[go: up one dir, main page]

WO2014040555A1 - Dérivé de noyau hétéroaromatique contenant de l'azote utile comme inhibiteur de la tyrosine kinase - Google Patents

Dérivé de noyau hétéroaromatique contenant de l'azote utile comme inhibiteur de la tyrosine kinase Download PDF

Info

Publication number
WO2014040555A1
WO2014040555A1 PCT/CN2013/083429 CN2013083429W WO2014040555A1 WO 2014040555 A1 WO2014040555 A1 WO 2014040555A1 CN 2013083429 W CN2013083429 W CN 2013083429W WO 2014040555 A1 WO2014040555 A1 WO 2014040555A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
alkyl
hydrogen atom
compound
methyl
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.)
Ceased
Application number
PCT/CN2013/083429
Other languages
English (en)
Chinese (zh)
Inventor
罗浩贤
张艳
张倩
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.)
KBP Biosciences Co Ltd
Original Assignee
KBP Biosciences 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 KBP Biosciences Co Ltd filed Critical KBP Biosciences Co Ltd
Priority to CN201380041846.3A priority Critical patent/CN104583195B/zh
Publication of WO2014040555A1 publication Critical patent/WO2014040555A1/fr
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/58Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin
    • 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
    • C07D401/04Heterocyclic 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 directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • 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
    • A61K31/4025Heterocyclic 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 not condensed and containing further heterocyclic rings, e.g. cromakalim
    • 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
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/48Two nitrogen 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
    • 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
    • C07D401/14Heterocyclic 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 three or more hetero rings
    • 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
    • C07D403/12Heterocyclic 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 chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/12Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • the present invention relates to the field of medical technology, and particularly relates to a nitrogen-containing heteroaromatic ring derivative as a tyrosine kinase inhibitor, a pharmaceutically acceptable salt thereof or a stereoisomer thereof, a preparation method of these compounds, a drug containing the same Compositions for the prevention and/or treatment of B cell-associated blood cancers in an individual (eg, B-cell chronic lymphocytic carcinoma, non-Hodgkin's lymphoma), inflammatory and autoimmune diseases (eg, rheumatoid arthritis, Methods for systemic lupus erythematosus, etc., and the preparation of these compounds for the prevention and/or treatment of B cell-associated blood cancer (eg, B-cell chronic lymphocytic carcinoma, non-Hodgkin's lymphoma), inflammatory and autoimmune diseases Use in drugs such as rheumatoid arthritis, systemic lupus erythematosus, etc.
  • Protein Shield Kinases constitute one of the largest families of human enzymes and regulate many different signaling processes by the addition of phosphate groups to protein shields (T. Hunter, Cell 1987 50: 823-829).
  • the tyrosine kinase phosphorylation protein is shielded in the hydroxyl portion of the tyrosine residue.
  • the tyrosine kinase family includes members that control cell growth, migration, and differentiation.
  • Abnormal kinase activity has been implicated in many human diseases, including cancer, autoimmune diseases, and inflammatory diseases.
  • protein shield kinases are key regulators of cell signaling, they provide the goal of regulating cell function with small molecule kinase inhibitors and are therefore a good drug design target.
  • selective and potent inhibitors of kinase activity can be used to study cellular signaling processes and to identify other therapeutically significant cell targets.
  • B cells play a key role in the pathogenesis of autoimmune and/or inflammatory diseases.
  • Protein shield-based therapeutics that deplete B cells, such as Rituxan are effective against autoantibody-induced inflammatory diseases such as rheumatoid arthritis (Rastetter et al, Annu Rev Med 2004 55: 477). Therefore, inhibitors of protein shield kinase that play a role in B cell activation should be useful therapeutic agents for B cell mediated pathology such as autoantibody production.
  • BCR B cell receptor
  • Btk is a member of the Tec family of tyrosine kinases and has been shown to be a key regulator of early B cell formation and activation and survival of mature B cells (Khan et al, Immunity 1995 3: 283; Ellmeier et al, J. Exp. Med. 2000 192 : 1611).
  • Human Btk mutations lead to the condition X-linked gamma globulin deficiency (XLA) (Lindvall et al Immunol. Rev. 2005 203: 200). These patients are immunocompromised and show impaired B cell maturation, reduced immunoglobulin and peripheral B cell levels, reduced T cell-independent immune response, and reduced calcium use following BCR stimulation.
  • XLA X-linked gamma globulin deficiency
  • Btk-deficient mice showed a significant improvement in disease progression in a preclinical mouse model of systemic lupus erythematosus (SLE). Furthermore, Btk-deficient mice are resistant to collagen-induced arthritis (Jansson and Holmdahl Clin. Exp. Immunol. 1993 94: 459). The dose-dependent efficacy of selective Btk inhibitors in the mouse arthritis model has been demonstrated (Z. Pan et al, Chem. Med Chem. 2007 2: 58).
  • Btk also has cellular expression that may be involved in disease processes in addition to B cells.
  • Btk is expressed by mast cells and Btk-deficient bone marrow-derived mast cells show impaired antigen-induced degranulation (Iwaki J. Biol. Chem. 2005 280: 40261). This shows that Btk can be used to treat pathological mast cell responses such as allergies and asthma.
  • monocytes from XLA patients lacking Btk activity show reduced TNFa production following stimulation (Horwood et al J Exp Med 2003 197: 1603).
  • TNFa-mediated inflammation can be modulated by small molecule Btk inhibitors.
  • Btk has been reported to play a role in apoptosis (Islam and Smith Immunol. Rev. 2000 178: 49), and thus Btk inhibitors will be effective in the treatment of certain B cell lymphomas and leukemias (Feldhahn et al. Exp. Med. 2005 201 : 1837).
  • Dasatinib which was launched in 2006, is a multi-inhibitor inhibitor, which has a strong inhibitory effect on Btk and is used to treat chronic myelogenous leukemia.
  • PCI-32765 which is in clinical phase III study, is also a multi-defect inhibitor, and its inhibitory effect on Btk is Irreversible, used to treat lymphoma, leukemia and autoimmune diseases.
  • CC-292 also known as AVL-292
  • Its irreversible selective inhibition of Btk is used to treat leukemia and
  • the present invention provides nitrogen-containing heteroaryl ring derivatives useful as tyrosine kinase inhibitors, which are excellent Btk inhibitors, and which can be used for the prevention and/or treatment of B cell-associated blood cancer, inflammatory and/or Or autoimmune disease.
  • the present invention provides a compound represented by the following formula (I), a pharmaceutically acceptable salt thereof or
  • ring A and ring B each independently represent a phenyl group, a 3- 7 membered cycloalkyl group, a 3- 7 membered heterocycloalkyl group having a N, 0, S hetero atom, a 4- 7 membered heteroaryl group or 6- 10-membered two-ring structure;
  • Li and L 2 each independently represent a covalent bond, -NH-, -N(C 1-3 alkyl) -, -0- , - S(0) m - , - N(C 1-3 alkyl) C(0)- , - C(0)N(C 1-3 alkyl) -, - N(C 1 -3 alkyl)S(0) 2 - or - S(0) 2 N(C 1- 3 alkyl) -;
  • A represents a covalent bond, a substituted or unsubstituted alkylene d_ 4 alkyl substituted amino
  • b represents -CO- or -S0 2 -;
  • c represents 1,3-propenylene, 1,1- or 1,2-vinylidene, ethynylene, or unsubstituted or unsubstituted or substituted by one or two methyl or trifluoromethyl groups a 1,3-butadiene-1,4-subunit substituted with one to four methyl or trifluoromethyl groups;
  • d represents a covalent bond or an alkylene group
  • e represents a hydrogen atom, an alkoxy group, an amino group, a 3- 7 membered cycloalkyl group, a 6- to 10-membered bicyclic structure, an alkylamino group or a bis-(Cw alkyl)amino group, wherein the alkyl moieties may be the same or different
  • L 3 represents a covalent bond, -NH-, -N(C 1-3 alkyl) -, -0- , - 0- C 1-3 alkylene-, -S-Ci -3 alkylene, - S(0) m - , - C(O)- , -NHC(O)- , - N(C 1-3 alkyl) C(O)- , - C(0)NH- , -C(0) N(Ci -3 alkyl) -, - NHS(0) 2 - , -N(Ci -3 alkyl) S(0) 2 - , -S(0) 2 NH- , -S(0) 2 N (Ci -3 alkyl) -, - OC(O)- or - C(0)0- ,
  • R4 represents a hydrogen atom, an alkyl group, -N(Cw alkyl) 2 , -NHC(0)0-(Cw alkyl), -OH, - 0(C 1-4 alkyl), - S(0) 2 (C 1-3 alkyl), 3- 7 membered cycloalkyl, phenyl or 5- 6-membered heteroaryl;
  • R 2 represents - L 4 - R 5 ,
  • L 4 represents a covalent bond, -NH-, -N(C 1-3 alkyl)-, - 0-C 1-3 alkylene-, -S-C 1-3 alkylene- or -S ( 0) m - ,
  • R 5 represents a hydrogen atom, C 2-4 alkenyl, C 2-4 alkynyl, C 1-4 alkyl, amino, -NH(C 1-3 alkyl), -N(Cw alkyl) 2 , - OH , - 0 (Cw alkyl), - C(0) (Cw alkyl) or 3- 7 membered cycloalkyl,
  • R 5 cannot be C 1-4 alkyl, - OH , - 0 (C 1-3 alkyl), - C(0) (C 1-3 alkyl),
  • L 4 is - 0- d_ 3 alkylene - when, R 5 is not a hydrogen atom;
  • alkyl moiety, cycloalkyl group, heteroaryl group may be further substituted by 1 to 4 ( ⁇ ,
  • the carbon atom of the cycloalkyl or bicyclic structure may be replaced by 1-4 identical or different N, NH, N(C 1-3 alkyl), 0, S(0) m , C(O);
  • the heteroaryl group has 1-4 hetero atoms, and each of them is independently selected from N, 0 or S;
  • n 0, 1 or 2;
  • p and q are independent representations of 0, 1, 2, 3 or 4.
  • the present invention provides the above-described formula (I) a compound, a pharmaceutically acceptable salt thereof or a stereoisomer thereof, wherein:
  • Ring A and Ring B each independently represent a phenyl group, a 3- 7 membered cycloalkyl group, a 3-7 membered heterocycloalkyl group having a N, 0, S hetero atom, a 4-7 membered heteroaryl group or a 6-10 member.
  • Li and L 2 each independently represent a covalent bond, -NH-, -N(C 1-3 alkyl) -, -0- , - S(0) m - , - N(C 1-3 alkyl) C(0)- , - C(0)N(C 1-3 alkyl) -, - N(C 1 -3 alkyl)S(0) 2 - or - S(0) 2 N(C 1- 3 alkyl) -;
  • A represents a covalent bond, a substituted or unsubstituted alkylene d_ 4 alkyl substituted amino
  • b represents -CO- or -S0 2 -;
  • c represents 1, 3-propenylene, 1,1- or 1,2-vinylidene, ethynylene, or unsubstituted, which is unsubstituted or substituted by one or two methyl or trifluoromethyl groups. Or a 1,3-butadiene-1,4-subunit substituted by one to four methyl groups or by a trifluoromethyl group;
  • d represents a covalent bond or an alkylene group
  • e represents a hydrogen atom, an alkoxy group, an amino group, a 3- 7 membered cycloalkyl group, a 6- to 10-membered bicyclic structure, an alkylamino group or a bis-(Cw alkyl)amino group, wherein the alkyl moieties may be the same or different;
  • L 3 represents a covalent bond, -NH-, -N(C 1-3 alkyl) -, -0- , - 0- C 1-3 alkylene-, -S-Ci -3 alkylene, - S(0) m - , - C(O)- , -NHC(O)- , - N(C 1-3 alkyl) C(O)- , - C(0)NH- , -C(0) N(Ci -3 alkyl) -, - NHS(0) 2 - , -N(Ci -3 alkyl) S(0) 2 - , -S(0) 2 NH- , - S(0) 2 N (C 1-3 alkyl) -, - OC(O)- or - C(0)0- ,
  • R4 represents a hydrogen atom, an alkyl group, -N(Cw alkyl) 2 , -NHC(0)0-(Cw alkyl), -OH, - 0(C 1-4 alkyl), - S(0) 2 (C 1-3 alkyl), 3- 7 membered cycloalkyl, phenyl or 5- 6-membered heteroaryl;
  • R 2 represents - L 4 - R 5 ,
  • L 4 represents -S-C 1-3 alkylene- or -S(0) m - ,
  • R 5 represents a hydrogen atom, C 2-4 alkenyl, C 2-4 alkynyl, C 1-4 alkyl, amino, -NH(C 1-3 alkyl), -N(Cw alkyl) 2 , - OH , - 0 (Cw alkyl), - C(0) (Cw alkyl) or 3- 7 membered cycloalkyl,
  • n 0, 1 or 2.
  • alkyl moiety, cycloalkyl group, heteroaryl group may be further substituted by 1 to 4 ( ⁇ ,
  • the carbon atom of the cycloalkyl or bicyclic structure may be replaced by 1-4 identical or different N, NH, N(C 1-3 alkyl), 0, S(0) m , C(O);
  • the heteroaryl group contains 1-4 heteroatoms, each independently selected N, 0 or S;
  • n 0, 1 or 2;
  • p and q are independent representations of 0, 1, 2, 3 or 4.
  • the present invention provides a compound represented by the above formula (I), a pharmaceutically acceptable salt thereof or a stereoisomer thereof, wherein:
  • Ring A and Ring B each independently represent a phenyl group, a 5- to 6-membered cycloalkyl group, a 5-6 membered heterocycloalkyl group having a N, 0, S hetero atom, a 5-6 membered heteroaryl group or 8-10 members.
  • Li and L 2 each independently represent a covalent bond, -NH-, -N(CH 3 )-, - ⁇ -, - S(0) m - , -N(CH 3 )C(0)-, -C (0)N(CH 3 ) -, - N(CH 3 )S(0) 2 - or - S(0) 2 N(CH 3 ) -;
  • a represents a covalent bond, an imino group that is unsubstituted or substituted with CH 3 ;
  • b represents -CO- or -S0 2 -;
  • c represents a 1,2-vinylidene or ethynylene group which is unsubstituted or substituted by one or two methyl groups;
  • d represents a covalent bond or a methylene group;
  • e represents a hydrogen atom, a methoxy group, an amino group, a piperidinyl group, a morpholinyl group, a pyrrolidinyl group, a piperazinyl group, a TV-methyl piperazinyl group, a 6- to 9-membered spiro ring structure, a 6- to 8-membered ring structure. , 6- to 8-membered bridged ring structure, methylamino or bis-(methyl)amino;
  • L 3 represents a covalent bond, -NH-, -N(C 1-3 alkyl)-, -0-, - 0-C 1-3 alkylene-, -S-Ci -3 alkylene, - S(0) m - , - C(O)- , -NHC(O)-, - C(0)NH- , - NHS(0) 2 - , - S(0) 2 NH- , - oc(o )- or - c(o)o- ,
  • R4 represents a hydrogen atom, an alkyl group, -N(Cw alkyl)2, -NHC(0)0-(Cw alkyl), -OH, - 0(C 1-4 alkyl), - S(0) 2 (C 1-3 alkyl), 5- 6-membered cycloalkyl, phenyl or 5- 6-membered heteroaryl;
  • R 2 represents - L 4 - R 5 ,
  • L 4 represents -S-C 1-3 alkylene- or -S(0) m - ,
  • R 5 represents a hydrogen atom, a C 1-4 alkyl group, an amino group, -NH(C 1-3 alkyl), -N(C 1-3 alkyl) 2 ,
  • alkyl moiety, cycloalkyl group, phenyl group, heteroaryl group, spiro ring structure, concentric ring structure, bridged ring structure may be further substituted by 1 to 4 ( ⁇ ,
  • the carbon atom of the cycloalkyl or bicyclic structure may be replaced by 1-4 identical or different N, NH, N(C 1-3 alkyl), 0, S(0) m , C(O);
  • heteroaryl group, the spiro ring structure, the concentric ring structure, and the bridged ring structure contain 1-4 hetero atoms, and each of them is independently selected from N, 0 or S;
  • n means 0 , 1 or 2 ;
  • p and q are independent representations of 0, 1, 2, 3 or 4.
  • the present invention provides a compound represented by the above formula (I), a pharmaceutically acceptable salt thereof or a stereoisomer thereof, wherein:
  • Ring A and ring B each independently represent a phenyl group, a 5-6 membered heterocycloalkyl group having a N hetero atom or a 5-6 membered heteroaryl group;
  • Li and L 2 are independently represented by -NH- , - 0- or - S(0) m -;
  • a represents a covalent bond or an imino group
  • c 1,2-vinylidene
  • d represents a covalent bond or a methylene group
  • e represents a hydrogen atom, piperidinyl, morpholinyl, pyrrolidinyl, piperazinyl or bis-(methyl)amino;
  • Ri represents a hydrogen atom, a gas atom, a methyl group, a methoxy group, a methylamino group or a di-(methyl)amino group which is unsubstituted or substituted with one to three atomic atoms;
  • R 3 represents a hydrogen atom, a halogen atom or -L 3 - R 4 ,
  • L 3 represents a covalent bond, -NH-, -N(C 1-3 alkyl)-, -0- , - 0- C 1-3 alkylene-, -S-Ci -3 alkylene-, - S(0) m - , - C(O)- , -NHC(O)- , - C(0)NH- , - OC(O)- or -C(0)0-
  • R4 represents a hydrogen atom, Methyl, ethyl, -N(C 1-3 alkyl) 2 , - NHC(0)0-CH 3 , - 0(CH 3 ) , -0(CH 2 CH 3 ) , - 0 (C(CH) 3 ) 3 ), -S(0) 2 -CH 3 , cyclopentyl, cyclohexane, pyrrolidinyl, tetrahydrofuranyl, piperidinyl, morpholinyl, piperazinyl,
  • methylthio group, ethylthio group, methylsulfonyl group, aminosulfonyl group, methylsulfinyl group, aminosulfinyl group may be further one to two of the same or different methoxy, pyrrolidinyl, tetra Hydroimidazolyl, piperidinyl, morpholinyl, piperazinyl,
  • the pyrimidinyl group may be further substituted by one to two identical or different,
  • Derivative atom methyl, amino, methylamino, bis-(methyl)amino, hydroxy, methoxy, methoxycarbonyl, carbamoyl, methylcarbamoyl or bis-(methyl)carbamoyl ;
  • n 0, 1 or 2;
  • p and q stand independently for 0, 1 or 2.
  • the present invention provides a compound represented by the above formula (I), a pharmaceutically acceptable salt thereof or a stereoisomer thereof, wherein:
  • Ring A and Ring B each independently represent phenyl, pyridyl, piperidinyl, imidazolyl, pyrazolyl, thiazolyl, thiazyl, 1,2,3-triazolyl or 1,2,4-tri Azolyl
  • Li and L 2 are independently represented by -NH- or - 0-;
  • a represents a covalent bond or an imino group
  • c 1,2-vinylidene
  • d represents a covalent bond or a methylene group
  • e represents a hydrogen atom, piperidinyl, pyrrolidinyl, morpholinyl, piperazinyl or bis-(methyl)amino;
  • R 3 represents a hydrogen atom, a fluorine atom, a chlorine atom or -L 3 - R 4 ,
  • L 3 represents a covalent bond, -NH-, - N (Ci -3 alkyl) -, - 0- , - 0- CH 2 CH 2 - , -S-CH 2 CH 2 - or - s(o) m -,
  • R4 represents a hydrogen atom, methyl, ethyl, -N(C 1-3 alkyl) 2 , -NHC(0)0-CH 3 , - 0(CH 3 ),
  • phenyl group, pyrrolyl group, imidazolyl group, thiazolyl group, oxazolyl group, thiadiazolyl group, pyridyl group Can be further replaced by one or two identical or different,
  • R 2 represents an ethylthio group which is unsubstituted or substituted by a methoxy group, a piperidinyl group or a morpholinyl group, a methylthio group, a methylsulfonyl group, an aminosulfonyl group, a methylsulfinyl group, an aminosulfinyl group.
  • m means 0, 1 or 2;
  • the present invention provides a compound represented by the above formula (I), a pharmaceutically acceptable salt thereof or a stereoisomer thereof, wherein:
  • Ring A and ring B each independently represent a phenyl group, a pyridyl group, a pyrazolyl group or a piperidinyl group;
  • Li and L 2 are independently represented by -NH-;
  • a represents an imino group
  • c 1,2-vinylidene
  • d represents a covalent bond or a methylene group
  • e represents a hydrogen atom, a pyrrolidinyl group, a morpholinyl group, a piperazinyl group or a bis-(methyl)amino group
  • Ri represents a hydrogen atom
  • R 3 represents - L 3 - R4 , L 3 represents a covalent bond, - 0- or - 0-CH 2 CH 2 -, R4 represents -N(CH 3 ) 2 , -0(CH 3 ), tetrahydrofuranyl, Phenyl, phenyl or pyridyl,
  • the phenyl group and the pyridyl group may be further substituted by one to two identical or different, and represent a carbamoyl group, a methylcarbamoyl group or a bis-(methyl)carbamoyl group;
  • R 2 represents an ethylthio group which is unsubstituted or substituted with a methoxy group or a morpholinyl group, a methylthio group, a methylsulfonyl group, an aminosulfonyl group, a methylsulfinyl group, an aminosulfinyl group;
  • p 1 ;
  • the present invention provides a compound represented by the above formula (I), a pharmaceutically acceptable salt thereof or a stereoisomer thereof, wherein:
  • Ring A represents phenyl
  • ring B represents phenyl, pyrazolyl or pyridyl
  • L 2 independently represents -NH-
  • a represents an imino group
  • b represents -CO-
  • c represents 1,2-vinylidene
  • d represents a covalent bond
  • e represents a hydrogen atom
  • R 3 represents - L 3 - R4 , L 3 represents a covalent bond, -NH-, - N(C 1-3 alkyl) -, -0-, - 0- C 1-3 alkylene-, - S - C 1-3 alkylene-, or - C(0)NH- , R4 represents a hydrogen atom, methyl, ethyl, - 0(CH 3 ), - 0(C(CH 3 ) 3 ) or morpholinyl; R 2 represents -L 4 - R 5 , and L 4 represents -S-C 1-3 alkylene- or -S(0) m - , R 5 represents a hydrogen atom or p represents 1; q represents 1.
  • the "d- 6 alkyl group” as used in the present invention means a linear or branched alkyl group having 1 to 6 carbon atoms, and includes “alkyl", “Cw alkyl” and the like, and examples thereof include, but are not limited to, For example, methyl, ethyl, n-propyl, isopropyl, n-butyl, 2-methylpropyl, 1-methylpropyl, 1,1-dimethylethyl and the like.
  • the terms “Cw alkyl” and “Cw alkyl” refer to specific examples containing 1 to 4, 1 to 3 carbon atoms in the above examples.
  • the "d- 6 alkylene group” as used in the present invention means a structure in which a straight or branched alkyl group having 1 to 6 carbon atoms is removed by one hydrogen atom, and means a divalent alkyl group.
  • the "alkylene chain” is a polymethylene group, that is, -(CH 2 )x- , wherein X is a positive integer, preferably 1 to 6, 1 to 4, 1 to 3, 1 to 2 or 2 to 3.
  • the substituted alkylene chain is a polymethylene group in which one or more methylene hydrogen atoms are replaced by a substituent.
  • C 1-4 alkylene “C 1-3 alkylene” and the like, examples of which include, but are not limited to, for example, methylene (-CH 2 - ), ethylene ( - CH 2 CH 2 - ), propylene (-CH 2 CH 2 CH 2 - ), butylene (-CH 2 CH 2 CH 2 CH 2 - ), and the like.
  • C 1-4 alkylene “C 1-3 alkylene” refers to a specific example containing 1 to 4, 1 to 3 carbon atoms in the above examples.
  • C 2 _ 4 alkenyl group as used in the present invention means a linear or branched alkenyl group having 2 to 4 carbon atoms; examples thereof include, but are not limited to, for example, a vinyl group, a 1-propenyl group, 2-propenyl, 1-methylvinyl, 1-butenyl, 2-butenyl, 3-butenyl, 1-methyl-1-propenyl, 2-methyl-i-propenyl, 1-methyl-2-propenyl, 2-methyl-2-propenyl.
  • C 2 _ 4 alkynyl group in the present invention means the number of carbon atoms containing a triple bond to 2-4 straight or branched alkynyl group; examples thereof include, but are not limited to, ethynyl, 2-propynyl Base, 2-butynyl, 3-butynyl, 1-methyl-2-propynyl, and the like.
  • the "d- 6 alkoxy group” of the present invention means a group in which the term “d- 6 alkyl group” is bonded to another structure through an oxygen atom, such as a methoxy group, an ethoxy group, a propoxy group, an isopropoxy group, or a butyl group. Oxyl, isobutoxy, tert-butoxy, sec-butoxy, pentyloxy, neopentyloxy, hexyloxy and the like. Alkoxy groups are preferred, and d- 3 alkoxy groups are more preferred.
  • the terms “d- 4 alkoxy", “d- 3 alkoxy” refer to the group “alkyl", "d- 3 alkyl” attached to the other structure through an oxygen atom.
  • the "d- 6 alkylamido" of the present invention means a group in which "d- 6 alkyl" is bonded to other structures via an acylamino group.
  • the "prime” as used in the present invention means a fluorine atom, a chlorine atom, a bromine atom or an iodine atom.
  • the "3- 7-membered cycloalkyl group” as used in the present invention means that the ring atoms are all carbon atoms, and a hydrogen atom-derived cyclic alkyl group is removed, including, for example, "3- 6-membered cycloalkyl group", "4".
  • cycloalkyl ""5- 7-membered cycloalkyl", "5-6-membered cycloalkyl”, examples of which include, but are not limited to, cyclopropyl, cyclobutane, cyclopentyl, cyclohexyl Alkyl, cycloheptyl, cyclooctyl, cyclopentanone, cyclohexanone, and the like.
  • the "3- 7-membered heterocycloalkyl group” as used in the present invention means a 3-7 membered cyclic group having one or more hetero atoms, and the "hetero atom” means a nitrogen atom, an oxygen atom, a sulfur atom, or the like. .
  • a 3- to 6-membered heterocyclic group is preferred, and a 5- to 6-membered heterocyclic group is more preferred.
  • "4-7-membered heteroaryl” means an aromatic group consisting of 4 to 7 ring atoms (having at least one hetero atom), including "5- to 7-membered heteroaryl""5- to 6-membered heteroaryl” Specific examples include, but are not limited to, furyl, thiyl, pyrrolyl, thiazolyl, thiadiazolyl, oxazolyl, oxadiazolyl, imidazolyl, pyrazolyl, pyridyl, pyrimidinyl, 1,4-dioxadienyl, 2//- 1,2-oxazinyl, 4//- 1,2-oxazinyl, 6//- 1,2-oxazinyl, 4 //- 1,3-oxazinyl, 6//-1,3-oxazinyl, 4//- 1,4-oxazinyl, pyridazinyl, pyridyl 11 -methyl, 1,
  • 6-10 membered bicyclic structure means a bicyclic group consisting of 6 to 10 ring atoms (may not contain or contain one or more heteroatoms:), including "7-10 membered bicyclic structure”,”8-10 “Secondary ring structure”, “6-9 element spiral ring structure”, “6-8 yuan parallel ring structure”, “6-8 yuan bridge ring structure”, etc.
  • aromatic bicyclic structures including, but not limited to, benzofuranyl, benzisofuranyl, benzothianyl, fluorenyl, benzoxazolyl, benzo Imidazolyl, carbazolyl, benzotriazolyl, quinolyl, isoquinolyl, acridinyl, phenanthryl, benzoxazinyl, pyridazinyl, quinazolinyl, quinoxalinyl, Phenazinyl, acridinyl, fluorenyl, naphthyridyl, 1 ,3-dihydrobenzofuranyl, benzo[[1.3]dioxolyl, isoindolyl, chromanyl, 1,2,3,4-tetrahydropyrrolo[3,4-c]pyrrolyl, 5,6-dihydroimidazole[1.2- ⁇ ]pyrazine-7(8
  • the "6-9-membered spirocyclic group" of the present invention means that at least two rings of a class share an atomic shape.
  • a 6- to 9-membered fused ring structure include, but are not limited to, spiro[3.3]heptyl, spiro[3.4]octyl, spiro[3.5]decyl, spiro[4.4]decyl, spiro[3.4]oct-6-ene , snail [3.5] ⁇ -6-alkenyl, spiro[4.4] ⁇ -6-alkenyl, spiro[4.4] ⁇ -2,7-dienyl, 2-oxaspiro[3.3]heptanyl, 6-oxaspiro[2.5]octyl, 4-oxa-7-aminospiro[2.5]octyl, 2-aminospiro[3.3]heptanyl, 2-oxa-6-aminospiro[3.3 Heptylalkyl, 2-
  • the "6- to 8-membered ring structure" as used in the present invention refers to a 6-8 membered cyclic group formed by two or more ring structures sharing two adjacent atoms with each other, and specific examples thereof include But not limited to: bicyclo [3.1.0] hexane, bicyclo [4.1.0] heptyl, bicyclo [2.2.0] hexane, bicyclo [3.2.0] heptyl, bicyclo [4.2.0] Octyl, bicyclo[3.1.0]hex-2-enyl, bicyclo[4.1.0]hept-3-enyl, bicyclo[3.2.0]hept-3-yl,bicyclo[ 4.2.0] Oct-3-enyl, benzofuranyl, benzoisofuranyl, benzothianyl, decyl, benzoxazolyl, benzimidazolyl, oxazolyl, benzotrien Azyl, quinolyl, isoquinolyl, acridiny
  • the "6- to 8-membered bridged ring structure" of the present invention refers to a 6- to 8-membered cyclic group formed by two or more ring-shaped structures sharing two non-adjacent atoms with each other, and a specific embodiment thereof Including, ⁇ , ⁇ , ⁇ , ⁇ , ⁇ , ⁇ , ⁇ , , ⁇ , ⁇ , 0,
  • i JSr (3, ⁇ , ⁇ , The above group may be further substituted by 1 to 4, ( ⁇ represents a halogen atom, a C alkyl group, an amino group, an alkylamino group, a bis-(C alkyl)amino group, a hydroxyl group, an alkoxy group, an alkoxycarbonyl group, an amino group.
  • represents a halogen atom, a C alkyl group, an amino group, an alkylamino group, a bis-(C alkyl)amino group, a hydroxyl group, an alkoxy group, an alkoxycarbonyl group, an amino group.
  • Formyl, alkylcarbamoyl, bis-(Cw alkyl)carbamoyl or 3- 6 membered cycloalkyl which may be the same or different.
  • Particularly preferred compounds of the formula (I) according to the invention include:
  • Particularly preferred compounds of the formula (I) according to the invention further comprise:
  • the invention provides a process for the preparation of a compound of the invention described above. In one embodiment, the invention provides a pharmaceutical group comprising a compound of the invention described above Compound.
  • the invention provides the use of a compound of the invention described above for the prevention and/or treatment of B cell associated blood cancer in an individual (eg, B cell chronic lymphocytic carcinoma, non-Hodgkin's lymphoma), inflammatory and autoimmune A method of disease (eg, rheumatoid arthritis, systemic lupus erythematosus, etc.).
  • a compound of the invention described above for the prevention and/or treatment of B cell associated blood cancer in an individual (eg, B cell chronic lymphocytic carcinoma, non-Hodgkin's lymphoma), inflammatory and autoimmune A method of disease (eg, rheumatoid arthritis, systemic lupus erythematosus, etc.).
  • the invention provides a compound of the invention described above for use in the prevention and/or treatment of B cell associated blood cancer (eg, B cell chronic lymphocytic carcinoma, non-Hodgkin's lymphoma), inflammatory and autoimmune Use in drugs for sexual diseases such as rheumatoid arthritis, systemic lupus erythematosus, etc.
  • B cell associated blood cancer eg, B cell chronic lymphocytic carcinoma, non-Hodgkin's lymphoma
  • inflammatory and autoimmune Use in drugs for sexual diseases such as rheumatoid arthritis, systemic lupus erythematosus, etc.
  • DMF dimethylformamide
  • THF tetrahydrofuran
  • DIEA V, V-diisopropylethylamine
  • BINAP 2,2,-bisdiphenylphosphino-1,1,-binaphthyl
  • Xantphos 4,5-bisdiphenylphosphine-9,9-dimethyloxaxime.
  • Intermediate 1 (1 equivalent) is mixed with a solvent amount of phosphorus oxychloride (at least 10 equivalents), about two equivalents of dimethylaniline are added, and the reaction is stirred by heating (90-110 ° C) for several hours until the intermediate 1 disappears. After cooling, pour into ice water, precipitate a solid, filter, and dry to give Intermediate 2. Alternatively, the reaction system is concentrated, and column chromatography is carried out to obtain Intermediate 2.
  • Method 1 Mix intermediate 2 (1 eq.) with starting material 2 (1.1-1.3 eq.), add a suitable solvent (such as tetrahydrofuran, n-butanol, etc.), and heat (80-120 ° C) to stir the reaction for several hours to the intermediate. 2 disappeared. Cooling, concentrating the reaction system, and column chromatography gave Intermediate 3.
  • a suitable solvent such as tetrahydrofuran, n-butanol, etc.
  • Method 2 Buch Buchwald-Hartwig coupling. Mix intermediate 2 (1 eq.) with starting material 2 (1-1.3 eq.), add the appropriate solvent (usually toluene, dioxane, etc.), catalytic amount of palladium catalyst (usually Pd 2 (dba) 3 , Pd (OAc) 2, etc., catalytic amount of ligand (usually Xantphos, BINAP, etc.), base (Cs 2 C0 3 , NaO Bu, etc.) (1 ⁇ 1 - 1.5 equivalents), fully substituted inert gas (such as nitrogen, argon) Gas, etc., heating (70-120 ° C) to stir the reaction for several hours until the intermediate 2 disappears. The mixture was cooled, concentrated, and subjected to column chromatography to give Intermediate 3.
  • solvent usually toluene, dioxane, etc.
  • catalytic amount of palladium catalyst usually Pd 2 (dba) 3 , Pd (OAc) 2, etc.
  • Method 1 Intermediate 3 (1 eq.) And starting material 3 (1 1.3 equivalents) were mixed, and a suitable solvent (e.g. tert-butanol, tert-amyl alcohol, etc.), addition of a base (e.g., DIEA and the like, 1- 1.5 eq. ), or a catalytic amount of acid (such as acetic acid, trifluoroacetic acid, etc.), heated (80-120 ° C), or heated under microwave (80-120 ° C) to stir the reaction for several hours until the intermediate 3 disappears. Cooling, concentrating the reaction system, and column chromatography gave Intermediate 4.
  • a suitable solvent e.g. tert-butanol, tert-amyl alcohol, etc.
  • a base e.g., DIEA and the like, 1- 1.5 eq.
  • a catalytic amount of acid such as acetic acid, trifluoroacetic acid, etc.
  • Method 2 Buchwald-Hartwig coupling. In the same way as in step 2 of step 3, only raw material 2 needs to be replaced with raw material 3.
  • R 2 , R 3 , Li, L 2 , a, b, c, d, e, p, q, A and B in the reaction scheme are as described above.
  • Method 1 The substrate (1 equivalent) is dissolved in methanol and water (or a mixed solvent of dichloromethane, methanol, and water), and an oxidizing agent (such as potassium persulfate complex salt, m-chloroperoxybenzoic acid, etc.) is added ( 1 equivalent), react to LCMS at room temperature to monitor the complete disappearance of the substrate, spin the solution, prepare the liquid phase for purification, or purify the column with silica gel to obtain the product.
  • an oxidizing agent such as potassium persulfate complex salt, m-chloroperoxybenzoic acid, etc.
  • Method 2 The substrate (1 equivalent) is dissolved in methanol, and opened to reflux in air for several hours. The product is purified by spin-drying or purified by silica gel column.
  • the substrate (1 equivalent) is dissolved in methanol and water (or a mixed solvent of dichloromethane, methanol, water), and an oxidizing agent (such as potassium persulfate complex salt, m-chloroperoxybenzoic acid, etc.) (greater than 2 equivalents) ), reacting at room temperature overnight or heating for several hours, until the substrate is completely disappeared by LCMS, the solution is spin-dried, purified by liquid phase preparation, or purified by silica gel column to obtain a product.
  • an oxidizing agent such as potassium persulfate complex salt, m-chloroperoxybenzoic acid, etc.
  • R 2 , R 3 , R 5 , Li, L 2 , a, b, c, d, e, p, q, A and B in the reaction scheme are as described above.
  • the compound of the formula (I) of the present invention, a stereoisomer thereof, can be used in the form of a free form or a pharmaceutically acceptable salt thereof.
  • the compound of the formula (I) of the present invention is basic and can form an acid salt with an inorganic acid or an organic acid.
  • the compound of the formula (I) of the present invention or a pharmaceutically acceptable salt thereof may exist in the form of an optical isomer due to the presence of an asymmetric carbon atom, and therefore, the present invention also includes these optical isomers and mixtures thereof.
  • the structures described herein are also intended to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational) forms) of the structure; for example, R and about each asymmetric center S configuration, Z and E double bond isomers, and Z and E conformers.
  • single stereochemical isomers as well as mixtures of enantiomers, diastereomers and geometric isomers (or conformational isomers) of the compounds of the invention are within the scope of the invention.
  • the compound of the formula (I), a pharmaceutically acceptable salt thereof or a stereoisomer thereof of the present invention may be combined with one or more pharmaceutically acceptable carriers to constitute a pharmaceutical composition.
  • the pharmaceutical composition can be formulated into a conventional preparation for clinical use, and can be administered to a patient in need of such treatment by oral or parenteral administration. Such as tablets, granules, capsules, powders, injections, inhalants, sublingual preparations, syrups, gels, ointments, suppositories, lotions, eye drops, nasal drops, sprays, transdermal formulations Wait.
  • These preparations can be prepared by a conventional method, by adding a pharmaceutically acceptable carrier such as an excipient, a binder, a moisturizer, a disintegrating agent, a thickener or the like.
  • the compound of the formula (I) of the present invention a pharmaceutically acceptable salt thereof or a stereoisomer thereof has a preferable BTK kinase inhibitory action, and is a drug which preferably has an excellent antitumor effect and a therapeutic effect of an autoimmune disease.
  • the compound of the formula (I), a pharmaceutically acceptable salt thereof or a stereoisomer thereof thereof is used for the preparation of a B cell-associated blood cancer (for example, B cell chronic lymphocytic carcinoma, non-Hodgkin's lymphoma), and autoimmunity Sexual diseases (such as rheumatoid arthritis, systemic lupus erythematosus, etc.) play an important role.
  • a B cell-associated blood cancer for example, B cell chronic lymphocytic carcinoma, non-Hodgkin's lymphoma
  • autoimmunity Sexual diseases such as rheumatoid arthritis, systemic lupus erythematosus, etc.
  • the compound of the formula (I), a pharmaceutically acceptable salt thereof or a stereoisomer thereof of the present invention is a kinase inhibitor, particularly a Btk inhibitor.
  • These inhibitors can be used to treat one or more diseases that respond to kinase inhibition in a mammal, including response to Btk inhibition and/or B cells. A disease that inhibits proliferation.
  • Btk inhibition a disease that inhibits proliferation.
  • the present invention includes a method for treating a mammal, such as a human, having a disease responsive to inhibition of Btk activity and/or inhibiting proliferation of B cells, the method comprising: administering to a mammal having such a disease an effective amount of at least A chemical entity provided herein.
  • the effective concentration can be determined experimentally, for example, by measuring the blood concentration of the compound, or theoretically by calculating the bioavailability.
  • Other kinases that may be affected in addition to Btk include, but are not limited to, other tyrosine kinases and serine/threonine kinases.
  • kinases play a significant role in controlling the signaling pathways of essential cellular processes such as proliferation, differentiation and death (apoptosis). Abnormal kinase activity has been implicated in various diseases including a variety of cancers, autoimmune and/or inflammatory diseases, and acute inflammatory responses. The versatile role of kinases in key cell signaling pathways provides a significant opportunity to identify new drugs that target the kinase and signal transduction pathways.
  • One embodiment includes a method of treating a patient having an autoimmune and/or inflammatory disease or an acute inflammatory response in response to inhibition of Btk activity and/or B cell proliferation.
  • Autoimmune and/or inflammatory diseases that can be affected by the use of the compounds and compositions according to the invention include, but are not limited to, psoriasis, allergies, localized enteritis, irritable bowel syndrome, Sjogren's disease, tissue grafts Rejection and hyperacute rejection of transplanted organs, asthma, systemic lupus erythematosus (and associated glomerulonephritis), dermatomyositis, multiple sclerosis, scleroderma, vasculitis (ANCA-related and other blood vessels) Inflammation, autoimmune hemolytic and thrombocytopenic symptoms, Goodpas syndrome (and associated glomerulonephritis and pulmonary hemorrhage), atherosclerosis, rheumatoid arthritis, chronic idiopathic thrombocytopenia Purpura (ITP), Edison's disease, Parkinson's disease, Alzheimer's disease, diabetes, septic shock and myasthenia gravis.
  • Anti-inflammatory drugs include, but are not limited to: NSAID, non-specific and COX-2 specific cyclooxygenase inhibitors, gold compounds, dermal steroids, methotrexate, tumor necrosis factor (TNF) receptor antagonists, immunization Inhibitor and methotrexate.
  • NSAIDs include, but are not limited to, ibuprofen, flurbiprofen, naproxen and naproxen sodium, diclofenac, a combination of diclofenac sodium and misoprostol, sulindac, benzopyrene, diflunis Willow, piroxicam, indomethacin, etodolac, fenoprofen calcium, ketoprofen, nabumetone sodium, sulfasalazine, tolbutin sodium and hydroxychloroquine.
  • NSAIDs also include COX-2 Specific inhibitors such as celecoxib, valdecoxib, remiclox and/or etoricoxib.
  • the anti-inflammatory agent is a salicylate or a salt.
  • Salicylates or salts include, but are not limited to, acetylsalicylic acid or aspirin, sodium salicylate, and choline salicylate and magnesium salicylate.
  • Anti-inflammatory drugs can also be Shield Steroids.
  • the skin shield steroid may be cortisone, dexamethasone, methylprednisolone, prednisolone, prednisolone sodium phosphate, or prednisone.
  • the anti-inflammatory agent is a gold compound such as gold thioudate or auranofin.
  • the invention also includes embodiments in which the anti-inflammatory agent is a metabolic inhibitor such as a dihydrofolate reductase inhibitor such as methotrexate or a dihydroorotate dehydrogenase inhibitor such as leflunomide.
  • a further embodiment of the invention relates to wherein the at least one anti-inflammatory compound is an anti-monoclonal antibody (such as eculizumab or pegizumab), a TNF antagonist such as entanercept or infliximab
  • the infliximab is an anti-TNFa monoclonal antibody.
  • a further embodiment of the invention relates to wherein at least one active agent is an immunosuppressive compound such as selected from the group consisting of methotrexate, leflunomide, cyclosporine, tacrolimus, azathioprine and mycophenolate
  • an immunosuppressive compound such as selected from the group consisting of methotrexate, leflunomide, cyclosporine, tacrolimus, azathioprine and mycophenolate
  • B-B and B-precursor expression of Btk have been implicated in the pathology of B-package malignancy, B cell malignancy including but not limited to B-cell lymphoma, lymphoma (including Hodgkin and non-Hodgkin's lymphoma) ), hair cell lymphoma, multiple myeloma, chronic and acute myeloid leukemia and chronic and acute lymphocytic leukemia.
  • Btk has been shown to be an inhibitor of Fas/APO-1 (CD-95) death-inducing signaling complex (DISC) in B-lineage lymphoid cells.
  • the fate of leukemia/lymphoma cells may lie in the balance between the reverse pre-apoptotic effect of caspase-activated caspase and the upstream anti-apoptotic regulatory mechanism including Btk and/or its substrate (Vassilev et al. , J. Biol. Chem. 1998, 274, 1646-1656).
  • Btk inhibitors can be used as chemical sensitizers, and thus can be used in combination with other chemotherapeutic drugs, particularly drugs which induce apoptosis, such as antitumor agents, immunosuppressive agents and the like.
  • chemotherapeutic agents that can be used in combination with chemical sensitizers include, but are not limited to, topoisomerase I inhibitors (such as camptothecin or topotecan), topoisomerase II inhibitors (such as daunorubicin) And etoposide:), alkylating agents (such as cyclophosphamide, melphalan and BCNU), tubulin-directed agents (such as Taxol and Vinblastine) and biological agents (such as antibodies such as anti-CD20 antibody, IDEC8, Immunotoxins and cytokines).
  • Btk activity has been associated with some leukemias that express the bcr-abl fusion gene resulting from partial translocation of chromosomes 9 and 22.
  • Btk is shielded by bcr-abl kinase, which triggers a downstream survival signal that prevents apoptosis in bcr- abl cells (N. Feldhahn et al., J. Exp. Med. 2005, 201(11), 1837-1852 ).
  • the compound of the formula (I) of the present invention or a pharmaceutically acceptable salt thereof has a good BTK kinase inhibitory action, has high safety, a large safety window and a small side effect;
  • control compound CC-292, was prepared in accordance with WO2009158571A1.
  • the compound of the present invention its chemical name, structural formula and preparation method are shown in the preparation examples of the respective compounds.
  • ATP adenosine triphosphate
  • BTK Bruton's tyrosine kinase
  • mg mg
  • mL ml
  • g microgram
  • microliter
  • mM millimoles per liter
  • EDTA ethylenediaminetetraacetic acid
  • DMSO dimethyl Ketosulfone.
  • HTRFR KinEASETM -TK purchased from Cisbio, lot number 62TK0PEB; BTK: purchased from Carna, Cat.No.08-080; ATP: purchased from Sigma, Cat. No. A7699, CAS
  • MgCl 2 purchased from Sigma, CAS No. 7786-30-3, Lot. No. 101M8701V; DMSO: purchased from Sigma, CAS No. 67-68-5, Lot. No. STBC0365V ;
  • Test reagent preparation available from Thermo, Cat. No. 249944, Lot. No. 1057825; 384-well plate: available from Greiner, Cat. No. 784075, Lot. No. ⁇ 1112 ⁇ 6 ⁇ . 2. Test reagent preparation
  • test sample was separately detected at 615 nm by a microplate reader.
  • the inhibitory activity of the compounds of the present invention against BTK kinase was significantly stronger than that of the control compound CC-292.
  • the compound of the present invention its chemical name and structural formula and preparation method are shown in the preparation examples.
  • mice Male, 6-8 week Balb/c mice.
  • CTG CellTiter-Glo (Cat. No. G7572, Promega), storing CTG buffer and CTG substrate at -20.
  • C It is recommended to prepare CTG reagents in the following ways:
  • CTG buffer before use and equilibrate to room temperature.
  • CTG buffer can be used. Melt well before use and store at room temperature for more than 48 hours.
  • the lyophilized CTG substrate was equilibrated to room temperature and 100 ml of buffer was taken into the amber bottle containing the substrate to obtain the CTG reagent. Gently mix until a uniform solution is obtained.
  • the substrate should be completely melted in one minute, dispensed and stored in a - 20 ° C refrigerator for a long period of time.
  • test compound gradient dilution solution First, 10 mM of the test compound stock solution was diluted 3 times in DMSO with a total of 10 concentrations. Then add ⁇ DMSO diluted compound to 90 ⁇ compound dilution buffer, and then take lO L
  • the 10% DMSO diluted compound was added to 90 ⁇ of the compound dilution buffer at a maximum concentration of 10 ⁇ M and a DMSO concentration of 0.1% for a total of 10 concentration gradients.
  • mice * The spleens of Balb/c mice were smashed, mashed in MACS buffer, and filtered through a 40 ⁇ nylon cell sieve to obtain a single cell suspension.
  • the obtained cell suspension was centrifuged at 400 g for five minutes, the supernatant was removed, and 1 ml of the red blood cell lysate was added at room temperature and the cell pellet was gently resuspended. Two minutes later, pre-chilled MACS buffer was added. The cell suspension was filtered through a 40 ⁇ cell sieve into a new centrifuge tube.
  • a biotinylated antibody mixture of ⁇ was added to every 10 7 cells. After mixing, incubate on ice for 20 minutes, add 30 ⁇ l of MACS buffer and 20 ⁇ l of avidin-containing magnetic beads per 10 7 cells and incubate on ice for 20 minutes. After centrifugation, the cells were resuspended in 500 ⁇ l of MACS buffer. The pre-cooled MACS sorting column was placed in a MACS sorter and the cell suspension was applied to a MACS sorting column. The cells under the flow of unbound antibody are collected. • Detection of cells before and after sorting by flow cytometry using PE anti-biotin antibody and CD45R (B220) antibody detection.
  • the data obtained will be analyzed using Excel 2007 and GraphPad Prism 5.0 software in order to calculate IC 5 .
  • the nonlinear S-curve regression will be used to fit the data to obtain a dose-response curve, and the GraphPad Prism 5.0 software will automatically give the IC 50 value.
  • V sample / V2 dissolved 3 ⁇ 4 pairs, X 100%
  • V sample is the reading of the compound treated well
  • V2 » 3 ⁇ 4 vs. 3 ⁇ 4 is the average of the solvent control well (V2) readings.
  • the compound of the present invention (Compound 1-106) has an IC 5 Q ⁇ 0.5 ⁇ for the inhibitory activity of Balb/c mouse B cells in vitro.
  • the IC 5 Q of some compounds is as follows: In vitro cytological inhibitory activity of some compounds of the invention against Balb/c mouse B cells
  • an ELISA protocol is used that evaluates compounds at different concentrations in spleen cells using a biotinylated probe compound that binds only to BTK that is not occupied by the compound. For the occupancy rate of BTK enzyme, calculate the %81 ⁇ occupancy rate (BTK Occupancy).
  • Mouse anti-BTK antibody (Becton Dickinson); goat anti-mouse HRP antibody (Becton Dickinson); cell lysate (Cell Signaling); Bruton tyrosine kinase (BTK) (Carna); streptavidin coated 96-well plate (Thermo); rat lymphocyte isolation kit (LTS 1083PK, Tianjin Haoyang Biological Products Technology Co., Ltd.); microplate reader (victor4, PE); centrifuge (5804R, Eppendorf); microscope ( CX31RTSF, Olympus); MACS sorter (MACS).
  • BTK Bruton tyrosine kinase
  • Probe compound solution (Probe): Weigh 1 mg of sample compound and prepare a concentration of 1 mM. Stock solution, diluted with sample diluent; Sample diluents: PBS containing 1% bovine serum albumin and 0.1% Tween-20; Washing solution: 0.05% Tween-20 PBS.
  • the cell concentration was diluted to 3 x 107 Cells/ml, 90 ⁇ l/well with PBS. Add compound ⁇ /well and incubate for 1 h at 37 °C. Centrifuge at 20 °C, 400g for 20min, discard the supernatant.
  • the protease inhibitor PMSF was added to the cell lysis buffer (note that PMSF was added before the lysate was used).
  • the lysate was added to each tube-enriched cell and mixed. According to the lysate instructions, the ice was lysed for 5 min and centrifuged at 14,000 g for 10 min. Add the supernatant ⁇ to the 96-well plate.
  • the OD value at 450 nm was detected. According to the OD value, use the microplate reader
  • the A 4 parameter logic curve calculates the amount of BTK in each sample.
  • the frozen plasma was pre-incubated in a 37 ° C constant temperature water bath and thawed for use.
  • the stability of the compound in plasma was evaluated by the retention percentage of the compound after incubation at each time point, and the concentration of the sample was expressed by the ratio of the peak area of the test compound to the internal standard peak area. Calculated as follows:
  • the concentration of the compound is determined as the final solution at the initial incubation.
  • the compound 23 of the present invention is more stable in plasma than the control drug CC-292.
  • the V patch clamp method detects the inhibition of the hERG potassium channel by the compound of the present invention.
  • the compound of the present invention (Compound 1-106): Its chemical name, structural formula and preparation method are shown in the preparation examples of the respective compounds.
  • Control compound CC-292, synthesized according to patent WO2009158571A1.
  • experimental method
  • Extracellular fluid mM: 7V ⁇ 2 hydroxyethylpiperazine-2-ethylethanesulfonic acid (HEPES) 10.
  • HEPES hydroxyethylpiperazine-2-ethylethanesulfonic acid
  • In-electrode solution (in mM): KC1 120, OH 31.25, CaC3 ⁇ 4 5.374, MgCI 2 L75, ethylene glycol-bis( ⁇ -aminoethyl ether) ⁇ ', ⁇ '"-tetraacetic acid (EGTA) 10, HEPES 10, Na. 2 -ATP 4, pH was adjusted to 7.2 with IN potassium hydroxide; osmotic pressure was adjusted to 280 290 mOsm; after filtration - 20 ° C storage.
  • the hERG current was recorded using whole cell patch clamp technique.
  • the cell suspension was applied to a 35 mm culture and placed on an inverted microscope stage. After the cells were attached, the cells were perfused with extracellular fluid at a flow rate of 12 mL/min.
  • the glass microelectrode is drawn in two steps by a microelectrode drawing device, and its water resistance is 2 - 5 ⁇ .
  • the clamping potential was maintained at -80 mV.
  • the voltage is stimulated to depolarize to +60 mV and then repolarized to -50 mV to induce the hERG tail current. All records are performed after the current has stabilized. Extracellular perfusion administration starts from a low concentration, each concentration is 5 ⁇ until the current is stable, and the next concentration is given.
  • Stable cell line CHO hERG was purchased from AVIV A.
  • the minimum sealing resistance is not less than 1GQ
  • the hERG current is not less than 0.4nA.
  • the test coverage includes the following aspects:
  • the hERG current was recorded on the CHO-K1 cell line stably expressing the hERG channel by manual patch clamp technique; the inhibition rate of each concentration was calculated from the hERG tail current; each compound was tested 5 for each concentration test 2 cells; one positive control drug.
  • n H slope; IC 5 . : The maximum half of the test substance is inhibited.
  • the positive control drug Amitriptyline is one of the most widely used drugs for blocking hERG currents, IC 5 for inhibition of hERG current in this study. For 3.15 ⁇ , this result is consistent with the results reported in the literature. This shows that the results of this trial are credible.
  • test compound Compound 1-106
  • SD rats To observe the toxicity of the test compound (Compound 1-106) administered to SD rats for 14 days, to understand the toxicity of the test substance and its sputum organs, to determine the dose of non-toxic reaction under the test conditions; Whether the test substance has delayed toxicity and whether the toxic reaction is reversible.
  • the main experimental group was the vehicle control group, the test substance (Compound 1-106), the low dose group, the middle dose group, the high dose group, and 10 animals each, male and female.
  • the middle dose group and the vehicle control group were additionally restored. There are 10 animals each, half male and half female.
  • the compound of the present invention (Compound 1-106) has good tolerance and high safety.
  • 5-Bromouracil (10.00 g, 52.4 mmol) was added to the above aqueous solution, and refluxed for 16 h. The solution was cooled to -5 ° C, and the pH was adjusted to 7 with concentrated hydrochloric acid to precipitate a solid, which was filtered and dried to give a pale yellow solid, 3.50 g, yield 33%
  • V-(2-chloroethyl)morpholine hydrochloride (9.3 g, 0.05 mol), thiourea (3.81 g, 0.05 mol) in 95% ethanol (50 mL), heated to reflux for 24 hours, cooled Concentrated to give a white solid which was taken directly to next.
  • tert-Butyl 3-(2-chloro-5-(methylthio)pyrimidin-4-ylamino)phenylcarbamate (0.25 g, 0.68 mmol) was dissolved in 5 mL of tert-amyl alcohol, then acetic acid 2 was added 6-Methoxypyridine-3-amine (0.088 g, 0.71 mmol) was added dropwise, and the mixture was stirred at 85 ° C for 1 h. The reaction was cooled to room temperature, then EtOAc (EtOAc) was evaporated.
  • N"-(3-Aminophenyl)-W 2 -(4-(2-methoxyethoxy)phenyl)-5-(methylsulfinyl)pyrimidine-2,4-diamine (1.8 g, 4.35 mmol) was dissolved in 20 mL of dry tetrahydrofuran, 4 mL of DIPEA was added to bring the pH to 10, then 4-bromobut-2-enoyl chloride (crude, about 4.35 mmol) was dissolved in 10 mL of dichloromethane. Slowly drip into the solution, and the reaction was stirred at 0 ° C for 2 h and placed for later use.
  • Methyl 4-bromocrotonate ( 1.775 g, 9.93 mmol) was dissolved in 20 mL of dichloromethane, then pyrrolidine ( 1.408 g, 19.86 mmol) was added to the solution at 0 ° C, the reaction was stirred for 2 h, and dried. , used directly in the next step.
  • Methyl 4-(pyrrolidin-1-yl)but-2-enoate ( 1.67 g, 9.9 mmol) was dissolved in 20 mL of methanol, then NaOH (0.792 g, 19.8 mmol) was added and the resulting solution was at 0°.
  • Stir for 2 h at C then add 50 mL of ethyl acetate and 50 mL of water, extract, and dilute the aqueous phase with 2 N Hydrochloric acid was adjusted to pH 6.0, then ethyl acetate (100 mL) was added and extracted three times. The obtained organic phase was dried and dried to give a yield of 0.52 g of pale red oil.
  • the crude product in the previous step was dissolved in 15 mL of methanol and 5 mL of water, then slowly added potassium hydride (oxone) (0.301 g, 0.49 mmol), reacted at room temperature for 25 min, the solution was spun dry, and added to 30 The organic layer was dried over anhydrous sodium sulfate (MgSO4), filtered, filtered, filtered,jjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjjj
  • 0.21 g of the previous step was dissolved in 20 mL of methanol and 2 mL of water, then slowly added potassium hydride (oxone) (0.301 g, 0.49 mmol) dissolved in 2 mL of water, and reacted at room temperature for 25 min. After quenching with 10 mL of a saturated aqueous solution of sodium thiosulfate, 50 mL of dichloromethane and 30 mL of water were added, and the organic phase was dried over anhydrous sodium sulfate, filtered, filtered and evaporated to dryness. 20:1) 0.096 g of a white solid was obtained.
  • 4-Bromocrotonic acid (0.106 g, 0.64 mmol) was dissolved in 10 mL of dichloromethane, and 2 drops of DMF were added, then 2 mL of oxalyl chloride was added at 0 ° C, stirred for 2 h, and dried, and was taken.
  • N 4 -(3-Aminophenyl)-TV 2 -(1-methyl-p-pyrazol-4-yl)-5-(methylthio)pyrimidine-2,4-diamine (0.21 g, 0.64 Ment) was dissolved in 20 mL of dry tetrahydrofuran, DIPEA (0.248 g, 1.92 mmol) was added to bring the pH to 10, then 4-bromo crotonyl chloride was dissolved in 5 mL of dichloromethane and slowly added dropwise to the system. The reaction was stirred at 0 ° C for 2 h and placed for later use.
  • V-(3-(2-(1-methyl-p-pyrazol-4-ylamino)-5-(methylthio)pyrimidin-4-ylamino)phenyl)-4-(pyrrolidin-1) -yl)but-2-enamide (0.10 g, 0.215 mmol) dissolved in 15 mL of methanol, then slowly added potassium peroxodisulfate complex (oxone) (0.146 g, 0.237 mmol) dissolved in 2 mL of water.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
PCT/CN2013/083429 2012-09-12 2013-09-12 Dérivé de noyau hétéroaromatique contenant de l'azote utile comme inhibiteur de la tyrosine kinase Ceased WO2014040555A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201380041846.3A CN104583195B (zh) 2012-09-12 2013-09-12 作为酪氨酸激酶抑制剂的含氮杂芳环衍生物

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CN201210337565.X 2012-09-12
CN201210337565 2012-09-12
CN201310015201.4 2013-01-16
CN201310015201 2013-01-16
CN201310187510.X 2013-05-20
CN201310187510 2013-05-20

Publications (1)

Publication Number Publication Date
WO2014040555A1 true WO2014040555A1 (fr) 2014-03-20

Family

ID=50277638

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2013/083429 Ceased WO2014040555A1 (fr) 2012-09-12 2013-09-12 Dérivé de noyau hétéroaromatique contenant de l'azote utile comme inhibiteur de la tyrosine kinase

Country Status (2)

Country Link
CN (1) CN104583195B (fr)
WO (1) WO2014040555A1 (fr)

Cited By (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015025197A1 (fr) 2013-08-22 2015-02-26 Jubilant Biosys Limited Composés de pyrimidine substituée, compositions et applications médicinales correspondantes
WO2015151006A1 (fr) 2014-03-29 2015-10-08 Lupin Limited Composés de purine substitués en tant qu'inhibiteurs de btk
WO2015170266A1 (fr) 2014-05-07 2015-11-12 Lupin Limited Composés pyrimidine substitués en tant qu'inhibiteurs de la tyrosine kinase de bruton (btk)
US20160102076A1 (en) * 2014-10-13 2016-04-14 Yuhan Corporation Compounds and compositions for modulating egfr mutant kinase activities
US9394281B2 (en) 2014-03-28 2016-07-19 Calitor Sciences, Llc Substituted heteroaryl compounds and methods of use
CN106349224A (zh) * 2016-08-03 2017-01-25 山东大学 一种含有4‑氨基‑(1h)‑吡唑结构的jak激酶抑制剂及其制备方法和应用
CN106608868A (zh) * 2015-10-21 2017-05-03 复旦大学 5-(硫醚基)嘧啶类化合物及其药用组合物和应用
WO2017111076A1 (fr) * 2015-12-24 2017-06-29 協和発酵キリン株式会社 COMPOSÉ AMIDE α, β INSATURÉ
US9938257B2 (en) 2015-09-11 2018-04-10 Calitor Sciences, Llc Substituted heteroaryl compounds and methods of use
CN108329274A (zh) * 2017-01-13 2018-07-27 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
EP3354647A4 (fr) * 2015-09-25 2019-04-03 Zhejiang Bossan Pharmaceutical Co. Ltd. Inhibiteur de l'egfr kinase, son procédé de préparation et son utilisation
US10457669B2 (en) 2015-10-21 2019-10-29 Otsuka Pharmaceutical Co., Ltd. Benzolactam compounds as protein kinase inhibitors
CN110770211A (zh) * 2017-06-23 2020-02-07 协和麒麟株式会社 α,β不饱和酰胺化合物
US10807951B2 (en) 2017-10-13 2020-10-20 The Regents Of The University Of California mTORC1 modulators
US11066404B2 (en) 2018-10-11 2021-07-20 Incyte Corporation Dihydropyrido[2,3-d]pyrimidinone compounds as CDK2 inhibitors
US11142518B2 (en) 2017-04-20 2021-10-12 Otsuka Pharmaceutical Co., Ltd. 6-pyrimidin-isoindole derivative as ERK1/2 inhibitor
WO2022140246A1 (fr) 2020-12-21 2022-06-30 Hangzhou Jijing Pharmaceutical Technology Limited Procédés et composés destinés à l'autophagie ciblée
CN114685593A (zh) * 2020-12-31 2022-07-01 鲁南制药集团股份有限公司 一种氟维司群制备方法及其中间体
US11384083B2 (en) 2019-02-15 2022-07-12 Incyte Corporation Substituted spiro[cyclopropane-1,5′-pyrrolo[2,3-d]pyrimidin]-6′(7′h)-ones as CDK2 inhibitors
US11427567B2 (en) 2019-08-14 2022-08-30 Incyte Corporation Imidazolyl pyrimidinylamine compounds as CDK2 inhibitors
US11440914B2 (en) 2019-05-01 2022-09-13 Incyte Corporation Tricyclic amine compounds as CDK2 inhibitors
US11447494B2 (en) 2019-05-01 2022-09-20 Incyte Corporation Tricyclic amine compounds as CDK2 inhibitors
US11472791B2 (en) 2019-03-05 2022-10-18 Incyte Corporation Pyrazolyl pyrimidinylamine compounds as CDK2 inhibitors
CN116351103A (zh) * 2023-03-31 2023-06-30 百林科(兰州)新材料有限公司 一种层析介质及其制备方法和应用
US11691981B2 (en) 2019-06-25 2023-07-04 InventisBio Co., Ltd. Heterocyclic compounds, preparation methods therefor, and methods of uses thereof
US11851426B2 (en) 2019-10-11 2023-12-26 Incyte Corporation Bicyclic amines as CDK2 inhibitors
US11919904B2 (en) 2019-03-29 2024-03-05 Incyte Corporation Sulfonylamide compounds as CDK2 inhibitors
US11976073B2 (en) 2021-12-10 2024-05-07 Incyte Corporation Bicyclic amines as CDK2 inhibitors
US11981671B2 (en) 2021-06-21 2024-05-14 Incyte Corporation Bicyclic pyrazolyl amines as CDK2 inhibitors
WO2025061148A1 (fr) * 2023-09-22 2025-03-27 江苏恒瑞医药股份有限公司 Composé hétéroaryle, son procédé de préparation et son utilisation en médecine
US12441707B2 (en) 2019-12-30 2025-10-14 Tyra Biosciences, Inc. Indazole compounds

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101902911A (zh) * 2007-10-19 2010-12-01 阿维拉制药公司 杂芳基化合物和其用途
CN102083800A (zh) * 2008-06-27 2011-06-01 阿维拉制药公司 杂芳基化合物和其用途
WO2011153553A2 (fr) * 2010-06-04 2011-12-08 The Regents Of The University Of California Méthodes et compositions pour l'inhibition de kinases

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101902911A (zh) * 2007-10-19 2010-12-01 阿维拉制药公司 杂芳基化合物和其用途
CN102083800A (zh) * 2008-06-27 2011-06-01 阿维拉制药公司 杂芳基化合物和其用途
WO2011153553A2 (fr) * 2010-06-04 2011-12-08 The Regents Of The University Of California Méthodes et compositions pour l'inhibition de kinases

Cited By (53)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3036231A4 (fr) * 2013-08-22 2017-01-18 Jubilant Biosys Ltd. Composés de pyrimidine substituée, compositions et applications médicinales correspondantes
WO2015025197A1 (fr) 2013-08-22 2015-02-26 Jubilant Biosys Limited Composés de pyrimidine substituée, compositions et applications médicinales correspondantes
US9394281B2 (en) 2014-03-28 2016-07-19 Calitor Sciences, Llc Substituted heteroaryl compounds and methods of use
US9399637B2 (en) 2014-03-28 2016-07-26 Calitor Sciences, Llc Substituted heteroaryl compounds and methods of use
US9403801B2 (en) 2014-03-28 2016-08-02 Calitor Sciences, Llc Substituted heteroaryl compounds and methods of use
WO2015151006A1 (fr) 2014-03-29 2015-10-08 Lupin Limited Composés de purine substitués en tant qu'inhibiteurs de btk
WO2015170266A1 (fr) 2014-05-07 2015-11-12 Lupin Limited Composés pyrimidine substitués en tant qu'inhibiteurs de la tyrosine kinase de bruton (btk)
USRE50528E1 (en) * 2014-10-13 2025-08-12 Yuhan Corporation Compounds and compositions for modulating EGFR mutant kinase activities
US20160102076A1 (en) * 2014-10-13 2016-04-14 Yuhan Corporation Compounds and compositions for modulating egfr mutant kinase activities
US9593098B2 (en) * 2014-10-13 2017-03-14 Yuhan Corporation Compounds and compositions for modulating EGFR mutant kinase activities
US9938257B2 (en) 2015-09-11 2018-04-10 Calitor Sciences, Llc Substituted heteroaryl compounds and methods of use
US10266521B2 (en) 2015-09-11 2019-04-23 Calitor Sciences, Llc Substituted heteroaryl compounds and methods of use
EP3354647A4 (fr) * 2015-09-25 2019-04-03 Zhejiang Bossan Pharmaceutical Co. Ltd. Inhibiteur de l'egfr kinase, son procédé de préparation et son utilisation
AU2016327857B2 (en) * 2015-09-25 2019-10-03 Zhejiang Bossan Pharmaceutical Co. Ltd. EGFR kinase inhibitor and preparation method and use thereof
US10457669B2 (en) 2015-10-21 2019-10-29 Otsuka Pharmaceutical Co., Ltd. Benzolactam compounds as protein kinase inhibitors
US11001575B1 (en) 2015-10-21 2021-05-11 Otsuka Pharmaceutical Co., Ltd. Benzolactam compounds as protein kinase inhibitors
US11939321B2 (en) 2015-10-21 2024-03-26 Otsuka Pharmaceutical Co., Ltd. Benzolactam compounds as protein kinase inhibitors
CN106608868A (zh) * 2015-10-21 2017-05-03 复旦大学 5-(硫醚基)嘧啶类化合物及其药用组合物和应用
CN108430972A (zh) * 2015-12-24 2018-08-21 协和发酵麒麟株式会社 α,β不饱和酰胺化合物
WO2017111076A1 (fr) * 2015-12-24 2017-06-29 協和発酵キリン株式会社 COMPOSÉ AMIDE α, β INSATURÉ
US10787428B2 (en) 2015-12-24 2020-09-29 Kyowa Kirin Co., Ltd. α,β-unsaturated amide compound
US11332455B2 (en) 2015-12-24 2022-05-17 Kyowa Kirin Co., Ltd. α,β-unsaturated amide compound
CN108430972B (zh) * 2015-12-24 2022-12-27 协和麒麟株式会社 α,β不饱和酰胺化合物
CN106349224A (zh) * 2016-08-03 2017-01-25 山东大学 一种含有4‑氨基‑(1h)‑吡唑结构的jak激酶抑制剂及其制备方法和应用
CN108329274A (zh) * 2017-01-13 2018-07-27 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
CN108329274B (zh) * 2017-01-13 2021-09-03 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
US11142518B2 (en) 2017-04-20 2021-10-12 Otsuka Pharmaceutical Co., Ltd. 6-pyrimidin-isoindole derivative as ERK1/2 inhibitor
US12030873B2 (en) 2017-04-20 2024-07-09 Otsuka Pharmaceutical Co., Ltd. 6-pyrimidin-isoindole derivative as ERK1/2 inhibitor
CN110770211B (zh) * 2017-06-23 2023-09-19 协和麒麟株式会社 α,β不饱和酰胺化合物
CN110770211A (zh) * 2017-06-23 2020-02-07 协和麒麟株式会社 α,β不饱和酰胺化合物
US10807951B2 (en) 2017-10-13 2020-10-20 The Regents Of The University Of California mTORC1 modulators
US11066404B2 (en) 2018-10-11 2021-07-20 Incyte Corporation Dihydropyrido[2,3-d]pyrimidinone compounds as CDK2 inhibitors
US11866432B2 (en) 2018-10-11 2024-01-09 Incyte Corporation Dihydropyrido[2,3-d]pyrimidinone compounds as CDK2 inhibitors
US11384083B2 (en) 2019-02-15 2022-07-12 Incyte Corporation Substituted spiro[cyclopropane-1,5′-pyrrolo[2,3-d]pyrimidin]-6′(7′h)-ones as CDK2 inhibitors
US11472791B2 (en) 2019-03-05 2022-10-18 Incyte Corporation Pyrazolyl pyrimidinylamine compounds as CDK2 inhibitors
US11919904B2 (en) 2019-03-29 2024-03-05 Incyte Corporation Sulfonylamide compounds as CDK2 inhibitors
US11440914B2 (en) 2019-05-01 2022-09-13 Incyte Corporation Tricyclic amine compounds as CDK2 inhibitors
US11447494B2 (en) 2019-05-01 2022-09-20 Incyte Corporation Tricyclic amine compounds as CDK2 inhibitors
US12173010B2 (en) 2019-06-25 2024-12-24 InventisBio Co., Ltd. Heterocyclic compounds, preparation methods therefor, and methods of uses thereof
EP3990445A4 (fr) * 2019-06-25 2023-07-05 InventisBio Co., Ltd. Composés hétérocycliques, leurs procédés de préparation et leurs méthodes d'utilisation
US11691981B2 (en) 2019-06-25 2023-07-04 InventisBio Co., Ltd. Heterocyclic compounds, preparation methods therefor, and methods of uses thereof
US12312331B2 (en) 2019-08-14 2025-05-27 Incyte Corporation Imidazolyl pyrimidinylamine compounds as CDK2 inhibitors
US11427567B2 (en) 2019-08-14 2022-08-30 Incyte Corporation Imidazolyl pyrimidinylamine compounds as CDK2 inhibitors
US11851426B2 (en) 2019-10-11 2023-12-26 Incyte Corporation Bicyclic amines as CDK2 inhibitors
US12466828B2 (en) 2019-10-11 2025-11-11 Incyte Corporation Bicyclic amines as CDK2 inhibitors
US12441707B2 (en) 2019-12-30 2025-10-14 Tyra Biosciences, Inc. Indazole compounds
WO2022140246A1 (fr) 2020-12-21 2022-06-30 Hangzhou Jijing Pharmaceutical Technology Limited Procédés et composés destinés à l'autophagie ciblée
CN114685593B (zh) * 2020-12-31 2024-01-23 鲁南制药集团股份有限公司 一种氟维司群制备方法及其中间体
CN114685593A (zh) * 2020-12-31 2022-07-01 鲁南制药集团股份有限公司 一种氟维司群制备方法及其中间体
US11981671B2 (en) 2021-06-21 2024-05-14 Incyte Corporation Bicyclic pyrazolyl amines as CDK2 inhibitors
US11976073B2 (en) 2021-12-10 2024-05-07 Incyte Corporation Bicyclic amines as CDK2 inhibitors
CN116351103A (zh) * 2023-03-31 2023-06-30 百林科(兰州)新材料有限公司 一种层析介质及其制备方法和应用
WO2025061148A1 (fr) * 2023-09-22 2025-03-27 江苏恒瑞医药股份有限公司 Composé hétéroaryle, son procédé de préparation et son utilisation en médecine

Also Published As

Publication number Publication date
CN104583195B (zh) 2018-08-17
CN104583195A (zh) 2015-04-29

Similar Documents

Publication Publication Date Title
WO2014040555A1 (fr) Dérivé de noyau hétéroaromatique contenant de l'azote utile comme inhibiteur de la tyrosine kinase
CA3087089C (fr) Composes cycliques fondus
AU2019249721B2 (en) Heteroaryl compounds as type II IRAK inhibitors and uses hereof
ES2415863T3 (es) Heterociclos sustituidos como inhibidores de Janus Quinasas
CN107922431B (zh) Hpk1抑制剂及其使用方法
TWI642667B (zh) 吡啶並嘧啶類衍生物、其製備方法及其在醫藥上的應用
KR102085121B1 (ko) 이미다조[1,2-b]피리다진계 화합물, 그를 포함하는 조성물 및 그의 용도
CA3086867A1 (fr) Composes de cycles fusionnes comme inhibiteurs de proteine ras
JP6507234B2 (ja) ブルトンチロシンキナーゼ(btk)によって介入される障害の処置における使用のためのピラゾールカルボキサミド化合物
JP6920202B2 (ja) Irak阻害剤としてのピリダジノン大環状化合物及びその使用
WO2014089913A1 (fr) Composé bicyclique fonctionnant en tant qu'inhibiteur de tyrosine kinase
JP2020522520A (ja) Ire1小分子阻害薬
WO2018045956A1 (fr) Composé de benzimidazole inhibiteur de kinase, son procédé de préparation et son application
EA024109B1 (ru) Ингибиторы протеинкиназ
JP2017536369A (ja) Irakインヒビターとしてのヘテロアリール化合物及びその使用
UA74850C2 (en) Amide derivatives as nmda receptor antagonists
WO2020125513A1 (fr) Composé macrocyclique servant d'inhibiteur de cdk, son procédé de préparation et son utilisation en médecine
JP2018535985A (ja) ヤヌスキナーゼ、並びにその組成物及びその使用
JP2023509155A (ja) ビフェニル系誘導体阻害剤、その調製方法及び使用
EP3965803A1 (fr) Inhibiteurs de kinase d'adhésion focale à petites molécules (fak)
JP2021520418A (ja) 高活性csf1r阻害薬化合物
KR20190035769A (ko) 브루톤 타이로신 카이나제 저해제의 석신에이트 형태 및 조성물
WO2010096395A1 (fr) Amides utilisés en tant qu'inhibiteurs des kinases
JP2021510693A (ja) Jak阻害剤としてのピラゾロピリミジン化合物
JP2019526593A (ja) 8−(アゼチジン−1−イル)−[1,2,4]トリアゾロ[1,5−a]ピリジニル化合物、組成物及びその使用方法

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: 13837604

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 13837604

Country of ref document: EP

Kind code of ref document: A1