US20230192691A1 - Heterocyclic compounds as btk inhibitors - Google Patents

Heterocyclic compounds as btk inhibitors Download PDF

Info

Publication number
US20230192691A1
US20230192691A1 US18/169,423 US202318169423A US2023192691A1 US 20230192691 A1 US20230192691 A1 US 20230192691A1 US 202318169423 A US202318169423 A US 202318169423A US 2023192691 A1 US2023192691 A1 US 2023192691A1
Authority
US
United States
Prior art keywords
mmol
alkyl
amino
mixture
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US18/169,423
Other languages
English (en)
Inventor
Xiangyang Chen
Yucheng Pang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Beijing Innocare Pharma Tech Co Ltd
Original Assignee
Beijing Innocare Pharma Tech Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Beijing Innocare Pharma Tech Co Ltd filed Critical Beijing Innocare Pharma Tech Co Ltd
Publication of US20230192691A1 publication Critical patent/US20230192691A1/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • the invention relates to heterocyclic compounds or their pharmaceutically acceptable salts thereof, suitable for regulating or inhibiting activities of Bruton tyrosine kinase (BTK) and its C481 mutant.
  • BTK Bruton tyrosine kinase
  • the present invention also relates to methods for preparing the compounds or their pharmaceutically acceptable salts thereof.
  • the present invention further relates to the uses and methods of use of the compounds or their pharmaceutically acceptable salts thereof in the treatment and/or prevention of cancer and autoimmune diseases.
  • BTK is an important non-receptor tyrosine kinase that mediates cell signal transduction, which exists in plasma cells including B-cells.
  • B-cells are activated through B-cell receptor (BCR) and BTK plays an important role in the BCR-mediated signaling pathway.
  • BCR B-cell receptor
  • BTK plays an important role in the BCR-mediated signaling pathway.
  • PLC phospholipase C
  • PLC phospholipase C
  • This signaling pathway can promote cell proliferation, adhesion and survival, and plays an important role in the development of B-cell lymphoma.
  • BTK inhibitors inhibit the proliferation of B lymphoma cells by inhibiting the activity of BTK, destroy adhesion of tumor cells, and promote tumor cell apoptosis, making BTK a compelling drug target for B-cell related cancers, such as non-Hodgkin's lymphoma (NHL), chronic lymphocytic leukemia (CLL)/small lymphocytic lymphoma (SLL), mantle cell lymphoma (MCL), Waldenstrom's macroglobulinemia (WM), marginal zone Lymphoma (MZL), central nervous system leukemia (CNSL), etc.
  • NHL non-Hodgkin's lymphoma
  • CLL chronic lymphocytic leukemia
  • SLL small lymphocytic lymphoma
  • MCL mantle cell lymphoma
  • W Waldenstrom's macroglobulinemia
  • MZL marginal zone Lymphoma
  • CNSL central nervous system leukemia
  • BTK inhibitors are currently on the market, including Abbvie/JNJ's ibrutinib, AZ's acalabrutinib, Beigene's zanubrutinib and Gilead/Ono's tirabrutinib, and more BTK inhibitors are in clinical research.
  • BTK inhibitors can also inhibit the production of B-cell autoantibodies and cytokines.
  • B-cells present their own antigens, promote T-cell activation, secrete inflammatory factors that cause tissue damage, and at the same time activate B-cells to produce a large number of antibodies to trigger an autoimmune response.
  • T- and B-cells interact to each other to form a positive feedback regulatory chain which leads to uncontrolled autoimmune responses and aggravates tissue pathological damage.
  • IL-10 interleukin 10
  • TGF- ⁇ 1 transforming growth factor ⁇ 1
  • BTK can be a drug target for autoimmune diseases, such as rheumatoid arthritis (RA), multiple sclerosis (MS), systemic lupus erythematosus (SLE), pemphigus, etc.
  • RA rheumatoid arthritis
  • MS multiple sclerosis
  • SLE systemic lupus erythematosus
  • pemphigus etc.
  • BTK inhibitors are still in clinical research.
  • Sanofi's rilzabrutinib and Merck Serono's evobrutinib have achieved effective results in the treatment of pemphigus and multiple sclerosis, respectively.
  • BTK inhibitors on the market and under research are irreversible inhibitors which inhibit the activity of BTK by covalently binding to the cysteine residue located at 481 of the BTK protein.
  • BTK's C481 mutation such as C481S
  • C481S makes ibrutinib lose its covalent binding point with the protein, resulting in a decrease in the activity of ibrutinib, thereby making patients resistant to the ibrutinib treatment.
  • BTK inhibitors which effectively inhibit the activities of BTK and its C481 mutant, thereby overcoming the drug resistance caused by the C481 mutation associated with irreversible BTK inhibitors.
  • CSF/plasma ratio refers to the ratio of a compound concentration in cerebrospinal fluid (CSF) vs. in plasma.
  • BBB blood-brain barrier
  • C x-y refers to a range of the number of carbon atoms, where x and y are both integers, for example, C 3-8 cycloalkyl stands for cycloalkyl having 3 to 8 carbon atoms.
  • Alkyl refers to a saturated straight-chain or branched-chain hydrocarbyl substituent containing 1 to 20 carbon atoms, for example, 1 to 8 carbon atoms, 1 to 6 carbon atoms, or 1 to 4 carbon atoms.
  • alkyl examples include but are not limited to 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 and 2-ethylbutyl.
  • Cycloalkyl refers to a saturated cyclic hydrocarbyl substituent containing 3 to 14 annular carbon atoms. Cycloalkyl may be a mono carbon ring substituent, typically containing 3 to 8, 3 to 7, or 3 to 6 carbon atoms. Unrestricted examples of monocyclic cycloalkyl include but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl and cycloheptyl. Cycloalkyl may also be a substituent with two or three mono rings that are fused together, such as decahydronaphthyl, bicyclo[2.2.2]octane and spiro[3.3]heptane.
  • Heterocyclyl or heterocycle refers to a saturated or partially unsaturated monocyclic or polycyclic group containing 3 to 20 annular atoms, for example, 3 to 14, 3 to 12, 3 to 10, 3 to 8, 3 to 6, or 5 to 6 annular atoms in which one or more of the annular atoms are selected from N, O and S(O) m (where m is an integer from 0 to 2).
  • it may have 3 to 12 annular atoms, more preferably 3 to 10 ring atoms, more preferably 4 to 7 ring atoms, more preferably 4 to 6 ring atoms, most preferably 5 or 6 ring atoms, wherein 1 to 4 are heteroatoms, 1 to 3 are heteroatoms, or 1 to 2 are heteroatoms.
  • monocyclic heterocyclyl examples include but are not limited to pyrrolidinyl, oxetanyl, piperidyl, piperazinyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydrothiopyranyl, morpholinyl, thiomorpholinyl, homopiperazinyl and azetidinyl.
  • Polycyclic heterocyclyl includes fused, bridged or spiro polycyclic heterocycle, such as octahydrocyclopenta[c]pyrrole, octahydropyrrole[1,2-a]pyrazine, 3,8-diazabicyclo[3.2.1]octane, 5-azaspiro[2.4]heptane and 2-oxa-7-azaspiro[3.5]nonane.
  • Aryl or aryl ring refers to an aromatic monocyclic or fused polycyclic group containing 6 to 14 carbon atoms, preferably 6- to 10-membered, such as phenyl and naphthyl, most preferably phenyl.
  • the aryl ring may be fused with a heteroaryl, heterocyclic or cycloalkyl ring. Unrestricted examples include but are not limited to:
  • Heteroaryl or heteroaryl ring refers to a heteroaromatic system containing 5 to 14 annular atoms, of which 1 to 4 annular atoms are selected from heteroatoms including O, S and N.
  • Heteroaryl preferably is 5- to 10-membered, and more preferably 5- or 6-membered, such as furyl, thienyl, pyridyl, pyrrolyl, pyrimidyl, pyrazinyl, pyrazolyl, imidazolyl, tetrazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, quinolinyl, isoquinolinyl, indolyl and isoindolyl.
  • the heteroaryl ring may be fused with an aryl, heterocyclyl or cycloalkyl ring. Unrestricted examples include but are not limited to:
  • Halogen refers to F, Cl, Br, or I.
  • Cyano refers to —CN.
  • heterocyclic optionally substituted by an alkyl group includes the case where the heterocyclic is substituted by an alkyl group and the case where the heterocyclic is not substituted by an alkyl group.
  • substitution refers to one or more hydrogen atoms in a group, for example, 1 to 3 hydrogen atoms that are independently substituted by a corresponding number of substituents.
  • the substituents are located only in their possible chemical positions understood by those skilled in the art.
  • the substituents include but are not limited to halogen, hydroxyl, cyano, nitro, oxo, —SFS, C 1-4 alkyl, C 1-4 alkoxyl, etc.
  • “Isomers” refer to compounds that have the same molecular formula, but their atomic binding position or spatial arrangement is different. Isomers with different arrangement of their atoms in space are called “stereoisomers”. Stereoisomers include optical isomers, geometric isomers, and conformational isomers.
  • Optical isomers include enantiomers and diastereomers.
  • An enantiomer is one of two stereoisomers that are mirror images of each other and are non-superposable.
  • a racemic mixture or racemate is one that has equal amounts of left- and right-handed enantiomers of a chiral molecule.
  • Diastereomers are stereoisomers that are not mirror images of one another and are non-superimposable on one another.
  • Compounds of the present invention may also have geometric isomers resulting from the distribution of substituents around carbon-carbon double bonds, carbon-nitrogen double bonds, cycloalkyl or heterocyclyl groups.
  • the substituents around the carbon-carbon double bond or carbon-nitrogen bond are designated to be in a Z or E configuration, and the substituents around the cycloalkyl or heterocycle are designated to be in a cis or trans configuration.
  • Compounds of the present invention may also show tautomerism, such as keto-enol tautomerism.
  • the present invention includes any tautomeric or stereoisomeric forms and mixtures thereof and is not limited to any tautomeric or stereoisomeric forms used in the compound nomenclature or chemical structural formulae.
  • isotopes include all stable isotopes of the atoms appearing in the compounds of the present invention. Isotopes include those atoms with the same atomic number but in different masses. Examples of isotopes suitable for incorporation into the compounds of the present invention are isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine and chlorine, for example but not limited to 2 H (D), 3 H, 13 C, 14 C, 15 N, 18 O, 17 O, 31 P, 32 P, 35 S, 18 F and 36 Cl.
  • the isotopically labeled compounds of the present invention can generally be prepared by conventional techniques known to those skilled in the art or by methods similar to those described in the embodiments using appropriate isotopically labeled reagents instead of non-isotopically labeled reagents. Such compounds have various potential uses, for example, as standards and reagents in the determination of biological activities. In the case of stable isotopes, such compounds have the potential to beneficially alter biological, pharmacological, or pharmacokinetic properties.
  • Deuterium 2 H (D) is a preferable isotope of the present invention. For example, hydrogen in methyl, methylene or methine can be replaced by deuterium.
  • Prodrugs refer to derivatives that are converted into biologically active compounds under the physiological condition in vivo, for example, by oxidation, reduction, and hydrolysis (each of which occurs with or without the participation of enzymes).
  • Examples of a prodrug are a compound of the present invention in which an amino is acylated, alkylated or phoshorylated, for example eicosanoyl amino, alanyl amino and pivaloyloxymethyl amino; a hydroxyl is acylated, alkylated or phoshorylated or converted into borate, for example acetoxy, palmitoyloxy, pivaloyloxy, succinyloxy, fumaroyloxy and alanyloxy; a carbonyl is esterified or amidated; and a thiol forms a disulfide bridge with a carrier molecule that selectively delivers the drug to the target and/or to the cytosol of cells, such as peptide.
  • Prodrugs may be prepared from the compounds of the present invention according to well-known methods.
  • “Pharmaceutically acceptable salts” refer to the salts made from compounds of the present invention with pharmaceutically acceptable bases or acids, including inorganic alkalis or acids and organic bases or acids, under the condition that the compounds contain one or more acidic or basic groups.
  • Compounds of the present invention that contain acidic groups can exist in form of salts, for example, as alkali metal salts, alkaline earth metal salts, or ammonium salts.
  • such salts include sodium salts, potassium salts, calcium salts, magnesium salts or ammonia or organic amine salts such as salts of ethylamine, ethanolamine, triethanolamine or amino acids.
  • Compounds of the present invention that contain basic groups can exist in form of salts as inorganic or organic acid salts.
  • acids examples include hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, nitric acid, methanesulfonic acid, p-toluenesulfonic acid, naphthalene disulfonic acid, oxalic acid, acetic acid, tartaric acid, lactic acid, salicylic acid, benzoic acid, formic acid, propanoic acid, pivalic acid, malonic acid, succinic acid, pimelic acid, fumaric acid, maleic acid, malic acid, sulfamic acid, phenylpropionic acid, gluconic acid, ascorbic acid, isonicotinic acid, citric acid, adipic acid and other acids known to those skilled in the art.
  • the present invention further includes internal salts in addition to the mentioned salt forms.
  • Each salt can be obtained by conventional methods known to those skilled in the art, for example by mixing a compound of the present invention with an organic or inorganic acid or base in a solvent or dispersant, or by anion exchange or cation exchange with another salt.
  • “Pharmaceutical composition” refers to a composition containing one or more of compounds of the present invention or their pharmaceutically acceptable salts, stable isotope derivatives, isomers, prodrugs, and mixtures thereof, and other components such as pharmaceutically acceptable carrier and excipients.
  • “Cancer or lymphoma or leukemia” includes but is not limited to B-cell malignancies, B-cell lymphoma, diffuse large B-cell lymphoma, chronic lymphocytic leukemia, small lymphocytic lymphoma, non-Hodgkin Lymphoma (such as ABC-DLBCL), mantle cell lymphoma, follicular lymphoma, Waldenstrom's macroglobulinemia, marginal zone lymphoma, central nervous system lymphoma, chronic lymphocytic lymphoma, B-cell prelymphocytic leukemia, plasma cell lymphoma, multiple myeloma, various solid tumors (such as melanoma, bone cancer, brain cancer, colon cancer, liver cancer, skin cancer, kidney cancer, lung cancer, muscle cancer, bladder cancer, digestive tract/stomach Intestinal cancer, breast cancer, ovarian cancer, head and neck cancer, prostate cancer), etc.
  • B-cell malignancies B-cell lymphoma, diffuse large
  • “Autoimmune or inflammatory disease” includes but is not limited to arthritis, multiple sclerosis, osteoporosis, inflammatory bowel disease, colitis, Crohn's disease, lupus, rheumatoid Arthritis, psoriatic arthritis, lupus nephritis, Sjogren's syndrome, IgG4-related diseases, idiopathic thrombocytopenic purpura, immune thrombocytopenia, Wright's syndrome, psoriasis, Behcet's disease, asthma, Pemphigus, diabetes, myasthenia gravis, Guillain-Barre syndrome, Graves' disease, Hashimoto's thyroiditis, vasculitis, autoimmune vasculitis, granuloma with multiple vasculitis, autoimmune hepatitis, etc.
  • “Therapeutically effective amount” refers to an amount of compounds of the present invention that can effectively inhibit activities of BTK and its C481 mutant, and/or treat or prevent the diseases mediated by BTK and its C481 mutant.
  • “Patients” refer to mammals, preferably humans.
  • the present invention are directed to compounds with a structure as shown in Formula (I) as reversible BTK inhibitors, which effectively inhibit the activities of BTK and its C481 mutant, thereby overcoming the drug resistance caused by the C481 mutation associated with irreversible BTK inhibitors.
  • the present invention provides heterocyclic compounds as shown in Formula (I) or their pharmaceutically acceptable salts, stable isotope derivatives, isomers, and prodrugs thereof,
  • indicates that A is connected to the benzene ring and indicates that A is connected to E;
  • Ring K is phenyl or pyridyl, where the phenyl and pyridyl are optionally substituted by one or more substituents selected from halogen, cyano, C 1-6 alkyl or —OR a ;
  • E is C 1-6 alkyl, C 3-10 cycloalkyl, 3-10 membered heterocyclyl, C 6-10 aryl or 5-10 membered heteroaryl, where the alkyl, cycloalkyl, heterocyclyl, aryl and heteroaryl are optionally substituted by one or more substituents selected from D, halogen, cyano, —OR b , —NR b R c , —COOR b , —C(O)R b , —C(O)NR b R c or R e ;
  • R e is C 1-6 alkyl, C 3-10 cycloalkyl or 3-10 membered heterocyclyl, where one or more hydrogens of the alkyl, cycloalkyl and heterocyclyl are optionally substituted by halogen, cyano, —OR b , —NR b R c , —COOR b , —C(O)R b or —C(O)NR b R c ;
  • R 1 is H, halogen, —OR a or C 1-6 alkyl
  • R a is C 1-6 alkyl, where one or more hydrogens of the alkyl are optionally substituted by D or fluorine;
  • R b and R c are each independently selected from H, C 1-6 alkyl, C 3-6 cycloalkyl or 4-6 membered heterocyclyl.
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • E is C 1-6 alkyl, wherein one or more hydrogens of the alkyl are optionally substituted by D or fluorine, for example —CH(CH 3 )(CF 3 ).
  • E is C 3-7 monocyclic cycloalkyl or 4-8 membered monocyclic heterocyclyl containing N and/or O.
  • E is
  • E is C 5-10 polycyclic cycloalkyl or 5-10 membered polycyclic heterocyclyl containing O.
  • E is
  • the monocyclic cycloalkyl, polycyclic cycloalkyl, monocyclic heterocyclyl, and polycyclic heterocyclyl are independently and optionally substituted by one or more substituents selected from D, halogen, —OR b , —NR b R c , —COOR b , —C(O)R b , —C(O)NR b R c or C 1-6 alkyl, where one or more hydrogens of the alkyl are further optionally substituted by halogen, —OR b or —NR b R c ; R b and R c are each independently selected from H, C 1-6 alkyl, C 3-6 cycloalkyl or 4-6 containing N and/or O containing heterocyclic group (e.g., morpholinyl).
  • substituents selected from D, halogen, —OR b , —NR b R c , —COOR b , —C(O
  • E is
  • G is H, fluorine, —OR b , —NR b R c , —COOR b , —C(O)R b , —C(O)NR b R c or C 1-2 alkyl, where one or more hydrogens of the alkyl group are optionally substituted by fluorine, —OH or —NH 2 ;
  • R b and R c are each independently selected from H, C 1-2 alkyl, C 3-6 cycloalkyl or N and/or O containing 4-6 membered heterocyclyl (for example, morpholinyl).
  • ring K is phenyl, where the phenyl is optionally substituted by one or two substituents selected from halogen or —OR a ; R a is C 1-2 alkyl, where one or more hydrogens of the alkyl is optionally substituted by D.
  • ring K is
  • R a is a C 1-2 alkyl group, where one or more hydrogens of the alkyl are optionally substituted by D.
  • R 1 is H.
  • R 1 is halogen
  • the present invention relates to compounds that are have a substantial brain penetration and have a blood-brain barrier permeability.
  • the present invention relates to CNS penetrant compounds that have CSF/plasma concentration ratio Kp, CSF ⁇ 0.15.
  • the present invention further relates to the following Compounds 1-29, and 31-54, or their pharmaceutically acceptable salts, prodrugs, stable isotope derivatives, isomers and mixtures thereof.
  • N-(4-(4-amino-7-(1,1,1-trifluoropropan-2-yl)imidazo[5,1-f][1,2,4]triazin- 5-yl)benzyl)-5-fluoro-2-methoxybenzamide 27.
  • Compounds of the present invention effectively inhibit the activities of BTK and its C481 mutant, preferably having an IC 50 of less than 100 nM, and more preferably less than 10 nM.
  • the present invention also relates to pharmaceutical compositions comprising compounds of Formula (I) or their pharmaceutically acceptable salts, stable isotope derivatives, isomers and prodrugs thereof, and one or more pharmaceutically acceptable carriers or excipients.
  • the present invention further relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of Formula (I) or its pharmaceutically acceptable salt, stable isotope derivative, isomer, prodrug and a mixture thereof, and at least one additional therapeutic agent, wherein the agent may be a small molecule chemotherapeutic drug (such as anti-inflammatory steroid drug, kinase targeting drug, apoptosis inhibitor, inflammation modulator, cytotoxic drug, DNA damage related drug) or a macromolecular immune and/or inflammation modulator (such as CD-20 antibody, CD19 antibody, PD-1 antibody).
  • chemotherapeutic drug such as anti-inflammatory steroid drug, kinase targeting drug, apoptosis inhibitor, inflammation modulator, cytotoxic drug, DNA damage related drug
  • a macromolecular immune and/or inflammation modulator such as CD-20 antibody, CD19 antibody, PD-1 antibody.
  • Compounds of Formula (I) and another therapeutic agent may be present in the same pharmaceutical composition or in different pharmaceutical compositions.
  • Compounds of Formula (I) and another agent may be administered simultaneously or sequentially in the same or different forms.
  • the present invention provides a method for treating or preventing diseases mediated by BTK or its C481 mutant.
  • the method comprises administering to a patient in need a therapeutically effective amount of compounds of Formula (I) or their pharmaceutically acceptable salts, stable isotope derivatives, isomers, prodrugs and mixtures thereof, or pharmaceutical compositions containing compounds of Formula (I).
  • the diseases include but are not limited to cancer, lymphoma, leukemia, autoimmune or inflammation diseases, such as B-cell malignancies, B-cell lymphoma, diffuse large B-cell lymphoma, chronic lymphocytic leukemia, small lymphocytic lymphoma, non-Hodgkin's lymphoma (such as ABC-DLBCL), mantle cell lymphoma, follicular lymphoma, Waldenstrom's macroglobulinemia, marginal zone lymphoma, central nervous system lymphoma, chronic lymphocytic lymphoma, B-cell prelymphocytic leukemia, plasma cell lymphoma, multiple myeloma, various solid tumors (such as lung cancer, prostate cancer, head and neck cancer, breast cancer, ovarian cancer, uterine cancer, pancreatic cancer, colon cancer, rectal cancer, stomach cancer, esophageal cancer, brain cancer, liver cancer, kidney cancer, skin cancer, muscle cancer, epithelial cancer, bladder cancer
  • the pharmaceutical composition may be in any dosage form, including but not limited to tablets, capsules, solutions, freeze-dried preparations and injectable.
  • the pharmaceutical formulation of the present invention may be administered in form of a dosage unit containing a predetermined amount of active ingredient.
  • a dosage unit may contain 1 mg to 1 g, preferably 5 mg to 700 mg, particularly preferably 10 mg to 500 mg of a compound of the present invention, depending on the disease being treated, the method of administration, as well as age, weight, and condition of the patients.
  • the pharmaceutical formulation may be prepared using methods well-known in the pharmaceutical field, for example, by formulating the active ingredient with one or more excipients or one or more adjuvants.
  • the pharmaceutical formulation of the present invention is suitable for administration by any appropriate method, such as by oral (including oral or sublingual) or parenteral (including subcutaneous, intramuscular, intravenous, or intradermal).
  • the present invention also provides methods for preparing compounds.
  • the preparation of compounds of the present invention may be accomplished by the following exemplary methods and examples, but these methods and examples should not be considered as limitation of the scope of the present invention in any way.
  • Compounds of the present invention may also be synthesized by synthetic techniques known to those skilled in the art or by combinations of methods known in the art and of the present invention.
  • the products obtained at each step of reaction are isolated by separation techniques known in the art, including but not limited to extraction, filtration, distillation, crystallization, and chromatographic separation.
  • the starting materials and chemical reagents used for syntheses may be conventionally made based on literature (for example, SciFinder) or purchased.
  • Heterocyclic compounds shown in Formula (I) of the present invention can be synthesized according to the route shown below: 1) bromination of A1 with NBS to give A2; 2) substitution of A2 by a protected amine (for example, 2,4-dimethoxybenzylamine) to give A3; 3) Mitsunobu reaction between A3 and E-OH to provide A4; 4) Suzuki coupling of A4 with a phenylboronic acid to give A5; 5) deprotection of A5 to give A6.
  • a functional group of E can be further derivatized to afford various target compounds.
  • an ester in E can be hydrolyzed by alkali (such as LiOH) to form an acid and E containing protected amine or alcohol can be deprotected to give amine or alcohol, and the amine can be further amidated to afford amides, etc.
  • alkali such as LiOH
  • Heterocyclic compounds shown in Formula (I) of the present invention may also be synthesized according to the route shown below: 1) amide formation between B1 and E-COOH to give B2; 2) cyclization of B2 in phosphine oxychloride under heated to give B3; 3) bromination of B3 with NBS to give B4; 4) substitution of B4 by a protected amine (for example, 2,4-dimethoxybenzylamine) under base catalysis to give B5; 5) Suzuki coupling of B5 with a phenylboronic acid to give B6; 6) deprotection of B6 to give B7.
  • a functional group of B7 can be further derivatized to afford various target compounds.
  • Heterocyclic compounds shown in Formula (I) of the present invention may also be synthesized according to the route shown below: 1) amide formation of C1 with E-COOH to give C2; 2) cyclization of C2 in phosphine oxychloride under heat to give C3; 3) iodination of C3 with NIS to give C4; 4) amination of C4 to give C5; 5) Suzuki coupling of C5 a phenylboronic acid to give C6. Similarly, a functional group of C6 can be further derivatized to afford various target compounds.
  • Heterocyclic compounds shown in Formula (I) of the present invention may also be synthesized according to the route shown below: 1) substitution of D1 (A is N or CH) by E-NH 2 under base catalysis to give D2; 2) further substitution of D2 by a protected amine (such as dibenzylamine) to give D3; 3) reduction of D3 to give D4 (for example, with a reducing agent Zn/NH 4 Cl); 4) cyclization of D4 with carbonyl diimidazole or triphosgene in the present of a base (such as NEt 3 ) to give D5; 5) deprotection of D5 to give D6; 6) Chan-Lam coupling of D6 with a phenylboronic acid to give D7. Similarly, a functional group of D7 can be further derivatized to afford various target compounds.
  • the starting materials in the present invention were synthesized according to methods known in the art or purchased from ABCR GmbH & Co. KG, Acros Organics, Aldrich Chemical Company, Accela ChemBio Inc., Beijing Ouhe, etc.
  • the structure of a compound was determined by nuclear magnetic resonance (NMR) or mass spectrometry (MS).
  • NMR determination used a Bruker ASCEND-400 NMR spectrometer.
  • the solvent for the determination was deuterated dimethyl sulfoxide (DMSO-d 6 ), deuterated chloroform (CDCl 3 ) or deuterated methanol (CD 3 OD).
  • the internal standard was tetramethylsilane (TMS) and the chemical shift was given in a unit of 10 ⁇ 6 (ppm).
  • MS determination used an Agilent SQD (ESI) mass spectrometer (Agilent 6120).
  • HPLC determination used Agilent 1260 DAD high pressure liquid chromatograph (column: Poroshell 120 EC-C18, 50 ⁇ 3.0 mm, 2.7 ⁇ m) or Waters Arc high pressure liquid chromatograph (column: Sunfire C18, 150 ⁇ 4.6 mm, 5 ⁇ m).
  • Thin layer chromatography used GF254 silica gel plates from Qingdao Haiyang Chemical Co., Ltd. with a thickness of 0.15 to 0.2 mm, and the separation/purification of products by thin layer chromatography used silica plates with a thickness 0.4 to 0.5 mm.
  • reactions were run in room temperature (20-30° C.) and under an atmosphere of argon or nitrogen using a balloon with a volume of about 1 L.
  • Hydrogenation was carried out under an atmosphere of hydrogen using a balloon with a volume of about 1 L that was attached to the reaction vessel after being vacuumed and filled with hydrogen repeatedly for 3 times.
  • the microwave reaction used a CEM Discover-SP microwave reactor.
  • the reaction was monitored using Agilent LCMS (1260/6120) or thin layer chromatography.
  • the solvent eluting systems for column chromatography and TLC included a) dichloromethane/methanol, b) petroleum ether/ethyl acetate, or other systems as indicated.
  • the ratio of the solvents was adjusted according to the polarity of the compound, and further adjusted by addition of a small amount of TEA, or an acidic or alkaline reagent as needed.
  • the compound purification was alternatively done using Waters' MS-guided automated preparation system (abbreviated as prep-HPLC) with a MS detector (SQD2), eluting at a flow rate of 20 mL/min in an appropriate acetonitrile/water (containing 0.1% TFA or formic acid) or acetonitrile/water (containing 0.05% of 25-28% ammonium hydroxide) gradient (XBridge-C18, 19 ⁇ 150 mm, 5 ⁇ m).
  • Some compounds were prepared as HCl salts after prep-HPLC purification by addition of 1 N HCl to the collected fractions, followed by drying under reduced pressure.
  • DMF refers to N,N-dimethylformamide
  • DIPEA N,N-diisopropylethylamine
  • DBU refers to 1,8-diazabicycloundec-7-ene.
  • NBS N-bromosuccinimide
  • NIS refers to N-iodosuccinimide.
  • Pd(dppf)Cl 2 refers to [1,1′-bis(diphenylphosphino)ferrocene]dichloropalladium.
  • HATU refers to 2-(7-azabenzotriazole)-N,N,N′,N′-tetramethylurea hexafluorophosphate.
  • the abbreviation LDA refers to lithium diisopropylamide.
  • Trifluoroacetic acid (5 mL) was added to a solution of 1g (80 mg, 0.13 mmol) in dichloromethane (2 mL) and the resulting mixture was heated to 40° C. and stirred for 2 hours. After cooling to room temperature, the solvent was removed under reduced pressure and the residue was purified by prep-HPLC to give the title compound 1 (21 mg, solid, 34%).
  • Step 4 N-(4-(4-amino-1-((1s,4s)-4-hydroxycyclohexyl)-1H-pyrazolo[4,3-c]pyridin-3-yl)benzyl)-5-fluoro-2-methoxybenzamide (Compound 3) and N-(4-(4-amino-1-((1r,4r)-4-hydroxycyclohexyl)-1H-pyrazolo[4,3-c]pyridin-3-yl)benzyl)-5-fluoro-2-methoxybenzamide (Compound 4)
  • the intermediate as shown below was synthesized according to the procedures for the first to fourth steps in Example 4, except that tetrahydro-2H-pyran-3-carboxylic acid was used instead of (1r,4r)-4-(methoxycarbonyl)cyclohexane-1-carboxylic acid in the first step.
  • the intermediate as shown below was synthesized according to the procedures for the first to the fifth steps in Example 5, except that methyl 4-methylpiperidine-4-carboxylate was used instead of methyl piperidine-4-carboxylate in the first step.
  • Compound 44 was synthesized according to the procedure in Example 5 (without the sixth step), except that morpholine was used instead of methyl piperidine-4-carboxylate in the first step.
  • Step 8 (5-(8-amino-1-bromoimidazo[1,5-a]pyrazin-3-yl)tetrahydro-2H-pyran-2-yl)methyl acetate (8i)
  • the intermediate as shown below was synthesized according to the procedures for the sixth to ninth step in Example 11, except that (1r,4r)-4-(methoxycarbonyl)cyclohexane-1-carboxylic acid was used instead of cyclopentanecarboxylic acid in the sixth step.
  • Lithium aluminum hydride (8 mg, 0.19 mmol) was added to a solution of 18f (50 mg, 0.093 mmol) in THE (3 mL) at 0° C. The mixture was gradually warmed to room temperature and stirred for 3 hours. It was quenched with water (0.1 mL) and filtered. The filtrate was removed from the solvent under reduced pressure and the residue was purified by prep-HPLC to give the title compound 18 (15.4 mg, solid, 33%).
  • Step 3 methyl (1r,4r)-4-(((3-chloropyrazin-2-yl)methyl)carbamoyl)-1,4-dimethylcyclohexane-1-carboxylate (32d) and methyl (1s,4s)-4-(((3-chloropyrazin-2-yl)methyl)carbamoyl)-1,4-dimethylcyclohexane-1-carboxylate (32e)
  • Step 4 through Step 7. methyl (1r,4r)-4-(8-((2,4-dimethoxybenzyl)amino)-1-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)imidazo[1,5-a]pyrazin-3-yl)-1,4-dimethylcyclohexane-1-carboxylate (32j)
  • 32j was synthesized according to the procedures for step 2 to 5 in Example 4, except that 32d was used instead of 5b.
  • Step 8 methyl (1r,4r)-4-(8-amino-1-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)imidazo[1,5-a]pyrazin-3-yl)-1,4-dimethylcyclohexane-1-carboxylate (32k)
  • Compound 39 was synthesized according to the procedures for the fourth to eighth step in Example 23, except that 32e was used instead of 32d in the fourth step.
  • Step 1 methyl 4-(((3-chloropyrazin-2-yl)methyl)carbamoyl)bicyclo[2.2.2]octane-1-carboxylate (33b)
  • Step 4 methyl 4-(8-((2,4-dimethoxybenzyl)amino)-1-iodoimidazo[1,5-a]pyrazin-3-yl)bicyclo[2.2.2]octane-1-carboxylate (33e)
  • Step 5 methyl 4-(8-((2,4-dimethoxybenzyl)amino)-1-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)imidazo[1,5-a]pyrazin-3-yl)bicyclo[2.2.2]octane-1-carboxylate (33f)
  • Step 6 methyl 4-(8-amino-1-(4-((5-fluoro-2-methoxybenzamido)methyl)phenyl)imidazo[1,5-a]pyrazin-3-yl)bicyclo[2.2.2]octane-1-carboxylate (33)
  • Step 1 dimethyl (1s,4s)-1,4-dimethylcyclohexane-1,4-dicarboxylate (46a) and dimethyl (1r,4r)-1,4-dimethylcyclohexane-1,4-dicarboxylate (46b)
  • 46i was synthesized according to the procedures for the first to sixth step in Example 18, except that 46c was used instead of (R)-1-((benzyloxy)carbonyl)pyrrolidine-3-carboxylic acid 22a in the third step.
  • 46 was synthesized according to the procedures for the first to the sixth step in Example 26, except that 46i was used instead of 33 in the third step.
  • Compound 47 as shown below was synthesized according to the procedures for the second to the ninth step in Example 30, except that 46b was used instead of 46a in the second step.
  • Compound 49 can be synthesized according to the procedures in Example 32.
  • the enzymatic activity of BTK is determined by detecting the phosphorylation level of the substrate in the kinase reaction.
  • the reaction buffer contains the enzyme reaction buffer (1 ⁇ ) from the kit, 5 mM MgCl 2 , 1 mM DTT, 10 nM SEB and 0.01% Tween-20; the kinase reaction solution contains human-derived recombinant BTK protein (Carna Biosciences, Catalog No.
  • the substrate reaction solution contains biotin-labeled tyrosine kinase substrate diluted to 0.5 ⁇ M with the reaction buffer and 40 ⁇ M ATP;
  • the detection buffer contains Eu 3+ -labeled cage antibody diluted to 0.05 ng/ ⁇ L and streptavidin-labeled XL665 antibody diluted to 31.25 nM with the reaction buffer;
  • the test compound is dissolved and diluted to 100 ⁇ M with DMSO, followed by a 4-fold serial dilution with DMSO to the lowest concentration of 6.1 nM and finally 40-time dilution with the reaction buffer for each concentration point. If the IC 50 value of the compound is very low, the initial concentration of the compound is reduced.
  • the group without BTK is the negative control (100% inhibition) and the group with BTK but no compound is the positive control (0% inhibition).
  • the inhibition curve is plotted and the corresponding IC 50 value of the test compound is calculated using XLfit software (ID Business Solutions Ltd., UK).
  • HTRF time-resolved fluorescence
  • the reaction buffer contains the enzyme reaction buffer (1 ⁇ ) from the kit, 5 mM MgCl 2 , 1 mM DTT, 10 nM SEB, and 0.01% Tween-20;
  • the kinase reaction solution contains human recombinant BTK C481S protein (purified in-house) diluted to 1.5 ng/ ⁇ L with the reaction buffer;
  • the substrate reaction solution contains biotin-labeled tyrosine kinase substrate diluted to 0.5 ⁇ M with the reaction buffer and 35 ⁇ M ATP;
  • the detection buffer contains Eu 3+ -labeled cage antibody diluted to 0.05 ng/ ⁇ L and streptavidin-labeled XL665 antibody diluted to 31.25 nM with the reaction buffer;
  • the test compound is dissolved and diluted to 100 ⁇ M with DMSO, followed by a 4-fold serial dilution with DMSO to the lowest concentration of 6.1 nM and finally 40-time dilution with the reaction buffer for
  • the group without BTK is the negative control (100% inhibition) and the group with BTK C481 S but no compound is the positive control (0% inhibition).
  • the inhibition curve is plotted and the corresponding IC 50 value of the test compound is calculated using XLfit software (TD Business Solutions Ltd., UK).
  • BTK IC 50 (nM)
  • BTK C481S IC 50 (nM) 1. 17 3.5 2. 1.7 2.5 3. 3.2 4.6 4. 2.3 3.0 5. 4.6 2.6 6. 4.6 3.3 7. 3.4 1.7 8. 1.2 0.8 9. 718 498 10. 14 3.6 11. 20 3.9 12. 3.0 13. 1.4 0.8 14. 3.2 2.9 15. 1.4 1.1 16. 1.8 1.1 17. 6.3 2.4 18. 0.7 0.8 19. 212 201 21. 1.7 1.8 22. 155 36 23. 1.0 3.0 24. 3.9 2.5 25. 1.1 1.5 26. 3.8 11 27. 2.7 1.3 28. 1.0 2.2 29. 1.2 2.1 31. 0.6 3.0 32. 2.4 6.3 33. 0.8 1.8 34.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Immunology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Rheumatology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Pain & Pain Management (AREA)
  • Epidemiology (AREA)
  • Communicable Diseases (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
US18/169,423 2020-08-20 2023-02-15 Heterocyclic compounds as btk inhibitors Pending US20230192691A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN202010836243.4A CN114075190A (zh) 2020-08-20 2020-08-20 杂环类btk抑制剂
CN202010836243.4 2020-08-20
PCT/CN2021/113535 WO2022037649A1 (en) 2020-08-20 2021-08-19 Heterocyclic compounds as btk inhibitors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/113535 Continuation WO2022037649A1 (en) 2020-08-20 2021-08-19 Heterocyclic compounds as btk inhibitors

Publications (1)

Publication Number Publication Date
US20230192691A1 true US20230192691A1 (en) 2023-06-22

Family

ID=80282666

Family Applications (2)

Application Number Title Priority Date Filing Date
US18/169,423 Pending US20230192691A1 (en) 2020-08-20 2023-02-15 Heterocyclic compounds as btk inhibitors
US18/169,408 Pending US20230212175A1 (en) 2020-08-20 2023-02-15 Bridged bicyclic compounds as btk inhibitors

Family Applications After (1)

Application Number Title Priority Date Filing Date
US18/169,408 Pending US20230212175A1 (en) 2020-08-20 2023-02-15 Bridged bicyclic compounds as btk inhibitors

Country Status (5)

Country Link
US (2) US20230192691A1 (zh)
EP (1) EP4200283A1 (zh)
JP (2) JP2023538090A (zh)
CN (2) CN114075190A (zh)
WO (2) WO2022037649A1 (zh)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021180107A1 (en) * 2020-03-12 2021-09-16 Fochon Pharmaceuticals, Ltd. Compounds useful as kinase inhibitors
CN114573586B (zh) * 2020-11-28 2023-11-03 杭州和正医药有限公司 一种抑制布鲁顿酪氨酸激酶活性的多环化合物、药物组合物及其应用
CN114634512B (zh) * 2020-12-16 2023-11-14 江苏恒瑞医药股份有限公司 作为布鲁顿酪氨酸激酶抑制剂的化合物、其制备方法和医药应用
CN116693458A (zh) * 2022-03-03 2023-09-05 深圳市塔吉瑞生物医药有限公司 环烷基或杂环基取代的杂芳基化合物及其组合物及用途

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2016009403A (es) * 2014-02-03 2016-09-16 Cadila Healthcare Ltd Compuestos heterociclicos.
GB201410430D0 (en) * 2014-06-11 2014-07-23 Redx Pharma Ltd Compounds
WO2016106623A1 (en) * 2014-12-31 2016-07-07 Merck Sharp & Dohme Corp. Benzamide imidazopyrazine btk inhibitors
WO2016106652A1 (en) * 2014-12-31 2016-07-07 Merck Sharp & Dohme Corp. Biarylether imidazopyrazine btk inhibitors
WO2016106624A1 (en) * 2014-12-31 2016-07-07 Merck Sharp & Dohme Corp. Tertiary alcohol imidazopyrazine btk inhibitors
KR20180109842A (ko) * 2015-09-16 2018-10-08 록쏘 온콜로지, 인코포레이티드 화합물
CN112424203B (zh) * 2018-07-20 2022-07-26 正大天晴药业集团股份有限公司 布鲁顿酪氨酸激酶抑制剂
CN110964016B (zh) * 2018-09-29 2021-05-28 南京药捷安康生物科技有限公司 氨基降茨烷衍生物及其制备方法与应用
AU2020207953A1 (en) * 2019-01-18 2021-08-26 Hangzhou Bangshun Pharmaceutical Co., Ltd. Bruton's tyrosine kinase inhibitors
WO2021180107A1 (en) * 2020-03-12 2021-09-16 Fochon Pharmaceuticals, Ltd. Compounds useful as kinase inhibitors

Also Published As

Publication number Publication date
WO2022037649A1 (en) 2022-02-24
JP2023538091A (ja) 2023-09-06
EP4200283A1 (en) 2023-06-28
WO2022037650A1 (en) 2022-02-24
CN114075190A (zh) 2022-02-22
JP2023538090A (ja) 2023-09-06
CN116670119A (zh) 2023-08-29
US20230212175A1 (en) 2023-07-06

Similar Documents

Publication Publication Date Title
US11285140B2 (en) Piperidin-4-yl azetidine derivatives as JAK1 inhibitors
US20230192691A1 (en) Heterocyclic compounds as btk inhibitors
US20220348594A1 (en) Heterocyclic compounds as immunomodulators
RU2695664C1 (ru) Производные имидазопиридина в качестве модуляторов активности tnf
RU2679609C1 (ru) Производные имидазопиридазина в качестве модуляторов активности tnf
RU2677697C1 (ru) Производные триазолопиридина в качестве модуляторов активности tnf
KR20190057132A (ko) Il-12, il-23 및/또는 ifn 알파 반응의 조정제로서 유용한 이미다조피리다진 화합물
RU2677698C1 (ru) Производные триазолопиридазина в качестве модуляторов активности tnf
NZ769326A (en) Heterocyclylamines as pi3k inhibitors
US11046658B2 (en) Aminopyrazine derivatives as PI3K-γ inhibitors
US20160272645A1 (en) Heterocyclic Compounds and Methods of Use
US10669286B2 (en) Fused pentacyclic imidazole derivatives as modulators of TNF activity
RU2697090C1 (ru) Производные тетрагидроимидазопиридина в качестве модуляторов активности tnf
US10550125B2 (en) Prodrugs of imidazotriazine compounds as CK2 inhibitors
JP2023521172A (ja) キナーゼ阻害剤
US20240043405A1 (en) Potent and selective irreversible inhibitors of irak1
US11884650B2 (en) Substituted heterocyclic compounds
WO2022241175A1 (en) Substituted heterocyclic compounds
NZ717505B2 (en) Heterocyclylamines as pi3k inhibitors
NZ765458B2 (en) Heterocyclylamines as pi3k inhibitors
NZ735378B2 (en) Heterocyclylamines as pi3k inhibitors
NZ751428B2 (en) Heterocyclylamines as pi3k inhibitors

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION UNDERGOING PREEXAM PROCESSING

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION