH (compound la, Example 1) and Fmoc-D- Asn(trt)-OH in place of Fmoc-Asn(trt)-OH to yield 0.15 g crude material of the title compound 3. LCMS: 230.1 (M+H) + .

Example 4: Synthesis of Co

The compound was synthesised using similar procedure as depicted in Example 2 for synthesising compound 2 using instead of H-Ser('Bu)-0'Bu (in synthesis of compound 2b) to yield 0.35 g crude material of the title compound. The crude solid material was purified using preparative HPLC described under experimental conditions. LCMS: 361.2 (M+H) + , HPLC: t R = 12.19 min.

Example 5: Synthesis of

The compound was synthesised using similar procedure as depicted in Example 4 (compound 4) using D-amino acids are linked up in reverse order. Boc-D-Thr('Bu)-OH was used in place of Boc-Ser('Bu)-OH, Fmoc-D-Asn(trt)-OH in place of Fmoc-Asn(trt)- OH and H-D-Ser('Bu)-0'Bu was used in place of H-Thr^Bu^O'Bu to yield 0.3 g crude material of the title compound. The cmde solid material was purified using preparative HPLC described under experimental conditions. LCMS: 361.3 (M+H) + . HPLC: t R = 13.58 min.

Example 6: Synthesis of Compound 6

The compound was synthesised using similar procedure as depicted in Example 2 by using H-Thr('Bu)-OMe instead of H-Ser('Bu)-0'Bu (in synthesis of compound 2b) to yield 0.2 g crude material of the title compound. The crude solid material was purified using preparative HPLC described under experimental conditions. LCMS: 375.1 (M+H) + , HPLC: t R = 1.84 min.

Example 7: Synthesis of Compound 7

Step 7a:

1f

7a

The compound 7a was synthesised using similar procedure as for compound 2a (Example 2, step 2a) using H-Thr('Bu)-OMe instead of H-Ser('Bu)-OtBu to get crude material which was further purified by silica gel column chromatography (Eluent: 0-50% ethyl acetate in he ane) to get 2.0 g of compound 7a (Yield: 74 %). LCMS: 829.2 (M+H) + .

Step 7b:

7a 7b

To a solution of compound 7a (0.35 g, 4.0 mmol) in THF (5 mL) was added lithium hydroxide (0.026 g, 0.63 mmol) at 0 °C and the mixture was stirred for 2 h at room temperature. The completion of the reaction was confirmed by TLC analysis. THF was evaporated from the reaction mass, and partitioned between water and ethyl acetate. Organic layer was washed with citric acid, brine solution, dried over Na 2 S0 4 and evaporated under reduced pressure to afford 7b, which was further purified by silica gel column chromatography (Eluent: 0-5% methanol in DCM) to get 0.3 g of product 7b (Yield: 86.7%). LCMS 815.2 (M+H) + .

Step 7c:

7b 7

Compound 7b (0.295 g, 0.39 mmol) was anchored to Rink amide resin (0.7 g, 0.55 mmol/g) using HOBT (0.072 g, 0.54 mmol) and DIC (0.068 g, 0.54 mmol) method in DMF (10 mL). The resin was stirred for 12 h at room temperature. The resin was washed with DCM, DMF and DCM and dried. The target compound was cleaved from the rink amide resin using TFA (5 mL) and catalytic amount of TIPS. The resin was allowed to remain at room temperature for 2 h with occasional stirring. After 2 h, TFA and TIPS were evaporated under nitrogen atmosphere and the resulting residue was washed with diethyl ether to yield 0.1 g crude material of the title compound 7. The crude solid material was purified using preparative HPLC described under experimental conditions. LCMS: 360.0 (M+H) + , HPLC: t R = 13.88 min.

Example 8: Synthesis of

The compound was synthesised using similar procedure as depicted in Example 2 (compound 2) using Fmoc-Glu(0'Bu)-OH instead of Fmoc-Asn(Trt)-OH to get 0.4 g crude material of the title compound. The crude solid material was purified using preparative HPLC described under experimental conditions. LCMS: 362.1 (M+H) + . HPLC: t R = 13.27 min.

The compounds in table 3 below were prepared based on the experimental procedures described above.

Table 3

Example 9: Rescue of mouse splenocyte proliferation in the presence of recombinant PD-L1/PD-L2

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 (HiMedia 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 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).

Protocol

Splenocyte preparation and culturing:

Splenocytes harvested in a 50 mL falcon tube by mashing mouse spleen in a 40μπι 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 + lmM sodium pyruvate + 10,000units/mL penicillin and 10,00C^g/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 (lxlO 5 cells cultured in high glucose DMEM medium) or recombinant human PDL- 1 (100 ng/mL) and test compounds. Splenocytes were stimulated with anti-mouse CD3 and anti- mouse CD28 antibody (^g/mL each), and the culture was further incubated for 72 h at 37 °C with 5% CO 2 . 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%.

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 4).

Table 4 Percent rescue of Percent rescue of splenocyte splenocyte

Compound Compound

proliferation proliferation No. No.

(@ 100 nM compound (@ 100 nM compound concentration) concentration)

1 93 14 75

2 50 15 83

4 89 16 72

5 67.6 17 55

6 84 18 64

7 55 19 88

8 67 20 69

9 34 21 47

10 49 22 55

11 90 23 74

12 64 24 52

13 74 25 91