US20160194295A1 - 1,3,4-Oxadiazole and 1,3,4-Thiadiazole Derivatives as Immunomodulators - Google Patents

1,3,4-Oxadiazole and 1,3,4-Thiadiazole Derivatives as Immunomodulators Download PDF

Info

Publication number
US20160194295A1
US20160194295A1 US14/916,290 US201414916290A US2016194295A1 US 20160194295 A1 US20160194295 A1 US 20160194295A1 US 201414916290 A US201414916290 A US 201414916290A US 2016194295 A1 US2016194295 A1 US 2016194295A1
Authority
US
United States
Prior art keywords
cancer
asn
amino acid
compound
carcinoma
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US14/916,290
Inventor
Pottayil Govindan Nair Sasikumar
Muralidhara Ramachandra
Seetharamaiah Setty Sudarshan Naremaddepalli
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Aurigene Oncology Ltd
Original Assignee
Aurigene Discovery Technologies Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Aurigene Discovery Technologies Ltd filed Critical Aurigene Discovery Technologies Ltd
Assigned to AURIGENE DISCOVERY TECHNOLOGIES LIMITED reassignment AURIGENE DISCOVERY TECHNOLOGIES LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: NAREMADDEPALLI, SEETHARAMAIAH SETTY SUDARSHAN, RAMACHANDRA, MURALIDHARA, SASIKUMAR, POTTAYIL GOVINDAN NAIR
Publication of US20160194295A1 publication Critical patent/US20160194295A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D285/00Heterocyclic compounds containing rings having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by groups C07D275/00 - C07D283/00
    • C07D285/01Five-membered rings
    • C07D285/02Thiadiazoles; Hydrogenated thiadiazoles
    • C07D285/04Thiadiazoles; Hydrogenated thiadiazoles not condensed with other rings
    • C07D285/121,3,4-Thiadiazoles; Hydrogenated 1,3,4-thiadiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4245Oxadiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/433Thidiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D271/00Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms
    • C07D271/02Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms not condensed with other rings
    • C07D271/101,3,4-Oxadiazoles; Hydrogenated 1,3,4-oxadiazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Definitions

  • the present invention relates to 1,3,4-oxadiazole and 1,3,4-thiadiazole compounds therapeutically useful as immune modulators.
  • the invention also relates to pharmaceutical compositions comprising the said 1,3,4-oxadiazole and 1,3,4-thiadiazole compounds as therapeutic agents.
  • PD-1 Programmed cell death-1
  • PD-L1 or PD-L2 are members 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).
  • PD-1 programmed cell death protein-1
  • Several PD-1 pathway inhibitors have shown robust activity in various phases of clinical trials (R D Harvey, Clinical Pharmacology & Therapeutics (2014); 96 2, 214-223).
  • PD-1 Programmed death-1
  • PD-L1 or PD-L2 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.
  • 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.
  • the present invention provides 1,3,4-oxadiazole and 1,3,4-thiadiazole compounds which are capable of suppressing and/or inhibiting the programmed cell death 1 (PD1) signalling pathway.
  • PD1 programmed cell death 1
  • 1,3,4-oxadiazole and 1,3,4-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) signalling pathway.
  • PD1 programmed cell death 1
  • the present invention provides a 1,3,4-oxadiazole and 1,3,4-thiadiazole compounds of formula (I):
  • R 1 is side chain of an amino acid selected from Ser, Thr, Phe, Ala or Asn;
  • X is S or O
  • R 2 is hydrogen or —CO-Aaa
  • Aaa is an amino acid residue selected from Ser, Asn or Thr; 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 Ser, Ala, Glu, Gln, Asn or Asp;
  • R 4 and R 5 independently are hydrogen or absent
  • composition comprising a compound of formula (I) or a pharmaceutically acceptable salt or a stereoisomer and processes for preparing thereof.
  • PD1 programmed cell death 1
  • the present invention provides 1,3,4-oxadiazole and 1,3,4-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.
  • the present invention relates to compounds of formula (I)
  • R 1 is side chain of an amino acid selected from Ser, Thr, Phe, Ala or Asn;
  • X is S or O
  • R 2 is hydrogen or —CO-Aaa
  • Aaa is an amino acid residue selected from Ser, Asn or Thr; 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 Ser, Ala, Glu, Gln, Asn or Asp;
  • R 4 and R 5 independently are hydrogen or absent
  • the present invention provides compounds of formula (IA)
  • R 1 is side chain of an amino acid selected from Ser, Thr, Phe, Ala or Asn;
  • X is S or O
  • R 2 is hydrogen or —CO-Aaa
  • R 3 is side chain of an amino acid selected from Ser, Ala, Glu, Gln, Asn or Asp;
  • Aaa is an amino acid residue selected from Ser, Asn or Thr; wherein a C-terminus thereof is a free terminus, is amidated or is esterified.
  • the present invention provides compounds of formula (IB)
  • R 1 is side chain of an amino acid selected from Ser, Thr, Phe, Ala or Asn;
  • R 3 is side chain of an amino acid selected from Ser, Ala, Glu, Gln, Asn or Asp;
  • Aaa is an amino acid residue selected from Ser, Asn or Thr; wherein a C-terminus thereof is a free terminus, is amidated or is esterified.
  • the present invention provides compounds of formula (IC)
  • R 1 is side chain of an amino acid selected from Ser, Thr, Phe, Ala or Asn;
  • R 3 is side chain of an amino acid selected from Ser, Ala, Glu, Gln, Asn or Asp;
  • Aaa is an amino acid residue selected from Ser, Asn or Thr; wherein a C-terminus thereof is a free terminus, is amidated or is esterified.
  • the present invention provides compounds of formula (I), wherein,
  • R 1 is side chain of Ser or Thr
  • R 2 is —CO-Aaa
  • Aaa is an amino acid residue Ser or Thr; wherein the C-terminus is free;
  • R 3 is side chain of Asn, Gln, Glu or Asp.
  • the compounds as disclosed in the present invention are formulated for pharmaceutical administration.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising the compound as disclosed, and a pharmaceutically acceptable carrier or a diluent.
  • the said pharmaceutical composition further comprising at least one of an anticancer agent, chemotherapy agent, or antiproliferative compound.
  • the present invention provides the compounds as disclosed in the present invention for use as a medicament.
  • the present invention provides the compounds as disclosed in the present invention for use as a medicament for the treatment of cancer or infectious disease.
  • the present invention provides the compounds as disclosed in the present invention for use as a medicament for the treatment 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 lymph
  • the present invention provides the compounds as disclosed in the present invention for use in the treatment of cancer.
  • the present invention provides the compounds as disclosed in the present invention for use in the treatment of infectious disease.
  • the present invention provides the compounds as disclosed in the present invention for use as a medicament for the treatment of bacterial infectious disease, a viral infectious disease or a fungal infectious disease.
  • 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.
  • 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 therapeutically effective amount of the compound of the present invention such that the immune response in the subject is modulated.
  • 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.
  • PD1 programmed cell death 1
  • 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
  • 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.
  • PD1 programmed cell death 1
  • 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) signalling pathway such that the subject is treated for the bacterial, viral and fungal infections.
  • PD1 programmed cell death 1
  • 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, men
  • 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, Naegleria fowleri, Acanthamoeba sp., Giardia lambia, Cryptosporidium sp
  • 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.
  • compositions are usually administered by oral or inhalation routes, but can be administered by parenteral administration route.
  • compositions can be administered, for example, by orally, intravenous infusion, topically, intraperitoneally, intravesically or intrathecally.
  • 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.
  • 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.
  • 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.
  • 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(s) of the present invention can be used with an existing chemotherapeutic concomitantly or in a mixture form.
  • 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.
  • 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.
  • the compound(s) of the present invention can be used with other immunomodulators and/or a potentiating agent concomitantly or in a mixture form.
  • the immunomodulator include various cytokines, vaccines and adjuvants.
  • these cytokines, vaccines and adjuvants that stimulates immune responses include but not limited to GM-C SF, M-CSF, G-CSF, interferon- ⁇ , ⁇ , or ⁇ , IL-1, IL-2, IL-3, IL-12, Poly (I:C) and C p G.
  • 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.
  • a mitosis inhibitor such as paclitaxel, Sunitinib (Sutent) or other antiangiogenic agents
  • an aromatase inhibitor such as letrozole
  • A2a adenosine receptor (A2AR) antagonist an angiogenesis inhibitor
  • anthracyclines oxaliplatin
  • doxorubicin TLR4 antagonists
  • 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.
  • amino acid refers to amino acids having L or D stereochemistry at the alpha carbon.
  • “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.
  • stereoisomer 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.
  • 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.
  • diastereomers are formed from the mixture by reaction with an optically active resolving agent.
  • 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.
  • optically active resolving agent for example dinitrobenzoylphenylglycine, cellulose triacetate or other derivatives of carbohydrates or chirally derivatised methacrylate polymers immobilised on silica gel.
  • 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).
  • domestic animals e.g., household pets including cats and dogs
  • non-domestic animals such as wildlife.
  • “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.
  • 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.
  • 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.
  • 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.
  • Analytical HPLC was performed using on ZIC HILIC 200 A° column (4.6 mm ⁇ 250 mm, 5 ⁇ m), 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 was performed using on SeQuant ZIC HILIC 200 A° column (10 mm ⁇ 250 mm, 5 ⁇ m), 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 AP1 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.
  • Fmoc group was deprotected by the addition of diethylamine (20.0 mL) to a solution of compound 1f (0.8 g, 1.45 mmol) in CH 2 Cl 2 (20.0 mL) at 0. The reaction was stirred at room temperature for 2 h. The resulting solution was concentrated in vacuum to get a thick gummy residue. The crude compound was purified by neutral alumina column chromatography (eluent: 2% methanol in chloroform) to afford 0.38 g of compound 1g (Yield: 80.0%): LCMS: 329.4 (M+H) + .
  • Lawesson's reagent (2.85 g, 7.03 mmol) was added to a solution of compound 2e (4 g, 4.68 mmol) in THF (40 mL) and stirred at 75° C. for 4 h. The completeness of the reaction was confirmed by TLC analysis. The reaction mixture was evaporated under reduced pressure and the obtained residue was partitioned between ice water and ethyl acetate. The organic layer was washed with NaHCO 3 solution followed brine solution.
  • Fmoc group on compound 3a was deprotected by adding diethylamine (3.8 mL) to the solution of compound 3a (1 g, 1.17 mmol) in CH 2 Cl 2 (3.8 mL). The reaction mixture was stirred at room temperature for 30 min. The resulting solution was concentrated in vacuum to get a thick 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 attain 0.62 g of compound 3b. LCMS: 630.5 (M+H) + .
  • the urea linkage was carried out by coupling of compound 3b (0.5 g, 7.9 mmol) in THF (10 m L) at room temperature with compound 4e (0.34 g, 7.9 mmol). The coupling was initiated by the addition of TEA (0.16 g, 15.8 mmol) in THF (10 m L) and the resultant mixture was stirred at room temperature. After 12 h, THF was evaporated from the reaction mass, and partitioned between water and ethyl acetate.
  • mice PD-L1 Recombinant mouse PD-L1 (rm-PDL-1, cat no: 1019-B7-100 and R&D Systems) were used as the source of PD-L1.
  • Working concentrations were titrated from 10 ⁇ M to 1 ⁇ M. (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-L1 cat no: 1019-B7-100).
  • Splenocytes harvested in a 50 mL falcon tube by mashing mouse spleen in a 40 ⁇ m 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 1 ⁇ PBS 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 800 ⁇ g 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 1 ⁇ PBS 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+1 mM sodium pyruvate+10,000 units/ml penicillin and 10,000 ⁇ g/ml streptomycin) and maintained in a CO 2 incubator with 5% CO 2 at 37° C.
  • RPMI complete media RPMI+10% fetal bovine serum+1 mM sodium pyruvate+10,000 units/ml penicillin and 10,000 ⁇ g/ml streptomycin
  • CFSE is a dye that passively diffuses into cells and binds to intracellular proteins. 1 ⁇ 10 6 cells/mL of harvested splenocytes were treated with 5 ⁇ M of CFSE in pre-warmed 1 ⁇ PBS/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 1 ⁇ 10 5 splenocytes added to wells containing either MDA-MB231 cells (1 ⁇ 10 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 (1 ⁇ 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 analyzed by flow cytometry with 488 nm excitation and 521 nm emission filters.
  • Percent splenocyte proliferation was analyzed 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 concentration of compound to anti-CD3/CD28 stimulated splenocytes in presence of ligand (PDL-1)

Abstract

The present invention relates to 1,3,4-oxadiazole and 1,3,4-thiadiazole compounds as therapeutic agents capable of inhibiting the programmed cell death 1 (PD1) signalling pathway. The invention also refers to derivatives of the therapeutic agents. The invention also encompasses the use of the said therapeutic agents and 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.

Description

  • This application claims the benefit of Indian provisional application number 4012/CHE/2013, filed on Sep. 6, 2013; which hereby incorporated by reference.
  • TECHNICAL FIELD
  • The present invention relates to 1,3,4-oxadiazole and 1,3,4-thiadiazole compounds therapeutically useful as immune modulators. The invention also relates to pharmaceutical compositions comprising the said 1,3,4-oxadiazole and 1,3,4-thiadiazole compounds 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 clinical trials (R D 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 U.S. Ser. No. 08/735,553 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 1,3,4-oxadiazole and 1,3,4-thiadiazole compounds which are capable of suppressing and/or inhibiting the programmed cell death 1 (PD1) signalling pathway.
  • SUMMARY OF INVENTION
  • In accordance with the present invention, 1,3,4-oxadiazole and 1,3,4-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) signalling pathway.
  • In one aspect, the present invention provides a 1,3,4-oxadiazole and 1,3,4-thiadiazole compounds of formula (I):
  • Figure US20160194295A1-20160707-C00001
  • wherein,
  • R1 is side chain of an amino acid selected from Ser, Thr, Phe, Ala or Asn;
  • X is S or O;
  • R2 is hydrogen or —CO-Aaa;
  • Aaa is an amino acid residue selected from Ser, Asn or Thr; wherein a C-terminus thereof is a free terminus, is amidated or is esterified;
  • R3 is side chain of an amino acid selected from Ser, Ala, Glu, Gln, Asn or Asp;
  • Figure US20160194295A1-20160707-P00001
    is an optional bond;
  • R4 and R5 independently are hydrogen or absent;
  • 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 a pharmaceutically acceptable salt or a stereoisomer and processes for preparing thereof.
  • In yet another aspect of the present invention, it provides use of 1,3,4-oxadiazole and 1,3,4-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 1,3,4-oxadiazole and 1,3,4-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)
  • Figure US20160194295A1-20160707-C00002
  • wherein,
  • R1 is side chain of an amino acid selected from Ser, Thr, Phe, Ala or Asn;
  • X is S or O;
  • R2 is hydrogen or —CO-Aaa;
  • Aaa is an amino acid residue selected from Ser, Asn or Thr; wherein a C-terminus thereof is a free terminus, is amidated or is esterified;
  • R3 is side chain of an amino acid selected from Ser, Ala, Glu, Gln, Asn or Asp;
  • Figure US20160194295A1-20160707-P00001
    is an optional bond;
  • R4 and R5 independently are hydrogen or absent;
  • or a pharmaceutically acceptable salt or a stereoisomer thereof.
  • In yet another embodiment, the present invention provides compounds of formula (IA)
  • Figure US20160194295A1-20160707-C00003
  • or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein,
  • R1 is side chain of an amino acid selected from Ser, Thr, Phe, Ala or Asn;
  • X is S or O;
  • R2 is hydrogen or —CO-Aaa;
  • R3 is side chain of an amino acid selected from Ser, Ala, Glu, Gln, Asn or Asp;
  • Aaa is an amino acid residue selected from Ser, Asn or Thr; wherein a C-terminus thereof is a free terminus, is amidated or is esterified.
  • In yet another further embodiment, the present invention provides compounds of formula (IB)
  • Figure US20160194295A1-20160707-C00004
  • or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein,
  • R1 is side chain of an amino acid selected from Ser, Thr, Phe, Ala or Asn;
  • R3 is side chain of an amino acid selected from Ser, Ala, Glu, Gln, Asn or Asp;
  • Aaa is an amino acid residue selected from Ser, Asn or Thr; wherein a C-terminus thereof is a free terminus, is amidated or is esterified.
  • In yet another further embodiment, the present invention provides compounds of formula (IC)
  • Figure US20160194295A1-20160707-C00005
  • or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein,
  • R1 is side chain of an amino acid selected from Ser, Thr, Phe, Ala or Asn;
  • R3 is side chain of an amino acid selected from Ser, Ala, Glu, Gln, Asn or Asp;
  • Aaa is an amino acid residue selected from Ser, Asn or Thr; wherein a C-terminus thereof is a free terminus, is amidated or is esterified.
  • In yet another further embodiment, the present invention provides compounds of formula (I), wherein,
  • R1 is side chain of Ser or Thr;
  • R2 is —CO-Aaa;
  • Aaa is an amino acid residue Ser or Thr; wherein the C-terminus is free;
  • R3 is side chain of Asn, Gln, Glu or Asp.
  • The embodiment 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) and (IA), in which X is O.
  • According to another embodiment, specifically provided are compounds of the formula (I) and (IA) in which X is S.
  • According to yet another embodiment, specifically provided are compounds of the formula (I) and (IA) in which R2 is hydrogen.
  • According to yet another embodiment, specifically provided are compounds of the formula (I) in which R4 and R5 are hydrogen.
  • 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 R2 is —CO-Ser.
  • According to yet another embodiment, specifically provided are compounds of the formula (I) in which R2 is —CO-Thr.
  • According to yet another embodiment, specifically provided are compounds of the formula (I), (IA), (IB) and (IC) in which R1 is side chain of Ser.
  • According to yet another embodiment, specifically provided are compounds of the formula (I), (IA), (IB) and (IC) in which R1 is side chain of Thr.
  • According to yet another embodiment, specifically provided are compounds of the formula (I), (IA) and (IC) in which R1 is side chain of Phe, Ala or Asn.
  • According to yet another embodiment, specifically provided are compounds of the formula (I), (IA), (IB) and (IC) in which R3 is side chain of Asn.
  • According to yet another embodiment, specifically provided are compounds of the formula (I), (IA) and (IB) in which R3 is side chain of Ser.
  • According to yet another embodiment, specifically provided are compounds of the formula (I), (IA) and (IC) in which R3 is side chain of Gln.
  • According to yet another embodiment, specifically provided are compounds of the formula (I), (IA) and (IC) in which R3 is side chain of Glu.
  • According to yet another embodiment, specifically provided are compounds of the formula (I), (IA) and (IC) in which R3 is side chain of Ala or Asp.
  • According to yet another embodiment, specifically provided are compounds of the formula (IB) and (IC) in which Aaa is Ser.
  • According to yet another embodiment, specifically provided are compounds of the formula (IC) in which Aaa is Thr.
  • According to yet another embodiment, specifically provided are compounds of the formula (I), (IA) and (TB) 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
    Compound
    No. Structure
     1.
    Figure US20160194295A1-20160707-C00006
     2.
    Figure US20160194295A1-20160707-C00007
     3.
    Figure US20160194295A1-20160707-C00008
     4.
    Figure US20160194295A1-20160707-C00009
     5.
    Figure US20160194295A1-20160707-C00010
     6.
    Figure US20160194295A1-20160707-C00011
     7.
    Figure US20160194295A1-20160707-C00012
     8.
    Figure US20160194295A1-20160707-C00013
     9.
    Figure US20160194295A1-20160707-C00014
    10.
    Figure US20160194295A1-20160707-C00015
    11.
    Figure US20160194295A1-20160707-C00016
    12.
    Figure US20160194295A1-20160707-C00017
    13.
    Figure US20160194295A1-20160707-C00018
    14.
    Figure US20160194295A1-20160707-C00019
    15.
    Figure US20160194295A1-20160707-C00020
    16.
    Figure US20160194295A1-20160707-C00021
    17.
    Figure US20160194295A1-20160707-C00022
    18.
    Figure US20160194295A1-20160707-C00023
    19.
    Figure US20160194295A1-20160707-C00024
    20.
    Figure US20160194295A1-20160707-C00025
  • or a pharmaceutically acceptable salt thereof or a stereoisomer thereof.
  • The compounds as disclosed in the present invention are formulated for pharmaceutical administration.
  • In one embodiment, the present invention provides a pharmaceutical composition comprising the compound as disclosed, and a pharmaceutically acceptable carrier or a diluent.
  • In another embodiment, the said pharmaceutical composition further comprising at least one of an anticancer agent, chemotherapy agent, or antiproliferative compound.
  • In one embodiment, the present invention provides the compounds as disclosed in the present invention for use as a medicament.
  • In another embodiment, the present invention provides the compounds as disclosed in the present invention for use as a medicament for the treatment of cancer or infectious disease.
  • In another embodiment, the present invention provides the compounds as disclosed in the present invention for use as a medicament for the treatment 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 another embodiment, the present invention provides the compounds as disclosed in the present invention for use in the treatment of cancer.
  • In another embodiment, the present invention provides the compounds as disclosed in the present invention for use in the treatment of infectious disease.
  • In one embodiment, the present invention provides the compounds as disclosed in the present invention for use as 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 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 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) signalling 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, Naegleria fowleri, 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(s) 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-C SF, M-CSF, G-CSF, interferon-α, β, or γ, IL-1, IL-2, IL-3, IL-12, Poly (I:C) and CpG.
  • In another embodiment, the potentiating agents includes cyclophosphamide and analogs of cyclophosphamide, anti-TGFβ 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 “amino” refers to —NH2 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.
  • “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” 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).
  • “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 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 Gln
    Aspartic acid Asp Phenylalanine Phe
    Alanine Ala Serine Ser
    Glutamic acid Glu Threonine Thr
  • The abbreviations used in the entire specification may be summarized hereinbelow with their particular meaning.
  • ° C. (degree Celsius); δ (delta); % (percentage); brine (NaCl solution); CH2Cl2/DCM (Dichloromethane); br s (Broad singlet); Cs2CO3 (Caesium carbonate); d (Doublet); DMF (Dimethyl formamide); DMSO (Dimethyl sulphoxide); DMSO-d6 (Deuterated DMSO); EDC.HCl/EDCI (1-(3-Dimethyl aminopropyl)-3-carbodiimide hydrochloride); Et2NH (Diethylamine); Fmoc (Fluorenylmethyloxycarbonyl chloride); g or gr (gram); H or H2 (Hydrogen); H2O (Water); HOBt/HOBT (1-Hydroxy benzotriazole); HCl (Hydrochloric acid); h or hr (Hours); Hz (Hertz); HPLC (High-performance liquid chromatography); I2 (Iodine); K2CO3 (Potassium carbonate); LCMS (Liquid chromatography mass spectroscopy); MeOH (Methanol); mmol (Millimoles); M (Molar); μl (Microlitre); mL (Millilitre); mg (Milligram); m (Multiplet); MHz (Megahertz); MS (ES) (Mass spectroscopy-electro spray); min. (Minutes); Na (Sodium); NaHCO3 (Sodium bicarbonate); NH2NH2.H2O (Hydrazine hydrate); NMM (N-methyl morpholine); Na2SO4 (Sodium sulphate); N2 (Nitrogen); NMR (Nuclear magnetic resonance spectroscopy); 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); tBu (tertiary butyl); TEA/Et3N (Triethyl amine); TLC (Thin Layer Chromatography); THF (Tetrahydrofuran); TIPS (Triisopropylsilane); TFA/CF3COOH (Trifluoroacetic acid); t (Triplet); tR=(Retention time); TPP (Triphenylphosphine); 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×250 mm, 5 μm), 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×250 mm, 5 μm), 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 AP1 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 1a:
  • Figure US20160194295A1-20160707-C00026
  • Potassium carbonate (7.9 g, 57.39 mmol) and Methyl iodide (1.3 mL, 21.04 mmol) were added to a solution of compound 1a (5.0 g, 19.13 mmol) in DMF (35 mL) and stirred at room temperature for 2 h. The completeness of the reaction was confirmed by TLC analysis. The reaction mixture was partitioned between water and ethyl acetate. Organic layer was washed with water, brine, dried over Na2SO4 and evaporated under reduced pressure to get 5.0 g of compound 1b (Yield: 96.1%). LCMS: 176.1 (M-Boc)+.
  • Step 1b:
  • Figure US20160194295A1-20160707-C00027
  • Hydrazine hydrate (7.2 mL) was added to a solution of compound 1b (5.0 g, 18.16 mmol) in methanol (30 mL) and stirred at room temperature for 2 h. The completeness of the reaction was confirmed by TLC analysis. The reaction mixture was evaporated under reduced pressure, the residue obtained was partitioned between water and ethyl acetate. Organic layer was washed with water, brine, dried over Na2SO4 and evaporated under reduced pressure to get 4.0 g of compound 1c (Yield: 80.0%). LCMS: 276.3 (M+H)+.
  • Step 1c:
  • Figure US20160194295A1-20160707-C00028
  • NMM (0.67 ml, 6.52 mmol) was slowly added to a stirred solution of 1c (1.2 g, 4.35 mmol), 1d (1.43 g, 4.35 mmol), HOBt (0.7 g, 5.22 mmol) and EDC.HCl (0.99 g, 5.22 mmol) in DMF (15 mL) at 0. The reaction mixture was stirred at room temperature for 12 h. The completeness of the reaction was confirmed by TLC analysis. The reaction was quenched with ice and the solid precipitated was filtered and dried under vacuum to obtain 2.0 g of pure product 1e (Yield: 83.3%). LCMS: 591.5 (M+Na)+.
  • Step 1d:
  • Figure US20160194295A1-20160707-C00029
  • To a stirred solution of 1e (1.5 g, 2.63 mmol) in dry THF (15.0 mL) and DMF (5.0 mL) triphenylphosphine (1.38 g, 5.27 mmol) and iodine (1.33 g, 5.27 mmol) were added at 0° C. After the iodine was completely dissolved, Et3N (1.52 mL, 10.54 mmol) was added to this reaction mixture at ice cold temperature. Reaction mixture was allowed to attain room temperature and stirred for 4 h. The completeness of the reaction was confirmed by TLC analysis. The reaction was quenched with ice water and extracted with ethyl acetate. Organic layer was washed with saturated sodium thiosulphate and brine solution. The separated Organic layer was dried over Na2SO4 and evaporated under reduced pressure to get residue, which was further purified by silica gel column chromatography (eluent: 30% ethyl acetate in hexane) to afford 0.8 g of compound 1f (Yield: 55%). LCMS: 551.3 (M+H)+.
  • Step 1e:
  • Figure US20160194295A1-20160707-C00030
  • Fmoc group was deprotected by the addition of diethylamine (20.0 mL) to a solution of compound 1f (0.8 g, 1.45 mmol) in CH2Cl2 (20.0 mL) at 0. The reaction was stirred at room temperature for 2 h. The resulting solution was concentrated in vacuum to get a thick gummy residue. The crude compound was purified by neutral alumina column chromatography (eluent: 2% methanol in chloroform) to afford 0.38 g of compound 1g (Yield: 80.0%): LCMS: 329.4 (M+H)+.
  • Step 1f:
  • Figure US20160194295A1-20160707-C00031
  • Compound 1g (0.38 g, 1.16 mmol), TEA (0.33 mL, 2.32 mmol) dissolved in DMF (10 mL) were added drop wise to a solution of 1h (0.55 g, 1.39 mmol) at 0 for urea bond formation and the mixture was stirred at room temperature for 2 h. The completeness of the reaction was confirmed by TLC analysis. The reaction was quenched with ice water, the solid precipitated was filtered and dried under vacuum to get crude compound, which was further purified by silica gel column chromatography (eluent: 0-35% ethyl acetate in hexane) to get 0.4 g of product 1i (Yield: 59.7%). LCMS: 586.4 (M+H)+.
  • Step 1g:
  • Figure US20160194295A1-20160707-C00032
  • To a solution of compound 1i (0.4 g, 0.68 mmol) in CH2Cl2 (5 m L), trifluoroacetic acid (5 mL) and catalytic amount of triisopropylsilane were added and stirred at room temperature for 3 h to remove the acid sensitive protecting groups. The resulting solution was concentrated under nitrogen and the solid material was purified by preparative HPLC method as described under experimental conditions (Yield: 0.05 g). LCMS: 318.0 (M+H)+; HPLC: tR=10.96 min.
  • Synthesis of Compound 1h (NO2—C6H4—OCO-Thr(tBu)-OtBu)
  • Figure US20160194295A1-20160707-C00033
  • To a solution of 4-nitrophenylchloroformate (4.79 g, 23.77 mmol) in DCM (25.0 mL) was added a solution of H-Thr(tBu)-OtBu (5.0 g, 21.61 mmol) TEA (6.2 mL, 43.22 mmol) in CH2Cl2 (25 mL) slowly 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 1.0 M sodium carbonate solution. The organic layer was dried over Na2SO4 and evaporated under reduced pressure to afford crude compound 1h, which was further purified by silica gel column chromatography (eluent: 0-5% ethyl acetate in hexane) to get 3.0 g of product 1h. 1H NMR (CDCl3, 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).
  • Example 2 Synthesis of Compound 2 Step 2a:
  • Figure US20160194295A1-20160707-C00034
  • NMM (1.8 mL, 18.15 mmol) was slowly added to a stirred solution of 1c (2.0 g, 7.26 mmol), 2d (4.3 g, 7.26 mmol), HOBt (1.17 g, 8.7 mmol) and EDC.HCl (1.66 g, 8.7 mmol) in DMF (15 mL) at 0. The reaction mixture was stirred at room temperature for 12 h. The completeness of the reaction was confirmed by TLC analysis. The reaction was quenched with ice, the solid precipitated was filtered and dried under vacuum to afford 3.7 g of pure product 2e (Yield: 59.6%). LCMS: 854.4 (M+H)+.
  • Step 2b:
  • Figure US20160194295A1-20160707-C00035
  • To a stirred solution of 2e (3.7 g, 4.33 mmol) dissolved in dry THF (25.0 mL) and DMF (10.0 mL), triphenylphosphine (2.28 g, 8.66 mmol) and iodine (2.2 g, 8.66 mmol) were added at 0. After the iodine was completely dissolved, Et3N (2.5 mL, 17.32 mmol) was added at same temperature. The reaction mixture was stirred at room temperature for 4 h. The completeness of the reaction was confirmed by TLC analysis. The reaction was quenched with ice water and extracted with ethyl acetate. The organic layer was washed with saturated sodium thiosulphate and brine solution. The separated organic layer was dried over Na2SO4 and evaporated under reduced pressure, which was further purified by silica gel column chromatography (eluent: 30% ethyl acetate in hexane) to get 2.0 g of compound 2f (Yield: 55%). LCMS: 858.4 (M+Na)+.
  • Step 2c:
  • Figure US20160194295A1-20160707-C00036
  • Diethylamine (30.0 mL) was added to a solution of compound 2f (2.0 g, 1.17 mmol) in CH2Cl2 (30.0 mL) at 0. The reaction mixture was stirred at room temperature for 1 h. The resulting solution was concentrated in vacuum to get a thick gummy residue. The crude compound was purified by neutral alumina column chromatography (eluent: 2% methanol in chloroform) to afford 1.0 g of compound 2g (Yield: 71.4%). LCMS: 614.5 (M+H)+.
  • Step 2d:
  • Figure US20160194295A1-20160707-C00037
  • Compound 2g (1.0 g, 1.63 mmol) and TEA (0.47 mL, 3.2 mmol) dissolved in DMF (10 m L) were added drop wise to a solution of 111 (0.7 g, 1.79 mmol) at 0. The reaction mixture was then allowed to reach room temperature and continued the stirring for 2 h. The completeness of the reaction was confirmed by TLC analysis. The reaction was quenched with ice water, the solid precipitated was filtered and dried under vacuum. The crude compound obtained was further purified by silica gel column chromatography (eluent: 0-30% ethyl acetate in hexane) to get 0.8 g of product 2i (Yield: 57.1%). LCMS: 871.6 (M+H)+.
  • Step 2e:
  • Figure US20160194295A1-20160707-C00038
  • To a solution of compound 21 (0.8 g, 0.92 mmol) in CH2Cl2 (6 m L), trifluoroacetic acid (6 mL) and catalytic amount of triisopropylsilane were added and stirred at room temperature for 3 h. The resulting solution was concentrated under nitrogen and the solid material was purified by preparative HPLC method described under experimental conditions (Yield: 0.065 g). HPLC: tR=12.01 min.; LCMS: 361.34 (M+H)+.
  • Example 3 Synthesis of Compound 3 Step 3a:
  • Figure US20160194295A1-20160707-C00039
  • Lawesson's reagent (2.85 g, 7.03 mmol) was added to a solution of compound 2e (4 g, 4.68 mmol) in THF (40 mL) and stirred at 75° C. for 4 h. The completeness of the reaction was confirmed by TLC analysis. The reaction mixture was evaporated under reduced pressure and the obtained residue was partitioned between ice water and ethyl acetate. The organic layer was washed with NaHCO3 solution followed brine solution. The organic layer was dried over Na2SO4, filtered and evaporated under reduced pressure to get residue which was further purified by silica gel column chromatography (eluent: 0-5% ethyl acetate in hexane) to afford 2.7 g of compound 3a (Yield: 67.66%). LCMS: 852.3 (M+H)+,
  • Step 3b:
  • Figure US20160194295A1-20160707-C00040
  • Fmoc group on compound 3a was deprotected by adding diethylamine (3.8 mL) to the solution of compound 3a (1 g, 1.17 mmol) in CH2Cl2 (3.8 mL). The reaction mixture was stirred at room temperature for 30 min. The resulting solution was concentrated in vacuum to get a thick 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 attain 0.62 g of compound 3b. LCMS: 630.5 (M+H)+.
  • Step 3c:
  • Figure US20160194295A1-20160707-C00041
  • To a solution of compound 3b (0.6 g) in CH2Cl2 (7.5 mL), trifluoroacetic acid (2.5 mL) and catalytic amount of triisopropylsilane were added and stirred at room temperature for 3 h. The resulting solution was concentrated in vacuum to get 0.13 g of compound 3 which was purified by preparative HPLC method described under experimental conditions. LCMS: 232.3 (M+H)+.
  • Example 4 Synthesis of Compound 4 Step 4a:
  • Figure US20160194295A1-20160707-C00042
  • The urea linkage was carried out by coupling of compound 3b (0.5 g, 7.9 mmol) in THF (10 m L) at room temperature with compound 4e (0.34 g, 7.9 mmol). The coupling was initiated by the addition of TEA (0.16 g, 15.8 mmol) in THF (10 m L) and the resultant mixture was stirred at room temperature. After 12 h, THF was evaporated from the reaction mass, and partitioned between water and ethyl acetate. The organic layer was washed with water, brine, dried over Na2SO4 and evaporated under reduced pressure to yield 4a, which was further purified by silica gel column chromatography (eluent: 0-50% ethyl acetate in hexane) to get 0.45 g of product 4a (Yield: 61.64%). LCMS: 921.8 (M+H)+.
  • Step 4b:
  • Figure US20160194295A1-20160707-C00043
  • To a solution of compound 4a (0.55 g) in methanol (20 mL), was added 10% Pd—C (0.15 g) under inert atmosphere. The mixture was stirred for 1 h under H2 atmosphere. The completion of the reaction was confirmed by TLC analysis. The Pd—C catalyst was then removed by filtration through a Celite® pad and washed with 20 mL of methanol. The combined organic filtrate on evaporation under reduced pressure resulted in the isolation of the product 4b (Yield: 0.42 g, 85.71%). LCMS: 831.5 (M+H)+.
  • Step 4c:
  • Figure US20160194295A1-20160707-C00044
  • To a solution of compound 4b (0.2 g, 0.3 mmol) in CH2Cl2 (5 mL), trifluoroacetic acid (5 mL) and catalytic amount of triisopropylsilane were added and stirred at room temperature for 3 h. The resulting solution was concentrated in vacuum and the solid material was purified by preparative HPLC method described under experimental conditions (Yield: 0.065 g). HPLC: tR=14.1 min.; LCMS: 377.3 (M+H)+.
  • Synthesis of Compound 4e, (NO2—C6H4—OCO-Thr(OtBu)-Bzl,)
  • Figure US20160194295A1-20160707-C00045
  • To a solution of compound Fmoc-Thr(tBu)-OH (15 g, 37.73 mmol) in 100 mL of DMF, Cs2CO3 (14.75 g, 45.2 mmol) was added and the resulting mixture was cooled to 0. To the cooled reaction mixture benzyl bromide (7.74 g, 45.2 mmol) was added and the solution was stirred at ice cold temperature for 30 min and then at room temperature for 12 h. The reaction mixture was concentrated under reduced pressure and diluted with ethyl acetate. The organic layer was washed with water followed by brine solution and dried over Na2SO4. The filtered solution was concentrated and purified by silica gel column chromatography (eluent: 0-30% ethyl acetate in hexane) to get 18 g of 4c as white solid. LCMS: 433.1 (M-OtBu)+, 397.2 (M-OBzl)+.
  • Fmoc group on compound 4c (25 g, 51.3 mmol) was deprotected by adding diethylamine (100 mL) to compound 4d (25 g, 51.3 mmol) in CH2Cl2 (100 mL) for 1 h with stirring at room temperature. The resulting solution was concentrated in vacuum and the thick residue was purified by neutral alumina column chromatography (eluent: 0-50% ethyl acetate in hexane then 0-5% methanol in chloroform) to afford 10.6 g of compound 4d. LCMS: 266.5 (M+H)+.
  • Figure US20160194295A1-20160707-C00046
  • To a solution of compound 4d (1.5 g, 5.65 mmol) in CH2Cl2 (25 mL) was added TEA (1.14 g, 11.3 mmol) and the solution was stirred at room temperature for 5-10 min. To this mixture a solution of 4-nitrophenyl chloroformate (1.4 g, 6.78 mmol) in CH2Cl2 (10 mL) was added and the resultant mixture was stirred at room temperature for 12 h. 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 sodium bisulphate solution followed by 1.0 M sodium carbonate solution. The organic layer was dried over Na2SO4, filtered and evaporated under reduced pressure to yield crude compound 4e, which was further purified by silica gel column chromatography (eluent: 0-20% ethyl acetate in hexane) to yield 0.7 g of product 4e. 1H NMR (DMSO-d6, 300 MHz): δ 1.04 (s, 9H), 1.16 (d, 3H), 4.11 (m, 1H), 5.11 (m, 3H), 6.91 (d, 2H), 7.40 (m, 5H), 8.10 (d, 2H), 8.26 (hr s, 1H).
  • The compounds in table 3 below were prepared based on the experimental procedures described above.
  • TABLE 3
    Compound LCMS HPLC
    No. Structure (M + H)+ tR in min
     5.
    Figure US20160194295A1-20160707-C00047
    391.1 12.43
     6.
    Figure US20160194295A1-20160707-C00048
    377.1
     7.
    Figure US20160194295A1-20160707-C00049
    361.1 12.21
     8.
    Figure US20160194295A1-20160707-C00050
    230.1 12.95
     9.
    Figure US20160194295A1-20160707-C00051
    375.4 11.55
    10.
    Figure US20160194295A1-20160707-C00052
    361.2 11.91
    11.
    Figure US20160194295A1-20160707-C00053
    361.1 12.08
    12.
    Figure US20160194295A1-20160707-C00054
    375.2 11.5 
    13.
    Figure US20160194295A1-20160707-C00055
    389.1 11.10
    14.
    Figure US20160194295A1-20160707-C00056
    347.1 12.58
    15.
    Figure US20160194295A1-20160707-C00057
    376.1 12.20
    16.
    Figure US20160194295A1-20160707-C00058
    375.2 11.91
    17.
    Figure US20160194295A1-20160707-C00059
    361.2 12.34
    18.
    Figure US20160194295A1-20160707-C00060
    362.1 12.50
    19.
    Figure US20160194295A1-20160707-C00061
    348.1 12.83
    20.
    Figure US20160194295A1-20160707-C00062
    391.1
  • The compounds shown in below table 4, which can be prepared by following similar procedure as described above with suitable modification known to the one ordinary skilled in the art are also included in the scope of the present application.
  • TABLE 4
    Com-
    pound
    No. Structure
    21.
    Figure US20160194295A1-20160707-C00063
    22.
    Figure US20160194295A1-20160707-C00064
    23.
    Figure US20160194295A1-20160707-C00065
    24.
    Figure US20160194295A1-20160707-C00066
    25.
    Figure US20160194295A1-20160707-C00067
    26.
    Figure US20160194295A1-20160707-C00068
    27.
    Figure US20160194295A1-20160707-C00069
    28.
    Figure US20160194295A1-20160707-C00070
    29.
    Figure US20160194295A1-20160707-C00071
  • 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 and 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 (10000 unit/ml)-Streptomycin (10,000 μg/ml) Liquid (GIBCO, Cat #15140-122); MEM Sodium Pyruvate solution 100 mM (100×), 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 (1 mL) (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); 40 μM 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 μL of Dimethyl sulfoxide (DMSO C2H6SO, SIGMA-D-5879) and aliquoted in to tubes for further use. Working concentrations were titrated from 10 μM to 1 μM. (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-L1 cat no: 1019-B7-100).
  • Protocol Splenocyte Preparation and Culturing:
  • Splenocytes harvested in a 50 mL falcon tube by mashing mouse spleen in a 40 μm 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 1×PBS 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 800×g 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 1×PBS 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+1 mM sodium pyruvate+10,000 units/ml penicillin and 10,000 μg/ml streptomycin) and maintained in a CO2 incubator with 5% CO2 at 37° C.
  • CFSE Proliferation Assay:
  • CFSE is a dye that passively diffuses into cells and binds to intracellular proteins. 1×106 cells/mL of harvested splenocytes were treated with 5 μM of CFSE in pre-warmed 1×PBS/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 1×105 splenocytes added to wells containing either MDA-MB231 cells (1×105 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 (1 μg/mL each), and the culture was further incubated for 72 h at 37° C. with 5% CO2. Cells were harvested and washed thrice with ice cold FACS buffer and % proliferation was analyzed by flow cytometry with 488 nm excitation and 521 nm emission filters.
  • Data Compilation, Processing and Inference:
  • Percent splenocyte proliferation was analyzed 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 concentration of compound to anti-CD3/CD28 stimulated splenocytes in presence of ligand (PDL-1)
  • TABLE 5
    Compound Percent rescue of splenocyte proliferation
    No. (@100 nM compound concentration)
    1 61.2
    2 80.3
    3 48.4
    4 60
    9 74
    10 58
    12 92
    13 75
    14 53
    15 69
    16 56
    17 53
    18 68

Claims (21)

1: A compound of formula (I)
Figure US20160194295A1-20160707-C00072
wherein,
R1 is a side chain of an amino acid Ser, Thr, Phe, Ala or Asn;
X is S or O;
R2 is hydrogen or —CO-Aaa;
Aaa is an amino acid residue Ser, Asn or Thr; wherein a C-terminus thereof is a free terminus, is amidated or is esterified;
R3 is a side chain of an amino acid Ser, Ala, Glu, Gln, Asn or Asp;
Figure US20160194295A1-20160707-P00001
is an optional bond;
R4 and R5 independently are hydrogen or absent;
or a pharmaceutically acceptable salt or a stereoisomer thereof.
2: The compound according to claim 1 is a compound of formula (IA):
Figure US20160194295A1-20160707-C00073
or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein,
R1 is a side chain of an amino acid Ser, Thr, Phe, Ala or Asn;
X is S or O;
R2 is hydrogen or —CO-Aaa;
R3 is a side chain of an amino acid Ser, Ala, Glu, Gln, Asn or Asp;
Aaa is an amino acid residue Ser, Asn or Thr; wherein a C-terminus thereof is a free terminus, is amidated or is esterified.
3: The compound according to claim 1, is a compound of formula (IB):
Figure US20160194295A1-20160707-C00074
or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein,
R1 is a side chain of an amino acid Ser, Thr, Phe, Ala or Asn;
R3 is a side chain of an amino acid Ser, Ala, Glu, Gln, Asn or Asp;
Aaa is an amino acid residue Ser, Asn or Thr; wherein a C-terminus thereof is a free terminus, is amidated or is esterified.
4: The compound according to claim 1, is a compound of formula (IC):
Figure US20160194295A1-20160707-C00075
or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein,
R1 is a side chain of an amino acid Ser, Thr, Phe, Ala or Asn;
R3 is a side chain of an amino acid Ser, Ala, Glu, Gln, Asn or Asp;
Aaa is an amino acid residue Ser, Asn or Thr; wherein a C-terminus thereof is a free terminus, is amidated or is esterified.
5: The compound according to claim 1, wherein X is O.
6: The compound according to claim 1, wherein X is S.
7: The compound according to claim 1, wherein
R1 is a side chain of Ser or Thr;
R2 is —CO-Aaa;
Aaa is an amino acid residue Ser or Thr; wherein the C-terminus is free;
R3 is a side chain of Asn, Gln, Glu or Asp.
8: The compound according to claim 1, wherein R2 is hydrogen.
9: A compound according to claim 1 selected from the group consisting of
Com- pound No. Structure  1.
Figure US20160194295A1-20160707-C00076
 2.
Figure US20160194295A1-20160707-C00077
 3.
Figure US20160194295A1-20160707-C00078
 4.
Figure US20160194295A1-20160707-C00079
 5.
Figure US20160194295A1-20160707-C00080
 6.
Figure US20160194295A1-20160707-C00081
 7.
Figure US20160194295A1-20160707-C00082
 8.
Figure US20160194295A1-20160707-C00083
 9.
Figure US20160194295A1-20160707-C00084
10.
Figure US20160194295A1-20160707-C00085
11.
Figure US20160194295A1-20160707-C00086
12.
Figure US20160194295A1-20160707-C00087
13.
Figure US20160194295A1-20160707-C00088
14.
Figure US20160194295A1-20160707-C00089
15.
Figure US20160194295A1-20160707-C00090
16.
Figure US20160194295A1-20160707-C00091
17.
Figure US20160194295A1-20160707-C00092
18.
Figure US20160194295A1-20160707-C00093
19.
Figure US20160194295A1-20160707-C00094
20.
Figure US20160194295A1-20160707-C00095
21.
Figure US20160194295A1-20160707-C00096
22.
Figure US20160194295A1-20160707-C00097
23.
Figure US20160194295A1-20160707-C00098
24.
Figure US20160194295A1-20160707-C00099
25.
Figure US20160194295A1-20160707-C00100
26.
Figure US20160194295A1-20160707-C00101
27.
Figure US20160194295A1-20160707-C00102
28.
Figure US20160194295A1-20160707-C00103
29.
Figure US20160194295A1-20160707-C00104
or a pharmaceutically acceptable salt or a stereoisomer thereof.
10: A pharmaceutical composition comprising at least one compound according to claim 1 or a pharmaceutically acceptable salt or a stereoisomer thereof, and a pharmaceutically acceptable carrier or excipient.
11: The pharmaceutical composition according to claim 10, comprising at least one additional pharmaceutical agent wherein the said additional pharmaceutical agent is an anticancer agent, chemotherapy agent, or antiproliferative compound.
12. (canceled)
13: A method of treating a cancer or infectious disease in a subject, comprising administering to the subject therapeutically effective amount of the pharmaceutical composition according to claim 10.
14: The method according to claim 13, wherein the cancer is 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, or environmentally induced cancers including those induced by asbestos, or combinations of said cancers.
15: The method according to claim 13, wherein the infectious disease is a bacterial infectious disease, a viral infectious disease or a fungal infectious disease.
16: A method of modulating an immune response mediated by PD-1 signaling pathway in a subject, comprising administering to the subject therapeutically effective amount of a compound according to claim 1.
17: A method of inhibiting growth of tumour cells and/or metastasis in a subject, comprising administering to the subject a therapeutically effective amount of a compound according to claim 1.
18: The method of claim 15, wherein the tumour cells are of a cancer selected from the group consisting of breast cancer, colon cancer, lung cancer, melanoma, prostate cancer and renal cancer.
19: The method of claim 15, wherein the tumour cells comprise 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, or environmentally induced cancers including those induced by asbestos, or combinations of said cancers.
20: A method of treating an infectious disease in a subject comprising administering to the subject a therapeutically effective amount of compound according to claim 1.
21: A method of treating bacterial, viral and fungal infections in a subject comprising administering to the subject a therapeutically effective amount of a compound according to claim 1.
US14/916,290 2013-09-06 2014-09-05 1,3,4-Oxadiazole and 1,3,4-Thiadiazole Derivatives as Immunomodulators Abandoned US20160194295A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
IN4012/CHE/2013 2013-09-06
IN4012CH2013 2013-09-06
PCT/IB2014/064281 WO2015033301A1 (en) 2013-09-06 2014-09-05 1,3,4-oxadiazole and 1,3,4-thiadiazole derivatives as immunomodulators

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2014/064281 A-371-Of-International WO2015033301A1 (en) 2013-09-06 2014-09-05 1,3,4-oxadiazole and 1,3,4-thiadiazole derivatives as immunomodulators

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/296,292 Continuation US9776978B2 (en) 2013-09-06 2016-10-18 1,3,4-oxadiazole and 1,3,4-thiadiazole derivatives as immunomodulators

Publications (1)

Publication Number Publication Date
US20160194295A1 true US20160194295A1 (en) 2016-07-07

Family

ID=52627870

Family Applications (3)

Application Number Title Priority Date Filing Date
US14/916,290 Abandoned US20160194295A1 (en) 2013-09-06 2014-09-05 1,3,4-Oxadiazole and 1,3,4-Thiadiazole Derivatives as Immunomodulators
US15/296,292 Expired - Fee Related US9776978B2 (en) 2013-09-06 2016-10-18 1,3,4-oxadiazole and 1,3,4-thiadiazole derivatives as immunomodulators
US15/722,194 Expired - Fee Related US10160736B2 (en) 2013-09-06 2017-10-02 1,3,4-oxadiazole and 1,3,4-thiadiazole derivatives as immunomodulators

Family Applications After (2)

Application Number Title Priority Date Filing Date
US15/296,292 Expired - Fee Related US9776978B2 (en) 2013-09-06 2016-10-18 1,3,4-oxadiazole and 1,3,4-thiadiazole derivatives as immunomodulators
US15/722,194 Expired - Fee Related US10160736B2 (en) 2013-09-06 2017-10-02 1,3,4-oxadiazole and 1,3,4-thiadiazole derivatives as immunomodulators

Country Status (25)

Country Link
US (3) US20160194295A1 (en)
EP (2) EP3041828B1 (en)
JP (1) JP2016532711A (en)
KR (1) KR20160081898A (en)
CN (1) CN105849092A (en)
AU (1) AU2014316684A1 (en)
CA (1) CA2922655A1 (en)
CU (1) CU24345B1 (en)
CY (1) CY1120769T1 (en)
DK (1) DK3041828T3 (en)
EA (1) EA029661B1 (en)
ES (1) ES2682040T3 (en)
HR (1) HRP20181251T1 (en)
HU (1) HUE039014T2 (en)
IL (1) IL244313A0 (en)
LT (1) LT3041828T (en)
MX (1) MX2016002971A (en)
PH (1) PH12016500405B1 (en)
PL (1) PL3041828T3 (en)
PT (1) PT3041828T (en)
RS (1) RS57559B1 (en)
SG (2) SG10201800508SA (en)
SI (1) SI3041828T1 (en)
TR (1) TR201811077T4 (en)
WO (1) WO2015033301A1 (en)

Cited By (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018098352A2 (en) 2016-11-22 2018-05-31 Jun Oishi Targeting kras induced immune checkpoint expression
US10160736B2 (en) 2013-09-06 2018-12-25 Aurigene Discovery Technologies Limited 1,3,4-oxadiazole and 1,3,4-thiadiazole derivatives as immunomodulators
US10568870B2 (en) 2016-04-07 2020-02-25 Chemocentryx, Inc. Reducing tumor burden by administering CCR1 antagonists in combination with PD-1 inhibitors or PD-L1 inhibitors
US10744118B2 (en) 2012-12-07 2020-08-18 Chemocentryx, Inc. Diazole lactams
US11414433B2 (en) 2018-05-11 2022-08-16 Incyte Corporation Heterocyclic compounds as immunomodulators
US11535615B2 (en) 2015-12-22 2022-12-27 Incyte Corporation Heterocyclic compounds as immunomodulators
US11566026B2 (en) 2016-12-22 2023-01-31 Incyte Corporation Heterocyclic compounds as immunomodulators
US11572366B2 (en) 2015-11-19 2023-02-07 Incyte Corporation Heterocyclic compounds as immunomodulators
US11608337B2 (en) 2016-05-06 2023-03-21 Incyte Corporation Heterocyclic compounds as immunomodulators
US11613536B2 (en) 2016-08-29 2023-03-28 Incyte Corporation Heterocyclic compounds as immunomodulators
US11673883B2 (en) 2016-05-26 2023-06-13 Incyte Corporation Heterocyclic compounds as immunomodulators
TWI808955B (en) * 2016-12-22 2023-07-21 美商英塞特公司 Heterocyclic compounds as immunomodulators
US11718605B2 (en) 2016-07-14 2023-08-08 Incyte Corporation Heterocyclic compounds as immunomodulators
US11753406B2 (en) 2019-08-09 2023-09-12 Incyte Corporation Salts of a PD-1/PD-L1 inhibitor
US11760756B2 (en) 2020-11-06 2023-09-19 Incyte Corporation Crystalline form of a PD-1/PD-L1 inhibitor
US11780836B2 (en) 2020-11-06 2023-10-10 Incyte Corporation Process of preparing a PD-1/PD-L1 inhibitor
US11866434B2 (en) 2020-11-06 2024-01-09 Incyte Corporation Process for making a PD-1/PD-L1 inhibitor and salts and crystalline forms thereof
US11866451B2 (en) 2019-11-11 2024-01-09 Incyte Corporation Salts and crystalline forms of a PD-1/PD-L1 inhibitor
US11873309B2 (en) 2016-06-20 2024-01-16 Incyte Corporation Heterocyclic compounds as immunomodulators

Families Citing this family (149)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PL3363790T3 (en) 2013-09-06 2020-07-27 Aurigene Discovery Technologies Limited 1,2,4-oxadiazole derivatives as immunomodulators
SG10202108921VA (en) 2015-03-10 2021-09-29 Aurigene Discovery Tech Ltd 1,2,4-oxadiazole and thiadiazole compounds as immunomodulators
BR112017019306A2 (en) * 2015-03-10 2018-05-08 Aurigene Discovery Technologies Limited 1,3,4-oxadiazole and thiadiazole compounds as immunomodulators
CA2979163A1 (en) * 2015-03-10 2016-09-15 Aurigene Discovery Technologies Limited 3-substituted 1,3,4-oxadiazole and thiadiazole compounds as immunomodulators
MY197262A (en) 2015-07-27 2023-06-08 Chong Kun Dang Pharmaceutical Corp 1,3,4-oxadiazole sulfonamide derivative compounds as histone deacetylase 6 inhibitor, and the pharmaceutical composition comprising the same
JP6538266B2 (en) 2015-07-27 2019-07-03 チョン クン ダン ファーマシューティカル コーポレーション 1,3,4-oxadiazole sulfamide derivative compounds as histone deacetylase 6 inhibitors and pharmaceutical compositions containing the same (1, 3, 4-Oxadiazole Sulfamide Derivatives Compounds as Histone Deacetylase 6 Inhibitor , And the Pharmaceutical Composition Composing the same)
KR101799009B1 (en) 2015-07-27 2017-11-17 주식회사 종근당 1,3,4-Oxadiazole Amide Derivative Compounds as Histone Deacetylase 6 Inhibitor, and the Pharmaceutical Composition Comprising the same
BR112018002304B1 (en) 2015-08-04 2023-12-19 Chong Kun Dang Pharmaceutical Corp COMPOUNDS OF 1,3,4-OXADIAZOLE DERIVATIVE AS A HISTONE DEACETYLASE 6 INHIBITOR AND THE PHARMACEUTICAL COMPOSITION COMPRISING THE SAME
MA44909A (en) 2015-09-15 2018-07-25 Acerta Pharma Bv THERAPEUTIC ASSOCIATION OF A CD19 INHIBITOR AND A BTK INHIBITOR
NZ740809A (en) 2015-10-12 2019-04-26 Chong Kun Dang Pharmaceutical Corp Oxadiazole amine derivative compounds as histone deacetylase 6 inhibitor, and the pharmaceutical composition comprising the same
US20170107216A1 (en) 2015-10-19 2017-04-20 Incyte Corporation Heterocyclic compounds as immunomodulators
PL3402503T3 (en) 2016-01-13 2021-04-19 Acerta Pharma B.V. Therapeutic combinations of an antifolate and a btk inhibitor
KR102401963B1 (en) 2016-06-27 2022-05-25 케모센트릭스, 인크. Immunomodulatory compounds
CN107573332B (en) 2016-07-05 2022-04-19 广州再极医药科技有限公司 Aromatic acetylene or aromatic vinyl compound, intermediate thereof, preparation method, pharmaceutical composition and application
WO2018047143A1 (en) * 2016-09-12 2018-03-15 Aurigene Discovery Technologies Limited Vista signaling pathway inhibitory compounds useful as immunomodulators
WO2018047139A1 (en) * 2016-09-12 2018-03-15 Aurigene Discovery Technologies Limited Compounds as modulators of tigit signalling pathway
WO2018051255A1 (en) * 2016-09-14 2018-03-22 Aurigene Discovery Technologies Limited Cyclic substituted-1,3,4-oxadiazole and thiadiazole compounds as immunomodulators
EP3515453A1 (en) 2016-09-22 2019-07-31 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for reprograming immune environment in a subject in need thereof
AU2017342536A1 (en) 2016-10-14 2019-05-02 Precision Biosciences, Inc. Engineered meganucleases specific for recognition sequences in the Hepatitis B virus genome
TWI788307B (en) 2016-10-31 2023-01-01 美商艾歐凡斯生物治療公司 Engineered artificial antigen presenting cells for tumor infiltrating lymphocyte expansion
MD3558990T2 (en) 2016-12-22 2023-02-28 Incyte Corp Tetrahydro imidazo[4,5-c]pyridine derivatives as PD-L1 internalization inducers
ES2899402T3 (en) 2016-12-22 2022-03-11 Incyte Corp Pyridine derivatives as immunomodulators
WO2018129533A1 (en) 2017-01-09 2018-07-12 Shuttle Pharmaceuticals, Llc Selective histone deacetylase inhibitors for the treatment of human disease
MA47215A (en) 2017-01-09 2019-11-13 Bioxcel Therapeutics Inc PREDICTIVE AND DIAGNOSTIC PROCEDURES FOR PROSTATE CANCER
US11584733B2 (en) 2017-01-09 2023-02-21 Shuttle Pharmaceuticals, Inc. Selective histone deacetylase inhibitors for the treatment of human disease
CN108395443B (en) * 2017-02-04 2021-05-04 广州丹康医药生物有限公司 Cyclic compounds inhibiting programmed death receptor ligand 1 and uses thereof
TW201841896A (en) * 2017-04-26 2018-12-01 大陸商南京聖和藥業股份有限公司 Heterocyclic compound serving as pd-l1 inhibitor
CN108863963B (en) * 2017-05-08 2022-05-27 南京圣和药物研发有限公司 Heterocyclic compounds as PD-L1 inhibitors
CN109096219B (en) * 2017-06-20 2023-03-21 广州丹康医药生物有限公司 Novel anti-PD-L1 compound, application thereof and composition containing same
EP3658173A1 (en) 2017-07-25 2020-06-03 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for modulating monocytopoiesis
IL272258B (en) 2017-07-28 2022-08-01 Chemocentryx Inc Immunomodulator compounds
CN111225665B (en) 2017-08-08 2023-12-08 凯莫森特里克斯股份有限公司 Macrocyclic immunomodulators
WO2019061324A1 (en) 2017-09-29 2019-04-04 Curis Inc. Crystal forms of immunomodulators
SG11202003081WA (en) 2017-10-11 2020-05-28 Aurigene Discovery Tech Ltd Crystalline forms of 3-substituted 1,2,4-oxadiazole
CN109678796B (en) * 2017-10-19 2023-01-10 上海长森药业有限公司 PD-1/PD-L1 small molecule inhibitor and preparation method and application thereof
CN111655260A (en) 2017-10-26 2020-09-11 南方研究院 Oxadiazoles and thiadiazoles as TGF-beta inhibitors
CN111372584A (en) 2017-11-03 2020-07-03 奥瑞基尼探索技术有限公司 Dual inhibitors of the TIM-3 and PD-1 pathways
CA3081675A1 (en) 2017-11-06 2019-05-09 Aurigene Discovery Technologies Limited Conjoint therapies for immunomodulation
JP2021502066A (en) 2017-11-06 2021-01-28 ジェネンテック, インコーポレイテッド Cancer diagnosis and therapy
KR102492187B1 (en) 2017-12-20 2023-01-27 인스티튜트 오브 오가닉 케미스트리 앤드 바이오케미스트리 에이에스 씨알 브이.브이.아이. 3'3' cyclic dinucleotides with phosphonate linkages that activate STING adapter proteins
AU2018392212B9 (en) 2017-12-20 2021-03-18 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3' cyclic dinucleotides with phosphonate bond activating the STING adaptor protein
SG11202005962YA (en) 2017-12-29 2020-07-29 Guangzhou Maxinovel Pharmaceuticals Co Ltd Aromatic vinyl or aromatic ethyl derivative, preparation method therefor, intermediate, pharmaceutical composition, and application
US11407723B2 (en) 2018-01-09 2022-08-09 Shuttle Pharmaceuticals, Inc. Selective histone deacetylase inhibitors for the treatment of human disease
EP3755311A4 (en) 2018-02-22 2021-11-10 ChemoCentryx, Inc. Indane-amines as pd-l1 antagonists
CN111788204B (en) 2018-02-26 2023-05-05 吉利德科学公司 Substituted pyrrolizine compounds as inhibitors of HBV replication
US20210030703A1 (en) 2018-03-12 2021-02-04 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of caloric restriction mimetics for potentiating chemo-immunotherapy for the treatment of cancers
PE20211911A1 (en) 2018-03-30 2021-09-28 Incyte Corp HETEROCYCLIC COMPOUNDS AS IMMUNOMODULATORS
WO2019195181A1 (en) 2018-04-05 2019-10-10 Gilead Sciences, Inc. Antibodies and fragments thereof that bind hepatitis b virus protein x
TWI818007B (en) 2018-04-06 2023-10-11 捷克科學院有機化學與生物化學研究所 2'3'-cyclic dinucleotides
TW202005654A (en) 2018-04-06 2020-02-01 捷克科學院有機化學與生物化學研究所 2'2'-cyclic dinucleotides
KR20200140867A (en) 2018-04-06 2020-12-16 인스티튜트 오브 오가닉 케미스트리 앤드 바이오케미스트리 에이에스 씨알 브이.브이.아이. 3'3'-cyclic dinucleotide
TW201945388A (en) 2018-04-12 2019-12-01 美商精密生物科學公司 Optimized engineered meganucleases having specificity for a recognition sequence in the hepatitis B virus genome
US20190359645A1 (en) 2018-05-03 2019-11-28 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotides comprising carbocyclic nucleotide
WO2019232319A1 (en) 2018-05-31 2019-12-05 Peloton Therapeutics, Inc. Compositions and methods for inhibiting cd73
SG11202012446UA (en) 2018-06-23 2021-01-28 Genentech Inc Methods of treating lung cancer with a pd-1 axis binding antagonist, a platinum agent, and a topoisomerase ii inhibitor
TW202011991A (en) 2018-07-18 2020-04-01 美商建南德克公司 Methods of treating lung cancer with a pd-1 axis binding antagonist, an antimetabolite, and a platinum agent
KR102316234B1 (en) 2018-07-26 2021-10-22 주식회사 종근당 1,3,4-Oxadiazole Derivative Compounds as Histone Deacetylase 6 Inhibitor, and the Pharmaceutical Composition Comprising the same
WO2020028097A1 (en) 2018-08-01 2020-02-06 Gilead Sciences, Inc. Solid forms of (r)-11-(methoxymethyl)-12-(3-methoxypropoxy)-3,3-dimethyl-8-0x0-2,3,8,13b-tetrahydro-1h-pyrido[2,1-a]pyrrolo[1,2-c] phthalazine-7-c arboxylic acid
TW202031273A (en) 2018-08-31 2020-09-01 美商艾歐凡斯生物治療公司 Treatment of nsclc patients refractory for anti-pd-1 antibody
EP3847154A1 (en) 2018-09-03 2021-07-14 F. Hoffmann-La Roche AG Carboxamide and sulfonamide derivatives useful as tead modulators
WO2020048942A1 (en) 2018-09-04 2020-03-12 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for enhancing cytotoxic t lymphocyte-dependent immune responses
WO2020058372A1 (en) 2018-09-19 2020-03-26 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical composition for the treatment of cancers resistant to immune checkpoint therapy
CN112930114B (en) 2018-09-20 2023-10-03 艾欧凡斯生物治疗公司 Amplification of TIL from cryopreserved tumor samples
WO2020070053A1 (en) 2018-10-01 2020-04-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of inhibitors of stress granule formation for targeting the regulation of immune responses
CN111057069B (en) * 2018-10-16 2024-01-26 武汉光谷通用名药物研究院有限公司 Cyclic compound, application and composition thereof
CN111100086B (en) * 2018-10-25 2022-07-01 南京圣和药业股份有限公司 1,3, 4-oxadiazole-2-cyclobutyl compound and preparation method thereof
TW202024042A (en) * 2018-10-25 2020-07-01 大陸商南京聖和藥業股份有限公司 1,3,4-oxadiazole-2-cyclobutyl compounds, preparation method therefor and application thereof
WO2020092528A1 (en) 2018-10-31 2020-05-07 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds having hpk1 inhibitory activity
MX2021005047A (en) 2018-10-31 2021-09-08 Gilead Sciences Inc Substituted 6-azabenzimidazole compounds as hpk1 inhibitors.
JP2022513592A (en) 2018-11-02 2022-02-09 シャンハイ マキシノベル ファーマシューティカルズ カンパニー リミテッド Biphenyl compounds, their intermediates, manufacturing methods, pharmaceutical compositions and uses
WO2020109355A1 (en) 2018-11-28 2020-06-04 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and kit for assaying lytic potential of immune effector cells
US20220018835A1 (en) 2018-12-07 2022-01-20 INSERM (Institut National de la Santé et de la Recherche Médicale Use of cd26 and cd39 as new phenotypic markers for assessing maturation of foxp3+ t cells and uses thereof for diagnostic purposes
US20220047556A1 (en) 2018-12-17 2022-02-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of sulconazole as a furin inhibitor
BR112021013157A8 (en) 2019-01-03 2022-12-06 Inst Nat Sante Rech Med USES OF AN NRP-1 INHIBITOR, USE OF A COMBINATION, USE OF A MULTIESPECIFIC ANTIBODY, EX VIVO METHOD TO PREDICT, USE OF AN INHIBITOR, MULTIESPECIFIC ANTIBODY, MODIFIED CELL POPULATION, EX VIVO METHOD OF PRODUCTION AND USE OF A POPULATION OF T CELLS
JP2022518399A (en) 2019-01-14 2022-03-15 ジェネンテック, インコーポレイテッド How to Treat Cancer with PD-1 Axial Binding Antagonists and RNA Vaccines
SG11202107606VA (en) 2019-01-15 2021-08-30 Inst Nat Sante Rech Med Mutated interleukin-34 (il-34) polypeptides and uses thereof in therapy
WO2020169472A2 (en) 2019-02-18 2020-08-27 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods of inducing phenotypic changes in macrophages
CN113543851A (en) 2019-03-07 2021-10-22 捷克共和国有机化学与生物化学研究所 2'3' -cyclic dinucleotides and their prodrugs
EP3935066A1 (en) 2019-03-07 2022-01-12 Institute of Organic Chemistry and Biochemistry ASCR, V.V.I. 3'3'-cyclic dinucleotides and prodrugs thereof
US11766447B2 (en) 2019-03-07 2023-09-26 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3′3′-cyclic dinucleotide analogue comprising a cyclopentanyl modified nucleotide as sting modulator
CN109824621A (en) * 2019-03-28 2019-05-31 中国药科大学 Furodiazole and thiadiazole compound and its preparation method and application
WO2020201362A2 (en) 2019-04-02 2020-10-08 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods of predicting and preventing cancer in patients having premalignant lesions
US20220160692A1 (en) 2019-04-09 2022-05-26 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of sk2 inhibitors in combination with immune checkpoint blockade therapy for the treatment of cancer
WO2020212484A1 (en) 2019-04-17 2020-10-22 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and compositions for treatment of nlrp3 inflammasome mediated il-1beta dependent disorders
TW202210480A (en) 2019-04-17 2022-03-16 美商基利科學股份有限公司 Solid forms of a toll-like receptor modulator
TWI751517B (en) 2019-04-17 2022-01-01 美商基利科學股份有限公司 Solid forms of a toll-like receptor modulator
KR20220002967A (en) 2019-04-19 2022-01-07 제넨테크, 인크. Anti-MERTK antibodies and methods of use thereof
KR20220009420A (en) 2019-05-15 2022-01-24 케모센트릭스, 인크. Triaryl Compounds for Treatment of PD-L1 Disease
EP3972695A1 (en) 2019-05-23 2022-03-30 Gilead Sciences, Inc. Substituted exo-methylene-oxindoles which are hpk1/map4k1 inhibitors
CA3141531A1 (en) 2019-05-24 2020-12-03 Pfizer Inc. Combination therapies using cdk inhibitors
CA3143104A1 (en) 2019-06-12 2020-12-17 Technical University Of Denmark Dissacharide formulations for controlled drug release
AU2020294781A1 (en) 2019-06-20 2021-12-23 Chemocentryx, Inc. Compounds for treatment of PD-L1 diseases
CN114206338A (en) 2019-07-10 2022-03-18 凯莫森特里克斯股份有限公司 Indanes as PD-L1 inhibitors
EP4017476A1 (en) 2019-08-19 2022-06-29 Gilead Sciences, Inc. Pharmaceutical formulations of tenofovir alafenamide
EP4037773A1 (en) 2019-09-30 2022-08-10 Incyte Corporation Pyrido[3,2-d]pyrimidine compounds as immunomodulators
WO2021067181A1 (en) 2019-09-30 2021-04-08 Gilead Sciences, Inc. Hbv vaccines and methods treating hbv
CA3151322A1 (en) 2019-10-01 2021-04-08 Silverback Therapeutics, Inc. Combination therapy with immune stimulatory conjugates
EP3800201A1 (en) 2019-10-01 2021-04-07 INSERM (Institut National de la Santé et de la Recherche Médicale) Cd28h stimulation enhances nk cell killing activities
US20220363776A1 (en) 2019-10-04 2022-11-17 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical composition for the treatment of ovarian cancer, breast cancer or pancreatic cancer
PE20221764A1 (en) 2019-10-16 2022-11-11 Chemocentryx Inc HETEROARYL-BIPHENYL AMINES FOR THE TREATMENT OF PD-L1 DISEASES
CA3152329A1 (en) 2019-10-16 2021-04-22 Pingchen Fan Heteroaryl-biphenyl amides for the treatment of pd-l1 diseases
TW202130618A (en) 2019-11-13 2021-08-16 美商建南德克公司 Therapeutic compounds and methods of use
US20230031465A1 (en) 2019-12-06 2023-02-02 Precision Biosciences, Inc. Optimized engineered meganucleases having specificity for a recognition sequence in the hepatitis b virus genome
KR20220125278A (en) 2020-01-03 2022-09-14 상하이 한서 바이오메디컬 컴퍼니 리미티드 Biphenyl derivative inhibitor, preparation method and use thereof
BR112022012918A2 (en) 2020-01-07 2022-09-06 Univ Texas VARIANTS OF IMPROVED HUMAN METHYLTHIOADENOSINE/ADENOSINE EXHAUST ENZYME FOR CANCER THERAPY
JP2023512654A (en) 2020-01-31 2023-03-28 ジェネンテック, インコーポレイテッド Methods for Inducing Neoepitope-Specific T Cells Using PD-1 Axis Binding Antagonists and RNA Vaccines
JP2023518433A (en) 2020-03-20 2023-05-01 ギリアード サイエンシーズ, インコーポレイテッド Prodrugs of 4'-C-substituted-2-halo-2'-deoxyadenosine nucleosides and methods of making and using them
WO2021216920A1 (en) 2020-04-22 2021-10-28 Iovance Biotherapeutics, Inc. Systems and methods for coordinating manufacturing of cells for patient-specific immunotherapy
KR20230042222A (en) 2020-05-26 2023-03-28 인쎄름 (엥스띠뛰 나씨오날 드 라 쌍떼 에 드 라 흐쉐르슈 메디깔) Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-COV-2) Polypeptides and Uses Thereof for Vaccine Purposes
US11787775B2 (en) 2020-07-24 2023-10-17 Genentech, Inc. Therapeutic compounds and methods of use
WO2022052926A1 (en) 2020-09-09 2022-03-17 广州再极医药科技有限公司 Aromatic ethylene compound and preparation method therefor, and intermediate, pharmaceutical composition, and application thereof
TW202233671A (en) 2020-10-20 2022-09-01 美商建南德克公司 Peg-conjugated anti-mertk antibodies and methods of use
AU2021369590A1 (en) 2020-10-28 2023-06-22 Ikena Oncology, Inc. Combination of an ahr inhibitor with a pdx inhibitor or doxorubicine
WO2022093981A1 (en) 2020-10-28 2022-05-05 Genentech, Inc. Combination therapy comprising ptpn22 inhibitors and pd-l1 binding antagonists
CA3195572A1 (en) 2020-11-04 2022-05-12 Heidelberg Pharma Research Gmbh Composition comprising a combination of immune checkpoint inhibitor and antibody-amatoxin conjugate for use in cancer therapy
WO2022101302A1 (en) 2020-11-12 2022-05-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Antibodies conjugated or fused to the receptor-binding domain of the sars-cov-2 spike protein and uses thereof for vaccine purposes
WO2022101463A1 (en) 2020-11-16 2022-05-19 INSERM (Institut National de la Santé et de la Recherche Médicale) Use of the last c-terminal residues m31/41 of zikv m ectodomain for triggering apoptotic cell death
US20220168293A1 (en) 2020-12-02 2022-06-02 Pfizer Inc. Time to resolution of axitinib-related adverse events
WO2022119830A1 (en) 2020-12-02 2022-06-09 Genentech, Inc. Methods and compositions for neoadjuvant and adjuvant urothelial carcinoma therapy
KR20230159480A (en) 2021-03-19 2023-11-21 하이델베르크 파마 리서치 게엠베하 B-lymphocyte specific amatoxin antibody conjugate
EP4322938A1 (en) 2021-04-14 2024-02-21 Institut National de la Santé et de la Recherche Médicale (INSERM) New method to improve nk cells cytotoxicity
JP2024516230A (en) 2021-04-30 2024-04-12 ジェネンテック, インコーポレイテッド Therapeutic and diagnostic methods and compositions for cancer
WO2022241134A1 (en) 2021-05-13 2022-11-17 Gilead Sciences, Inc. COMBINATION OF A TLR8 MODULATING COMPOUND AND ANTI-HBV siRNA THERAPEUTICS
EP4351564A1 (en) 2021-06-11 2024-04-17 Gilead Sciences, Inc. Combination mcl-1 inhibitors with anti-cancer agents
AU2022290855A1 (en) 2021-06-11 2023-12-07 Gilead Sciences, Inc. Combination mcl-1 inhibitors with anti-body drug conjugates
EP4359411A1 (en) 2021-06-23 2024-05-01 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
IL309378A (en) 2021-06-23 2024-02-01 Gilead Sciences Inc Diacylglyercol kinase modulating compounds
WO2022271684A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
CN117396478A (en) 2021-06-23 2024-01-12 吉利德科学公司 Diacylglycerol kinase modulating compounds
AU2022302170A1 (en) 2021-07-02 2023-12-21 F. Hoffmann-La Roche Ag Methods and compositions for treating cancer
WO2023280790A1 (en) 2021-07-05 2023-01-12 INSERM (Institut National de la Santé et de la Recherche Médicale) Gene signatures for predicting survival time in patients suffering from renal cell carcinoma
WO2023010094A2 (en) 2021-07-28 2023-02-02 Genentech, Inc. Methods and compositions for treating cancer
CA3224180A1 (en) 2021-07-28 2023-02-02 F. Hoffmann-La Roche Ag Methods and compositions for treating cancer
TW202321308A (en) 2021-09-30 2023-06-01 美商建南德克公司 Methods for treatment of hematologic cancers using anti-tigit antibodies, anti-cd38 antibodies, and pd-1 axis binding antagonists
WO2023080900A1 (en) 2021-11-05 2023-05-11 Genentech, Inc. Methods and compositions for classifying and treating kidney cancer
WO2023088968A1 (en) 2021-11-17 2023-05-25 INSERM (Institut National de la Santé et de la Recherche Médicale) Universal sarbecovirus vaccines
TW202332429A (en) 2021-11-24 2023-08-16 美商建南德克公司 Therapeutic compounds and methods of use
WO2023097194A2 (en) 2021-11-24 2023-06-01 Genentech, Inc. Therapeutic compounds and methods of use
WO2023191816A1 (en) 2022-04-01 2023-10-05 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2023219613A1 (en) 2022-05-11 2023-11-16 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
EP4282435A1 (en) 2022-05-23 2023-11-29 Danmarks Tekniske Universitet Formulations of active pharmaceutical ingredients and excipients in icells via hydrophobic ion pairing
EP4282448A1 (en) 2022-05-23 2023-11-29 Danmarks Tekniske Universitet Porous expanding biocompatible scaffolds
WO2023240058A2 (en) 2022-06-07 2023-12-14 Genentech, Inc. Prognostic and therapeutic methods for cancer
WO2024015897A1 (en) 2022-07-13 2024-01-18 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2024020432A1 (en) 2022-07-19 2024-01-25 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2024049949A1 (en) 2022-09-01 2024-03-07 Genentech, Inc. Therapeutic and diagnostic methods for bladder cancer
WO2024052356A1 (en) 2022-09-06 2024-03-14 Institut National de la Santé et de la Recherche Médicale Inhibitors of the ceramide metabolic pathway for overcoming immunotherapy resistance in cancer
WO2024077095A1 (en) 2022-10-05 2024-04-11 Genentech, Inc. Methods and compositions for classifying and treating bladder cancer
WO2024077166A1 (en) 2022-10-05 2024-04-11 Genentech, Inc. Methods and compositions for classifying and treating lung cancer

Family Cites Families (36)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001014557A1 (en) 1999-08-23 2001-03-01 Dana-Farber Cancer Institute, Inc. Pd-1, a receptor for b7-4, and uses therefor
EP2388590A1 (en) 2001-04-02 2011-11-23 Dana Farber Cancer Institute PD-1, a receptor for B7-4, and uses thereof
AU2002258941A1 (en) 2001-04-20 2002-11-05 Mayo Foundation For Medical Education And Research Methods of enhancing cell responsiveness
AU2002348394A1 (en) 2001-10-25 2003-05-06 Merck And Co., Inc. Tyrosine kinase inhibitors
WO2003042402A2 (en) 2001-11-13 2003-05-22 Dana-Farber Cancer Institute, Inc. Agents that modulate immune cell activation and methods of use thereof
DE10161767T1 (en) 2002-07-03 2018-06-07 Honjo Tasuku Immunopotentiating compositions containing an anti-PD-L1 antibody
US7521051B2 (en) 2002-12-23 2009-04-21 Medimmune Limited Methods of upmodulating adaptive immune response using anti-PD-1 antibodies
AU2006244885B2 (en) 2005-05-09 2011-03-31 E. R. Squibb & Sons, L.L.C. Human monoclonal antibodies to programmed death 1(PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
US20080021217A1 (en) 2006-07-20 2008-01-24 Allen Borchardt Heterocyclic inhibitors of rho kinase
US20080242694A1 (en) 2006-09-18 2008-10-02 D Sidocky Neil R Amino-substituted heterocycles, compositions thereof, and methods of treatment therewith
SI2170959T1 (en) 2007-06-18 2014-04-30 Merck Sharp & Dohme B.V. Antibodies to human programmed death receptor pd-1
US20090264315A1 (en) * 2007-07-26 2009-10-22 Texas A&M University System Dipeptide mimics, libraries combining two dipeptide mimics with a third group, and methods for production thereof
TW200940537A (en) 2008-02-26 2009-10-01 Astrazeneca Ab Heterocyclic urea derivatives and methods of use thereof
US20130225527A1 (en) 2008-05-21 2013-08-29 Ariad Pharmaceuticals, Inc. Phosphorus Derivatives as Kinase Inhibitors
CN114835812A (en) 2008-12-09 2022-08-02 霍夫曼-拉罗奇有限公司 anti-PD-L1 antibodies and their use for enhancing T cell function
HUE037159T2 (en) 2009-11-24 2018-08-28 Medimmune Ltd Targeted binding agents against b7-h1
US8907053B2 (en) * 2010-06-25 2014-12-09 Aurigene Discovery Technologies Limited Immunosuppression modulating compounds
CN103732238A (en) * 2011-06-08 2014-04-16 奥瑞基尼探索技术有限公司 Therapeutic compounds for immunomodulation
US9006230B2 (en) 2011-08-30 2015-04-14 Wockhardt Ltd. 1,6-diazabicyclo [3,2,1] octan-7-one derivatives and their use in the treatment of bacterial infections
JP2015505850A (en) 2011-12-14 2015-02-26 シアトル ジェネティックス, インコーポレイテッド Novel antibody drug conjugates (ADC) and their use
US20140343017A1 (en) 2012-01-25 2014-11-20 Kabushiki Kaisha Yakult Honsha Pyrrole compound
WO2013131018A1 (en) 2012-03-02 2013-09-06 Zalicus Pharmaceuticals Ltd. Biaryl inhibitors of the sodium channel
EP2822957A1 (en) 2012-03-07 2015-01-14 Aurigene Discovery Technologies Limited Peptidomimetic compounds as immunomodulators
JP2015512910A (en) 2012-03-29 2015-04-30 オーリジーン ディスカバリー テクノロジーズ リミテッドAurigene Discovery Technologies Limited Immunomodulatory cyclic compounds derived from the BC loop of human PD1
EA201590197A1 (en) 2012-08-23 2015-07-30 Алиос Биофарма, Инк. COMPOUNDS FOR THE TREATMENT OF PARAMIX VIRAL VIRAL INFECTIONS
CN107892719B (en) 2012-10-04 2022-01-14 达纳-法伯癌症研究所公司 Human monoclonal anti-PD-L1 antibodies and methods of use
EP2906684B8 (en) 2012-10-10 2020-09-02 Sangamo Therapeutics, Inc. T cell modifying compounds and uses thereof
AR093984A1 (en) 2012-12-21 2015-07-01 Merck Sharp & Dohme ANTIBODIES THAT JOIN LEGEND 1 OF SCHEDULED DEATH (PD-L1) HUMAN
AU2014229283B2 (en) 2013-03-14 2016-07-28 Novartis Ag 3-pyrimidin-4-yl-oxazolidin-2-ones as inhibitors of mutant IDH
EA029661B1 (en) 2013-09-06 2018-04-30 Ауриген Дискавери Текнолоджиз Лимитед 1,3,4-oxadiazole and 1,3,4-thiadiazole derivatives as immunomodulators
PL3363790T3 (en) * 2013-09-06 2020-07-27 Aurigene Discovery Technologies Limited 1,2,4-oxadiazole derivatives as immunomodulators
CN112552401B (en) 2013-09-13 2023-08-25 广州百济神州生物制药有限公司 anti-PD 1 antibodies and their use as therapeutic and diagnostic agents
CA2979163A1 (en) 2015-03-10 2016-09-15 Aurigene Discovery Technologies Limited 3-substituted 1,3,4-oxadiazole and thiadiazole compounds as immunomodulators
BR112017019306A2 (en) 2015-03-10 2018-05-08 Aurigene Discovery Technologies Limited 1,3,4-oxadiazole and thiadiazole compounds as immunomodulators
WO2016149160A1 (en) 2015-03-15 2016-09-22 Sunshine Lake Pharma Co., Ltd. Substituted aminopyrimidine compounds and methods of use
WO2018051255A1 (en) 2016-09-14 2018-03-22 Aurigene Discovery Technologies Limited Cyclic substituted-1,3,4-oxadiazole and thiadiazole compounds as immunomodulators

Cited By (23)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11759454B2 (en) 2012-12-07 2023-09-19 Chemocentryx, Inc. Diazole lactams
US10744118B2 (en) 2012-12-07 2020-08-18 Chemocentryx, Inc. Diazole lactams
US10160736B2 (en) 2013-09-06 2018-12-25 Aurigene Discovery Technologies Limited 1,3,4-oxadiazole and 1,3,4-thiadiazole derivatives as immunomodulators
US11572366B2 (en) 2015-11-19 2023-02-07 Incyte Corporation Heterocyclic compounds as immunomodulators
US11535615B2 (en) 2015-12-22 2022-12-27 Incyte Corporation Heterocyclic compounds as immunomodulators
US11866435B2 (en) 2015-12-22 2024-01-09 Incyte Corporation Heterocyclic compounds as immunomodulators
US10568870B2 (en) 2016-04-07 2020-02-25 Chemocentryx, Inc. Reducing tumor burden by administering CCR1 antagonists in combination with PD-1 inhibitors or PD-L1 inhibitors
US11744822B2 (en) 2016-04-07 2023-09-05 Chemocentryx, Inc. Reducing tumor burden by administering CCR1 antagonists in combination with PD-1 inhibitors or PD-L1 inhibitors
US11608337B2 (en) 2016-05-06 2023-03-21 Incyte Corporation Heterocyclic compounds as immunomodulators
US11673883B2 (en) 2016-05-26 2023-06-13 Incyte Corporation Heterocyclic compounds as immunomodulators
US11873309B2 (en) 2016-06-20 2024-01-16 Incyte Corporation Heterocyclic compounds as immunomodulators
US11718605B2 (en) 2016-07-14 2023-08-08 Incyte Corporation Heterocyclic compounds as immunomodulators
US11613536B2 (en) 2016-08-29 2023-03-28 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2018098352A2 (en) 2016-11-22 2018-05-31 Jun Oishi Targeting kras induced immune checkpoint expression
TWI808955B (en) * 2016-12-22 2023-07-21 美商英塞特公司 Heterocyclic compounds as immunomodulators
US11787793B2 (en) 2016-12-22 2023-10-17 Incyte Corporation Heterocyclic compounds as immunomodulators
US11566026B2 (en) 2016-12-22 2023-01-31 Incyte Corporation Heterocyclic compounds as immunomodulators
US11414433B2 (en) 2018-05-11 2022-08-16 Incyte Corporation Heterocyclic compounds as immunomodulators
US11753406B2 (en) 2019-08-09 2023-09-12 Incyte Corporation Salts of a PD-1/PD-L1 inhibitor
US11866451B2 (en) 2019-11-11 2024-01-09 Incyte Corporation Salts and crystalline forms of a PD-1/PD-L1 inhibitor
US11760756B2 (en) 2020-11-06 2023-09-19 Incyte Corporation Crystalline form of a PD-1/PD-L1 inhibitor
US11780836B2 (en) 2020-11-06 2023-10-10 Incyte Corporation Process of preparing a PD-1/PD-L1 inhibitor
US11866434B2 (en) 2020-11-06 2024-01-09 Incyte Corporation Process for making a PD-1/PD-L1 inhibitor and salts and crystalline forms thereof

Also Published As

Publication number Publication date
PH12016500405A1 (en) 2016-05-16
HRP20181251T1 (en) 2018-10-05
EP3041828A4 (en) 2017-01-25
SG11201601679TA (en) 2016-04-28
HUE039014T2 (en) 2018-12-28
CY1120769T1 (en) 2019-12-11
PL3041828T3 (en) 2018-10-31
US9776978B2 (en) 2017-10-03
EP3041828A1 (en) 2016-07-13
US20180086726A1 (en) 2018-03-29
CU24345B1 (en) 2018-05-08
SG10201800508SA (en) 2018-02-27
PH12016500405B1 (en) 2016-05-16
ES2682040T3 (en) 2018-09-18
CA2922655A1 (en) 2015-03-12
AU2014316684A1 (en) 2016-04-28
IL244313A0 (en) 2016-04-21
DK3041828T3 (en) 2018-08-13
EA201600234A1 (en) 2016-08-31
PT3041828T (en) 2018-10-09
LT3041828T (en) 2018-09-10
US20170101387A1 (en) 2017-04-13
EP3385257A1 (en) 2018-10-10
CU20160028A7 (en) 2017-02-02
JP2016532711A (en) 2016-10-20
EP3041828B1 (en) 2018-05-23
MX2016002971A (en) 2016-10-07
US10160736B2 (en) 2018-12-25
KR20160081898A (en) 2016-07-08
WO2015033301A1 (en) 2015-03-12
RS57559B1 (en) 2018-10-31
CN105849092A (en) 2016-08-10
EA029661B1 (en) 2018-04-30
TR201811077T4 (en) 2018-08-27
SI3041828T1 (en) 2018-10-30

Similar Documents

Publication Publication Date Title
US10160736B2 (en) 1,3,4-oxadiazole and 1,3,4-thiadiazole derivatives as immunomodulators
US10961205B2 (en) 1,2,4-oxadiazole derivatives as immunomodulators
US10106581B2 (en) Cyclic peptidomimetic compounds as immunomodulators
WO2015036927A1 (en) Immunomodulating peptidomimetic derivatives
NZ718711B2 (en) 1,2,4-oxadiazole derivatives as immunomodulators

Legal Events

Date Code Title Description
AS Assignment

Owner name: AURIGENE DISCOVERY TECHNOLOGIES LIMITED, INDIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SASIKUMAR, POTTAYIL GOVINDAN NAIR;RAMACHANDRA, MURALIDHARA;NAREMADDEPALLI, SEETHARAMAIAH SETTY SUDARSHAN;SIGNING DATES FROM 20160223 TO 20160225;REEL/FRAME:037889/0301

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION