WO2018133151A1 - 用作布鲁顿酪氨酸激酶抑制剂的化合物及其制备方法和应用 - Google Patents

用作布鲁顿酪氨酸激酶抑制剂的化合物及其制备方法和应用 Download PDF

Info

Publication number
WO2018133151A1
WO2018133151A1 PCT/CN2017/074108 CN2017074108W WO2018133151A1 WO 2018133151 A1 WO2018133151 A1 WO 2018133151A1 CN 2017074108 W CN2017074108 W CN 2017074108W WO 2018133151 A1 WO2018133151 A1 WO 2018133151A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
substituted
unsubstituted
compound
formula
Prior art date
Application number
PCT/CN2017/074108
Other languages
English (en)
French (fr)
Inventor
李英富
黄浩喜
刘冠锋
陈垌珲
任俊峰
苏忠海
Original Assignee
成都倍特药业有限公司
成都海博锐药业有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 成都倍特药业有限公司, 成都海博锐药业有限公司 filed Critical 成都倍特药业有限公司
Priority to EP17893058.2A priority Critical patent/EP3572414A4/en
Priority to US16/468,658 priority patent/US10793576B2/en
Priority to JP2019531745A priority patent/JP6884863B2/ja
Publication of WO2018133151A1 publication Critical patent/WO2018133151A1/zh

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Definitions

  • the invention relates to the technical field of medicinal chemistry, in particular to a compound used as a Bruton tyrosine kinase inhibitor and a preparation method and application thereof.
  • BTK is a member of the non-receptor protein tyrosine kinase family.
  • BCR B cell receptor
  • Ibrutinib is in the treatment of chronic lymphocytic leukemia (CLL), mantle cell lymphoma (MCL) and Macroglobulinemia has improved the survival of certain malignancies and avoided many side effects caused by traditional chemotherapy.
  • CLL chronic lymphocytic leukemia
  • MCL mantle cell lymphoma
  • Macroglobulinemia has improved the survival of certain malignancies and avoided many side effects caused by traditional chemotherapy.
  • CLL chronic lymphocytic leukemia
  • MCL mantle cell lymphoma
  • Macroglobulinemia has improved the survival of certain malignancies and avoided many side effects caused by traditional chemotherapy.
  • CLL chronic lymphocytic leukemia
  • MCL mantle cell lymphoma
  • Macroglobulinemia has improved the survival of certain malignancies and avoided many side effects caused by traditional chemotherapy.
  • CLL chronic lymphocytic leukemia
  • MCL mantle cell lymphoma
  • Macroglobulinemia has improved
  • ibrutinib the most common side effects of ibrutinib are bleeding, infection, cytopenia, atrial fibrillation, second primary malignancy, tumor lysis syndrome, embryo or fetal toxicity.
  • the above bleeding events (such as subdural hematoma, gastrointestinal bleeding, hematuria and post-program bleeding) occurred in 6%, and the bleeding mechanism is still unclear; the probability of infection of grade 3 or above is 14% to 26%; Grade 3 or 4 cytopenia, including neutropenia (19% to 29%), thrombocytopenia (5% to 17%), and anemia (0 to 9%); incidence of atrial fibrillation and atrial flutter 6% to 9%, especially in patients with heart disease, acute infections, patients with a history of atrial fibrillation; the incidence of other malignant tumors is 5% to 14%, including non-skin cancer (1% to 3%), the most common It is a non-melanoma skin cancer (4% to 11%); patients treated with ibrutin
  • the technical problem to be solved by the present invention is to provide a compound for use as a Bruton tyrosine kinase inhibitor, a preparation method and application thereof, and a compound prepared by comparison with ibrutinib which is currently on the market.
  • Some Bruton's tyrosine kinase inhibitor compounds disclosed in this patent have better activity and higher cardiac safety, especially in terms of pharmacokinetics, after oral administration in animals, regardless of blood concentration, exposure, half-life Parameters such as oral bioavailability have obvious advantages. Therefore, the Bruton tyrosine kinase inhibitor compound disclosed in this patent has broad clinical application prospects.
  • Substituted as used in the present invention means that one or more hydrogen atoms in the given structure are replaced by a substituent.
  • the substituents may be the same or different.
  • the above substituents may be optionally substituted by one or more of the same or different secondary substituents.
  • the secondary substituents described therein may Yes, but not limited to, fluorine, chlorine, bromine, iodine, cyano, nitro, alkyl, haloalkyl, heteroalkyl, alkenyl, alkynyl, hydroxy, decyl, amino, amido, alkylamino Acyl, aryl, heteroaryl and the like.
  • the heteroalkyl group may be, but not limited to, a hydroxyalkyl group, an alkoxyalkyl group, an aminoalkyl group, an alkylaminoalkyl group, a cyanoalkyl group, an alkoxy group, an alkylthio group and the like.
  • N-containing spiro group means that two rings share one carbon atom, wherein two rings are independently a carbocyclic or heterocyclic ring, and at least one of them is an N-containing heterocyclic ring.
  • the carbocyclic and heterocyclic rings may be independently substituted, wherein the substituent may be, but not limited to, fluorine, chlorine, bromine, iodine, cyano, nitro, alkyl, haloalkyl, heteroalkyl, chain Alkenyl, alkynyl, hydroxy, decyl, amino, amido, alkylamino, aryl, heteroaryl, and the like.
  • the heteroalkyl group may be, but not limited to, a hydroxyalkyl group, an alkoxyalkyl group, an aminoalkyl group, an alkylaminoalkyl group, a cyanoalkyl group, an alkoxy group, an alkylthio group and the like.
  • the "N-bridged ring-containing group" as used in the present invention means a bicyclic or polycyclic heterocyclic compound which shares two or more carbon atoms.
  • the heterocyclic compound may be substituted, wherein the substituent may be, but not limited to, fluorine, chlorine, bromine, iodine, cyano, nitro, alkyl, haloalkyl, heteroalkyl, alkenyl, alkyne Base, hydroxy, thiol, amino, amido, alkylaminoacyl, aryl, heteroaryl and the like.
  • the heteroalkyl group may be, but not limited to, a hydroxyalkyl group, an alkoxyalkyl group, an aminoalkyl group, an alkylaminoalkyl group, a cyanoalkyl group, an alkoxy group, an alkylthio group and the like.
  • a substituent is attached to the ring system formed on the ring by a chemical bond (as shown in the following scheme), indicating that the substituent may be substituted at any substitutable position on the ring.
  • formula a represents that R 1 may be substituted for any position on the spiro group that may be substituted, i.e., anywhere from 1 to 7, as shown in formula b.
  • the bridge atom is chemically bonded to the ring system formed on the ring (as shown in the figure below), which means that the bridge atom can be attached to any connectable C atom or hetero atom on the ring.
  • the formula c indicates that the bridge atom E can be bonded to any C atom or hetero atom of the bridgeable atom on the six-membered ring, that is, any position of 1-3.
  • the bridge atom E can also Connect with X or Q as shown in equation d.
  • the chemical bond is directly attached to the ring and interrupted by a bend line (as shown in e), indicating that the chemical bond is attached to a substituent and can be located at any position that can be substituted.
  • the formula e represents that the substituent may be at any position from 1 to 3, and when X or Q is N or C, the substituent may be bonded to X or Q.
  • isomeric forms thereof e.g., enantiomeric, diastereomeric, and geometric (or conformational).
  • R for example, the R, S configuration containing an asymmetric center, the (Z), (E) isomer of a double bond, and the conformational isomer of (Z), (E).
  • individual stereochemical isomers of the compounds of the invention, or enantiomers, diastereomers thereof, or mixtures of geometric isomers (or conformational isomers) are within the scope of the invention.
  • the "pharmaceutically acceptable salt” as used herein means an organic acid salt and a mineral acid salt of the compound of the present invention.
  • the pharmaceutically acceptable acid capable of forming salts thereof includes, but is not limited to, hydrochloric acid, sulfuric acid, phosphoric acid, hydrobromic acid, nitric acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, L-valine, Butyric acid, glycolic acid, acetic acid, acid, citric acid, propionic acid, malic acid, aspartic acid, malonic acid, succinic acid, tartaric acid, ethanedisulfonic acid, methanesulfonic acid, fumaric acid, benzoic acid, Lactic acid, succinic acid, adipic acid, ⁇ -ketoglutaric acid, lactobionic acid, maleic acid, 1,5-naphthalenedisulfonic acid, salicylic acid, acetylsalicylic acid,
  • solvent refers to an association of one or more solvent molecules with a compound of the invention.
  • Solvent-forming solvents include, but are not limited to, water, isopropanol, ethanol, methanol, acetone, acetonitrile, tetrahydrofuran, diisopropyl ether, dichloromethane, dimethyl sulfoxide, ethyl acetate, acetic acid, aminoethanol.
  • the compounds provided by the present invention, and pharmaceutical formulations thereof, have potential utility for the treatment of Bruton's tyrosine kinases, particularly diseases or conditions modulated by BTK receptors. It has the structure represented by formula (I) or an isomer thereof, a pharmaceutically acceptable solvate or salt:
  • Y is selected from substituted or unsubstituted aryl or heteroaryl
  • R is selected from substituted or unsubstituted alkenyl or alkynyl
  • M is selected from a substituted or unsubstituted N-containing spiro group or an N-containing ring-containing group, and the N atom is bonded to a carbonyl group;
  • M is selected from the group represented by formula (II), A is a substituted or unsubstituted spiro group or a bridged ring group, and an amino group is bonded to a carbonyl group;
  • M is preferably a substituted or unsubstituted C5-15 N-containing spiro group or N-containing bridged ring group, more preferably a substituted or unsubstituted C5-12 N-containing spiro group or N-containing bridge
  • the cyclic group is further preferably a substituted or unsubstituted C5-C8 N-containing or N-bridged ring-containing group.
  • the M is selected from any of the following groups:
  • n 1 , n 2 , and m are independently 0, 1, or 2;
  • n 1, 2 or 3;
  • X, Q are independently selected from CR 2 R 3 , NR 4 , O, S or S(O) 2 ;
  • R 1 , R 2 , R 3 , R 4 are independently selected from H, substituted or unsubstituted C 1-10 alkyl, substituted or unsubstituted C 1-10 heteroalkyl, C 1-10 carbonyl group, substituted or unsubstituted C 3 ⁇ 10 cycloalkyl, substituted or unsubstituted C3-10 heterocycloalkyl.
  • R 1 , R 2 , R 3 , R 4 are independently selected from H, substituted or unsubstituted C1-8 alkyl, substituted or unsubstituted C1-8 heteroalkyl, C1-8 carbonyl group, substituted or Unsubstituted C3-8 cycloalkyl, substituted or unsubstituted C3-8 heterocycloalkyl.
  • R 1 , R 2 , R 3 , R 4 are independently selected from H, substituted or unsubstituted C1-3 alkyl, substituted or unsubstituted C1-3 alkyl, C1-33 carbonyl group, substituted or Unsubstituted C3-6 cycloalkyl, substituted or unsubstituted C3-6 heterocycloalkyl.
  • R 1 , R 2 , R 3 , R 4 are independently selected from the group consisting of H, fluorine, chlorine, bromine, iodine, hydroxyl, thiol, cyano, amino, methyl, ethyl, trifluoromethyl, acetyl Base, isopropyl, trifluoroacetyl, isobutyl, cyclopropyl, epoxybutyl.
  • R 1 , R 2 , R 3 , R 4 may also be substituted by a secondary substituent which may be, but is not limited to, fluorine, chlorine, bromine, iodine, cyano, nitro, alkane Base, haloalkyl, heteroalkyl, alkenyl, alkynyl, hydroxy, decyl, amino, amido, alkylamino, aryl, heteroaryl, and the like.
  • a secondary substituent which may be, but is not limited to, fluorine, chlorine, bromine, iodine, cyano, nitro, alkane Base, haloalkyl, heteroalkyl, alkenyl, alkynyl, hydroxy, decyl, amino, amido, alkylamino, aryl, heteroaryl, and the like.
  • the heteroalkyl group may be, but not limited to, a hydroxyalkyl group, an alkoxyalkyl group, an aminoalkyl group, an alkylaminoalkyl group, a cyanoalkyl group, an alkoxy group, an alkylthio group and the like.
  • the M is selected from the group consisting of:
  • P is selected from CR 5 R 6 , NR 7 or O;
  • R 7 is a substituted or unsubstituted C 1-8 alkyl group, a substituted or unsubstituted C 1-8 heteroalkyl group, a substituted or unsubstituted C 3-8 cycloalkyl group, a substituted or unsubstituted C 3-8 heterocycloalkyl group, or
  • R 7 is a substituted or unsubstituted C1-6 alkyl group, a substituted or unsubstituted C1-6 heteroalkyl group, a substituted or unsubstituted C3-6 cycloalkyl group, a substituted or unsubstituted C3-6 heterocycloalkyl group, or
  • R 7 is H, cyano, methyl, ethyl, isopropyl, acetyl, trifluoroacetyl, cyclopropyl or epoxybutyl.
  • R 5 , R 6 , R 8 are independently selected from substituted or unsubstituted C1-8 alkyl, or substituted or unsubstituted C1-8 heteroalkyl, substituted or unsubstituted C3-8 cycloalkyl, substituted or unsubstituted C3 ⁇ 8 heterocycloalkyl;
  • R 5 , R 6 , R 8 are independently selected from substituted or unsubstituted C1-6 alkyl, or substituted or unsubstituted C1-6 alkyl, substituted or unsubstituted C3-6 cycloalkyl, substituted or Unsubstituted C3-6 heterocycloalkyl.
  • R 5 , R 6 , and R 8 are independently selected from the group consisting of H, fluorine, chlorine, bromine, iodine, hydroxyl, thiol, amino, methyl, ethyl, trifluoromethyl, acetyl, isopropyl, Trifluoroacetyl, isobutyl, cyclopropyl or epoxybutyl.
  • R 5 , R 6 , R 7 , R 8 may also be substituted by a secondary substituent which may be, but is not limited to, fluorine, chlorine, bromine, iodine, cyano, nitro, alkane Base, haloalkyl, heteroalkyl, alkenyl, alkynyl, hydroxy, decyl, amino, amido, alkylamino, aryl, heteroaryl, and the like.
  • a secondary substituent which may be, but is not limited to, fluorine, chlorine, bromine, iodine, cyano, nitro, alkane Base, haloalkyl, heteroalkyl, alkenyl, alkynyl, hydroxy, decyl, amino, amido, alkylamino, aryl, heteroaryl, and the like.
  • the heteroalkyl group may be, but not limited to, a hydroxyalkyl group, an alkoxyalkyl group, an aminoalkyl group, an alkylaminoalkyl group, a cyanoalkyl group, an alkoxy group, an alkylthio group and the like.
  • the M is selected from the group consisting of:
  • R is a substituted or unsubstituted alkenyl or alkynyl group. Preferred are the following groups:
  • R' is H, substituted or unsubstituted C1-8 alkyl, substituted or unsubstituted C1-8 heteroalkyl, substituted or unsubstituted C1-8 cycloalkyl or substituted or unsubstituted C1-8 Heterocycloalkyl;
  • R' is H, a substituted or unsubstituted C1-6 alkyl group, a substituted or unsubstituted C1-6 alkyl network, a substituted or unsubstituted C1-6 alkyl group or a substituted or unsubstituted C1 ⁇ 6 heterocycloalkyl;
  • R' is H, methyl, ethyl, N,N-dimethylaminomethyl, N-methyl-N-cyclopropylmethyl, methoxymethyl, ethoxymethyl, Trifluoromethoxymethyl, N,N-dicyclopropylmethyl or N-methyl-N-ethylmethyl.
  • R" is H, nitro, fluorine, chlorine, bromine, iodine or cyano.
  • the Y is a substituted or unsubstituted aryl or heteroaryl group, preferably a substituted or unsubstituted C5-10 aryl or heteroaryl group, more preferably a substituted or unsubstituted C6-8 aryl or heteroaryl group.
  • Aryl is a substituted or unsubstituted aryl or heteroaryl group, preferably a substituted or unsubstituted C5-10 aryl or heteroaryl group, more preferably a substituted or unsubstituted C6-8 aryl or heteroaryl group.
  • the substituent of the phenyl group is selected from a substituted or unsubstituted amide group, a substituted or unsubstituted alkyl group, a substituted or unsubstituted ether group.
  • the Y is selected from the group consisting of:
  • the substituent may be in the ortho, meta or para position.
  • R 9 is a trifluoromethyl group or a methyl group
  • R 10 , R 11 and R 12 are independently selected from substituted or unsubstituted aryl or heteroaryl.
  • R 10 , R 11 and R 12 are independently selected from substituted or unsubstituted C5-10 aryl or heteroaryl, more preferably, R 10 , R 11 and R 12 are independently selected from substituted or non-substituted. Substituted C5-8 aryl or heteroaryl.
  • R 10 , R 11 and R 12 are independently selected from substituted or unsubstituted phenyl, pyridyl, piperidinyl, piperazinyl or pyrimidinyl;
  • the substituent of the above group is selected from the group consisting of a nitro group, a hydroxyl group, a decyl group, a fluorine, a chlorine, a bromine, an iodine, a cyano group, a substituted or unsubstituted C10-10 alkyl group, a substituted or unsubstituted C1-10 heteroalkyl group, a substituted or not Substituting a C3-10 cycloalkyl group, a substituted or unsubstituted C3-10 heterocycloalkyl group.
  • the A is a substituted or unsubstituted spiro group or a bridged ring group, preferably a substituted or unsubstituted C6-15 spiro ring group or a bridged ring group.
  • the A is adamantyl.
  • the Y is a substituted or unsubstituted 2-aminopyridylbenzamide group
  • M is a C5-10 spiro or bridged ring group
  • R is a vinyl group or Propynyl.
  • H on the pyridyl group of Y may be optionally substituted with fluorine, chlorine, bromine, iodine, methyl or trifluoromethyl, the substitution may be ortho Substitution, meta substitution or para substitution.
  • the Y is a diphenylethanol group
  • M is a spiro or bridged ring group of C5-10
  • R is a vinyl or propynyl group
  • the Y is a diphenyl ether group
  • M is a spiro or bridged ring group of C5-10
  • R is a vinyl or propynyl group
  • the Y is a 2-aminopyridylbenzamide group
  • M is a C5-10 bridged ring group
  • R is a vinyl group
  • the Y is a diphenylethanol group
  • M is a bridged ring group of C5-10
  • R is a vinyl group
  • the Y is a diphenyl ether group
  • M is a bridged ring group of C5-10
  • R is a vinyl group
  • the Y is a trifluoromethyl substituted 2-aminopyridylbenzamide group
  • M is a C5-10 bridged ring group
  • R is a vinyl group.
  • the compound has any of the following structures or a stereoisomer or cis-trans isomer thereof:
  • the invention also provides a preparation method of the above compound, comprising the following steps:
  • the compound represented by the above formula (VI) can be reacted with 3-chloropropionyl chloride or acryloyl chloride under the action of a base by direct condensation or direct condensation to eliminate the olefination of hydrogen chloride to obtain the formula (I-2).
  • the compound represented by the above formula (VI) and the enoic acid derivative represented by the formula (VII) are reacted under a condensing agent to obtain a compound of the formula (I-3) or the formula (I-4), R. "H or fluorine, chlorine, bromine, iodine, respectively;
  • the compound of the above formula (VI) and the cyanoacetic acid or nitroacetic acid are subjected to a condensing agent to obtain an amide compound, and then subjected to a brain text reaction with the aldehyde compound represented by the formula (VIII) to obtain a formula (I- 4) a compound shown; R" is a nitro group or a cyano group;
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising the above compound or a salt thereof or a compound prepared by the above production method or a salt thereof, and a pharmaceutically acceptable carrier, excipient, diluent, adjuvant, vehicle or they The combination.
  • the present invention also provides the use of the above compound or the compound prepared by the above production method or the above pharmaceutical composition for the preparation of a medicament for treating or ameliorating a BTK-mediated disease.
  • the BTK-mediated disease is selected from any one or more of the group consisting of an immune, an autoimmune, an inflammatory disease, an allergy, an infectious disease, a proliferative disorder, and a cancerous disease.
  • the BTK-mediated disease is selected from the group consisting of rheumatoid arthritis, infectious arthritis, teratogenic arthritis, gouty arthritis, spondylitis, pancreatitis, chronic bronchitis , acute bronchitis, allergic bronchitis, toxic bronchitis, pediatric asthma, allergic alveolitis, allergic or non-allergic rhinitis, chronic sinusitis, cystic fibrosis or mucous viscous disease, cough, emphysema , interstitial lung disease, alveolitis, nasal polyps, pulmonary edema, various causes of pneumonia, lupus erythematosus, systemic scleroderma, sarcoidosis, sub-diffuse large B-cell lymphoma, mantle cell lymphoma ( MCL), chronic lymphocytic lymphoma, extranodal marginal B-cell lymphoma, B-cell chronic rheumatoid arthritis,
  • compositions of the present invention may be used alone or in combination with other drugs when applied to the preparation of a medicament for treating or ameliorating BTK-mediated diseases.
  • the present invention provides a compound having the structure of the formula (I), or an isomer thereof, a pharmaceutically acceptable solvate thereof, or a salt thereof, for use as Bruton's tyrosine kinase inhibitor
  • the agent has a high inhibitory activity against BTK and has a small adverse reaction.
  • DMF stands for N,N-dimethylformamide
  • NBS represents N-bromosuccinimide
  • DCM dichloromethane
  • TEA means triethylamine
  • THF represents tetrahydrofuran
  • TFA represents trifluoroacetic acid
  • EA means ethyl acetate
  • PE represents petroleum ether
  • Et2O represents diethyl ether
  • DIEA represents N,N-diisopropylethylamine
  • HBTU represents benzotriazole-N,N,N',N'-tetramethylurea hexafluorophosphate
  • TLC means thin layer chromatography
  • KOAc means potassium acetate
  • X-phos represents 2-dicyclohexylphosphino-2,4,6-triisopropylbiphenyl
  • Pd 2 (dba) 3 represents tris(dibenzylidenefluorenone) dipalladium
  • Pd(pph 3 ) 4 represents triphenylphosphine palladium
  • n-BuLi represents t-butyllithium
  • EDCI represents 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
  • DIPEA represents N,N-diisopropylethylamine
  • STAB represents sodium triacetoxyborohydride
  • DMSO means dimethyl sulfoxide
  • FAM represents carboxyfluorescein
  • ATP represents adenine nucleoside triphosphate
  • MEM means minimum essential medium
  • FBS means fetal bovine serum
  • IMDM means Iscove’s Modified Dulbecco’s Media
  • RPMI 1640medium represents Roswell Park Memorial Institute 1640 medium
  • HEPES represents 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid
  • EGTA represents ethylene glycol diethyl ether diamine tetraacetic acid
  • Na-ATP represents adenosine nucleoside triphosphate.
  • reaction was stirred at -78 °C for 10 min.
  • DMF (84 ml, 1092 mmol) was dissolved in THF (50 mL) and added dropwise to the reaction mixture, and the temperature of the reaction system was controlled at -70 ° C to -78 ° C for 10 min, and the reaction was stirred at -78 ° C for 2 h.
  • MeOH 800 mL was added to the reaction mixture, and the mixture was added, and NaBH 4 (33 g, 868 mmol) was added portionwise to the reaction mixture, and the reaction mixture was allowed to rise to room temperature and stirred for 2 h.
  • Step 4 Preparation of (S)-6-(((3-chloropyrazin-2-yl)methyl)carbamoyl)-5-azaspiro[2.4]heptane-5-carboxylic acid tert-butyl ester
  • Step 5 Preparation of (S)-6-(8-chloroimidazo[1,5-a]pyrazin-3-yl)-5-azaspiro[2.4]heptane-5-carboxylic acid tert-butyl ester
  • Step 6 (S)-6-(1-Bromo-8-chloroimidazo[1,5-a]pyrazin-3-yl)-5-azaspiro[2.4]heptane-5-carboxylic acid Preparation of butyl ester
  • Step 7 (S)-6-(8-Amino-1-bromoimidazo[1,5-a]pyrazin-3-yl)-5-azaspiro[2.4]heptane-5-carboxylic acid Preparation of butyl ester
  • Step 8 (S)-6-(8-Amino-1-(4-(pyridin-2-ylcarbamoyl)phenyl)imidazo[1,5-a]pyrazin-3-yl)-5 -Preparation of aza-spiro[2.4]heptane-5-carboxylic acid tert-butyl ester
  • Step 9 (S)-4-(8-Amino-3-(5-azaspiro[2.4]heptan-6-yl)imidazo[1,5-a]pyrazine-1-yl)-N -(pyridin-2-yl) Preparation of benzamide
  • Step 10 a(S)-4-(8-Amino-3-(5-(2-butynyl)-5-azaspiro[2.4]heptan-6-yl)imidazo[1,5- Preparation of a]pyrazine-1-yl)-N-(pyridin-2-yl)benzamide
  • the structure of the product was characterized by nuclear magnetic resonance and mass spectrometry. The results are as follows:
  • the structure of the product was characterized by nuclear magnetic resonance and mass spectrometry. The results are as follows:
  • Example 1 Using the following compounds as starting materials, the following compounds were prepared by the preparation method of Example 1 or Example 2. The structures and nuclear magnetic characterization data of the compounds are shown in Table 1. Table 1 shows the structures of the compounds prepared in Examples 3 to 21 of the present application. And a summary of structural analysis data.
  • Example 22 4-(3-((1R,3S,4S)-2-acryloyl-2-azabicyclo[2.2.1]heptan-3-yl)-8-aminoimidazo[1,5 -a]Preparation of pyrazin-1-yl)-N-(pyridin-2-yl)benzamide
  • Step 5 (1R,3S,4S)-3-(8-Amino-1-(4-(pyridin-2-ylcarbamoyl)phenyl)imidazo[1,5-a]pyrazine-3- Of 2-(2-dicyclobicyclo[2.2.1]heptane-2-carboxylic acid tert-butyl ester
  • Step 6 4-(8-Amino-3-((1R,3S,4S)-2-azabicyclo[2.2.1]heptan-3-yl)imidazo[1,5-a]pyrazine- Preparation of 1-yl)-N-(pyridin-2-yl)benzamide
  • Step 7 4-(3-((1R,3S,4S)-2-acryloyl-2-azabicyclo[2.2.1]heptan-3-yl)-8-aminoimidazo[1,5- Preparation of a]pyrazine-1-yl)-N-(pyridin-2-yl)benzamide
  • the structure of the product was characterized by nuclear magnetic resonance and mass spectrometry. The results are as follows:
  • the structure of the product was characterized by nuclear magnetic resonance and mass spectrometry. The results are as follows:
  • Example 22 The following compounds were prepared by the preparation method of Example 22 or Example 23.
  • the structure and nuclear magnetic characterization data of the compound are shown in Table 2.
  • Table 2 summarizes the structure and structural analysis data of the compounds prepared in Examples 24 to 39 of the present application.
  • Step 1 4-(4,4,5,5-Tetramethyl-1,3,2-dioxaborolan-2-yl)-N-(4-(trifluoromethyl)pyridine-2- Preparation of benzamide
  • Step 2 (S)-6-(8-Amino-1-(4-((4-(trifluoromethyl)pyridin-2-yl)carbamoyl)phenyl)imidazo[1,5-a Preparation of pyrazin-3-yl)-5-azaspiro[2.4]heptane-5-carboxylic acid tert-butyl ester
  • Step 3 (S)-4-(8-Amino-3-(5-azaspiro[2.4]heptan-6-yl)imidazo[1,5-a]pyrazine-1-yl)-N -(4-(Trifluoromethyl)pyridin-2-yl)benzamide preparation
  • Step 4 (S)-4-(8-Amino-3-(5-(2-butynyl)-5-azaspiro[2.4]heptan-6-yl)imidazo[1,5-a Preparation of pyrazin-1-yl l)-N-(4-(trifluoromethyl)piperidin-2-yl)benzamide
  • the structure of the product was characterized by nuclear magnetic resonance and mass spectrometry. The results are as follows:
  • the structure of the product was characterized by nuclear magnetic resonance and mass spectrometry. The results are as follows:
  • Example 40 or Example 41 Using a similar structural compound as a raw material, the preparation method of Example 40 or Example 41 was used to prepare the following.
  • the structure and the nuclear magnetic characterization data of the compound are shown in Table 3.
  • Table 3 is a summary of the structure and structural analysis data of the compounds prepared in Examples 42 to 72 of the present application.
  • phenylmagnesium bromide (10.05 mL, 30.15 mmol, 3M in Et 2 O) was added dropwise to a solution of 4-bromoacetophenone (5 g, 25.13 mmol) in 30 mL of THF at -45 °C. ), and the reaction mixture was further stirred at -45 °C reaction 1h, TLC showed starting material the reaction was completed, slowly added saturated NH 4 Cl the reaction was quenched, the reaction stirring was continued for 0.5h.
  • Step 2 Preparation of 1-phenyl-1-(4-(4,4,5,5-tetramethyl-1,3,2-dioxaborolan-2-yl)phenyl)ethanol
  • Step 3 (6S)-6-(8-Amino-1-(4-(1-hydroxy-1-phenylethyl)phenyl)imidazo[1,5-a]pyrazin-3-yl)- Preparation of 5-Azaspiro[2.4]heptane-5-carboxylic acid tert-butyl ester
  • Step 4 1-(4-(8-Amino-3-((S)-5-azaspiro[2.4]heptan-6-yl)imidazo[1,5-a]pyrazin-1-yl Preparation of phenyl)-1-phenylethanol
  • Step 5 1-((6S)-6-(8-Amino-1-(4-(1-hydroxy-1-phenylethyl)phenyl)imidazo[1,5-a]pyrazin-3-yl Preparation of 5-azaspiro[2.4]heptane-5-yl)2-butynoyl-1-one
  • the structure of the product was characterized by nuclear magnetic resonance and mass spectrometry. The results are as follows:
  • the structure of the product was characterized by nuclear magnetic resonance and mass spectrometry. The results are as follows:
  • Example 73 The following compounds were prepared by the preparation method of Example 73 or Example 74 using a similar structural compound as a starting material.
  • the structure and nuclear magnetic characterization data of the compound are shown in Table 4.
  • Table 4 shows the compounds prepared in Examples 75-91 of the present application. Summary of structure and structural analysis data.
  • Step 1 (S)-6-(8-Amino-1-(4-phenoxyphenyl)imidazo[1,5-a]pyrazin-3-yl)-5-azaspiro[2.4] Preparation of tert-butyl heptane-5-carboxylate
  • Step 2 (S)-1-(4-Phenoxyphenyl)-3-(5-azaspiro[2.4]heptan-6-yl)imidazo[1,5-a]pyrazine-8 - Preparation of amines
  • Step 3 (S)-1-(6-(8-amino-1-(4-phenoxyphenyl)imidazo[1,5-a]pyrazin-3-yl)-5-azaspiro[ Preparation of 2.4]heptane-5-yl)-2-butyn-1-one
  • the structure of the product was characterized by nuclear magnetic resonance and mass spectrometry. The results are as follows:
  • the structure of the product was characterized by nuclear magnetic resonance and mass spectrometry. The results are as follows:
  • Example 92 The following compounds were prepared by the preparation method of Example 92 or Example 93 using a similar structural compound as a starting material.
  • the structure and nuclear magnetic characterization data of the compound are shown in Table 5.
  • Table 5 shows the compounds prepared in Examples 94-111 of the present application. Summary of structure and structural analysis data.
  • Ethyl bromocrotonate (90% purity, 2.8 g, 12.95 mmol) was added to a solution of cyclopropylamine (2.2 g, 38.85 mol), K 2 CO 3 (3.6 g, 25.9 mmol) in 20 mL THF. , stirred at room temperature overnight, after completion by TLC of the reaction, the above reaction mixture was added water to quench the reaction, EA (15mLx3). the organic was washed with saturated NaCl phase was used were combined, dried over anhydrous Na 2 SO 4 sufficiently dried and evaporated in vacuo to give 1.1 g Target compound was used in the next reaction without further purification.
  • Step 4 (S,E)-4-(8-Amino-3-(5-(4-(cyclopropyl(methyl)amino)-2-butenoyl)-5-azaspiro[2.4] Preparation of heptane-6-yl)imidazo[1,5-a]pyrazin-1-yl)-N-(piperidin-2-yl)benzamide
  • Step 1 (S)-4-(8-Amino-3-(5-(2-cyanoacetyl)-5-azaspiro[2.4]heptan-6-yl)imidazo[1,5-a Preparation of pyrazin-1-yl)-N-(pyridin-2-yl)benzamide
  • Step 2 (S,Z)-4-(8-Amino-3-(5-(2-cyano-4-methyl-2-pentenoyl)-5-azaspiro[2.4]heptane- Preparation of 6-yl)imidazo[1,5-a]pyrazin-1-yl)-N-(pyridin-2-yl)benzamide
  • Test Example 1 In vitro BTK inhibitory kinase activity assay
  • Mobility-Shift Assay a microfluidic chip technology that applies the basic idea of capillary electrophoresis to a microfluidic environment.
  • the substrate used for the experiment is a fluorescently labeled peptide, which is enzymed in the reaction system. Under the action of the substrate, the substrate is transformed into a product, and the charge it carries also changes accordingly.
  • the Mobility-Shift Assay separates the two by using the difference in charge between the substrate and the product, and separately detects it. The test results are expressed by the conversion rate.
  • the 10 concentrations of the compound were diluted 10-fold with 1 ⁇ kinase buffer; wherein the kinase buffer contained hydroxyethylpiperazine ethanesulfonic acid at a concentration of 50 mmol/L and a pH of 7.5, 0.01%.
  • adding a substrate solution to the 384-well plate adding 10 ⁇ l of a 2.5-fold substrate solution to the 384-well reaction plate;
  • Test Example 2 Determination of in vitro cell proliferation inhibitory activity of compounds using different cell lines
  • test compounds were diluted with DMSO to prepare a final concentration of 10 mM mother liquor for use.
  • the logarithmic growth phase cells were collected, counted, and the cells were resuspended in complete medium, and the cell concentration was adjusted to an appropriate concentration (determined according to the cell density optimization test results), and 96-well plates were seeded, and 100 ⁇ l of the cell suspension was added to each well. The cells were incubated for 24 hours at 37 ° C in a 100% relative humidity, 5% CO 2 incubator.
  • test compound was diluted with the medium to the corresponding concentration of action set, and the cells were added at 25 ⁇ l/well. The cells were incubated for 72 hours at 37 ° C in a 100% relative humidity, 5% CO 2 incubator.
  • the medium was aspirated, and the complete medium containing 10% of the detection reagent was added and incubated in a 37 ° C incubator for 1-4 hours.
  • the absorbance at a wavelength of 450 nm was measured on a SpectraMax M5 Microplate Reader with gentle shaking, and the absorbance at 650 nm was used as a reference to calculate the inhibition rate.
  • the logarithmic growth phase cells were collected, counted, and the cells were resuspended in complete medium, and the cell concentration was adjusted to an appropriate concentration (determined according to the cell density optimization test results), and 96-well plates were seeded, and 90 ⁇ l of the cell suspension was added to each well.
  • the cells were incubated for 24 hours at 37 ° C in a 100% relative humidity, 5% CO 2 incubator.
  • test compound was diluted with the medium to the corresponding concentration of action set, and the cells were added at 10 ⁇ l/well. The cells were incubated for 72 hours at 37 ° C in a 100% relative humidity, 5% CO 2 incubator.
  • tumor cell growth inhibition rate % [(Ac-As) / (Ac-Ab)] ⁇ 100%
  • Test Example 3 hERG experiment to investigate potential cardiotoxicity
  • CHO hERG cells were grown in a Petri dish containing the above culture solution, and cultured in an incubator containing 5% CO 2 at 37 °C. CHO hERG cells were transferred to circular glass slides placed in petri dishes 24 to 48 hours prior to electrophysiological experiments and grown under the same culture and culture conditions as above. The density of CHO hERG cells on each circular slide requires that most cells be independent, individual requirements.
  • This experiment used a manual patch clamp system (HEKA EPC-10 signal amplifier and digital conversion system, purchased from HEKA Electronics, Germany) for the recording of whole cell currents.
  • a circular slide with CHO hERG cells grown on it was placed in an electrophysiology recording trough under an inverted microscope.
  • the extracellular fluid was continuously perfused in the recording tank (about 1 ml per minute).
  • the experimental procedure uses conventional whole-cell patch clamp current recording techniques. Unless otherwise stated, the experiments were carried out at regular room temperature ( ⁇ 25 ° C). The cells were clamped at a voltage of -80 mV.
  • the cell clamp voltage was depolarized to +20 mV to activate the hERG potassium channel, and after 5 seconds it was clamped to -50 mV to eliminate inactivation and generate tail current.
  • the tail current peak is used as the value of the hERG current magnitude.
  • the hERG potassium current recorded in the above steps is stabilized after continuous extracellular fluid perfusion in the recording tank.
  • the drug to be tested can be superimposed and filled until the inhibitory effect of the drug on the hERG current reaches a steady state.
  • the recent three consecutive current recording lines are recombined as a criterion for judging whether or not the state is stable. After reaching a steady state, rinse with extracellular fluid until the hERG current returns to the size before the drug is added.
  • Cisapride (cisapride, purchased from Sigma) was used in the experiment as a positive control to ensure that the cells used were of normal quality.
  • IC 50 of the compound we selected the following concentrations (30, 10, 3, 1, 0.3, and 0.1 ⁇ M) for testing. Prior to testing, the cells were first diluted to a 10, 3, 1, 0.3, and 0.1 mM stock solution in gradual dilution with DMSO and diluted to the final ⁇ M test concentration with extracellular fluid. The final concentration of DMSO in each of the other compound solutions was 0.1% except that the DMSO concentration in the 30 ⁇ M compound test solution was 0.3%. The positive control Cisapride (cisapride) was tested at a concentration of 0.1 [mu]M. All compound solutions were sonicated and shaken for 5 to 10 minutes to ensure complete dissolution of the compound.
  • test data was analyzed by HEKA Patchmaster, Microsoft Excel and data analysis software provided by Graphpad Prism.
  • Test Example 4 Pharmacokinetic test of the compound of the present invention
  • the time points were: before administration, 5 min, 15 min, 30 min, 1 h, 2 h, 4 h, 6 h after administration.
  • 3 whole blood samples were collected at each time point, and the collected amount was about 0.2 mL, and anticoagulated by heparin sodium. Blood samples were immediately placed on ice and centrifuged to separate plasma within 30 minutes (centrifugation conditions: 8000 rpm, 6 minutes, 2-8 ° C). The collected plasma was stored at –70 °C prior to analysis.
  • Example 3 0.62 0.27 1619.79 1492.19 3,420.38 52.55
  • Example 5 0.13 0.10 303.06 278.20 7278.25 20.64
  • Example 11 0.47 0.33 1612.51 1583.67 2187.37 34.59
  • Example 22 0.60 0.25 1156.00 1038.16 3510.37 36.56
  • Example 96 0.55 0.36 1589.06 1580.79 1960.88 34.53 Ibrutinib 0.26 0.33 217.10 218.09 1200.00 2.67 ACP-196 0.13 0.08 266.02 266.08 7489.19 19.75
  • the compound of the present invention has a significant inhibitory effect on BTK activity, and the cardiotoxicity is extremely small compared with the marketed drug ibrutinib or the clinical phase ACP-196, and the pharmacokinetic aspect is also It has obvious advantages and can be used as a BTK inhibitor, and has broad application prospects against malignant tumor diseases or inflammatory diseases.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)

Abstract

本发明提供了具有式(Ⅰ)所示结构的化合物,或其异构体、药学上可接受的溶剂化物、盐,用作布鲁顿酪氨酸激酶抑制剂,其对于BTK具有较高的抑制活性,同时具有较小的不良反应。

Description

用作布鲁顿酪氨酸激酶抑制剂的化合物及其制备方法和应用
本申请要求于2017年01月20日提交中国专利局、申请号为201710044771.4、发明名称为“用作布鲁顿酪氨酸激酶抑制剂的化合物及其制备方法和应用”的中国专利申请的优先权,其全部内容通过引用结合在本申请中。
技术领域
本发明涉及药物化学技术领域,尤其涉及一种用作布鲁顿酪氨酸激酶抑制剂的化合物及其制备方法和应用。
背景技术
自从2012年美国血液学会会议中报道了依鲁替尼对多种B细胞淋巴瘤有良好的治疗效果之后,人们就已经认识到BTK作为一种非受体蛋白酪氨酸激酶家族的成员,是除了T淋巴细胞和自然杀伤细胞之外的所有造血细胞类型中表达的关键信号酶。在B细胞信号传导途径中扮演至关重要的角色,与B淋巴细胞发育、分化、信号传递和存活密切相关。BTK在B细胞受体(BCR)信号通路的重要作用,使其成为B细胞恶性肿瘤治疗的热门靶点。
虽然依鲁替尼在治疗慢性淋巴细胞白血病(CLL)、套细胞淋巴瘤(MCL)和
Figure PCTCN2017074108-appb-000001
巨球蛋白血症方面已经改善了某些恶性肿瘤的生存期,并且避免了传统化疗导致的多种副作用。但是在临床给药过程中,依然存在易被代谢、生物利用度低(仅仅7~23%)、临床给药量大(560mg/天,4片/次)、持续疗程长(一个疗程28天,有时候需要几个疗程)、服用短时间内就会产生很强的耐药性等问题。
另外依鲁替尼最常见的副作用主要是出血、感染、血细胞减少、心房颤动、第二原发性恶性肿瘤、肿瘤溶解综合征、胚胎或胎儿毒性等。以上的出血事件(如硬膜下血肿,胃肠道出血,血尿和后程序出血)发生率达6%,出血机制还不清楚;发生3或3级以上的感染概率为14%~26%;出现3或4级的血细胞减少,包括中性粒细胞减少(19%~29%),血小板减少(5%~17%)和贫血(0~9%);出现心房颤动和心房扑动的几率为6%~9%,尤其是心脏病患者,急性感染患者,房颤病史的患者;出现其他恶性肿瘤几率为5%~14%,包括非皮肤癌(1%~3%),最常见的是非黑色素瘤皮肤癌(4%~11%);经依鲁替尼治疗的患者出现过肿瘤溶解综合征。
所以进一步开发既能增加病人顺应性、又能维持高效性、高安全性、且将不良反应最小化的新BTK抑制剂是十分必要的。
发明内容
有鉴于此,本发明要解决的技术问题在于提供一种用作布鲁顿酪氨酸激酶抑制剂的化合物及其制备方法和应用,制备的化合物与目前已经上市的依鲁替尼相比,本专利公布的一些布鲁顿酪氨酸激酶抑制剂化合物具有较好的活性、更高的心脏安全性,尤其是在药代方面,在动物口服后,不管是血药浓度、暴露量、半衰期、口服生物利用度等参数都具有明显的优势。因此本专利公布的布鲁顿酪氨酸激酶抑制剂化合物具有广泛的临床应用前景。
定义和一般术语
本发明所述的“取代”,表示所给结构中的一个或多个氢原子被取代基所取代。当所给出的结构式中不止一个位置能被取代基所取代时,所述取代基可以相同或不同。上述取代基可以任选的被一个或多个相同或不同的次级取代基所取代。其中所述的次级取代基可以 是,但并不限于,氟,氯,溴,碘,氰基,硝基,烷基,卤素烷基,杂烷基,链烯基,炔基,羟基,巯基,氨基,酰胺基,烷氨基酰基,芳基,杂芳基等。所述杂烷基可以是,但不限于,羟基烷基,烷氧基烷基,氨基烷基,烷氨基烷基,氰基烷基,烷氧基,烷硫基等。
本发明所述的“含N螺环基团”表示两个环共用一个碳原子,其中两个环独立的为碳环或杂环,且至少有一个为含N杂环。所述碳环和杂环可以独立的被取代,其中取代基可以是,但并不限于,氟,氯,溴,碘,氰基,硝基,烷基,卤素烷基、杂烷基,链烯基,炔基,羟基,巯基,氨基,酰胺基,烷氨基酰基,芳基,杂芳基等。所述杂烷基可以是,但不限于,羟基烷基,烷氧基烷基,氨基烷基,烷氨基烷基,氰基烷基,烷氧基,烷硫基等。
本发明所述的“含N桥环基团”表示共用两个或两个以上碳原子的双环或多环杂环化合物。所述杂环化合物可以被取代,其中取代基可以是,但并不限于,氟,氯,溴,碘,氰基,硝基,烷基,卤素烷基、杂烷基,链烯基,炔基,羟基,巯基,氨基,酰胺基,烷氨基酰基,芳基,杂芳基等。所述杂烷基可以是,但不限于,羟基烷基,烷氧基烷基,氨基烷基,烷氨基烷基,氰基烷基,烷氧基,烷硫基等。
如本发明所描述的,取代基用化学键连接到环上形成的环体系(如下图所示),代表取代基在环上任何可取代的位置都可以取代。例如,式a表示R1可取代螺环基团上任何可能被取代的位置,即1~7的任意位置,如式b所示。
Figure PCTCN2017074108-appb-000002
桥原子用化学键连接到环上形成的环体系(如下图所示),代表桥原子可与环上任意可连接的C原子或杂原子连接。例如式c表示桥原子E可与六元环上任意的可连接桥原子的C原子或杂原子连接,即1~3的任意位置,当X或Q为N或C时,桥原子E也可以与X或Q连接,如式d所示。
Figure PCTCN2017074108-appb-000003
化学键直接连接到环上,并用弯线打断(如图e所示),表示该化学键连接有取代基,并且可以位于任意可被取代的位置。例如式e表示取代基可以位于1~3的任意位置,当X或Q为N或C时,取代基可以与X或Q相连。
Figure PCTCN2017074108-appb-000004
本发明所述的“异构体”指,本发明所描述的结构式包括其所有的同分异构形式(如对映异构,非对映异构,和几何异构(或构象异构)):例如含有不对称中心的R、S构型,双键的(Z)、(E)异构体,和(Z)、(E)的构象异构体。因此,本发明的化合物的单个立体化学异构体或其对映异构体,非对映异构体,或几何异构体(或构象异构体)的混合物都属于本发明的范围。
本发明所述的“药学上可接受的盐”是指本发明的化合物的有机酸盐和无机酸盐。所述药学上可接受的能与之成盐的酸包括但不限于盐酸、硫酸、磷酸、氢溴酸、硝酸、苯磺酸、对甲基苯磺酸、柠檬酸、L-脯氨酸、丁酸、羟乙酸、乙酸、已酸、壬酸、丙酸、苹果酸、天门冬氨酸、丙二酸、琥珀酸、酒石酸、乙二磺酸、甲磺酸、富马酸、苯甲酸、乳酸、丁二磺酸、已二酸、α-酮戊二酸、乳糖酸、马来酸、1,5-萘二磺酸、水杨酸、乙酰水杨酸、2-萘磺酸、苯乙酸、烟酸、1-羟基-2-萘甲酸、樟脑酸、2-羟基乙磺酸、扁桃酸、苦味酸、肉桂酸或草酸等。
本发明所述的“溶剂化物”是指一个或多个溶剂分子与本发明的化合物所形成的缔合物。形成溶剂化物的溶剂包括,但并不限于,水,异丙醇,乙醇,甲醇,丙酮、乙腈、四氢呋喃、异丙醚、二氯甲烷、二甲亚砜,乙酸乙酯,乙酸,氨基乙醇。
化合物
本发明提供的化合物,及其药物制剂,对布鲁顿酪氨酸激酶,尤其是BTK受体调节的疾病或病症的治疗具有潜在的用途。其具有式(Ⅰ)所示结构或其异构体、药学上可接受的溶剂化物或盐:
Figure PCTCN2017074108-appb-000005
其中,Y选自取代或非取代的芳基或杂芳基;
R选自取代或非取代的烯基或炔基;
M选自取代或非取代的含N螺环基团或含N桥环基团,且N原子与羰基相连;
或者M选自式(Ⅱ)所示基团,A为取代或非取代的螺环基团或桥环基团,且氨基与羰基相连;
Figure PCTCN2017074108-appb-000006
其中,M优选为取代或非取代的C5~15的含N螺环基团或含N桥环基团,更优选为取代或非取代的C5~12的含N螺环基团或含N桥环基团,进一步优选为取代或非取代的C5-C8的含N螺环或含N桥环基团。
在本发明的某些具体实施例中,所述M选自以下任一基团:
Figure PCTCN2017074108-appb-000007
其中,n1、n2、m独立的为0、1或2;
n为1、2或3;
X、Q独立的选自CR2R3、N-R4、O、S或S(O)2
R1、R2、R3、R4独立的选自H、取代或未取代C1~10烷基,取代或未取代C1~10杂烷基,C1~10羰基基团,取代或未取代C3~10环烷基,取代或未取代C3~10杂环烷基。
优选的,R1、R2、R3、R4独立的选自H、取代或未取代C1~8烷基,取代或未取代C1~8杂烷基,C1~8羰基基团,取代或未取代C3~8环烷基,取代或未取代C3~8杂环烷基。
进一步的,R1、R2、R3、R4独立的选自H、取代或未取代C1~3烷基,取代或未取代C1~3杂烷基,C1~3羰基基团,取代或未取代C3~6环烷基,取代或未取代C3~6杂环烷基。
更优选的,R1、R2、R3、R4独立的选自H、氟、氯、溴、碘、羟基、巯基、氰基、氨基、甲基、乙基、三氟甲基、乙酰基、异丙基、三氟乙酰基、异丁基、环丙基、环氧丁基。
上述R1、R2、R3、R4还可以被次级取代基取代,所述次级取代基可以是,但并不限于,氟,氯,溴,碘,氰基,硝基,烷基,卤代烷基、杂烷基,链烯基,炔基,羟基,巯基,氨基,酰胺基,烷氨基酰基,芳基,杂芳基等。所述杂烷基可以是,但不限于,羟基烷基,烷氧基烷基,氨基烷基,烷氨基烷基,氰基烷基,烷氧基,烷硫基等。
在本发明另外一些具体实施例中,所述M选自以下基团:
Figure PCTCN2017074108-appb-000008
其中,P选自CR5R6,N-R7或O;
R7为取代或非取代C1~8烷基,取代或非取代C1~8杂烷基,取代或未取代C3~8环烷基,取代或未取代C3~8杂环烷基,或
Figure PCTCN2017074108-appb-000009
优选的,R7为取代或非取代C1~6烷基,取代或非取代C1~6杂烷基,取代或未取代C3~6环烷基,取代或未取代C3~6杂环烷基,或
Figure PCTCN2017074108-appb-000010
更优选的,R7为H、氰基、甲基、乙基、异丙基、乙酰基、三氟乙酰基、环丙基或环氧丁基。
R5、R6、R8独立的选自取代或非取代C1~8烷基,或取代或非取代C1~8杂烷基,取代或未取代C3~8环烷基,取代或未取代C3~8杂环烷基;
优选的,R5、R6、R8独立的选自取代或非取代C1~6烷基,或取代或非取代C1~6杂烷基,取代或未取代C3~6环烷基,取代或未取代C3~6杂环烷基。
更优选的,R5、R6、R8独立的选自H、氟、氯、溴、碘、羟基、巯基、氨基、甲基、乙基、三氟甲基、乙酰基、异丙基、三氟乙酰基、异丁基、环丙基或环氧丁基。
上述R5、R6、R7、R8还可以被次级取代基取代,所述次级取代基可以是,但并不限于,氟,氯,溴,碘,氰基,硝基,烷基,卤代烷基、杂烷基,链烯基,炔基,羟基,巯基,氨基,酰胺基,烷氨基酰基,芳基,杂芳基等。所述杂烷基可以是,但不限于,羟基烷基,烷氧基烷基,氨基烷基,烷氨基烷基,氰基烷基,烷氧基,烷硫基等。
在本发明的另外一些具体实施中,所述M选自以下具体基团:
Figure PCTCN2017074108-appb-000011
Figure PCTCN2017074108-appb-000012
所述R为取代或非取代的烯基或炔基。优选为以下基团:
Figure PCTCN2017074108-appb-000013
其中,R'为H、取代或非取代的C1~8烷基、取代或非取代的C1~8杂烷基,取代或非取代的C1~8环烷基或取代或非取代的C1~8杂环烷基;
优选的,R'为H、取代或非取代的C1~6烷基、取代或非取代的C1~6杂烷基,取代或非取代的C1~6环烷基或取代或非取代的C1~6杂环烷基;
更优选的,R'为H、甲基、乙基、N,N-二甲氨基甲基、N-甲基-N-环丙基甲基、甲氧基甲基、乙氧基甲基、三氟甲氧基甲基、N,N-二环丙基甲基或N-甲基-N-乙基甲基。
R"为H、硝基、氟、氯、溴、碘或氰基。
所述Y为取代或非取代的芳基或杂芳基,优选为取代或非取代的C5~10的芳基或杂芳基,更优选为取代或非取代的C6~8的芳基或杂芳基。
当所述Y选自取代苯基时,所述苯基的取代基选自取代或非取代酰胺基团、取代或非取代烷基、取代或非取代醚基。
在本发明的某些具体实施例中,所述Y选自以下基团:
Figure PCTCN2017074108-appb-000014
所述取代基可以位于邻位、间位或对位。
其中,R9为三氟甲基或甲基;
R10,R11和R12独立的选自取代或非取代的芳基或杂芳基。
优选的,R10,R11和R12独立的选自取代或非取代的C5~10的芳基或杂芳基,更优选 的,R10,R11和R12独立的选自取代或非取代的C5~8的芳基或杂芳基.
在本发明的某些具体实施例中,所述R10,R11和R12独立的选自取代或非取代的苯基、吡啶基、哌啶基、哌嗪基或嘧啶基;
上述基团的取代基选自硝基、羟基、巯基、氟、氯、溴、碘、氰基、取代或非取代C1~10烷基、取代或非取代C1~10杂烷基,取代或未取代C3~10环烷基,取代或未取代C3~10杂环烷基。
所述A为取代或非取代的螺环基团或桥环基团,优选为取代或非取代的C6~15的螺环基团或桥环基团。在本发明的某些具体实施例中,所述A为金刚烷基。
在本发明的某些具体实施例中,所述Y为取代或非取代的2-氨基吡啶基苯甲酰胺基团,M为C5-10的螺环或桥环基团,R为乙烯基或丙炔基。在本发明的某些具体实施例中,所述Y的吡啶基团上的H可以任选的被氟、氯、溴、碘、甲基或三氟甲基取代,所述取代可以为邻位取代、间位取代或对位取代。
在本发明的另外一些具体实施例中,所述Y为二苯基乙醇基团,M为C5-10的螺环或桥环基团,R为乙烯基或丙炔基;
在本发明的另外一些具体实施例中,所述Y为二苯醚基团,M为C5-10的螺环或桥环基团,R为乙烯基或丙炔基;
在本发明的另外一些具体实施例中,所述Y为2-氨基吡啶基苯甲酰胺基团,M为C5-10的桥环基团,R为乙烯基;
在本发明的另外一些具体实施例中,所述Y为二苯基乙醇基团,M为C5-10的桥环基团,R为乙烯基;
在本发明的另外一些具体实施例中,所述Y为二苯醚基团,M为C5-10的桥环基团,R为乙烯基;
在本发明的另外一些具体实施例中,所述Y为三氟甲基取代的2-氨基吡啶基苯甲酰胺基团,M为C5-10的桥环基团,R为乙烯基。
在本发明的某些具体实施例中,所述化合物具有以下任一结构或其立体异构体或顺反异构体:
Figure PCTCN2017074108-appb-000015
Figure PCTCN2017074108-appb-000016
Figure PCTCN2017074108-appb-000017
Figure PCTCN2017074108-appb-000018
Figure PCTCN2017074108-appb-000019
Figure PCTCN2017074108-appb-000020
Figure PCTCN2017074108-appb-000021
Figure PCTCN2017074108-appb-000022
Figure PCTCN2017074108-appb-000023
本发明还提供了上述化合物的制备方法,包括以下步骤:
1)以2-氯吡嗪为原料,在碱性化合物作用下,反应生成3-氯-2-吡嗪甲醇;
2)将3-氯-2-吡嗪甲醇通过盖布瑞尔合成法生成(3-氯吡嗪-2-基)甲胺;
3)将(3-氯吡嗪-2-基)甲胺与式(Ⅲ)所示的螺环羧酸或桥环羧酸进行反应,制备得到酰胺类化合物;
4)将酰胺类化合物在三氯氧磷的作用下进行闭环反应,然后经NBS溴代,得到式(Ⅳ)所示化合物;
5)将上述式(Ⅳ)所示化合物在醇和氨水的作用下,发生氨基化反应;
6)将上述氨基化反应后的产物与式(Ⅴ-1)所示硼酸或式(Ⅴ-2)所示硼酸酯通过Suzuki偶联反应,得到式(Ⅵ)所示化合物;
7)将上述式(Ⅵ)所示化合物与取代的或未取代的2-丁炔酸在缩合剂作用下发生缩 合反应,得到式(Ⅰ-1)所示化合物;
或者将上述式(Ⅵ)所示化合物与3-氯丙酰氯或烯丙酰氯在碱的作用下通过直接缩合或者直接缩合后再消除氯化氢烯烃化的方式进行反应,得到式(Ⅰ-2)所示化合物;
或者将上述式(Ⅵ)所示化合物与式(Ⅶ)所示的烯酸类衍生物在缩合剂作用下进行反应,得到式(Ⅰ-3)或式(Ⅰ-4)所示化合物,R"分别为H或氟、氯、溴、碘;
或者将上述式(Ⅵ)所示化合物与氰基乙酸或硝基乙酸在缩合剂的作用下得到酰胺化合物,然后再与式(Ⅷ)所示醛类化合物发生脑文格反应得到式(Ⅰ-4)所示化合物;R"为硝基或氰基;
Figure PCTCN2017074108-appb-000024
Figure PCTCN2017074108-appb-000025
上述Y、M、R'的范围同上,在此不再赘述。
本发明还提供了一种药物组合物,包括上述化合物或其盐或上述制备方法制备的化合物或其盐,以及药学上可接受的载体,赋形剂,稀释剂,辅剂,媒介物或它们的组合。
本发明还提供了上述化合物或上述制备方法制备的化合物或上述药物组合物在制备用于治疗或减轻BTK介导的疾病的药物中的应用。
在本发明的某些具体实施例中,所述BTK介导的疾病选自免疫、自身免疫、炎症疾病、过敏症、感染性疾病、增生性病症和癌症疾病中的任意一种或多种。
在本发明的另外一些具体实施例中,所述BTK介导的疾病选自风湿性关节炎、感染性关节炎、致畸性关节炎、痛风性关节炎、脊椎炎、胰腺炎、慢性支气管炎、急性支气管炎、过敏性支气管炎、毒性支气管炎、儿科哮喘、变应性肺泡炎、过敏性或非过敏性鼻炎、慢性鼻窦炎、囊性纤维化或粘液粘稠病、咳嗽、肺气肿、间质性肺病、肺泡炎、鼻息肉、肺水肿、各种原因的肺炎、红斑狼疮、全身性硬皮病、结节病、亚弥型漫性大B细胞淋巴瘤、套细胞淋巴瘤(MCL)、慢性淋巴细胞淋巴瘤、结外边缘带B细胞淋巴瘤、B细胞慢性淋巴细胞白血病(CLL)、B细胞幼淋巴细胞白血病、成熟B细胞的急性成淋巴细胞性白血病、17p缺失的慢性淋巴细胞白血病、
Figure PCTCN2017074108-appb-000026
巨球蛋白血症、淋巴质浆细胞淋巴瘤、脾脏边缘带淋巴瘤、浆细胞性骨髓瘤、浆细胞瘤、结内边缘带B细胞淋巴瘤、外套细胞淋巴瘤、血管内大B细胞淋巴瘤和原发性渗出性淋巴瘤中的一种或多种。
本发明上述化合物或组合物在应用于制备用于治疗或减轻BTK介导的疾病的药物时,可以单独使用,或与其他药物联合使用。
与现有技术相比,本发明提供了具有式(Ⅰ)所示结构的化合物,或其异构体、药学上可接受的溶剂化物、或其盐,用作布鲁顿酪氨酸激酶抑制剂,其对于BTK具有较高的抑制活性,同时具有较小的不良反应。
具体实施方式
为了进一步说明本发明,下面结合实施例对本发明提供的用作布鲁顿酪氨酸激酶抑制剂的化合物及其制备方法和应用进行详细描述。
下面的缩写具有如下所示的意义:
DMF表示N,N-二甲基甲酰胺;
NBS表示N-溴代丁二酰亚胺;
DCM表示二氯甲烷;
TEA表示三乙胺;
THF表示四氢呋喃;
TFA表示三氟乙酸;
EA表示乙酸乙酯;
PE表示石油醚;
MeOH表示甲醇;
EtOH表示乙醇;
Et2O表示乙醚;
DIEA表示N,N-二异丙基乙胺;
HBTU表示苯并三氮唑-N,N,N',N'-四甲基脲六氟磷酸盐;
TLC表示薄层层析法;
KOAc表示乙酸钾;
X-phos表示2-二环己基磷-2,4,6-三异丙基联苯;
Pd2(dba)3表示三(二亚苄基茚丙酮)二钯;
Pd(pph3)4表示三苯基磷钯;
n-BuLi表示叔丁基锂;
EDCI表示1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐;
DIPEA表示N,N-二异丙基乙胺;
2-BuOH表示异丁醇;
STAB表示三乙酰氧基硼氢化钠;
DMSO表示二甲基亚砜;
FAM表示羧基荧光素;
ATP表示腺嘌呤核苷三磷酸;
MEM表示minimum essential medium;
FBS表示胎牛血清;
IMDM表示Iscove’s Modified Dulbecco’s Media;
PS表示Penicillin-Streptomycin Solution;
RPMI 1640medium表示Roswell Park Memorial Institute 1640培养基;
HEPES表示4-(2-羟乙基)-1-哌嗪乙磺酸;
EGTA表示乙二醇二乙醚二胺四乙酸;
Na-ATP表示腺嘌呤核苷三磷酸钠。
实施例1:(S)-4-(8-氨基-3-(5-(2-丁炔酰基)-5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺的制备
合成步骤如下所示:
Figure PCTCN2017074108-appb-000027
步骤1:3-氯-2-吡嗪甲醇的制备
Figure PCTCN2017074108-appb-000028
氮气保护下,向装有2,2,6,6-四甲基哌啶(72.8g,516.3mmol)的500mL干燥的THF溶液中,在-78℃下慢慢滴加n-BuLi(346mL,553.7mmol,1.6mol/L THF),加毕,反应混合物自然升温至0℃继续搅拌反应20min,然后再把反应混合物降温至-78℃,向反应混合物中逐滴滴加2-氯吡嗪(50g,436.3mmol)的THF(100mL)溶液,30min加完,反应混合物颜色由浅黄色变成深棕色,在-78℃下继续搅拌反应10min。量取DMF(84ml,1092mmol)溶于THF(50mL)逐滴滴加到反应混合物中,控制反应体系温度在-70℃~-78℃,10min加毕,再在-78℃下搅拌反应2h。然后向反应混合物中加入MeOH(800mL),加毕,再向反应混合物中逐份加入NaBH4(33g,868mmol),加毕,待反应混合物自然升高到室温并继续搅拌2h,TLC显示原料反应完全以后,反应液用饱和NH4Cl淬灭,DCM(1L×3)萃取三次,有机相用水洗涤,无水Na2SO4充分干燥,真空蒸发后通过硅胶柱(PE/EA=100/1~5/1)纯化得到60g目标化合物,为黄色油状物。
步骤2:2-((3-氯吡嗪-2-基)甲基)异吲哚啉-1,3-二酮的制备
Figure PCTCN2017074108-appb-000029
在氮气保护下,向装有3-氯-2-吡嗪甲醇(60g,414.9mmol),三苯基膦(130.4g,497.9mmol)和邻苯二甲酰亚胺(73.2g,497.9mmol)的500mL THF溶液中,在-5℃下慢慢滴加DIAD(100.6g,497.9mmol),加毕,该反应混合物在20℃下搅拌3h,TLC显示反应完成后,水加入反应混合物中然后用EA萃取(1L×3),有机相用无水Na2SO4充分干燥,真空蒸发后通过快速色谱法(PE/EA=30/1)纯化得到90.8g目标化合物,为白色固体。
步骤3:(3-氯吡嗪-2-基)甲胺的制备
Figure PCTCN2017074108-appb-000030
在氮气保护下,向装有2-((3-氯吡嗪-2-基)甲基)异吲哚啉-1,3-二酮(90g,328.5mmol)的DCM/MeOH(800mL/400mL)混合溶液中加入水合肼(41.1g,821.5mmol);该反应混合物在室温下搅拌过夜,反应体系中有大量白色固体生成,抽滤,滤饼用乙酸乙酯洗涤,滤液浓缩,再抽滤,滤饼用乙酸乙酯洗涤,滤液用无水Na2SO4充分干燥,真空蒸发后得到38.2g目标化合物,为黄色油状物。
步骤4:(S)-6-(((3-氯吡嗪-2-基)甲基)氨甲酰基)-5-氮杂螺[2.4]庚烷-5-甲酸叔丁酯的制备
Figure PCTCN2017074108-appb-000031
在氮气保护下,向装有(3-氯吡嗪-2-基)甲胺(2.86g,20mmol)、(S)-5-叔丁氧羰基-5-氮杂螺[2.4]庚烷-6-羧酸(4.82g,20mmol)、HOBt(3.51g,26mmol)和TEA(3.64g,36mmol)的30mL DMF溶液中(0℃)逐份加EDCI(4.97g,26mmol),该反应混合物在室温下搅拌反应过夜,TLC显示原料反应完全以后,加水淬灭反应,EA萃取(50mL×3),有机相用饱和食盐水反洗,无水Na2SO4充分干燥,真空蒸发后通过柱层析(PE/EA=5/1~3/1)纯化得到6.4g目标化合物,为黄色固体。
步骤5:(S)-6-(8-氯咪唑并[1,5-a]吡嗪-3-基)-5-氮杂螺[2.4]庚烷-5-甲酸叔丁酯的制备
Figure PCTCN2017074108-appb-000032
冰盐浴下,向装有(S)-6-(((3-氯吡嗪-2-基)甲基)氨甲酰基)-5-氮杂螺[2.4]庚烷-5-甲酸叔丁酯(6.3g,17.18mmol)的DMF/EA(7.5mL/50mL)混合溶液中慢慢滴加POCl3(12.6mL,103.08mmol),加毕,该反应混合物在室温下搅拌2h,TLC显示原料反应完全以后,将反应液慢慢加入Na2CO3(6mol/L)的溶液中,保持pH大于8,分出有机相,水相用EA(20mL x 3)萃取,合并上述有机相用无水Na2SO4充分干燥,真空蒸发后通过柱层析(PE/EA=3/1)纯化得到5.52g目标化合物,为白色固体。
步骤6:(S)-6-(1-溴-8-氯咪唑并[1,5-a]吡嗪苯-3-基)-5-氮杂螺[2.4]庚烷-5-甲酸叔丁酯的制备
Figure PCTCN2017074108-appb-000033
冰盐浴下,向装有(S)-6-(8-氯咪唑并[1,5-a]吡嗪-3-基)-5-氮杂螺[2.4]庚烷-5-甲酸叔丁酯(5.5g,15.77mmol)的50mL DMF溶液中逐份加入NBS(2.95g,16.56mmol),该反应混合物在冰盐浴下搅拌1h,TLC显示原料反应完成以后,将反应液慢慢加入到NaHCO3(1mol/L)溶液中淬灭反应,用EA(20mL×3)萃取,有机相用饱和NaCl洗涤,无水Na2SO4充分干燥,真空蒸发后通过柱层析(PE/EA=5/1)纯化得到6.07g目标化合物,为红棕色固体。
步骤7:(S)-6-(8-胺基-1-溴咪唑并[1,5-a]吡嗪-3-基)-5-氮杂螺[2.4]庚烷-5-甲酸叔丁酯的制备
Figure PCTCN2017074108-appb-000034
室温下,向装有(S)-6-(1-溴-8-氯咪唑并[1,5-a]吡嗪-3-基)-5-氮杂螺[2.4]庚烷-5-甲酸叔丁酯(5.5g,12.86mmol)的高压反应釜中加入15mL 2-BuOH和30mL氨水,该反应混合物在90℃搅拌反应15h,TLC显示原料反应完全以后,将反应液真空浓缩得到固体粗产品,用EA/PE(5/1)打浆得纯品3.94g目标化合物,为浅黄色固体。
步骤8:(S)-6-(8-氨基-1-(4-(吡啶-2-基氨甲酰基)苯基)咪唑并[1,5-a]吡嗪-3-基)-5-氮杂螺[2.4]庚烷-5-甲酸叔丁酯的制备
Figure PCTCN2017074108-appb-000035
氮气保护下,向装有2-氨基吡啶(5g,53.19mmol)、4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)苯甲酸(13.20g,53.19mmol)、DIPEA(13.72g,106.38mmol)的50mL DMF(0℃)溶液中,逐份加入HBTU(26.21g,,69.15mmol),该反应混合物在室温下搅拌反应过夜,TLC显示原料反应完全以后,向反应溶液中加入水淬灭,用EA(30mL×3)萃取,有机相用饱和食盐水反洗,无水Na2SO4干燥,真空蒸发后通过柱层析(PE/EA=20/1)纯化得到13.1g目标化合物,为类白色固体。
Figure PCTCN2017074108-appb-000036
氮气保护下,向装有(S)-6-(8-氨基-1-溴咪唑并[1,5-a]吡嗪-3-基)-5-氮杂螺[2.4]庚烷-5-甲酸叔丁酯(3.5g,8.57mmol)、N-(吡啶-2-基)-4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)苯甲酰胺(3.33g,10.28mmol)、Na2CO3(1.82g,17.14mmol)的dioxane/EtOH/water(36mL/12mL/12mL)混合溶液中,加入Pd(PPh3)4(496.89mg,,0.43mmol),该反应混合物在90℃下搅拌反应过夜,TLC显示原料反应完全以后,向反应溶液中加入水淬灭,用EA(40mL×3)萃取,有机相用饱和食盐水反洗,无水Na2SO4干燥,真空蒸发后通过柱层析(DCM/MeOH=60/1)纯化得到2.71g目标化合物,为淡黄色固体。
步骤9:(S)-4-(8-氨基-3-(5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基) 苯甲酰胺的制备
Figure PCTCN2017074108-appb-000037
向装有(S)-6-(8-氨基-1-(4-(吡啶-2-基氨甲酰基)苯基)咪唑并[1,5-a]吡嗪-3-基)-5-氮杂螺[2.4]庚烷-5-甲酸叔丁酯(2.71g,5.16mmol)的DCM(20mL)溶液中,加入TFA(3mL),该反应混合物在室温下搅拌反应过夜,TLC显示原料反应完全以后,反应体系浓缩,用Na2CO3(3mol/L)调pH至8,用DCM/MeOH(10/1)萃取,有机相无水Na2SO4干燥,真空蒸发后通过柱层析(DCM/MeOH=60/1~10/1)纯化得到1.98g目标化合物,为白色固体。
步骤10:a(S)-4-(8-氨基-3-(5-(2-丁炔酰基)-5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺的制备
Figure PCTCN2017074108-appb-000038
氮气保护下,向装有(S)-4-(8-氨基-3-(5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺(90mg,0.212mmol)、2-丁炔酸(19.6mg,0.233mmol)、DIPEA(82mg,0.636mmol)的DMF(3mL)溶液中,加入HBTU(96.4mg,0.254mmol),该反应混合物在室温下搅拌反应1h,TLC显示原料反应完全以后,向反应体系中加水淬灭,用EA(10mL×3)萃取,有机相饱和食盐水反洗,无水Na2SO4干燥,真空蒸发后通过制备硅胶板(DCM/MeOH=20/1)纯化得到55mg目标化合物,为黄色固体。
采用核磁共振以及质谱对产物结构进行表征,结果如下:
1H NMR(400MHz,d6-DMSO)δ0.58-0.77(4H,m),1.58(1H,s),1.99(2H,s),2.24-2.33(2H,m),3.51(0.4H,d,J=16.4Hz),3.61(0.4H,d,J=11.6Hz),3.61(0.6H,d,J=10.8Hz),3.84(0.6H,d,J=10.8Hz),5.56-5.59(0.6H,m),5.79-5.82(0.4H,m),6.14-6.20(2H,m),7.11-7.20 (2H,m),7.73-7.79(2.6H,m),7.84-7.89(1.4H,m),8.16(2H,dd,J=8.4Hz,2.8Hz),8.23(1H,d,J=8.4Hz),8.41(1H,dd,J=4.8Hz,1.2Hz),10.85(1H,s).
EM(计算值):491.2;MS(ESI)m/e(M+1H)+:492.2。
可见本申请制备得到的化合物与上述反应式中的化合物结构一致。
实施例2:(S)-4-(3-(5-丙烯酰基-5-氮杂螺[2.4]庚烷-6-基)-8-氨基咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺的制备
Figure PCTCN2017074108-appb-000039
氮气保护下,向装有(S)-4-(8-氨基-3-(5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺(120mg,0.28mmol)、TEA(113.12mg,1.12mmol)的DCM(10mL)溶液中,0℃下逐滴加入3-氯丙酰氯(35.6mg,0.28mmol),该反应混合物在室温下搅拌反应过夜,TLC显示原料反应完全以后,向反应体系中加水淬灭,用EA(10mL×3)萃取,有机相无水Na2SO4干燥,真空蒸发后通过制备硅胶板(DCM/MeOH=15/1)纯化得到40mg目标化合物,为黄色固体。
采用核磁共振以及质谱对产物结构进行表征,结果如下:
1H NMR(400MHz,d6-DMSO)δ0.56-0.76(4H,m),2.24-2.26(1.5H,m),3.61-3.66(1.5H,m),3.87(1H,d,J=10.4Hz),5.60-5.68(2H,m),6.05-6.20(3H,m),6.55(1H,dd,J=16.8Hz,10.4Hz),7.11-7.20(2H,m),7.66-7.74(2H,m),7.83-7.88(2H,m),8.15(2H,d,J=8.4Hz),8.22(1H,d,J=8.4Hz),8.41(1H,d,J=3.6Hz),10.84(1H,s).
EM(计算值):479.2;MS(ESI)m/e(M+1H)+:480.2。
可见本申请制备得到的化合物与上述化合物结构一致。
实施例3~21
采用以下化合物为原料,以实施例1或实施例2的制备方法,制备得到以下化合物,所述化合物的结构以及核磁表征数据见表1,表1是本申请实施例3~21制备的化合物结构以及结构分析数据汇总。
Figure PCTCN2017074108-appb-000040
根据文献Tetrahedron Letters 57(2016)599–602合成制备;
Figure PCTCN2017074108-appb-000041
根据文献Tetrahedron Letters 57(2016)599–602合成制备;
Figure PCTCN2017074108-appb-000042
根据文献Bioorganic&Medicinal Chemistry Letters 14(2004)6107-6111合成制备;
Figure PCTCN2017074108-appb-000043
根据专利WO2014/140081A1合成制备;
Figure PCTCN2017074108-appb-000044
参照专利WO2011/35332合成制备;
Figure PCTCN2017074108-appb-000045
参照文献Journal of Organic Chemistry;vol.59;nb.2;(1994);p.276–277合成制备;
其他关键中间体片断均通过直接采购或定制合成。
表1 实施例3~21制备的化合物结构以及结构分析数据
Figure PCTCN2017074108-appb-000046
Figure PCTCN2017074108-appb-000047
Figure PCTCN2017074108-appb-000048
Figure PCTCN2017074108-appb-000049
Figure PCTCN2017074108-appb-000050
实施例22:4-(3-((1R,3S,4S)-2-丙烯酰基-2-氮杂双环[2.2.1]庚烷-3-基)-8-氨基咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺的制备
合成步骤如下所示:
Figure PCTCN2017074108-appb-000051
步骤1:(1R,3R,4S)-3-(((3-氯吡嗪-2-基)甲基)胺甲酰基)-2-氮杂双环[2.2.1]庚烷-2-甲酸叔丁酯的制备
Figure PCTCN2017074108-appb-000052
在氮气保护下,向装有(3-氯吡嗪-2-基)甲胺(3.43g,24mmol)、(1R,3R,4S)-2-(叔丁氧羰基)-2-氮杂双环[2.2.1]庚烷-3-羧酸(5.80g,20mmol)、HOBt(4.21g,31.2mmol)和TEA(4.37g,43.2mmol)的30mL DMF溶液中(0℃)逐份加EDCI(5.97g,31.2mmol),该反应混合物在室温下搅拌反应过夜,TLC显示原料反应完全以后,加水淬灭反应,EA萃取(50mL×3),有机相用饱和食盐水反洗,无水Na2SO4充分干燥,真空蒸发后通过柱层析(PE/EA=5/1~3/1)纯化得到8.0g目标化合物,为棕色固体。
步骤2:(1R,3S,4S)-3-(8-氯咪唑并[1,5-a]吡嗪-3-基)-2-氮杂双环[2.2.1]庚烷-2-甲酸叔丁 酯的制备
Figure PCTCN2017074108-appb-000053
冰盐浴下,向装有(1R,3R,4S)-3-(((3-氯吡嗪-2-基)甲基)胺甲酰基)-2-氮杂双环[2.2.1]庚烷-2-甲酸叔丁酯(6.3g,17.18mmol)的DMF/EA(7.5mL/50mL)混合溶液中慢慢滴加POCl3(12.6mL,103.08mmol),加毕,该反应混合物在室温下搅拌2h,TLC显示原料反应完全以后,将反应液慢慢加入Na2CO3(6mol/L)的溶液中,保持pH大于8,分出有机相,水相用EA(20mL x 3)萃取,合并上述有机相用无水Na2SO4充分干燥,真空蒸发后通过柱层析(PE/EA=3/1)纯化得到5.6g目标化合物,为黄色固体。
步骤3:(1R,3S,4S)-3-(1-溴-8-氯咪唑并[1,5-a]吡嗪-3-基)-2-氮杂双环[2.2.1]庚烷-2-甲酸叔丁酯的制备
Figure PCTCN2017074108-appb-000054
冰盐浴下,向装有(1R,3S,4S)-3-(8-氯咪唑并[1,5-a]吡嗪-3-基)-2-氮杂双环[2.2.1]庚烷-2-甲酸叔丁酯(4.95g,14.19mmol)的50mL DMF溶液中逐份加入NBS(2.66g,14.9mmol),该反应混合物在冰盐浴下搅拌1h,TLC显示原料反应完成以后,将反应液慢慢加入到NaHCO3(1mol/L)溶液中淬灭反应,用EA(20mL×3)萃取,有机相用饱和NaCl洗涤,无水Na2SO4充分干燥,真空蒸发后通过柱层析(PE/EA=5/1)纯化得到5.2g目标化合物,为淡黄色固体。
步骤4:(1R,3S,4S)-3-(8-氨基-1-溴咪唑并[1,5-a]吡嗪-3-基)-2-氮杂双环[2.2.1]庚烷-2-甲酸叔丁酯的制备
Figure PCTCN2017074108-appb-000055
室温下,向装有(1R,3S,4S)-3-(1-溴-8-氯咪唑并[1,5-a]吡嗪-3-基)-2-氮杂双环[2.2.1]庚烷-2-甲酸叔丁酯(4.4g,10.29mmol)的高压反应釜中加入15mL 2-BuOH和30mL氨水,该反应混合物在90℃搅拌反应15h,TLC显示原料反应完全以后,将反应液真空浓缩得到固体粗产品,用EA/PE(5/1)打浆得纯品3.2g目标化合物,为淡黄色固体。
步骤5:(1R,3S,4S)-3-(8-氨基-1-(4-(吡啶-2-基氨甲酰基)苯基)咪唑并[1,5-a]吡嗪-3-基)-2-氮杂双环[2.2.1]庚烷-2-甲酸叔丁酯的制备
Figure PCTCN2017074108-appb-000056
氮气保护下,向装有(1R,3S,4S)-3-(8-氨基-1-溴咪唑并[1,5-a]吡嗪-3-基)-2-氮杂双环[2.2.1]庚烷-2-甲酸叔丁酯(3.5g,8.57mmol)、N-(吡啶-2-基)-4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)苯甲酰胺(3.33g,10.28mmol)、Na2CO3(1.82g,17.14mmol)的dioxane/EtOH/water(36mL/12mL/12mL)混合溶液中,加入Pd(PPh3)4(496.89mg,,0.43mmol),该反应混合物在90℃下搅拌反应过夜,TLC显示原料反应完全以后,向反应溶液中加入水淬灭,用EA(40mL×3)萃取,有机相用饱和食盐水反洗,无水Na2SO4干燥,真空蒸发后通过柱层析(DCM/MeOH=60/1)纯化得到2.8g目标化合物,为淡黄色固体。
步骤6:4-(8-氨基-3-((1R,3S,4S)-2-氮杂双环[2.2.1]庚烷-3-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺的制备
Figure PCTCN2017074108-appb-000057
向装有(1R,3S,4S)-3-(8-氨基-1-(4-(吡啶-2-基氨甲酰基)苯基)咪唑并[1,5-a]吡嗪-3-基)-2- 氮杂双环[2.2.1]庚烷-2-甲酸叔丁酯(2.98g,5.68mmol)的DCM(20mL)溶液中,加入TFA(3.5mL),该反应混合物在室温下搅拌反应过夜,TLC显示原料反应完全以后,反应体系浓缩,用Na2CO3(3mol/L)调pH至8,用DCM/MeOH(10/1)萃取,有机相无水Na2SO4干燥,真空蒸发后通过柱层析(DCM/MeOH=60/1~10/1)纯化得到2.0g目标化合物,为白色固体。
步骤7:4-(3-((1R,3S,4S)-2-丙烯酰基-2-氮杂双环[2.2.1]庚烷-3-基)-8-氨基咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺的制备
Figure PCTCN2017074108-appb-000058
氮气保护下,向装有4-(8-氨基-3-((1R,3S,4S)-2-氮杂双环[2.2.1]庚烷-3-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺(180mg,0.42mmol)、TEA(170mg,1.68mmol)的DCM(10mL)溶液中,0℃下逐滴加入3-氯丙酰氯(53.4mg,0.42mmol),该反应混合物在室温下搅拌反应过夜,TLC显示原料反应完全以后,向反应体系中加水淬灭,用EA(10mL×3)萃取,有机相无水Na2SO4干燥,真空蒸发后通过制备硅胶板(DCM/MeOH=15/1)纯化得到48mg目标化合物,为黄色固体。
采用核磁共振以及质谱对产物结构进行表征,结果如下:
1H NMR(400MHz,d6-DMSO)δ1.49-1.55(2H,m),1.74-1.80(3H,m),2.57-2.58(1H,m),2.67-2.73(1H,m),4.66(0.2H,s),4.74(0.8H,s),5.04(0.2H,s),5.25(0.8H,s),5.44-5.47(0.2H,m),5.65-5.68(0.8H,m),5.85(0.2H,dd,J=16.8Hz,10.4Hz),6.06-6.13(3H,m),6.76(0.8H,dd,J=16.8Hz,10.4Hz),7.13(1H,d,J=5.2Hz),7.16-7.20(1H,m),7.71(2H,dd,J=8.4Hz,4.0Hz),7.82-7.88(1H,m),7.92(1H,d,J=5.2Hz),8.15(2H,d,J=8.4Hz),8.22(1H,d,J=8.4Hz),8.40-8.41(1H,m),10.84-10.85(1H,m).
EM(计算值):479.2;MS(ESI)m/e(M+1H)+:480.2
可见本申请制备得到的化合物与上述反应式中的化合物结构一致。
实施例23:4-(8-氨基-3-((1R,3S,4S)-2-(2-丁炔酰基)-2-氮杂双环[2.2.1]庚烷-3-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺的制备
Figure PCTCN2017074108-appb-000059
氮气保护下,向装有4-(8-氨基-3-((1R,3S,4S)-2-氮杂双环[2.2.1]庚烷-3-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺(180mg,0.424mmol)、2-丁炔酸(39.2mg,0.466mmol)、DIPEA(164mg,1.27mmol)的DMF(10mL)溶液中,加入HBTU(192.8mg,0.51mmol),该反应混合物在室温下搅拌反应1h,TLC显示原料反应完全以后,向反应体系中加水淬灭,用EA(10mL×3)萃取,有机相饱和食盐水反洗,无水Na2SO4干燥,真空蒸发后通过制备硅胶板(DCM/MeOH=20/1)纯化得到95mg目标化合物,为黄色固体。
采用核磁共振以及质谱对产物结构进行表征,结果如下:
1H NMR(400MHz,d6-DMSO)δ1.46-1.50(2H,m),1.67-1.86(4H,m),2.03(2H,s),2.60-2.63(2H,m),4.56-4.63(1H,m),5.01-5.21(1H,m),6.15-6.20(2H,m),7.12-7.20(2H,m),7.71-7.76(2H,m),7.84-7.88(2H,m),8.15-8.18(2H,m),8.22(1H,d,J=8.4Hz),8.40-8.42(1H,m),10.85-10.86(1H,m).
EM(计算值):491.2;MS(ESI)m/e(M+1H)+:492.2。
可见本申请制备得到的化合物与上述反应式中的化合物结构一致。
实施例24~39
以实施例22或实施例23的制备方法,制备得到以下化合物,所述化合物的结构以及核磁表征数据见表2,表2是本申请实施例24~39制备的化合物结构以及结构分析数据汇总。
表2 实施例24~39制备的化合物结构以及结构分析数据
Figure PCTCN2017074108-appb-000060
Figure PCTCN2017074108-appb-000061
Figure PCTCN2017074108-appb-000062
Figure PCTCN2017074108-appb-000063
Figure PCTCN2017074108-appb-000064
实施例40
(S)-4-(8-氨基-3-(5-(2-丁炔酰基)-5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-1-基l)-N-(4-(三氟甲基)哌啶-2-基)苯甲酰胺的制备
合成步骤如下所示:
Figure PCTCN2017074108-appb-000065
步骤1:4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺的制备
Figure PCTCN2017074108-appb-000066
氮气保护下,向装有4-(三氟甲基)吡啶-2-胺(4g,24.69mmol)、4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)苯甲酸(6.13g,24.69mmol)、DIPEA(6.37g,49.38mmol)的30mLDMF(0℃)溶液中,逐份加入HBTU(11.23g,,29.63mmol),该反应混合物在室温下搅拌反应过夜,TLC显示原料反应完全以后,向反应溶液中加入水淬灭,用EA(20mL×3)萃取,有机相用饱和食盐水反洗,无水Na2SO4干燥,真空蒸发后通过柱层析(PE/EA=20/1)纯化得到7.75g目标化合物,为类白色固体。
步骤2:(S)-6-(8-氨基-1-(4-((4-(三氟甲基)吡啶-2-基)氨甲酰基)苯基)咪唑并[1,5-a]吡嗪-3-基)-5-氮杂螺[2.4]庚烷-5-甲酸叔丁酯制备
Figure PCTCN2017074108-appb-000067
氮气保护下,向装有(S)-6-(8-氨基-1-溴咪唑并[1,5-a]吡嗪-3-基)-5-氮杂螺[2.4]庚烷-5-甲酸叔丁酯(3.5g,8.57mmol)、4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺(4.03g,10.28mmol)、Na2CO3(1.82g,17.14mmol)的dioxane/EtOH/water(36mL/12mL/12mL)混合溶液中,加入Pd(PPh3)4(496.89mg,,0.43mmol),该反应混合物在90℃下搅拌反应过夜,TLC显示原料反应完成以后,向反应溶液中加入水淬灭,用EA(40mL×3)萃取,有机相用饱和食盐水反洗,无水Na2SO4干燥,真空蒸发后通过柱层析(DCM/MeOH=60/1)纯化得到3.2g目标化合物,为淡黄色固体。
步骤3:(S)-4-(8-氨基-3-(5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺制备
Figure PCTCN2017074108-appb-000068
向装有(S)-6-(8-氨基-1-(4-((4-(三氟甲基)吡啶-2-基)氨甲酰基)苯基)咪唑并[1,5-a]吡嗪-3-基)-5-氮杂螺[2.4]庚烷-5-甲酸叔丁酯(3.2g,5.39mmol)的DCM(20mL)溶液中,加入TFA(3mL),该反应混合物在室温下搅拌反应过夜,TLC显示原料反应完成以后,反应体系浓缩,用Na2CO3(3mol/L)调pH至8,用DCM/MeOH(10/1)萃取,有机相无水Na2SO4干燥,真空蒸发后通过柱层析(DCM/MeOH=60/1~10/1)纯化得到2.53g目标化合物,为白色固体。
步骤4:(S)-4-(8-氨基-3-(5-(2-丁炔酰基)-5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-1-基l)-N-(4-(三氟甲基)哌啶-2-基)苯甲酰胺的制备
Figure PCTCN2017074108-appb-000069
氮气保护下,向装有(S)-4-(8-氨基-3-(5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺(100mg,0.203mmol)、2-丁炔酸(20.5mg,0.244mmol)、DIPEA(78.56mg,0.609mmol)的DMF(5mL)溶液中,加入HBTU(92.5mg,0.244mmol),该反应混合物在室温下搅拌反应1h,TLC显示原料反应完成以后,向反应体系中加水淬灭,用EA(15mL×3)萃取,有机相饱和食盐水反洗,无水Na2SO4干燥,真空蒸发后通过制备硅胶板(DCM/MeOH=20/1)纯化得到60mg目标化合物,为黄色固体。
采用核磁共振以及质谱对产物结构进行表征,结果如下:
1H NMR(400MHz,d6-DMSO)δ0.58-0.79(4H,m),1.58(1H,s),1.97(2H,s),2.24-2.33(2H,m),3.50-3.85(2H,m),5.57-5.59(0.65H,m),5.80-5.82(0.35H,m),6.14-6.22(2H,m),7.14 (0.65H,d,J=4.8Hz),7.18(0.35H,d,J=4.8Hz),7.58(1H,d,J=4.8Hz),7.74-7.79(2H,m),7.89-8.04(1H,m),8.17-8.20(2H,m),8.62(1H,s),8.70(1H,d,J=5.2Hz),10.82(1H,s).
EM(计算值):559.2;MS(ESI)m/e(M+1H)+:560.2
可见本申请制备得到的化合物与上述化合物结构一致。
实施例41
(S)-4-(3-(5-丙烯酰基l-5-氮杂螺[2.4]庚烷-6-基)-8-氨基咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺的制备
Figure PCTCN2017074108-appb-000070
氮气保护下,向装有(S)-4-(8-氨基-3-(5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-1-基)-N-(4-(三氟甲基)吡啶-2-基)苯甲酰胺(100mg,0.203mmol)、TEA(103.1mg,1.02mmol)的DCM(10mL)溶液中,0℃下逐滴加入3-氯丙酰氯(25.8mg,0.203mmol),该反应混合物在室温下搅拌反应过夜,TLC显示原料反应完成以后,向反应体系中加水淬灭,用EA(10mL×3)萃取,有机相无水Na2SO4干燥,真空蒸发后通过制备硅胶板(DCM/MeOH=15/1)纯化得到32mg目标化合物,为黄色固体。
采用核磁共振以及质谱对产物结构进行表征,结果如下:
1H NMR(400MHz,d6-DMSO)δ0.56-0.76(4H,m),2.24-2.28(1.5H,m),3.61-3.66(1.5H,m),3.87(1H,d,J=10.4Hz),5.60-5.66(2H,m),6.05-6.19(3H,m),6.57(1H,dd,J=16.4Hz,10.4Hz),7.15(0.7H,d,J=5.2Hz),7.19(0.3H,d,J=5.2Hz),7.52-7.56(1H,m),7.76(2H,dd,J=8.0Hz,4.0Hz),7.83(0.7H,d,J=5.2Hz),7.97(0.3H,d,J=5.2Hz),8.17(2H,d,J=8.0Hz),8.57(1H,s),8.71(1H,d,J=5.2Hz),11.12(1H,s).
EM(计算值):547.2;MS(ESI)m/e(M+1H)+:548.2
可见本申请制备得到的化合物与上述化合物结构一致。
实施例42~72
采用类似结构化合物为原料,以实施例40或实施例41的制备方法,制备得到以下化 合物,所述化合物的结构以及核磁表征数据见表3,表3是本申请实施例42~72制备的化合物结构以及结构分析数据汇总。
表3 实施例42~72制备的化合物结构以及结构分析数据汇总
Figure PCTCN2017074108-appb-000071
Figure PCTCN2017074108-appb-000072
Figure PCTCN2017074108-appb-000073
Figure PCTCN2017074108-appb-000074
Figure PCTCN2017074108-appb-000075
Figure PCTCN2017074108-appb-000076
Figure PCTCN2017074108-appb-000077
Figure PCTCN2017074108-appb-000078
实施例73
1-((6S)-6-(8-氨基-1-(4-(1羟基-1-苯乙基)苯基)咪唑并[1,5-a]吡嗪-3-基)-5-氮杂螺[2.4]庚烷-5-基)2-丁炔酰基-1-酮的制备
合成步骤如下所示:
Figure PCTCN2017074108-appb-000079
步骤1:1-(4-溴苯基)-1-苯乙醇的制备
Figure PCTCN2017074108-appb-000080
氮气保护中,在-45℃下,向装有4-溴苯乙酮(5g,25.13mmol)的30mL THF溶液中逐滴加入苯基溴化镁(10.05mL,30.15mmol,3M in Et2O),该反应混合物在-45℃下继续搅拌反应1h,TLC显示原料反应完全,慢慢加入饱和NH4Cl萃灭反应,继续搅拌反应0.5h。水相用EA萃取(30mLx3),合并有机相用饱和食盐水反洗,无水Na2SO4充分干燥,真空蒸发后通过柱层析(PE/EA=60/1~10/1)纯化得到5.8g目标化合物。
步骤2:1-苯基-1-(4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)苯基)乙醇的制备
Figure PCTCN2017074108-appb-000081
氮气保护下,向装有1-(4-溴苯基)-1-苯乙醇(5.2g,18.76mmol)、频哪醇硼酸酯(6.19g, 24.39mmol)、KOAc(3.68g,37.52mmol)和X-Phos(894.2mg,1.876mmol)的30mL dioxane溶液中,加入Pd2(dba)3(858.9mg,,0.938mmol),该反应混合物在90℃下搅拌反应过夜,TLC显示原料反应完成以后,向反应溶液中加入水淬灭,用EA(20mLX3)萃取,有机相用饱和食盐水反洗,无水Na2SO4干燥,真空蒸发后通过柱层析(PE/EA=60/1~10/1)纯化得到4.8g目标化合物,为类白色固体。
步骤3:(6S)-6-(8-氨基-1-(4-(1-羟基-1-苯乙基)苯基)咪唑并[1,5-a]吡嗪-3-基)-5-氮杂螺[2.4]庚烷-5-甲酸叔丁酯的制备
Figure PCTCN2017074108-appb-000082
氮气保护下,向装有(S)-6-(8-氨基-1-溴咪唑并[1,5-a]吡嗪-3-基)-5-氮杂螺[2.4]庚烷-5-甲酸叔丁酯(1g,2.45mmol)、1-苯基-1-(4-(4,4,5,5-四甲基-1,3,2-二氧硼戊环-2-基)苯基)乙醇(872.2mg,2.69mmol)、Na2CO3(519.4mg,4.9mmol)的dioxane/EtOH/water(12mL/4mL/4mL)混合溶液中,加入Pd(PPh3)4(141.56mg,,0.1225mmol),该反应混合物在90℃下搅拌反应过夜,TLC显示原料反应完全以后,向反应溶液中加入水淬灭,用EA(10mL×3)萃取,有机相用饱和食盐水反洗,无水Na2SO4干燥,真空蒸发后通过柱层析(DCM/MeOH=60/1~30/1)纯化得到837.1mg目标化合物,为淡黄色固体。
步骤4:1-(4-(8-氨基-3-((S)-5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-1-基)苯基)-1-苯基乙醇的制备
Figure PCTCN2017074108-appb-000083
在室温下,向装有(6S)-6-(8-氨基-1-(4-(1-羟基-1-苯乙基)苯基)咪唑并[1,5-a]吡嗪-3-基)-5-氮杂螺[2.4]庚烷-5-甲酸叔丁酯(837mg,1.59mmol)的15mL DCM溶液中慢慢滴加TFA(1mL),该反应混合物在室温下搅拌3h,TLC显示原料反应完成以后,反应体系浓缩,用Na2CO3(3mol/L)调pH至8,用DCM/MeOH(10/1)萃取,有机相无水Na2SO4干燥,真空蒸发后得到642.8mg目标化合物,为白色固体。
步骤5:1-((6S)-6-(8-氨基-1-(4-(1羟基-1-苯乙基)苯基)咪唑并[1,5-a]吡嗪-3-基)-5-氮杂螺[2.4]庚烷-5-基)2-丁炔酰基-1-酮的制备
Figure PCTCN2017074108-appb-000084
氮气保护下,向装1-(4-(8-氨基-3-((S)-5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-1-基)苯基)-1-苯基乙醇(80mg,0.188mmol)、2-丁炔酸(19mg,0.226mmol)、DIPEA(72.8mg,0.564mmol)的DMF(3mL)溶液中,加入HBTU(85.7mg,0.226mmol),该反应混合物在室温下搅拌反应2h,TLC显示原料反应完全以后,向反应体系中加水淬灭,用EA(15mL×3)萃取,有机相饱和食盐水反洗,无水Na2SO4干燥,真空蒸发后通过制备硅胶板(DCM/MeOH=15/1)纯化得到20mg目标化合物,为黄色固体。
采用核磁共振以及质谱对产物结构进行表征,结果如下:
1H NMR(400MHz,d6-DMSO)δ0.55-0.73(4H,m),1.58(1H,s),1.90(3H,s),2.01(2H,s),2.25-2.32(2H,m),3.52(0.3H,d,J=16.4Hz),3.62-3.65(0.3H,m),3.73(0.7H,d,J=10.8Hz),3.82(0.7H,d,J=10.8Hz),5.57-5.59(1H,m),5.71(1H,s),6.10-6.20(2H,m),7.08(1H,d,J=4.8Hz),7.12-7.18(1H,m),7.27-7.33(2H,m),7.44-7.55(6H,m),7.78(0.3H,d,J=4.8Hz),7.86(0.7H,d,J=4.8Hz).
EM(计算值):491.2;MS(ESI)m/e(M+1H)+:492.2
可见本申请制备得到的化合物与上述化合物结构一致。
实施例74
1-((6S)-6-(8-氨基-1-(4-(1-羟基-1-苯乙基)苯基)咪唑并[1,5-a]吡嗪-3-羟基)-5-氮杂螺[2.4]庚烷-5-基)-2-丙烯-1-酮的制备
Figure PCTCN2017074108-appb-000085
氮气保护下,向装有1-(4-(8-氨基-3-((S)-5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-1-基)苯基)-1-苯基乙醇(80mg,0.188mmol)、TEA(94.9mg,0.94mmol)的DCM(10mL)溶液中,0℃下逐滴加入3-氯丙酰氯(23.9mg,0.188mmol),该反应混合物在室温下搅拌反应过夜,TLC显示原料反应完全以后,向反应体系中加水淬灭,用EA(10mL×3)萃取,有机相无水Na2SO4干燥,真空蒸发后通过制备硅胶板(DCM/MeOH=15/1)纯化得到15mg目标化合物,为黄色固体。
采用核磁共振以及质谱对产物结构进行表征,结果如下:
1H NMR(400MHz,d6-DMSO)δ0.58-0.73(4H,m),1.91(3H,s),2.22-2.32(2H,m),3.50-3.67(0.6H,m),3.75-3.80(1.4H,d,J=10.8Hz),5.57-5.60(1H,m),5.65-5.67(1H,m),5.71(1H,s),6.08-6.20(3H,m),6.76(1H,dd,J=16.8Hz,10.4Hz),7.06(1H,d,J=4.8Hz),7.10-7.18(1H,m),7.28-7.33(2H,m),7.46-7.55(6H,m),7.77(0.4H,d,J=4.8Hz),7.86(0.6H,d,J=4.8Hz).
EM(计算值):479.2;MS(ESI)m/e(M+1H)+:480.2
可见本申请制备得到的化合物与上述化合物结构一致。
实施例75~91
采用类似结构化合物为原料,以实施例73或实施例74的制备方法,制备得到以下化合物,所述化合物的结构以及核磁表征数据见表4,表4是本申请实施例75~91制备的化合物结构以及结构分析数据汇总。
表4 实施例75~91制备的化合物结构以及结构分析数据汇总
Figure PCTCN2017074108-appb-000086
Figure PCTCN2017074108-appb-000087
Figure PCTCN2017074108-appb-000088
Figure PCTCN2017074108-appb-000089
Figure PCTCN2017074108-appb-000090
实施例92
(S)-1-(6-(8氨基-1-(4-苯氧基苯基)咪唑并[1,5-a]吡嗪-3-基)-5-氮杂螺[2.4]庚烷-5-基)-2-丁炔-1-酮的制备
合成步骤如下所示:
Figure PCTCN2017074108-appb-000091
步骤1:(S)-6-(8-氨基-1-(4-苯氧基苯基)咪唑并[1,5-a]吡嗪-3-基)-5-氮杂螺[2.4]庚烷-5-甲酸叔丁酯的制备
Figure PCTCN2017074108-appb-000092
氮气保护下,向装有(S)-6-(8-氨基-1-溴咪唑并[1,5-a]吡嗪-3-基)-5-氮杂螺[2.4]庚烷-5-甲酸叔丁酯(1g,2.45mmol)、4,4,5,5-四甲基-2-(4-苯氧基苯基)-1,3,2-二氧硼戊环(796.7mg,2.69mmol)、Na2CO3(519.4mg,4.9mmol)的dioxane/EtOH/water(12mL/4mL/4mL)混合溶液中,加入Pd(PPh3)4(141.56mg,,0.1225mmol),该反应混合物在90℃下搅拌反应3h,TLC显示原料反应完全以后,向反应溶液中加入水淬灭,用EA(10mL×3)萃取,有机相用饱和食盐水反洗,无水Na2SO4干燥,真空蒸发后通过柱层析(DCM/MeOH=60/1~30/1)纯化得到865.6mg目标化合物,为淡黄色固体。
步骤2:(S)-1-(4-苯氧基苯基)-3-(5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-8-胺的制备
Figure PCTCN2017074108-appb-000093
在室温下,向装有(S)-6-(8-氨基-1-(4-苯氧基苯基)咪唑并[1,5-a]吡嗪-3-基)-5-氮杂螺[2.4]庚烷-5-甲酸叔丁酯(865mg,1.74mmol)的15mL DCM溶液中慢慢滴加TFA(1mL),该反应混合物在室温下搅拌3h,TLC显示原料反应完成以后,反应体系浓缩,用Na2CO3(3mol/L)调pH至8,用DCM/MeOH(10/1)萃取,有机相无水Na2SO4干燥,真空蒸发后得到690mg目标化合物,为白色固体。
步骤3:(S)-1-(6-(8氨基-1-(4-苯氧基苯基)咪唑并[1,5-a]吡嗪-3-基)-5-氮杂螺[2.4]庚烷-5-基)-2-丁炔-1-酮的制备
Figure PCTCN2017074108-appb-000094
氮气保护下,向装(S)-1-(4-苯氧基苯基)-3-(5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-8-胺(80mg,0.20mmol)、2-丁炔酸(20.2mg,0.24mmol)、DIPEA(77.4mg,0.60mmol)的DMF(3mL)溶液中,加入HBTU(91mg,0.24mmol),该反应混合物在室温下搅拌反应2h,TLC显示原料反应完成以后,向反应体系中加水淬灭,用EA(15mL×3)萃取,有机相饱和食盐水反洗,无水Na2SO4干燥,真空蒸发后通过制备硅胶板(DCM/MeOH=25/1)纯化得到30mg目标化合物,为白色固体。
采用核磁共振以及质谱对产物结构进行表征,结果如下:
1H NMR(400MHz,d6-DMSO)δ0.55-0.76(4H,m),1.58(1H,s),2.00(2H,s),2.26-2.33 (2H,m),3.51(0.4H,d,J=16.4Hz),3.63(0.4H,d,J=11.6Hz),3.70(0.6H,d,J=10.8Hz),3.84(0.6H,d,J=10.8Hz),5.57-5.59(0.6H,m),5.77-5.82(0.4H,m),6.14-6.21(2H,m),7.03-7.19(6H,m),7.41-7.44(2H,m),7.57-7.61(2H,m),7.82(0.6H,d,J=5.2Hz),7.97(0.4H,d,J=5.2Hz).
EM(计算值):463.2;MS(ESI)m/e(M+1H)+:464.2
可见本申请制备得到的化合物与上述化合物结构一致。
实施例93
(S)-1-(6-(8-氨基-1-(4-苯氧基苯基)咪唑并[1,5-a]吡嗪-3-基)-5-氮杂螺[2.4]庚烷-5-基)-2-丙烯-1-酮的制备
Figure PCTCN2017074108-appb-000095
氮气保护下,向装有(S)-1-(4-苯氧基苯基)-3-(5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-8-胺(80mg,0.20mmol)、TEA(101mg,1.0mmol)的DCM(10mL)溶液中,0℃下逐滴加入3-氯丙酰氯(25.4mg,0.20mmol),该反应混合物在室温下搅拌反应过夜,TLC显示原料反应完成以后,向反应体系中加水淬灭,用EA(10mL×3)萃取,有机相无水Na2SO4干燥,真空蒸发后通过制备硅胶板(DCM/MeOH=25/1)纯化得到20mg目标化合物,为白色固体。
采用核磁共振以及质谱对产物结构进行表征,结果如下:
1H NMR(400MHz,d6-DMSO)δ1.25-1.54(5H,m),2.47-2.55(1H,m),2.68-2.70(1H,m),4.61(0.25H,s),4.73(0.75H,s),5.01(0.75H,s),5.19(0.25H,s),5.44(0.25H,d,J=10.4Hz),5.65(0.75H,dd,J=10.4Hz,2.4Hz),5.70(0.25H,dd,J=16.8Hz,10.4Hz),6.04-6.15(3H,m),6.74(0.75H,dd,J=16.8Hz,10.4Hz),7.03-7.19(6H,m),7.42-7.44(2H,m),7.55-7.61(2H,m),7.81(0.75H,d,J=5.2Hz),7.97(0.25H,d,J=5.2Hz).
EM(计算值):451.2;MS(ESI)m/e(M+1H)+:452.2
可见本申请制备得到的化合物与上述化合物结构一致。
实施例94~111
采用类似结构化合物为原料,以实施例92或实施例93的制备方法,制备得到以下化合物,所述化合物的结构以及核磁表征数据见表5,表5是本申请实施例94~111制备的化合物结构以及结构分析数据汇总。
表5 实施例94~111制备的化合物结构以及结构分析数据汇总
Figure PCTCN2017074108-appb-000096
Figure PCTCN2017074108-appb-000097
Figure PCTCN2017074108-appb-000098
Figure PCTCN2017074108-appb-000099
Figure PCTCN2017074108-appb-000100
实施例112
(S,E)-4-(8-氨基-3-(5-(4-(二甲氨基)2-丁烯酰基)-5-氮杂螺[2.4庚烷-6-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺的制备
Figure PCTCN2017074108-appb-000101
在氮气保护下,向装有(S)-4-(8-氨基-3-(5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺(100mg,0.235mmol)、(E)-4-(二甲氨基)-2-丁烯酸盐酸盐(46.7mg,0.282mmol)和DIPEA(121.26mg,0.94mmol)的DMF(3mL)溶液中,加入HBTU(106.9mg,0.282mmol),该反应混合物在室温下搅拌反应3h,TLC显示原料反应完成以后,向反应体系中加水淬灭,用EA(15mL×3)萃取,有机相饱和食盐水反洗,无水Na2SO4干燥,真空蒸发后通过制备硅胶板(DCM/MeOH=20/1)纯化得到20mg目标化合物,为淡黄色固体。
结构分析数据:
1H NMR(400MHz,d6-DMSO)δ0.56-0.73(4H,m),1.82(2H,s),2.13(4H,s),2.22-2.26(1.5H,m),3.05(2H,d,J=4.4Hz),3.62-3.66(1.5H,m),3.87(1H,d,J=10.4Hz),5.59-5.66(1H,m),6.03-6.15(2H,m),6.37-6.54(2H,m),7.14-7.21(2H,m),7.71(2H,d,J=8.4Hz),7.86-7.92(2H,m),8.15(2H,d,J=8.4Hz),8.24(1H,d,J=8.4Hz),8.40(1H,dd,J=4.8Hz,1.8Hz),10.85(1H,s).
EM(计算值):536.3;MS(ESI)m/e(M+1H)+:537.3。
实施例113
4-(8-氨基-3-((1S)-2-((E)-4-(二甲氨基)-2-丁烯酰基)八氢环戊烷[c]吡咯-1-基l)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺的制备
Figure PCTCN2017074108-appb-000102
在氮气保护下,向装有4-(8-氨基-3-((1S)-八氢环戊烷[c]吡咯-1-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺(100mg,0.228mmol)、(E)-4-(二甲氨基)-2-丁烯酸盐酸盐(45.4mg,0.274mmol)和DIPEA(117.6mg,0.912mmol)的DMF(3mL)溶液中,加入HBTU(86.4mg,0.228mmol),该反应混合物在室温下搅拌反应3h,TLC显示原料反应完成以后,向反应体系中加水淬灭,用EA(15mL×3)萃取,有机相饱和食盐水反洗,无水Na2SO4干燥,真空蒸发后通过制备硅胶板(DCM/MeOH=20/1)纯化得到28mg目标化合物,为淡黄色固体。
结构分析数据:
1H NMR(400MHz,d6-DMSO)δ1.45-1.60(3H,m),1.77-1.84(3H,m),1.85-2.03(2H,m),2.13(4H,s),2.68-2.71(1H,m),2.89-2.92(1H,m),3.05(2H,d,J=4.4Hz),3.63-3.67(1H,m),3.88-3.94(1H,m),5.43(1H,d,J=2.0Hz),6.03-6.18(2H,m),6.41-6.54(2H,m),7.15(1H,d,J=5.2Hz),7.18-7.20(1H,m),7.71(2H,d,J=8.4Hz),7.86-7.92(2H,m),8.13(2H,d,J=8.4Hz),8.23(1H,d,J=8.4Hz),8.42(1H,dd,J=4.8Hz,1.8Hz),10.84(1H,s).
EM(计算值):550.3;MS(ESI)m/e(M+1H)+:551.3
实施例114
(S,E)-4-(8-氨基-3-(5-(4-(环丙基(甲基)氨基)-2-丁烯酰基)-5-氮杂螺[2.4]庚烷-6-yl基)咪唑并[1,5-a]吡嗪-1-基)-N-(哌啶-2-基)苯甲酰胺的合成步骤如下所示:
Figure PCTCN2017074108-appb-000103
步骤1:(E)-4-(环丙基氨基)-2-丁炔酸乙酯的制备
Figure PCTCN2017074108-appb-000104
室温下,向装有环丙胺(2.2g,38.85mol)、K2CO3(3.6g,25.9mmol)的20mL THF溶液中加入溴代巴豆酸乙酯(90%purity,2.8g,12.95mmol),室温搅拌过夜,TLC检测反应完成以后,向上述反应混合物加水淬灭反应,EA(15mLx3)萃取,合并有机相后用饱和NaCl洗涤,无水Na2SO4充分干燥,真空蒸发后,得到1.1g目标化合物,不用进一步纯化直接用于下一步反应。
步骤2:(E)-4-(环丙基(甲基)氨基)-2-丁炔酸乙酯的制备
Figure PCTCN2017074108-appb-000105
在氮气保护下,向装有(E)-4-(环丙基氨基)-2-丁炔酸乙酯(1.1g,6.5mmol)的DCM溶液中加入甲醛水溶液(38wt%,2.6g,32.5mmol),冰盐浴下,逐份加入STAB(4.1g,19.5mmol),此反应混合物自然升至室温并搅拌3h,TLC检测反应完全以后,再慢慢滴加水淬灭反应,DCM(10mLx4)萃取,合并有机相后用饱和NaCl洗涤,无水Na2SO4充分干燥,真空蒸发后通过柱层析(PE/EA=20/1)纯化得到860mg目标化合物,为黄色油状物。
步骤3:(E)-4-(环丙基(甲基)氨基-2-丁烯酸的制备
Figure PCTCN2017074108-appb-000106
室温下,向装有(E)-4-(环丙基(甲基)氨基)-2-丁炔酸乙酯(860mg,4.7mmol)、的THF/水(5mL/3mL)的混合溶液中,加入LiOH(451.2mg,18.8mmol),该反应混合物在室温下搅拌5h,TLC检测反应完成,DCM/MeOH(10/1,10mL x 3)萃取,有机相用无水Na2SO4充分干燥,真空蒸发后得到750mg目标化合物,为白色固体。
步骤4:(S,E)-4-(8-氨基-3-(5-(4-(环丙基(甲基)氨基)-2-丁烯酰基)-5-氮杂螺[2.4]庚烷-6-yl基)咪唑并[1,5-a]吡嗪-1-基)-N-(哌啶-2-基)苯甲酰胺的制备
Figure PCTCN2017074108-appb-000107
在氮气保护下,向装有(S)-4-(8-氨基-3-(5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺(100mg,0.235mmol)、(E)-4-(环丙基(甲基)氨基-2-丁烯酸(48.2mg,0.282mmol)和DIPEA(121.26mg,0.94mmol)的DMF(3mL)溶液中,加入HBTU(106.9mg,0.282mmol),该反应混合物在室温下搅拌反应3h,TLC显示原料反应完成以后,向反应体系中加水淬灭,用EA(15mLX3)萃取,有机相饱和食盐水反洗,无水Na2SO4干燥,真空蒸发后通过制备硅胶板(DCM/MeOH=20/1)纯化得到45mg目标化合物,为淡黄色固体。
结构分析数据:
1H NMR(400MHz,d6-DMSO)δ0.24-0.39(4H,m),0.56-0.69(4H,m),1.44-1.46(1H,m),2.24-2.27(1.5H,m),3.23(3H,s),3.62-3.75(3.5H,m),3.85(1H,d,J=10.4Hz),5.65-5.68(1H,m),6.05-6.17(2H,m),6.37-6.50(2H,m),7.11-7.22(2H,m),7.66-7.71(2H,m),7.83-7.87(2H,m),8.13(2H,,J=8.4Hz),8.20(1H,d,J=8.4Hz),8.41(1H,d,J=3.6Hz),10.86(1H,s).
EM(计算值):562.3;MS(ESI)m/e(M+1H)+:563.3
实施例115
4-(8-氨基-3-((1S)-2-((E)-4-(环丙基(甲基)氨基)-2-丁烯酰基)八氢环戊烷[c]吡咯-1-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺的制备
Figure PCTCN2017074108-appb-000108
在氮气保护下,向装有4-(8-氨基-3-((1S)-八氢环戊烷[c]吡咯-1-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺(100mg,0.228mmol)(E)-4-(环丙基(甲基)氨基-2-丁烯酸(46.9mg,0.274mmol)和DIPEA(117.6mg,0.912mmol)的DMF(3mL)溶液中,加入HBTU(86.4mg,0.228mmol),该反应混合物在室温下搅拌反应3h,TLC显示原料反应完成以后,向反应体系中加水淬灭,用EA(15mL×3)萃取,有机相饱和食盐水反洗,无水Na2SO4干燥,真空蒸发后通过制备硅胶板(DCM/MeOH=20/1)纯化得到55mg目标化合物,为淡黄色固体。
结构分析数据:
1H NMR(400MHz,d6-DMSO)δ0.24-0.39(4H,m),1.48-1.59(3H,m),1.79-1.85(3H,m),1.91-2.03(2H,m),2.13(2H,s),2.67-2.71(1H,m),2.88-2.92(1H,m),3.04(2H,d,J=4.4Hz),3.63-3.66(1H,m),3.88-3.92(1H,m),5.45(1H,d,J=2.0Hz),6.03-6.15(2H,m),6.41-6.54(2H,m),7.14(1H,d,J=5.2Hz),7.17-7.20(1H,m),7.71(2H,d,J=8.4Hz),7.86-7.91(2H,m),8.13(2H,d,J=8.4Hz),8.25(1H,d,J=8.4Hz),8.42(1H,dd,J=4.8Hz,1.8Hz),10.86(1H,s).
EM(计算值):576.3;MS(ESI)m/e(M+1H)+:577.3
实施例116
(S,Z)-4-(8-氨基-3-(5-(2-氰基-4-甲基-2-戊烯酰基)-5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺的合成步骤如下所示:
Figure PCTCN2017074108-appb-000109
步骤1:(S)-4-(8-氨基-3-(5-(2-氰乙酰基)-5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺的制备
Figure PCTCN2017074108-appb-000110
在氮气保护下,向装有(S)-4-(8-氨基-3-(5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺(300mg,0.705mmol)、2-氰基乙酸(71.9mg,0.846mmol)和DIPEA(272.8mg,2.115mmol)的DMF(10mL)溶液中,加入HBTU(320.6mg,0.846mmol),该反应混合物在室温下搅拌反应3h,TLC显示原料反应完全以后,向反应体系中加水淬灭,用EA(15mL×3)萃取,有机相饱和食盐水反洗,无水Na2SO4干燥,真空蒸发后通过柱层析(DCM/MeOH=60/1~20/1)纯化得到150mg目标化合物,为黄色固体。
步骤2:(S,Z)-4-(8-氨基-3-(5-(2-氰基-4-甲基-2-戊烯酰基)-5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺的制备
Figure PCTCN2017074108-appb-000111
在氮气保护下,向装有(S)-4-(8-氨基-3-(5-(2-氰乙酰基)-5-氮杂螺[2.4]庚烷-6-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺(120mg,0.244mmol)、哌啶(24.3mg,0.293mmol)的MeOH(5mL)溶液中,加入异丁醛(26.4mg,0.366mmol),该反应混合物在室温下搅拌反应26h,TLC显示原料反应完全以后,向反应体系中加水淬灭,用DCM(15mL×3)萃取,无水Na2SO4干燥,真空蒸发后通过硅胶板(DCM/MeOH=20/1)纯化得到30mg目标化合物,为黄色固体。
结构分析数据:
1H NMR(400MHz,d6-DMSO)δ0.56-0.76(4H,m),1.04-1.09(6H,m),2.24-2.26(1.5H,m),2.78-2.81(1H,m),3.61-3.66(1.5H,m),3.87(1H,d,J=10.4Hz),5.60-5.68(1H,m),6.05-6.20(2H,m),6.74-6.77(1H,m),7.11-7.20(2H,m),7.66-7.74(2H,m),7.83-7.88(2H,m),8.15(2H,,J=8.4Hz),8.22(1H,d,J=8.4Hz),8.41(1H,d,J=3.6Hz),10.84(1H,s).
EM(计算值):546.2;MS(ESI)m/e(M+1H)+:547.3
实施例117
4-(8-氨基-3-((1S)-2-((Z)-2-氰基-4-甲基-2-戊烯酰基l)八氢环戊烷[c]吡咯-1-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺的制备
Figure PCTCN2017074108-appb-000112
合成方法同实施例116。通过制备硅胶板(DCM/MeOH=20/1)纯化得到20mg目标化合物,为黄色固体。
结构分析数据:
1H NMR(400MHz,d6-DMSO)δ1.04-1.10(6H,m),1.48-1.61(3H,m),1.76-1.79(1H,m),1.85-2.02(2H,m),2.65-2.69(1H,m),2.77-2.82(1H,m),2.90-2.93(1H,m),3.63-3.66(1H,m),3.91-4.00(1H,m),5.44-5.46(1H,m),6.08-6.17(2H,m),6.74-6.76(1H,m),7.13(1H,d,J=5.2Hz),7.20(1H,dd,J=6.8Hz,5.2Hz),7.70-7.73(2H,m),7.84-7.90(2H,m),8.13(2H,d,J=8.4Hz),8.22(1H,d,J=8.4Hz),8.40(1H,d,J=4.0Hz),10.85(1H,brs).
EM(计算值):560.3;MS(ESI)m/e(M+1H)+:561.3
实施例118
4-(3-((1R,2S,4S)-7-丙烯酰基-7-氮杂双环[2.2.1]庚烷-2-基)-8-氨基咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺的制备
合成方法同实施例2.通过制备硅胶板(DCM/MeOH=20/1)纯化得到15mg目标化合物,为类白色固体。
结构分析数据:
1H NMR(400MHz,d6-DMSO)δ1.49-1.57(2H,m),1.74-1.85(3H,m),2.33-2.39(1H,m),4.56-4.74(2H,m),5.15-5.17(0.3H,m),5.28-5.32(0.7H,m),5.55-5.69(1H,m),6.06-6.18(3H,m),6.72(1H,dd,J=16.8Hz,10.4Hz),7.13(1H,d,J=5.2Hz),7.16-7.21(1H,m),7.71(2H,dd,J=8.4Hz,4.0Hz),7.83-7.87(1H,m),7.91(1H,d,J=5.2Hz),8.15(2H,d,J=8.4Hz),8.20(1H,d,J=8.4Hz),8.40-8.42(1H,m),10.84(1H,s).
EM(计算值):479.2;MS(ESI)m/e(M+1H)+:479.2
实施例119
4-(8-氨基-3-((1R,2S,4S)-7-(2-丁炔酰基)-7-氮杂双环[2.2.1]庚烷-2-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺的制备
Figure PCTCN2017074108-appb-000114
合成方法同实施例1。通过制备硅胶板(DCM/MeOH=20/1)纯化得到12mg目标化合物,为类白色固体。
结构分析数据:
1H NMR(400MHz,d6-DMSO)δ1.40-1.55(3H,m),1.73-1.82(3H,m),2.02(2H,s), 2.33-2.40(1H,m),4.53-4.64(2H,m),5.13-5.17(0.4H,m),5.28-5.32(0.6H,m),6.11-6.23(2H,m),7.15(1H,d,J=5.2Hz),7.19-7.22(1H,m),7.71(2H,dd,J=8.4Hz,4.0Hz),7.85-7.87(1H,m),7.93(1H,d,J=5.2Hz),8.15(2H,d,J=8.4Hz),8.24(1H,d,J=8.4Hz),8.39-8.42(1H,m),10.83(1H,s).
EM(计算值):491.2;MS(ESI)m/e(M+1H)+:492.2
实施例120
4-(3-(2-丙烯酰基-2-氮杂双环[2.2.2]辛烷-1-基)-8-氨基咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺的制备
Figure PCTCN2017074108-appb-000115
合成方法同实施例2。通过制备硅胶板(DCM/MeOH=20/1)纯化得到10mg目标化合物,为淡黄色固体。
结构分析数据:
1H NMR(400MHz,d6-DMSO)δ1.22-1.55(3H,m),1.74-1.80(2H,m),2.07-2.23(3H,m),2.37-2.41(1H,m),3.25-3.28(1H,m),3.81-3.84(1H,m),5.65-5.69(1H,m),6.06-6.20(3H,m),6.75(1H,dd,J=16.8Hz,10.4Hz),7.13(1H,d,J=5.2Hz),7.16-7.21(1H,m),7.70(2H,dd,J=8.4Hz,4.0Hz),7.83-7.88(1H,m),7.90(1H,d,J=5.2Hz),8.15(2H,d,J=8.4Hz),8.21(1H,d,J=8.4Hz),8.39-8.41(1H,m),10.84-10.85(1H,m).
EM(计算值):493.2;MS(ESI)m/e(M+1H)+:494.2
实施例121
4-(8-氨基-3-(2-(2-丁炔酰基)-2-氮杂双环[2.2.2]辛烷-1-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺的制备
Figure PCTCN2017074108-appb-000116
合成方法同实施例1。通过制备硅胶板(DCM/MeOH=20/1)纯化得到13mg目标化合物,为淡黄色固体。
结构分析数据:
1H NMR(400MHz,d6-DMSO)δ1.25-1.53(4H,m),1.70-1.76(2H,m),2.01(2H,s),2.07-2.24(3H,m),2.35-2.41(1H,m),3.25-3.26(1H,m),3.81-3.84(1H,m),6.06-6.20(2H,m),7.14(1H,d,J=5.2Hz),7.15-7.23(1H,m),7.68(2H,dd,J=8.4Hz,4.0Hz),7.81-7.85(1H,m),7.90(1H,d,J=5.2Hz),8.16(2H,d,J=8.4Hz),8.20(1H,d,J=8.4Hz),8.39-8.41(1H,m),10.86-10.88(1H,m).
EM(计算值):505.2;MS(ESI)m/e(M+1H)+:506.2
实施例122
4-(3-(2-丙烯酰基-2-氮杂双环[2.2.1]庚烷-1-基)-8-氨基咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺的制备
Figure PCTCN2017074108-appb-000117
合成方法同实施例2。通过制备硅胶板(DCM/MeOH=20/1)纯化得到20mg目标化合物,为淡黄色固体。
结构分析数据:
1H NMR(400MHz,d6-DMSO)δ1.25-1.51(3H,m),1.69-1.72(1H,m),1.90-1.93(1H,m),2.61-2.70(2H,m),3.03-3.07(1H,m),3.43-3.47(1H,m),5.61-5.68(1H,m),6.10-6.19(3H,m),6.73-6.75(1H,m),7.11-7.19(2H,m),7.73-7.86(4H,m),8.15(2H,dd,J=8.4Hz,2.8Hz),8.20 (1H,d,J=8.4Hz),8.41(1H,dd,J=4.8Hz,1.2Hz),10.81(1H,s).
EM(计算值):479.2;MS(ESI)m/e(M+1H)+:480.2
实施例123
4-(8-氨基-3-(2-(2-丁炔酰基)-2-氮杂双环[2.2.1]庚烷-1-基)咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺的制备
Figure PCTCN2017074108-appb-000118
合成方法同实施例1。通过制备硅胶板(DCM/MeOH=20/1)纯化得到15mg目标化合物,为淡黄色固体。
结构分析数据:
1H NMR(400MHz,d6-DMSO)δ1.31-1.56(4H,m),1.66-1.72(1H,m),1.95-1.98(1H,m),2.02(2H,s),2.59-2.72(2H,m),3.07-3.09(1H,m),3.44-3.47(1H,m),6.10-6.19(2H,m),7.12-7.19(2H,m),7.71-7.76(2H,m),7.84-7.89(2H,m),8.15-8.17(2H,m),8.21(1H,d,J=8.4Hz),8.40-8.42(1H,m),10.85-10.87(1H,m).
EM(计算值):491.2;MS(ESI)m/e(M+1H)+:492.2
实施例124
(S)-3-(3-(5-丙烯酰基-5-氮杂螺环[2.4]庚烷-6-基)-8-氨基咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺的制备
Figure PCTCN2017074108-appb-000119
合成方法同实施例2。通过制备硅胶板(DCM/MeOH=20/1)纯化得到20mg目标化合物,为淡黄色固体。
结构分析数据:
1H NMR(400MHz,d6-DMSO)δ0.58-0.72(4H,m),2.23-2.25(1.6H,m),3.63-3.66(1.4H,m),3.85(1H,d,J=10.4Hz),5.62-5.68(2H,m),6.05-6.17(3H,m),6.54(1H,dd,J=16.8Hz,10.4Hz),7.12-7.20(2H,m),7.78-7.83(3H,m),7.83-7.86(2H,m),8.21(1H,d,J=8.4Hz),8.41(1H,d,J=3.6Hz),8.68(1H,s),10.82(1H,s).
EM(计算值):479.2;MS(ESI)m/e(M+1H)+:480.2
实施例125
(S)-2-(3-(5-丙烯酰基-5-氮杂螺环[2.4]庚烷-6-基)-8-氨基咪唑并[1,5-a]吡嗪-1-基)-N-(吡啶-2-基)苯甲酰胺的制备
Figure PCTCN2017074108-appb-000120
合成方法同实施例2。通过制备硅胶板(DCM/MeOH=20/1)纯化得到18mg目标化合物,为淡黄色固体。
结构分析数据:
1H NMR(400MHz,d6-DMSO)δ0.55-0.76(4H,m),2.23-2.26(1.5H,m),3.61-3.65(1.5H,m),3.86(1H,d,J=10.4Hz),5.62-5.68(2H,m),6.07-6.19(3H,m),6.55(1H,dd,J=16.8Hz, 10.4Hz),7.13-7.20(2H,m),7.58-7.62(2H,m),7.70-7.72(1H,m),7.83-7.88(2H,m),8.12-8.14(1H,m),8.21(1H,d,J=8.4Hz),8.40(1H,d,J=3.6Hz),10.84(1H,s).
EM(计算值):479.2;MS(ESI)m/e(M+1H)+:480.2
实施例126
(1S,4S)-叔丁基-5-丙烯酰基-6-(8-氨基-1-(4-(吡啶-2-氨甲酰基)苯基)咪唑并[1,5-a]吡嗪-3-基)-2,5-二氮杂双环[2.2.1]庚烷-2-甲酸酯的制备
Figure PCTCN2017074108-appb-000121
合成方法同实施例2。通过制备硅胶板(DCM/MeOH=20/1)纯化得到25mg目标化合物,为淡黄色固体。
结构分析数据:
1H NMR(400MHz,d6-DMSO)δ1.47-1.52(9H,m),1.99-2.01(1H,m),2.82-2.84(1H,m),3.24-3.48(2H,m),4.38-4.52(1H,m),4.96-5.24(2H,m),5.46-5.52(0.4H,m),5.65-5.76(1H,m),6.00-6.30(3H,m),6.86(0.6H,dd,J=16.8Hz,10.4Hz),7.17-7.25(2H,m),7.65-7.75(3H,m),7.84-7.88(1H,m),8.14-8.23(3H,m),8.40-8.42(1H,m),10.85(1H,s).
EM(计算值):580.3;MS(ESI)m/e(M+1H)+:581.3
药效试验
试验例1:体外BTK抑制激酶活性试验
1:试验原理:
微流体芯片技术的迁移率检测技术(Mobility-Shift Assay),该技术将毛细管电泳的基本理念应用到微流体环境中,用于实验的底物是带有荧光标记的多肽,在反应体系中酶的作用下,底物转变为产物,其所带的电荷也发生了相应的变化,Mobility-Shift Assay正是利用底物和产物所带电荷的不同,将二者进行分离,并分别进行检测,检测结果由转化率表达出来。
2:试验方法:
(1)配置待测样品:用100%DMSO稀释至反应终浓度的50倍,即25umol/L;
(2)稀释:25umol/L为起始浓度,然后以4倍浓度稀释,稀释10个浓度梯度;
(3)阳性对照和阴性对照孔中分别加入100%DMSO;
(4)将配好的10个浓度的化合物分别用1倍激酶缓冲液稀释10倍;其中激酶缓冲液中包含浓度为50mmol/L,pH为7.5的羟乙基哌嗪乙硫磺酸、0.01%的十二烷基聚乙二醇醚、10mmol/L的氯化镁、2mmol/L的二硫苏糖醇;
(5)配制2.5倍酶溶液:将激酶加入1倍激酶缓冲液,形成2.5倍酶溶液;
(6)配制2.5倍的底物溶液:将FAM标记的多肽和ATP加入1倍激酶缓冲液,形成2.5倍底物溶液;
(7)向384孔板中加入酶溶液:384孔反应板中已有5μl的10%DMSO溶解的5倍化合物,然后再加入10μl的2.5倍酶溶液,室温下孵育10分钟;
(8)向384孔板中加入底物溶液:在384孔反应板中加入10μl的2.5倍底物溶液;
(9)激酶反应和终止:28℃下孵育1h,然后加25μl终止液终止反应;其中终止液中包含浓度为100mmol/L,pH为7.5的羟乙基哌嗪乙硫磺酸、0.015%的十二烷基聚乙二醇醚、0.2%的3号表面试剂、20mmol/L的乙二胺四乙酸;
(10)Caliper读取数据:Caliper上读取转化率数据;
试验例2:选用不同的细胞株对化合物进行体外细胞增殖抑制活性的测定
1:培养基配制见表6:
表6 培养基配制
细胞系 培养基
TMD8 MEM+10%FBS
OCI-Ly10 IMDM+10%FBS+1%PS+L-Glu
DOHH2 RPMI1640+10%FBS
2:化合物配置:
待测化合物分别用DMSO稀释配成终浓度为10mM母液备用。
3:IC50测定
3.1.CCK-8方法检测TMD8和OCI-ly10细胞
收集对数生长期细胞,计数,用完全培养基重新悬浮细胞,调整细胞浓度至合适浓度(依照细胞密度优化试验结果确定),接种96孔板,每孔加100μl细胞悬液。细胞在37℃,100%相对湿度,5%CO2培养箱中孵育24小时。
用培养基将待测化合物稀释至所设置的相应作用浓度,按25μl/孔加入细胞。细胞置于37℃,100%相对湿度,5%CO2培养箱中孵育72小时。
吸弃培养基,加入含10%检测试剂的完全培养基置于37℃培养箱中孵育1-4小时。轻轻震荡后在SpectraMax M5 Microplate Reader上测定450nm波长处的吸光度,以650nm处吸光度作为参比,计算抑制率。
3.2CTG assay方法检测DOHH2细胞
收集对数生长期细胞,计数,用完全培养基重新悬浮细胞,调整细胞浓度至合适浓度(依照细胞密度优化试验结果确定),接种96孔板,每孔加90μl细胞悬液。细胞在37℃,100%相对湿度,5%CO2培养箱中孵育24小时。
用培养基将待测化合物稀释至所设置的相应作用浓度,按10μl/孔加入细胞。细胞置于37℃,100%相对湿度,5%CO2培养箱中孵育72小时。
吸弃培养基,加入30ul的检测试剂,避光摇板10分钟,裂解细胞。之后在室温孵育2分钟,在Envision上测定,计算抑制率。
4:数据处理
按下式计算药物对肿瘤细胞生长的抑制率:肿瘤细胞生长抑制率%=[(Ac-As)/(Ac-Ab)]×100%
As:样品的OA(细胞+检测试剂+待测化合物)
Ac:阴性对照的OA(细胞+检测试剂+DMSO)
Ab:阳性对照的OA(培养基+检测试剂+DMSO)
运用软件Graphpad Prism 5或XLfit进行IC50曲线拟合并计算出IC50值。
化合物对BTK激酶抑制活性和体外细胞增殖抑制活性结果见表7:
表7 化合物对BTK激酶抑制活性和体外细胞增殖抑制活性结果
Figure PCTCN2017074108-appb-000122
Figure PCTCN2017074108-appb-000123
Figure PCTCN2017074108-appb-000124
Figure PCTCN2017074108-appb-000125
Figure PCTCN2017074108-appb-000126
试验例3:hERG实验考察潜在的心脏毒性
1:细胞培养
CHO hERG细胞生长于含上述培养液的培养皿中,并在37℃、含5%CO2的培养箱中进行培养。电生理实验之前24到48小时,CHO hERG细胞被转移到放置于培养皿中的圆形玻璃片上,并在以上相同的培养液及培养条件下生长。每个圆形玻片上CHO hERG细胞的密度需要达到绝大多数细胞是独立、单个的要求。
2:实验溶液配制
细胞内液和外液的组成成分
Figure PCTCN2017074108-appb-000127
3:电生理记录系统
本实验采用手动膜片钳系统(HEKA EPC-10信号放大器及数字转换系统,购自德国HEKA Electronics)作全细胞电流的记录。表面生长有CHO hERG细胞的圆形玻片被放置于倒置显微镜下的电生理记录槽中。记录槽内以细胞外液作持续灌流(大约每分钟1毫升)。实验过程采用常规全细胞膜片钳电流记录技术。如无特殊说明,实验都是在常规室温下进行(~25℃)。细胞钳制在-80mV的电压下。细胞钳制电压去极化到+20mV以激活hERG钾通道,5秒后再钳制到-50mV以消除失活并产生尾电流。尾电流峰值用作hERG电流大小的数值。上述步骤所记录的hERG钾电流在记录槽内持续的细胞外液灌流下达到稳定后 则可以叠加灌流待测试的药物,直到药物对hERG电流的抑制作用达到稳定状态。一般以最近的连续3个电流记录线重合作为判断是否稳定状态的标准。达到稳定态势以后以细胞外液灌流冲洗直到hERG电流回复到加药物之前的大小。一个细胞上可以测试一个或多个药物,或者同一种药物的多个浓度,但是在不同药物之间需要以细胞外液冲洗。Cisapride(西沙必利,购自Sigma)被用于实验中作为阳性对照以保证所使用的细胞质量正常。
4:化合物的处理与稀释
为了取得化合物的IC50,我们选择了下列浓度(30,10,3,1,0.3和0.1μM)来作测试。在试验之前,首先用DMSO以梯度稀释的方式稀释成10,3,1,0.3和0.1mM的贮备液,再用细胞外液稀释成最终的μM测试浓度。除了30μM的化合物测试溶液中的DMSO浓度为0.3%以外,其它各浓度化合物溶液中DMSO的最终浓度都为0.1%。阳性对照Cisapride(西沙比利)的测试浓度为0.1μM。所有的化合物溶液都经过常规的5到10分钟超声和振荡以保证化合物完全溶解。
5:数据处理
试验数据由HEKA Patchmaster,Microsoft Excel以及Graphpad Prism提供的数据分析软件进行分析。
部分化合物心脏hERG钾电流检测结果见表8:
表8 部分化合物心脏hERG钾电流检测结果
实施例 hERG IC50值(um)
3 >30
5 >30
11 >30
18 >30
22 >30
47 >30
79 >30
96 >30
依鲁替尼 1.5
试验例4:本发明化合物的药代动力学测试
SD大鼠,雄性(购于上海西普尔-必凯实验动物有限公司)。各受试化合物分别以口服(10mg/kg,每组3只)和静脉(1mg/kg,每组3只)两种给药方式单次给予SD大鼠进行药代动力学研究,受试化合物使用DMSO/9%的氯化钠注射液=5/95(V/V)溶解,并经涡 旋1min,超声1min之后配制成给药溶液。口服给药前动物需禁食16-17小时,并于给药4小时后恢复给食。SD大鼠经口服与静脉给药后,经颈静脉或心脏穿刺采集药代动力学样本,采集时间点为:给药前、给药后5min、15min、30min、1h、2h、4h、6h、8h和24h,每个时间点采集3个全血样本,采集量约0.2mL,并经肝素钠抗凝。血液样本采集后立即置于冰上,于30分钟之内离心分离血浆(离心条件:8000转/分钟,6分钟,2-8℃)。收集的血浆分析前存放于–70℃。取50μL血浆样品至1.5mL离心管中,加入250μL内标溶液(空白不加内标补加相同体积的甲醇),涡旋混匀,15000转/分钟离心5分钟,取200μL上清液加入到96孔进样板中,经LC-MS/MS进样分析。
本发明部分化合物的药代动力学测试结果如下表9所示:
表9 本发明部分化合物的药代动力学测试结果
实施例 T1/2(iv)h Tmax(po)h Cmax(po)ng/ml AUC(po)ng/ml*h Cl(iv)ml/hr/kg F(po)%
实施例3 0.62 0.27 1619.79 1492.19 3420.38 52.55
实施例5 0.13 0.10 303.06 278.20 7278.25 20.64
实施例11 0.47 0.33 1612.51 1583.67 2187.37 34.59
实施例22 0.60 0.25 1156.00 1038.16 3510.37 36.56
实施例47 0.30 0.25 1121.14 971.12 2277.10 21.89
实施例79 0.52 0.21 1105.72 770.13 3450.68 27.12
实施例96 0.55 0.36 1589.06 1580.79 1960.88 34.53
依鲁替尼 0.26 0.33 217.10 218.09 1200.00 2.67
ACP-196 0.13 0.08 266.02 266.08 7489.19 19.75
从以上成药性研究数据可以看出,本发明化合物对BTK活性具有明显的抑制作用,与已上市药物依鲁替尼或临床三期的ACP-196相比,心脏毒性极小,药代方面也具有明显的优势,可用作BTK抑制剂,具有广阔的抗恶性肿瘤疾病或炎症疾病的应用前景。
以上实施例的说明只是用于帮助理解本发明的方法及其核心思想。应当指出,对于本技术领域的普通技术人员来说,在不脱离本发明原理的前提下,还可以对本发明进行若干改进和修饰,这些改进和修饰也落入本发明权利要求的保护范围内。

Claims (18)

  1. 一种用作布鲁顿酪氨酸激酶抑制剂的化合物,其特征在于,具有式(Ⅰ)所示结构或其异构体、药学上可接受的溶剂化物或盐:
    Figure PCTCN2017074108-appb-100001
    其中,Y选自取代或非取代的芳基或杂芳基;
    R选自取代或非取代的烯基或炔基;
    M选自取代或非取代的含N螺环基团或含N桥环基团,且N原子与羰基相连;
    或者M选自式(Ⅱ)所示基团,A为取代或非取代的螺环基团或桥环基团,且氨基与羰基相连;
    Figure PCTCN2017074108-appb-100002
  2. 根据权利要求1所述的化合物,其特征在于,所述M选自取代或非取代的C5~15的含N螺环基团或含N桥环基团;A为取代或非取代的C5~15的螺环基团或桥环基团。
  3. 根据权利要求1所述的化合物,其特征在于,所述M选自以下基团:
    Figure PCTCN2017074108-appb-100003
    其中,n1、n2、m独立的为0、1或2;
    n为1、2或3;
    X、Q独立的选自CR2R3、N-R4、O、S或S(O)2
    R1、R2、R3、R4独立的选自H、取代或未取代C1~10烷基,取代或未取代C1~10杂烷基,C1~10羰基基团,取代或未取代C3~10环烷基,取代或未取代C3~10杂环烷基。
  4. 根据权利要求3所述的化合物,其特征在于,所述M选自以下基团:
    Figure PCTCN2017074108-appb-100004
    其中,P选自CR5R6,N-R7或O;
    R7为取代或非取代C1~8烷基,取代或非取代C1~8杂烷基,取代或未取代C3~8环烷基,取代或未取代C3~8杂环烷基,或
    Figure PCTCN2017074108-appb-100005
    R5、R6、R8独立的选自取代或非取代C1~8烷基,或取代或非取代C1~8杂烷基,取代或未取代C3~8环烷基,取代或未取代C3~8杂环烷基。
  5. 根据权利要求3所述的化合物,其特征在于,所述M选自以下基团:
    Figure PCTCN2017074108-appb-100006
    Figure PCTCN2017074108-appb-100007
  6. 根据权利要求1所述的化合物,其特征在于,所述R为取代或非取代的烯基或炔基。
  7. 根据权利要求6所述的化合物,其特征在于,所述R选自以下基团:
    Figure PCTCN2017074108-appb-100008
    其中,R'为H、取代或非取代的C1~8烷基、取代或非取代的C1~8杂烷基,取代或非取代的C1~8环烷基或取代或非取代的C1~8杂环烷基;
    R"为H、硝基、卤素或氰基。
  8. 根据权利要求1所述的化合物,其特征在于,所述Y为取代或非取代的C5~10的芳基或杂芳基。
  9. 根据权利要求1所述的化合物,其特征在于,所述Y选自取代苯基,所述苯基的取代基选自取代或非取代酰胺基团、取代或非取代烷基、取代或非取代醚基。
  10. 根据权利要求9所述的化合物,其特征在于,所述Y选自以下基团:
    Figure PCTCN2017074108-appb-100009
    其中,R9为三氟甲基或甲基;
    R10,R11和R12独立的选自取代或非取代的芳基或杂芳基。
  11. 根据权利要求10所述的化合物,其特征在于,所述R10,R11和R12独立的选自取代或非取代的苯基、吡啶基、哌啶基、哌嗪基或嘧啶基;
    上述基团的取代基选自硝基、羟基、巯基、氟、氯、溴、碘、氰基、取代或非取代C1~10烷基、取代或非取代C1~10杂烷基,取代或未取代C3~10环烷基,取代或未取代C3~10杂环烷基。
  12. 根据权利要求1所述的化合物,其特征在于,所述A为金刚烷基。
  13. 根据权利要求1所述的化合物,其特征在于,具有以下任一结构或其立体异构体或顺反异构体:
    Figure PCTCN2017074108-appb-100010
    Figure PCTCN2017074108-appb-100011
    Figure PCTCN2017074108-appb-100012
    Figure PCTCN2017074108-appb-100013
    Figure PCTCN2017074108-appb-100014
    Figure PCTCN2017074108-appb-100015
    Figure PCTCN2017074108-appb-100016
    Figure PCTCN2017074108-appb-100017
    Figure PCTCN2017074108-appb-100018
  14. 权利要求1~13任一项所述的化合物的制备方法,其特征在于,包括以下步骤:
    1)以2-氯吡嗪为原料,在碱性化合物作用下,反应生成3-氯-2-吡嗪甲醇;
    2)将3-氯-2-吡嗪甲醇通过盖布瑞尔合成法生成(3-氯吡嗪-2-基)甲胺;
    3)将(3-氯吡嗪-2-基)甲胺与式(Ⅲ)所示的螺环羧酸或桥环羧酸进行反应,制备得到 酰胺类化合物;
    4)将酰胺类化合物在三氯氧磷的作用下进行闭环反应,然后经NBS溴代,得到式(Ⅳ)所示化合物;
    5)将上述式(Ⅳ)所示化合物在醇和氨水的作用下,发生氨基化反应;
    6)将上述氨基化反应后的产物与式(Ⅴ-1)所示硼酸或式(Ⅴ-2)所示硼酸酯通过Suzuki偶联反应,得到式(Ⅵ)所示化合物;
    7)将上述式(Ⅵ)所示化合物与取代的或未取代的2-丁炔酸在缩合剂作用下发生缩合反应,得到式(Ⅰ-1)所示化合物;
    或者将上述式(Ⅵ)所示化合物与3-氯丙酰氯或烯丙酰氯在碱的作用下通过直接缩合或者直接缩合后再消除氯化氢烯烃化的方式进行反应,得到式(Ⅰ-2)所示化合物;
    或者将上述式(Ⅵ)所示化合物与式(Ⅶ)所示的烯酸类衍生物在缩合剂作用下进行反应,得到式(Ⅰ-3)或式(Ⅰ-4)所示化合物,R"分别为H或氟、氯、溴、碘;
    或者将上述式(Ⅵ)所示化合物与氰基乙酸或硝基乙酸在缩合剂的作用下得到酰胺化合物,然后再与式(Ⅷ)所示醛类化合物发生脑文格反应得到式(Ⅰ-4)所示化合物;R"为硝基或氰基;
    Figure PCTCN2017074108-appb-100019
    Figure PCTCN2017074108-appb-100020
    其中,Y为取代或非取代的芳基或杂芳基;
    M为取代或非取代的含N螺环基团或含N桥环基团,且N原子与羰基相连;
    或者M为式(Ⅱ)所示基团,A为取代或非取代的螺环基团或桥环基团,且氨基与羰基相连;
    Figure PCTCN2017074108-appb-100021
    R'为H、取代或非取代的C1~8烷基、取代或非取代的C1~8杂烷基,取代或非取代的C1~8环烷基或取代或非取代的C1~8杂环烷基。
  15. 一种药物组合物,包括权利要求1~13任意一项所述的化合物或其盐或权利要求14所述的制备方法制备的化合物或其盐,以及药学上可接受的载体,赋形剂,稀释剂,辅剂,媒介物或它们的组合。
  16. 权利要求1~13任一项所述的化合物或权利要求14所述的制备方法制备的化合物或权利要求15所述的药物组合物在制备用于治疗或减轻BTK介导的疾病的药物中的应用。
  17. 根据权利要求16所述的应用,其特征在于,所述BTK介导的疾病选自免疫、自身免疫、炎症疾病、过敏症、感染性疾病、增生性病症和癌症疾病中的任意一种或多种。
  18. 根据权利要求17所述的应用,其特征在于,所述BTK介导的疾病选自风湿性关节炎、感染性关节炎、致畸性关节炎、痛风性关节炎、脊椎炎、胰腺炎、慢性支气管炎、急性支气管炎、过敏性支气管炎、毒性支气管炎、儿科哮喘、变应性肺泡炎、过敏性或非过敏性鼻炎、慢性鼻窦炎、囊性纤维化或粘液粘稠病、咳嗽、肺气肿、间质性肺病、肺泡炎、鼻息肉、肺水肿、各种原因的肺炎、红斑狼疮、全身性硬皮病、结节病、亚弥型漫性大B细胞淋巴瘤、套细胞淋巴瘤、慢性淋巴细胞淋巴瘤、结外边缘带B细胞淋巴瘤、B细胞慢性淋巴细胞白血病、B细胞幼淋巴细胞白血病、成熟B细胞的急性成淋巴细胞性白血病、17p缺失的慢性淋巴细胞白血病、
    Figure PCTCN2017074108-appb-100022
    巨球蛋白血症、淋巴质浆细胞淋巴瘤、脾脏边缘带淋巴瘤、浆细胞性骨髓瘤、浆细胞瘤、结内边缘带B细胞淋巴瘤、外套细胞淋巴瘤、血管内大B细胞淋巴瘤和原发性渗出性淋巴瘤中的一种或多种。
PCT/CN2017/074108 2017-01-20 2017-02-20 用作布鲁顿酪氨酸激酶抑制剂的化合物及其制备方法和应用 WO2018133151A1 (zh)

Priority Applications (3)

Application Number Priority Date Filing Date Title
EP17893058.2A EP3572414A4 (en) 2017-01-20 2017-02-20 COMPOUND USED AS A BRUTON TYROSINE KINASE INHIBITOR, PROCESS OF PREPARATION AND APPLICATION
US16/468,658 US10793576B2 (en) 2017-01-20 2017-02-20 Compound used as Bruton's tyrosine kinase inhibitor and preparation method and application thereof
JP2019531745A JP6884863B2 (ja) 2017-01-20 2017-02-20 ブルトン型チロシンキナーゼ阻害剤として使用される化合物、並びにその調製方法及び応用

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201710044771.4A CN106831787B (zh) 2017-01-20 2017-01-20 用作布鲁顿酪氨酸激酶抑制剂的化合物及其制备方法和应用
CN201710044771.4 2017-01-20

Publications (1)

Publication Number Publication Date
WO2018133151A1 true WO2018133151A1 (zh) 2018-07-26

Family

ID=59119508

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2017/074108 WO2018133151A1 (zh) 2017-01-20 2017-02-20 用作布鲁顿酪氨酸激酶抑制剂的化合物及其制备方法和应用

Country Status (6)

Country Link
US (1) US10793576B2 (zh)
EP (1) EP3572414A4 (zh)
JP (1) JP6884863B2 (zh)
CN (1) CN106831787B (zh)
TW (1) TWI668221B (zh)
WO (1) WO2018133151A1 (zh)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019239374A1 (en) * 2018-06-13 2019-12-19 Acerta Pharma B.V. Imidazopyrazine inhibitors of interleukin-2-inducible t-cell kinase
WO2020188015A1 (en) 2019-03-21 2020-09-24 Onxeo A dbait molecule in combination with kinase inhibitor for the treatment of cancer
WO2021089791A1 (en) 2019-11-08 2021-05-14 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of cancers that have acquired resistance to kinase inhibitors
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
EP3851439A4 (en) * 2018-09-14 2022-06-08 Chia Tai Tianqing Pharmaceutical Group Co., Ltd. BTK INHIBITOR CONTAINING FURO[3,4-B]PYRROLE
WO2022140246A1 (en) 2020-12-21 2022-06-30 Hangzhou Jijing Pharmaceutical Technology Limited Methods and compounds for targeted autophagy
WO2023122594A1 (en) * 2021-12-21 2023-06-29 Corcept Therapeutics Incorporated Bicyclic indazole glucocorticoid receptor antagonists
WO2023122600A1 (en) * 2021-12-21 2023-06-29 Corcept Therapeutics Incorporated Piperazine indazole glucocorticoid receptor antagonists
JP7438962B2 (ja) 2018-03-23 2024-02-27 ベイジン タイド ファーマシューティカル カンパニー リミテッド 受容体阻害剤、同阻害剤を含む医薬組成物及びその使用
TWI835476B (zh) 2021-12-21 2024-03-11 美商科賽普特治療學股份有限公司 哌嗪吲唑糖皮質素受體拮抗劑

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102384924B1 (ko) * 2017-07-12 2022-04-08 주식회사 대웅제약 신규한 1h-피라졸로피리딘 유도체 및 이를 포함하는 약학 조성물
WO2019013562A1 (ko) * 2017-07-12 2019-01-17 주식회사 대웅제약 신규한 1h-피라졸로피리딘 유도체 및 이를 포함하는 약학 조성물
CN108191871B (zh) * 2018-01-02 2020-02-18 成都倍特药业有限公司 新型布鲁顿酪氨酸激酶抑制剂及其制备方法和应用
US11325921B2 (en) 2018-03-30 2022-05-10 Sumitomo Dainippon Pharma Co., Ltd. Optically active crosslinked cyclic secondary amine derivative
CN109053780B (zh) * 2018-07-05 2020-09-29 浙江合聚生物医药有限公司 一种抗肿瘤药物Acalabrutinib关键中间体的制备方法
WO2020015735A1 (zh) * 2018-07-20 2020-01-23 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
TWI767148B (zh) * 2018-10-10 2022-06-11 美商弗瑪治療公司 抑制脂肪酸合成酶(fasn)
CN111620875B (zh) * 2019-02-28 2021-12-28 成都倍特药业股份有限公司 咪唑并吡嗪类化合物的制备工艺
CN113544130B (zh) * 2019-05-31 2024-01-09 西藏海思科制药有限公司 一种btk抑制剂环衍生物及其制备方法和药学上的应用
CN112142762A (zh) * 2019-06-27 2020-12-29 成都倍特药业股份有限公司 一种布鲁顿酪氨酸激酶抑制剂的晶体及其制备方法和用途
CN112209934B (zh) * 2019-07-12 2023-09-08 正大天晴药业集团股份有限公司 含有氮杂螺庚烷的btk抑制剂
WO2021008441A1 (zh) * 2019-07-12 2021-01-21 正大天晴药业集团股份有限公司 含有5-氮杂螺庚烷的btk抑制剂
CN112209933B (zh) * 2019-07-12 2023-09-08 正大天晴药业集团股份有限公司 含有4-氮杂螺庚烷的btk抑制剂
CN110606848A (zh) * 2019-08-27 2019-12-24 药雅科技(上海)有限公司 一种5-氮杂吲哚衍生物Bruton′s酪氨酸激酶抑制剂及其制备方法与用途
WO2021110142A1 (en) * 2019-12-04 2021-06-10 Henan Normal University Substituted imidazolecarboxamide as bruton's tyrosine kinase inhibitors
CN115210241B (zh) * 2020-03-09 2023-09-08 成都倍特药业股份有限公司 咪唑并吡嗪化合物枸橼酸盐固体形态
CN111471048B (zh) * 2020-04-30 2021-06-15 成都海博为药业有限公司 一种具有含氮桥环、螺环或并环结构的化合物及其用途
WO2021262587A1 (en) 2020-06-22 2021-12-30 Corcept Therapeutics Incorporated Quaternary indazole glucocorticoid receptor antagonists
CN115702898B (zh) * 2021-08-04 2024-02-09 成都倍特药业股份有限公司 一种btk抑制剂固体制剂及其制备方法

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011035332A1 (en) 2009-09-21 2011-03-24 Chemocentryx, Inc. Pyrrolidinone carboxamide derivatives as chemerin-r ( chemr23 ) modulators
WO2013113097A1 (en) * 2012-01-31 2013-08-08 Beta Pharma Canada Inc. Cyclic molecules as bruton's tyrosine kinase inhibitors
WO2014140081A1 (en) 2013-03-14 2014-09-18 Boehringer Ingelheim International Gmbh Substituted bicyclic 1-carboxylic-acid (benzyl-cyano-methyl)-amides inhibitors of cathepsin c
WO2015132799A2 (en) * 2014-02-03 2015-09-11 Cadila Healthcare Limited Novel heterocyclic compounds

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1500982A1 (en) 2003-07-24 2005-01-26 ASML Netherlands B.V. Lithographic apparatus and device manufacturing method
JP2011520970A (ja) * 2008-05-19 2011-07-21 オーエスアイ・フアーマスーテイカルズ・インコーポレーテツド 置換されたイミダゾピラジン類およびイミダゾトリアジン類
LT2734522T (lt) * 2011-07-19 2019-02-11 Merck Sharp & Dohme B.V. 4-imidazopiridazin-1-il-benzamidai ir 4-imidazotriazin-1-il-benzamidai kaip btk-inhibitoriai
US8940893B2 (en) 2013-03-15 2015-01-27 Boehringer Ingelheim International Gmbh Heteroaromatic compounds as BTK inhibitors
CA2918242C (en) * 2013-07-31 2022-06-21 Merck Patent Gmbh Pyridines, pyrimidines, and pyrazines, as btk inhibitors and uses thereof
US9937171B2 (en) 2014-04-11 2018-04-10 Acerta Pharma B.V. Methods of blocking the CXCR-4/SDF-1 signaling pathway with inhibitors of bruton's tyrosine kinase
WO2016106628A1 (en) * 2014-12-31 2016-07-07 Merck Sharp & Dohme Corp. Btk inhibitors
CN105913859A (zh) 2015-12-12 2016-08-31 乐视致新电子科技(天津)有限公司 音源输入模式提示方法及音频播放设备
CN105732638B (zh) * 2016-01-22 2018-01-30 成都倍特药业有限公司 一种具有螺环或桥环结构的布鲁顿酪氨酸激酶抑制剂及其制备方法

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011035332A1 (en) 2009-09-21 2011-03-24 Chemocentryx, Inc. Pyrrolidinone carboxamide derivatives as chemerin-r ( chemr23 ) modulators
WO2013113097A1 (en) * 2012-01-31 2013-08-08 Beta Pharma Canada Inc. Cyclic molecules as bruton's tyrosine kinase inhibitors
WO2014140081A1 (en) 2013-03-14 2014-09-18 Boehringer Ingelheim International Gmbh Substituted bicyclic 1-carboxylic-acid (benzyl-cyano-methyl)-amides inhibitors of cathepsin c
WO2015132799A2 (en) * 2014-02-03 2015-09-11 Cadila Healthcare Limited Novel heterocyclic compounds

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
JOURNAL OF ORGANIC CHEMISTRY, vol. 59, no. 2, 1994, pages 276 - 277
See also references of EP3572414A4
TETRAHEDRON LETTERS, vol. 57, 2016, pages 599 - 602

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP7438962B2 (ja) 2018-03-23 2024-02-27 ベイジン タイド ファーマシューティカル カンパニー リミテッド 受容体阻害剤、同阻害剤を含む医薬組成物及びその使用
WO2019239374A1 (en) * 2018-06-13 2019-12-19 Acerta Pharma B.V. Imidazopyrazine inhibitors of interleukin-2-inducible t-cell kinase
US11891405B2 (en) 2018-09-14 2024-02-06 Chia Tai Tianqing Pharmaceutical Group Co., Ltd. Furo[3,4-b]pyrrole-containing BTK inhibitor
EP3851439A4 (en) * 2018-09-14 2022-06-08 Chia Tai Tianqing Pharmaceutical Group Co., Ltd. BTK INHIBITOR CONTAINING FURO[3,4-B]PYRROLE
AU2019339994B2 (en) * 2018-09-14 2023-02-16 Chia Tai Tianqing Pharmaceutical Group Co., Ltd. Furo[3,4-b]pyrrole-containing BTK inhibitor
JP7493495B2 (ja) 2018-09-14 2024-05-31 チア タイ ティエンチン ファーマシューティカル グループ カンパニー リミテッド フロ[3,4-b]ピロール含有BTK阻害剤
WO2020188015A1 (en) 2019-03-21 2020-09-24 Onxeo A dbait molecule in combination with kinase inhibitor for the treatment of cancer
WO2021089791A1 (en) 2019-11-08 2021-05-14 INSERM (Institut National de la Santé et de la Recherche Médicale) Methods for the treatment of cancers that have acquired resistance to kinase inhibitors
WO2021148581A1 (en) 2020-01-22 2021-07-29 Onxeo Novel dbait molecule and its use
WO2022140246A1 (en) 2020-12-21 2022-06-30 Hangzhou Jijing Pharmaceutical Technology Limited Methods and compounds for targeted autophagy
WO2023122594A1 (en) * 2021-12-21 2023-06-29 Corcept Therapeutics Incorporated Bicyclic indazole glucocorticoid receptor antagonists
WO2023122600A1 (en) * 2021-12-21 2023-06-29 Corcept Therapeutics Incorporated Piperazine indazole glucocorticoid receptor antagonists
TWI835476B (zh) 2021-12-21 2024-03-11 美商科賽普特治療學股份有限公司 哌嗪吲唑糖皮質素受體拮抗劑

Also Published As

Publication number Publication date
US10793576B2 (en) 2020-10-06
CN106831787A (zh) 2017-06-13
EP3572414A1 (en) 2019-11-27
EP3572414A4 (en) 2020-07-22
US20190315758A1 (en) 2019-10-17
CN106831787B (zh) 2018-10-23
TWI668221B (zh) 2019-08-11
TW201827434A (zh) 2018-08-01
JP2020505323A (ja) 2020-02-20
JP6884863B2 (ja) 2021-06-09

Similar Documents

Publication Publication Date Title
TWI668221B (zh) 用作布魯頓酪氨酸激酶抑制劑的化合物及其製備方法和用途
CN106083887B (zh) 抑制btk和/或jak3激酶活性的化合物
JP6576325B2 (ja) ヘテロアリール化合物およびそれらの使用
RU2674701C2 (ru) Аминопиридазиноновые соединения в качестве ингибиторов протеинкиназы
TWI398439B (zh) 特定經取代醯胺、其製法及其用途
CN109983007A (zh) 酰胺类衍生物抑制剂及其制备方法和应用
RU2569635C2 (ru) Замещенные пиридопиразины как новые ингибиторы syk
JP6948659B1 (ja) ピリダジニルチアアゾールカルボキシアミド化合物
AU2015266453C1 (en) Alk kinase inhibitor, and preparation method and use thereof
CA2818545A1 (en) Heterocyclic-substituted pyrrolopyridines and pyrrolopyrimidines as jak inhibitors
TWI824403B (zh) 一種雜環化合物、其中間體、其製備方法及其應用
KR20230028522A (ko) 헤테로고리 화합물 및 이의 용도
CN109867675B (zh) 一种吡咯并嘧啶衍生的化合物、药物组合物以及其用途
WO2014146249A1 (en) Geminally substituted cyanoethylpyrazolo pyridones as janus kinase inhibitors
TWI723480B (zh) 用作fgfr4抑制劑的稠環衍生物
EP4194457A1 (en) Compound for targeting and degrading protein, and preparation method therefor and use thereof
TWI804266B (zh) Tyk2抑制劑及其用途
JP6885962B2 (ja) 1,4−ジカルボニル−ピペリジル誘導体
JP2022515309A (ja) 置換アリール化合物、その製造方法及び用途
EP2981264B1 (en) Inhibitors of bruton's tyrosine kinase
CN110114355A (zh) 用于治疗癌症的噁唑衍生物
WO2023241608A1 (en) Csf-1r inhibitors and uses thereof
WO2024017372A1 (zh) 一种吲哚酮衍生物及其应用
TW202237124A (zh) 作為mrgprx2拮抗劑的化合物
WO2020135488A1 (zh) 2,4-二氨基嘧啶衍生物及其用途

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: 17893058

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2019531745

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2017893058

Country of ref document: EP

Effective date: 20190820