WO2019052440A1 - 氘原子取代的吲哚甲酰胺类衍生物、其制备方法及其在医药上的应用 - Google Patents

氘原子取代的吲哚甲酰胺类衍生物、其制备方法及其在医药上的应用 Download PDF

Info

Publication number
WO2019052440A1
WO2019052440A1 PCT/CN2018/105008 CN2018105008W WO2019052440A1 WO 2019052440 A1 WO2019052440 A1 WO 2019052440A1 CN 2018105008 W CN2018105008 W CN 2018105008W WO 2019052440 A1 WO2019052440 A1 WO 2019052440A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
atom
compound
formula
halogen
Prior art date
Application number
PCT/CN2018/105008
Other languages
English (en)
French (fr)
Inventor
刘�东
陈磊
陆标
刘苏星
张儒民
贺峰
陶维康
Original Assignee
江苏恒瑞医药股份有限公司
上海恒瑞医药有限公司
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 江苏恒瑞医药股份有限公司, 上海恒瑞医药有限公司 filed Critical 江苏恒瑞医药股份有限公司
Priority to AU2018331125A priority Critical patent/AU2018331125A1/en
Priority to BR112020004611-6A priority patent/BR112020004611A2/pt
Priority to JP2020514271A priority patent/JP2020533337A/ja
Priority to EP18856441.3A priority patent/EP3683206A4/en
Priority to CN201880004431.1A priority patent/CN109963836B/zh
Priority to CA3075324A priority patent/CA3075324A1/en
Priority to US16/645,533 priority patent/US20200277278A1/en
Publication of WO2019052440A1 publication Critical patent/WO2019052440A1/zh

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • A61K31/404Indoles, e.g. pindolol
    • A61K31/405Indole-alkanecarboxylic acids; Derivatives thereof, e.g. tryptophan, indomethacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/4151,2-Diazoles
    • A61K31/41551,2-Diazoles non condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/04Indoles; Hydrogenated indoles
    • C07D209/10Indoles; Hydrogenated indoles with substituted hydrocarbon radicals attached to carbon atoms of the hetero ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/05Isotopically modified compounds, e.g. labelled
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152

Definitions

  • the invention belongs to the field of medicine, and relates to an oxime (D) atom-substituted anthraquinone derivative, a preparation method thereof and application thereof in medicine.
  • the present invention relates to a fluorene-substituted anthracene amide derivative represented by the general formula (I), a process for producing the same, a pharmaceutical composition containing the same, which is used as an ROR agonist and its use for prevention and / or use in drugs for treating tumors or cancer.
  • Retinoid-related orphan receptors are members of the nuclear receptor family and are a class of ligand-dependent transcription factors that regulate a variety of physiological and biochemical processes, including reproductive development. Metabolism, regulation of the immune system, etc. (Mech Dev. 1998 Jan, 70 (1-2: 147-53; EMBO J. 1998 Jul 15, 17(14): 3867-77).
  • the ROR family includes three types of ROR ⁇ , ROR ⁇ and ROR ⁇ (Curr Drug Targets Inflamm Allergy. 2004 Dec, 3(4): 395-412), in which ROR ⁇ can be expressed in many tissues including thymus, liver, kidney, fat and skeletal muscle (Immunity.1998 Dec, 9 (6) ): 797-806.).
  • ROR ⁇ 1 ROR ⁇ 1
  • ROR ⁇ t ROR ⁇ 2
  • Th17 can also recruit cytotoxic CD8+ T cells and natural killer cells into the tumor microenvironment to kill tumor cells for anti-tumor purposes (Blood.2009 Aug 6,114(6):1141-9; Clin Cancer Res.2008 Jun 1,14 (11): 3254-61). Therefore, activation of ROR ⁇ t may become a new anti-tumor therapy.
  • ROR ⁇ t a small molecule drug developed by Lycera Corp.
  • LYC-54143 activates ROR ⁇ t to regulate the differentiation of Th17 and Tc17 cells, promote the expression of other cytokines such as IL-17, and increase the activity of T cells.
  • activated ROR ⁇ t can regulate the expression of various genes in the immune system, inhibit the expression of PD-1 in the cell, thereby reducing immunosuppression and increasing anticancer activity (Oncoimmunology.2016 Nov 4,5(12):e1254854; ACS Chem Biol.
  • the present inventors have designed a hydrazine atom-substituted anthracene amide compound having a structure represented by the general formula (I), and the substituent contains a ruthenium atom, so that the compound of the present invention has unexpected pharmacological absorption activity and pharmacological action.
  • the compound of the present invention has unexpected pharmacological absorption activity and pharmacological action.
  • the invention also provides a pharmacodynamic test, which exhibits good antitumor activity when the compound of the invention is administered alone, and additionally, the compound of the invention exhibits a synergistic effect when combined with the PD-1 antibody, and opens up a new effect for improving the therapeutic effect of the immunotherapy. way.
  • G 1 , G 2 and G 3 are the same or different and are each independently selected from C, CH, CH 2 and N;
  • Ring A is selected from the group consisting of an aryl group, a heteroaryl group, a cycloalkyl group, and a heterocyclic group;
  • Ring B is an aryl or heteroaryl group
  • R 1 is the same or different and is each independently selected from the group consisting of a H atom, a D atom, a halogen, an alkyl group, a halogenated alkyl group, an alkoxy group, a halogenated alkoxy group, a cyano group, an amino group, a nitro group, a hydroxyl group, and a hydroxyalkyl group;
  • R 2 is a haloalkyl group
  • R 3 and R 4 are the same or different and are each independently selected from the group consisting of H atom, D atom, halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, hydroxyalkyl, cyano, amino, nitro, hydroxy a cycloalkyl, a heterocyclic group, an aryl group and a heteroaryl group, wherein said alkyl group, hydroxyalkyl group, cycloalkyl group, heterocyclic group, aryl group and heteroaryl group are each independently optionally selected from D Substituting one or more substituents of an atom, a hydroxyl group, a halogen, an alkyl group, an amino group, and -OR 11 ;
  • R 5 is selected from the group consisting of H atom, alkyl group, halogenated alkyl group, amino group, hydroxyl group, hydroxyalkyl group, cycloalkyl group, heterocyclic group, aryl group and heteroaryl group, wherein the alkyl group, cycloalkyl group, heterocyclic group And an aryl group and a heteroaryl group are optionally substituted with one or more substituents selected from the group consisting of a D atom, a halogen, a hydroxyl group, a cycloalkyl group, and a heterocyclic group;
  • R 6 are the same or different and are each independently selected from the group consisting of H atom, D atom, halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, naphthenic a base, a heterocyclic group, an aryl group and a heteroaryl group;
  • R 7 is selected from the group consisting of a H atom, an alkyl group, a halogenated alkyl group, a cycloalkyl group, a heterocyclic group, an aryl group, and a heteroaryl group, wherein the alkyl group, the cycloalkyl group, the heterocyclic group, the aryl group, and the heteroaryl group are Selected from D atom, halogen, hydroxy, amino, cyano, nitro, alkoxy, haloalkoxy, -OR 11 , -C(O)R 11 , -C(O)OR 11 , -NR 12 Substituting one or more substituents of R 13 , —C(O)NR 12 R 13 , —S(O) m R 11 , cycloalkyl and heterocyclyl;
  • R 8 and R 9 are the same or different and are each independently selected from the group consisting of a H atom, a D atom, a halogen, an alkyl group, a halogenated alkyl group, an alkoxy group, a cyano group, an amino group, a nitro group, a hydroxyl group, and a hydroxyalkyl group;
  • R 10 is the same or different and is each independently selected from the group consisting of a H atom, a D atom, a halogen, an alkyl group, a halogenated alkyl group, an alkoxy group, a halogenated alkoxy group, a cyano group, an amino group, a nitro group, a hydroxyl group, a hydroxyalkyl group, a cycloalkane. a base, a heterocyclic group, an aryl group and a heteroaryl group;
  • R 11 is selected from the group consisting of a H atom, a D atom, an alkyl group, a halogenated alkyl group, a hydroxyalkyl group, a cycloalkyl group, and a heterocyclic group, wherein the alkyl group, hydroxyalkyl group, cycloalkyl group, and heterocyclic group are optionally selected. Substituted from one or more substituents of a D atom, a halogen, a hydroxyl group, a cycloalkyl group, and a heterocyclic group;
  • R 12 and R 13 are the same or different and are each independently selected from the group consisting of a H atom, a D atom, an alkyl group, a halogenated alkyl group, a hydroxyl group, and a hydroxyalkyl group, wherein the alkyl group and the hydroxyalkyl group are optionally selected from a D atom. Substituting one or more substituents of a halogen, a hydroxyl group, a cycloalkyl group and a heterocyclic group;
  • the condition is that the compound represented by the formula (I) contains at least one D atom, and in particular, at least one of the substituents R 3 , R 4 , R 5 , R 7 , R 8 and R 9 contains at least one D atom. ;
  • n 0, 1 or 2;
  • n 0, 1, 2, 3 or 4;
  • s 0, 1, 2 or 3;
  • t 0, 1, 2 or 3.
  • the compound of the formula (I) is a compound of the formula (II):
  • G is CH or N
  • R 1 , R 3 to R 9 , n and t are as defined in the formula (I).
  • the compound of the formula (I), wherein R 7 is selected from the group consisting of alkyl, haloalkyl, cycloalkyl and heterocyclyl, wherein the alkyl group is optional Substituted by one or more substituents selected from the group consisting of D atoms, halogens, hydroxy groups, amino groups, cyano groups, nitro groups, alkoxy groups, haloalkoxy groups, and hydroxyalkyl groups.
  • the compound of the formula (I) is a compound of the formula (III):
  • L 1 is an alkylene group, wherein the alkylene group is optionally substituted with one or more substituents of a halogen and a D atom;
  • R 14 is selected from the group consisting of D atom, halogen, hydroxy, amino, cyano, nitro, alkoxy, haloalkoxy and hydroxyalkyl;
  • R 1 , R 5 , R 6 , R 11 , n and t are as defined in the formula (I).
  • the compound of the formula (I) is a compound of the formula (IV):
  • L 1 is an alkylene group, wherein the alkylene group is optionally substituted with one or more substituents of a halogen and a D atom;
  • R 14 is selected from the group consisting of D atom, halogen, hydroxy, amino, cyano, nitro, alkoxy, haloalkoxy and hydroxyalkyl;
  • R 1 , R 5 , R 6 , R 11 , n and t are as defined in the formula (I).
  • the compound of the formula (I) wherein R 1 is the same or different and each independently is selected from the group consisting of a H atom, a D atom, a halogen and an alkyl group.
  • the compound of the formula (I), wherein R 5 is an alkyl group, wherein the alkyl group is optionally selected from the group consisting of a D atom, a halogen and a hydroxyl group. Or substituted with multiple substituents.
  • the compound of the formula (I) wherein R 6 are the same or different and are each independently selected from a H atom, a D atom or a halogen.
  • Typical compounds of formula (I) include, but are not limited to:
  • a tautomer a meso form, a racemate, an enantiomer, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof.
  • Another aspect of the invention relates to a compound of the formula (V) which is an intermediate for the preparation of a compound of the formula (I),
  • G 1 , G 2 and G 3 are the same or different and are each independently selected from C, CH, CH 2 and N;
  • Ring A is selected from the group consisting of an aryl group, a heteroaryl group, a cycloalkyl group, and a heterocyclic group;
  • R 1 is the same or different and is each independently selected from the group consisting of a H atom, a D atom, a halogen, an alkyl group, a halogenated alkyl group, an alkoxy group, a halogenated alkoxy group, a cyano group, an amino group, a nitro group, a hydroxyl group, and a hydroxyalkyl group;
  • R 2 is a haloalkyl group
  • R 6 are the same or different and are each independently selected from the group consisting of H atom, D atom, halogen, alkyl, haloalkyl, alkoxy, haloalkoxy, cyano, amino, nitro, hydroxy, hydroxyalkyl, naphthenic a base, a heterocyclic group, an aryl group and a heteroaryl group;
  • R 7 is selected from the group consisting of a H atom, an alkyl group, a halogenated alkyl group, a cycloalkyl group, a heterocyclic group, an aryl group, and a heteroaryl group, wherein the alkyl group, the cycloalkyl group, the heterocyclic group, the aryl group, and the heteroaryl group are Selected from D atom, halogen, hydroxy, amino, cyano, nitro, alkoxy, haloalkoxy, -OR 11 , -C(O)R 11 , -C(O)OR 11 , -NR 12 Substituting one or more substituents of R 13 , —C(O)NR 12 R 13 , —S(O) m R 11 , cycloalkyl and heterocyclyl;
  • R 8 and R 9 are the same or different and are each independently selected from the group consisting of a H atom, a D atom, a halogen, an alkyl group, a halogenated alkyl group, an alkoxy group, a cyano group, an amino group, a nitro group, a hydroxyl group, and a hydroxyalkyl group;
  • R 11 is selected from the group consisting of a H atom, a D atom, an alkyl group, a halogenated alkyl group, a hydroxyalkyl group, a cycloalkyl group, and a heterocyclic group, wherein the alkyl group, hydroxyalkyl group, cycloalkyl group, and heterocyclic group are optionally selected. Substituted from one or more substituents of a D atom, a halogen, a hydroxyl group, a cycloalkyl group, and a heterocyclic group;
  • R 12 and R 13 are the same or different and are each independently selected from the group consisting of a H atom, a D atom, an alkyl group, a halogenated alkyl group, a hydroxyl group, and a hydroxyalkyl group, wherein the alkyl group and the hydroxyalkyl group are optionally selected from a D atom. Substituting one or more substituents of a halogen, a hydroxyl group, a cycloalkyl group and a heterocyclic group;
  • n 0, 1 or 2;
  • n 0, 1, 2, 3 or 4;
  • t 0, 1, 2 or 3.
  • the compound of the formula (V), wherein at least one of the substituents R 7 , R 8 and R 9 contains one or more D atoms.
  • Typical compounds of formula (V) include, but are not limited to:
  • a tautomer a meso form, a racemate, an enantiomer, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof.
  • Another aspect of the invention relates to a process for the preparation of a compound of formula (I), which process comprises:
  • a compound of the formula (V) is condensed with a compound of the formula (VI) or a pharmaceutically acceptable salt thereof to give a compound of the formula (I);
  • Ring A, Ring B, G 1 to G 3 , R 1 to R 10 , n, s and t are as defined in the formula (I).
  • Another aspect of the invention relates to a process for the preparation of a compound of formula (II), the process comprising:
  • a compound of the formula (II-3) is condensed with a compound of the formula (II-4) or a pharmaceutically acceptable salt thereof to give a compound of the formula (II);
  • R 1 , R 3 to R 9 , n and t are as defined in the formula (II).
  • Another aspect of the invention relates to a process for the preparation of a compound of the formula (III) which comprises:
  • a compound of the formula (III-3) is condensed with a compound of the formula (III-4) or a pharmaceutically acceptable salt thereof to give a compound of the formula (III);
  • R 1 , R 5 , R 6 , R 11 , R 14 , L 1 , n and t are as defined in the formula (III); R 8 and R 9 are as defined in the formula (I).
  • Another aspect of the invention relates to a process for the preparation of a compound of formula (IV), the process comprising:
  • a compound of the formula (III-3) is condensed with a compound of the formula (IV-1) or a pharmaceutically acceptable salt thereof to give a compound of the formula (IV);
  • R 1 , R 5 , R 6 , R 11 , R 14 , L 1 , n and t are as defined in the formula (IV); R 8 and R 9 are as defined in the formula (I).
  • Another aspect of the invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound of the formula (I) or a tautomer thereof, a mesogen, a racemate, an enantiomer a form, a diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt, and one or more pharmaceutically acceptable carriers, diluents or excipients.
  • the present invention also relates to a process for the preparation of the pharmaceutical composition
  • the pharmaceutical composition comprising the compound of the formula (I) or a tautomer, a mesophil, a racemate, an enantiomer thereof
  • the diastereomer, or a mixture thereof, or a pharmaceutically acceptable salt thereof is admixed with a pharmaceutically acceptable carrier, diluent or excipient.
  • the pharmaceutical composition further comprises an anti-PD-1 antibody, preferably an anti-murine PD-1 antibody.
  • the invention further relates to a compound of the formula (I) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer or a mixture thereof, or Use of a pharmaceutically acceptable salt, or a pharmaceutical composition comprising the same, in the manufacture of a ROR agonist.
  • the invention further relates to a compound of the formula (I) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer or a mixture thereof, or Use of a pharmaceutically acceptable salt, or a pharmaceutical composition comprising the same, for the preparation of a medicament for the prevention and/or treatment of a tumor or cancer, in particular as a ROR agonist for the preparation of a medicament for the prevention and/or treatment of a tumor or cancer Use in.
  • the invention further relates to a compound of the formula (I) or a tautomer, a mesomer, a racemate, an enantiomer, a diastereomer or a mixture thereof, or Use of a pharmaceutically acceptable salt (as an ROR agonist), or a combination of a pharmaceutical composition comprising the same and an anti-PD-1 antibody, for the manufacture of a medicament for the prevention and/or treatment of a tumor or cancer.
  • the invention further relates to a compound of the formula (I) or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer or a mixture thereof, or A pharmaceutically acceptable salt, or a pharmaceutical composition comprising the same, for use as a medicament.
  • the present invention also relates to a compound of the formula (I) or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer or a mixture thereof, or A pharmaceutically acceptable salt, or a pharmaceutical composition comprising the same, as an ROR agonist.
  • the present invention also relates to a compound of the formula (I) or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer or a mixture thereof, or A pharmaceutically acceptable salt, or a pharmaceutical composition comprising the same, for use in the prevention and/or treatment of a tumor or cancer, in particular as an ROR agonist for the prevention and/or treatment of a tumor or cancer.
  • the present invention also relates to a compound of the formula (I) or a tautomer, a mesophil, a racemate, an enantiomer, a diastereomer or a mixture thereof, or A pharmaceutically acceptable salt, or a pharmaceutical composition comprising the same, in combination with an anti-PD-1 antibody for use in the prevention and/or treatment of a tumor or cancer.
  • the present invention also relates to a method for the treatment of preventing and/or treating a tumor or cancer comprising administering to a patient in need thereof a therapeutically effective amount of a compound of the formula (I) or a tautomer thereof as an ROR agonist. a form, a meso form, a racemate, an enantiomer, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same.
  • the present invention also relates to a method for the prophylactic and/or therapeutic treatment of preventing tumors or cancer comprising administering to a patient in need thereof a therapeutically effective amount of a compound of the formula (I) or a tautomer thereof, internal elimination A form of a rot, a racemate, an enantiomer, a diastereomer or a mixture thereof, or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition comprising the same and an anti-PD-1 antibody.
  • the tumor or cancer of the present invention is selected from the group consisting of non-Hodgkin's lymphoma, diffuse large B-cell lymphoma, follicular lymphoma, synovial sarcoma, breast cancer, cervical cancer, colon cancer, lung cancer, stomach cancer, rectal cancer, Pancreatic cancer, brain cancer, skin cancer, oral cancer, prostate cancer, bone cancer, kidney cancer, ovarian cancer, bladder cancer, liver cancer, fallopian tube tumor, ovarian tumor, peritoneal tumor, melanoma, solid tumor, glioma, nerve Glioblastoma, hepatocellular carcinoma, papillary renal tumor, head and neck tumor, leukemia, lymphoma, myeloma, and non-small cell lung cancer.
  • the active ingredient-containing pharmaceutical composition may be in a form suitable for oral administration, such as tablets, dragees, lozenges, aqueous or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules, or syrups or Tincture.
  • Oral compositions can be prepared according to any method known in the art for preparing pharmaceutical compositions, such compositions may contain one or more ingredients selected from the group consisting of sweeteners, flavoring agents, coloring agents, and preservatives, To provide a pleasing and tasty pharmaceutical preparation. Tablets contain the active ingredient and non-toxic pharmaceutically acceptable excipients suitable for the preparation of tablets for mixing.
  • excipients can be inert excipients, granulating agents, disintegrating agents, binders, and lubricants. These tablets may be uncoated or may be coated by masking the taste of the drug or delaying disintegration and absorption in the gastrointestinal tract, thus providing a sustained release effect over a longer period of time.
  • Oral formulations can also be provided in soft gelatine capsules in which the active ingredient is mixed with an inert solid diluent or the active ingredient in admixture with a water-soluble vehicle or an oil vehicle.
  • the aqueous suspension contains the active substance and excipients suitable for the preparation of the aqueous suspension for mixing. Such excipients are suspending, dispersing or wetting agents.
  • the aqueous suspensions may also contain one or more preservatives, one or more coloring agents, one or more flavoring agents, and one or more sweetening agents.
  • the oil suspension can be formulated by suspending the active ingredient in vegetable oil, or mineral oil.
  • the oil suspension may contain a thickening agent.
  • the above sweeteners and flavoring agents may be added to provide a palatable preparation. These compositions can be preserved by the addition of an antioxidant.
  • compositions of the invention may also be in the form of an oil-in-water emulsion.
  • the oil phase can be a vegetable oil, or a mineral oil or a mixture thereof.
  • Suitable emulsifiers can be naturally occurring phospholipids, and emulsions can also contain sweeteners, flavoring agents, preservatives, and antioxidants.
  • Such formulations may also contain a demulcent, a preservative, a colorant, and an antioxidant.
  • the pharmaceutical compositions of the invention may be in the form of a sterile injectable aqueous solution.
  • acceptable vehicles or solvents that may be employed are water, Ringer's solution, and isotonic sodium chloride solution.
  • the sterile injectable preparation may be a sterile injectable oil-in-water microemulsion in which the active ingredient is dissolved in the oily phase.
  • the injection or microemulsion is injected into the bloodstream of the patient by topical injection.
  • the solution and microemulsion are preferably administered in a manner that maintains a constant circulating concentration of the compound of the invention.
  • a continuous intravenous delivery device can be used.
  • An example of such a device is the Deltec CADD-PLUS.TM.5400 intravenous pump.
  • compositions of the invention may be in the form of a sterile injectable aqueous or oily suspension for intramuscular and subcutaneous administration.
  • the suspension may be formulated according to known techniques using those suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension prepared in a parenterally acceptable non-toxic diluent or solvent.
  • sterile fixed oils may conveniently be employed as a solvent or suspension medium. Any blended fixed oil can be used for this purpose.
  • fatty acids can also be prepared as injections.
  • the compounds of the invention may be administered in the form of a suppository for rectal administration.
  • These pharmaceutical compositions can be prepared by mixing the drug with a suitable non-irritating excipient which is solid at ordinary temperatures but liquid in the rectum and thus dissolves in the rectum to release the drug.
  • the dosage of the drug to be administered depends on a variety of factors including, but not limited to, the following factors: the activity of the particular compound used, the age of the patient, the weight of the patient, the health of the patient, the behavior of the patient. , the patient's diet, the time of administration, the mode of administration, the rate of excretion, the combination of drugs, etc.; in addition, the optimal treatment modality such as the mode of treatment, the daily dosage of the compound of formula (I) or the pharmaceutically acceptable salt
  • the type can be verified according to traditional treatment options.
  • alkyl refers to a saturated aliphatic hydrocarbon group which is a straight or branched chain group containing from 1 to 20 carbon atoms, preferably an alkyl group having from 1 to 12 carbon atoms, more preferably from 1 to 6 carbons.
  • the alkyl group of the atom is a saturated aliphatic hydrocarbon group which is a straight or branched chain group containing from 1 to 20 carbon atoms, preferably an alkyl group having from 1 to 12 carbon atoms, more preferably from 1 to 6 carbons.
  • Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 1 ,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2- Methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3 - dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-dimethylbutyl, n-heptyl, 2 -methylhexyl, 3-methylhexyl, 4-methylhexyl,
  • lower alkyl groups having from 1 to 6 carbon atoms, non-limiting examples including methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl Base, n-pentyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethyl Butyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl, 2-ethylbutyl, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl Base, 2,3-dimethylbutyl and the like.
  • the alkyl group may be substituted or unsubstituted, and when substituted, the substituent may be substituted at any available point of attachment, which is independently selected from the group consisting of a ruthenium atom, an alkyl group, an alkenyl group, an alkynyl group, an alkoxy group.
  • alkylthio alkylamino, halogen, fluorenyl, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkane Thio, heterocycloalkylthio, oxo, carboxyl, carboxylate, -OR 11 , -C(O)R 11 , -C(O)OR 11 , -NR 12 R 13 , -C(O Substituting one or more substituents of NR 12 R 13 and -S(O) m R 11 .
  • alkylene refers to a saturated straight or branched aliphatic hydrocarbon radical having two residues derived from the removal of two hydrogen atoms from the same carbon atom of the parent alkane or two different carbon atoms.
  • Non-limiting examples of alkylene include, but are not limited to, methylene (-CH 2 -), 1,1-ethylene (-CH(CH 3 )-), 1,2-ethylene (-CH 2 ) CH 2 )-, 1,1-propylene (-CH(CH 2 CH 3 )-), 1,2-propylene (-CH 2 CH(CH 3 )-), 1,3-propylene (-CH 2 CH 2 CH 2 -), 1,4-butylene (-CH 2 CH 2 CH 2 CH 2 -) and 1,5-pentylene (-CH 2 CH 2 CH 2 CH 2 CH 2 - )Wait.
  • the alkylene group may be substituted or unsubstituted, and when substituted, the substituent may be substituted at any available point of attachment, which is independently optionally selected from the group consisting of a halogen atom, an alkyl group, an alkenyl group, Alkynyl, alkoxy, alkylthio, alkylamino, halogen, decyl, hydroxy, nitro, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkane Oxyl, cycloalkylthio, heterocycloalkylthio, oxo, -OR 11 , -C(O)R 11 , -C(O)OR 11 , -NR 12 R 13 , -C(O)NR Substituting one or more substituents of 12 R 13 and -S(O) m R 11 .
  • alkoxy refers to -O-(alkyl) and -O-(unsubstituted cycloalkyl), wherein alkyl and cycloalkyl are as defined above.
  • alkoxy groups include: methoxy, ethoxy, propoxy, butoxy, cyclopropoxy, cyclobutoxy, cyclopentyloxy, cyclohexyloxy.
  • the alkoxy group may be optionally substituted or unsubstituted, and when substituted, is independently selected from the group consisting of a halogen atom, an alkyl group, an alkenyl group, an alkynyl group, an alkoxy group, an alkylthio group, an alkylamino group, a halogen, Mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxy, Carboxyl ester group, -OR 11 , -C(O)R 11 , -C(O)OR 11 , -NR 12 R 13 , -C(O)NR 12 R 13 and -S(O) m R 11 Substituted by one or more substituents.
  • cycloalkyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent containing from 3 to 20 carbon atoms, preferably from 3 to 12 carbon atoms, more preferably from 3 to 6 carbon atoms. One carbon atom.
  • Non-limiting examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatriene
  • a polycycloalkyl group includes a spiro ring, a fused ring, and a cycloalkyl group.
  • spirocycloalkyl refers to a polycyclic group that shares a carbon atom (referred to as a spiro atom) between 5 to 20 members of a single ring, which may contain one or more double bonds, but none of the rings have a fully conjugated ⁇ electronic system. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • the spirocycloalkyl group is classified into a monospirocycloalkyl group, a bispirocycloalkyl group or a polyspirocycloalkyl group, preferably a monospirocycloalkyl group and a bispirocycloalkyl group, depending on the number of common spiro atoms between the rings.
  • spirocycloalkyl groups include:
  • fused cycloalkyl refers to 5 to 20 members, and each ring in the system shares an all-carbon polycyclic group of an adjacent pair of carbon atoms with other rings in the system, wherein one or more of the rings may contain one or Multiple double bonds, but none of the rings have a fully conjugated ⁇ -electron system. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • fused cycloalkyl groups include:
  • bridged cycloalkyl refers to an all-carbon polycyclic group of 5 to 20 members, any two rings sharing two carbon atoms which are not directly bonded, which may contain one or more double bonds, but none of the rings have complete Conjugate ⁇ -electron system. It is preferably 6 to 14 members, more preferably 7 to 10 members. Depending on the number of constituent rings, it may be classified into a bicyclic, tricyclic, tetracyclic or polycyclic bridged cycloalkyl group, preferably a bicyclic ring, a tricyclic ring or a tetracyclic ring, and more preferably a bicyclic ring or a tricyclic ring.
  • bridged cycloalkyl groups include:
  • the cycloalkyl ring may be fused to an aryl, heteroaryl or heterocycloalkyl ring, wherein the ring to which the parent structure is attached is a cycloalkyl group, non-limiting examples include indanyl, tetrahydronaphthalene Base, benzocycloheptyl and the like.
  • the cycloalkyl group may be optionally substituted or unsubstituted, and when substituted, it is independently selected from the group consisting of a halogen atom, an alkyl group, an alkenyl group, an alkynyl group, an alkoxy group, an alkylthio group, an alkylamino group, a halogen, Sulfhydryl, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo Base, carboxyl group, carboxylate group, -OR 11 , -C(O)R 11 , -C(O)OR 11 , -NR 12 R 13 , -C(O)NR 12 R 13 and -S(O) Substituted by one or more substituents in m R 11 .
  • heterocyclyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent containing from 3 to 20 ring atoms wherein one or more ring atoms are selected from nitrogen, oxygen or S(O).
  • a hetero atom of m (where m is an integer of 0 to 2), but excluding the ring moiety of -OO-, -OS- or -SS-, the remaining ring atoms being carbon.
  • ring atoms Preferably comprising from 3 to 12 ring atoms, wherein from 1 to 4 are heteroatoms; most preferably from 3 to 8 ring atoms, wherein from 1 to 3 are heteroatoms; most preferably from 3 to 6 ring atoms, wherein from 1 to 2 It is a hetero atom.
  • monocyclic heterocyclic groups include pyrrolidinyl, imidazolidinyl, tetrahydrofuranyl, tetrahydrothiophenyl, dihydroimidazolyl, dihydrofuranyl, dihydropyrazolyl, dihydropyrrolyl, piperidine.
  • the group, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, pyranyl and the like are preferably piperidinyl, piperazinyl or morpholinyl.
  • Polycyclic heterocyclic groups include spiro, fused, and bridged heterocyclic groups.
  • spiroheterocyclyl refers to a polycyclic heterocyclic group in which one atom (called a spiro atom) is shared between 5 to 20 members of a single ring, wherein one or more ring atoms are selected from nitrogen, oxygen or S (O). ) m (where m is an integer 0 to 2) heteroatoms, and the remaining ring atoms are carbon. It may contain one or more double bonds, but none of the rings have a fully conjugated pi-electron system. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • the spiroheterocyclyl group is classified into a monospiroheterocyclic group, a dispiroheterocyclic group or a polyspirocyclic group according to the number of shared spiro atoms between the ring and the ring, and is preferably a monospiroheterocyclic group and a dispiroheterocyclic group.
  • spiroheterocyclyl groups include:
  • fused heterocyclyl refers to 5 to 20 members, and each ring in the system shares an adjacent pair of atomic polycyclic heterocyclic groups with other rings in the system, and one or more rings may contain one or more Double bond, but none of the rings have a fully conjugated ⁇ -electron system in which one or more ring atoms are heteroatoms selected from nitrogen, oxygen or S(O) m (where m is an integer from 0 to 2), and the remaining rings
  • the atom is carbon. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • fused heterocyclic groups include:
  • bridge heterocyclyl refers to a polycyclic heterocyclic group of 5 to 14 members, any two rings sharing two atoms which are not directly bonded, which may contain one or more double bonds, but none of the rings have a total A ⁇ -electron system of a yoke in which one or more ring atoms are heteroatoms selected from nitrogen, oxygen or S(O) m (where m is an integer from 0 to 2), the remaining ring atoms being carbon. It is preferably 6 to 14 members, more preferably 7 to 10 members.
  • bridge heterocyclic groups include:
  • the heterocyclyl ring may be fused to an aryl, heteroaryl or cycloalkyl ring, wherein the ring to which the parent structure is attached is a heterocyclic group, non-limiting examples of which include:
  • the heterocyclic group may be optionally substituted or unsubstituted, and when substituted, is independently selected from the group consisting of a halogen atom, an alkyl group, an alkenyl group, an alkynyl group, an alkoxy group, an alkylthio group, an alkylamino group, a halogen, Sulfhydryl, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, oxo Base, carboxyl group, carboxylate group, -OR 11 , -C(O)R 11 , -C(O)OR 11 , -NR 12 R 13 , -C(O)NR 12 R 13 and -S(O) Substituted by one or more substituents in m R 11 .
  • aryl refers to a 6 to 14 membered all-carbon monocyclic or fused polycyclic ring (ie, a ring that shares a pair of adjacent carbon atoms) having a conjugated ⁇ -electron system, preferably 6 to 10 members, such as benzene. Base and naphthyl. More preferred is phenyl.
  • the aryl ring may be fused to a heteroaryl, heterocyclic or cycloalkyl ring, wherein the ring to which the parent structure is attached is an aryl ring, non-limiting examples of which include:
  • the aryl group may be substituted or unsubstituted, and when substituted, it is independently selected from the group consisting of a halogen atom, an alkyl group, an alkenyl group, an alkynyl group, an alkoxy group, an alkylthio group, an alkylamino group, a halogen, a fluorenyl group, a hydroxyl group.
  • heteroaryl refers to a heteroaromatic system containing from 1 to 4 heteroatoms, from 5 to 14 ring atoms, wherein the heteroatoms are selected from the group consisting of oxygen, sulfur and nitrogen.
  • the heteroaryl group is preferably 5 to 10 members, and has 1 to 3 hetero atoms; more preferably 5 or 6 members, and 1 to 2 hetero atoms; preferably, for example, imidazolyl, furyl, thienyl, thiazolyl, pyridyl Azolyl, oxazolyl, pyrrolyl, tetrazolyl, pyridyl, pyrimidinyl, thiadiazole, pyrazinyl, etc., preferably imidazolyl, tetrazolyl, pyridyl, thienyl, pyrazolyl or pyrimidinyl , thiazolyl; more selective pyridyl.
  • the heteroaryl ring may be fuse
  • the heteroaryl group may be optionally substituted or unsubstituted, and when substituted, is independently selected from the group consisting of a halogen atom, an alkyl group, an alkenyl group, an alkynyl group, an alkoxy group, an alkylthio group, an alkylamino group, a halogen, Mercapto, hydroxy, nitro, cyano, cycloalkyl, heterocycloalkyl, aryl, heteroaryl, cycloalkoxy, heterocycloalkoxy, cycloalkylthio, heterocycloalkylthio, carboxy, Carboxyl ester group, -OR 11 , -C(O)R 11 , -C(O)OR 11 , -NR 12 R 13 , -C(O)NR 12 R 13 and -S(O) m R 11 Substituted by one or more substituents.
  • haloalkyl refers to an alkyl group substituted by one or more halogens, wherein alkyl is as defined above.
  • haloalkoxy refers to an alkoxy group substituted by one or more halogens, wherein alkoxy is as defined above.
  • hydroxyalkyl refers to an alkyl group substituted with a hydroxy group, wherein alkyl is as defined above.
  • hydroxy refers to an -OH group.
  • halogen means fluoro, chloro, bromo or iodo.
  • amino means -NH 2.
  • cyano refers to -CN.
  • nitro refers to -NO 2 .
  • carboxylate group refers to -C(O)O(alkyl) or -C(O)O(cycloalkyl), wherein alkyl, cycloalkyl are as defined above.
  • acyl halide refers to a compound containing a -C(O)-halogen group.
  • the invention also includes various compounds of formula (I) in deuterated form.
  • Each of the available hydrogen atoms attached to the carbon atom can be independently replaced by a deuterium atom.
  • Those skilled in the art will be able to synthesize a compound of formula (I) in deuterated form with reference to the relevant literature.
  • Commercially available deuterated starting materials can be used in the preparation of the compounds of formula (I) in deuterated form, or they can be synthesized using deuterated reagents using conventional techniques, including but not limited to deuterated borane, triterpenoids. Borane tetrahydrofuran solution, deuterated lithium aluminum hydride, deuterated iodoethane and deuterated iodomethane.
  • heterocyclic group optionally substituted by an alkyl group means that an alkyl group may be, but is not necessarily, present, and the description includes the case where the heterocyclic group is substituted with an alkyl group and the case where the heterocyclic group is not substituted with an alkyl group.
  • Substituted refers to one or more hydrogen atoms in the group, preferably up to 5, more preferably 1 to 3, hydrogen atoms, independently of each other, substituted by a corresponding number of substituents. It goes without saying that the substituents are only in their possible chemical positions, and those skilled in the art will be able to determine (by experiment or theory) substitutions that may or may not be possible without undue effort. For example, an amino group or a hydroxyl group having a free hydrogen may be unstable when combined with a carbon atom having an unsaturated (e.g., olefinic) bond.
  • “Pharmaceutical composition” means a mixture comprising one or more of the compounds described herein, or a physiologically/pharmaceutically acceptable salt or prodrug thereof, and other chemical components, as well as other components such as physiological/pharmaceutically acceptable carriers. And excipients.
  • the purpose of the pharmaceutical composition is to promote the administration of the organism, which facilitates the absorption of the active ingredient and thereby exerts biological activity.
  • “Pharmaceutically acceptable salt” refers to a salt of a compound of the invention which is safe and effective for use in a mammal and which possesses the desired biological activity.
  • the preparation method of the salt used includes the following steps:
  • a compound of the formula (I-1) and a compound of the formula (I-2) are subjected to a nucleophilic substitution reaction under basic conditions to give a compound of the formula (I-3);
  • the compound of the formula (I-3) is hydrolyzed under basic conditions to give a compound of the formula (V);
  • the third step the compound of the formula (V) and the compound of the formula (VI) or a pharmaceutically acceptable salt thereof are subjected to a condensation reaction under basic conditions in the presence of a condensing agent to obtain a compound of the formula (I).
  • X is a halogen
  • R a is an alkyl group, preferably a methyl group or an ethyl group
  • Ring A, Ring B, G 1 to G 3 , R 1 to R 10 , n, s and t are as defined in the formula (I).
  • the reagents providing basic conditions include organic bases including, but not limited to, triethylamine, N,N-diisopropylethylamine, n-butyllithium, lithium diisopropylamide, and inorganic bases. , potassium acetate, sodium acetate, aqueous ammonia, sodium t-butoxide or potassium t-butoxide, said inorganic bases including but not limited to sodium hydride, sodium hydroxide, potassium hydroxide, lithium hydroxide, potassium phosphate, sodium carbonate, Sodium bicarbonate, potassium carbonate or cesium carbonate.
  • Condensing agents include, but are not limited to, 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride, N,N'-dicyclohexylcarbodiimide, N,N'-di Propylcarbodiimide, O-benzotriazole-N,N,N',N'-tetramethyluronium tetrafluoroborate, 1-hydroxybenzotriazole, 1-hydroxy-7- Azobenzotriazole, O-benzotriazole-N,N,N',N'-tetramethyluron hexafluorophosphate, 2-(7-oxobenzotriazole)-N,N , N', N'-tetramethylurea hexafluorophosphate, 2-(7-azobenzotriazole)-N,N,N',N'-tetramethylurea hexafluorophosphate, benzene And triazol-1-
  • the preparation method of the salt used includes the following steps:
  • a compound of the formula (II-1) and a compound of the formula (I-2) are subjected to a nucleophilic substitution reaction under basic conditions to obtain a compound of the formula (II-2);
  • the compound of the formula (II-2) is hydrolyzed under basic conditions to give a compound of the formula (II-3);
  • the compound of the formula (II-3) and the compound of the formula (II-4) or a pharmaceutically acceptable salt thereof are subjected to a condensation reaction under basic conditions in the presence of a condensing agent to obtain a formula (II).
  • X is a halogen
  • R a is an alkyl group, preferably a methyl group or an ethyl group
  • R 1 , R 3 to R 9 , n and t are as defined in the formula (II).
  • the reagents providing basic conditions include organic bases including, but not limited to, triethylamine, N,N-diisopropylethylamine, n-butyllithium, lithium diisopropylamide, and inorganic bases. , potassium acetate, sodium acetate, aqueous ammonia, sodium t-butoxide or potassium t-butoxide, said inorganic bases including but not limited to sodium hydride, sodium hydroxide, potassium hydroxide, lithium hydroxide, potassium phosphate, sodium carbonate, Sodium bicarbonate, potassium carbonate or cesium carbonate.
  • Condensing agents include, but are not limited to, 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride, N,N'-dicyclohexylcarbodiimide, N,N'-di Propylcarbodiimide, O-benzotriazole-N,N,N',N'-tetramethyluronium tetrafluoroborate, 1-hydroxybenzotriazole, 1-hydroxy-7- Azobenzotriazole, O-benzotriazole-N,N,N',N'-tetramethyluron hexafluorophosphate, 2-(7-oxobenzotriazole)-N,N , N', N'-tetramethylurea hexafluorophosphate, 2-(7-azobenzotriazole)-N,N,N',N'-tetramethylurea hexafluorophosphate, benzene And triazol-1-
  • the preparation method of the salt used includes the following steps:
  • a compound of the formula (II-1) and a compound of the formula (III-1) are subjected to a nucleophilic substitution reaction under basic conditions to obtain a compound of the formula (III-2);
  • the compound of the formula (III-2) is hydrolyzed under basic conditions to give a compound of the formula (III-3);
  • the compound of the formula (III-3) and the compound of the formula (III-4) or a pharmaceutically acceptable salt thereof are subjected to a condensation reaction under basic conditions in the presence of a condensing agent to obtain a formula (III).
  • X is a halogen
  • R a is an alkyl group, preferably a methyl group or an ethyl group
  • R 1 , R 5 , R 6 , R 11 , R 14 , L 1 , n and t are as defined in the formula (III).
  • the reagents providing basic conditions include organic bases including, but not limited to, triethylamine, N,N-diisopropylethylamine, n-butyllithium, lithium diisopropylamide, and inorganic bases. , potassium acetate, sodium acetate, aqueous ammonia, sodium t-butoxide or potassium t-butoxide, said inorganic bases including but not limited to sodium hydride, sodium hydroxide, potassium hydroxide, lithium hydroxide, potassium phosphate, sodium carbonate, Sodium bicarbonate, potassium carbonate or cesium carbonate.
  • Condensing agents include, but are not limited to, 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride, N,N'-dicyclohexylcarbodiimide, N,N'-di Propylcarbodiimide, O-benzotriazole-N,N,N',N'-tetramethyluronium tetrafluoroborate, 1-hydroxybenzotriazole, 1-hydroxy-7- Azobenzotriazole, O-benzotriazole-N,N,N',N'-tetramethyluron hexafluorophosphate, 2-(7-oxobenzotriazole)-N,N , N', N'-tetramethylurea hexafluorophosphate, 2-(7-azobenzotriazole)-N,N,N',N'-tetramethylurea hexafluorophosphate, benzene And triazol-1-
  • R 1 , R 5 , R 6 , R 11 , R 14 , L 1 , n and t are as defined in the formula (IV).
  • the reagents providing basic conditions include organic bases including, but not limited to, triethylamine, N,N-diisopropylethylamine, n-butyllithium, lithium diisopropylamide, and inorganic bases. , potassium acetate, sodium acetate, aqueous ammonia, sodium t-butoxide or potassium t-butoxide, said inorganic bases including but not limited to sodium hydride, sodium hydroxide, potassium hydroxide, lithium hydroxide, potassium phosphate, sodium carbonate, Sodium bicarbonate, potassium carbonate or cesium carbonate.
  • Condensing agents include, but are not limited to, 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride, N,N'-dicyclohexylcarbodiimide, N,N'-di Propylcarbodiimide, O-benzotriazole-N,N,N',N'-tetramethyluronium tetrafluoroborate, 1-hydroxybenzotriazole, 1-hydroxy-7- Azobenzotriazole, O-benzotriazole-N,N,N',N'-tetramethyluron hexafluorophosphate, 2-(7-oxobenzotriazole)-N,N , N', N'-tetramethylurea hexafluorophosphate, 2-(7-azobenzotriazole)-N,N,N',N'-tetramethylurea hexafluorophosphate, benzene And triazol-1-
  • Figure 1 Effect of the compound of Example 1 alone or in combination with anti-mouse-PD-1 antibody on the growth of MC38 colorectal tumors in C57BL/6 mice.
  • the structure of the compound is determined by nuclear magnetic resonance (NMR) or/and mass spectrometry (MS).
  • NMR shift ( ⁇ ) is given in units of 10 -6 (ppm).
  • NMR was measured using a Bruker AVANCE-400 nuclear magnetic apparatus, and the solvent was deuterated dimethyl sulfoxide (DMSO-d 6 ), deuterated chloroform (CDCl 3 ), deuterated methanol (CD 3 OD), internal standard was four.
  • DMSO-d 6 dimethyl sulfoxide
  • CDCl 3 deuterated chloroform
  • CD 3 OD deuterated methanol
  • TMS Methyl silane
  • MS-2020 The measurement of MS was carried out using a SHIMAZU liquid chromatography-mass spectrometer (manufacturer: Shimazu, model: LC-20AD, LCMS-2020).
  • the thin layer chromatography silica gel plate uses Agela MF254 silica gel plate, and the silica gel plate used for thin layer chromatography (TLC) has a specification of 0.15 mm to 0.2 mm, and the thin layer chromatography separation and purification product has a specification of 0.4 mm to 0.5 mm.
  • the CombiFlash Rapid Preparer uses the Combiflash Rf200 (TELEDYNE ISCO).
  • the known starting materials of the present invention may be synthesized by or according to methods known in the art, or may be purchased from ABCR GmbH & Co. KG, Acros Organics, Aldrich Chemical Company, Accela ChemBio Inc, Dari Chemicals, Shanghai Bi De Pharmaceutical Technology Co., Ltd. and other companies.
  • the reactions can all be carried out under an argon atmosphere or a nitrogen atmosphere.
  • An argon atmosphere or a nitrogen atmosphere means that the reaction flask is connected to an argon or nitrogen balloon having a volume of about 1 L.
  • the hydrogen atmosphere means that the reaction flask is connected to a hydrogen balloon of about 1 L volume.
  • the pressurized hydrogenation reaction was carried out using a Parr Model 3916EKX hydrogenation apparatus and a clear blue QL-500 type hydrogen generator or a HC2-SS type hydrogenation apparatus.
  • the hydrogenation reaction is usually evacuated, charged with hydrogen, and operated three times.
  • the microwave reaction used a CEM Discover-S Model 908860 microwave reactor.
  • the solution means an aqueous solution.
  • reaction temperature is room temperature and is 20 ° C to 30 ° C.
  • the progress of the reaction in the examples was monitored by thin layer chromatography (TLC), the developing agent used for the reaction, the column chromatography eluent system used for the purification of the compound, and the thin layer chromatography developing solvent system including: A: Methylene chloride/methanol system, B: n-hexane/ethyl acetate system, C: n-hexane/ethyl acetate/ethanol, D: petroleum ether/ethyl acetate, the volume ratio of the solvent is adjusted according to the polarity of the compound It can also be adjusted by adding a small amount of alkaline or acidic reagents such as triethylamine and acetic acid.
  • TLC thin layer chromatography
  • A Methylene chloride/methanol system
  • B n-hexane/ethyl acetate system
  • C n-hexane/ethyl acetate/ethanol
  • D petroleum ether/ethyl acetate
  • the compound 1h (530 mg, 1.24 mmol) was dissolved in 8 mL of tetrahydrofuran, and a 1 M solution of tri-bromoborane tetrahydrofuran (1.52 mL, 1.52 mmol) was added at room temperature, and the reaction was stirred at room temperature for 30 minutes, and the mixture was heated to 75 ° C and stirred for 45 minutes. 0.6 mL of a 1 M solution of tridecaneborane tetrahydrofuran was added, and the reaction was stirred at 75 ° C for 45 minutes. The reaction mixture was added to 5 mL of methanol, and the mixture was stirred at 75 ° C for 10 min.
  • the deuterated lithium aluminum hydride (66 mg, 1.57 mmol) was suspended in 2 mL of tetrahydrofuran, cooled to 0 ° C, and 1 mL of a pre-formed compound 2b (0.25 g, 0.81 mmol) in tetrahydrofuran was added dropwise, and the reaction was stirred at 0 ° C for 2 hours. .
  • a small amount of a mixed solvent of methanol and water (V: V 1:1) was added to the reaction mixture, and the resulting suspension was filtered through Celite, and the filtrate was purified using a CombiFlash rapid preparation apparatus to eluent system B to obtain a residue.
  • the title compound 2c (86 mg, yield: 39.6%).
  • reaction liquid was purified by high performance liquid chromatography (Shimadzu SPD-20A high pressure liquid chromatography, Phenomenex Gemini-NX5uM C18 21.2 ⁇ 100 mm column, elution system: trifluoroacetic acid, water, acetonitrile) to give the title compound 2 ( 3 mg, yield: 26.7%).
  • Sodium hydroxide (410 mg, 10.28 mmol) was dissolved in water (20 mL), and 5 mL of potassium dihydrate hydrate (60 mg, 137.11 ⁇ mol) was added, and 15 mL of pre-formed tert-butyl carbamate (98%, 1.5 g) , 12.00 mmol) of the n-propanol solution was added to the above sodium hydroxide solution, cooled in an ice water bath, t-butyl hypochlorite (1.2 g, 10.28 mmol) was added dropwise, stirred for 5 minutes, and 5 mL of pre-formed hydrogenated quinidine 1 was added.
  • the reaction solution was concentrated under reduced pressure and purified by high-purity liquid chromatography (Waters 2767-SQ high-pressure liquid chromatography, sunfire OBD, 150*19 mm 5nfi column, elution system: ammonium hydrogencarbonate, water, acetonitrile).
  • Example 6 Using the synthesis procedure of Example 6, the starting compound 6b was replaced with deuterated methyl iodide to give the title compound 7.
  • the compound 3 g (10 mg, 25.01 ⁇ mol) was dissolved in 2 mL of N,N-dimethylformamide, and the compound 2-amino-2-(4-ethanesulfonylphenyl)ethanol 8a (8.67 mg, 37.83 ⁇ mol) was added.
  • N,N-diisopropylethylamine (6.47 mg, 50.03 ⁇ mol) followed by 2-(7-azobenzotriazole)-N,N,N',N'-tetramethylurea hexafluoro Phosphate ester (11.77 mg, 50.03 ⁇ mol) was stirred at room temperature for 2 hours.
  • Example 6 A synthetic procedure similar to that of Example 6 can be employed to synthesize from the corresponding starting materials.
  • the reaction solution was concentrated under reduced pressure and purified by high-purity liquid chromatography (Waters 2767-SQ high pressure liquid chromatography, sunfire OBD, 150*19 mm 5nfi column, elution system: ammonium hydrogencarbonate, water, acetonitrile).
  • the corresponding fractions were collected and concentrated under reduced pressure to give the title compound 23 (100 mg, yield: 25.57%) and 24 (10 mg, yield: 2%).
  • Test Example 1 Determination of in vitro activity of ROR ⁇ by the compound of the present invention
  • TR-FRET ROR ⁇ co-activation system (Life Technologies)
  • Modulation of RORy activity by the compounds of the invention was screened using a LanthaScreen TR-FRET (Time Resolved Fluorescence Energy Resonance Transfer) ROR ⁇ co-activation system.
  • LanthaScreen TR-FRET Time Resolved Fluorescence Energy Resonance Transfer
  • the Complete TR-FRET Coregulator (Life Technologies) was first formulated to contain a final concentration of 5 mM DTT. The final concentration of DMSO was 2%. The test compound was serially diluted to 2 x final concentration in intact buffer D containing 2% DMSO at a maximum dose of 60 ⁇ m. 10 ⁇ l/well was added to the test well of a 384-well plate (PerkinElmer). Two parallel control wells were placed at the same concentration for each test compound. Prepare 4X ROR ⁇ LBD (AB Vector). The ROR ⁇ LBD concentration was diluted to 1 ng/ ⁇ L using intact buffer D. 5 ⁇ l/well was added to the test well of a 384-well assay plate.
  • the negative control wells were 5 [mu]L of intact buffer D without ROR[gamma]LBD.
  • Fluorescence readings were detected using a Tecan Infinite M1000, and a logarithmic curve of the ratio of the emission wavelength of 520 nm / 495 nm to the concentration of the compound was plotted by GraphPad Prism 6.0 software to calculate the EC 50 value of the test compound.
  • Table 1 EC 50 values of the compounds of the invention for ROR ⁇ in vitro activity
  • the compounds of the present invention have significant agonistic effects on the in vitro activity of ROR ⁇ .
  • Test Example 2 Quantitative Analysis of Activity of IL-17A Enzyme-Linked Immunosorbent Assay
  • PBMC Human peripheral blood mononuclear cells
  • PBMC peripheral blood mononuclear cells
  • Table 2 EC 50 values of quantitative analysis of IL-17A enzyme-linked immunosorbent assay of the compounds of the present invention
  • the compounds of the present invention have a significant regulatory effect on the activity of IL-17A enzyme-linked immunoassay.
  • Test Example 3 Mouse pharmacokinetic test of the compound of the present invention
  • mice Using mice as test animals, the concentration of the drug in the plasma at different times after administration of the compound of Example 1 and Example 6 by intragastric administration was determined by LC/MS/MS method. The pharmacokinetic behavior of the compounds of the invention in mice was investigated and their pharmacokinetic characteristics were evaluated.
  • a certain amount of the drug was weighed, and 5% by volume of DMSO, 5% by volume of Tween 80 and 90% of physiological saline were placed in a 0.1 mg/ml colorless clear liquid.
  • mice were intragastrically administered overnight after fasting, and the dose was 2.0 mg/kg, and the administration volume was 0.2 ml/10 g.
  • Example 1 The compound of Example 1 and the compound of Example 6 were administered by gavage in mice, and 0.1 ml of blood was collected before administration and after 0.25, 0.5, 1.0, 2.0, 4.0, 6.0, 8.0, 11.0, 24.0 hours after administration (each time point) Three animals were placed in heparinized tubes, and the plasma was separated by centrifugation at 3500 rpm for 10 minutes and stored at -20 °C.
  • the content of the test compound in the plasma of mice after different doses of the drug was measured: 25 ⁇ l of the mouse plasma at each time after administration, 80 ⁇ l of camptothecin (100 ng/mL), 200 ⁇ l of acetonitrile, vortex The mixture was spun for 5 minutes, centrifuged for 10 minutes (3600 rpm), and plasma samples were taken for 1 ⁇ l of the supernatant for LC/MS/MS analysis.
  • the pharmacokinetic parameters of the compounds of the invention are as follows:
  • the compounds of the present invention have better pharmacological absorption and have pharmacokinetic advantages.
  • the MC38 mouse model was used to evaluate the inhibitory effect of the compound of Example 1 on the growth of MC38 colorectal tumors.
  • mice purchased from Charles River Lab (USA), were purchased at 20-25 grams, 7-9 weeks old. 10 / cage feeding, temperature 23 ⁇ 1 ° C constant temperature, humidity 50 ⁇ 60%, free to eat water. Everything is treated and used in accordance with the Laboratory Animal Care and Use Committee (IACUC approved guidelines). After the animals were purchased, the experiment was started after 7 days of adaptive feeding.
  • IACUC approved guidelines Laboratory Animal Care and Use Committee
  • Anti-mouse PD-1 (CD279) antibody was purchased from BioXcell (clone RMP1-14; catalog number BP0146)
  • IgG2a isotype control antibody was purchased from BioXcell (clone 2A3; catalog number BE0089)
  • mice After adaptive breeding in mice, the groups are as follows:
  • 1.Q3dx4 means to be administered every three days for a total of four times
  • QDx20 represents one dose per day for 20 consecutive days
  • IgG2a and PD-1 antibodies are administered by intraperitoneal injection, and the compound of Example 1 and the carrier are administered orally.
  • mice Female C57BL/6 mice (20-25 grams, 7-9 weeks old) were used in the experiment.
  • the in vivo antitumor activity of the compound of Example 1 or the compound of Example 1 in combination with an anti-mouse-PD-1 antibody was evaluated by detecting the growth of the isotype MC38 colorectal tumor in inbred C57BL/6 mice. 500,000 (5 ⁇ 10 5 ) MC38 cells were implanted subcutaneously into the right abdomen of each mouse. When the tumor grew to 40-80 mm 3 (Day 6), the mice were randomly divided into the above four groups. Group III and Group IV were administered once a day (Compound of Example 1, 0.5 mg/kg) once for 20 consecutive days.
  • mice bearing MC38 tumors were injected intraperitoneally (ip) with anti-mouse PD-1 (CD279) antibody (BioXcell) on day 6, 9, 12, and 15 (5 mg/ Kg).
  • Group I control group
  • the carrier CMC-Na drug formulation is administered in the same manner as the compound of Example 1, the administration mode of the IgG2a isotype control antibody and the anti-mouse PD-1 ( CD279)
  • the antibodies are identical.
  • tumor volume (mm 3 ) l ⁇ w ⁇ h ⁇ 0.5236, where 1 represents the length of the tumor, w represents the width of the tumor, and h represents the height of the tumor.
  • the unit is mm.
  • TGI As shown in Fig. 1, when 0.5 mg/kg of the compound of Example 1 was administered alone, the TGI was 22.5%. When the anti-mouse PD-1 (CD279) antibody (5 mg/kg) was injected alone, the TGI was 39.8%. The compound of Example 1 exhibited a synergistic effect when administered in combination with an anti-mouse PD-1 monoclonal antibody (5 mg/kg) (TGI was 65.5% at 0.5 mg/kg administration).
  • Example 1 alone exhibits antitumor activity in the isogenic MC38 colorectal tumor model, while the compound of Example 1 exhibits a synergistic effect with the PD-1 antibody combination, which also indicates that Example 1 Compounds have biological activities consistent with ROR ⁇ activation (rather than inhibition), opening up new avenues for improving the efficacy of immunotherapy.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Indole Compounds (AREA)

Abstract

一种涉及氘原子取代的吲哚甲酰胺类衍生物、其制备方法及其在医药上的应用。特别地,涉及一种通式(I)所示的氘原子取代的吲哚甲酰胺类衍生物、其制备方法、含有该衍生物的药物组合物,其作为ROR激动剂的用途以及其用于预防和/或治疗肿瘤或癌症的用途,其中通式(I)中的各取代基与说明书中的定义相同。

Description

氘原子取代的吲哚甲酰胺类衍生物、其制备方法及其在医药上的应用 技术领域
本发明属于医药领域,涉及氘(D)原子取代的吲哚甲酰胺类衍生物、其制备方法及其在医药上的应用。特别地,本发明涉及通式(I)所示的氘原子取代的吲哚甲酰胺类衍生物、其制备方法、含有该衍生物的药物组合物,其作为ROR激动剂以及其用于预防和/或治疗肿瘤或癌症的药物中的用途。
背景技术
维甲酸相关孤儿核受体(Retinoid-related orphan receptors,ROR)是核受体家族的成员之一,也是一类配体依赖的转录因子,它能够调控多种生理和生化过程,包括生殖发育、新陈代谢、免疫系统调节等(Mech Dev.1998 Jan,70(1-2:147-53;EMBO J.1998 Jul 15,17(14):3867-77)。ROR家族包括三种类型RORα、RORβ和RORγ(Curr Drug Targets Inflamm Allergy.2004Dec,3(4):395-412),其中RORγ可以在许多组织中表达,包括胸腺、肝脏、肾脏、脂肪和骨骼肌等(Immunity.1998 Dec,9(6):797-806.)。
RORγ有两种亚型:RORγ1和RORγt(RORγ2),其中RORγ1在许多组织中表达,如:胸腺、肌肉、肾脏和肝脏中表达,而RORγt则只在免疫细胞内表达(Eur J Immunol.1999 Dec,29(12):4072-80)。已有文献报道,RORγt能够调节在免疫细胞分化的过程中T细胞的存活,并能激活和促进CD4+、CD8+的细胞分化成辅助T细胞17(Th17)和细胞毒性T细胞(Tc17)(J Immunol.2014 Mar 15,192(6):2564-75),其中TH17和Tc17细胞是一类效应细胞,通过分泌白介素17(IL-17)和其他炎症因子(如IL-21)促进炎症反应、增强获得性免疫反应和自身免疫应答。此外,现有研究证明,通过将Th17和Tc17细胞移植到荷瘤小鼠中,可以明显抑制移植瘤的生长(J Immunol.2010 Apr 15,184(8):4215-27)。Th17还可以招募细胞毒性CD8+T细胞和自然杀伤细胞进入肿瘤微环境,从而杀死肿瘤细胞,达到抗肿瘤的目的(Blood.2009 Aug 6,114(6):1141-9;Clin Cancer Res.2008 Jun 1,14(11):3254-61)。因此,激活RORγt,有可能成为新的抗肿瘤疗法。
目前,已有医药公司开发出RORγt的激动剂,比如Lycera Corp.公司开发的小分子药物LYC-54143。临床前研究表明,LYC-54143可通过两条不同的通路抑制肿瘤生长,表现出优越的抗癌活性。首先,LYC-54143激活RORγt后可通过传统途径调节Th17和Tc17细胞的分化,促进IL-17等其他细胞因子的表达,提高T细胞活性。另外,激活的RORγt可以调节免疫系统中的多种基因表达,抑制细胞检查受体PD-1的表达,从而降低免疫抑制,提高抗癌活性(Oncoimmunology.2016 Nov 4,5(12):e1254854;ACS Chem Biol.2016 Apr 15,11(4):1012-8)。虽然该小分子激动目前已经进入临床II期,但是有关该靶点激动剂的药物仍然非常少,并且无上市药 物出现,已公开的专利有如WO2015171558、WO2008152260、WO2007068580、WO2007068579、WO2005056516、WO2005056510、WO2005066116、WO00228810,仍需要继续开发更高效的新的RORγt激动剂,以期为患者提供新的有效的抗癌药物。
本发明人设计具有通式(I)所示结构的氘原子取代的吲哚甲酰胺类化合物,取代基中含有氘原子,使得本发明化合物取得意料不到的药代吸收活性和药效作用。同时,在环A的邻位有较大的取代基(例如:三氟甲基),表现出显著的激动ROR的作用。本发明还提供了药效试验,单独施用本发明化合物时表现出良好的抑瘤活性,另外,本发明化合物与PD-1抗体联合用药时表现出协同作用,为提高免疫治疗的疗效开辟了新途径。
发明内容
本发明的目的在于提供一种通式(I)所示的化合物:
Figure PCTCN2018105008-appb-000001
或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用的盐,
其中:
Figure PCTCN2018105008-appb-000002
为双键或单键;
G 1、G 2和G 3相同或不同,且各自独立地选自C、CH、CH 2和N;
环A选自芳基、杂芳基、环烷基和杂环基;
环B为芳基或杂芳基;
R 1相同或不同,且各自独立地选自H原子、D原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、氰基、氨基、硝基、羟基和羟烷基;
R 2为卤代烷基;
R 3和R 4相同或不同,且各自独立地选自H原子、D原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟烷基、氰基、氨基、硝基、羟基、环烷基、杂环基、芳基和杂芳基,其中所述的烷基、羟烷基、环烷基、杂环基、芳基和杂芳基各自独立地任选被选自D原子、羟基、卤素、烷基、氨基和-OR 11中的一个或多个取代基所取代;
R 5选自H原子、烷基、卤代烷基、氨基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基,其中所述的烷基、环烷基、杂环基、芳基和杂芳基任选被选自D 原子、卤素、羟基、环烷基和杂环基中的一个或多个取代基所取代;
R 6相同或不同,且各自独立地选自H原子、D原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、氰基、氨基、硝基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基;
R 7选自H原子、烷基、卤代烷基、环烷基、杂环基、芳基和杂芳基,其中所述的烷基、环烷基、杂环基、芳基和杂芳基任选被选自D原子、卤素、羟基、氨基、氰基、硝基、烷氧基、卤代烷氧基、-OR 11、-C(O)R 11、-C(O)OR 11、-NR 12R 13、-C(O)NR 12R 13、-S(O) mR 11、环烷基和杂环基中的一个或多个取代基所取代;
R 8和R 9相同或不同,且各自独立地选自H原子、D原子、卤素、烷基、卤代烷基、烷氧基、氰基、氨基、硝基、羟基和羟烷基;
R 10相同或不同,且各自独立地选自H原子、D原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、氰基、氨基、硝基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基;
R 11选自H原子、D原子、烷基、卤代烷基、羟烷基、环烷基和杂环基,其中所述的烷基、羟烷基、环烷基和杂环基任选被选自D原子、卤素、羟基、环烷基和杂环基中的一个或多个取代基所取代;
R 12和R 13相同或不同,且各自独立地选自H原子、D原子、烷基、卤代烷基、羟基和羟烷基,其中所述的烷基和羟烷基任选被选自D原子、卤素、羟基、环烷基和杂环基中的一个或多个取代基所取代;
条件是通式(I)所示的化合物含有至少一个D原子,特别地,在取代基R 3、R 4、R 5、R 7、R 8、R 9中至少一个取代基含有至少一个D原子;
m为0、1或2;
n为0、1、2、3或4;
s为0、1、2或3;且
t为0、1、2或3。
在本发明一个优选的实施方案中,所述的通式(I)所示的化合物,其中环A选自苯基、吡啶基、咪唑基、吡唑基、哌啶基和吗啉基;且环B为苯基或吡啶基。
在本发明一个优选的实施方案中,所述的通式(I)所示的化合物,其中
Figure PCTCN2018105008-appb-000003
选自:
Figure PCTCN2018105008-appb-000004
Figure PCTCN2018105008-appb-000005
在本发明一个优选的实施方案中,所述的通式(I)所示的化合物为通式(II)所 示的化合物:
Figure PCTCN2018105008-appb-000006
其中:
G为CH或N;
R 1、R 3~R 9、n和t如通式(I)中所定义。
在本发明一个优选的实施方案中,所述的通式(I)所示的化合物,其中R 4为H原子或D原子;R 3选自H原子、D原子和烷基,其中所述的烷基任选被选自D原子、羟基、卤素、氨基和-OR 11中的一个或多个取代基所取代;R 11如通式(I)中所定义。
在本发明一个优选的实施方案中,所述的通式(I)所示的化合物,其中R 7选自烷基、卤代烷基、环烷基和杂环基,其中所述的烷基任选被选自D原子、卤素、羟基、氨基、氰基、硝基、烷氧基、卤代烷氧基和羟烷基中的一个或多个取代基所取代。
在本发明一个优选的实施方案中,所述的通式(I)所示的化合物,其中R 8和R 9相同或不同,且各自独立地为H原子或D原子。
在本发明一个优选的实施方案中,所述的通式(I)所示的化合物为通式(III)所示的化合物:
Figure PCTCN2018105008-appb-000007
其中:
L 1为亚烷基,其中所述的亚烷基任选被卤素和D原子中的一个或多个取代基所取代;
R 14选自D原子、卤素、羟基、氨基、氰基、硝基、烷氧基、卤代烷氧基和羟烷基;
R 1、R 5、R 6、R 11、n和t如通式(I)中所定义。
在本发明一个优选的实施方案中,所述的通式(I)所示的化合物为通式(IV) 所示的化合物:
Figure PCTCN2018105008-appb-000008
其中:
L 1为亚烷基,其中所述的亚烷基任选被卤素和D原子中的一个或多个取代基所取代;
R 14选自D原子、卤素、羟基、氨基、氰基、硝基、烷氧基、卤代烷氧基和羟烷基;
R 1、R 5、R 6、R 11、n和t如通式(I)中所定义。
在本发明一个优选的实施方案中,所述的通式(I)所示的化合物,其中R 1相同或不同,且各自独立地选自H原子、D原子、卤素和烷基。
在本发明一个优选的实施方案中,所述的通式(I)所示的化合物,其中R 5为烷基,其中所述的烷基任选被选自D原子、卤素和羟基中的一个或多个取代基所取代。
在本发明一个优选的实施方案中,所述的通式(I)所示的化合物,其中R 6相同或不同,且各自独立地选自H原子、D原子或卤素。
典型的通式(I)的化合物,包括但不限于:
Figure PCTCN2018105008-appb-000009
Figure PCTCN2018105008-appb-000010
Figure PCTCN2018105008-appb-000011
Figure PCTCN2018105008-appb-000012
Figure PCTCN2018105008-appb-000013
或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐。
本发明的另一方面涉及一种通式(V)所示的化合物,其为制备通式(I)化合物的中间体,
Figure PCTCN2018105008-appb-000014
或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,
其中:
Figure PCTCN2018105008-appb-000015
为双键或单键;
G 1、G 2和G 3相同或不同,且各自独立地选自C、CH、CH 2和N;
环A选自芳基、杂芳基、环烷基和杂环基;
R 1相同或不同,且各自独立地选自H原子、D原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、氰基、氨基、硝基、羟基和羟烷基;
R 2为卤代烷基;
R 6相同或不同,且各自独立地选自H原子、D原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、氰基、氨基、硝基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基;
R 7选自H原子、烷基、卤代烷基、环烷基、杂环基、芳基和杂芳基,其中所述的烷基、环烷基、杂环基、芳基和杂芳基任选被选自D原子、卤素、羟基、氨基、氰基、硝基、烷氧基、卤代烷氧基、-OR 11、-C(O)R 11、-C(O)OR 11、-NR 12R 13、-C(O)NR 12R 13、-S(O) mR 11、环烷基和杂环基中的一个或多个取代基所取代;
R 8和R 9相同或不同,且各自独立地选自H原子、D原子、卤素、烷基、卤代烷基、烷氧基、氰基、氨基、硝基、羟基和羟烷基;
R 11选自H原子、D原子、烷基、卤代烷基、羟烷基、环烷基和杂环基,其中所述的烷基、羟烷基、环烷基和杂环基任选被选自D原子、卤素、羟基、环烷基和杂环基中的一个或多个取代基所取代;
R 12和R 13相同或不同,且各自独立地选自H原子、D原子、烷基、卤代烷基、羟基和羟烷基,其中所述的烷基和羟烷基任选被选自D原子、卤素、羟基、环烷基和杂环基中的一个或多个取代基所取代;
m为0、1或2;
n为0、1、2、3或4;且
t为0、1、2或3。
在本发明一个优选的实施方案中,所述通式(V)所示的化合物,其中取代基 R 7、R 8和R 9中至少一个取代基含有一个或多个D原子。
典型的通式(V)的化合物,包括但不限于:
Figure PCTCN2018105008-appb-000016
Figure PCTCN2018105008-appb-000017
或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐。
本发明的另一方面涉及一种制备通式(I)所示的化合物的方法,该方法包括:
Figure PCTCN2018105008-appb-000018
通式(V)化合物与通式(VI)化合物或其可药用的盐发生缩合反应,得到通式(I)化合物;
其中:
Figure PCTCN2018105008-appb-000019
环A、环B、G 1~G 3、R 1~R 10、n、s和t如通式(I)中所定义。
本发明的另一方面涉及一种制备通式(II)所示的化合物的方法,该方法包括:
Figure PCTCN2018105008-appb-000020
通式(II-3)化合物与通式(II-4)化合物或其可药用的盐发生缩合反应,得到通式(II)化合物;
其中:
G、R 1、R 3~R 9、n和t如通式(II)中所定义。
本发明的另一方面涉及一种制备通式(III)所示的化合物的方法,该方法包括:
Figure PCTCN2018105008-appb-000021
通式(III-3)化合物与通式(III-4)化合物或其可药用的盐发生缩合反应,得到通式(III)化合物;
其中:
R 1、R 5、R 6、R 11、R 14、L 1、n和t如通式(III)中所定义;R 8、R 9如通式(I)中所定义。
本发明的另一方面涉及一种制备通式(IV)所示的化合物的方法,该方法包括:
Figure PCTCN2018105008-appb-000022
通式(III-3)化合物与通式(IV-1)化合物或其可药用的盐发生缩合反应,得到通式(IV)化合物;
其中:
R 1、R 5、R 6、R 11、R 14、L 1、n和t如通式(IV)中所定义;R 8、R 9如通式(I)中所定义。
本发明的另一方面涉及一种药物组合物,其含有治疗有效剂量的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或可药用的盐,以及一种或多种药学上可接受的载体、稀释剂或赋形剂。本发明还涉及一种制备所述药物组合物的方法,其包括将通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、或其混合物形式、或其可药用的盐与药学上可接受的载体、稀释剂或赋形剂相混合。在本发明一个实施方案中,所述药物组合物进一步含有抗PD-1抗体,优选抗鼠PD-1抗体。
本发明进一步涉及通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,或包含其的药物组合物在制备ROR激动剂中的用途。
本发明进一步涉及通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,或包含其的药物组合物在制备用于预防和/或治疗肿瘤或癌症的药物中的用途,特别是作为 ROR激动剂在制备用于预防和/或治疗肿瘤或癌症的药物中的用途。
本发明进一步涉及通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐(作为ROR激动剂),或包含其的药物组合物与抗PD-1抗体的组合在制备用于预防和/或治疗肿瘤或癌症的药物中的用途。
本发明进一步涉及通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,或包含其的药物组合物,其用作药物。
本发明还涉及通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,或包含其的药物组合物,其作为ROR激动剂。
本发明还涉及通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,或包含其的药物组合物,其用于预防和/或治疗肿瘤或癌症,特别是作为ROR激动剂用于预防和/或治疗肿瘤或癌症。
本发明还涉及通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式、或其可药用盐,或包含其的药物组合物与抗PD-1抗体的组合,其用于预防和/或治疗肿瘤或癌症。
本发明还涉及一种治疗预防和/或治疗肿瘤或癌症的方法,其包括向需要其的患者施用治疗有效剂量的作为ROR激动剂的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,或包含其的药物组合物。
本发明还涉及一种治疗预防和/或治疗预防肿瘤或癌症的方法,其包括向需要其的患者施用治疗有效剂量的通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,或包含其的药物组合物和抗PD-1抗体。
本发明所述的肿瘤或癌症选自非霍奇金淋巴瘤、弥漫大B细胞淋巴瘤、滤泡性淋巴瘤、滑膜肉瘤、乳腺癌、宫颈癌、结肠癌、肺癌、胃癌、直肠癌、胰腺癌、脑癌、皮肤癌、口腔癌、前列腺癌、骨癌、肾癌、卵巢癌、膀胱癌、肝癌、输卵管肿瘤、卵巢瘤、腹膜肿瘤、黑色素瘤、实体瘤、神经胶质瘤、神经胶母细胞瘤、肝细胞癌、乳突肾性瘤、头颈部肿瘤、白血病、淋巴瘤、骨髓瘤和非小细胞肺癌。
含活性成分的药物组合物可以是适用于口服的形式,例如片剂、糖锭剂、锭剂、水或油混悬液、可分散粉末或颗粒、乳液、硬或软胶囊,或糖浆剂或酏剂。可按照本领域任何已知制备药用组合物的方法制备口服组合物,此类组合物可含有一种或多种选自以下的成分:甜味剂、矫味剂、着色剂和防腐剂,以提供悦目和可口的药用制剂。片剂含有活性成分和用于混合的适宜制备片剂的无毒的可药 用的赋形剂。这些赋形剂可以是惰性赋形剂,造粒剂、崩解剂,粘合剂,和润滑剂,。这些片剂可以不包衣或可通过掩盖药物的味道或在胃肠道中延迟崩解和吸收,因而在较长时间内提供缓释作用的已知技术将其包衣。
也可用其中活性成分与惰性固体稀释剂或其中活性成分与水溶性载体或油溶媒混合的软明胶胶囊提供口服制剂。
水悬浮液含有活性物质和用于混合的适宜制备水悬浮液的赋形剂。此类赋形剂是悬浮剂,分散剂或湿润剂。水混悬液也可以含有一种或多种防腐剂、一种或多种着色剂、一种或多种矫味剂和一种或多种甜味剂。
油混悬液可通过使活性成分悬浮于植物油,或矿物油配制而成。油悬浮液可含有增稠剂。可加入上述的甜味剂和矫味剂,以提供可口的制剂。可通过加入抗氧化剂保存这些组合物。
本发明的药物组合物也可以是水包油乳剂的形式。油相可以是植物油,或矿物油或其混合物。适宜的乳化剂可以是天然产生的磷脂,乳剂也可以含有甜味剂、矫味剂、防腐剂和抗氧剂。此类制剂也可含有缓和剂、防腐剂、着色剂和抗氧剂。
本发明的药物组合物可以是无菌注射水溶液形式。可以使用的可接受的溶媒或溶剂有水、林格氏液和等渗氯化钠溶液。无菌注射制剂可以是其中活性成分溶于油相的无菌注射水包油微乳可通过局部大量注射,将注射液或微乳注入患者的血流中。或者,最好按可保持本发明化合物恒定循环浓度的方式给予溶液和微乳。为保持这种恒定浓度,可使用连续静脉内递药装置。这种装置的实例是Deltec CADD-PLUS.TM.5400型静脉注射泵。
本发明的药物组合物可以是用于肌内和皮下给药的无菌注射水或油混悬液的形式。可按已知技术,用上述那些适宜的分散剂或湿润剂和悬浮剂配制该混悬液。无菌注射制剂也可以是在肠胃外可接受的无毒稀释剂或溶剂中制备的无菌注射溶液或混悬液。此外,可方便地用无菌固定油作为溶剂或悬浮介质。为此目的,可使用任何调和固定油。此外,脂肪酸也可以制备注射剂。
可按用于直肠给药的栓剂形式给予本发明化合物。可通过将药物与在普通温度下为固体但在直肠中为液体,因而在直肠中会溶化而释放药物的适宜的无刺激性赋形剂混合来制备这些药物组合物。
如本领域技术人员所熟知的,药物的给药剂量依赖于多种因素,包括但并非限定于以下因素:所用具体化合物的活性、患者的年龄、患者的体重、患者的健康状况、患者的行为、患者的饮食、给药时间、给药方式、排泄的速率、药物的组合等;另外,最佳的治疗方式如治疗的模式、通式化合物(I)的日用量或可药用的盐的种类可以根据传统的治疗方案来验证。
发明的详细说明
除非有相反陈述,在说明书和权利要求书中使用的术语具有下述含义。
术语“烷基”指饱和脂肪族烃基团,其为包含1至20个碳原子的直链或支链基团,优选含有1至12个碳原子的烷基,更优选含有1至6个碳原子的烷基。非限制性实例包括甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基、正庚基、2-甲基己基、3-甲基己基、4-甲基己基、5-甲基己基、2,3-二甲基戊基、2,4-二甲基戊基、2,2-二甲基戊基、3,3-二甲基戊基、2-乙基戊基、3-乙基戊基、正辛基、2,3-二甲基己基、2,4-二甲基己基、2,5-二甲基己基、2,2-二甲基己基、3,3-二甲基己基、4,4-二甲基己基、2-乙基己基、3-乙基己基、4-乙基己基、2-甲基-2-乙基戊基、2-甲基-3-乙基戊基、正壬基、2-甲基-2-乙基己基、2-甲基-3-乙基己基、2,2-二乙基戊基、正癸基、3,3-二乙基己基、2,2-二乙基己基,及其各种支链异构体等。更优选的是含有1至6个碳原子的低级烷基,非限制性实施例包括甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基等。烷基可以是取代的或非取代的,当被取代时,取代基可以在任何可使用的连接点上被取代,其独立地选自氘原子、烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基、羧酸酯基、-OR 11、-C(O)R 11、-C(O)OR 11、-NR 12R 13、-C(O)NR 12R 13和-S(O) mR 11中的一个或多个取代基所取代。
术语“亚烷基”指饱和的直链或支链脂肪族烃基,其具有2个从母体烷的相同碳原子或两个不同的碳原子上除去两个氢原子所衍生的残基,其为包含1至20个碳原子的直链或支链基团,优选含有1至12个碳原子,更优选含有1至6个碳原子的亚烷基。亚烷基的非限制性实例包括但不限于亚甲基(-CH 2-)、1,1-亚乙基(-CH(CH 3)-)、1,2-亚乙基(-CH 2CH 2)-、1,1-亚丙基(-CH(CH 2CH 3)-)、1,2-亚丙基(-CH 2CH(CH 3)-)、1,3-亚丙基(-CH 2CH 2CH 2-)、1,4-亚丁基(-CH 2CH 2CH 2CH 2-)和1,5-亚戊基(-CH 2CH 2CH 2CH 2CH 2-)等。亚烷基可以是取代的或非取代的,当被取代时,取代基可以在任何可使用的连接点上被取代,所述取代基独立地任选选自氘原子、烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、-OR 11、-C(O)R 11、-C(O)OR 11、-NR 12R 13、-C(O)NR 12R 13和-S(O) mR 11中的一个或多个取代基所取代。
术语“烷氧基”指-O-(烷基)和-O-(非取代的环烷基),其中烷基和环烷基的定义 如上所述。烷氧基的非限制性实例包括:甲氧基、乙氧基、丙氧基、丁氧基、环丙氧基、环丁氧基、环戊氧基、环己氧基。烷氧基可以是任选取代的或非取代的,当被取代时,其独立地选自氘原子、烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基、羧酸酯基、-OR 11、-C(O)R 11、-C(O)OR 11、-NR 12R 13、-C(O)NR 12R 13和-S(O) mR 11中的一个或多个取代基所取代。
术语“环烷基”指饱和或部分不饱和单环或多环环状烃取代基,环烷基环包含3至20个碳原子,优选包含3至12个碳原子,更优选包含3至6个碳原子。单环环烷基的非限制性实例包括环丙基、环丁基、环戊基、环戊烯基、环己基、环己烯基、环己二烯基、环庚基、环庚三烯基、环辛基等;多环环烷基包括螺环、稠环和桥环的环烷基。
术语“螺环烷基”指5至20元的单环之间共用一个碳原子(称螺原子)的多环基团,其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。优选为6至14元,更优选为7至10元。根据环与环之间共用螺原子的数目将螺环烷基分为单螺环烷基、双螺环烷基或多螺环烷基,优选为单螺环烷基和双螺环烷基。更优选为4元/4元、4元/5元、4元/6元、5元/5元或5元/6元单螺环烷基。螺环烷基的非限制性实例包括:
Figure PCTCN2018105008-appb-000023
术语“稠环烷基”指5至20元,系统中的每个环与体系中的其他环共享毗邻的一对碳原子的全碳多环基团,其中一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环稠环烷基,优选为双环或三环,更优选为5元/5元或5元/6元双环烷基。稠环烷基的非限制性实例包括:
Figure PCTCN2018105008-appb-000024
术语“桥环烷基”指5至20元,任意两个环共用两个不直接连接的碳原子的全碳多环基团,其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环桥环烷基,优选为双环、三环或四环,更有选为双环或三环。桥环烷基的非限制性实例包括:
Figure PCTCN2018105008-appb-000025
所述环烷基环可以稠合于芳基、杂芳基或杂环烷基环上,其中与母体结构连接在一起的环为环烷基,非限制性实例包括茚满基、四氢萘基、苯并环庚烷基等。环烷基可以是任选取代的或非取代的,当被取代时,其独立地选自氘原子、烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基、羧酸酯基、-OR 11、-C(O)R 11、-C(O)OR 11、-NR 12R 13、-C(O)NR 12R 13和-S(O) mR 11中的一个或多个取代基所取代。
术语“杂环基”指饱和或部分不饱和单环或多环环状烃取代基,其包含3至20个环原子,其中一个或多个环原子为选自氮、氧或S(O) m(其中m是整数0至2)的杂原子,但不包括-O-O-、-O-S-或-S-S-的环部分,其余环原子为碳。优选包含3至12个环原子,其中1~4个是杂原子;最优选包含3至8个环原子,其中1~3是杂原子;最优选包含3至6个环原子,其中1~2是杂原子。单环杂环基的非限制性实例包括吡咯烷基、咪唑烷基、四氢呋喃基、四氢噻吩基、二氢咪唑基、二氢呋喃基、二氢吡唑基、二氢吡咯基、哌啶基、哌嗪基、吗啉基、硫代吗啉基、高哌嗪基、吡喃基等,优选哌啶基、哌嗪基或吗啉基。多环杂环基包括螺环、稠环和桥环的杂环基。
术语“螺杂环基”指5至20元的单环之间共用一个原子(称螺原子)的多环杂环基团,其中一个或多个环原子为选自氮、氧或S(O) m(其中m是整数0至2)的杂原子,其余环原子为碳。其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统。优选为6至14元,更优选为7至10元。根据环与环之间共用螺原子的数目将螺杂环基分为单螺杂环基、双螺杂环基或多螺杂环基,优选为单螺杂环基和双螺杂环基。更优选为3元/6元、4元/4元、4元/5元、4元/6元、5元/5元或5元/6元单螺杂环基。螺杂环基的非限制性实例包括:
Figure PCTCN2018105008-appb-000026
术语“稠杂环基”指5至20元,系统中的每个环与体系中的其他环共享毗邻的一对原子的多环杂环基团,一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统,其中一个或多个环原子为选自氮、氧或S(O) m(其 中m是整数0至2)的杂原子,其余环原子为碳。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环稠杂环基,优选为双环或三环,更优选为5元/5元或5元/6元双环稠杂环基。稠杂环基的非限制性实例包括:
Figure PCTCN2018105008-appb-000027
术语“桥杂环基”指5至14元,任意两个环共用两个不直接连接的原子的多环杂环基团,其可以含有一个或多个双键,但没有一个环具有完全共轭的π电子系统,其中一个或多个环原子为选自氮、氧或S(O) m(其中m是整数0至2)的杂原子,其余环原子为碳。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环桥杂环基,优选为双环、三环或四环,更有选为双环或三环。桥杂环基的非限制性实例包括:
Figure PCTCN2018105008-appb-000028
所述杂环基环可以稠合于芳基、杂芳基或环烷基环上,其中与母体结构连接在一起的环为杂环基,其非限制性实例包括:
Figure PCTCN2018105008-appb-000029
等。
杂环基可以是任选取代的或非取代的,当被取代时,其独立地选自氘原子、烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氧代基、羧基、羧酸酯基、-OR 11、-C(O)R 11、-C(O)OR 11、-NR 12R 13、-C(O)NR 12R 13和-S(O) mR 11中的一个或多个取代基所取代。
术语“芳基”指具有共轭的π电子体系的6至14元全碳单环或稠合多环(也就是共享毗邻碳原子对的环)基团,优选为6至10元,例如苯基和萘基。更优选苯基。所述芳基环可以稠合于杂芳基、杂环基或环烷基环上,其中与母体结构连接在一 起的环为芳基环,其非限制性实例包括:
Figure PCTCN2018105008-appb-000030
芳基可以是取代的或非取代的,当被取代时,其独立地选自氘原子、烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基、羧酸酯基、-OR 11、-C(O)R 11、-C(O)OR 11、-NR 12R 13、-C(O)NR 12R 13和-S(O) mR 11中的一个或多个取代基所取代。
术语“杂芳基”指包含1至4个杂原子、5至14个环原子的杂芳族体系,其中杂原子选自氧、硫和氮。杂芳基优选为5至10元,含1至3个杂原子;更优选为5元或6元,含1至2个杂原子;优选例如咪唑基、呋喃基、噻吩基、噻唑基、吡唑基、噁唑基、吡咯基、四唑基、吡啶基、嘧啶基、噻二唑、吡嗪基等,优选为咪唑基、四唑基、吡啶基、噻吩基、吡唑基或嘧啶基、噻唑基;更有选吡啶基。所述杂芳基环可以稠合于芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为杂芳基环,其非限制性实例包括:
Figure PCTCN2018105008-appb-000031
杂芳基可以是任选取代的或非取代的,当被取代时,其独立地选自氘原子、烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、巯基、羟基、硝基、氰基、环烷基、杂环烷基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、羧基、羧酸酯基、-OR 11、-C(O)R 11、-C(O)OR 11、-NR 12R 13、-C(O)NR 12R 13和-S(O) mR 11中的一个或多个取代基所取代。
术语“卤代烷基”指被一个或多个卤素取代的烷基,其中烷基如上所定义。
术语“卤代烷氧基”指被一个或多个卤素取代的烷氧基,其中烷氧基如上所定义。
术语“羟烷基”指被羟基取代的烷基,其中烷基如上所定义。
术语“羟基”指-OH基团。
术语“卤素”指氟、氯、溴或碘。
术语“氨基”指-NH 2
术语“氰基”指-CN。
术语“硝基”指-NO 2
术语“氧代基”指=O。
术语“羰基”指C=O。
术语“羧基”指-C(O)OH。
术语“羧酸酯基”指-C(O)O(烷基)或-C(O)O(环烷基),其中烷基、环烷基如上所定义。
术语“酰卤”指含有-C(O)-卤素的基团的化合物。
本发明还包括各种氘化形式的式(I)化合物。与碳原子连接的各个可用的氢原子可独立地被氘原子替换。本领域技术人员能够参考相关文献合成氘化形式的式(I)化合物。在制备氘代形式的式(I)化合物时可使用市售的氘代起始物质,或它们可使用常规技术采用氘代试剂合成,氘代试剂包括但不限于氘代硼烷、三氘代硼烷四氢呋喃溶液、氘代氢化锂铝、氘代碘乙烷和氘代碘甲烷等。
“任选”或“任选地”意味着随后所描述的事件或环境可以但不必发生,该说明包括该事件或环境发生或不发生的场合。例如,“任选被烷基取代的杂环基团”意味着烷基可以但不必须存在,该说明包括杂环基团被烷基取代的情形和杂环基团不被烷基取代的情形。
“取代的”指基团中的一个或多个氢原子,优选为最多5个,更优选为1~3个氢原子彼此独立地被相应数目的取代基取代。不言而喻,取代基仅处在它们的可能的化学位置,本领域技术人员能够在不付出过多努力的情况下确定(通过实验或理论)可能或不可能的取代。例如,具有游离氢的氨基或羟基与具有不饱和(如烯属)键的碳原子结合时可能是不稳定的。
“药物组合物”表示含有一种或多种本文所述化合物或其生理学上/可药用的盐或前体药物与其他化学组分的混合物,以及其他组分例如生理学/可药用的载体和赋形剂。药物组合物的目的是促进对生物体的给药,利于活性成分的吸收进而发挥生物活性。
“可药用盐”是指本发明化合物的盐,这类盐用于哺乳动物体内时具有安全性和有效性,且具有应有的生物活性。
本发明化合物的合成方法
为了完成本发明的目的,本发明采用如下技术方案:
方案一
本发明通式(I)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用的盐的制备方法,包括以下步 骤:
Figure PCTCN2018105008-appb-000032
第一步,通式(I-1)化合物和通式(I-2)化合物在碱性条件下,发生亲核取代反应,得到通式(I-3)化合物;
第二步,通式(I-3)化合物在碱性条件下水解,得到通式(V)化合物;
第三步,通式(V)化合物和通式(VI)化合物或其可药用的盐,在碱性条件下,在缩合剂存在下,发生缩合反应,得到通式(I)化合物,
其中:
X为卤素;
R a为烷基,优选为甲基或乙基;
Figure PCTCN2018105008-appb-000033
环A、环B、G 1~G 3、R 1~R 10、n、s和t如通式(I)中所定义。
提供碱性条件的试剂包括有机碱和无机碱类,所述的有机碱类包括但不限于三乙胺、N,N-二异丙基乙胺、正丁基锂、二异丙基氨基锂、醋酸钾、醋酸钠、氨水、叔丁醇钠或叔丁醇钾,所述的无机碱类包括但不限于氢化钠、氢氧化钠、氢氧化钾、氢氧化锂、磷酸钾、碳酸钠、碳酸氢钠、碳酸钾或碳酸铯。
缩合剂包括但不限于1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐、N,N'-二环己基碳化二亚胺、N,N'-二异丙基碳二酰亚胺、O-苯并三氮唑-N,N,N',N'-四甲基脲四氟硼酸酯、1-羟基苯并三唑、1-羟基-7-偶氮苯并三氮唑、O-苯并三氮唑-N,N,N',N'-四甲脲六氟磷酸酯、2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯、2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯、苯并三氮唑-1-基氧基三(二甲基氨基)磷鎓六氟磷酸盐或六氟磷酸苯并三唑-1-基-氧基三吡咯烷基磷,优选为1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐和1-羟基苯并三唑。
方案二
本发明通式(II)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用的盐的制备方法,包括以下步骤:
Figure PCTCN2018105008-appb-000034
第一步,通式(II-1)化合物和通式(I-2)化合物在碱性条件下,发生亲核取代反应,得到通式(II-2)化合物;
第二步,通式(II-2)化合物在碱性条件下水解,得到通式(II-3)化合物;
第三步,通式(II-3)化合物和通式(II-4)化合物或其可药用的盐,在碱性条件下,在缩合剂存在下发生缩合反应,得到通式(II)化合物,
其中:
X为卤素;
R a为烷基,优选为甲基或乙基;
G、R 1、R 3~R 9、n和t如通式(II)中所定义。
提供碱性条件的试剂包括有机碱和无机碱类,所述的有机碱类包括但不限于三乙胺、N,N-二异丙基乙胺、正丁基锂、二异丙基氨基锂、醋酸钾、醋酸钠、氨水、叔丁醇钠或叔丁醇钾,所述的无机碱类包括但不限于氢化钠、氢氧化钠、氢氧化钾、氢氧化锂、磷酸钾、碳酸钠、碳酸氢钠、碳酸钾或碳酸铯。
缩合剂包括但不限于1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐、N,N'-二环己基碳化二亚胺、N,N'-二异丙基碳二酰亚胺、O-苯并三氮唑-N,N,N',N'-四甲基脲四氟硼酸酯、1-羟基苯并三唑、1-羟基-7-偶氮苯并三氮唑、O-苯并三氮唑-N,N,N',N'-四甲脲六氟磷酸酯、2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯、2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯、苯并三氮唑-1-基氧基三(二甲基氨基)磷鎓六氟磷酸盐或六氟磷酸苯并三唑-1-基-氧基三吡咯烷基磷,优选为1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐和1-羟基苯并三唑。
方案三
本发明通式(III)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用的盐的制备方法,包括以下步骤:
Figure PCTCN2018105008-appb-000035
第一步,通式(II-1)化合物和通式(III-1)化合物在碱性条件下,发生亲核取代反应,得到通式(III-2)化合物;
第二步,通式(III-2)化合物在碱性条件下水解,得到通式(III-3)化合物;
第三步,通式(III-3)化合物和通式(III-4)化合物或其可药用的盐,在碱性条件下,在缩合剂存在下发生缩合反应,得到通式(III)化合物,
其中:
X为卤素;
R a为烷基,优选为甲基或乙基;
R 1、R 5、R 6、R 11、R 14、L 1、n和t如通式(III)中所定义。
提供碱性条件的试剂包括有机碱和无机碱类,所述的有机碱类包括但不限于三乙胺、N,N-二异丙基乙胺、正丁基锂、二异丙基氨基锂、醋酸钾、醋酸钠、氨水、叔丁醇钠或叔丁醇钾,所述的无机碱类包括但不限于氢化钠、氢氧化钠、氢氧化钾、氢氧化锂、磷酸钾、碳酸钠、碳酸氢钠、碳酸钾或碳酸铯。
缩合剂包括但不限于1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐、N,N'-二环己基碳化二亚胺、N,N'-二异丙基碳二酰亚胺、O-苯并三氮唑-N,N,N',N'-四甲基脲四氟硼酸酯、1-羟基苯并三唑、1-羟基-7-偶氮苯并三氮唑、O-苯并三氮唑-N,N,N',N'-四甲脲六氟磷酸酯、2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯、2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯、苯并三氮唑-1-基氧基三(二甲基氨基)磷鎓六氟磷酸盐或六氟磷酸苯并三唑-1-基-氧基三吡咯烷基磷,优选为1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐和1-羟基苯并三唑。
方案四
本发明通式(IV)所示的化合物或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用的盐的制备方法,包括以下步骤:
Figure PCTCN2018105008-appb-000036
通式(III-3)化合物和通式(IV-1)化合物或其可药用盐,在碱性条件下,在缩合剂存在下发生缩合反应,得到通式(IV)化合物,
其中:
R 1、R 5、R 6、R 11、R 14、L 1、n和t如通式(IV)中所定义。
提供碱性条件的试剂包括有机碱和无机碱类,所述的有机碱类包括但不限于三乙胺、N,N-二异丙基乙胺、正丁基锂、二异丙基氨基锂、醋酸钾、醋酸钠、氨水、叔丁醇钠或叔丁醇钾,所述的无机碱类包括但不限于氢化钠、氢氧化钠、氢氧化钾、氢氧化锂、磷酸钾、碳酸钠、碳酸氢钠、碳酸钾或碳酸铯。
缩合剂包括但不限于1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐、N,N'-二环己基碳化二亚胺、N,N'-二异丙基碳二酰亚胺、O-苯并三氮唑-N,N,N',N'-四甲基脲四氟硼酸酯、1-羟基苯并三唑、1-羟基-7-偶氮苯并三氮唑、O-苯并三氮唑-N,N,N',N'-四甲脲六氟磷酸酯、2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯、2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯、苯并三氮唑-1-基氧基三(二甲基氨基)磷鎓六氟磷酸盐或六氟磷酸苯并三唑-1-基-氧基三吡咯烷基磷,优选为1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐和1-羟基苯并三唑。
附图说明
图1:实施例1化合物单独使用或者与抗鼠-PD-1抗体联合用药在C57BL/6小鼠中对MC38结肠直肠肿瘤生长的影响。
具体实施方式
以下结合实施例进一步描述本发明,但这些实施例并非限制着本发明的范围。
实施例
化合物的结构是通过核磁共振(NMR)或/和质谱(MS)来确定的。NMR位移(δ)以10 -6(ppm)的单位给出。NMR的测定是用Bruker AVANCE-400核磁仪,测定溶剂为氘代二甲基亚砜(DMSO-d 6),氘代氯仿(CDCl 3),氘代甲醇(CD 3OD),内标为四甲基硅烷(TMS)。
MS的测定用SHIMAZU液相色谱-质谱仪(生产商:Shimazu,型号:LC-20AD,LCMS-2020)。
HPLC使用岛津SPD-20A高压液相色谱仪(Phenomenex Gemini-NX 5uM C18 21.2×100mm色谱柱)、岛津LC-20AD高压液相色谱仪(Phenomenex Luna 3uM C18  50X2mm色谱柱)、Agilent HPLC 1200DAD、Agilent HPLC 1200VWD或Waters HPLC e2695-2489高压液相色谱仪。
薄层层析硅胶板使用Agela MF254硅胶板,薄层色谱法(TLC)使用的硅胶板采用的规格是0.15mm~0.2mm,薄层层析分离纯化产品采用的规格是0.4mm~0.5mm。
柱层析一般使用ISCO TELEDYNE或AGELA预制硅胶柱。
CombiFlash快速制备仪使用Combiflash Rf200(TELEDYNE ISCO)。
激酶平均抑制率及IC 50值的测定用NovoStar酶标仪(德国BMG公司)。
本发明的已知的起始原料可以采用或按照本领域已知的方法来合成,或可购买自ABCR GmbH&Co.KG,Acros Organics,Aldrich Chemical Company,韶远化学科技(Accela ChemBio Inc)、达瑞化学品、上海毕得医药科技有限公司等公司。
实施例中无特殊说明,反应能够均在氩气氛或氮气氛下进行。
氩气氛或氮气氛是指反应瓶连接一个约1L容积的氩气或氮气气球。
氢气氛是指反应瓶连接一个约1L容积的氢气气球。
加压氢化反应使用Parr 3916EKX型氢化仪和清蓝QL-500型氢气发生器或HC2-SS型氢化仪。
氢化反应通常抽真空,充入氢气,反复操作3次。
微波反应使用CEM Discover-S 908860型微波反应器。
实施例中无特殊说明,溶液是指水溶液。
实施例中无特殊说明,反应的温度为室温,为20℃~30℃。
实施例中的反应进程的监测采用薄层色谱法(TLC),反应所使用的展开剂,纯化化合物采用的柱层析的洗脱剂的体系和薄层色谱法的展开剂体系包括:A:二氯甲烷/甲醇体系,B:正己烷/乙酸乙酯体系,C:正己烷/乙酸乙酯/乙醇,D:石油醚/乙酸乙酯,溶剂的体积比根据化合物的极性不同而进行调节,也可以加入少量的三乙胺和醋酸等碱性或酸性试剂进行调节。
实施例1
2-[[4-氯-2-(三氟甲基)苯基]甲基]-1-(2,2-二氘代-2-氟-乙基)-N-[(1R)-1-(4-乙磺酰基苯基)-2-羟基-乙基]吲哚-5-甲酰胺1
Figure PCTCN2018105008-appb-000037
Figure PCTCN2018105008-appb-000038
第一步
(4-氯-2-(三氟甲基)苯基)甲醇1b
将4-氯-2-(三氟甲基)苯甲醛1a(10g,48mmol,采用专利申请“WO2011021492”公开的方法制备而得)溶于100mL乙醇中,分批加入硼氢化钠(1.83g,48mmol),搅拌反应2小时。反应液减压浓缩,所得残余物中加入水,用乙酸乙酯萃取,有机相用饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得标题化合物1b(9.5g,产率:95%)。
第二步
1-(溴甲基)-4-氯-2-(三氟甲基)苯1c
将化合物1b(9.5g,45.2mmol)溶于100mL二氯甲烷中,滴加入三溴化磷(24.5g,90.5mmol),搅拌反应2小时。反应液中加入水,分离有机相,水相用二氯甲烷萃取,合并有机相,依次用水、饱和碳酸氢钠溶液和饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得粗品标题化合物1c(10.5g),产品不经纯化直接进行下一步反应。
第三步
2-(4-氯-2-(三氟甲基)苄基)-1H-吲哚-5-甲酸甲酯1e
将1H-吲哚-5-甲酸甲酯1d(4.4g,25.8mmol,采用公知的方法“Huaxue Shiji,2015,37(7),585-589,594”制备而得)溶于40mL N,N-二甲基乙酰胺中,加入二(乙腈)二氯化钯(1.34g,5.16mmol)、双环[2.2.1]-2-庚烯(4.85g,51.6mmol)和碳酸氢钠(4.25g,51mmol),再加入粗品化合物1c(7.4g,27mmol),升温至70℃搅拌反应12小时。反应液冷却至室温,加入200mL水,用乙酸乙酯萃取三次,合并有机相,依次用水、饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得标题化合物1e(8.1g,产率:87%)。MS m/z(ESI):368.1[M+1]。
第四步
1-(2-(叔丁氧基)-2-氧代乙基)-2-(4-氯-2-(三氟甲基)苄基)-1H-吲哚-5-甲酸甲酯1g
将化合物1e(21.6mg,0.059mmol),2-溴乙酸叔丁酯1f(43.4μL,0.294mmol,采用公知的方法“Tetrahedron,2007,63(2),337-346”制备而得)和碳酸铯(95.7mg,0.294mmol)加入到1mL N,N-二甲基甲酰胺中,升温至110℃微波搅拌反应1.5小时。反应结束后得标题化合物1g的反应液,不作处理直接进行下一步反应。
第五步
2-(2-(4-氯-2-(三氟甲基)苄基)-5-(甲氧基羰基)-1H-吲哚-1-基)乙酸1h
向上述化合物1g的反应液中加入1mL甲醇,加入1mL 2M的氢氧化钾溶液,搅拌反应16小时。反应液用6M盐酸调节pH小于3,用乙酸乙酯萃取三次,合并有机相,用饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用高效液相色谱法(岛津SPD-20A高压液相色谱仪,Phenomenex Gemini-NX 5uM C18 21.2×100mm色谱柱,洗脱体系:三氟乙酸,水,乙腈)纯化所得残余物,得到标题化合物1h(10mg,产率:40%)。
第六步
2-[[4-氯-2-(三氟甲基)苯基]甲基]-1-(2,2-二氘代-2-羟基-乙基)吲哚-5-羧酸甲酯1i
将化合物1h(530mg,1.24mmol)溶于8mL四氢呋喃中,室温下加入1M三氘代硼烷四氢呋喃溶液(1.52mL,1.52mmol),室温搅拌反应30分钟,升温至75℃搅拌反应45分钟,补加0.6mL 1M三氘代硼烷四氢呋喃溶液,75℃搅拌反应45分钟。反应液加入5mL甲醇,75℃搅拌10分钟,减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得标题化合物1i(365mg,产率:70%)。
MS m/z(ESI):414[M+1]。
第七步
2-[[4-氯-2-(三氟甲基)苯基]甲基]-1-(2,2-二氘代-2-氟-乙基)吲哚-5-羧酸甲酯1j
将化合物1i(365mg,0.88mmol)溶于10mL二氯甲烷中,干冰丙酮浴冷却至-78℃,加入二乙胺基三氟化硫(0.23mL,1.76mmol)并搅拌反应1小时,于-78℃下补加二乙胺基三氟化硫(0.115mL,0.88mmol)继续搅拌反应1小时,升温至0℃(冰水浴)搅拌反应1小时。反应液中加入20mL二氯甲烷,用饱和碳酸氢钠溶液洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得标题化合物1j(158mg,产率:43%)。
MS m/z(ESI):416[M+1]。
第八步
2-[[4-氯-2-(三氟甲基)苯基]甲基]-1-(2,2-二氘代-2-氟-乙基)吲哚-5-羧酸1k
将化合物1j(158mg,0.38mmol)溶于16mL甲醇和四氢呋喃(V/V=1:1)的混合溶剂中,加入10mL 2M的氢氧化钾溶液,搅拌反应16小时,补加2mL 2M的氢氧化钾溶液,搅拌反应1小时,补加氢氧化钾(0.4g,7.13mmol),搅拌反应2小时,继续补加0.2g氢氧化钾固体,室温搅拌反应1小时。反应液中滴加6M盐 酸至pH小于2,加入50mL水,用乙酸乙酯萃取(30mL×3),合并有机相,用饱和氯化钠溶液洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,得粗品标题化合物1k(160mg),产品不经纯化直接进行下一步反应。
MS m/z(ESI):402[M+1]。
第九步
2-[[4-氯-2-(三氟甲基)苯基]甲基]-1-(2,2-二氘代-2-氟-乙基)-N-[(1R)-1-(4-乙磺酰基苯基)-2-羟基-乙基]吲哚-5-甲酰胺1
将粗品化合物1k(160mg,0.4mmol)和(R)-2-氨基-2-(4-(乙磺酰基)苯基)乙醇1l(211mg,0.8mmol,采用专利申请“US9481674”公开的方法制备而得)溶于50mL二氯甲烷中,加入1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐(153mg,0.8mmol)、1-羟基苯并三唑(108mg,0.8mmol)和三乙胺(280μL,2.0mmol),搅拌反应48小时。反应液中加入20mL二氯甲烷,用20mL水洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系C纯化所得残余物,得标题化合物1(180mg,产率:70%)。
MS m/z(ESI):613[M+1]。
1H NMR(500MHz,CDCl 3)δ8.08(s,1H),7.91-7.85(d,2H),7.77-7.64(m,2H),7.59(d,2H),7.42(dd,1H),7.32(d,1H),7.12-7.10(d,1H),7.09-7.07(d,1H),6.29(s,1H),5.33(dt,1H),5.30(s,1H),4.31(s,3H),4.25(s,1H),4.12-3.97(m,2H),3.09(q,2H),1.28(t,3H)。
实施例2
1-环丙基-2-[二氘代-[5-(三氟甲基)吡唑-1-基]甲基]-N-[(1R)-1-(4-乙基磺酰基苯基)-2-羟基-乙基]吲哚-5-甲酰胺2
Figure PCTCN2018105008-appb-000039
第一步
5-溴-1-环丙基-1H-吲哚-2-羧酸乙酯2b
将5-溴-1H-吲哚-2-羧酸乙酯2a(0.76g,2.84mmol,采用专利申请“WO2014060386”公开的方法制备而得)和环丙基硼酸(1.22g,14.2mmol,采用专 利申请“WO2008024843”公开的方法制备而得)溶解于8mL 1,2-二氯乙烷中,加入醋酸铜(1.13g,5.96mmol)、2,2’-联吡啶(0.98g,6.25mmol)和碳酸钠(0.66g,6.25mmol),80℃搅拌反应16小时。反应液过滤,二氯甲烷洗涤滤饼,合并滤洗液,减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物2b(920mg,产率:100%)。
MS m/z(ESI):308[M+1]。
第二步
(5-溴-1-环丙基-吲哚-2-基)-二氘代-甲醇2c
将氘代氢化锂铝(66mg,1.57mmol)悬浮于2mL四氢呋喃中,冷却至0℃,滴加1mL预制的化合物2b(0.25g,0.81mmol)的四氢呋喃溶液,反应体系在0℃搅拌反应2小时。反应液中加入少量甲醇和水(V:V=1:1)的混合溶剂,产生的悬浊液用硅藻土过滤,滤液用CombiFlash快速制备仪以洗脱剂体系B纯化所得残余物,得到标题化合物2c(86mg,产率:39.6%)。
MS m/z(ESI):268[M+1]。
第三步
5-溴-1-环丙基-2-[二氘代-[5-(三氟甲基)吡唑-1-基]甲基]吲哚2e
将化合物2c(80mg,0.3mmol)和5-(三氟甲基)-1H-吡唑2d(81mg,0.54mmol,采用公知的方法“Journal of Heterocyclic Chemistry,2010,47(2),301-308”制备而得)溶解于3mL四氢呋喃中,加入三苯基膦(142mg,0.54mmol)和二乙基偶氮二羧酸酯(94mg,0.54mmol),搅拌反应16小时。向反应液中加入30mL乙酸乙酯,依次用水(10mL)、饱和氯化钠溶液(10mL)洗涤,无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物2e(30mg,产率:25.97%)。
MS m/z(ESI):386[M+1]。
第四步
1-环丙基-2-[二氘代-[5-(三氟甲基)吡唑-1-基]甲基]吲哚-5-甲酸2f
将化合物2e(30mg,0.078mmol)、六羰基钼(40mg,0.15mmol)、反式二-(M)-双[2-(二邻甲苯基膦)苄基]乙酸二钯(II)(20mg,0.02mmol)、三叔丁基膦四氟硼酸盐(20mg,0.069mmol)和1,8-二氮杂双环[5.4.0]十一碳-7-烯(50μL,0.34μmol)加入水(50μL)和1,4-二氧六环(0.6mL)混合溶剂中,微波150℃搅拌反应15分钟。反应液用CombiFlash快速制备仪以洗脱剂体系A纯化,得到标题化合物2f(13mg,产率:47.5%)。
MS m/z(ESI):352[M+1]。
第五步
1-环丙基-2-[二氘代-[5-(三氟甲基)吡唑-1-基]甲基]-N-[(1R)-1-(4-乙基磺酰基苯基)-2-羟基-乙基]吲哚-5-甲酰胺2
将化合物2f(7mg,0.02mmol)溶于0.8mL N,N-二甲基甲酰胺中,加入化合物1l(7mg,0.03mmol)、N,N-二异丙基乙胺(20μL,0.12μmmol)和2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(15mg,0.095mmol),搅拌反应2小时。反应液用高效液相色谱法(岛津SPD-20A高压液相色谱仪,Phenomenex Gemini-NX5uM C18 21.2×100mm色谱柱,洗脱体系:三氟乙酸,水,乙腈)纯化,得到标题化合物2(3mg,产率:26.7%)。
MS m/z(ESI):563[M+1]。
1H NMR(500MHz,CDCl 3)δ7.95(d,1H),7.82(d,2H),7.64(dd,1H),7.61-7.49(m,4H),7.02(d,1H),6.64(d,1H),6.22(s,1H),5.28-5.23(m,1H),4.02(dd,1H),3.95(dd,1H),3.16(tt,1H),3.06-3.00(m,3H),1.26-1.12(m,5H),1.08-0.96(m,2H)。
实施例3
2-[[4-氯-2-(三氟甲基)苯基]甲基]-N-[二氘代-(4-乙磺酰基苯基)甲基]-1-(2-氟代乙基)吲哚-5-甲酰胺3
Figure PCTCN2018105008-appb-000040
第一步
二氘代-(4-乙硫基苯基)甲胺3b
将4-(乙硫基)苯甲腈3a(1.36g,8.34mmol,采用专利申请“采用专利申请“lo-(4-公开的方法制备而得)溶于50mL四氢呋喃中,分批加入氘代氢化锂铝(0.668g,17mmol),搅拌反应0.5小时。反应液中加入少量水淬灭反应,再加入50mL水,过滤,滤饼用50mL乙酸乙酯洗涤,将滤液分液,水相用乙酸乙酯萃取(20mL×2),合并有机相,用饱和氯化钠溶液洗涤(20mL),无水硫酸镁干燥,过滤,滤液减压浓缩,得粗品标题化合物3b(1.47g),产物不经纯化直接下一步反应。
第二步
N-[二氘代-(4-乙硫基苯基)甲基]氨基甲酸叔丁酯3c
将粗品化合物3b(1.47g,8.68mmol)溶于100mL二氯甲烷中,加入二碳酸二叔丁酯(2.84g,13.03mmol),搅拌反应0.5小时。反应液用水洗涤(20mL),无水硫酸钠干燥,过滤,得标题化合物3c的溶液,不经处理直接下一步反应。
第三步
N-[二氘代-(4-乙磺酰基苯基)甲基]氨基甲酸叔丁酯3d
向上述含化合物3c的溶液中加入间氯过氧苯甲酸(7.02g,43.4mmol),搅拌反应16小时。反应液依次用饱和硫代硫酸钠溶液(30mL×2),饱和碳酸氢钠溶液洗涤(30mL),无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得标题化合物3d(1.97g,产率:75.5%)
第四步
二氘代-(4-乙磺酰基苯基)甲胺盐酸盐3e
将化合物3d(1.97g,6.53mmol)溶于15mL二氯甲烷中,加入2M的氯化氢乙醚溶液(20mL,40mmol),搅拌反应2小时,补加2M的氯化氢乙醚溶液(15mL,30mmol),搅拌反应16小时。反应液过滤,收集滤饼,得粗品标题化合物3e(1.465g),产物不经纯化直接下一步反应。
第五步
2-(4-氯-2-(三氟甲基)苄基)-1-(2-氟代乙基)-1H-吲哚-5-甲酸甲酯3f
将化合物1e(0.3g,815.77μmol),1-溴-2-氟乙烷(310.7mg,2.45mmol)溶于10mL N,N-二甲基甲酰胺,加入碳酸铯(797.38mg,2.45mmol),微波100℃搅拌1小时。冷却反应液,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物3f(0.25g,产率:74.06%)。
MS m/z(ESI):414.1[M+1]。
第六步
2-(4-氯-2-(三氟甲基)苄基)-1-(2-氟代乙基)-1H-吲哚-5-甲酸3g
将化合物3f(0.25g,604.17μmol)溶于20mL甲醇中,加入1.5mL 4M的氢氧化钠溶液,回流搅拌反应1小时。反应液冷却至室温,滴加入1M盐酸调节pH为3~4,加入水和乙酸乙酯各20mL,再用乙酸乙酯萃取(20mL酸乙酯,合并有机相,无水硫酸钠干燥。过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物3g(0.24g,产率:99.4%)。
MS m/z(ESI):400.1[M+1]。
第七步
2-[[4-氯-2-(三氟甲基)苯基]甲基]-N-[二氘代-(4-乙磺酰基苯基)甲基]-1-(2-氟代乙基)吲哚-5-甲酰胺3
将化合物3g(10mg,25.01μmol)溶于1mL N,N-二甲基甲酰胺中,加入粗品化 合物3e(25mg,0.124mmol)和三乙胺(28μL,0.2mmol),再加入1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐(12mg,0.0625mmol)和1-羟基苯并三唑(8.5mg,0.0625mmol),搅拌反应16小时。反应液减压浓缩,用高效液相色谱法(岛津SPD-20A高压液相色谱仪,Phenomenex Gemini-NX 5uM C18 21.2×100mm色谱柱,洗脱体系:三氟乙酸,水,乙腈)纯化所得残余物,制得标题化合物3(2.6mg,17.8%)。MS m/z(ESI):583[M+1]。
实施例4
1-环丙基-N-[二氘代-(4-乙磺酰基苯基)甲基]-2-[[2-(三氟甲基)苯基]甲基]吲哚-5-甲酰胺4
Figure PCTCN2018105008-appb-000041
第一步
2-(2-三氟甲基)苄基)-1H-吲哚-5-甲酸甲酯4b
将化合物1d(400mg,2.29mmol)、1-(溴甲基)-2-(三氟甲基)苯4a(574mg,2.4mmol,采用专利申请“WO2015176640”公开的方法制备而得)、双(乙腈)二氯化钯(II)(118mg,0.46mmol)、双环[2.2.1]-2-庚烯(429mg,4.6mmol)和碳酸氢钠(384mg,4.6mmol)加入到10mL N,N-二甲基乙酰胺中,氩气氛下,加热至70℃,搅拌反应16小时。反应液冷却至室温,倒入水中,用乙酸乙酯萃取三次,合并有机相,依次用水、饱和氯化钠溶液洗涤,无水硫酸钠干燥。过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物4b(570mg,产率:74.9%)。
第二步
1-环丙基-2-(2-(三氟甲基)苄基)-1H-吲哚-5-甲酸甲酯4c
将化合物4b(64mg,192μmol)、环丙基硼酸(100mg,1150μmol)、2,2’-联吡啶(63mg,404μmol)、醋酸铜(73mg,404μmol)和碳酸钠(43mg,404μmol)加入到5mL四氢呋喃中,氩气氛下,60℃搅拌反应16小时。反应液过滤,滤液用乙酸乙酯萃取,无水硫酸钠干燥。过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂 体系B纯化所得残余物,得到标题化合物4c(70mg,产率:98%)。
MS m/z(ESI):374[M+1]。
第三步
1-环丙基-2-(2-(三氟甲基)苄基)-1H-吲哚-5-甲酸4d
将化合物4c(70mg,188μmol)和2M氢氧化钾溶液(1.5mL,3.0mmol)加入1.5mL甲醇中,搅拌反应16小时。反应液减压浓缩除去甲醇,所得残余物中滴加1M盐酸调节pH为3,用乙酸乙酯萃取,合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,得到粗品标题化合物4d(70mg),产物不经纯化直接用于下一步反应。
MS m/z(ESI):360[M+1]。
第四步
1-环丙基-N-[二氘代-(4-乙磺酰基苯基)甲基]-2-[[2-(三氟甲基)苯基]甲基]吲哚-5-甲酰胺4
采用实施例3第七步的合成步骤,将原料化合物3g替换为粗品化合物4d,制得标题化合物4(1.0mg)。
MS m/z(ESI):543[M+1]。
实施例5
2-[[4-氯-2-(三氟甲基)苯基]甲基]-1-(2,2-二氘代-2-羟基-乙基)-N-[(1R)-1-(4-乙磺酰基苯基)-2-羟基-乙基]吲哚-5-甲酰胺5
Figure PCTCN2018105008-appb-000042
第一步
2-[[4-氯-2-(三氟甲基)苯基]甲基]-1-(2,2-二氘代-2-羟基-乙基)吲哚-5-甲酸5a
采用实施例1第八步的合成步骤,将原料化合物1j替换为化合物1i,制得标题化合物5a(3.3mg)。
MS m/z(ESI):400[M+1]
第二步
2-[[4-氯-2-(三氟甲基)苯基]甲基]-1-(2,2-二氘代-2-羟基-乙基)-N-[(1R)-1-(4-乙磺酰基苯基)-2-羟基-乙基]吲哚-5-甲酰胺5
采用实施例1第九步的合成步骤,将原料化合物1k替换为化合物5a,制得标 题化合物5(2.0mg)。
MS m/z(ESI):611[M+1]。
1H NMR(500MHz,CDCl 3)δ8.06(d,1H),7.92-7.86(m,2H),7.71(d,1H),7.69(dd,1H),7.63-7.57(m,2H),7.41(dd,1H),7.39(d,1H),7.11(d,1H),7.09-7.05(m,1H),6.27-6.23(m,1H),5.34(dt,1H),4.36(s,2H),4.16(s,2H),4.13-3.99(m,2H),3.10(q,2H),1.29(t,3H)。
实施例6
2-[[4-氯-2-(三氟甲基)苯基]甲基]-1-(2-氟乙基)-N-[(1R)-2-羟基-1-[4-(1,1,2,2,2-五氘代乙基磺酰基)苯基]乙基]吲哚-5-甲酰胺6
Figure PCTCN2018105008-appb-000043
第一步
1-溴-4-(1,1,2,2,2-五氘代乙基硫基)苯6c
将4-溴苯硫酚6a(1g,5.29mmol,Adamas)、1,1,1,2,2-五氘代-2-碘-乙烷6b(1.1g,6.35mmol,Sigma)和碳酸铯(5.25g,15.87mmol)加入50mL N,N-二甲基甲酰胺中,搅拌16小时。反应液过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系D纯化所得残余物,得标题化合物6c(1.1g,产率93.62%)。
第二步
1-溴-4-(1,1,2,2,2-五氘代乙基磺酰基)苯6d
将化合物6c(1.17g,4.95mmol)溶于20mL甲醇中,加入5mL预制的过氧硫酸氢钾复合盐(6.1g,9.90mmol)溶液,搅拌反应2小时。反应液过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系D纯化所得残余物,得标题化合物6d(0.9g,产率71.52%)。
MS m/z(ESI):271[M+18]。
第三步
1-(1,1,2,2,2-五氘代乙基磺酰基)-4-乙烯基-苯6e
将化合物6d(0.8g,3.46mmol)、4,4,5,5-四甲基-2-乙烯基-1,3,2-二氧杂戊硼烷(1g,6.92mmol,韶远化学科技(上海)有限公司)溶于12mL 1,4-二氧六环和水(V:V=5:1)的混合溶剂中,加入四三苯基膦钯(400mg,346.24μmol)和碳酸铯(2.3g,6.92mmol),氩气氛下,反应体系于95℃搅拌18小时。反应液冷却至室温,用乙酸乙酯萃取(30mL×2),合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系D纯化所得残余物,得标题化合物6e(0.6g,产率86.09%)。
MS m/z(ESI):219[M+18]。
第四步
N-[2-羟基1-[4-(1,1,2,2,2-五氘代乙基磺酰基)苯基]乙基]氨基甲酸叔丁酯6f
将氢氧化钠(410mg,10.28mmol)溶于水(20mL)中,取其中5mL溶解二水锇酸钾(60mg,137.11μmol)备用,将15mL预制的氨基甲酸叔丁酯(98%,1.5g,12.00mmol)的正丙醇溶液加入上述氢氧化钠溶液,冰水浴冷却,滴加次氯酸叔丁酯(1.2g,10.28mmol),搅拌5分钟,加入5mL预制的氢化奎尼定1,4-(2,3-二氮杂萘)二醚(0.17g,205.67μmol)的正丙醇溶液,加入化合物6e(0.6g,3.43mmol)和上述二水锇酸钾溶液,反应体系于25℃搅拌1小时。反应液减压浓缩,加入20mL水,用乙酸乙酯萃取(30mL×2),合并有机相,用无水硫酸钠干燥,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系D纯化所得残余物,得标题化合物6f(R/S=14/1)(0.5g,产率43.62%)。
MS m/z(ESI):235[M+1-100],279[M+1-56]。
第五步
2-氨基-2-[4-(1,1,2,2,2-五氘代乙基磺酰基)苯基]乙醇盐酸盐6g
将化合物6f(0.5g,1.5mmol)溶于15mL甲醇,加入3mL浓盐酸,搅拌反应1小时。反应液减压浓缩,得粗品标题化合物6g(R/S=14/1)(350mg,产率:86.46%)。MS m/z(ESI):235[M+1]。
第六步
2-[[4-氯-2-(三氟甲基)苯基]甲基]-1-(2-氟乙基)-N-[(1R)-2-羟基-1-[4-(1,1,2,2,2-五氘代乙基磺酰基)苯基]乙基]吲哚-5-甲酰胺6
将化合物3g(0.15g,500.29μmol)和粗品化合物6g(150mg,650.38μmol)溶于5mL N,N-二甲基甲酰胺中,加入2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(0.4g,1mmol)和N,N-二异丙基乙胺(0.2g,1mmol),搅拌反应1小时。反应液减压浓缩,用高效液相色谱法纯化(Waters 2767-SQ高压液相色谱仪,sunfire OBD,150*19mm 5nfi色谱柱,洗脱体系:碳酸氢铵,水,乙腈)所得残余物,得粗品标题化合物6(70mg),将粗品进行手性制备(分离条件:手性制备柱Amylose-120*250mm,5um;流动相:乙醇/正己烷=40/60(v/v),流速:20mL/分钟),收集其相应组分,减压浓缩,得到标题化合物6(55.8mg,产率:18.10%)。
MS m/z(ESI):616[M+1]。
手性HPLC:保留时间13.798分钟,(色谱柱:Lux Amylose-1(AD)4.6*150mm 5um;流动相:乙醇(含0.1%二乙胺)/正己烷=40/60(v/v))。
1HNMR(400MHz,CDCl 3)δ8.11(s,1H),7.88-7.86(d,2H),7.75-7.72(m,2H),7.61-7.59(d,2H),7.47-7.44(d,1H),7.34-7.32(m,1H),7.29-7.27(m,1H),7.13-7.11(d,1H),6.30(s,1H),5.33-5.32(m,1H),4.69-4.66(m,1H),4.57-4.55(m,1H),4.34(s,3H),4.30-4.27(m,1H),4.08-4.05(dd,1H),4.01-3.97(dd,1H)。
实施例7
2-[[4-氯-2-(三氟甲基)苯基]甲基]-1-(2-氟乙基)-N-[(1R)-2-羟基-1-[4-(三氘代甲基磺酰基)苯基]乙基]吲哚-5-甲酰胺
Figure PCTCN2018105008-appb-000044
采用实施例6的合成步骤,将原料化合物6b替换为氘代碘甲烷,制得标题化合物7。
实施例8
2-[[4-氯-2-(三氟甲基)苯基]甲基]-N-[(1R)-1-(4-乙磺酰基苯基)-2-(三氘代甲氧基)乙基]-1-(2-氟乙基)吲哚-5-甲酰胺8
Figure PCTCN2018105008-appb-000045
第一步
2-(4-氯-2-(三氟甲基)苄基)-N-(1-(4-(乙磺酰基)苯基)-2-羟乙基)-1-(2-氟代乙基)-1H-吲哚-5-甲酰胺8b
将化合物3g(10mg,25.01μmol)溶于2mL N,N-二甲基甲酰胺中,加入化合物2-氨基-2-(4-乙磺酰基苯基)乙醇8a(8.67mg,37.83μmol)和N,N-二异丙基乙胺(6.47mg,50.03μmol),再加入2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(11.77mg,50.03μmol),室温搅拌反应2小时。减压浓缩反应液,用高效液相色谱法(Waters 2767-SQ高压液相色谱仪,sunfire OBD,150*19mm 5nfi色谱柱,洗脱体系:碳酸氢铵,水,乙腈)纯化所得残余物,制得标题化合物8b(7.9mg,51.7%)。MS m/z(ESI):611.5[M+1]。
第二步
(R)-2-(4-氯-2-(三氟甲基)苄基)-N-(1-(4-(乙磺酰基)苯基)-2-羟乙基)-1-(2-氟代乙基)-1H-吲哚-5-甲酰胺8c
将化合物8b(120mg,0.197mmol)进行手性制备(分离条件:Superchiral S-AD(Chiralway),2cm I.D.×25cm Length,5um;流动相:二氧化碳/乙醇/二乙胺=60/40/0.05(v/v/v),流速:50g/min),收集较长保留时间11.747分钟的组分,减压浓缩,得到标题化合物8c(52mg)。
MS m/z(ESI):611.0[M+1]。
第三步
2-[[4-氯-2-(三氟甲基)苯基]甲基]-N-[(1R)-1-(4-乙磺酰基苯基)-2-(三氘代甲氧基)乙基]-1-(2-氟乙基)吲哚-5-甲酰胺8
将化合物8c(10.5mg,0.017mmol),氧化银(11mg,0.047mmol),氘代碘甲烷(20uL,0.32mmol)和1mL乙腈混合,搅拌48小时补加氧化银(11mg,0.047mmol),氘代碘甲烷(20uL,0.32mmol),搅拌24小时。过滤,用高效液相色谱法(岛津SPD-20A高压液相色谱仪,Phenomenex Gemini-NX 5uM C18 21.2×100mm色谱柱,洗脱体系:三氟乙酸,水,乙腈)纯化所得残余物,制得标题化合物8(3mg)。
MS m/z(ESI):628[M+1]。
实施例9
2-[[4-氯-2-(三氟甲基)苯基]甲基]-N-[(1R)-1-(4-乙磺酰基苯基)-2-羟基乙基]-1-(1,1,2,2-四氘代-2-氟-乙基)吲哚-5-甲酰胺9
Figure PCTCN2018105008-appb-000046
第一步
2-[[4-氯-2-(三氟甲基)苯基]甲基]-1-(1,1,2,2-四氘代-2-羟基-乙基)吲哚-5-甲酸甲酯9b
将化合物1e(21.6mg,59μmol),溶于1mL N,N-二甲基甲酰胺,加入钠氢(23.5mg,0.59mmol),室温搅拌45分钟,加入化合物2-溴-1,1,2,2-四氘代-乙醇9a(17mg,172μmol)升温至50℃搅拌3小时。补加化合物9a(17mg,172μmol),50℃搅拌16小时,冷却反应液,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物9b(1mg,产率:4%)。
MS m/z(ESI):415.1[M+1]。
第二-四步
2-[[4-氯-2-(三氟甲基)苯基]甲基]-N-[(1R)-1-(4-乙磺酰基苯基)-2-羟基乙基]-1-(1,1,2,2-四氘代-2-氟-乙基)吲哚-5-甲酰胺9
采用实施例1第七-九步的合成步骤,将原料化合物1i替换为化合物9b,制得标题化合物9。
实施例10
2-[[4-氯-2-(三氟甲基)苯基]甲基]-N-[(1R)-1-(4-乙磺酰基苯基)-2-羟基乙基]-1-(1,1,2,2,2-五氘代乙基)吲哚-5-甲酰胺10
Figure PCTCN2018105008-appb-000047
第一步
2-[[4-氯-2-(三氟甲基)苯基]甲基]-1-(1,1,2,2,2-五氘代乙基)吲哚-5-甲酸甲酯10a
采用实施例3第五步的合成步骤,将原料化合物1-溴-2-氟乙烷替换为1,1,1,2,2-五氘代-2-碘-乙烷,制得标题化合物10a(8mg)。
MS m/z(ESI):401[M+1]。
第二步
2-[[4-氯-2-(三氟甲基)苯基]甲基]-N-[(1R)-1-(4-乙磺酰基苯基)-2-羟基乙基]-1-(1,1,2,2,2-五氘代乙基)吲哚-5-甲酰胺10
采用实施例1第八-九步的合成步骤,将原料化合物1j替换为化合物10a,1l替换为化合物10c(采用专利申请“WO2017024018”公开的方法制备而得),制得标题化合物10(1.5mg)。
MS m/z(ESI):598[M+1]。
实施例11
2-[[4-氯-2-(三氟甲基)苯基]甲基]-1-(1,1-二氘代乙基)-N-[(1R)-1-(4-乙磺酰基苯基)-2-羟基乙基]吲哚-5-甲酰胺11
Figure PCTCN2018105008-appb-000048
采用实施例10的合成步骤,将原料化合物1,1,1,2,2-五氘代-2-碘-乙烷替换为 化合物1,1,-二氘代-碘乙烷,制得标题化合物11(1.5mg)。
MS m/z(ESI):595[M+1]。
实施例12
2-[[4-氯-2-(三氟甲基)苯基]甲基]-N-[(1R)-1-(4-乙磺酰基苯基)-2-羟基乙基]-1-(1,1,2,2-四氘代-2-羟基-乙基)吲哚-5-甲酰胺12
Figure PCTCN2018105008-appb-000049
采用实施例1第九步的合成步骤,将原料化合物1k替换为化合物9b,制得标题化合物12(1.5mg)。
MS m/z(ESI):613[M+1]。
实施例13
2-[[4-氯-2-(三氟甲基)苯基]甲基]-1-(2,2-二氘代-2-氟-乙基)-N-[(1R)-1-(5-乙基磺酰基-2-吡啶)-2-羟基-乙基]吲哚-5-甲酰胺13
Figure PCTCN2018105008-appb-000050
采用实施例1第九步的合成步骤,将原料化合物1k替换为化合物(R)-2-氨基-2-(5-(乙基磺酰基)吡啶-2-基)乙醇(采用专利申请“US20160122318”公开的方法制备而得),制得标题化合物13。
以下实施例14-22
可以采用类似实施例6的合成步骤,从相应的原料合成。
实施例14
1-(2-氟乙基)-2-[[4-氟-2-(三氟甲基)苯基]甲基]-N-[(1R)-2-羟基-1-[4-(三氘代甲基磺酰基)苯基]乙基]吲哚-5-甲酰胺14
Figure PCTCN2018105008-appb-000051
实施例15
2-[[4-氯-2-(三氟甲基)苯基]甲基]-1-(2-氟乙基)-N-[(1R)-2-羟基-1-[5-(1,1,2,2,2-五氘代乙基磺酰基)-2-吡啶基]乙基]吲哚-5-甲酰胺15
Figure PCTCN2018105008-appb-000052
实施例16
1-(2-氟乙基)-2-[[4-氟-2-(三氟甲基)苯基]甲基]-N-[(1R)-2-羟基-1-[5-(1,1,2,2,2-五氘代乙基磺酰基)-2-吡啶基]乙基]吲哚-5-甲酰胺16
Figure PCTCN2018105008-appb-000053
实施例17
1-环丙基-N-[(1R)-2-羟基-1-[4-(1,1,2,2,2-五氘代乙基磺酰基)苯基]乙基]-2-[[2-(三氟甲基)-3-吡啶基]甲基]吲哚-5-甲酰胺17
Figure PCTCN2018105008-appb-000054
实施例18
2-[[4-氯-2-(三氟甲基)苯基]甲基]-1-(2-氟乙基)-N-[(1R)-2-羟基-1-[5-(三氘代甲基磺酰基)-2-吡啶基]乙基]吲哚-5-甲酰胺18
Figure PCTCN2018105008-appb-000055
实施例19
2-[4-氯-2-(三氟甲基)苯基]甲基]-1-环丙基-N-[(1R)-2-羟基-1-[4-(1,1,2,2,2-五氘代乙基磺酰基)苯基]乙基]吲哚-5-甲酰胺19
Figure PCTCN2018105008-appb-000056
实施例20
2-[[4-氯-2-(三氟甲基)苯基]甲基]-1-(2-羟基乙基)-N-[(1R)-2-羟基-1-[4-(1,1,2,2,2-五 氘代乙基磺酰基)苯基]乙基]吲哚-5-甲酰胺20
Figure PCTCN2018105008-appb-000057
实施例21
2-[[4-氯-2-(三氟甲基)苯基]-二氘代-甲基]-N-[(1R)-1-(4-乙基磺酰基苯基]-2-羟基-乙基]-1-(2-氟乙基)吲哚-5-甲酰胺21
Figure PCTCN2018105008-appb-000058
实施例22
2-[[4-氯-2-(三氟甲基)苯基]甲基]-N-[(1R)-2,2-二氘代-1-(4-乙磺酰基苯基)-2-羟基乙基]-1-(2-氟乙基)吲哚-5-甲酰胺22
Figure PCTCN2018105008-appb-000059
实施例23,24
1-(2-氟乙基)-2-[[4-氟-2-(三氟甲基)苯基]甲基]-N-[(1R)-2-羟基-1-[4-(1,1,2,2,2-五氘代乙基磺酰基)苯基]乙基]吲哚-5-甲酰胺23
1-(2-氟乙基)-2-[[4-氟-2-(三氟甲基)苯基]甲基]-N-[(1S)-2-羟基-1-[4-(1,1,2,2,2-五氘代乙基磺酰基)苯基]乙基]吲哚-5-甲酰胺24
Figure PCTCN2018105008-appb-000060
第一步
2-(4-氟-2-(三氟甲基)苄基)-1H-吲哚-5-甲酸甲酯23b
将化合物1d(1.3g,7.42mmol),1-(溴甲基)-4-氟-2-(三氟甲基)苯23a(2.29g,8.90mmol)溶于20mL N,N-二甲基乙酰胺中,加入双(乙腈)二氯化钯(II)(385.03mg,1.48mmol),双环[2.2.1]-2-庚烯(698.67mg,7.42mmol)和碳酸钠(1.57g,14.84mmol),氩气氛下,加热至80℃搅拌反应17小时。冷却反应液,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物23b(2.0g,产率:76.72%)。
MS m/z(ESI):352.1[M+1]。
第二步
2-(4-氟-2-(三氟甲基)苄基)-1-(2-氟代乙基)-1H-吲哚-5-甲酸甲酯23c
将化合物23b(0.14g,398.53μmol),1-溴-2-氟乙烷(151.8mg,1.20mmol)溶于10mL N,N-二甲基甲酰胺,加入碳酸铯(389.54mg,1.20mmol),微波条件下100℃反应1小时。冷却反应,过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系B纯化所得残余物,得到标题化合物23c(0.135g,产率:85.3%)。
MS m/z(ESI):398.0[M+1]。
第三步
2-(4-氟-2-(三氟甲基)苄基)-1-(2-氟代乙基)-1H-吲哚-5-甲酸23d
将化合物23c(0.13g,339.76μmol)溶于15mL甲醇中,加入1.5mL 4N的氢氧化钠溶液,回流搅拌反应1小时。反应液冷却至室温,滴加入1M盐酸调节pH为3~4,加入水和乙酸乙酯各20mL,乙酸乙酯萃取(20mL×2),合并有机相,无水硫酸钠干燥。过滤,滤液减压浓缩,用硅胶柱色谱法以洗脱剂体系A纯化所得残余物,得到标题化合物23d(0.125g,产率:96.0%)。
MS m/z(ESI):384.0[M+1]。
第四步
1-(2-氟乙基)-2-[[4-氟-2-(三氟甲基)苯基]甲基]-N-[(1R)-2-羟基-1-[4-(1,1,2,2,2-五氘代乙基磺酰基)苯基]乙基]吲哚-5-甲酰胺23
1-(2-氟乙基)-2-[[4-氟-2-(三氟甲基)苯基]甲基]-N-[(1S)-2-羟基-1-[4-(1,1,2,2,2-五氘代乙基磺酰基)苯基]乙基]吲哚-5-甲酰胺24
将化合物23d(250mg,652.21μmol)溶于10mL N,N-二甲基甲酰胺中,加入化合物6g(211.93mg,782.65μmol)和N,N-二异丙基乙胺(168.58mg,1.30mmol),再加入2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(371.76mg,978.32μmol),室温搅拌反应2小时。减压浓缩反应液,用高效液相色谱法纯化(Waters2767-SQ高压液相色谱仪,sunfire OBD,150*19mm 5nfi色谱柱,洗脱体系:碳酸氢铵,水,乙腈)所得残余物,得粗品化合物140mg,将粗品进行手性制备(分离条件:手性制备柱Amylose-1 20*250mm,5um;流动相:乙醇/正己烷=40/60(v/v),流速:20mL/分钟),收集其相应组分,减压浓缩,得到标题化合物23(100mg,产率:25.57%)和24(10mg,产率:2%)。
化合物23:
MS m/z(ESI):600.0[M+1]。
手性HPLC分析:保留时间8.384分钟,手性纯度:100%(色谱柱:Phenomenex Lux Cellulose-1(OD)150*4.6mm,5um(带保护柱);流动相:正己烷/乙醇(0.1%二乙胺)=60/40(v/v))。
1HNMR(400MHz,CDCl 3)δ8.12(s,1H),7.94-7.92(d,2H),7.77-7.74(d,1H),7.65-7.63(d,2H),7.50-7.47(d,1H),7.38-7.36(d,1H),7.29-7.27(m,1H),7.13-7.11(m,2H),6.32(s,1H),5.41-5.37(m,1H),4.70-4.69(m,1H),4.58-4.56(m,1H),4.39-4.37(m,1H),4.35(s,2H),4.32-4.30(m,1H),4.08-4.05(dd,1H),4.01-3.97(dd,1H)。
化合物24:
MS m/z(ESI):600.0[M+1]。
手性HPLC分析:保留时间9.978分钟,手性纯度:100%(色谱柱:Phenomenex Lux Cellulose-1(OD)150*4.6mm,5um(带保护柱);流动相:正己烷/乙醇(0.1%二乙胺)=60/40(v/v))。
1HNMR(400MHz,CDCl 3)δ8.13(s,1H),7.93-7.91(d,2H),7.77-7.74(d,1H),7.65-7.63(d,2H),7.50-7.47(d,1H),7.38-7.36(d,1H),7.29-7.27(m,1H),7.13-7.11(m, 2H),6.32(s,1H),5.41-5.37(m,1H),4.70-4.69(m,1H),4.58-4.56(m,1H),4.39-4.37(m,1H),4.35(s,2H),4.32-4.30(m,1H),4.10-4.07(dd,1H),4.01-3.97(dd,1H)。
生物学评价
以下结合测试例进一步描述解释本发明,但这些实施例并非意味着限制本发明的范围。
测试例1、本发明化合物对RORγ体外活性的测定
一、实验材料及仪器
1.
Figure PCTCN2018105008-appb-000061
TR-FRET RORγ共激活体系(Life Technologies)
2.RORγLBD(AB Vector)
3.DMSO(SigmaAldrich)
4.酶标仪(Tecan)
二、实验步骤
采用LanthaScreen TR-FRET(时间分辨荧光能量共振转移)RORγ共激活体系筛选本发明的化合物对RORγ活性的调节。
首先配制完整缓冲液D(complete TR-FRET Coregulator)(Life Technologies)包含终浓度5mM DTT。DMSO终浓度为2%。将待测化合物在含有2%DMSO的完整缓冲液D中连续稀释为2x终浓度,最高剂量为的60μm。10μl/孔加入384孔板的试验孔(PerkinElmer)。每个检测化合物在相同浓度下设置2个平行对照孔。准备4X RORγLBD(AB Vector)。使用完整缓冲液D稀释RORγLBD浓度为1ng/μL。5μl/孔加入384孔测定板的试验孔。阴性对照孔为5μL完整缓冲液D,无RORγLBD。使用完全缓冲液D配制含有0.6μM荧光素-D22(4X)和8nM铽(Tb)标记的抗GST抗体(4X)(Life Technologies)混合液,将5μL混合液加入到384孔板中。总反应体系为20μL。在振荡器上轻轻混匀该384孔板并在室温下避光孵育2-4小时。
使用Tecan Infinite M1000检测荧光读数,通过GraphPad Prism 6.0软件绘制发射波长520nm/495nm的比值与化合物浓度的对数曲线,计算待测化合物的EC 50值。
本发明化合物对RORγ体外活性通过以上的试验进行测定,测得的EC 50值见表1。
表1 本发明化合物对RORγ体外活性的EC 50
实施例编号 EC 50(nM) Emax(%)
1 9 77%
2 16 89%
3 17 98%
4 5 98%
5 9 73%
6 33 102%
8 34 85%
11 21 84%
13 9 77%
结论:本发明化合物对RORγ体外活性具有明显的激动作用。
测试例2、本发明化合物对IL-17A酶联免疫定量分析活性测定
一、实验材料及仪器
1.人外周血单核细胞(PBMC)(Zenbio)
2.淋巴细胞培养基(Zenbio)
3.TexMACS(Miltenyi Biotec)
4.人Cytostim(Miltenyi Biotec)
5.人IL-17酶联免疫试剂盒(R&D Systems)
6.CO 2培养箱(Fisher Scientific)
7.离心机(Fisher Scientific)
8..酶标仪(Tecan)
二、实验步骤
将冻存的人外周血单核细胞(PBMC)在预热的淋巴细胞培养基中快速复苏,离心1000rpm,10min,除去细胞培养上清,将细胞轻轻悬浮于TexMACS培养基中,计数细胞。在细胞悬液中按比例加入T细胞激活试剂cytostim(10μl/ml),然后以1×10 5外周血单核细胞/孔的密度将细胞种植于96孔细胞培养板中。使用TexMACS培养基梯度稀释待测化合物,分别加入各实验孔中,每组2-3个平行孔。准备只含细胞不含cytostim的阴性对照孔,以得到背景读数。将细胞培养板放置于5%二氧化碳37℃培养箱孵育3天。药物处理3天后收取细胞培养上清液,离心去除悬浮物。然后使用IL-17A酶联免疫试剂盒定量上清液中IL-17A。使用GraphPad Prism 6.0计算待测化合物的EC 50值。
本发明化合物对IL-17A酶联免疫定量分析通过以上的试验进行测定,测得的EC 50值见表2。
表2 本发明化合物对IL-17A酶联免疫定量分析的EC 50
实施例编号 EC 50(nM) Emax(%)
1 22 88%
3 103 102%
4 20 79%
5 32 82%
6 37 100%
13 22 88%
结论:本发明化合物对IL-17A酶联免疫定量分析活性具有明显的调节作用。
药代动力学评价
测试例3、本发明化合物的小鼠药代动力学测试
1、摘要
以小鼠为受试动物,应用LC/MS/MS法测定了小鼠灌胃给予实施例1化合物和实施例6后不同时刻血浆中的药物浓度。研究本发明化合物在小鼠体内的药代动力学行为,评价其药动学特征。
2、试验方案
2.1试验药品
实施例1化合物和实施例6。
2.2试验动物
C57小鼠18只,雌性,平均分为2组,购自上海杰思捷实验动物有限公司,动物生产许可证号:SCXK(沪)2013-0006。
2.3药物配制
称取一定量药物,加5%体积的DMSO、5%体积的吐温80和90%生理盐水配置成0.1mg/ml无色澄清透明液体。
2.4给药
C57小鼠禁食过夜后灌胃给药,给药剂量均为2.0mg/kg,给药体积均为0.2ml/10g。
3、操作
小鼠灌胃给药实施例1化合物和实施例6化合物,于给药前及给药后0.25,0.5,1.0,2.0,4.0,6.0,8.0,11.0,24.0小时采血0.1ml(每个时间点3只动物),置于肝素化试管中,3500转/分钟离心10分钟分离血浆,于-20℃保存。
测定不同浓度的药物灌胃给药后小鼠血浆中的待测化合物含量:取给药后各时刻的小鼠血浆25μl,加入内标溶液喜树碱80μl(100ng/mL),乙腈200μl,涡旋混合5分钟,离心10分钟(3600转/分钟),血浆样品取上清液1μl进行LC/MS/MS分析。
4、药代动力学参数结果
本发明化合物的药代动力学参数如下:
Figure PCTCN2018105008-appb-000062
Figure PCTCN2018105008-appb-000063
结论:本发明化合物的药代吸收较好,具有药代动力学优势。
药效学评价
测试例4、RORγ激动剂在同种型MC38结肠直肠肿瘤小鼠模型的药效
1.实验目的
用MC38小鼠模型来评估实施例1化合物对MC38结肠直肠肿瘤生长的抑制作用。
2.实验方法和实验材料
2.1.实验动物和饲养条件
实验用雌性C57BL/6小鼠,购自Charles River Lab(美国),购入时20-25克,7-9周龄。10只/笼饲养,温度23±1℃恒温,湿度50~60%,自由进食进水。一切按照实验动物护理和使用委员会(IACUC批准指南)进行护理和使用。动物购进后,进行7天适应性饲养后开始实验。
2.2.实验药品
实施例1化合物
抗鼠PD-1(CD279)抗体购自BioXcell(克隆RMP1-14;目录编号BP0146)
IgG2a同型对照抗体购自BioXcell(克隆2A3;目录编号BE0089)
2.3.实验设计和实验方法
2.3.1.动物分组:
小鼠适应性饲养后,分组如下:
Figure PCTCN2018105008-appb-000064
注:1.Q3dx4代表每隔三天给药,总共给四次;
2.QDx20代表每天给药1次,连续给药20天;
3.IgG2a和PD-1抗体是腹腔注射给药,实施例1化合物和载体是口服给药。
2.3.2.实验方法:
实验使用雌性C57BL/6小鼠(20-25克,7-9周龄)。通过检测同种型MC38结肠直肠肿瘤在自交系C57BL/6小鼠的生长情况评估单独使用实施例1化合物或实施例1化合物与抗鼠-PD-1抗体联合用药的体内抗肿瘤活性。将五十万(5×10 5) MC38细胞植入每只小鼠的右侧腹部皮下,当肿瘤生长至40-80mm 3后(第6天),将小鼠随机分为上述四组,第III组和第IV组每天给药(实施例1化合物,0.5mg/kg)一次,连续给药20天。在第II组和第IV组治疗实验中,在第6,9,12,15天分别对携带MC38肿瘤的小鼠腹腔(i.p.)注射抗鼠PD-1(CD279)抗体(BioXcell)(5mg/kg)。第I组(对照组)是载体CMC-Na药剂配方与IgG2a同型对照抗体,载体CMC-Na药剂配方的用药方式与实施例1化合物相同,IgG2a同型对照抗体的用药方式与抗鼠PD-1(CD279)抗体相同。
2.4.数据表达:
用卡尺在三个维度中测量肿瘤体积,然后根据下式计算:肿瘤体积(mm 3)=l×w×h×0.5236,其中,1表示肿瘤长度,w表示肿瘤的宽度,h表示肿瘤的高度,单位为毫米。肿瘤生长抑制率TGI%=100x(TV 对照-TV 肿瘤)/(TV 对照-TV 初始),其中TV 对照=对照组的肿瘤体积;TV 肿瘤=治疗组的肿瘤体积;TV 初始=6天时起始肿瘤体积。
3.结果和讨论:
如图1所示,单独给药0.5mg/kg实施例1化合物时,TGI为22.5%。单独注射抗鼠PD-1(CD279)抗体(5mg/kg)时,TGI为39.8%。当与抗鼠PD-1单克隆抗体(5mg/kg)联合用药时,实施例1化合物表现出协同效应(给药0.5mg/kg时,TGI为65.5%)。这些数据表明了在同基因的MC38结肠直肠肿瘤模型中,单独施用实施例1化合物表现出抑瘤活性,同时实施例1化合物与PD-1抗体联合用药表现出协同作用,这也表明实施例1化合物具有与RORγ激活(而非抑制)一致的生物活性,为提高免疫治疗的疗效开辟了新途径。

Claims (22)

  1. 一种通式(I)所示的化合物:
    Figure PCTCN2018105008-appb-100001
    或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用的盐,
    其中:
    Figure PCTCN2018105008-appb-100002
    为双键或单键;
    G 1、G 2和G 3相同或不同,且各自独立地选自C、CH、CH 2和N;
    环A选自芳基、杂芳基、环烷基和杂环基;
    环B为芳基或杂芳基;
    R 1相同或不同,且各自独立地选自H原子、D原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、氰基、氨基、硝基、羟基和羟烷基;
    R 2为卤代烷基;
    R 3和R 4相同或不同,且各自独立地选自H原子、D原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、羟烷基、氰基、氨基、硝基、羟基、环烷基、杂环基、芳基和杂芳基,其中所述的烷基、羟烷基、环烷基、杂环基、芳基和杂芳基各自独立地任选被选自D原子、羟基、卤素、烷基、氨基和-OR 11中的一个或多个取代基所取代;
    R 5选自H原子、烷基、卤代烷基、氨基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基,其中所述的烷基、环烷基、杂环基、芳基和杂芳基任选被选自D原子、卤素、羟基、环烷基和杂环基中的一个或多个取代基所取代;
    R 6相同或不同,且各自独立地选自H原子、D原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、氰基、氨基、硝基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基;
    R 7选自H原子、烷基、卤代烷基、环烷基、杂环基、芳基和杂芳基,其中所述的烷基、环烷基、杂环基、芳基和杂芳基任选被选自D原子、卤素、羟基、氨基、氰基、硝基、烷氧基、卤代烷氧基、-OR 11、-C(O)R 11、-C(O)OR 11、-NR 12R 13、-C(O)NR 12R 13、-S(O) mR 11、环烷基和杂环基中的一个或多个取代基所取代;
    R 8和R 9相同或不同,且各自独立地选自H原子、D原子、卤素、烷基、卤代烷基、烷氧基、氰基、氨基、硝基、羟基和羟烷基;
    R 10相同或不同,且各自独立地选自H原子、D原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、氰基、氨基、硝基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基;
    R 11选自H原子、D原子、烷基、卤代烷基、羟烷基、环烷基和杂环基,其中所述的烷基、羟烷基、环烷基和杂环基任选被选自D原子、卤素、羟基、环烷基和杂环基中的一个或多个取代基所取代;
    R 12和R 13相同或不同,且各自独立地选自H原子、D原子、烷基、卤代烷基、羟基和羟烷基,其中所述的烷基和羟烷基任选被选自D原子、卤素、羟基、环烷基和杂环基中的一个或多个取代基所取代;
    条件是通式(I)所示的化合物含有至少一个D原子;
    m为0、1或2;
    n为0、1、2、3或4;
    s为0、1、2或3;且
    t为0、1、2或3。
  2. 根据权利要求1所述的通式(I)所示的化合物,其中环A选自苯基、吡啶基、咪唑基、吡唑基、哌啶基和吗啉基;且环B为苯基或吡啶基。
  3. 根据权利要求1或2所述的通式(I)所示的化合物,其为通式(II)所示的化合物:
    Figure PCTCN2018105008-appb-100003
    其中:
    G为CH或N;
    R 1、R 3~R 9、n和t如权利要求1中所定义。
  4. 根据权利要求1~3中任一项所述的通式(I)所示的化合物,其中R 4为H原子或D原子;且R 3选自H原子、D原子和烷基,其中所述的烷基任选被选自D原子、羟基、卤素、氨基和-OR 11中的一个或多个取代基所取代;R 11如权利要求1中所定义。
  5. 根据权利要求1~4中任一项所述的通式(I)所示的化合物,其中R 7选自烷基、卤代烷基、环烷基和杂环基,其中所述的烷基任选被选自D原子、卤素、羟基、氨基、氰基、硝基、烷氧基、卤代烷氧基和羟烷基中的一个或多个取代基所取代。
  6. 根据权利要求1~5中任一项所述的通式(I)所示的化合物,其中R 8和R 9相同或不同,且各自独立地为H原子或D原子。
  7. 根据权利要求1~6中任一项所述的通式(I)所示的化合物,其为通式(III)所示的化合物:
    Figure PCTCN2018105008-appb-100004
    其中:
    L 1为亚烷基,其中所述的亚烷基任选被选自卤素和D原子中的一个或多个取代基所取代;
    R 14选自D原子、卤素、羟基、氨基、氰基、硝基、烷氧基、卤代烷氧基和羟烷基;
    R 1、R 5、R 6、R 11、n和t如权利要求1中所定义。
  8. 根据权利要求1~7中任一项所述的通式(I)所示的化合物,其为通式(IV)所示的化合物:
    Figure PCTCN2018105008-appb-100005
    其中:
    L 1为亚烷基,其中所述的亚烷基任选被选自卤素和D原子中的一个或多个取 代基所取代;
    R 14选自D原子、卤素、羟基、氨基、氰基、硝基、烷氧基、卤代烷氧基和羟烷基;
    R 1、R 5、R 6、R 11、n和t如权利要求1中所定义。
  9. 根据权利要求1~8中任一项所述的通式(I)所示的化合物,其中R 1相同或不同,且各自独立地选自H原子、D原子、卤素和烷基。
  10. 根据权利要求1~9中任一项所述的通式(I)所示的化合物,其中R 5为烷基,其中所述的烷基任选被选自D原子、卤素和羟基中的一个或多个取代基所取代。
  11. 根据权利要求1~10中任一项所述的通式(I)所示的化合物,其中R 6相同或不同,且各自独立地选自H原子、D原子和卤素。
  12. 根据权利要求1~11中任一项所述的通式(I)所示的化合物,其选自:
    Figure PCTCN2018105008-appb-100006
    Figure PCTCN2018105008-appb-100007
  13. 一种通式(V)所示化合物:
    Figure PCTCN2018105008-appb-100008
    或其互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体或其混合物形式,或其可药用盐,
    其中:
    Figure PCTCN2018105008-appb-100009
    为双键或单键;
    G 1、G 2和G 3相同或不同,且各自独立地选自C、CH、CH 2和N;
    环A选自芳基、杂芳基、环烷基和杂环基;
    R 1相同或不同,且各自独立地选自H原子、D原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、氰基、氨基、硝基、羟基和羟烷基;
    R 2为卤代烷基;
    R 6相同或不同,且各自独立地选自H原子、D原子、卤素、烷基、卤代烷基、烷氧基、卤代烷氧基、氰基、氨基、硝基、羟基、羟烷基、环烷基、杂环基、芳基和杂芳基;
    R 7选自H原子、烷基、卤代烷基、环烷基、杂环基、芳基和杂芳基,其中所述的烷基、环烷基、杂环基、芳基和杂芳基任选被选自D原子、卤素、羟基、氨基、氰基、硝基、烷氧基、卤代烷氧基、-OR 11、-C(O)R 11、-C(O)OR 11、-NR 12R 13、-C(O)NR 12R 13、-S(O) mR 11、环烷基和杂环基中的一个或多个取代基所取代;
    R 8和R 9相同或不同,且各自独立地选自H原子、D原子、卤素、烷基、卤代烷基、烷氧基、氰基、氨基、硝基、羟基和羟烷基;
    R 11选自H原子、D原子、烷基、卤代烷基、羟烷基、环烷基和杂环基,其中所述的烷基、羟烷基、环烷基和杂环基任选被选自D原子、卤素、羟基、环烷基和杂环基中的一个或多个取代基所取代;
    R 12和R 13相同或不同,且各自独立地选自H原子、D原子、烷基、卤代烷基、羟基和羟烷基,其中所述的烷基和羟烷基任选被选自D原子、卤素、羟基、环烷基和杂环基中的一个或多个取代基所取代;
    m为0、1或2;
    n为0、1、2、3或4;且
    t为0、1、2或3。
  14. 根据权利要求13所述的通式(V)所示的化合物,其中取代基R 7、R 8和R 9中至少一个取代基含有一个或多个D原子。
  15. 根据权利要求13所述的通式(V)所示的化合物,其选自:
    Figure PCTCN2018105008-appb-100010
  16. 一种制备根据权利要求1所述的通式(I)化合物的方法,该方法包括:
    Figure PCTCN2018105008-appb-100011
    通式(V)化合物与通式(VI)化合物或其可药用的盐发生缩合反应,得到通式(I)化合物;
    其中:
    Figure PCTCN2018105008-appb-100012
    环A、环B、G 1~G 3、R 1~R 10、n、s和t如权利要求1中所定义。
  17. 一种药物组合物,其含有治疗有效量的根据权利要求1~12中任一项所述的通式(I)所示的化合物,以及一种或多种药学上可接受的载体、稀释剂或赋形剂。
  18. 根据权利要求17所述的药物组合物,其进一步含有抗PD-1抗体。
  19. 根据权利要求1~12中任一项所述的通式(I)所示的化合物或根据权利要求17所述的药物组合物在制备ROR激动剂中的用途。
  20. 根据权利要求1~12中任一项所述的通式(I)所示的化合物或根据权利要求17所述的药物组合物在制备用于预防和/或治疗肿瘤或癌症的药物中的用途。
  21. 根据权利要求1~12中任一项所述的通式(I)所示的化合物或根据权利要求17所述的药物组合物与抗PD-1抗体的组合在制备用于预防和/或治疗肿瘤或癌症的药物中的用途。
  22. 根据权利要求20或21所述的用途,其中所述的肿瘤或癌症为实体瘤和血液瘤,优选选自非霍奇金淋巴瘤、弥漫大B细胞淋巴瘤、滤泡性淋巴瘤、滑膜肉瘤、乳腺癌、宫颈癌、结肠癌、肺癌、胃癌、直肠癌、胰腺癌、脑癌、皮肤癌、口腔癌、前列腺癌、骨癌、肾癌、卵巢癌、膀胱癌、肝癌、输卵管肿瘤、卵巢瘤、腹膜肿瘤、黑色素瘤、神经胶质瘤、神经胶母细胞瘤、肝细胞癌、乳突肾性瘤、头颈部肿瘤、白血病、淋巴瘤、骨髓瘤和非小细胞肺癌。
PCT/CN2018/105008 2017-09-12 2018-09-11 氘原子取代的吲哚甲酰胺类衍生物、其制备方法及其在医药上的应用 WO2019052440A1 (zh)

Priority Applications (7)

Application Number Priority Date Filing Date Title
AU2018331125A AU2018331125A1 (en) 2017-09-12 2018-09-11 Deuterium atom-substituted indole formamide derivative, preparation method therefor, and medical applications thereof
BR112020004611-6A BR112020004611A2 (pt) 2017-09-12 2018-09-11 derivado de indol formamida substituído por átomo de deutério, método de preparação do mesmo e aplicações médicas do mesmo
JP2020514271A JP2020533337A (ja) 2017-09-12 2018-09-11 重水素原子置換インドールホルムアミド誘導体、その調製方法、およびその医療用途
EP18856441.3A EP3683206A4 (en) 2017-09-12 2018-09-11 DEUTERIUM ATOM SUBSTITUTED INDOLFORMAMIDE DERIVATIVE, MANUFACTURING METHOD FOR ITS AND MEDICAL APPLICATIONS THEREOF
CN201880004431.1A CN109963836B (zh) 2017-09-12 2018-09-11 氘原子取代的吲哚甲酰胺类衍生物、其制备方法及其在医药上的应用
CA3075324A CA3075324A1 (en) 2017-09-12 2018-09-11 Deuterium atom-substituted indole formamide derivative, preparation method therefor, and medical applications thereof
US16/645,533 US20200277278A1 (en) 2017-09-12 2018-09-11 Deuterium Atom-Substituted Indole Formamide Derivative, Preparation Method Therefor, and Medical Applications Thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201710817140.1 2017-09-12
CN201710817140 2017-09-12

Publications (1)

Publication Number Publication Date
WO2019052440A1 true WO2019052440A1 (zh) 2019-03-21

Family

ID=65723209

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2018/105008 WO2019052440A1 (zh) 2017-09-12 2018-09-11 氘原子取代的吲哚甲酰胺类衍生物、其制备方法及其在医药上的应用

Country Status (9)

Country Link
US (1) US20200277278A1 (zh)
EP (1) EP3683206A4 (zh)
JP (1) JP2020533337A (zh)
CN (1) CN109963836B (zh)
AU (1) AU2018331125A1 (zh)
BR (1) BR112020004611A2 (zh)
CA (1) CA3075324A1 (zh)
TW (1) TW201912627A (zh)
WO (1) WO2019052440A1 (zh)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020140960A1 (zh) * 2019-01-04 2020-07-09 江苏恒瑞医药股份有限公司 吲哚甲酰胺类衍生物的晶型及其制备方法
WO2020182109A1 (zh) * 2019-03-11 2020-09-17 江苏恒瑞医药股份有限公司 氘原子取代的吲哚甲酰胺类衍生物的晶型及其制备方法
CN112745268A (zh) * 2019-10-31 2021-05-04 江苏恒瑞医药股份有限公司 苯并咪唑衍生物的晶型及制备方法

Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000022810A1 (de) 1998-10-09 2000-04-20 Deutsche Telekom Ag Verfahren zum generieren von digitalen wasserzeichen für elektronische dokumente
WO2005056516A1 (en) 2003-12-08 2005-06-23 Galderma Research & Development, S.N.C. Biphenyl derivatives useful as ligands that activate the rar receptors, process for preparing them and use thereof in human medicine and in cosmetics
WO2005056510A2 (en) 2003-12-08 2005-06-23 Galderma Research & Development, S.N.C. Novel ligands that are activators of the rar receptors, use in human medicine and in cosmetics
WO2005066116A1 (en) 2003-12-30 2005-07-21 Allergan, Inc. Disubstituted chalcone oximes as selective agonists of rarϝ retinoid receptors
WO2007068580A1 (fr) 2005-12-15 2007-06-21 Galderma Research & Development Derives biphenyliques agonistes selectifs du recepteur rar-gamma
WO2007068579A1 (fr) 2005-12-15 2007-06-21 Galderma Research & Development Derives biphenyliques agonistes selectifs du recepteur rar-gamma
WO2008024843A2 (en) 2006-08-25 2008-02-28 Viropharma Incorporated Combination therapy method for treating hepatitis c virus infection and pharmaceutical compositions for use therein
WO2008152260A2 (fr) 2007-05-11 2008-12-18 Galderma Research & Development Nouveaux ligands agonistes des recepteurs rars, utilisation en medecine humaine ainsi qu'en cosmetique
WO2011021492A1 (ja) 2009-08-18 2011-02-24 セントラル硝子株式会社 ホルミル基置換芳香族化合物の製造方法
WO2014060386A1 (en) 2012-10-18 2014-04-24 F. Hoffmann-La Roche Ag Indol-amide compounds as beta-amyloid inhbitors
WO2015171558A2 (en) 2014-05-05 2015-11-12 Lycera Corporation BENZENESULFONAMIDO AND RELATED COMPOUNDS FOR USE AS AGONISTS OF RORγ AND THE TREATEMENT OF DISEASE
WO2015176640A1 (en) 2014-05-22 2015-11-26 Merck Sharp & Dohme Corp. Antidiabetic tricyclic compounds
US20160122318A1 (en) 2014-11-05 2016-05-05 Vitae Pharmaceuticals, Inc. Isoindoline inhibitors of ror-gamma
US9481674B1 (en) 2016-06-10 2016-11-01 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma
WO2017024018A1 (en) 2015-08-05 2017-02-09 Vitae Pharmaceuticals, Inc. Modulators of ror-gamma
WO2017157332A1 (zh) * 2016-03-18 2017-09-21 江苏恒瑞医药股份有限公司 芳香酰胺类衍生物、其制备方法及其在医药上的应用

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW201906816A (zh) * 2017-07-06 2019-02-16 大陸商江蘇恆瑞醫藥股份有限公司 吲哚甲醯胺類衍生物、其製備方法及其在醫藥上的應用

Patent Citations (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2000022810A1 (de) 1998-10-09 2000-04-20 Deutsche Telekom Ag Verfahren zum generieren von digitalen wasserzeichen für elektronische dokumente
WO2005056516A1 (en) 2003-12-08 2005-06-23 Galderma Research & Development, S.N.C. Biphenyl derivatives useful as ligands that activate the rar receptors, process for preparing them and use thereof in human medicine and in cosmetics
WO2005056510A2 (en) 2003-12-08 2005-06-23 Galderma Research & Development, S.N.C. Novel ligands that are activators of the rar receptors, use in human medicine and in cosmetics
WO2005066116A1 (en) 2003-12-30 2005-07-21 Allergan, Inc. Disubstituted chalcone oximes as selective agonists of rarϝ retinoid receptors
WO2007068580A1 (fr) 2005-12-15 2007-06-21 Galderma Research & Development Derives biphenyliques agonistes selectifs du recepteur rar-gamma
WO2007068579A1 (fr) 2005-12-15 2007-06-21 Galderma Research & Development Derives biphenyliques agonistes selectifs du recepteur rar-gamma
WO2008024843A2 (en) 2006-08-25 2008-02-28 Viropharma Incorporated Combination therapy method for treating hepatitis c virus infection and pharmaceutical compositions for use therein
WO2008152260A2 (fr) 2007-05-11 2008-12-18 Galderma Research & Development Nouveaux ligands agonistes des recepteurs rars, utilisation en medecine humaine ainsi qu'en cosmetique
WO2011021492A1 (ja) 2009-08-18 2011-02-24 セントラル硝子株式会社 ホルミル基置換芳香族化合物の製造方法
WO2014060386A1 (en) 2012-10-18 2014-04-24 F. Hoffmann-La Roche Ag Indol-amide compounds as beta-amyloid inhbitors
WO2015171558A2 (en) 2014-05-05 2015-11-12 Lycera Corporation BENZENESULFONAMIDO AND RELATED COMPOUNDS FOR USE AS AGONISTS OF RORγ AND THE TREATEMENT OF DISEASE
WO2015176640A1 (en) 2014-05-22 2015-11-26 Merck Sharp & Dohme Corp. Antidiabetic tricyclic compounds
US20160122318A1 (en) 2014-11-05 2016-05-05 Vitae Pharmaceuticals, Inc. Isoindoline inhibitors of ror-gamma
WO2017024018A1 (en) 2015-08-05 2017-02-09 Vitae Pharmaceuticals, Inc. Modulators of ror-gamma
WO2017157332A1 (zh) * 2016-03-18 2017-09-21 江苏恒瑞医药股份有限公司 芳香酰胺类衍生物、其制备方法及其在医药上的应用
US9481674B1 (en) 2016-06-10 2016-11-01 Vitae Pharmaceuticals, Inc. Dihydropyrrolopyridine inhibitors of ROR-gamma

Non-Patent Citations (15)

* Cited by examiner, † Cited by third party
Title
ACS CHEM BIOL., vol. 11, no. 4, 15 April 2016 (2016-04-15), pages 1012 - 8
BLOOD, vol. 114, no. 6, 6 August 2009 (2009-08-06), pages 1141 - 9
CLIN CANCER RES., vol. 14, no. 11, 1 June 2008 (2008-06-01), pages 3254 - 61
CURR DRUG TARGETS INFLAMM ALLERGY, vol. 3, no. 4, December 2004 (2004-12-01), pages 395 - 412
EMBO J., vol. 17, no. 14, 15 July 1998 (1998-07-15), pages 3867 - 77
EUR J IMMUNOL., vol. 29, no. 12, December 1999 (1999-12-01), pages 4072 - 80
HUAXUE SHIJI, vol. 37, no. 7, 2015, pages 585 - 589,594
IMMUNITY, vol. 9, no. 6, December 1998 (1998-12-01), pages 797 - 806
J IMMUNOL., vol. 184, no. 8, 15 April 2010 (2010-04-15), pages 4215 - 27
J IMMUNOL., vol. 192, no. 6, 15 March 2014 (2014-03-15), pages 2564 - 75
JOURNAL OF HETEROCYCLIC CHEMISTRY, vol. 47, no. 2, 2010, pages 301 - 308
MECH DEV., vol. 70, no. 1-2, January 1998 (1998-01-01), pages 147 - 53
ONCOIMMUNOLOGY, vol. 5, no. 12, 4 November 2016 (2016-11-04), pages e1254854
See also references of EP3683206A4
TETRAHEDRON, vol. 63, no. 2, 2007, pages 337 - 346

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020140960A1 (zh) * 2019-01-04 2020-07-09 江苏恒瑞医药股份有限公司 吲哚甲酰胺类衍生物的晶型及其制备方法
CN113227048A (zh) * 2019-01-04 2021-08-06 江苏恒瑞医药股份有限公司 吲哚甲酰胺类衍生物的晶型及其制备方法
CN113227048B (zh) * 2019-01-04 2022-04-12 江苏恒瑞医药股份有限公司 吲哚甲酰胺类衍生物的晶型及其制备方法
WO2020182109A1 (zh) * 2019-03-11 2020-09-17 江苏恒瑞医药股份有限公司 氘原子取代的吲哚甲酰胺类衍生物的晶型及其制备方法
CN113365980A (zh) * 2019-03-11 2021-09-07 江苏恒瑞医药股份有限公司 氘原子取代的吲哚甲酰胺类衍生物的晶型及其制备方法
CN113365980B (zh) * 2019-03-11 2022-04-12 江苏恒瑞医药股份有限公司 氘原子取代的吲哚甲酰胺类衍生物的晶型及其制备方法
CN112745268A (zh) * 2019-10-31 2021-05-04 江苏恒瑞医药股份有限公司 苯并咪唑衍生物的晶型及制备方法
CN112745268B (zh) * 2019-10-31 2022-09-16 江苏恒瑞医药股份有限公司 苯并咪唑衍生物的晶型及制备方法

Also Published As

Publication number Publication date
CN109963836B (zh) 2022-11-22
TW201912627A (zh) 2019-04-01
EP3683206A4 (en) 2021-01-13
BR112020004611A2 (pt) 2020-09-24
CN109963836A (zh) 2019-07-02
JP2020533337A (ja) 2020-11-19
US20200277278A1 (en) 2020-09-03
EP3683206A1 (en) 2020-07-22
CA3075324A1 (en) 2019-03-21
AU2018331125A1 (en) 2020-03-05

Similar Documents

Publication Publication Date Title
RU2742770C1 (ru) Производное индола и формамида, способ его получения и применение в медицине
US10676438B2 (en) KCNQ2-5 channel activator
WO2016155545A1 (zh) 含氨磺酰基的1,2,5-噁二唑类衍生物、其制备方法及其在医药上的应用
WO2019052440A1 (zh) 氘原子取代的吲哚甲酰胺类衍生物、其制备方法及其在医药上的应用
WO2019158051A1 (zh) 一种作为吲哚胺-2,3-双加氧酶抑制剂的螺环化合物
JP2020193235A (ja) ヒドロキシトリアジン化合物及びその医薬用途
WO2019154294A1 (zh) 吡唑并[1,5-a][1,3,5]三嗪-2-胺类衍生物、其制备方法及其在医药上的应用
CN114149423A (zh) 四氢吡啶并嘧啶二酮类衍生物、其制备方法及其在医药上的应用
WO2022111498A1 (zh) 嘧啶二酮类衍生物、其制备方法及其在医药上的应用
FR2930249A1 (fr) Nouveaux derives de 3-aminoalkyl-1,3-dihydro-2h-indol-2-one, leur preparation et leur application en therapeutique.
CN109485595B (zh) 亲水性基团取代的吲哚甲酰胺类衍生物、其制备方法及其在医药上的应用
WO2018121551A1 (zh) 氮杂双环基取代的三唑类衍生物、其制备方法及其在医药上的应用
WO2020182109A1 (zh) 氘原子取代的吲哚甲酰胺类衍生物的晶型及其制备方法
JP2023551319A (ja) 5-ht7セロトニン受容体活性阻害用ビフェニルピロリジン及びビフェニルジヒドロイミダゾール誘導体並びにこれを有効成分として含む薬学組成物

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: 18856441

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2018331125

Country of ref document: AU

Date of ref document: 20180911

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 3075324

Country of ref document: CA

Ref document number: 2020514271

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112020004611

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 2018856441

Country of ref document: EP

Effective date: 20200414

ENP Entry into the national phase

Ref document number: 112020004611

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20200306