WO2015036927A1 - Immunomodulating peptidomimetic derivatives - Google Patents

Immunomodulating peptidomimetic derivatives Download PDF

Info

Publication number
WO2015036927A1
WO2015036927A1 PCT/IB2014/064380 IB2014064380W WO2015036927A1 WO 2015036927 A1 WO2015036927 A1 WO 2015036927A1 IB 2014064380 W IB2014064380 W IB 2014064380W WO 2015036927 A1 WO2015036927 A1 WO 2015036927A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
compound
compound according
carcinoma
pharmaceutically acceptable
Prior art date
Application number
PCT/IB2014/064380
Other languages
French (fr)
Inventor
Pottayil Govindan Nair Sasikumar
Muralidhara Ramachandra
Seetharamaiah Setty Sudarshan Naremaddepalli
Original Assignee
Aurigene Discovery Technologies Limited
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 Limited filed Critical Aurigene Discovery Technologies Limited
Publication of WO2015036927A1 publication Critical patent/WO2015036927A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/0606Dipeptides with the first amino acid being neutral and aliphatic the side chain containing heteroatoms not provided for by C07K5/06086 - C07K5/06139, e.g. Ser, Met, Cys, Thr
    • C07K5/06069Ser-amino acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/0606Dipeptides with the first amino acid being neutral and aliphatic the side chain containing heteroatoms not provided for by C07K5/06086 - C07K5/06139, e.g. Ser, Met, Cys, Thr
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to immunomodulating peptidomimetic derivatives.
  • the invention also relates to pharmaceutical compositions comprising the said peptidomimetic compounds and their derivatives 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 (Hyun-Tak Jin, 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
  • 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 peptidomimetic compounds which are capable of suppressing and/or inhibiting the programmed cell death 1 (PD1) signalling pathway.
  • PD1 programmed cell death 1
  • peptidomimetic compounds or a pharmaceutically acceptable salt or a stereoisomer thereof which are capable of suppressing and/or inhibiting the programmed cell death 1 (PD1) signalling pathway.
  • the present invention provides a peptidomimetic compounds of formula (I):
  • x, y and z are integers, each independently selected from 1 to 2; each of [Aaal], [Aaa2] and [Aaa3] represents x, y and z independently selected amino acid residues, with a proviso that [Aaal] is not selected from Ser, Thr, Ala or Leu if
  • [Aaa2] is selected from Asp, Asn, Glu or Gin;
  • [Aaa3] is selected from Ser, Asp, Ala, He, Phe, Trp, Glu or Thr;
  • [Aaal], [Aaa2] and [Aaa3] are optionally substituted by hydroxyl;
  • Ri and R 2 are independently hydrogen or alkyl;
  • P represents free N-terminus, alkylated N-terminus or acylated N-terminus of [Aaal];
  • Q represents free C-terminus, amidated C-terminus or esterified C-terminus of [Aaa3];
  • the bond between [Aaal] and -NRi- and/or between [Aaa2] and -NR 2 - is a peptide bond or a reduced peptide bond.
  • composition comprising a compound of formula (I) or a pharmaceutically acceptable salt or a stereoisomer and processes for preparing thereof.
  • peptidomimetic 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.
  • the present invention provides peptidomimetic 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)
  • x, y and z are integers, each independently selected from 1 to 2;
  • each of [Aaal], [Aaa2] and [Aaa3] represents x, y and z independently selected amino acid residues, with a proviso that [Aaal] is not selected from Ser, Thr, Ala or Leu if
  • [Aaa2] is selected from Asp, Asn, Glu or Gin;
  • [Aaa3] is selected from Ser, Asp, Ala, He, Phe, Trp, Glu or Thr;
  • [Aaal], [Aaa2] and [Aaa3] are optionally substituted by hydroxyl;
  • Ri and R 2 are independently hydrogen or alkyl;
  • P represents free N-terminus, alkylated N-terminus or acylated N-terminus of [Aaal];
  • Q represents free C-terminus, amidated C-terminus or esterified C-terminus of [Aaa3];
  • the bond between [Aaal] and -NRi- and/or between [Aaa2] and -NR 2 - is a peptide bond or a reduced peptide bond.
  • the present invention provides compounds of formula (IA)
  • the present invention provides peptidomimetic compounds of formula (IB):
  • each of Ri ' , R 2 ' and R3 independently represents side chain of an amino acid with the proviso that Ri ' is not a side chain of Ser, Thr, Ala or Leu if R 2 ' is a side chain of Asp, Asn, Glu or Gin and R 3 is a side chain of Ser, Asp, Ala, He, Phe, Trp, Glu or Thr;
  • R4 is -OR or -NH 2 ;
  • R is hydrogen, alkyl or arylalkyl
  • R 5 and R 6 are independently selected from hydrogen, alkyl or acyl
  • each of R7 and R7' independently represents hydrogen or alkyl
  • the present invention provides compounds of the formula (I) or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein, [Aaal ] is selected from the group consisting of: Ser, Ala, Cys, Asp, Phe, Pro, Asn and Lys.
  • the present invention provides compounds of the formula (I) or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein, [Aaa2] is selected from the group consisting of: Ala, Asn, Phe, Lys, Thr and Phe;
  • the present invention provides compounds of the formula (I) or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein, [Aaa3] is selected from the group consisting of: Thr, Ala, Phe, Ser, Lys, Gly, val and Asn.
  • 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 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 inventioncapable 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, meningoco
  • coli legionella, diphtheria, salmonella, bacilli, cholera, tetanus, botulism, anthrax, plague, leptospirosis, and Lyme's disease bacteria, pathogenic infection by the fungi Candida (albicans, krusei, glabrata, tropicalis, etc.), Cryptococcus neoformans, Aspergillus (fumigatus, niger, etc.), Genus Mucorales (mucor, absidia, rhizophus), Sporothrix schenkii, Blastomyces dermatitidis, Paracoccidioides brasiliensis, Coccidioides immitis and Histoplasma capsulatum, and pathogenic infection by the parasites Entamoeba histolytica, Balantidium coli, Naegleriafowleri, Acanthamoeba sp., Giardia lambia, Cryptosporidium sp., Pneum
  • 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-CSF, M-CSF, G-CSF, interferon-a, ⁇ , 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
  • acyl refers to RC(O)-, wherein R is alkyl as defined below.
  • examples of acyl group include, but are not limited to -C(0)CH 3 , -C(0)CH 2 CH 3j -C(0)(CH 2 ) 2 CH 3 , -C(0)(CH 2 ) 3 CH 3 , -C(0)(CH 2 ) 4 CH 3 or -C(0)(CH 2 ) 5 CH 3 .
  • alkyl refers to a hydrocarbon chain radical that includes solely carbon and hydrogen atoms in the backbone, containing no unsaturation, having from one to twenty carbon atoms (i.e., Ci-2o alkyl) or one to ten carbon atoms (i.e. , Ci-io alkyl) or one to five carbon atoms (i.e., C 1 -5 alkyl) and which is attached to the rest of the molecule by a single bond, e.g., including but not limited to methyl, ethyl, propyl, butyl, isobutyl, sec-butyl, tert-butyl, isopentyl or neopentyl. Unless set forth or recited to the contrary, all alkyl groups described or claimed herein may be straight chain or branched, substituted or unsubstituted.
  • aminoated C-terminus refers to that the C-terminal of the amino acid in amide form.
  • amide form refers to primary, secondary and/or tertiary amides and may be represented by the formula -C(0)NR x R y , wherein each of R x and R y independently represents hydrogen or alkyl.
  • 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.
  • esters refers to (Q-Ce) linear or branched alkyl, (C4- Cio)aryl, (C4-Cio)heteroaryl or arylalkyl esters.
  • esteerified C-terminus refers to that the C-terminal of the amino acid in ester form.
  • free C-terminus refers to that the C-terminal of the amino acid in -CO 2 H form.
  • hydroxyl refers to -OH group.
  • the term "optionally substituted” refers to the replacement of one or more hydrogen radicals in a given structure with the radical of a specified substituent including, but not limited to: alkyl, alkoxy, acyl, halo, and hydroxyl. It is understood that the substituent may be further substituted.
  • “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” means 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 method Analytical HPLC was performed using on ZIC HILIC 200A° column (4.6 mm x 250 mm, 5 ⁇ ), 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.
  • the urea linkage was carried out using coupling of le (1 g, 2.8 mmol) in THF (20 mL) at room temperature, with If (1.24 g, 2.8 mmol).
  • the coupling was initiated by the addition of TEA (0.42 g, 4.2 mmol) and the resultant mixture was stirred at room temperature.
  • THF was evaporated from the reaction mass, and partitioned between water and ethyl acetate. Organic layer was washed with water, brine, dried over Na 2 SC>4 and evaporated under reduced pressure to yield crude compound which was further washed with diethyl ether to yield 1.4 g of lg.
  • Fmoc group on li was deprotected by adding diethylamine (40.0 mL) to li (10.0 g, 20.5 mmol) in CH 2 CI 2 (40.0 mL). After an hour the resulting solution was concentrated in vacuum and the thick-residue was purified by column chromatography over neutral alumina (Eluent: 0-50% ethyl acetate in hexane then 0-5% methanol in chloroform) to yield 3.9 g + .
  • reaction mixture was diluted with CH 2 CI 2 (50 mL) and washed with 1.0 M of sodium bisulphate (50 mL x 2) and 1.0 M sodium carbonate (50 mL x 2), dried over Na 2 S0 4 and evaporated under reduced pressure to yield crude compound If, which was further purified by silica gel column chromatography (eluent: 0-20% ethyl acetate in Hexane) to yield 0.8 g of If.
  • the compound 2 was synthesised by similar procedure as depicted in Example 1 using Boc-L-Ala-OH instead of Boc-L-Ser(tBu)-OH (compound la) and L-Ala-OtBu instead of L-Thr(tBu)-OBzl (compound lj) to yield 1.5 g of crude compound 2.
  • the compound 4 was synthesised by similar procedure as depicted in Example 1 using Boc-L-Phe-OH instead of Boc-L-Ser(tBu)-OH (compound l a) and Cbz-Asn-OH instead of Cbz-Ala-OH (compound lc).
  • the compound 5 was synthesised by similar procedure as depicted in Example 1 using Boc-L-Asp(OtBu)-OH instead of Boc-L-Ser(tBu)-OH (compound l a).
  • the compound 6 was synthesised by similar procedure as depicted in Example 1 using Boc-L-Phe-OH instead of Boc-L-Ser(tBu)-OH (compound l a), Cbz-Phe-OH instead of Cbz-Ala-OH (compound lc) and H-Phe-OtBu instead of L-Thr(tBu)-OBzl (compound lj).
  • Example 7 Synthesis of
  • the compound 7 was synthesised by similar procedure as depicted in Example 1 using Boc-L-Hyp-OMe instead of Boc-L-Ser(tBu)-OH and Fmoc-Asn-OH instead of Cbz-Ala- OH (compound lc).
  • the compound 8 was synthesised by similar procedure as depicted in Example 1 using Boc-Lys(Boc)-OH instead of Boc-L-Ser(tBu)-OH (compound la) and Fmoc-Asn-OH instead of Cbz-Ala-OH (compound lc).
  • the compound 9 was synthesised using by procedure as depicted in Example 1 using Fmoc-Lys(Boc)-OH instead of Cbz-L-Ala-OH (compound lc).
  • the compound 10 was synthesised using similar procedure as depicted in Example 1 using Fmoc-Asn(Trt)-OH instead of Cbz-L-Ala-OH (compound lc) and H-Lys(Boc)- OtBu instead of L-Thr(tBu)-OBzl (compound lj).
  • the compound 11 was synthesised by similar procedure as depicted in Example 1 using compound llf in place compound lb (Example 1 , step la) and Cbz-Asn-OH in place of Cbz-Ala-OH to yield 0.25 g crude material of the title compound 11, LCMS: 450.2 (M+H) + .
  • Fmoc group was deprotected by the addition of diethylamine (13.5 mL) to a solution of compound 12e (4.5 g, 7.54 mmol) in CH 2 CI 2 at -10 °C. The reaction was stirred at room temperature for 3 h. Progress of reaction was monitored by TLC. After completion, the resulting solution was concentrated in vacuum to get a thick gummy residue. The crude compound was purified by silica gel (60-120mesh) column chromatography (eluent: 5% methanol in DCM) to afford 2.2 g of compound 12f, LCMS: 375.5 (M+H) + , 397.4 (M+Na) + .
  • the urea linkage was carried out using coupling of 12f (1.5 g, 4.0 mmol) in DMF at room temperature, with 12g (1.72 g, 4.0 mmol).
  • the coupling was initiated by the addition of TEA (0.81 g, 8.0 mmol) and the resultant mixture was stirred at room temperature for 30 min. Progress of reaction was monitored by TLC. After completion, the reaction mass was partitioned between ethyl acetate and water. Organic layer was washed with citric acid, brine, dried over Na 2 S0 4 and evaporated under reduced pressure to give crude compound. The resulting solid was washed with ether and dried to afford 1.1 g of Compound 12h.
  • 12k Fmoc group was deprotected by the addition of diethylamine (30 mL) to a solution of compound 12k (10 g, 14.5 mmol) in CH 2 CI 2 at -10 °C. The reaction was stirred at room temperature for 3 h. Progress of reaction was monitored by TLC. After completion, the resulting solution was concentrated in vacuum to get a thick gummy residue. The crude compound was purified by silica gel (60-120mesh) column chromatography (eluent: 5% methanol in DCM) to afford 6.0 g of compound 121.
  • the compound 14 was synthesised by similar procedure as depicted in Example 12 using Boc-Phe-OH instead of Boc-Asp(OtBu)-OH (compound 12d); Fmoc-Asn-OH instead of Fmoc-Ala-OH (compound 12a) and H-Gly-OtBu instead of H-Asn(Trt)-OBzl (compound 12 1).
  • rm-PDL-1 Recombinant mouse PD-Ll (rm-PDL-1, cat no: 1019-B7-100; R&D Systems) were used as the source of PD-Ll.
  • Working concentrations were titrated from 10 ⁇ to 1 ⁇ . (eBioscience-650850-85); 0.05% Trypsin and 0.02% EDTA (SIGMA 59417C); 96-well format ELISA plates (Corning CLS3390); BD FACS caliber (E6016); Recombinant mouse B7-H1/PDL1 Fc Chimera, (rm-PD-Ll cat no: 1019-B7-100).
  • Splenocytes harvested in a 50 mL falcon tube by mashing mouse spleen in a 40 ⁇ cell strainer were further treated with 1 mL ACK lysis buffer for 5 min at room temperature. After washing with 9 mL of RPMI complete media, cells were re-suspended in 3 mL of lxPBS in a 15 mL tube. 3 mL of Histopaque was added carefully to the bottom of the tube without disturbing overlaying splenocyte suspension. After centrifuging at 800xg for 20 min at room temperature, the opaque layer of splenocytes was collected carefully without disturbing / mixing the layers. Splenocytes were washed twice with cold lxPBS followed by total cell counting using Trypan Blue exclusion method and used further for cell based assays.
  • Splenocytes were cultured in RPMI complete media (RPMI + 10% fetal bovine serum + 1 mM sodium pyruvate + 10,000units/ml penicillin and 10,000 ⁇ g/ml streptomycin) and maintained in a CO 2 incubator with 5% C(3 ⁇ 4 at 37°C.
  • CFSE is a dye that passively diffuses into cells and binds to intracellular proteins.
  • lxlO 6 cells/mL of harvested splenocytes were treated with 5 ⁇ of CFSE in pre-warmed lxPBS/0.1% BSA solution for 10 min at 37°C. Excess CFSE was quenched using 5 volumes of ice-cold culture media to the cells and incubated on ice for 5 min.
  • CFSE labelled splenocytes were further given three washes with ice cold complete RPMI media.
  • CFSE labelled lxlO 5 splenocytes added to wells containing either MDA-MB231 cells (lxlO 5 cells cultured in high glucose DMEM medium) or recombinant human PDL- 1 (100 ng/mL) and test compounds.
  • Splenocytes were stimulated with anti-mouse CD3 and anti- mouse CD28 antibody (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

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to immunomodulating peptidomimetic derivatives 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

IMMUNOMODULATING PEPTIDOMIMETIC DERIVATIVES
This application claims the benefit of Indian provisional application number 4044/CHE/2013, filed on September 10, 2013 and Indian provisional application number 4045/CHE/2013, filed on September 10, 2013; which hereby incorporated by reference. TECHNICAL FIELD
The present invention relates to immunomodulating peptidomimetic derivatives. The invention also relates to pharmaceutical compositions comprising the said peptidomimetic compounds and their derivatives as therapeutic agents.
BACKGROUND OF THE INVENTION
Programmed cell death- 1 (PD-1) is a member of the CD28 superfamily that delivers negative signals upon interaction with its two ligands, PD-L1 or PD-L2. PD-1 and its ligands are broadly expressed and exert a wider range of immunoregulatory roles in T cells activation and tolerance compared with other CD28 members. PD-1 and its ligands are involved in attenuating infectious immunity and tumor immunity, and facilitating chronic infection and tumor progression. The biological significance of PD- 1 and its ligand suggests the therapeutic potential of manipulation of PD- 1 pathway against various human diseases (Hyun-Tak Jin, 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 (RD Harvey, Clinical Pharmacology & Therapeutics (2014); 96 2, 214-223).
Programmed death-1 (PD-1) is a co-receptor that is expressed predominantly by T cells. The binding of PD-1 to its ligands, PD-L1 or PD-L2, is vital for the physiological regulation of the immune system. A major functional role of the PD-1 signaling pathway is the inhibition of self-reactive T cells, which serve to protect against autoimmune diseases. Elimination of the PD-1 pathway can therefore result in the breakdown of immune tolerance that can ultimately lead to the development of pathogenic autoimmunity. Conversely, tumor cells can at times co-opt the PD-1 pathway to escape from immunosurveillance mechanisms. Therefore, blockade of the PD-1 pathway has become an attractive target in cancer therapy. Current approaches include six agents that are either PD-1 and PD-L1 targeted neutralizing antibodies or fusion proteins. More than forty clinical trials are underway to better define the role of PD-1 blockade in variety of tumor types. (Ariel Pedoeem et al., Clinical Immunology (2014), 153(1), 145-152).
International applications WO 01/14557, WO 02/079499, WO 2002/086083, WO 03/042402, WO 2004/004771 , WO 2004/056875, WO2006121168, WO2008156712, WO2010077634, WO2011066389, WO2014055897, WO2014059173, WO2014100079 and US patent US08735553 report PD-1 or PD-L1 inhibitory antibodies or fusion proteins.
Further, International applications, WO2011161699, WO2012/168944, WO2013144704 and WO2013132317report 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 peptidomimetic 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, provided are peptidomimetic compounds or a pharmaceutically acceptable salt or a stereoisomer thereof, which are capable of suppressing and/or inhibiting the programmed cell death 1 (PD1) signalling pathway.
In one aspect, the present invention provides a peptidomimetic compounds of formula (I):
Rl R2
P— [Aaal]x— N— N— [Aaa2]y— C(O)— [Aaa3]z— Q (I)
or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein, x, y and z are integers, each independently selected from 1 to 2; each of [Aaal], [Aaa2] and [Aaa3] represents x, y and z independently selected amino acid residues, with a proviso that [Aaal] is not selected from Ser, Thr, Ala or Leu if
(i) [Aaa2] is selected from Asp, Asn, Glu or Gin;
(ii) [Aaa3] is selected from Ser, Asp, Ala, He, Phe, Trp, Glu or Thr; and
(iii) each of x, y and z is 1 ;
wherein, [Aaal], [Aaa2] and [Aaa3] are optionally substituted by hydroxyl; Ri and R2 are independently hydrogen or alkyl;
P represents free N-terminus, alkylated N-terminus or acylated N-terminus of [Aaal];
Q represents free C-terminus, amidated C-terminus or esterified C-terminus of [Aaa3]; and
the bond between [Aaal] and -NRi- and/or between [Aaa2] and -NR2- is a peptide bond or a reduced peptide bond.
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 peptidomimetic 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 peptidomimetic 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)
Rl R2
I I
P— [Aaal]x— N— N— [Aaa2]y— C(O)— [Aaa3]z— Q (I)
or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein, x, y and z are integers, each independently selected from 1 to 2;
each of [Aaal], [Aaa2] and [Aaa3] represents x, y and z independently selected amino acid residues, with a proviso that [Aaal] is not selected from Ser, Thr, Ala or Leu if
(i) [Aaa2] is selected from Asp, Asn, Glu or Gin;
(ii) [Aaa3] is selected from Ser, Asp, Ala, He, Phe, Trp, Glu or Thr; and
(iii) each of x, y and z is 1 ;
wherein, [Aaal], [Aaa2] and [Aaa3] are optionally substituted by hydroxyl; Ri and R2 are independently hydrogen or alkyl;
P represents free N-terminus, alkylated N-terminus or acylated N-terminus of [Aaal];
Q represents free C-terminus, amidated C-terminus or esterified C-terminus of [Aaa3]; and
the bond between [Aaal] and -NRi- and/or between [Aaa2] and -NR2- is a peptide bond or a reduced peptide bond.
In yet another embodiment, the present invention provides compounds of formula (IA)
H
-[Aaal]x— N— N— [Aaa2]y— C(O)— [Aaa3]z— Q
H
(IA)
or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein, x, y, z, [Aaal], [Aaa2], [Aaa3], P and Q are same as defined in compound of formula (I). In yet another embodiment, the present invention provides peptidomimetic compounds of formula (IB):
Figure imgf000006_0001
or a stereoisomer or a pharmaceutically acceptable salt thereof;
wherein;
each of Ri ' , R2' and R3 independently represents side chain of an amino acid with the proviso that Ri ' is not a side chain of Ser, Thr, Ala or Leu if R2' is a side chain of Asp, Asn, Glu or Gin and R3 is a side chain of Ser, Asp, Ala, He, Phe, Trp, Glu or Thr;
R4 is -OR or -NH2;
R is hydrogen, alkyl or arylalkyl;
R5 and R6 are independently selected from hydrogen, alkyl or acyl;
each of R7 and R7' independently represents hydrogen or alkyl;
both Ra and Ra' represent hydrogen; or together represent an oxo (=0) group; both Rb and Rb' represent hydrogen; or together represent an oxo (=0) group. In yet another embodiment, the present invention provides compounds of the formula (I) or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein, [Aaal ] is selected from the group consisting of: Ser, Ala, Cys, Asp, Phe, Pro, Asn and Lys.
In yet another embodiment, the present invention provides compounds of the formula (I) or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein, [Aaa2] is selected from the group consisting of: Ala, Asn, Phe, Lys, Thr and Phe;
In yet another embodiment, the present invention provides compounds of the formula (I) or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein, [Aaa3] is selected from the group consisting of: Thr, Ala, Phe, Ser, Lys, Gly, val and Asn.
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), in which Ri is hydrogen. According to another embodiment, specifically provided are compounds of the formula (I), in which R2 is hydrogen.
According to yet another embodiment, specifically provided are compounds of the formula (I) and (IA), in which P is free N-terminus.
According to yet another embodiment, specifically provided are compounds of the formula (I) and (IA), in which Q is free C-terminus.
According to yet another embodiment, specifically provided are compounds of the formula (I), in which x, y and z are independently are 1.
According to yet another embodiment, specifically provided are compounds of the formula (I), in which x, y and z do not simultaneously have a value of 1.
According to yet another embodiment, specifically provided are compounds of the formula (I) and (IA), in which x is 2, y is 1 and z is 1.
According to yet another embodiment, specifically provided are compounds of the formula (I) and (IA), in which x is 1, y is 1 and z is 2.
According to yet another embodiment, specifically provided are compounds of the formula (I) and (IA), in which [Aaal] is selected from the group of: Ser, Ala, Asp, Phe and Lys.
According to yet another embodiment, specifically provided are compounds of the formula (I) and (IA), in which [Aaa2] is selected from the group of: Ala and Asn.
According to yet another embodiment, specifically provided are compounds of the formula (I) and (IA), in which [Aaa3] selected from the group of: Thr, Ala, Lys and Gly.
According to yet another embodiment, specifically provided are compounds of the formula (I) and (IA) in which one, more or all amino acid/s is/are D amino acid/s.
In an embodiment, specific compounds of formula (I) or a pharmaceutically acceptable salt or a stereoisomer thereof; without any limitation are enumerated in table 1. Table 1
Figure imgf000008_0001
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, use of compounds as disclosed in the present invention for use as a medicament.
In another embodiment, use of compounds as disclosed in the present invention in the manufacture of a medicament for the treatment of cancer or infectious disease.
In another embodiment, use of the compounds as disclosed in the present invention in the manufacture of 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 inventioncapable 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, Naegleriafowleri, Acanthamoeba sp., Giardia lambia, Cryptosporidium sp., Pneumocystis carinii, Plasmodium vivax, Babesia microti, Trypanosoma brucei, Trypanosoma cruzi, Leishmania donovani, Toxoplasma gondi, Nippostrongylus brasiliensis.
The compounds of the present invention may be used as single drugs or as a pharmaceutical composition in which the compound is mixed with various pharmacologically acceptable materials.
The pharmaceutical composition is usually administered by oral or inhalation routes, but can be administered by parenteral administration route. In the practice of this invention, compositions can be administered, for example, by orally, intravenous infusion, topically, intraperitoneally, intravesically or intrathecally. Examples of the parenteral administration includes but not limited to intraarticular (in the joints), intravenous, intramuscular, intradermal, intraperitoneal, and subcutaneous routes, include aqueous and non-aqueous, isotonic sterile injection solutions, which can contain antioxidants, buffers, bacteriostats, and solutes that render the formulation isotonic with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives. Oral administration, parenteral administration, subcutaneous administration and intravenous administration are the preferred methods of administration.
The dosage of the compounds of the present invention varies depending on age, weight, symptom, therapeutic efficacy, dosing regimen and/or treatment time. Generally, they may be administered by oral or inhalation routes, in an amount of 1 mg to 100 mg per time, from once a couple of days, once 3 days, once 2 days, once a day to a couple of times a day, in the case of an adult, or continuously administered by oral or inhalation routes from 1 to 24 hours a day. Since the dosage is affected by various conditions, an amount less than the above dosage may sometimes work well enough, or higher dosage may be required in some cases.
The compounds of the present invention may be administered in combination with other drugs for (1) complementation and/or enhancement of prevention and/or therapeutic efficacy of the preventive and/or therapeutic drug of the present invention, (2) dynamics, absorption improvement, dosage reduction of the preventive and/or therapeutic drug of the present invention, and/or (3) reduction of the side effects of the preventive and/or therapeutic drug of the present invention.
A concomitant medicine comprising the compounds of the present invention and other drug may be administered as a combination preparation in which both components are contained in a single formulation, or administered as separate formulations. The administration by separate formulations includes simultaneous administration and administration with some time intervals. In the case of the administration with some time intervals, the compound of the present invention can be administered first, followed by another drug or another drug can be administered first, followed by the compound of the present invention. The administration method of the respective drugs may be the same or different.
The dosage of the other drug can be properly selected, based on a dosage that has been clinically used. The compounding ratio of the compound of the present invention and the other drug can be properly selected according to age and weight of a subject to be administered, administration method, administration time, disorder to be treated, symptom and combination thereof. For example, the other drug may be used in an amount of 0.01 to 100 parts by mass, based on 1 part by mass of the compound of the present invention. The other drug may be a combination of two or more kind of arbitrary drugs in a proper proportion. The other drug that complements and/or enhances the preventive and/or therapeutic efficacy of the compound of the present invention includes not only those that have already been discovered, but those that will be discovered in future, based on the above mechanism.
Diseases on which this concomitant use exerts a preventive and/or therapeutic effect are not particularly limited. The concomitant medicine can be used for any diseases, as long as it complements and/or enhances the preventive and/or therapeutic efficacy of the compound of the present invention.
The compound(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-CSF, M-CSF, G-CSF, interferon-a, 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-TGF and Imatinib (Gleevac), a mitosis inhibitor, such as paclitaxel, Sunitinib (Sutent) or other antiangiogenic agents, an aromatase inhibitor, such as letrozole, an A2a adenosine receptor (A2AR) antagonist, an angiogenesis inhibitor, anthracyclines, oxaliplatin, doxorubicin, TLR4 antagonists, and IL-18 antagonists. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as is commonly understood by one of skill in art to which the subject matter herein belongs. As used herein, the following definitions are supplied in order to facilitate the understanding of the present invention.
As used herein, the term "acyl" refers to RC(O)-, wherein R is alkyl as defined below. Examples of acyl group include, but are not limited to -C(0)CH3, -C(0)CH2CH3j -C(0)(CH2)2CH3, -C(0)(CH2)3CH3, -C(0)(CH2)4CH3 or -C(0)(CH2)5CH3.
As used herein the term "alkyl" refers to a hydrocarbon chain radical that includes solely carbon and hydrogen atoms in the backbone, containing no unsaturation, having from one to twenty carbon atoms (i.e., Ci-2o alkyl) or one to ten carbon atoms (i.e. , Ci-io alkyl) or one to five carbon atoms (i.e., C1-5 alkyl) and which is attached to the rest of the molecule by a single bond, e.g., including but not limited to methyl, ethyl, propyl, butyl, isobutyl, sec-butyl, tert-butyl, isopentyl or neopentyl. Unless set forth or recited to the contrary, all alkyl groups described or claimed herein may be straight chain or branched, substituted or unsubstituted.
As used herein, the term "amidated C-terminus" refers to that the C-terminal of the amino acid in amide form.
As used herein, the term "amide form" refers to primary, secondary and/or tertiary amides and may be represented by the formula -C(0)NRxRy, wherein each of Rx and Ry independently represents hydrogen or alkyl.
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.
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 "ester" refers to (Q-Ce) linear or branched alkyl, (C4- Cio)aryl, (C4-Cio)heteroaryl or arylalkyl esters. As used herein the term "esterified C-terminus" refers to that the C-terminal of the amino acid in ester form.
As used herein the term "free C-terminus' refers to that the C-terminal of the amino acid in -CO2H form.
As used herein, the term "hydroxyl" refers to -OH group.
As used herein, the term "including" as well as other forms, such as "include", "includes," and "included," is not limiting.
As used herein, the term "optionally substituted" refers to the replacement of one or more hydrogen radicals in a given structure with the radical of a specified substituent including, but not limited to: alkyl, alkoxy, acyl, halo, and hydroxyl. It is understood that the substituent may be further substituted.
"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" means sufficient amount of the compound(s) of the present invention that (i) treats or prevents the particular disease, disorder or syndrome (ii) attenuates, ameliorates or eliminates one or more symptoms of the particular disease, disorder or syndrome or (iii) prevents or delays the onset of one or more symptoms of the particular disease, disorder or syndrome described herein. In the case of cancer, the therapeutically effective amount of the drug may decrease the number of cancer cells; decrease the cancer size; inhibit (i.e., slow to some extent and alternatively stop) cancer cell infiltration into peripheral organs; suppress (i.e., slow to some extent and alternatively stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer. In the case of infectious disease states, the therapeutic effective amount is an amount sufficient to decrease or alleviate an infectious diseases, the symptoms of an infections caused by bacterial, viral and fungal.
Naturally-occurring amino acids are identified throughout the specification by the conventional three-letter abbreviations indicated in the below table 2.
Table 2 (Amino acid codes)
Figure imgf000016_0001
Glutamic acid Glu Serine Ser
Glutamine Gin Threonine Thr
Glycine Gly Valine Val
The abbreviations used in the entire specification may be summarized hereinbelow with their particular meaning.
°C (degree Celsius); δ (delta); % (percentage); brine (NaCl solution); CH2CI2/DCM (Dichloromethane); BnBr (Benzyl bromide); br s (Broad singlet); Boc (Tert-butyloxycarbonyl); Bzl (Benzyloxy-carbonyl); CS2CO3 (Caesium carbonate); d (Doublet); DIPEA (Ν,Ν-Diisopropylethylamine); DMF (Dimethyl formamide); DMSO (Dimethyl sulphoxide); DMSO-d6 (Deuterated DMSO); EDC.HC1/EDCI (1 -(3 -Dimethyl aminopropyl)-3-carbodiimide hydrochloride); Et2NH (Diethylamine); Fmoc (9- Fluorenylmethyloxycarbonyl); g or gr (gram); H or ¾ (Hydrogen); ¾0 (Water); HOBt/HOBT (1 -Hydroxy benzotriazole); HCl (Hydrochloric acid); h or hr (Hours); Hz (Hertz); HPLC (High-performance liquid chromatography); K2CO3 (Potassium carbonate); LCMS (Liquid chromatography mass spectroscopy); MeOH (Methanol); mmol (Millimoles); M (Molar); μΐ (Microlitre); mL (Millilitre); mg (Milligram); m (Multiplet); MHz (Megahertz); MS (ES) (Mass spectroscopy-electro spray); rnin/min (Minutes); Na (Sodium); NaHC03 (Sodium bicarbonate); NH2NH2.H20 (Hydrazine hydrate); NMM (N- methyl morpholine); Na2S04 (Sodium sulphate); N2 (Nitrogen); NMR (Nuclear magnetic resonance spectroscopy); Pd/C (Palladium on carbon); PD-L1 (Programmed death-ligand 1); PD-L2 (Programmed cell death 1 ligand 2); prep- HPLC/preparative HPLC (Preparative High-performance liquid chromatography); S (Singlet); 'Bu/tBu (Tertiary butyl); TEA/Et3N (Triethylamine); TLC (Thin Layer Chromatography); THF (Tetrahydrofuran); TIPS (Triisopropylsilane); TFA/CF3COOH (Trifluoroacetic acid); t (Triplet); tR = (Retention time); TPP (Triphenylphosphine); Trt (Trityl or Triphenylmethyl), etc.
EXPERIMENTAL
An embodiment of the present invention provides the preparation of compounds of formula (I) according to the procedures of the following 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 200A° column (4.6 mm x 250 mm, 5 μπι), Flow rate: 1.0 mL / min. The elution conditions used are: Buffer A: 5 mmol ammonium acetate, Buffer B: Acetonitrile, Equilibration of the column with 90 % buffer B and elution by a gradient of 90 % to 40 % buffer B during 30 min.
Preparative HPLC Method: The crude material was purified by preparative HPLC using ZIC HILIC 200A° column (21.2 mm x 150 mm, 5 μπι). The elution conditions used are Eluent: A: 5 mmol ammonium acetate B: Acetonitrile, Flow rate: 20 mL / min. The compound was eluted by gradient elution 0-2 min = 90 % buffer B, 2-20 min = 90- 40 % buffer B with a flow rate of 20 mL/min.
Example 1: Synthesis of compound 1
Step la:
Figure imgf000018_0001
Potassium carbonate (3.9 g, 28 mmol) and methyl iodide (3.26 g, 22mmol) were added to a solution of la (5.0 g, 19.15 mmol) in DMF (50 mL) and stirred at room temperature for 3 h. The completeness of the reaction was confirmed by TLC analysis. DMF was evaporated from the reaction mass and partitioned between water and ethyl acetate. Organic layer was washed with water, brine, dried over Na2S04 and evaporated under reduced pressure to yield crude compound. The resulting crude compound was dissolved in methanol (50 mL), 99% hydrazine hydrate (12.5 mL) was added to the crude compound solution and stirred at room temperature for 6 h. The completeness of the reaction was confirmed by TLC analysis. The reaction mixture was evaporated under reduced pressure to yield 5 g of compound lb (Yield: 96.15%), LCMS: 276.3 (M+H)+. Step lb:
Figure imgf000019_0001
1c 1d
DIPEA (4.3 g, 33.5 mmol) was added slowly to a stirred solution of lc (3 g, 13.4 mmol), HOBt (2.2 g, 16.1 mmol) and EDC.HCl (3.1 g, 16.1 mmol) in DCM (50 mL) at room temperature. To the above reaction mixture lb (3.7 g, 13.4 mmol) was added slowly and stirred at room temperature for 12 h. The completeness of the reaction was confirmed by TLC analysis. The reaction mixture was then quenched with ice, precipitate was filtered and the solid re-crystallized with CH2C12 to yield 6 g of Id, (Yield: 93 %), LCMS: 481.4 (M+H)+.
Step lc:
Figure imgf000019_0002
To a solution of Id (1.5 g) in methanol (20 mL) under inert atmosphere 10% Pd/C (0.5 g) was added and stirred for 1 h under H2 atmosphere. The completion of the reaction was confirmed by TLC analysis. The Pd/C catalyst was removed by filtration through a Celite® pad, and washed with 50 mL of methanol. The combined organic filtrate on evaporation under reduced pressure to yield 1 g of le (Yield: 93%), LCMS: 347.2 (M+H)+.
Step Id:
Figure imgf000019_0003
1e 1g
The urea linkage was carried out using coupling of le (1 g, 2.8 mmol) in THF (20 mL) at room temperature, with If (1.24 g, 2.8 mmol). The coupling was initiated by the addition of TEA (0.42 g, 4.2 mmol) and the resultant mixture was stirred at room temperature. After the completion of 20 h, THF was evaporated from the reaction mass, and partitioned between water and ethyl acetate. Organic layer was washed with water, brine, dried over Na2SC>4 and evaporated under reduced pressure to yield crude compound which was further washed with diethyl ether to yield 1.4 g of lg. LCMS: 638.2 (M+H)+. Step le:
Figure imgf000020_0001
ig in To a solution of lg (1.4 g) in methanol (10 mL) under inert atmosphere, 10% Pd-C (0.4 g) was added and stirred at room temperature 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, which was then washed with 40 mL of methanol. The combined organic filtrate was evaporated under reduced pressure to obtain the desired 1.1 g of lh. LCMS: 548.1 (M+H)+
Step If:
Figure imgf000020_0002
1 h 1
To a solution of lh (1 g) in CH2CI2 (10 mL), trifluoroacetic acid (10 mL) and catalytic amount of triisopropylsilane were added and stirred for 3 h at room temperature. The resulting solution was concentrated in vacuum to yield 0.2 g of compound 1 (Yield:
32.6%), LCMS: 336.2 (M+H)+ The crude solid material was purified as preparative
HPLC method described under experimental conditions (yield: 30 mg, HPLC: tR= 13.9 min).
Figure imgf000020_0003
To a solution of Fmoc-Thr(tBu)-OH (15.0 g, 37.7 mmol) in 100.0 mL of DMF, Cs2C03 (14.8 g, 45.2 mmol) was added and the resutling mixture was cooled to 0 °C. To the reaction mixture benzyl bromide (7.74 g, 345.2 mmol) was added and the solution was stirred for 30 min at ice cold temperature followed by at room temperature for 12 h. The reaction mixture was concentrated under reduced pressure and diluted with ethyl acetate (150 mL). The organic layer was washed with water (2 x 100 mL), brine (1 x 100 mL) and dried over Na2S04. The filtered solution was concentrated and purified by silica gel column chromatogrophy (Eluent: 0-30% ethyl acetate in Hexane) to yeild 18.5 g of li as a white solid. LCMS: 433.1 (M-0'BU+H)+, 397.2 (M-OBzl)+.
Fmoc group on li was deprotected by adding diethylamine (40.0 mL) to li (10.0 g, 20.5 mmol) in CH2CI2 (40.0 mL). After an hour the resulting solution was concentrated in vacuum and the thick-residue was purified by column chromatography over neutral alumina (Eluent: 0-50% ethyl acetate in hexane then 0-5% methanol in chloroform) to yield 3.9 g +.
Figure imgf000021_0001
To a stirred solution of lj (1.6 g, 6.0 mmol) in CH2C12 (30 mL), TEA (1.2 g, 12.0 mmol) was added followed by the addition of 4-nitrophenyl chloroformate (1.3 g, 6.6 mmol) in CH2CI2 (10 mL). 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, the reaction mixture was diluted with CH2CI2 (50 mL) and washed with 1.0 M of sodium bisulphate (50 mL x 2) and 1.0 M sodium carbonate (50 mL x 2), dried over Na2S04 and evaporated under reduced pressure to yield crude compound If, which was further purified by silica gel column chromatography (eluent: 0-20% ethyl acetate in Hexane) to yield 0.8 g of If. lU NMR (DMSO-d6, 300 MHz): δ 8.23-8.27 (m, 2H), 7. ,27 '-7.39 (d, 7H), 5.99-6.01 (m, 1H), 5.10-5.25 (m, 2H), 4.28-4.31 (m, 2H), 1.28-1.30 (d, 3H), 1.12 (s, 9H).
Example 2: Synthesis of co
Figure imgf000021_0002
The compound 2 was synthesised by similar procedure as depicted in Example 1 using Boc-L-Ala-OH instead of Boc-L-Ser(tBu)-OH (compound la) and L-Ala-OtBu instead of L-Thr(tBu)-OBzl (compound lj) to yield 1.5 g of crude compound 2. The crude solid material (0.5 g) was purified as preparative HPLC method described under experimental conditions (yield: 0.17 g, analytical HPLC: tR = 15.2 min), LCMS: 290.5 (M+H)+.
Example 3: Synthesis of
Figure imgf000022_0001
The compound 3 was synthesised by similar procedure as depicted in Example 1 using Boc-L-Cys(tBu)-OH instead of Boc-L-Ser(tBu)-OH (compound l a) and Cbz-Asn-OH instead of Cbz-Ala-OH (compound lc). Yield: 2 g (crude); LCMS: 434.4 (M+H20)+. Example 4: Synthesis of compound 4
Figure imgf000022_0002
The compound 4 was synthesised by similar procedure as depicted in Example 1 using Boc-L-Phe-OH instead of Boc-L-Ser(tBu)-OH (compound l a) and Cbz-Asn-OH instead of Cbz-Ala-OH (compound lc). The crude solid material (0.06 g) was purified as preparative HPLC method described under experimental conditions. LCMS: 439.2 (M+H)+, HPLC: tR = 8.63 min.
Example 5: Synthesis of compound 5
Figure imgf000022_0003
The compound 5 was synthesised by similar procedure as depicted in Example 1 using Boc-L-Asp(OtBu)-OH instead of Boc-L-Ser(tBu)-OH (compound l a). The crude solid material (0.35 g) was purified as preparative HPLC method described under experimental conditions. LCMS: 364.1 (M+H)+, HPLC: tR = 12.19 min.
Example 6: Synthesis of compound 6
Figure imgf000023_0001
The compound 6 was synthesised by similar procedure as depicted in Example 1 using Boc-L-Phe-OH instead of Boc-L-Ser(tBu)-OH (compound l a), Cbz-Phe-OH instead of Cbz-Ala-OH (compound lc) and H-Phe-OtBu instead of L-Thr(tBu)-OBzl (compound lj). The crude solid material (0.18 g) was purified as preparative HPLC method described under experimental conditions. LCMS: 518.5 (M+H)+, HPLC: tR = 4.8 min. Example 7: Synthesis of
Figure imgf000023_0002
The compound 7 was synthesised by similar procedure as depicted in Example 1 using Boc-L-Hyp-OMe instead of Boc-L-Ser(tBu)-OH and Fmoc-Asn-OH instead of Cbz-Ala- OH (compound lc). The crude solid material (0.6 g) was purified as preparative HPLC method described under experimental conditions. LCMS: 405.2 (M+H)+, HPLC: tR = 15.15 min.
Example 8: Synthesis of compound 8
Figure imgf000023_0003
The compound 8 was synthesised by similar procedure as depicted in Example 1 using Boc-Lys(Boc)-OH instead of Boc-L-Ser(tBu)-OH (compound la) and Fmoc-Asn-OH instead of Cbz-Ala-OH (compound lc). The crude solid material (0.15 g) was purified as preparative HPLC method described under experimental conditions. LCMS: 420.5 (M+H)+, HPLC: tR = 20.15 min.
Example 9: Synthesis of compound 9
Figure imgf000024_0001
The compound 9 was synthesised using by procedure as depicted in Example 1 using Fmoc-Lys(Boc)-OH instead of Cbz-L-Ala-OH (compound lc). The crude solid material (1 g) was purified as preparative HPLC method described under experimental conditions LCMS: 393.2 (M+H)+, HPLC: tR = 9.95 min.
Example 10: Synthesis of compound 10
Figure imgf000024_0002
The compound 10 was synthesised using similar procedure as depicted in Example 1 using Fmoc-Asn(Trt)-OH instead of Cbz-L-Ala-OH (compound lc) and H-Lys(Boc)- OtBu instead of L-Thr(tBu)-OBzl (compound lj). The crude solid material (1 g) was purified as preparative HPLC method described under experimental conditions. LCMS: 406.2 (M+H)+, HPLC: tR = 16.91 min.
Example 11: Synthesis of compound 11
Figure imgf000024_0003
Step 11a:
Figure imgf000024_0004
Potassium carbonate (7.0 g, 50.8 mmol) and methyl iodide (2.5 g, 18.6 mmol) were added to a solution of compound 11a (5.0 g, 16.9 mmol) in DMF (35 mL) and stirred at room temperature for 3 h. The completeness of the reaction was confirmed by TLC analysis. DMF was evaporated from the reaction mass, and partitioned between water and ethyl acetate. Organic layer was washed with water, brine, dried over Na2S04 and evaporated under reduced pressure get the desired 4.4 g of compound lib (Yield: 84.6%). LCMS: 310.1 (M+H)+ .
Step lib:
Figure imgf000025_0001
To a solution of compound lib (4.4 g) in methanol (25 mL) under inert atmosphere 10% Pd/C (0.8 g) was added and stirred for 10 h under ¾ atmosphere. The completion of the reaction was confirmed by TLC analysis. The Pd/C catalyst was then removed by filtration through a Celite® bed and washed with 50 mL of methanol. The combined organic filtrate on evaporation under reduced pressure to yield 2.0 g of compound 11c (Yield: 80%). LCMS: 176.1 (M+H)+.
Step 11c:
Figure imgf000025_0002
DIPEA (2.2 g, 17.1 mmol) was added slowly to a stirred solution of 11c (1.1 g, 5.7 mmol), HOBt (0.9 g, 6.8 mmol) and EDC.HC1 (1.3 g, 6.8 mmol) in DMF (10 mL). The reaction mixture was stirred at room temperature for 5 min. To the above reaction mixture compound lid (1.0 g, 5.7 mmol) was added slowly and stirred at room temperature for 12 h. The completeness of the reaction was confirmed by TLC analysis. The reaction mixture was then quenched with ice and the precipitate was filtered. The solid re-crystallized with CH2CI2 to yield 1.1 g of compound lie, (Yield: 57.8%). LCMS: 347.4 (M+H)+.
Step lid:
Figure imgf000025_0003
To a solution of compound lie (1.1 g, 3.17 mmol) in methanol (10 mL), 99% hydrazine hydrate (1.2 mL) was added and stirred at room temperature for 6 h. The completeness of the reaction was confirmed by TLC analysis. The reaction mixture was evaporated under reduced pressure to yield lg of compound llf (Yield: 91.15%), LCMS: 347.4 (M+H)+.
The compound 11 was synthesised by similar procedure as depicted in Example 1 using compound llf in place compound lb (Example 1 , step la) and Cbz-Asn-OH in place of Cbz-Ala-OH to yield 0.25 g crude material of the title compound 11, LCMS: 450.2 (M+H)+.
Example 12: Synthesis of compound 12
Step 12a:
Figure imgf000026_0001
12a 12b
Potassium carbonate (4.4 g, 32.2 mmol) and methyl iodide (2.5 g, 17.7 mmol) were added to a solution of compound 12a (5.0 g, 16.1 mmol) in DMF 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 Na2SC>4 and evaporated under reduced pressure to get 5.0 g of compound 12b, LCMS: 326.5 (M+H)+
Step 12b:
Figure imgf000026_0002
12b 12c
Hydrazine hydrate (6.9 g, 138 mmol) was added to a solution of compound 12b (4.5 g, 13.8 mmol) in methanol 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 Na2S04 and evaporated under reduced pressure yielded 3.2 g of compound 12c. LCMS: 326.2 (M+H)+ .
Step 12c:
Figure imgf000026_0003
12c 12d 12e DIPEA (3.2 g, 25.3 mmol) was added slowly to a stirred solution of 12d (2.7 g, 9.2 mmol), HOBt (1.36 g, 10.1 mmol) and EDC.HC1 (1.93 g, 10.1 mmol) in DCM and stirred at room temperature. To the above reaction mixture 12c (3.0 g, 9.2 mmol) was added slowly and stirred at room temperature for 1 h. The completeness of the reaction was confirmed by TLC analysis. The reaction mass was partitioned between ethyl acetate and water. Organic layer was washed with citric acid, NaHCOs, brine, and dried over Na2S04 and evaporated under reduced pressure to give crude compound. The crude was washed with diethyl ether and dried to yield 4.52 g of compound 12e; LCMS: 619.4(M+Na) +.
Figure imgf000027_0001
Fmoc group was deprotected by the addition of diethylamine (13.5 mL) to a solution of compound 12e (4.5 g, 7.54 mmol) in CH2CI2 at -10 °C. The reaction was stirred at room temperature for 3 h. Progress of reaction was monitored by TLC. After completion, the resulting solution was concentrated in vacuum to get a thick gummy residue. The crude compound was purified by silica gel (60-120mesh) column chromatography (eluent: 5% methanol in DCM) to afford 2.2 g of compound 12f, LCMS: 375.5 (M+H)+, 397.4 (M+Na)+.
Step 12e:
Figure imgf000027_0002
The urea linkage was carried out using coupling of 12f (1.5 g, 4.0 mmol) in DMF at room temperature, with 12g (1.72 g, 4.0 mmol). The coupling was initiated by the addition of TEA (0.81 g, 8.0 mmol) and the resultant mixture was stirred at room temperature for 30 min. Progress of reaction was monitored by TLC. After completion, the reaction mass was partitioned between ethyl acetate and water. Organic layer was washed with citric acid, brine, dried over Na2S04 and evaporated under reduced pressure to give crude compound. The resulting solid was washed with ether and dried to afford 1.1 g of Compound 12h.
Step 12f:
Figure imgf000028_0001
To a solution of compound 12h (1.1 g, 1.65 mmol) in methanol, was added 10% Pd-C (0.15 g) under inert atmosphere. The mixture was stirred for 1 h under H2 atmosphere at room temperature. 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 methanol (2 x 50 mL). The combined organic filtrate on evaporation under reduced pressure resulted in the isolation of 0.7 g compound 12i, LCMS: 576.3 (M+H)+, 598.4 (M+H)+
Step 12g:
Figure imgf000028_0002
To a stirred solution of 12j (10 g, 16.7 mmol) in DMF, potassium carbonate (4.62 g, 33.6 mmol) and BnBr (3.2 g, 18.5 mmol) were added and continued reaction at room temperature for 2 h. The progress of reaction was monitored by TLC. After completion, the reaction mass was partitioned between ethyl acetate and water. Organic layer was washed with water, brine, and dried over Na2SC>4 and evaporated under reduced pressure. The resulting solid was washed with n-hexane and dried to yield 10.0 g of compound 12k.
Step 12h:
Figure imgf000028_0003
12k Fmoc group was deprotected by the addition of diethylamine (30 mL) to a solution of compound 12k (10 g, 14.5 mmol) in CH2CI2 at -10 °C. The reaction was stirred at room temperature for 3 h. Progress of reaction was monitored by TLC. After completion, the resulting solution was concentrated in vacuum to get a thick gummy residue. The crude compound was purified by silica gel (60-120mesh) column chromatography (eluent: 5% methanol in DCM) to afford 6.0 g of compound 121.
Step 12i:
Figure imgf000029_0001
DIPEA (4.8 g, 3.8 mmol) was added slowly to a stirred solution of 12i (0.83 g, 1.5 mmol), HOBt (0.22 g, 1.65 mmol) and EDC.HCI (0.32 g, 1.65 mmol) in DCM and stirred at room temperature. To the above reaction mixture 121 (0.47 g, 1.5 mmol) was added slowly and stirred at room temperature for 1 h. The completeness of the reaction was confirmed by TLC analysis. The reaction mass was partitioned between ethyl acetate and water. Organic layer was washed with citric acid, NaHC03, brine, and dried over Na2S04 and evaporated under reduced pressure to give crude compound. The crude was washed with diethyl ether and dried to yield 1.0 g of compound 12m.
Step 12j:
Figure imgf000029_0002
To a solution of compound 12m (1.0 g, 0.98 mmol) in methanol, was added 10% Pd-C (0.1 g) under inert atmosphere. The mixture was stirred for 1 h under ¾ atmosphere at room temperature. 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 methanol (2 x 50 mL). The combined organic filtrate on evaporation under reduced pressure resulted in the isolation of 0.77 g compound 12n, LCMS: 932.7 (M+H)+, 954.8 (M+Na)+.
Step 12k:
Figure imgf000030_0001
To a solution of compound 12n (0.77 g, 0.77 mmol) in CH2CI2, trifluoroacetic acid (2.5 mL) and catalytic amount of triisopropylsilane were added and stirred at room temperature for 2 h. The resulting solution was concentrated under nitrogen and the solid material was further washed with diethyl ether, dried under vacuum to afford 0.5 g of crude compound. Crude compound was further purified by preparative HPLC method described under experimental conditions to yield pure compound 12, LCMS: 478.2. (M+H)+.
Example 13: Synthesis of compound 13
Ste
Figure imgf000030_0002
1 3a 1 3b
1 3c
Compound 13b (2.5 g, 5.9 mmol), TEA (1.2 g, 11.8 mmol) dissolved in DMF were added drop wise to a solution of 13a (1.5 g, 5.9 mmol) at 0 °C for urea bond formation and the mixture was stirred at room temperature for 30 min. The progress of the reaction was monitored by TLC analysis. After completion, the reaction mass was partitioned between ethyl acetate and water. Organic layer was washed with water, brine, and dried over Na2S04 and evaporated under reduced pressure to give crude compound. The crude was further crystallized from hexane to yield 1.2 g of compound 13c.
Step 13b:
Figure imgf000030_0003
13c 3d
To a solution of compound 13c (1.2 g, 2.35 mmol) in methanol (20 mL), was added 10% Pd-C (0.15 g) under inert atmosphere. The mixture was stirred for 1 h under ¾ atmosphere at room temperature. 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 methanol (2 x 75 mL). The combined organic filtrate on evaporation under reduced pressure resulted in the isolation of 0.7 g compound 13d, LCMS: 419.4 (M+H)+. Step 13c:
Figure imgf000031_0001
Compound 13e was synthesised by similar procedure as depicted for compound 1 If in step 1 Id of Example 11 using Boc-Ser(OtBu)-OH instead of Boc-Ala-OH (compound l id) and Fmoc-Asn(Trt)-OH instead of Cbz-Ser(OtBu)-OH (compound 11a). Yield: 2.5 g; LCMS: 654.3 (M+Na)+.
DIPEA (0.35 g, 2.8 mmol) was slowly added to a stirred solution of compound 13d (0.46 g, 1.1 mmol), compound 13e (0.7 g, 1.1 mmol), HOBt (0.16 g, 1.2 mmol) and EDC.HCI (0.23 g, 1.2 mmol) in DCM at 0°C. The reaction mixture was stirred at room temperature for 1 h. The completeness of the reaction was confirmed by TLC analysis. The reaction mass was partitioned between ethyl acetate and water. Organic layer was washed with citric acid, NaHCC>3, brine, and dried over Na2SC¼ and evaporated under reduced pressure to give crude compound. The crude was washed with diethyl ether and dried to afford 0.82 g of compound 13f. LCMS: 1032.9 (M+H)+.
Step 13d:
Figure imgf000031_0002
To a solution of compound 13f (0.8 g, 0.77 mmol) in CH2CI2, trifluoroacetic acid (1 mL) and catalytic amount of triisopropylsilane were added and stirred at room temperature for 2 h. The resulting solution was concentrated under nitrogen and the solid material was further washed with diethyl ether, dried under vacuum to afford 0.5 g of crude compound. Crude compound was further purified by preparative HPLC method described under experimental conditions to yield pure compound 13, LCMS: 466.3 (M+H)+, HPLC: tR = 16.41 min.
Example 14: Synthesis of compound 14
Figure imgf000032_0001
The compound 14 was synthesised by similar procedure as depicted in Example 12 using Boc-Phe-OH instead of Boc-Asp(OtBu)-OH (compound 12d); Fmoc-Asn-OH instead of Fmoc-Ala-OH (compound 12a) and H-Gly-OtBu instead of H-Asn(Trt)-OBzl (compound 12 1). The crude solid material was purified as preparative HPLC method described under experimental conditions. LCMS: 496.4 (M+H)+, HPLC: tR = 11.6 min.
The compounds shown in below table 3, 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 3
Figure imgf000032_0002
Rescue of mouse splenocvte proliferation in the presence of recombinant PD- L1/PD-L2:
Recombinant mouse PD-Ll (rm-PDL-1, cat no: 1019-B7-100; R&D Systems) were used as the source of PD-Ll.
Requirement:
Mouse splenocytes harvested from 6-8 weeks old C57 BL6 mice; RPMI 1640 (GIBCO, Cat # 11875); DMEM with high glucose (GIBCO, Cat # D6429); Fetal Bovine Serum [Hyclone, Cat # SH30071.03]; Penicillin (10000unit/ml)-Streptomycin(10,000μg/ml) Liquid (GIBCO, Cat # 15140-122); MEM Sodium Pyruvate solution lOOmM (lOOx), Liquid (GIBCO, Cat # 11360); Nonessential amino acid (GIBCO, Cat # 11140); L- Glutamine (GIBCO, Cat # 25030); Anti-CD3 antibody (eBiosciences - 16-0032); Anti- CD28 antibody (eBiosciences - 16-0281); ACK lysis buffer (lmL) (GIBCO, Cat # - A10492); Histopaque (density- 1.083 gm/mL) (SIGMA 10831); Trypan blue solution (SIGMA-T8154); 2 mL Norm Ject Luer Lock syringe- (Sigma 2014-12); 40 μΜ nylon cell strainer (BD FALCON 35230); Hemacytometer (Bright line-SIGMA Z359629); FACS Buffer (PBS/0.1% BSA): Phosphate Buffered Saline (PBS) pH 7.2 (HiMedia TS1006) with 0.1 % Bovine Serum Albumin (BSA) (SIGMA A7050) and sodium azide (SIGMA 08591); 5 mM stock solution of CFSE: CFSE stock solution was prepared by diluting lyophilized CFSE with 180 of Dimethyl sulfoxide (DMSO C2H6SO, SIGMA-D-5879) and aliquoted in to tubes for further use. Working concentrations were titrated from 10 μΜ to 1 μΜ. (eBioscience-650850-85); 0.05% Trypsin and 0.02% EDTA (SIGMA 59417C); 96-well format ELISA plates (Corning CLS3390); BD FACS caliber (E6016); Recombinant mouse B7-H1/PDL1 Fc Chimera, (rm-PD-Ll cat no: 1019-B7-100).
Protocol
Splenocyte preparation and culturing:
Splenocytes harvested in a 50 mL falcon tube by mashing mouse spleen in a 40 μπι cell strainer were further treated with 1 mL ACK lysis buffer for 5 min at room temperature. After washing with 9 mL of RPMI complete media, cells were re-suspended in 3 mL of lxPBS in a 15 mL tube. 3 mL of Histopaque was added carefully to the bottom of the tube without disturbing overlaying splenocyte suspension. After centrifuging at 800xg for 20 min at room temperature, the opaque layer of splenocytes was collected carefully without disturbing / mixing the layers. Splenocytes were washed twice with cold lxPBS followed by total cell counting using Trypan Blue exclusion method and used further for cell based assays.
Splenocytes were cultured in RPMI complete media (RPMI + 10% fetal bovine serum + 1 mM sodium pyruvate + 10,000units/ml penicillin and 10,000μg/ml streptomycin) and maintained in a CO2 incubator with 5% C(¾ at 37°C.
CFSE Proliferation assay:
CFSE is a dye that passively diffuses into cells and binds to intracellular proteins. lxlO6 cells/mL of harvested splenocytes were treated with 5 μΜ of CFSE in pre-warmed lxPBS/0.1% BSA solution for 10 min at 37°C. Excess CFSE was quenched using 5 volumes of ice-cold culture media to the cells and incubated on ice for 5 min. CFSE labelled splenocytes were further given three washes with ice cold complete RPMI media. CFSE labelled lxlO5 splenocytes added to wells containing either MDA-MB231 cells (lxlO5 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 4
Figure imgf000034_0001

Claims

We claim:
1. A compound of formula (I)
Ri R2
P— [Aaal]x— N— N— [Aaa2]y— C(O)— [Aaa3]z— Q (I)
or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein, x, y and z are integers, each independently selected from 1 to 2;
each of [Aaal], [Aaa2] and [Aaa3] represents x, y and z independently selected amino acid residues, with a proviso that [Aaal] is not selected from Ser, Thr, Ala or Leu if
(i) [Aaa2] is selected from Asp, Asn, Glu or Gin;
(ii) [Aaa3] is selected from Ser, Asp, Ala, He, Phe, Trp, Glu or Thr; and
(iii) each of x, y and z is 1 ;
wherein, [Aaal], [Aaa2] and [Aaa3] are optionally substituted by hydroxyl;
Ri and R2 are independently hydrogen or alkyl;
P represents free N-terminus, alkylated N-terminus or acylated N-terminus of [Aaal];
Q represents free C-terminus, amidated C-terminus or esterified C-terminus of [Aaa3]; and
the bond between [Aaal] and -NRi- and/or between [Aaa2] and -NR2- is a peptide bond or a reduced peptide bond.
The compound according to claim 1 , wherein the compound of formula (I) is compound of formula (IA):
H
P— [Aaal]x— N— N— [Aaa2]y— C(O)— [Aaa3]z— Q
H
(IA)
or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein, x, y, z, [Aaal], [Aaa2], [Aaa3], P and Q are same as defined in claiml.
The compound according to any one of claims 1 to 2, wherein x, y and z are independently 1.
The compound according to any one of claims 1 to 2, wherein x, y and z simultaneously do not have a value of 1.
5. The compound according to any one of claims 1 , 2 or 4, wherein x is 1 or 2, y is 1 and z is 1 or 2.
6. The compound according to claim 1 , or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein [Aaal] is selected from the group consisting of: Ser, Ala, Cys, Asp, Phe, Pro, Asn and Lys.
7. The compound according to claim 1 , or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein [Aaa2] is selected from the group consisting of: Ala, Asn, Phe, Lys, Thr and Phe.
8. The compound according to claim 1 , or a pharmaceutically acceptable salt or a stereoisomer thereof; wherein [Aaa3] is selected from the group consisting of: Thr, Ala, Phe, Ser, Lys, Gly, val and Asn.
9. The compound according to any one of claims 1 to 8, wherein P is free N-terminus and Q is free C-terminus.
10. A compound is selected from the group consisting of
Figure imgf000036_0001
Figure imgf000037_0001
or a pharmaceutically acceptable sa t or a stereoisomer thereof.
11. A pharmaceutical composition comprising at least one compound according to any one of claims 1 to 10 or a pharmaceutically acceptable salt or a stereoisomer thereof, and a pharmaceutically acceptable carrier or excipient.
12. The pharmaceutical composition according to claim 11, further comprising at least one of an anticancer agent, chemotherapy agent, or antiproliferative compound.
13. Use of a compound according to any one of claims 1 to 10, or a pharmaceutically acceptable salt or a stereoisomer thereof, as a medicament.
14. Use of a compound according to any one of claims 1 to 10, or a pharmaceutically acceptable salt or a stereoisomer thereof, in the manufacture of a medicament for the treatment of cancer or infectious disease.
15. The use according to claim 14, wherein the cancer is selected from 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.
16. The use according to claim 14, wherein the infectious disease is a bacterial infectious disease, a viral infectious disease or a fungal infectious disease.
17. 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 any one of claims 1 to 10.
18. 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 any one of claims 1 to 10.
19. The method of claim 18, 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.
20. The method of claim 18, wherein the tumour cells are of a cancer selected from the list consisting of 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.
A method of treating an infectious disease in a subject comprising administering to the subject a therapeutically effective amount of compound according to any one of claims 1 to 10.
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 any one of claims 1 to 10.
A method of treating cancer in a subject comprising administering to the subject a therapeutically effective amount of compound according to any one of claims 1 to 10.
PCT/IB2014/064380 2013-09-10 2014-09-10 Immunomodulating peptidomimetic derivatives WO2015036927A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
IN4044/CHE/2013 2013-09-10
IN4045/CHE/2013 2013-09-10
IN4045CH2013 2013-09-10
IN4044CH2013 2013-09-10

Publications (1)

Publication Number Publication Date
WO2015036927A1 true WO2015036927A1 (en) 2015-03-19

Family

ID=52665145

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2014/064380 WO2015036927A1 (en) 2013-09-10 2014-09-10 Immunomodulating peptidomimetic derivatives

Country Status (1)

Country Link
WO (1) WO2015036927A1 (en)

Cited By (108)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017122175A1 (en) 2016-01-13 2017-07-20 Acerta Pharma B.V. Therapeutic combinations of an antifolate and a btk inhibitor
WO2018006795A1 (en) 2016-07-05 2018-01-11 广州再极医药科技有限公司 Aromatic acetylene or aromatic ethylene compound, intermediate, preparation method, pharmaceutical composition and use thereof
WO2018055080A1 (en) 2016-09-22 2018-03-29 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
WO2019020593A1 (en) 2017-07-25 2019-01-31 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for modulating monocytopoiesis
WO2019090263A1 (en) 2017-11-06 2019-05-09 Genentech, Inc. Diagnostic and therapeutic methods for cancer
WO2019128918A1 (en) 2017-12-29 2019-07-04 广州再极医药科技有限公司 Aromatic vinyl or aromatic ethyl derivative, preparation method therefor, intermediate, pharmaceutical composition, and application
WO2019139921A1 (en) 2018-01-09 2019-07-18 Shuttle Pharmaceuticals, Inc. Selective histone deacetylase inhibitors for the treatment of human disease
WO2019165374A1 (en) 2018-02-26 2019-08-29 Gilead Sciences, Inc. Substituted pyrrolizine compounds as hbv replication inhibitors
US10415015B2 (en) 2016-10-31 2019-09-17 Iovance Biotherapeutics, Inc. Engineered artificial antigen presenting cells for tumor infiltrating lymphocyte expansion
CN110305200A (en) * 2018-03-20 2019-10-08 上海大学 A kind of little albumen and its application
WO2019195181A1 (en) 2018-04-05 2019-10-10 Gilead Sciences, Inc. Antibodies and fragments thereof that bind hepatitis b virus protein x
WO2019193542A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotides
WO2019193543A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotides
WO2019193533A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'2'-cyclic dinucleotides
WO2019200247A1 (en) 2018-04-12 2019-10-17 Precision Biosciences, Inc. Optimized engineered meganucleases having specificity for a recognition sequence in the hepatitis b virus genome
WO2019211799A1 (en) 2018-05-03 2019-11-07 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotide analogue comprising a cyclopentanyl modified nucleotide
WO2019232319A1 (en) 2018-05-31 2019-12-05 Peloton Therapeutics, Inc. Compositions and methods for inhibiting cd73
WO2019246557A1 (en) 2018-06-23 2019-12-26 Genentech, Inc. Methods of treating lung cancer with a pd-1 axis binding antagonist, a platinum agent, and a topoisomerase ii inhibitor
WO2020018789A1 (en) 2018-07-18 2020-01-23 Genentech, Inc. Methods of treating lung cancer with a pd-1 axis binding antagonist, an antimetabolite, and a platinum agent
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
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
WO2020051099A1 (en) 2018-09-03 2020-03-12 Genentech, Inc. Carboxamide and sulfonamide derivatives useful as tead modulators
WO2020061429A1 (en) 2018-09-20 2020-03-26 Iovance Biotherapeutics, Inc. Expansion of tils from cryopreserved tumor samples
US10618916B2 (en) 2018-05-11 2020-04-14 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2020088357A1 (en) 2018-11-02 2020-05-07 上海再极医药科技有限公司 Diphenyl-like compound, intermediate thereof, preparation method therefor, pharmaceutical composition thereof and uses thereof
WO2020092621A1 (en) 2018-10-31 2020-05-07 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds as hpk1 inhibitors
WO2020092528A1 (en) 2018-10-31 2020-05-07 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds having hpk1 inhibitory activity
WO2020096682A2 (en) 2018-08-31 2020-05-14 Iovance Biotherapeutics, Inc. Treatment of nsclc patients refractory for anti-pd-1 antibody
US10662416B2 (en) 2016-10-14 2020-05-26 Precision Biosciences, Inc. Engineered meganucleases specific for recognition sequences in the hepatitis B virus genome
US10669271B2 (en) 2018-03-30 2020-06-02 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2020123453A2 (en) 2018-12-11 2020-06-18 Theravance Biopharma R&D Ip, Llc Alk5 inhibitors
WO2020141199A1 (en) 2019-01-03 2020-07-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for enhancing cd8+ t cell-dependent immune responses in subjects suffering from cancer
WO2020150152A1 (en) 2019-01-14 2020-07-23 Genentech, Inc. Methods of treating cancer with a pd-1 axis binding antagonist and an rna vaccine
US10744118B2 (en) 2012-12-07 2020-08-18 Chemocentryx, Inc. Diazole lactams
WO2020178770A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotides and prodrugs thereof
WO2020178768A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotide analogue comprising a cyclopentanyl modified nucleotide as sting modulator
WO2020178769A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotides and prodrugs thereof
US10793565B2 (en) 2016-12-22 2020-10-06 Incyte Corporation Heterocyclic compounds as immunomodulators
US10800768B2 (en) 2016-12-22 2020-10-13 Incyte Corporation Heterocyclic compounds as immunomodulators
US10806785B2 (en) 2016-12-22 2020-10-20 Incyte Corporation Immunomodulator compounds and methods of use
WO2020214995A1 (en) 2019-04-19 2020-10-22 Genentech, Inc. Anti-mertk antibodies and their methods of use
WO2020214663A1 (en) 2019-04-17 2020-10-22 Gilead Sciences, Inc. Solid forms of a toll-like receptor modulator
WO2020214652A1 (en) 2019-04-17 2020-10-22 Gilead Sciences, Inc. Solid forms of a toll-like receptor modulator
WO2020237025A1 (en) 2019-05-23 2020-11-26 Gilead Sciences, Inc. Substituted exo-methylene-oxindoles which are hpk1/map4k1 inhibitors
EP3747472A1 (en) 2015-09-15 2020-12-09 Acerta Pharma B.V. Therapeutic combinations of a cd19 inhibitor and a btk inhibitor
WO2021034804A1 (en) 2019-08-19 2021-02-25 Gilead Sciences, Inc. Pharmaceutical formulations of tenofovir alafenamide
US10966999B2 (en) 2017-12-20 2021-04-06 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3′3′ cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
WO2021067181A1 (en) 2019-09-30 2021-04-08 Gilead Sciences, Inc. Hbv vaccines and methods treating hbv
WO2021097110A1 (en) 2019-11-13 2021-05-20 Genentech, Inc. Therapeutic compounds and methods of use
WO2021102468A1 (en) 2019-11-22 2021-05-27 Theravance Biopharma R&D Ip, Llc Substituted 1,5-naphthyridines or quinolines as alk5 inhibitors
WO2021113765A1 (en) 2019-12-06 2021-06-10 Precision Biosciences, Inc. Optimized engineered meganucleases having specificity for a recognition sequence in the hepatitis b virus genome
US11034667B2 (en) 2017-01-09 2021-06-15 Shuttle Pharmaceuticals, Inc. Selective histone deacetylase inhibitors for the treatment of human disease
WO2021136354A1 (en) 2020-01-03 2021-07-08 上海翰森生物医药科技有限公司 Biphenyl derivative inhibitor, preparation method therefor and use thereof
WO2021155149A1 (en) 2020-01-31 2021-08-05 Genentech, Inc. Methods of inducing neoepitope-specific t cells with a pd-1 axis binding antagonist and an rna vaccine
WO2021188959A1 (en) 2020-03-20 2021-09-23 Gilead Sciences, Inc. Prodrugs of 4'-c-substituted-2-halo-2'-deoxyadenosine nucleosides and methods of making and using the same
WO2021203131A1 (en) 2020-03-31 2021-10-07 Theravance Biopharma R&D Ip, Llc Substituted pyrimidines and methods of use
WO2021216920A1 (en) 2020-04-22 2021-10-28 Iovance Biotherapeutics, Inc. Systems and methods for coordinating manufacturing of cells for patient-specific immunotherapy
WO2021253041A1 (en) 2020-06-10 2021-12-16 Theravance Biopharma R&D Ip, Llc Naphthyridine derivatives useful as alk5 inhibitors
US11203610B2 (en) 2017-12-20 2021-12-21 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2′3′ cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
WO2022020716A1 (en) 2020-07-24 2022-01-27 Genentech, Inc. Heterocyclic inhibitors of tead for treating cancer
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
WO2022086957A1 (en) 2020-10-20 2022-04-28 Genentech, Inc. Peg-conjugated anti-mertk antibodies and methods of use
WO2022094567A1 (en) 2020-10-28 2022-05-05 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
WO2022119830A1 (en) 2020-12-02 2022-06-09 Genentech, Inc. Methods and compositions for neoadjuvant and adjuvant urothelial carcinoma therapy
US11401279B2 (en) 2019-09-30 2022-08-02 Incyte Corporation Pyrido[3,2-d]pyrimidine compounds as immunomodulators
US11407749B2 (en) 2015-10-19 2022-08-09 Incyte Corporation Heterocyclic compounds as immunomodulators
US11465981B2 (en) 2016-12-22 2022-10-11 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2022232503A1 (en) 2021-04-30 2022-11-03 Genentech, Inc. 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
WO2022251359A1 (en) 2021-05-26 2022-12-01 Theravance Biopharma R&D Ip, Llc Bicyclic inhibitors of alk5 and methods of use
WO2022261301A1 (en) 2021-06-11 2022-12-15 Gilead Sciences, Inc. Combination mcl-1 inhibitors with anti-cancer agents
WO2022261310A1 (en) 2021-06-11 2022-12-15 Gilead Sciences, Inc. Combination mcl-1 inhibitors with anti-body drug conjugates
US11535615B2 (en) 2015-12-22 2022-12-27 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2022271659A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271684A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271650A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271677A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2023279092A2 (en) 2021-07-02 2023-01-05 Genentech, Inc. Methods and compositions for treating cancer
WO2023010094A2 (en) 2021-07-28 2023-02-02 Genentech, Inc. Methods and compositions for treating cancer
WO2023010095A1 (en) 2021-07-28 2023-02-02 F. Hoffmann-La Roche Ag Methods and compositions for treating cancer
US11572366B2 (en) 2015-11-19 2023-02-07 Incyte Corporation Heterocyclic compounds as immunomodulators
US11584733B2 (en) 2017-01-09 2023-02-21 Shuttle Pharmaceuticals, Inc. Selective histone deacetylase inhibitors for the treatment of human disease
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
WO2023051926A1 (en) 2021-09-30 2023-04-06 BioNTech SE Treatment involving non-immunogenic rna for antigen vaccination and pd-1 axis binding antagonists
WO2023056403A1 (en) 2021-09-30 2023-04-06 Genentech, Inc. 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
WO2023097195A1 (en) 2021-11-24 2023-06-01 Genentech, Inc. Therapeutic indazole compounds and methods of use in the treatment of cancer
WO2023097194A2 (en) 2021-11-24 2023-06-01 Genentech, Inc. Therapeutic compounds and methods of use
US11673883B2 (en) 2016-05-26 2023-06-13 Incyte Corporation 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
WO2023191816A1 (en) 2022-04-01 2023-10-05 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
US11780836B2 (en) 2020-11-06 2023-10-10 Incyte Corporation Process of preparing a PD-1/PD-L1 inhibitor
WO2023219613A1 (en) 2022-05-11 2023-11-16 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
WO2023240058A2 (en) 2022-06-07 2023-12-14 Genentech, Inc. Prognostic and therapeutic methods for cancer
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
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
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
WO2024091991A1 (en) 2022-10-25 2024-05-02 Genentech, Inc. Therapeutic and diagnostic methods for multiple myeloma
US11981921B2 (en) 2022-04-15 2024-05-14 Iovance Biotherapeutics, Inc. TIL expansion processes using specific cytokine combinations and/or AKTi treatment

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012168944A1 (en) * 2011-06-08 2012-12-13 Aurigene Discovery Technologies Limited Therapeutic compounds for immunomodulation
CN103096915A (en) * 2010-06-25 2013-05-08 奥瑞基尼探索技术有限公司 Immunosuppression modulating compounds
WO2013132317A1 (en) * 2012-03-07 2013-09-12 Aurigene Discovery Technologies Limited Peptidomimetic compounds as immunomodulators

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103096915A (en) * 2010-06-25 2013-05-08 奥瑞基尼探索技术有限公司 Immunosuppression modulating compounds
WO2012168944A1 (en) * 2011-06-08 2012-12-13 Aurigene Discovery Technologies Limited Therapeutic compounds for immunomodulation
WO2013132317A1 (en) * 2012-03-07 2013-09-12 Aurigene Discovery Technologies Limited Peptidomimetic compounds as immunomodulators

Cited By (133)

* 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
EP3747472A1 (en) 2015-09-15 2020-12-09 Acerta Pharma B.V. Therapeutic combinations of a cd19 inhibitor and a btk inhibitor
US11407749B2 (en) 2015-10-19 2022-08-09 Incyte Corporation Heterocyclic compounds as immunomodulators
US11572366B2 (en) 2015-11-19 2023-02-07 Incyte Corporation Heterocyclic compounds as immunomodulators
US11866435B2 (en) 2015-12-22 2024-01-09 Incyte Corporation Heterocyclic compounds as immunomodulators
US11535615B2 (en) 2015-12-22 2022-12-27 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2017122175A1 (en) 2016-01-13 2017-07-20 Acerta Pharma B.V. Therapeutic combinations of an antifolate and a btk inhibitor
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
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
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
WO2018006795A1 (en) 2016-07-05 2018-01-11 广州再极医药科技有限公司 Aromatic acetylene or aromatic ethylene compound, intermediate, preparation method, pharmaceutical composition and use thereof
US11078192B2 (en) 2016-07-05 2021-08-03 Guangzhou Maxinovel Pharmaceuticals Co., Ltd. Aromatic acetylene or aromatic ethylene compound, intermediate, preparation method, pharmaceutical composition and use thereof
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
WO2018055080A1 (en) 2016-09-22 2018-03-29 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
US11274285B2 (en) 2016-10-14 2022-03-15 Precision Biosciences, Inc. Engineered meganucleases specific for recognition sequences in the Hepatitis B virus genome
US10662416B2 (en) 2016-10-14 2020-05-26 Precision Biosciences, Inc. Engineered meganucleases specific for recognition sequences in the hepatitis B virus genome
US10415015B2 (en) 2016-10-31 2019-09-17 Iovance Biotherapeutics, Inc. Engineered artificial antigen presenting cells for tumor infiltrating lymphocyte expansion
US11667890B2 (en) 2016-10-31 2023-06-06 Iovance Biotherapeutics, Inc. Engineered artificial antigen presenting cells for tumor infiltrating lymphocyte expansion
US10800768B2 (en) 2016-12-22 2020-10-13 Incyte Corporation Heterocyclic compounds as immunomodulators
US10793565B2 (en) 2016-12-22 2020-10-06 Incyte Corporation Heterocyclic compounds as immunomodulators
US10806785B2 (en) 2016-12-22 2020-10-20 Incyte Corporation Immunomodulator compounds and methods of use
US11566026B2 (en) 2016-12-22 2023-01-31 Incyte Corporation Heterocyclic compounds as immunomodulators
US11339149B2 (en) 2016-12-22 2022-05-24 Incyte Corporation Heterocyclic compounds as immunomodulators
US11787793B2 (en) 2016-12-22 2023-10-17 Incyte Corporation Heterocyclic compounds as immunomodulators
US11465981B2 (en) 2016-12-22 2022-10-11 Incyte Corporation Heterocyclic compounds as immunomodulators
US11034667B2 (en) 2017-01-09 2021-06-15 Shuttle Pharmaceuticals, Inc. Selective histone deacetylase inhibitors for the treatment of human disease
EP4046989A1 (en) 2017-01-09 2022-08-24 Shuttle Pharmaceuticals, Inc. Selective histone deacetylase inhibitors for the treatment of human disease
US11584733B2 (en) 2017-01-09 2023-02-21 Shuttle Pharmaceuticals, Inc. Selective histone deacetylase inhibitors for the treatment of human disease
WO2019020593A1 (en) 2017-07-25 2019-01-31 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for modulating monocytopoiesis
WO2019090263A1 (en) 2017-11-06 2019-05-09 Genentech, Inc. Diagnostic and therapeutic methods for cancer
US11203610B2 (en) 2017-12-20 2021-12-21 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2′3′ cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
US10966999B2 (en) 2017-12-20 2021-04-06 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3′3′ cyclic dinucleotides with phosphonate bond activating the sting adaptor protein
US11384048B2 (en) 2017-12-29 2022-07-12 Guangzhou Maxinovel Pharmaceuticals Co., Ltd. Aromatic vinyl or aromatic ethyl derivative, preparation method therefor, intermediate, pharmaceutical composition, and application
WO2019128918A1 (en) 2017-12-29 2019-07-04 广州再极医药科技有限公司 Aromatic vinyl or aromatic ethyl derivative, preparation method therefor, intermediate, pharmaceutical composition, and application
WO2019139921A1 (en) 2018-01-09 2019-07-18 Shuttle Pharmaceuticals, Inc. Selective histone deacetylase inhibitors for the treatment of human disease
WO2019165374A1 (en) 2018-02-26 2019-08-29 Gilead Sciences, Inc. Substituted pyrrolizine compounds as hbv replication inhibitors
CN110305200B (en) * 2018-03-20 2021-02-26 上海大学 Small protein and application thereof
CN110305200A (en) * 2018-03-20 2019-10-08 上海大学 A kind of little albumen and its application
US10669271B2 (en) 2018-03-30 2020-06-02 Incyte Corporation Heterocyclic compounds as immunomodulators
US11124511B2 (en) 2018-03-30 2021-09-21 Incyte Corporation 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
US11149052B2 (en) 2018-04-06 2021-10-19 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2′3′-cyclic dinucleotides
WO2019193542A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotides
WO2019193543A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotides
US11292812B2 (en) 2018-04-06 2022-04-05 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3′3′-cyclic dinucleotides
WO2019193533A1 (en) 2018-04-06 2019-10-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'2'-cyclic dinucleotides
US11788077B2 (en) 2018-04-12 2023-10-17 Precision Biosciences, Inc. Polynucleotides encoding optimized engineered meganucleases having specificity for a recognition sequence in the Hepatitis B virus genome
WO2019200247A1 (en) 2018-04-12 2019-10-17 Precision Biosciences, Inc. Optimized engineered meganucleases having specificity for a recognition sequence in the hepatitis b virus genome
US11142750B2 (en) 2018-04-12 2021-10-12 Precision Biosciences, Inc. Optimized engineered meganucleases having specificity for a recognition sequence in the Hepatitis B virus genome
WO2019211799A1 (en) 2018-05-03 2019-11-07 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotide analogue comprising a cyclopentanyl modified nucleotide
US10906920B2 (en) 2018-05-11 2021-02-02 Incyte Corporation Heterocyclic compounds as immunomodulators
US10618916B2 (en) 2018-05-11 2020-04-14 Incyte Corporation Heterocyclic compounds as immunomodulators
US11414433B2 (en) 2018-05-11 2022-08-16 Incyte Corporation Heterocyclic compounds as immunomodulators
WO2019232319A1 (en) 2018-05-31 2019-12-05 Peloton Therapeutics, Inc. Compositions and methods for inhibiting cd73
WO2019246557A1 (en) 2018-06-23 2019-12-26 Genentech, Inc. Methods of treating lung cancer with a pd-1 axis binding antagonist, a platinum agent, and a topoisomerase ii inhibitor
WO2020018789A1 (en) 2018-07-18 2020-01-23 Genentech, Inc. Methods of treating lung cancer with a pd-1 axis binding antagonist, an antimetabolite, and a platinum agent
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
WO2020096682A2 (en) 2018-08-31 2020-05-14 Iovance Biotherapeutics, Inc. Treatment of nsclc patients refractory for anti-pd-1 antibody
WO2020051099A1 (en) 2018-09-03 2020-03-12 Genentech, Inc. Carboxamide and sulfonamide derivatives useful as tead modulators
WO2020061429A1 (en) 2018-09-20 2020-03-26 Iovance Biotherapeutics, Inc. Expansion of tils from cryopreserved tumor samples
EP4371987A1 (en) 2018-10-31 2024-05-22 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds as hpk1 inhibitors
WO2020092528A1 (en) 2018-10-31 2020-05-07 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds having hpk1 inhibitory activity
WO2020092621A1 (en) 2018-10-31 2020-05-07 Gilead Sciences, Inc. Substituted 6-azabenzimidazole compounds as hpk1 inhibitors
WO2020088357A1 (en) 2018-11-02 2020-05-07 上海再极医药科技有限公司 Diphenyl-like compound, intermediate thereof, preparation method therefor, pharmaceutical composition thereof and uses thereof
WO2020123453A2 (en) 2018-12-11 2020-06-18 Theravance Biopharma R&D Ip, Llc Alk5 inhibitors
EP4059569A1 (en) 2019-01-03 2022-09-21 Institut National De La Sante Et De La Recherche Medicale (Inserm) Methods and pharmaceutical compositions for enhancing cd8+ t cell-dependent immune responses in subjects suffering from cancer
WO2020141199A1 (en) 2019-01-03 2020-07-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for enhancing cd8+ t cell-dependent immune responses in subjects suffering from cancer
WO2020150152A1 (en) 2019-01-14 2020-07-23 Genentech, Inc. Methods of treating cancer with a pd-1 axis binding antagonist and an rna vaccine
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
WO2020178770A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotides and prodrugs thereof
WO2020178769A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 2'3'-cyclic dinucleotides and prodrugs thereof
WO2020178768A1 (en) 2019-03-07 2020-09-10 Institute Of Organic Chemistry And Biochemistry Ascr, V.V.I. 3'3'-cyclic dinucleotide analogue comprising a cyclopentanyl modified nucleotide as sting modulator
WO2020214663A1 (en) 2019-04-17 2020-10-22 Gilead Sciences, Inc. Solid forms of a toll-like receptor modulator
WO2020214652A1 (en) 2019-04-17 2020-10-22 Gilead Sciences, Inc. Solid forms of a toll-like receptor modulator
WO2020214995A1 (en) 2019-04-19 2020-10-22 Genentech, Inc. Anti-mertk antibodies and their methods of use
WO2020237025A1 (en) 2019-05-23 2020-11-26 Gilead Sciences, Inc. Substituted exo-methylene-oxindoles which are hpk1/map4k1 inhibitors
US11753406B2 (en) 2019-08-09 2023-09-12 Incyte Corporation Salts of a PD-1/PD-L1 inhibitor
WO2021034804A1 (en) 2019-08-19 2021-02-25 Gilead Sciences, Inc. Pharmaceutical formulations of tenofovir alafenamide
US11401279B2 (en) 2019-09-30 2022-08-02 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
US11866451B2 (en) 2019-11-11 2024-01-09 Incyte Corporation Salts and crystalline forms of a PD-1/PD-L1 inhibitor
WO2021097110A1 (en) 2019-11-13 2021-05-20 Genentech, Inc. Therapeutic compounds and methods of use
WO2021102468A1 (en) 2019-11-22 2021-05-27 Theravance Biopharma R&D Ip, Llc Substituted 1,5-naphthyridines or quinolines as alk5 inhibitors
WO2021113765A1 (en) 2019-12-06 2021-06-10 Precision Biosciences, Inc. Optimized engineered meganucleases having specificity for a recognition sequence in the hepatitis b virus genome
WO2021136354A1 (en) 2020-01-03 2021-07-08 上海翰森生物医药科技有限公司 Biphenyl derivative inhibitor, preparation method therefor and use thereof
WO2021155149A1 (en) 2020-01-31 2021-08-05 Genentech, Inc. Methods of inducing neoepitope-specific t cells with a pd-1 axis binding antagonist and an rna vaccine
WO2021188959A1 (en) 2020-03-20 2021-09-23 Gilead Sciences, Inc. Prodrugs of 4'-c-substituted-2-halo-2'-deoxyadenosine nucleosides and methods of making and using the same
WO2021203131A1 (en) 2020-03-31 2021-10-07 Theravance Biopharma R&D Ip, Llc Substituted pyrimidines and methods of use
WO2021216920A1 (en) 2020-04-22 2021-10-28 Iovance Biotherapeutics, Inc. Systems and methods for coordinating manufacturing of cells for patient-specific immunotherapy
WO2021253041A1 (en) 2020-06-10 2021-12-16 Theravance Biopharma R&D Ip, Llc Naphthyridine derivatives useful as alk5 inhibitors
WO2022020716A1 (en) 2020-07-24 2022-01-27 Genentech, Inc. Heterocyclic inhibitors of tead for treating cancer
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
WO2022086957A1 (en) 2020-10-20 2022-04-28 Genentech, Inc. Peg-conjugated anti-mertk antibodies and methods of use
WO2022093981A1 (en) 2020-10-28 2022-05-05 Genentech, Inc. Combination therapy comprising ptpn22 inhibitors and pd-l1 binding antagonists
WO2022094567A1 (en) 2020-10-28 2022-05-05 Ikena Oncology, Inc. Combination of an ahr inhibitor with a pdx inhibitor or doxorubicine
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
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
WO2022119830A1 (en) 2020-12-02 2022-06-09 Genentech, Inc. Methods and compositions for neoadjuvant and adjuvant urothelial carcinoma therapy
WO2022232503A1 (en) 2021-04-30 2022-11-03 Genentech, Inc. 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
WO2022251359A1 (en) 2021-05-26 2022-12-01 Theravance Biopharma R&D Ip, Llc Bicyclic inhibitors of alk5 and methods of use
US11957693B2 (en) 2021-06-11 2024-04-16 Gilead Sciences, Inc. Combination MCL-1 inhibitors with anti-cancer agents
US11931424B2 (en) 2021-06-11 2024-03-19 Gilead Sciences, Inc. Combination MCL-1 inhibitors with anti-body drug conjugates
WO2022261310A1 (en) 2021-06-11 2022-12-15 Gilead Sciences, Inc. Combination mcl-1 inhibitors with anti-body drug conjugates
WO2022261301A1 (en) 2021-06-11 2022-12-15 Gilead Sciences, Inc. Combination mcl-1 inhibitors with anti-cancer agents
WO2022271659A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271684A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271650A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2022271677A1 (en) 2021-06-23 2022-12-29 Gilead Sciences, Inc. Diacylglyercol kinase modulating compounds
WO2023279092A2 (en) 2021-07-02 2023-01-05 Genentech, Inc. Methods and compositions for treating cancer
WO2023010095A1 (en) 2021-07-28 2023-02-02 F. Hoffmann-La Roche Ag Methods and compositions for treating cancer
WO2023010094A2 (en) 2021-07-28 2023-02-02 Genentech, Inc. Methods and compositions for treating cancer
WO2023056403A1 (en) 2021-09-30 2023-04-06 Genentech, Inc. Methods for treatment of hematologic cancers using anti-tigit antibodies, anti-cd38 antibodies, and pd-1 axis binding antagonists
WO2023052531A1 (en) 2021-09-30 2023-04-06 BioNTech SE Treatment involving non-immunogenic rna for antigen vaccination and pd-1 axis binding antagonists
WO2023051926A1 (en) 2021-09-30 2023-04-06 BioNTech SE Treatment involving non-immunogenic rna for antigen vaccination 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
WO2023097194A2 (en) 2021-11-24 2023-06-01 Genentech, Inc. Therapeutic compounds and methods of use
WO2023097195A1 (en) 2021-11-24 2023-06-01 Genentech, Inc. Therapeutic indazole compounds and methods of use in the treatment of cancer
WO2023191816A1 (en) 2022-04-01 2023-10-05 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
US11981921B2 (en) 2022-04-15 2024-05-14 Iovance Biotherapeutics, Inc. TIL expansion processes using specific cytokine combinations and/or AKTi treatment
WO2023219613A1 (en) 2022-05-11 2023-11-16 Genentech, Inc. Dosing for treatment with anti-fcrh5/anti-cd3 bispecific antibodies
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
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
WO2024091991A1 (en) 2022-10-25 2024-05-02 Genentech, Inc. Therapeutic and diagnostic methods for multiple myeloma

Similar Documents

Publication Publication Date Title
US10961205B2 (en) 1,2,4-oxadiazole derivatives as immunomodulators
US10160736B2 (en) 1,3,4-oxadiazole and 1,3,4-thiadiazole derivatives as immunomodulators
US10106581B2 (en) Cyclic peptidomimetic compounds as immunomodulators
WO2015036927A1 (en) Immunomodulating peptidomimetic derivatives
WO2015044900A1 (en) Therapeutic immunomodulating compounds
NZ718711B2 (en) 1,2,4-oxadiazole derivatives as immunomodulators

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14843752

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14843752

Country of ref document: EP

Kind code of ref document: A1