WO2011045340A1 - Ido inhibitors and therapeutic uses thereof - Google Patents

Ido inhibitors and therapeutic uses thereof Download PDF

Info

Publication number
WO2011045340A1
WO2011045340A1 PCT/EP2010/065336 EP2010065336W WO2011045340A1 WO 2011045340 A1 WO2011045340 A1 WO 2011045340A1 EP 2010065336 W EP2010065336 W EP 2010065336W WO 2011045340 A1 WO2011045340 A1 WO 2011045340A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
formula
compound
pharmaceutically acceptable
ido
Prior art date
Application number
PCT/EP2010/065336
Other languages
French (fr)
Inventor
Ute Roehrig
Awad Loay
Olivier Michielin
Benoit Van Den Eynde
Luc Pilotte
Vincent Stroobant
Pierre Larrieu
Pierre Vogel
Vincent Zoete
Original Assignee
Ludwig Institute For Cancer Research Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ludwig Institute For Cancer Research Ltd filed Critical Ludwig Institute For Cancer Research Ltd
Publication of WO2011045340A1 publication Critical patent/WO2011045340A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • This invention relates to inhibitors. of indoleamine 2,3-dioxygenase (IDO) and their use in the treatment of cancer or infections, either alone or in combination with additional therapeutic agents,
  • IDO indoleamine 2,3-dioxygenase
  • IDO indoleamine 2,3-dioxygenase
  • R 1 represents hydrogen or cyclohexyl
  • R 2 represents hydrogen or alkyl C 1-6
  • R 1 and R 2 do not both simultaneously represent hydrogen
  • the compound of formula IA may be made by reductive amination of a compound of formula i in which Rt represents H with cyclohexanone and a hydride reducing agent, for example, NaBH(OAc)j, in a solvent which is inert to the reactive conditions, such as a polar, aprotic solvent, for example, dicioroethane.
  • Salts of compounds of formula I may be formed by reacting the free acid, or a salt thereof, with one or more equivalents of the appropriate base. The reaction may be carried out in a solvent or medium in which the salt is insoluble or in a solvent in which the salt is soluble, e.g. ethanol, tetrahydrofuran or diethyl ether, which may be removed in vacuo, or by freeze drying.
  • the reaction may also be a metathetical process or it may be carried out on an ion exchange resin.
  • Pharmaceutically acceptable salts of the compounds of formula 1 thereof include salts with strong acids, e.g., HC1, HBr, etc, and salts with weak acids, eg organic acids, for example carboxylic acids, such as acetic acid, benzoic acids, as well as sulphonic acids.
  • Non-toxic physiologically acceptable salts are preferred, although other salts are also useful, e.g. in isolating or purifying the product.
  • the compounds of formula I may be used alone or in combination with at least one additional therapeutic agent.
  • the at least one additional therapeutic agent may be an antineoplastic chemotherapy agent.
  • Suitable antineoplastic chemotherapeutic agent is selected from the group consisting of cyclophosphamide, methotrexate, fluorouracil, doxorubicin, vincristine, ifosfamide, cisplatin, gemcytabine, busulfan, ara-C, and combinations thereof.
  • the at least one additional therapeutic agent may be radiation therapy.
  • the radiation therapy may be localized radiation therapy delivered to the tumour or may be total body Irradiation.
  • the compounds of the invention may be used as an adjuvant to the therapeutic vaccination of various cancers.
  • Cancers that may be mentioned include melanoma, colon cancer, pancreatic cancer, breast cancer, prostate cancer, lung cancer, ieukemia, brain tumours, lymphoma, sarcoma, ovarian cancer, and Kaposi's sarcoma.
  • cancers and tumours include adrenocorticocancer, basal cell carcinoma, bladder cancer, bowel cancer, brain and CK!S tumors, breast cancers, B- cell lymphoma, carcinoid tumours, cervical cancer, childhood cancers, chondrosarcoma, choriocarcinoma, chronic myeloid leukemia, rectal cancers, endocrine cancers, endometrial cancer, esophageal cancer, Ewing's sarcoma, eye cancer, gastric cancer or carcinoma, gastrointestinal cancers, genitourinary cancers, glioma, gynecological cancers, head and neck cancers, hepatocellular cancer, Hodgkins disease, hypopharynx cancer, islet cell cancer, kidney cancer, laryngeal cancer, liver cancer, lung cancer (including small-cell lung carcinoma and non-small-cell carcinoma), lymphoma, male breast cancer, melanoma, mesothelioma, multiple
  • IDO In addition to cancers, IDO plays a role in several diseases, including Clamydia psittaci infection and Streptococcus pyogenes infection, systemic lupus erythematosus, rheumatoid arthritis, Alzheimer's disease, Huntington's disease, Parkinson's disease, lyme neuroborreliosis, late lyme encephalopathy, Tourette's syndrome, systemic sclerosis, multiple sclerosis, coronary heart disease, T-cell mediated immune diseases, chronic infections (viral, bacterial, fungal and microbial), depression, neurological disorders, cancer tumors, and cataracts. Inhibitors of IDO may be used to treat these diseases.
  • IDO inhibitors may be used to treat include, but are not limited to, human immunodeficiency virus (HIV) and AIDS-reiated cancers.
  • HIV human immunodeficiency virus
  • the compounds may also be used as adjuvants to bone marrow transplantation or peripheral blood stem cell transplantation and in immunotherapy by adoptive transfer,
  • the infection may be selected from the group consisting of a viral infection, infection with an intracellular parasite, and infection with an intracellular bacteria.
  • Particular viral infections include human immunodeficiency virus or cytomegalovirus
  • Particular intracellular parasite infections may be selected from the group consisting of Leishmania donovani, Leishmania tropica, Leishmania major. Leishmania aethiopica, Leishmania mexicana, Plasmodium falciparum, Plasmodium vivax, Plasmodium ovale, and Plasmodium malariae.
  • Particular intracellular bacterial infections may be selected from the group consisting of Mycobacterium leprae, Mycobacterium tuberculosis, Listeria monocytogenes, and Toxplasma gondii.
  • the at least one additional therapeutic agent may be a vaccine, for example, an anti-viral vaccine, a vaccine against HIV, a vaccine against tuberculosis, a vaccine against malaria.
  • the vaccine may also be a tumour vaccine or a melanoma vaccine.
  • the tumour vaccine comprises genetically modified tumour cells or genetically modified cell lines. In such cases, preferably the genetically modified tumour cells or genetically modified cell line has been transfected to express granulocyte-macrophage stimulating factor (GM-CSF),
  • GM-CSF granulocyte-macrophage stimulating factor
  • the vaccine may comprise one or more immunogenic peptides, preferably immunogenic peptides of cancer-testis antigens (CTAgs).
  • CTAgs cancer-testis antigens
  • CTAg proteins include MAGE, BAGE, GAGE, SSX, NY-ESO-1, LAGE, SCP, CTSP, CT7, CT8, CT9, CT10, CT 11, SAGE, OY-TES-1, NY-SAR-35 and NY-BR-1.
  • MAGE proteins include MAGE-A1, A3, A4, A5, A6, A8, A9, A10, A12, Bl , B2, B3, B4, C1, C2 and C3 proteins-
  • SSX proteins exist, including SSX1, SSX2, SSX3 and SSX5.
  • tumour vaccine may comprise dendritic cells.
  • the additional therapeutic agent may be a cytokine, for example a granulocyte- macrophage colony stimulating factor (GM-CSF) or flt3-Hgand.
  • GM-CSF granulocyte- macrophage colony stimulating factor
  • flt3-Hgand flt3-Hgand.
  • the compound of formula I or a pharmaceutically acceptable salt thereof is administered in an amount effective to increase the delayed type hypersensitivity reaction to tumour antigen, delay the time to relapse of post-transplant malignancy, increase relapse free survival time post-transplant, and/or increase long-term post- transplant survival.
  • the compound of formula I or a pharmaceutically acceptable salt thereof is administered prior to full hematopoetic reconstitution.
  • the compounds of formula I or a pharmaceutically acceptable salt thereof for use in the method will generally be administered in the form of a pharmaceutical composition.
  • a pharmaceutical composition including preferably less than 80% w/w, more preferably less than 50% w/w, e.g. 0.1 to 20%, of the compound of formula I or a pharmaceutically acceptable salt thereof, in admixture with a pharmaceutically acceptable diluent or carrier.
  • a pharmaceutically acceptable diluent or carrier preferably less than 80% w/w, more preferably less than 50% w/w, e.g. 0.1 to 20%, of the compound of formula I or a pharmaceutically acceptable salt thereof, in admixture with a pharmaceutically acceptable diluent or carrier.
  • the compounds of formula I or a pharmaceutically acceptable salt thereof is to be used in aqueous solution, e.g. for infusion, it may be necessary to incorporate other excipients, In particular there may be mentioned chelating or sequestering agents, antioxidants, tonicity adjusting agents, pH-modifying agents and buffering agents.
  • Solutions containing the compound of formula I or a pharmaceutically acceptable salt thereof may, if desired, be evaporated, e.g, by freeze drying or spray drying, to give a solid composition, which may be reconstituted prior to use.
  • the compound of formula 1 or a pharmaceutically acceptable salt thereof preferably is in a form having a mass median diameter of from 0.01 to ⁇ ⁇ m.
  • the compositions may also contain suitable preserving, stabilising and wetting agents, solubilisers, e.g. a water-soluble cellulose polymer such as hydroxypropyl methylce!lulose, or a water-soluble glycol such as propylene glycol, sweetening and colouring agents and flavourings. Where appropriate, the compositions may be formulated in sustained release form.
  • the content of the compound of formula I or a pharmaceutically acceptable salt thereof in a pharmaceutical composition is generally about 0.01 -about 99.9wt%, preferably about 0.1 -about 50wt%, relative to the entire preparation.
  • the dose of the compound of formula I or a pharmaceutically acceptable salt thereof is determined in consideration of age, body weight, general health condition, diet, administration time, administration method, clearance rate, combination of drugs, the level of disease for which the patient is under treatment then, and other factors.
  • the dose varies depending on the target disease, condition, subject of administration, administration method and the like, for oral administration as a therapeutic agent for the treatment of cancer in a patient suffering from such a disease is from 0.01 mg - 10 g, preferably 0.1 - 100 mg, is preferably administered in a single dose or in 2 or 3 portions per day.
  • the compound of formula I or a pharmaceutically acceptable salt thereof may be used at their normal therapeutic doses, e.g., as set out in pharmacopoeias or prescribing guides, such as the Physicians' Desk Reference (PDR).
  • the compound of formula I or a pharmaceutically acceptable salt thereof supplements the activity of the additional therapeutic agent(s) in a synergistic fashion, such that the additional therapeutic agent(s) can be administered at a lower dose than is normally used.
  • IR spectra were recorded on a Perkin Elmer Paragon 1000 FT-IR spectrometer. Mass spectra were recorded on a Nermag R 10- IOC in chemical ionisation mode. Electrospray mass analyses were recorded on a Finnigan MAT SSQ 7 IOC spectrometer in positive ionisation mode. 1 H-NMR spectra were recorded on a Bruker DPX-400 FT, Bruker ARX-400 FT, or Bruker AMX-600 spectrometer. All 1 H signal assignments were confirmed by COSY spectra.
  • the coding region for human IDO (Ala2-Gly403) was cloned into a derivative of plasmid pET9 (Novagen).
  • the recombinant plasmid, pETIDO encodes a histidine tag at the N-terminus of IDO.
  • the bacterial strain BL21 AI (Invitrogen) was used for overexpression of IDO and transformed with the plasmid pETIDO.
  • the transformed cells were grown on a rotary shaker at 37 C and 220 rpm to an OD600 of 1.2, in LB medium supplemented with kanamycin 25 ⁇ g/ml, L-tryptophan 50 ⁇ g/ml and bovine hemin (Sigma) 10 ⁇ .
  • the culture was cooled in a water/ice bath and supplemented again with L-tryptophan 50 ⁇ g/ml, bovine hemin 10 ⁇ .
  • the expression of his-tagged IDO was induced by the addition of 1% (w/v) arabinose. Induced cells were grown at 20 C, 60 rpm for 20 h. Cells (1 1 culture) were collected by centrifugation, resuspended in 40 ml of Mes 25 mM, KC1 150 mM, imidazole 10 mM, protease inhibitors (complete EDTA free, Roche Applied Science), pH 6.5, and disrupted with a French press. The extract was clarified by centrifugation and filtration on a 0.22 ⁇ filter.
  • the enzyme was purified by IMAC using Ni 2 ⁇ as ligand and an IMAC HITRAP column (5 ml; GE Healthcare). Briefly, the extract was loaded on the column with buffer Mes 25 mM, KC1 150 mM, imidazole 10 mM, pH 6.5. The column was washed with 50 ml of the same buffer with imidazole adjusted to 100 mM. Finally, the protein was eluted with Mes 25 mM, KC1 150 mM, EDTA 50 mM, pH 6.5. The buffer was then exchanged to Mes 25 mM, KCi 150 mM, pH 6.5 using a HITRAP desalting column (GE Healthcare). The purity of the enzyme was estimated to be > 95% based on SDS PAGE gel and Coomassie blue staining. The ratio of absorbance 404 nm/280 nm of the protein was around 1,9.
  • the compound of formula I ⁇ was synthesized from 4-amino-1-naphthol hydrochloride and cyclohexanone according to the reductive amination procedure to afford the hydrochloric salt as a white solid in 82% yield.
  • Mp 276-278 °C.
  • TLC R 0.42 (40% diethyl ether/pentane).
  • the enzymatic inhibition assays were performed as described by Takikawa et al. [Takikawa, O.; Kuroiwa, T.; Yamazaki, F.; Kido, R. J Biol Chem, 1988, 263, 2041- 2048.] with some modifications. Briefly, the reaction mixture (100 ⁇ ) contained potassium phosphate buffer (100 mM, pH 6.5) ascorbic acid (20 raM), catalase (200 units/ml), methylene blue (10 ⁇ ), purified recombinant IDO (l ng/ ⁇ l), and L- Trp (200 ⁇ ). The inhibitors were serially diluted ranging from 0.1 to 1000 ⁇ .
  • the reaction was carried out at 37 °C for 60 min and stopped by the addition of 30% (w/v) trichloroacetic acid (40 ⁇ ).
  • 30% (w/v) trichloroacetic acid 40 ⁇ .
  • the tubes were incubated at 50 °C for 30 min, followed by a centrifugation at 1OOOOg for 20 min.
  • 100 ⁇ of supernatant from each probe was transfered to another tube for HPLC analysis.
  • the mobile phase for HPLC measurements consisted of 50% sodium citrate buffer (40 mM, pH 2.25) and 50% methanol with 400 ⁇ SDS.
  • the flow rate through the S5-ODS1 column was chosen to be 1 ml/min, and kynurenine was detected at a wavelength of 365 nm.
  • a plasmid construct encoding murine IDO was transfected into mouse mastocytoma line P815B.
  • Mouse IDO shares a 62% sequence identity with human IDO, and the active site residues mentioned in the Introduction are 100% conserved.
  • a plasmid construct encoding human IDO was transfected into human HEK-293 cells, and clone 293-hIDO clone 17 [Uyttenhove et al] was selected and used in the assay.
  • the assay was performed in 96-well flat bottom plates seeded with 2 x 10 5 cells in a final volume of 200 ⁇ . To determine whether compounds were significant IDO inhibitors, the cells were incubated overnight at 37 °C in HBSS (Hanks Balanced Salt Solution, Invitrogen) supplemented with 50 ⁇ L-Trp and 2, 20 or 200 ⁇ of the compound. To determine the IC , the cells were incubated for 8 h at 37 °C in HBSS supplemented with 80 ⁇ L-Tryptophan and a titration of the compound ranging from 0.312 to 80 ⁇ or from 1.56 to 400 ⁇ .
  • the plates were then centrifuged for 10 min at 300 g, and 150 ⁇ l of the supernatant were collected.
  • the supernatant was analyzed by HPLC to measure the concentration of tryptophan and kynurenine.
  • HPLC analysis 50 ⁇ of supernatant were mixed with 500 ⁇ l acetonitrile to precipitate the proteins. After centrifugation, the supernatant was collected, concentrated on a speedvac, resuspended in a final volume of 100 ⁇ water and injected in the HPLC (CI 8 column). Trp was detected at an absorption wavelength of 280 nm, kynurenine at 360 nm.

Landscapes

  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Use use of the compounds of formula I, and pharmaceutically acceptable salts thereof (Formula I), in which R1 represents hydrogen or cyclohexyϊ and R2 represents hydrogen or alkyl C1-6 provided that R1 and R2 do not both simultaneously represent hydrogen; in the treatment of diseases in which inhibition of IDO plays a therapeutic role, eg in the treatment of cancer.

Description

IDO Inhibitors and Therapeutic uses thereof
This invention relates to inhibitors. of indoleamine 2,3-dioxygenase (IDO) and their use in the treatment of cancer or infections, either alone or in combination with additional therapeutic agents,
The heme-containing enzyme indoleamine 2,3-dioxygenase (IDO, EC 1.13.1 1.52) has been implicated in the establishment of pathological immune tolerance by tumors. IDO catalyzes the initial and rate-limiting step in the catabolism of tryptophan (Tip) along the kynurenine pathway. By depleting Trp locally, IDO blocks the proliferation of T lymphocytes, which are extremely sensitive to Trp shortage. The observation that many human tumors constitutively express IDO introduced the hypothesis that its inhibition could enhance the effectiveness of cancer immunotherapy. Results from in vitro and in vivo studies suggest that the efficacy of therapeutic vaccination of cancer patients may indeed be improved by concomitant administration of an IDO inhibitor.
According to the invention, we provide use of the compounds of formula I, and pharmaceutically acceptable salts thereof,
Figure imgf000003_0001
in which
R1 represents hydrogen or cyclohexyl and
R2 represents hydrogen or alkyl C1-6
provided that R1 and R2 do not both simultaneously represent hydrogen;
in the treatment of diseases in which inhibition of IDO plays a therapeutic role, particularly those conditions mentioned herein.
The compound of formula I in which R1 represents H is known and may be prepared by methods previously described in the literature, for example, Bunce et al, Journal of Organic Chemistry, 52, pp 4214-23 (1987), Soroka et al, Journal of Photochemistry and Photobiology A - Chemistry 64, pp 171-182 (1992) and Knolker et al, Synthesis- Stuttgart 1995, pp 397-408 (1995). The compound of formula I in which Rl is cyclohexyl has not previously been described in the literature.
Accordingly, we provide the compound of formula ΪΑ
Figure imgf000004_0001
and pharmaceutically acceptable salts thereof.
We also provide the use of compound IA and pharmaceutically acceptable salts thereof as a pharmaceutical. We also provide the use of compounds IB and pharmaceutical iy acceptable salts thereof as a pharmaceutical
Figure imgf000004_0002
in which R2 is as defined above and preferably represents C¾ The compound of formula IA may be made by reductive amination of a compound of formula i in which Rt represents H with cyclohexanone and a hydride reducing agent, for example, NaBH(OAc)j, in a solvent which is inert to the reactive conditions, such as a polar, aprotic solvent, for example, dicioroethane. Salts of compounds of formula I may be formed by reacting the free acid, or a salt thereof, with one or more equivalents of the appropriate base. The reaction may be carried out in a solvent or medium in which the salt is insoluble or in a solvent in which the salt is soluble, e.g. ethanol, tetrahydrofuran or diethyl ether, which may be removed in vacuo, or by freeze drying. The reaction may also be a metathetical process or it may be carried out on an ion exchange resin.
Pharmaceutically acceptable salts of the compounds of formula 1 thereof include salts with strong acids, e.g., HC1, HBr, etc, and salts with weak acids, eg organic acids, for example carboxylic acids, such as acetic acid, benzoic acids, as well as sulphonic acids. Non-toxic physiologically acceptable salts are preferred, although other salts are also useful, e.g. in isolating or purifying the product. The compounds of formula I may be used alone or in combination with at least one additional therapeutic agent.
The at least one additional therapeutic agent may be an antineoplastic chemotherapy agent. Suitable antineoplastic chemotherapeutic agent is selected from the group consisting of cyclophosphamide, methotrexate, fluorouracil, doxorubicin, vincristine, ifosfamide, cisplatin, gemcytabine, busulfan, ara-C, and combinations thereof.
Alternatively, the at least one additional therapeutic agent may be radiation therapy. The radiation therapy may be localized radiation therapy delivered to the tumour or may be total body Irradiation.
The compounds of the invention may be used as an adjuvant to the therapeutic vaccination of various cancers. Cancers that may be mentioned include melanoma, colon cancer, pancreatic cancer, breast cancer, prostate cancer, lung cancer, ieukemia, brain tumours, lymphoma, sarcoma, ovarian cancer, and Kaposi's sarcoma. Other cancers and tumours that may be mentioned include adrenocorticocancer, basal cell carcinoma, bladder cancer, bowel cancer, brain and CK!S tumors, breast cancers, B- cell lymphoma, carcinoid tumours, cervical cancer, childhood cancers, chondrosarcoma, choriocarcinoma, chronic myeloid leukemia, rectal cancers, endocrine cancers, endometrial cancer, esophageal cancer, Ewing's sarcoma, eye cancer, gastric cancer or carcinoma, gastrointestinal cancers, genitourinary cancers, glioma, gynecological cancers, head and neck cancers, hepatocellular cancer, Hodgkins disease, hypopharynx cancer, islet cell cancer, kidney cancer, laryngeal cancer, liver cancer, lung cancer (including small-cell lung carcinoma and non-small-cell carcinoma), lymphoma, male breast cancer, melanoma, mesothelioma, multiple myeloma, nasopharyngeal cancer, neuroblastoma, non-Hodgkins lymphoma, non-melanoma skin cancer, osteosarcoma, ovarian cancer, pancreas cancer, pituitary cancer, prostate cancer, renai cell carcinoma, retinoblastoma,, rhabdomyosarcoma, sarcoma, skin cancer, squamous cell carcinoma, stomach cancer, testicular cancer or seminoma, thymus cancer, thyroid cancer, transitional cell cancer, trophoblastic cancer, uterine cancer, vaginal cancer, Waldenstrom's macroglobulinemia, and Wilm' s tumor, colorectum, cervix, endometrium, ovary, testis, mesothelial lining, white blood cell (including lymphoma and leukemia) esophagus, muscle, connective tissue, adrenal gland, bone, glioblastoma, and cutaneous basocelluiar carcinoma.
In addition to cancers, IDO plays a role in several diseases, including Clamydia psittaci infection and Streptococcus pyogenes infection, systemic lupus erythematosus, rheumatoid arthritis, Alzheimer's disease, Huntington's disease, Parkinson's disease, lyme neuroborreliosis, late lyme encephalopathy, Tourette's syndrome, systemic sclerosis, multiple sclerosis, coronary heart disease, T-cell mediated immune diseases, chronic infections (viral, bacterial, fungal and microbial), depression, neurological disorders, cancer tumors, and cataracts. Inhibitors of IDO may be used to treat these diseases. Other diseases that IDO inhibitors may be used to treat include, but are not limited to, human immunodeficiency virus (HIV) and AIDS-reiated cancers. The compounds may also be used as adjuvants to bone marrow transplantation or peripheral blood stem cell transplantation and in immunotherapy by adoptive transfer,
Where the compounds are used in the treatment of an infection, the infection may be selected from the group consisting of a viral infection, infection with an intracellular parasite, and infection with an intracellular bacteria.
Particular viral infections include human immunodeficiency virus or cytomegalovirus, Particular intracellular parasite infections may be selected from the group consisting of Leishmania donovani, Leishmania tropica, Leishmania major. Leishmania aethiopica, Leishmania mexicana, Plasmodium falciparum, Plasmodium vivax, Plasmodium ovale, and Plasmodium malariae. Particular intracellular bacterial infections may be selected from the group consisting of Mycobacterium leprae, Mycobacterium tuberculosis, Listeria monocytogenes, and Toxplasma gondii.
When the compounds are used in combination, the at least one additional therapeutic agent may be a vaccine, for example, an anti-viral vaccine, a vaccine against HIV, a vaccine against tuberculosis, a vaccine against malaria. The vaccine may also be a tumour vaccine or a melanoma vaccine. Preferably, the tumour vaccine comprises genetically modified tumour cells or genetically modified cell lines. In such cases, preferably the genetically modified tumour cells or genetically modified cell line has been transfected to express granulocyte-macrophage stimulating factor (GM-CSF),
Alternatively, the vaccine may comprise one or more immunogenic peptides, preferably immunogenic peptides of cancer-testis antigens (CTAgs). Such CTAgs and immunogenic peptides thereof are well known in the art, see Scanlan et ai, Cancer Immun. 2004; 4:1 and Simpson et al ., Nat Rev Cancer. 2005; 5:615. CTAg proteins include MAGE, BAGE, GAGE, SSX, NY-ESO-1, LAGE, SCP, CTSP, CT7, CT8, CT9, CT10, CT 11, SAGE, OY-TES-1, NY-SAR-35 and NY-BR-1. Several MAGE proteins are known, including MAGE-A1, A3, A4, A5, A6, A8, A9, A10, A12, Bl , B2, B3, B4, C1, C2 and C3 proteins- Several SSX proteins exist, including SSX1, SSX2, SSX3 and SSX5.
Further, the tumour vaccine may comprise dendritic cells.
Further, the additional therapeutic agent may be a cytokine, for example a granulocyte- macrophage colony stimulating factor (GM-CSF) or flt3-Hgand.
According to the invention we further provide a method of treating a subject receiving a bone marrow transplant or peripheral blood stem cell transplant comprising administering a therapeutically effective amount of compound of formula 1 or a pharmaceutically acceptable salt thereof to such a subjeet.
Preferably, the compound of formula I or a pharmaceutically acceptable salt thereof is administered in an amount effective to increase the delayed type hypersensitivity reaction to tumour antigen, delay the time to relapse of post-transplant malignancy, increase relapse free survival time post-transplant, and/or increase long-term post- transplant survival. Preferably, the compound of formula I or a pharmaceutically acceptable salt thereof is administered prior to full hematopoetic reconstitution.
The compounds of formula I or a pharmaceutically acceptable salt thereof for use in the method will generally be administered in the form of a pharmaceutical composition.
Thus, according to a further aspect of the invention there is provided a pharmaceutical composition including preferably less than 80% w/w, more preferably less than 50% w/w, e.g. 0.1 to 20%, of the compound of formula I or a pharmaceutically acceptable salt thereof, in admixture with a pharmaceutically acceptable diluent or carrier. We also provide a process for the production of such a pharmaceutical composition which comprises mixing the ingredients. Examples of pharmaceutical formulations which may be used, and suitable diluents or carriers, are as follows;
for intravenous injection or infusion - purified water or saline solution;
for inhalation compositions - coarse lactose;
for tablets, capsules and dragees - macrocrystalline cellulose, calcium phosphate, diatomaceous earth, a sugar such as lactose, dextrose or mannitoi, talc, stearic acid, starch, sodium bicarbonate and/or gelatin;
for suppositories - natural or hardened oils or waxes.
When the compounds of formula I or a pharmaceutically acceptable salt thereof is to be used in aqueous solution, e.g. for infusion, it may be necessary to incorporate other excipients, In particular there may be mentioned chelating or sequestering agents, antioxidants, tonicity adjusting agents, pH-modifying agents and buffering agents.
Solutions containing the compound of formula I or a pharmaceutically acceptable salt thereof may, if desired, be evaporated, e.g, by freeze drying or spray drying, to give a solid composition, which may be reconstituted prior to use.
When not in solution, the compound of formula 1 or a pharmaceutically acceptable salt thereof preferably is in a form having a mass median diameter of from 0.01 to Ι Ομm. The compositions may also contain suitable preserving, stabilising and wetting agents, solubilisers, e.g. a water-soluble cellulose polymer such as hydroxypropyl methylce!lulose, or a water-soluble glycol such as propylene glycol, sweetening and colouring agents and flavourings. Where appropriate, the compositions may be formulated in sustained release form. The content of the compound of formula I or a pharmaceutically acceptable salt thereof in a pharmaceutical composition is generally about 0.01 -about 99.9wt%, preferably about 0.1 -about 50wt%, relative to the entire preparation.
The dose of the compound of formula I or a pharmaceutically acceptable salt thereof is determined in consideration of age, body weight, general health condition, diet, administration time, administration method, clearance rate, combination of drugs, the level of disease for which the patient is under treatment then, and other factors.
While the dose varies depending on the target disease, condition, subject of administration, administration method and the like, for oral administration as a therapeutic agent for the treatment of cancer in a patient suffering from such a disease is from 0.01 mg - 10 g, preferably 0.1 - 100 mg, is preferably administered in a single dose or in 2 or 3 portions per day. Where the compound of formula I or a pharmaceutically acceptable salt thereof is used in combination with other therapeutic agents, these may be used at their normal therapeutic doses, e.g., as set out in pharmacopoeias or prescribing guides, such as the Physicians' Desk Reference (PDR). In certain cases, the compound of formula I or a pharmaceutically acceptable salt thereof supplements the activity of the additional therapeutic agent(s) in a synergistic fashion, such that the additional therapeutic agent(s) can be administered at a lower dose than is normally used.
The potential activity of the compound of formula 1 or a pharmaceutically acceptable salt thereof in the treatment of cancer or infections has been demonstrated in the following predictive experiments, which demonstrate that the compound of formula I is an IDO inhibitor.
Experimental Methods
Chemistry Reagents were purchased from Acros, Fluka, Senn, Sigma-Aldrich or Merck and used without further purification. For extraction and chromatography all solvents were distilled prior to use. Anhydrous THF, diethyl ether, and toluene were distilled from sodium and benzophenone, dichloromethane from CaH2, and methanol from magnesium. Reactions were monitored by thin layer chromatography (TLC) using silica gel plates (Merck 60 F254). Developed TLC plates were visualized with UV light (254 nm) or molybdic reagent (21 g of (ΝΗ4)6Μ07O24.4Η2O, 1 g of Ce(S04)2, 31 ml of H2S04 and 470 ml of H20). Flash chromatography (FC) was conducted using silica gel 60 A, 230-400 mesh (Merck 9385). Melting points were measured with a Mettler FP52 and are uncorrected. Optical rotations were measured with a JASCO DIP-370 digital polarimeter. UV spectra were recorded on a Kontron Uvikon 810 CW spectrophotometer. IR spectra were recorded on a Perkin Elmer Paragon 1000 FT-IR spectrometer. Mass spectra were recorded on a Nermag R 10- IOC in chemical ionisation mode. Electrospray mass analyses were recorded on a Finnigan MAT SSQ 7 IOC spectrometer in positive ionisation mode. 1H-NMR spectra were recorded on a Bruker DPX-400 FT, Bruker ARX-400 FT, or Bruker AMX-600 spectrometer. All 1H signal assignments were confirmed by COSY spectra. !3C-NMR spectra were recorded on a Bruker DPX-400 FT (100.61 MHz) or on a Bruker ARX-400 FT (100.61 MHz) spectrometer. All 13C signal assignments were confirmed by HMQC spectra. Chemical shifts are given in ppm, relative to an internal standard such as residual solvent signals. Coupling constants are given in Hertz. High resolution mass spectra were recorded by ESI-TOF-HRMS or MALDI-TOF-HRMS.
Protein expression and purification of recombinant human IDO
The coding region for human IDO (Ala2-Gly403) was cloned into a derivative of plasmid pET9 (Novagen). The recombinant plasmid, pETIDO, encodes a histidine tag at the N-terminus of IDO. The bacterial strain BL21 AI (Invitrogen) was used for overexpression of IDO and transformed with the plasmid pETIDO. The transformed cells were grown on a rotary shaker at 37 C and 220 rpm to an OD600 of 1.2, in LB medium supplemented with kanamycin 25 μg/ml, L-tryptophan 50 μg/ml and bovine hemin (Sigma) 10 μΜ. The culture was cooled in a water/ice bath and supplemented again with L-tryptophan 50 μg/ml, bovine hemin 10 μΜ. The expression of his-tagged IDO was induced by the addition of 1% (w/v) arabinose. Induced cells were grown at 20 C, 60 rpm for 20 h. Cells (1 1 culture) were collected by centrifugation, resuspended in 40 ml of Mes 25 mM, KC1 150 mM, imidazole 10 mM, protease inhibitors (complete EDTA free, Roche Applied Science), pH 6.5, and disrupted with a French press. The extract was clarified by centrifugation and filtration on a 0.22 μηι filter. The enzyme was purified by IMAC using Ni as ligand and an IMAC HITRAP column (5 ml; GE Healthcare). Briefly, the extract was loaded on the column with buffer Mes 25 mM, KC1 150 mM, imidazole 10 mM, pH 6.5. The column was washed with 50 ml of the same buffer with imidazole adjusted to 100 mM. Finally, the protein was eluted with Mes 25 mM, KC1 150 mM, EDTA 50 mM, pH 6.5. The buffer was then exchanged to Mes 25 mM, KCi 150 mM, pH 6.5 using a HITRAP desalting column (GE Healthcare). The purity of the enzyme was estimated to be > 95% based on SDS PAGE gel and Coomassie blue staining. The ratio of absorbance 404 nm/280 nm of the protein was around 1,9.
4-(CyclohexyIamino)-1-naphthol hydrochloride (Compound of Formula IA).
The compound of formula IΑ was synthesized from 4-amino-1-naphthol hydrochloride and cyclohexanone according to the reductive amination procedure to afford the hydrochloric salt as a white solid in 82% yield. Mp = 276-278 °C. TLC R = 0.42 (40% diethyl ether/pentane). 1H NMR (400 MHz, DMSO-d6 + D20): δ 8.47 (d, 1H, 3J(H- C(8), H-C(7)) = 8.5, H-C(8)); 8.19 (d, 1H, 3J(H-C(5), H-C(6)) = 8.5, H~C(5)); 7.78- 7.64 (m, 4H, H-C(2), H-C(3), H-C(6), H-C(7)); 3.50 (m, 1H, H-C(l ')); 2.15 (d, 2H, 3 J(H-C(2'), H-C(l ')) - 3J(H-C(6'), H-C(l ')) = 8.5, H-C(2')-ax , H-C(6')-i?x); 1.77 (d, 2H, 3J(H-C(2'), H-QT)) = 3J(H-C(6'), H-C(l ')) = 8.5, H-C(2')-eq, H-C(6')-<w); 1.68- 1.54 (m, 2H, H-C(4')); 1.30-1.06 (m, 4H, H-C(3'), H-C(5')). 13C NMR (100.6 MHz, DMSO-d6): δ 151.7 (s, C(l)); 128.2 (d, !J(C,H) = 156, C(7)); 127.2 (s, C(4)); 126.9 (d, 'J(C,H) = 162, C(6)); 126.7 (s, C(9)); 126.6 (d, V(C,H) - 129, C(8)); 124.0 (d, SJ(C,H) = 128, C(5)); 121.9 (d, 'J(C,H) = 156, C(2)); 121.9 (d, !J(C,H) = 156, C(3)); 1 17.0 (s, C(10)); 61.3 (d, 'J(C,H) = 125, C(F)); 29.3 (t, 'J(C,H) = 137, C(2!), C(6')); 25.0 (t, 1 J(C,H) = 137, C(3'), C(5')); 24.4 (t, 'J(C,H) - 137, C(4')). ESI-TOF-HRMS m/z calcd. for (M + H) C16H20NO 242.1545, found 242.1544. Enzymatic Assay
The enzymatic inhibition assays were performed as described by Takikawa et al. [Takikawa, O.; Kuroiwa, T.; Yamazaki, F.; Kido, R. J Biol Chem, 1988, 263, 2041- 2048.] with some modifications. Briefly, the reaction mixture (100 μΐ) contained potassium phosphate buffer (100 mM, pH 6.5) ascorbic acid (20 raM), catalase (200 units/ml), methylene blue (10 μΜ), purified recombinant IDO (l ng/μl), and L- Trp (200 μΜ). The inhibitors were serially diluted ranging from 0.1 to 1000 μΜ. The reaction was carried out at 37 °C for 60 min and stopped by the addition of 30% (w/v) trichloroacetic acid (40 μΐ). To convert the product of Trp dioxygenation by IDO, N- formylkynurenine, to spectroscopically detectable kynurenine, the tubes were incubated at 50 °C for 30 min, followed by a centrifugation at 1OOOOg for 20 min. Lastly, 100 μΐ of supernatant from each probe was transfered to another tube for HPLC analysis. The mobile phase for HPLC measurements consisted of 50% sodium citrate buffer (40 mM, pH 2.25) and 50% methanol with 400 μΜ SDS. The flow rate through the S5-ODS1 column was chosen to be 1 ml/min, and kynurenine was detected at a wavelength of 365 nm.
Cellular Assay
Cell Lines
A plasmid construct encoding murine IDO was transfected into mouse mastocytoma line P815B. Clone P185B-mIDO clone 6, [Uyttenhove et al, Nat Med, 9(10) ppl269- 1274 ((2003)] which overexpresses IDO, was selected and used for the cellular assay. Mouse IDO shares a 62% sequence identity with human IDO, and the active site residues mentioned in the Introduction are 100% conserved. For the assays evaluating inhibition of human IDO, a plasmid construct encoding human IDO was transfected into human HEK-293 cells, and clone 293-hIDO clone 17 [Uyttenhove et al] was selected and used in the assay.
Assay
The assay was performed in 96-well flat bottom plates seeded with 2 x 105 cells in a final volume of 200 μΐ. To determine whether compounds were significant IDO inhibitors, the cells were incubated overnight at 37 °C in HBSS (Hanks Balanced Salt Solution, Invitrogen) supplemented with 50 μΜ L-Trp and 2, 20 or 200 μΜ of the compound. To determine the IC , the cells were incubated for 8 h at 37 °C in HBSS supplemented with 80 μΜ L-Tryptophan and a titration of the compound ranging from 0.312 to 80 μΜ or from 1.56 to 400 μΜ. The plates were then centrifuged for 10 min at 300 g, and 150 μl of the supernatant were collected. The supernatant was analyzed by HPLC to measure the concentration of tryptophan and kynurenine. For HPLC analysis, 50 μΐ of supernatant were mixed with 500 μl acetonitrile to precipitate the proteins. After centrifugation, the supernatant was collected, concentrated on a speedvac, resuspended in a final volume of 100 μΐ water and injected in the HPLC (CI 8 column). Trp was detected at an absorption wavelength of 280 nm, kynurenine at 360 nm. In the conditions used for IC50 estimation, about 50% of the initial amount of tryptophan was degraded in the absence of inhibitor, and an equimolar amount of kynurenine was produced. The percentages of inhibition of tryptophan degradation and kynurenine production by the compounds were calculated in reference to this maximal activity. The initial wells containing the cells in the remaining volume of 50 μΐ were used to estimate cell viability in a classical MTT assay. To that end, 50 μΐ of culture medium (Iscove medium with 10% FCS and amino acids) were added to the wells together with 50 μΐ of MTT. After 3-4 h of incubation at 37 °C, 100 μΐ of SDS/DMF were added to dissolve the crystals of formazan blue. The absorbance at 570 nm/650 nm was measured after overnight incubation at 37 °C.
Experimental Results
In the enzymatic model of IDO inhibition, the following activities were observed:
Figure imgf000014_0001

Claims

Claims
1. Use of the compounds of formula I, and pharmaceutically acceptable salts thereof,
Figure imgf000015_0002
in which
Ri represents hydrogen or cyclohexyl and
R.2 represents hydrogen or alkyl C1-6
provided that R\ and R2 do not both simultaneously represent hydrogen;
in the treatment of diseases in which inhibition of IDO plays a therapeutic role, particularly those conditions mentioned herein.
2. A compound of formula I according to claim 1 in combination with at least one additional therapeutic agent.
3. Use of a compound of formula I according to claim 1 as an adjuvant to the therapeutic vaccination of a cancer or tumour, including melanoma, colon cancer, pancreatic cancer, breast cancer, prostate cancer, lung cancer, leukemia, brain tumours, lymphoma, sarcoma, ovarian cancer, and Kaposi's sarcoma.
4. The compound of formula I A
Figure imgf000015_0001
and pharmaceutically acceptable salts thereof.
5. Use of compounds IB and pharmaceutically acceptable salts thereof as a pharmaceutical
Figure imgf000016_0001
in which R2 is as defined in claim 1 and preferably represents CH3
PCT/EP2010/065336 2009-10-13 2010-10-13 Ido inhibitors and therapeutic uses thereof WO2011045340A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GB0917926.8 2009-10-13
GBGB0917926.8A GB0917926D0 (en) 2009-10-13 2009-10-13 Ido inhibitors and therapeutic uses thereof

Publications (1)

Publication Number Publication Date
WO2011045340A1 true WO2011045340A1 (en) 2011-04-21

Family

ID=41402964

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2010/065336 WO2011045340A1 (en) 2009-10-13 2010-10-13 Ido inhibitors and therapeutic uses thereof

Country Status (2)

Country Link
GB (1) GB0917926D0 (en)
WO (1) WO2011045340A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017140835A1 (en) 2016-02-19 2017-08-24 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for the treatment of obesity

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009127669A2 (en) * 2008-04-15 2009-10-22 Ludwig Institute For Cancer Research Ltd Ido inhibitors and therapeutic uses thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009127669A2 (en) * 2008-04-15 2009-10-22 Ludwig Institute For Cancer Research Ltd Ido inhibitors and therapeutic uses thereof

Non-Patent Citations (12)

* Cited by examiner, † Cited by third party
Title
BUNCE ET AL., JOURNAL OF ORGANIC CHEMISTRY, vol. 52, 1987, pages 4214 - 23
DATABASE PubChem [online] 26 October 2006 (2006-10-26), "4-methoxynaphthalen-1-amine - Compound Summary", XP002612121, retrieved from NCBI Database accession no. 11074644 *
DATABASE PubChem [online] 9 August 2005 (2005-08-09), "4-Butoxy-1-naphthalenamine - Compound Summary", XP002612120, retrieved from NCBI Database accession no. 3041748 *
ELSLAGER E F ET AL: "synthetic Schistosomicides. VI. 4-Substituted 1-(Dialkylaminoalkylamino)naphthalenes", JOURNAL OF MEDICINAL CHEMISTRY, vol. 7, 1 January 1964 (1964-01-01), AMERICAN CHEMICAL SOCIETY, WASHINGTON, US, pages 658 - 662, XP002312532, ISSN: 0022-2623, DOI: 10.1021/JM00335A022 *
KNÖLKER ET AL., SYNTHESIS-STUTTGART 1995, 1995, pages 397 - 408
KUMAR S ET AL: "Structure based development of phenylimidazole-derived inhibitors of indoleamine 2,3-dioxygenase", JOURNAL OF MEDICINAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY, WASHINGTON, US, vol. 51, no. 16, 28 August 2008 (2008-08-28), pages 4968 - 4977, XP002558480, ISSN: 0022-2623, [retrieved on 20080730], DOI: DOI:10.1021/JM800512Z *
SCANLAN ET AL., CANCER IMMUN., vol. 4, 2004, pages 1
SIMPSON ET AL., NAT REV CANCER., vol. 5, 2005, pages 615
SOROKA ET AL., JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY A - CHEMISTRY, vol. 64, 1992, pages 171 - 182
TAKIKAWA, 0; KUROIWA, T.; YAMAZAKI, F.; KIDO, R., J BIOL CHEM, vol. 263, 1988, pages 2041 - 2048
UYTTENHOVE CATHERINE ET AL: "Evidence for a tumoral immune resistance mechanism based on tryptophan degradation by indoleamine 2,3-dioxygenase", NATURE MEDICINE, vol. 9, no. 10, 1 October 2003 (2003-10-01), NATURE PUBLISHING GROUP, NEW YORK, NY, US, pages 1269 - 1274, XP002559000, ISSN: 1078-8956, [retrieved on 20030921], DOI: 10.1038/NM934 *
UYTTENHOVE ET AL., NAT. MED, vol. 9, no. 10, 2003, pages 1269 - 1274

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017140835A1 (en) 2016-02-19 2017-08-24 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods and pharmaceutical compositions for the treatment of obesity

Also Published As

Publication number Publication date
GB0917926D0 (en) 2009-11-25

Similar Documents

Publication Publication Date Title
US20110112282A1 (en) Ido inhibitors and therapeutic uses thereof
KR100831133B1 (en) Geldanamycin Derivative and Method of Treating Cancer Using Same
CA3016161A1 (en) Small molecule ire1-alpha inhibitors
EP3773729A1 (en) Bi-functional molecules to degrade circulating proteins
AU2020356793B2 (en) PH/glutathione-responsive β-carbolines/cycloketene derivatives and their preparation and application
JP2016169161A (en) Novel imidazo pyridine compound
US10011614B2 (en) Bis-β-carboline compound and preparation method, pharmaceutical composition and use thereof
WO2020224652A1 (en) Ubiquitin-specific protease inhibitors, and preparation method therefor and application thereof
NZ744387A (en) Quinazolinone derivative, preparation method therefor, pharmaceutical composition, and applications
US20210047290A1 (en) Spiro compound as indoleamine-2,3-dioxygenase inhibitor
WO2011045341A1 (en) Ido inhibitors and therapeutic uses thereof
US20200277278A1 (en) Deuterium Atom-Substituted Indole Formamide Derivative, Preparation Method Therefor, and Medical Applications Thereof
JP7039579B2 (en) 4-Anilino-quinoline compound as an anticancer agent
CA3152757A1 (en) 4-amino-imidazoquinoline compounds and use thereof
TW200524847A (en) Compound having anti-HCV action and its producing method
JPH11510822A (en) Substituted tetralylmethylene-oxindole homologs as tyrosine kinase inhibitors
WO2011045340A1 (en) Ido inhibitors and therapeutic uses thereof
US10351586B2 (en) Cytidine derivative dimers and applications thereof
US20230250054A1 (en) Selective histone deacetylase 6 inhibitors
CN113004356B (en) Novel genipin derivative, and preparation method and application thereof
CA2918290A1 (en) New anti-malarial agents
CN112142716B (en) 5-membered heteroaryl substituted pyrazine derivative and application thereof
US9499552B2 (en) Pyrazolo[1,5-A]pyrimidine derivative and use of anti-tumor thereof
US11629121B2 (en) Benzylideneacetone derivative and use thereof
US20160102066A1 (en) Benzothiazole derivative and anti-tumor use thereof

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: 10771397

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: 10771397

Country of ref document: EP

Kind code of ref document: A1

32PN Ep: public notification in the ep bulletin as address of the adressee cannot be established

Free format text: NOTING OF LOSS OF RIGHTS PURSUANT TO RULE 112(1) EPC (EPO FORM 1205A DATED 23.08.2012)

122 Ep: pct application non-entry in european phase

Ref document number: 10771397

Country of ref document: EP

Kind code of ref document: A1