CN108329274B - Bruton's tyrosine kinase inhibitors - Google Patents

Bruton's tyrosine kinase inhibitors Download PDF

Info

Publication number
CN108329274B
CN108329274B CN201810020706.2A CN201810020706A CN108329274B CN 108329274 B CN108329274 B CN 108329274B CN 201810020706 A CN201810020706 A CN 201810020706A CN 108329274 B CN108329274 B CN 108329274B
Authority
CN
China
Prior art keywords
compound
alkyl
reaction
added
dmso
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201810020706.2A
Other languages
Chinese (zh)
Other versions
CN108329274A (en
Inventor
张寅生
任景
高勇
赵大敏
周宇
王庆璘
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Chia Tai Tianqing Pharmaceutical Group Co Ltd
Original Assignee
Chia Tai Tianqing Pharmaceutical Group Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Chia Tai Tianqing Pharmaceutical Group Co Ltd filed Critical Chia Tai Tianqing Pharmaceutical Group Co Ltd
Publication of CN108329274A publication Critical patent/CN108329274A/en
Application granted granted Critical
Publication of CN108329274B publication Critical patent/CN108329274B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F5/00Compounds containing elements of Groups 3 or 13 of the Periodic Table
    • C07F5/02Boron compounds
    • C07F5/025Boronic and borinic acid compounds
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/48Two nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic Table
    • C07F7/02Silicon compounds
    • C07F7/08Compounds having one or more C—Si linkages
    • C07F7/0803Compounds with Si-C or Si-Si linkages
    • C07F7/081Compounds with Si-C or Si-Si linkages comprising at least one atom selected from the elements N, O, halogen, S, Se or Te
    • C07F7/0812Compounds with Si-C or Si-Si linkages comprising at least one atom selected from the elements N, O, halogen, S, Se or Te comprising a heterocyclic ring
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic Table
    • C07F7/02Silicon compounds
    • C07F7/08Compounds having one or more C—Si linkages
    • C07F7/0803Compounds with Si-C or Si-Si linkages
    • C07F7/081Compounds with Si-C or Si-Si linkages comprising at least one atom selected from the elements N, O, halogen, S, Se or Te
    • C07F7/0812Compounds with Si-C or Si-Si linkages comprising at least one atom selected from the elements N, O, halogen, S, Se or Te comprising a heterocyclic ring
    • C07F7/0816Compounds with Si-C or Si-Si linkages comprising at least one atom selected from the elements N, O, halogen, S, Se or Te comprising a heterocyclic ring said ring comprising Si as a ring atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic Table
    • C07F7/02Silicon compounds
    • C07F7/08Compounds having one or more C—Si linkages
    • C07F7/0803Compounds with Si-C or Si-Si linkages
    • C07F7/0825Preparations of compounds not comprising Si-Si or Si-cyano linkages
    • C07F7/083Syntheses without formation of a Si-C bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F7/00Compounds containing elements of Groups 4 or 14 of the Periodic Table
    • C07F7/02Silicon compounds
    • C07F7/08Compounds having one or more C—Si linkages
    • C07F7/0834Compounds having one or more O-Si linkage
    • C07F7/0836Compounds with one or more Si-OH or Si-O-metal linkage

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The application belongs to the field of pharmaceutical chemistry, and particularly relates to a compound shown in a formula (I) or a pharmaceutically acceptable salt thereof, a preparation method thereof and a pharmaceutical composition containing the compound. The application also relates to the use of these compounds or their pharmaceutically acceptable salts and pharmaceutical compositions comprising these compounds in the treatment of bruton's tyrosine kinase related diseases.

Description

Bruton's tyrosine kinase inhibitors
Technical Field
The application belongs to the field of pharmaceutical chemistry, and particularly relates to a Bruton's tyrosine kinase inhibitor, a preparation method thereof, a pharmaceutical composition containing the compound, and application of the Bruton's tyrosine kinase inhibitor in treatment of Bruton's tyrosine kinase related diseases.
Background
Bruton's Tyrosine Kinase (BTK) is mainly expressed in B cells, distributed in the lymphatic, hematopoietic and blood systems, first discovered in 1993, is a member of the non-receptor tyrosine kinase Tec family, which also includes Tec, ITK/TSK/EMT and BMX, and they have high structural homology. BTK plays a crucial role in B cell signaling pathways that link cell surface B Cell Receptor (BCR) stimulation to downstream intracellular responses, and is a key regulator of B cell development, activation, signaling, and survival. In recent years, researches on B cells, particularly B cell non-Hodgkin lymphoma and rheumatoid arthritis show that BTK is often abnormally expressed.
The development of small molecule targeted drugs based on the BTK signaling pathway provides a brand new approach for the treatment of B cell tumors such as leukemia, primary myeloma and B cell immune diseases. The only BTK inhibitor, Ibrutinib, currently marketed, is approved for the treatment of cll (chloroonly leukemia) and mcl (mantle cell lymphoma), rheumatoid arthritis and other immune diseases in clinical research for indications. Other BTK inhibitors include acalabrutinib, evocrutinib, poseltininib, spebrutinib, and the like, and most of them are in early clinical development.
The BTK binding site is mutated to cause the generation of drug resistance, and in addition, the Ibrutinib has more side effects, such as causing second primary malignant tumors including non-skin cancer and non-melanoma skin cancer, the bleeding event occurrence rate reaches 6 percent, and the infection probability of grade 3 or above is 14 to 26 percent. Clinical data show that some patients are prone to Progressive Disease (PD) after drug withdrawal, such as Richter Syndrome (RS) or CLL, and the cells of such patients mostly have 17p chromosome loss or complex karyotype. Molecular biological results reveal that the patients have cell changes after the Ibrutinib is stopped, and intracellular gene mutation is reported to cause the drug resistance mechanism of the Ibrutinib. Therefore, more BTK inhibitors are clinically needed for treating diseases such as tumors and the like, and simultaneously, adverse events can be overcome.
Detailed Description
The present application relates to compounds of formula (I) or a pharmaceutically acceptable salt thereof,
Figure BDA0001543445930000011
wherein the content of the first and second substances,
R1selected from hydrogen, halogen, -CN, C1-6Alkyl, halo C1-6Alkyl radical, C1-6Alkoxy, -C (O) C1-6Alkyl or-C (O) NHC1-6An alkyl group;
R2selected from hydrogen, hydroxy, C1-6Alkoxy, halogen, cyano;
l is selected from NH, NHNH, O;
a is optionally halogen, hydroxy, C1-6Alkyl, halogen substituted C1-6Alkyl, or C1-6Alkoxy-substituted
Figure BDA0001543445930000021
Wherein X-Y is
Figure BDA0001543445930000022
Or
Figure BDA0001543445930000023
m is 1 or 2, R4Selected from hydroxy, C1-4Alkyl radical, C1-4An alkoxy group;
or A is
Figure BDA0001543445930000024
Wherein R is3Selected from hydrogen, hydroxy-substituted C1-6Alkyl radical, C1-4Alkoxy radical C1-4Alkyl, silyl, silicon-containing heterocycles, and silyloxy C1-6Alkyl, said silyl, said silicon-containing heterocycle, and said silyloxy C1-6Alkyl is optionally substituted by hydroxy, cyano, nitro, amino, halogen, or C1-4Alkyl is substituted with the proviso that when R3Is hydroxy, hydroxy-substituted alkylOr alkoxyalkyl, R1Is halo C1-6Alkyl, preferably fluoro C1-4Alkyl, more preferably trifluoromethyl; when R is3When hydrogen, L is NHNH.
In some embodiments, R1Selected from hydrogen, fluorine, chlorine, -CN, C1-4Alkyl, fluoro C1-4Alkyl, chloro C1-4Alkyl, and C1-4An alkoxy group. In some embodiments, R1Selected from fluorine, chlorine, C1-4Alkyl, fluoro C1-4Alkyl, and chloro C1-4An alkyl group. In some embodiments, R1Selected from the group consisting of fluoro, chloro, methyl, and trifluoromethyl.
In some embodiments, R2Selected from hydrogen, hydroxy, and C1-4An alkoxy group; in some embodiments, R2Selected from hydrogen, and methoxy.
In some embodiments, A is optionally substituted with halogen, hydroxy, or C1-6Alkyl substituted
Figure BDA0001543445930000025
Wherein X-Y is
Figure BDA0001543445930000026
Or
Figure BDA0001543445930000027
m is 1 or 2, R4Selected from hydroxy, C1-4Alkyl radical, C1-4An alkoxy group.
In some embodiments, A is optionally halogen, or C1-4Alkyl substituted
Figure BDA0001543445930000028
In some embodiments, a is optionally substituted with fluoro or methyl
Figure BDA0001543445930000029
In some embodiments, X-Y is
Figure BDA00015434459300000210
In some embodiments, R4Is a hydroxyl group.
In some embodiments, a is
Figure BDA0001543445930000031
Figure BDA0001543445930000032
Or
Figure BDA0001543445930000033
In some embodiments, a is
Figure BDA0001543445930000034
Wherein R is3Selected from hydroxy, hydroxy-substituted C1-6Alkyl, and C1-4Alkoxy radical C1-4Alkyl radical, R1Is halo C1-6Alkyl, preferably fluoro C1-4Alkyl, more preferably trifluoromethyl; in some embodiments, a is
Figure BDA0001543445930000035
Wherein R is3Selected from hydroxy-substituted C1-4Alkyl, and C1-4Alkoxy radical C1-4Alkyl radical, R1Is halo C1-6Alkyl, preferably fluoro C1-4Alkyl, more preferably trifluoromethyl; in some embodiments, a is
Figure BDA0001543445930000036
Wherein R is3Selected from the group consisting of hydroxy-substituted methyl, hydroxy-substituted isopropyl, methoxymethyl, R1Is trifluoromethyl.
In some embodiments, a is
Figure BDA0001543445930000037
Wherein R is3Is hydrogen and L is NHNH.
In some embodiments, a is
Figure BDA0001543445930000038
Wherein R is3Selected from the group consisting of silyl, silicon-containing heterocycles, and silyloxy C1-6Alkyl, said silyl, said silicon-containing heterocycle, or said silyloxy C1-6Alkyl is optionally substituted by hydroxy, cyano, nitro, amino, halogen, or C1-4Alkyl substitution.
In some embodiments, a is
Figure BDA0001543445930000039
Wherein R is3Selected from the group consisting of silyl, silicon-containing heterocycles, and silyloxy C1-4Alkyl, said silyl, said silicon-containing heterocycle, and said silyloxy C1-4Alkyl optionally substituted by hydroxy, or C1-4Alkyl substitution.
In some embodiments, a is
Figure BDA00015434459300000310
Wherein R is3Is selected from
Figure BDA00015434459300000311
Figure BDA00015434459300000312
And
Figure BDA00015434459300000313
in some embodiments, the compounds of formula (I) of the present application relate to compounds of formula (II) or a pharmaceutically acceptable salt thereof,
Figure BDA0001543445930000041
wherein R is1、R2L, X-Y and m are as defined above.
In some embodiments, the compounds of formula (I) of the present application relate to compounds of formula (III) or a pharmaceutically acceptable salt thereof,
Figure BDA0001543445930000042
wherein R is1、R2、R3And L is as defined above.
In some embodiments, the compounds of formula (I) of the present application relate to the following compounds or pharmaceutically acceptable salts thereof:
Figure BDA0001543445930000043
Figure BDA0001543445930000051
Figure BDA0001543445930000061
in another aspect, the present application relates to a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt thereof, as described herein. In some embodiments, the pharmaceutical compositions of the present application further comprise a pharmaceutically acceptable excipient.
In another aspect, the present application relates to a method of treating a BTK mediated disease in a mammal, comprising administering to a mammal, preferably a human, in need of such treatment a therapeutically effective amount of a compound of formula (I) or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof.
In another aspect, the present application relates to the use of a compound of formula (I), or a pharmaceutically acceptable salt thereof, or a pharmaceutical composition thereof, in the manufacture of a medicament for the prevention or treatment of a BTK-mediated disease.
Definition of
The following terms used in the present application have the following meanings, unless otherwise specified. A particular term should not be considered as ambiguous or unclear without special definition, but rather construed according to ordinary meaning in the art. When a trade name appears herein, it is intended to refer to its corresponding commodity or its active ingredient.
The term "substituted" means that any one or more hydrogen atoms on a particular atom is replaced with a substituent, so long as the valence of the particular atom is normal and the substituted compound is stable. When the substituent is oxo (i.e., ═ O), meaning that two hydrogen atoms are substituted, oxo does not occur on the aryl.
The terms "optionally" or "optionally" mean that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not. For example, ethyl is "optionally" substituted with halo, meaning that ethyl may be unsubstituted (CH)2CH3) Monosubstituted (e.g. CH)2CH2F) Polysubstituted (e.g. CHFCH)2F、CH2CHF2Etc.) or completely substituted (CF)2CF3). It will be appreciated by those skilled in the art that any group containing one or more substituents will not incorporate any substitution or substitution pattern which is sterically impossible and/or cannot be synthesized.
Herein Cm-nIt is the moiety that has an integer number of carbon atoms in the given range. E.g. "C1-6By "is meant that the group can have 1 carbon atom, 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms, or 6 carbon atoms.
When any variable (e.g., R) occurs more than one time in the composition or structure of a compound, its definition in each case is independent. Thus, for example, if a group is substituted with 2R, then there are separate options for each R.
The term "halo" or "halogen" refers to fluorine, chlorine, bromine and iodine.
The term "hydroxy" refers to an-OH group.
The term "cyano" refers to the group — CN.
The term "amino" refers to the group-NH2A group.
The term "nitro" means-NO2A group.
The term "alkyl" refers to a group of formula CnH2n+1A hydrocarbon group of (1). The alkyl group may be linear or branched. For example, the term "C1-6Alkyl "means an alkyl group having 1 to 6 carbon atoms (e.g., methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 1-methylbutyl, 2-methylbutyl, 3-methylbutyl, neopentyl, hexyl, 2-methylpentyl, and the like). Similarly, the alkyl portion (i.e., alkyl) of alkoxy, alkylamino, dialkylamino, alkylsulfonyl and alkylthio groups have the same definitions as above.
The term "silyl" refers to a group formed by replacement of a carbon atom in an alkyl group with a silicon atom. In some embodiments, the carbon atom of the alkyl group having 1 to 6 carbon atoms is replaced with a silicon atom. In some embodiments, the carbon atom of the alkyl group having 1 to 4 carbon atoms is replaced with a silicon atom. In some embodiments, the silane group has 1 silicon atom.
The term "alkoxy" refers to-O-alkyl.
The term "cycloalkyl" refers to a carbon ring that is fully saturated and may exist as a single ring, a bridged ring, or a spiro ring. Unless otherwise indicated, the carbocycle is typically a 3 to 10 membered ring. Non-limiting examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, norbornyl (bicyclo [2.2.1] heptyl), bicyclo [2.2.2] octyl, adamantyl, and the like.
The term "heterocycloalkyl" refers to a cyclic group that is fully saturated and may exist as a monocyclic, bicyclic, or spiro ring. Unless otherwise indicated, the heterocyclic ring is typically a 3 to 7 membered ring containing 1 to 3 heteroatoms (preferably 1 or 2 heteroatoms) independently selected from sulfur, oxygen and/or nitrogen. Examples of 3-membered heterocycloalkyl include, but are not limited to, oxiranyl, thietanyl, cycloazethyl, non-limiting examples of 4-membered heterocycloalkyl include, but are not limited to, azetidinyl, oxetanyl, thietanyl, examples of 5-membered heterocycloalkyl include, but are not limited to, tetrahydrofuranyl, tetrahydrothienyl, pyrrolidinyl, isoxazolidinyl, oxazolidinyl, isothiazolidinyl, thiazolidinyl, imidazolidinyl, tetrahydropyrazolyl, pyrrolinyl, dihydrofuranyl, dihydrothienyl, examples of 6-membered heterocycloalkyl include, but are not limited to, piperidinyl, tetrahydropyranyl, tetrahydrothiopyranyl, morpholinyl, piperazinyl, 1, 4-thioxanyl, 1, 4-dioxanyl, thiomorpholinyl, 1, 2-dithianyl, 1, 4-dithianyl, dihydropyridinyl, tetrahydropyranyl, dihydropyranyl, and mixtures thereof, Examples of tetrahydropyranyl, thiopyranyl, 7-membered heterocycloalkyl include, but are not limited to, azepanyl, oxepanyl, thiepanyl. Monocyclic heterocycloalkyl groups having 5 or 6 ring atoms are preferred.
The term "silicon-containing heterocycle" refers to a group formed by replacing a carbon atom in a cycloalkyl group or a heterocycloalkyl group with a silicon atom.
The term "treating" means administering a compound or formulation described herein to prevent, ameliorate or eliminate a disease or one or more symptoms associated with the disease, and includes:
(i) preventing the occurrence of a disease or condition in a mammal, particularly when such mammal is susceptible to the disease condition, but has not yet been diagnosed as having the disease condition;
(ii) inhibiting the disease or disease state, i.e., arresting its development;
(iii) alleviating the disease or condition, i.e., causing regression of the disease or condition.
The term "therapeutically effective amount" means an amount of a compound of the present application that (i) treats or prevents a particular disease, condition, or disorder, (ii) alleviates, ameliorates, or eliminates one or more symptoms of a particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of a particular disease, condition, or disorder described herein. The amount of a compound of the present application that constitutes a "therapeutically effective amount" varies depending on the compound, the disease state and its severity, the mode of administration, and the age of the mammal to be treated, but can be routinely determined by those skilled in the art with their own knowledge and this disclosure.
The term "pharmaceutically acceptable" is intended to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
As the pharmaceutically acceptable salt, for example, a metal salt, an ammonium salt, a salt with an organic base, a salt with an inorganic acid, a salt with an organic acid, a salt with a basic or acidic amino acid, and the like can be mentioned.
The term "pharmaceutical composition" refers to a mixture of one or more compounds of the present application or salts thereof and pharmaceutically acceptable excipients. The purpose of the pharmaceutical composition is to facilitate administration of the compounds of the present application to an organism.
The term "pharmaceutically acceptable adjuvants" refers to those adjuvants which do not have a significant irritating effect on the organism and do not impair the biological activity and properties of the active compound. Suitable adjuvants are well known to those skilled in the art, such as carbohydrates, waxes, water-soluble and/or water-swellable polymers, hydrophilic or hydrophobic materials, gelatin, oils, solvents, water, and the like.
The words "comprise" or "comprise" and variations thereof such as "comprises" or "comprising," are to be understood in an open, non-exclusive sense, i.e., "including but not limited to.
The compounds and intermediates of the present application may also exist in different tautomeric forms, and all such forms are included within the scope of the present application. The term "tautomer" or "tautomeric form" refers to structural isomers of different energies that can interconvert via a low energy barrier. For example, proton tautomers (also referred to as proton transfer tautomers) include interconversion via proton migration, such as keto-enol and imine-enamine isomerizations. A specific example of a proton tautomer is an imidazole moiety, wherein the proton can migrate between two ring nitrogens. Valence tautomers include interconversion by recombination of some of the bonding electrons.
The present application also includes isotopically-labeled compounds of the present application, which are identical to those recited herein, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature. Examples of isotopes that can be incorporated into compounds of the present application include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, iodine and chlorine,such as respectively2H、3H、11C、13C、14C、13N、15N、15O、17O、18O、31P、32P、35S、18F、123I、125I and36cl, and the like.
Certain isotopically-labelled compounds of the present application (e.g. with3H and14c-labeled ones) can be used in compound and/or substrate tissue distribution assays. Tritiated (i.e. by tritiation)3H) And carbon-14 (i.e.14C) Isotopes are particularly preferred for their ease of preparation and detectability. Positron emitting isotopes, such as15O、13N、11C and18f can be used in Positron Emission Tomography (PET) studies to determine substrate occupancy. Isotopically labeled compounds of the present application can generally be prepared by following procedures analogous to those disclosed in the schemes and/or in the examples below, by substituting an isotopically labeled reagent for a non-isotopically labeled reagent.
In addition, heavier isotopes are used (such as deuterium (i.e., deuterium)2H) Substitution may provide certain therapeutic advantages resulting from greater metabolic stability (e.g., increased in vivo half-life or reduced dosage requirements), and thus may be preferred in certain circumstances where deuterium substitution may be partial or complete, partial deuterium substitution meaning that at least one hydrogen is substituted with at least one deuterium.
The compounds of the present application may be asymmetric, e.g., having one or more stereoisomers. Unless otherwise indicated, all stereoisomers include, for example, enantiomers and diastereomers. The compounds of the present application containing asymmetric carbon atoms can be isolated in optically active pure form or in racemic form. The optically active pure form can be resolved from a racemic mixture or synthesized by using chiral starting materials or chiral reagents.
The pharmaceutical compositions of the present application can be prepared by combining the compounds of the present application with suitable pharmaceutically acceptable excipients, for example, can be formulated into solid, semi-solid, liquid or gaseous formulations, such as tablets, pills, capsules, powders, granules, ointments, emulsions, suspensions, suppositories, injections, inhalants, gels, microspheres, aerosols, and the like.
Typical routes of administration of a compound of the present application or a pharmaceutically acceptable salt thereof or a pharmaceutical composition thereof include, but are not limited to, oral, rectal, topical, inhalation, parenteral, sublingual, intravaginal, intranasal, intraocular, intraperitoneal, intramuscular, subcutaneous, intravenous administration.
The pharmaceutical compositions of the present application can be manufactured by methods well known in the art, such as conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, lyophilizing, and the like.
In some embodiments, the pharmaceutical composition is in an oral form. For oral administration, the pharmaceutical compositions may be formulated by mixing the active compounds with pharmaceutically acceptable excipients well known in the art. These adjuvants enable the compounds of the present application to be formulated as tablets, pills, lozenges, dragees, capsules, liquids, gels, slurries, suspensions and the like, for oral administration to a patient.
Solid oral compositions may be prepared by conventional mixing, filling or tableting methods. For example, it can be obtained by the following method: the active compounds are mixed with solid adjuvants, optionally the mixture obtained is milled, if desired with further suitable adjuvants, and the mixture is then processed to granules, to give tablets or dragee cores. Suitable excipients include, but are not limited to: binders, diluents, disintegrants, lubricants, glidants, sweeteners or flavoring agents, and the like.
The pharmaceutical compositions may also be adapted for parenteral administration, as sterile solutions, suspensions or lyophilized products in suitable unit dosage forms.
In all methods of administration of the compounds of the general formula I described herein, the daily dose is from 0.01 to 200mg/kg body weight, preferably from 0.02 to 100mg/kg body weight, in single or divided doses.
The compounds of the present application may be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, embodiments formed by combinations thereof with other chemical synthetic methods, and equivalents thereof known to those skilled in the art, with preferred embodiments including, but not limited to, the examples of the present application.
The chemical reactions of the embodiments herein are carried out in a suitable solvent that is compatible with the chemical changes of the present application and the reagents and materials required therefor. In order to obtain the compounds of the present application, it is sometimes necessary for a person skilled in the art to modify or select the synthesis steps or reaction schemes based on the existing embodiments.
An important consideration in the art of synthetic route planning is the selection of suitable protecting Groups for reactive functional Groups, for example, reference may be made to Greene's Protective Groups in Organic Synthesis (4th Ed.) Hoboken, New Jersey: John Wiley & Sons, Inc. all references cited in this application are incorporated herein in their entirety.
In some embodiments, the compounds of formula (II-1) herein can be prepared by one skilled in the art of organic synthesis via scheme 1 using standard methods in the art:
Figure BDA0001543445930000101
the compound of formula 2 is reacted with a compound of formula 3 (either commercially available or synthesized with reference to CN 2015109825451) under basic conditions to provide an intermediate compound of formula 4. The compound of formula 4 is reacted with boron-containing fragment 5 (commercially available or synthesized with reference to WO2011022337a 1) under acidic conditions to give the final product compound of formula (II-1).
The following abbreviations are used in this application:
BTK: bruton's tyrosine kinase
TLC: thin layer chromatography
eq: represents an equivalent weight
EA: ethyl acetate
PE: petroleum ether
DIPEA: n, N-diisopropylethylamine
EtOH: ethanol
THF: tetrahydrofuran (THF)
DCM: methylene dichloride
HEPES (high efficiency particulate air): 4-hydroxyethyl piperazine ethanesulfonic acid
DTT: dithiothreitol
EGTA: ethylene glycol diethyl ether diamine tetraacetic acid
EDTA-EDTA
DMSO, DMSO: dimethyl sulfoxide
XphOS: 2-dicyclohexylphosphonium-2 ',4',6' -triisopropylbiphenyl
(dba)3Pd2: tris (dibenzylideneacetone) dipalladium
MeOH: methanol
DMF: n, N-dimethylformamide
BnBr: benzyl bromide
DIBAL-H: diisobutylaluminum hydride
OD value: optical density
For clarity, the invention is further illustrated by examples, which do not limit the scope of the application. All reagents used herein were commercially available and used without further purification.
The nuclear magnetic resonance chromatogram (NMR) is measured by using a BRUKER AVANCE III HD 500M nuclear magnetic resonance instrument, the chemical shift takes tetramethylsilane (TMS ═ delta 0.00) as an internal standard, and the format of the nuclear magnetic resonance hydrogen spectrum data record is as follows: proton number, peak type (s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet), coupling constant (in hertz Hz). The instrument used for mass spectrometry was AB SCIEX Triple TOF 4600 or Shimadzu LC-30AD \ Qtrap 3200.
Examples
Example 1: n- (3- ((2- (1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) amino) -5- (trifluoromethyl) pyrimidin-4-yl) phenyl) acrylamide
Figure BDA0001543445930000111
Step (1): under the protection of nitrogen, the mixture is turned to the reverse directionThe intermediate 3-1(3.2g, 19.73mmol, 1eq) was added to a flask and dissolved in n-butanol (20mL), the intermediate 2-1(4.28g, 19.73mmol, 1eq) and DIPEA (3.06g, 23.68mmol, 1.2eq) were added separately at 0 deg.C, the reaction was carried out at room temperature for 3 hours, and the disappearance of the starting material point was detected by TLC. Evaporating most of the reaction solvent to dryness, and adding saturated Na2CO3The aqueous solution was adjusted to pH 10, extracted with EA, dried over anhydrous sodium sulfate, filtered and evaporated to dryness. Passing through a silica gel column, PE: EA elution (5:1 to 2:1) was combined and concentrated to give 6.35g of crude product. Intermediate 4-14.08 g was prepared by high pressure preparative chromatography.
1H-NMR(300M,DMSO-d6):10.22(s,1H),9.57(s,1H),8.58(s,1H),7.79(s,1H),7.49-7.52(d,1H),7.33-7.38(t,1H),7.13-7.16(d,1H),6.40-6.49(m,1H),6.23-6.29(d,1H),5.75-5.78(d,1H).13C-NMR(126M,DMSO-d6):163.14,162.30,157.59,156.71-156.49(m,2C),139.27,137.19,131.71,128.71,126.93,123.17(t),120.91,117.01,116.61.HR-ESIMS:m/z 343.0566 [M+H]+.
Step (2): intermediate 4-1(600mg, 1.75mmol, 1eq) and intermediate 5-1(391mg, 2.1mmol, 1.2eq) were placed in a 35mL microwave tube, 10mL of n-butanol solvent was added, and the starting material was dissolved. 1N HCl 0.175mL was added. Microwave reaction at 110 deg.c for 1 hr, and TLC shows the material is reacted completely. Ethyl acetate (20mL) was added to dilute the reaction. Extracting, washing the organic phase with saturated sodium bicarbonate solution and saturated salt solution, drying with anhydrous sodium sulfate, filtering, and evaporating to obtain yellowish oily substance. Purifying with EtOH \ THF 8:1 mixed solvent to obtain compound I-1350 mg.
1H NMR(500MHz,DMSO-d6)δ10.24(s,1H),9.75(brs,1H),9.13(s,1H),8.73(brs,1H),8.36(s,1H),7.82(d,J=8.5Hz,2H),7.54(d,J=8.5Hz,2H),7.38-7.26(m,1H),7.21(d,J=8.0Hz,1H),7.05(brs,1H),6.45(dd,J=16.9,10.1Hz,1H),6.24(dd,J=16.9,2.1Hz,1H),5.74(dd,J=10.1,2.1Hz,1H),4.88(s,2H).HR-ESIMS:456.1452([M+H]+).
Example 2: n- (3- ((2- (1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-4-yl) amino) -5- (trifluoromethyl) pyrimidin-4-yl) phenyl) acrylamide
Figure BDA0001543445930000121
Intermediate 4-1(1.000g, 2.918mmol,1eq) and 5-2(0.500g, 3.357mmol,1.15eq) were added to a 35mL microwave tube, solvent N-butanol 10mL was added, 1N HCl 0.7mL was added. Microwave reaction at 110 deg.c for 1 hr, and TLC shows the material is reacted completely. To the reaction mixture was added 200mL of ethyl acetate, and the mixture was washed with 50mL of a saturated aqueous solution of sodium hydrogencarbonate and 50mL of a saturated brine in this order, and the organic phase was dried over anhydrous sodium sulfate. Filtering, concentrating the filtrate, making sand, and performing column chromatography to obtain compound I-2100 mg.
1H NMR(500MHz,DMSO-d6)δ10.20(s,1H),9.18(s,1H),9.09(s,1H),8.65(s,1H),8.34(s,1H),7.82(s,1H),7.64(d,J=7.5Hz,1H),7.48(d,J=7.5Hz,1H),7.40(d,J=6.5Hz,1H),7.24-7.21(m,1H),7.14-7.13(m,1H),6.50-6.44(m,1H),6.25(d,J=17,1H),5.75(d,J=,10Hz,1H),4.98(s,2H).MS(ESI):m/z 456.1[M+H]+.
Example 3: n- (3- ((5-fluoro-2- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxaborzol-4-yl) amino) pyrimidin-4-yl) amino) acrylamide
Figure BDA0001543445930000122
Step (1): adding the intermediate 2-2(12.00g, 71.87mmol,1.0eq) and 3-1(13.99g, 86.24mmol, 1.2eq) into a reaction bottle, adding n-butanol (100mL), adding DIPEA (13.93g, 107.80mmol, 1.5eq) under stirring, reacting at room temperature under nitrogen protection overnight, and detecting the disappearance of a raw material point by TLC. Most of the reaction solvent is evaporated to dryness, water is added into the residue, solid is separated out by ultrasonic treatment, the filtration is carried out, and the filter cake is dried to obtain 4-223.21 g of intermediate.
1H-NMR(500M,DMSO-d6):10.20(s,1H),10.02(s,1H),8.31-8.32(d,J=3.5Hz,1H),8.01(s,1H),7.44(s,1H),7.42(s,1H),7.31-7.35(t,J=8.5Hz,1H),6.45-6.51(m,1H),6.26-6.29(dd,J1=17Hz,1.5Hz,1H),5.75-5.78(dd,J=10Hz,1.5Hz,1H).HR-ESIMS:m/z 293.0598[M+H]+.
Step (2): intermediate 4-2(574mg, 1.961mmol, 1eq) and 5-2(0.400g,2.157mmol,1.1eq) were added to a 35mL microwave tube, solvent N-butanol 10mL was added, 1N HCl 0.7mL was added. Microwave reaction at 110 deg.c for 1 hr, and TLC shows the material is reacted completely. To the reaction mixture was added 200mL of ethyl acetate, and the mixture was washed with 50mL of a saturated aqueous solution of sodium hydrogencarbonate and 50mL of a saturated brine in this order, and the organic phase was dried over anhydrous sodium sulfate. Filtering, concentrating the filtrate, making sand, and performing column chromatography to obtain compound I-370 mg.
1H NMR(500MHz,DMSO-d6)δ10.22(s,1H),9.37(s,1H),9.08(s,1H),8.64(s,1H),8.10(s,1H),8.06(s,1H),7.79(d,J=7Hz,1H),7.44-7.39(m,3H),7.24-7.23(m,1H),7.18-7.15(m,1H),6.54-6.49(m,1H),6.26(d,J=16.5Hz,1H),5.75(d,J=,10.5Hz,1H),5.03(s,2H).MS(ESI):m/z 406.1[M+H]+.
Example 4: n- (3- ((2- (1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-5-yl) amino) -5- (trifluoromethyl) pyrimidin-4-yl) phenyl) acrylamide
Figure BDA0001543445930000131
Intermediate 4-1(500mg, 1.459mmol, 1eq) and 5-3(271mg,1.824mmol,1.25eq) were charged to a 50mL reaction flask, solvent n-butanol 10mL was added, and trifluoroacetic acid (500mg, 4.377mmol, 3eq) was added. The reaction was heated to reflux for 2 hours and TLC showed that all starting materials were completely reacted. Cooling to room temperature, filtering, washing the filter cake with water, draining, adjusting pH of the filter cake to be alkaline with 100mL of saturated sodium bicarbonate aqueous solution, extracting with 10% methanol dichloromethane solution, mixing the organic phases, drying with anhydrous sodium sulfate, filtering, concentrating the filtrate, preparing sand, and performing column chromatography to obtain the compound I-4261 mg.
1H NMR(500MHz,DMSO-d6)δ10.22(s,1H),9.87(s,1H),8.90(s,1H),8.39(s,1H),7.78(s,1H),7.67(s,1H),7.62(d,J=7Hz,1H),7.46-7.39(m,2H),7.33(m,1H),7.13(d,J=6.5Hz,1H),6.45-6.40(m,1H),6.26(d,J=16.5Hz,1H),5.75(d,J=,10Hz,1H),4.66(s,2H).MS(ESI):m/z 456.1[M+H]+,454.3(M-H)-.
Example 5: n- (3- ((5-fluoro-2- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxaborzol-5-yl) amino) pyrimidin-4-yl) amino) acrylamide
Figure BDA0001543445930000141
Intermediate 4-2(500mg, 1.708mmol, 1eq) and 5-3(318mg,2.135mmol,1.25eq) were charged to a 50mL reaction flask, solvent n-butanol 10mL was added, and trifluoroacetic acid (584mg, 4.377mmol, 3eq) was added. The reaction was heated to reflux for 2 hours and TLC showed that a small amount of starting material was not reacted. Cooling to room temperature, adjusting pH to alkaline with 100mL saturated sodium bicarbonate solution, extracting with ethyl acetate for 3 times, mixing organic phases, drying with anhydrous sodium sulfate, filtering, concentrating the filtrate, making sand, and performing column chromatography to obtain compound I-5120 mg.
1H NMR(500MHz,DMSO-d6)δ10.13(s,1H),9.46(s,1H),9.37(s,1H),8.86(s,1H),8.14(d,J=3Hz,1H),7.93(s,1H),7.88(s,1H),7.52-7.43(m,4H),7.34-7.31(m,1H),6.48-6.42(m,1H),6.25(d,J=17Hz,1H),5.75(d,J=10Hz,1H),4.80(s,2H).MS(ESI):m/z 406.1[M+H]+,428.0[M+Na]+,404.2(M-H)-.
Example 6: n- (3- ((5-fluoro-2- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxaborzol-6-yl) amino) pyrimidin-4-yl) amino) acrylamide
Figure BDA0001543445930000142
Intermediate 4-2(500mg, 1.708mmol, 1eq) and 5-1(318mg,2.135mmol,1.25eq) were charged to a 50mL reaction flask, solvent n-butanol 10mL was added, and trifluoroacetic acid (584mg, 4.377mmol, 3eq) was added. The reaction was heated to reflux for 2 hours and TLC showed the starting material was completely reacted. Cooling to room temperature, adjusting pH to alkaline with 100mL saturated sodium bicarbonate solution, extracting with 10% methanol dichloromethane solution for 3 times, mixing organic phases, drying with anhydrous sodium sulfate, filtering, concentrating the filtrate, making sand, and performing column chromatography to obtain compound I-6130 mg.
1H NMR(500MHz,DMSO-d6)δ10.06(s,1H),9.38(s,1H),9.12(s,1H),9.07(s,1H),8.10(d,J=3.5Hz,1H),7.94(s,1H),7.87(s,1H),7.83-7.82(m,1H),7.62(d,J=8Hz,1H),7.40(d,J=8Hz,1H),7.27(t,J=8Hz,1H),7.20(d,J=8Hz,1H),6.48-6.43(m,1H),6.25(dd,J=1.5,16Hz,1H),5.75(dd,J=1.5,10Hz,1H),4.91(s,2H).MS(ESI):m/z 406.1[M+H]+.
Example 7: n- (3- ((5-chloro-2- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxaborazol-6-yl) amino) pyrimidin-4-yl) amino) phenyl) acryloyl amide
Figure BDA0001543445930000151
Step (1): the intermediate 2-3(10.00g, 54.52mmol,1.0eq) and 3-1(10.61g, 65.42mmol,1.2eq) were added to a reaction flask, n-butanol (100mL) was added, DIPEA (10.57g, 81.78mmol, 1.5eq) was added with stirring, and the mixture was reacted at 50 ℃ in an oil bath under nitrogen protection for 3 hours with disappearance of the starting material point detected by TLC. Most of the reaction solvent is evaporated to dryness, the residue is added with diluted hydrochloric acid to be adjusted to weak acidity, solid is precipitated by ultrasonic treatment, the filtration is carried out, and the filter cake is dried to obtain 4-317.93 g of intermediate.
1H-NMR(500M,DMSO-d6):10.20(s,1H),9.55(s,1H),8.38(s,1H),7.92(s,1H),7.46-7.48(d,J=8Hz,1H),7.33-7.36(t,J=8Hz,1H),7.28-7.30(d,J=8Hz,1H),6.44-6.50(m,1H),6.26-6.29(dd,J=17Hz,1.5Hz,1H),5.76-5.78(dd,J=10Hz,1.5Hz,1H).HR-ESIMS:
309.0305([M+H]+).
Step (2): intermediate 4-3(500mg, 1.617mmol, 1eq) and 5-1(289mg,1.941mmol,1.2eq) were added to a 35mL microwave reaction tube, 12mL solvent 2-butanol was added, and trifluoroacetic acid (663mg, 5.822mmol, 3eq) was added. Microwave, 140 ℃, and reacting for 1 hour, wherein TLC shows that the raw materials are completely reacted. Cooling to room temperature, filtering, adjusting pH of the filtrate to alkaline with 100mL saturated sodium bicarbonate solution, extracting with 10% methanol dichloromethane solution for 3 times, combining organic phases, drying with anhydrous sodium sulfate, filtering, concentrating the filtrate, making sand, and performing column chromatography to obtain compound I-780 mg.
1H NMR(500MHz,DMSO-d6)δ10.29(s,1H),9.40(s,1H),9.09(s,1H),8.82(s,1H),8.15(s,1H),8.02(s,1H),7.85(dd,J=1.5and 8Hz,1H),7.82(s,1H),7.52(d,J=8Hz,1H),7.40(d,J=9Hz,1H),7.29(t,J=8Hz 1H),7.15(d,J=8.5Hz,1H),6.50(dd,J=10and 17Hz,1H),6.25(dd,J=2and 17Hz,1H),5.75(dd,J=2and 10Hz,1H),4.90(s,2H).MS(ESI):m/z 422.0[M+H]+.
Example 8: n- (3- ((2-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxaborzol-6-yl) amino) -5-methylpyrimidin-4-yl) amino) phenyl)
Figure BDA0001543445930000152
Step (1): adding the intermediate 2-4(10.00g, 61.35mmol,1.0eq) and 3-1(14.92g, 92.02mmol,1.5eq) into a reaction bottle, adding n-butanol (100mL), adding DIPEA (15.86g, 122.70mmol, 2.0eq) under stirring, refluxing in an oil bath for 12 hours under the protection of nitrogen, and detecting the disappearance of a raw material point by TLC. Most of the reaction solvent is evaporated to dryness, the residue is adjusted to be faintly acid by adding diluted hydrochloric acid, DCM-MeOH (10:1 extraction) is added, an organic layer is washed by saturated saline water and dried, then a silica gel column is passed, and DCM-MeOH is eluted, so that 4-410.00 g of an intermediate is obtained.
1H-NMR(500M,DMSO-d6):10.16(s,1H),8.89(s,1H),8.04(s,1H),7.97(s,1H),7.37-7.40(t,J=7Hz,2H),7.30-7.33(t,J=8Hz,1H),6.45-6.51(m,1H),6.26-6.30(dd,J=17Hz,1.5Hz,1H),5.75-5.77(dd,J=10Hz,1.5Hz,1H),2.18(s,3H).HR-ESIMS:m/z 289.0845[M+H]+.
Step (2): intermediate 4-4(500mg, 1.732mmol, 1eq) and 5-1(309mg, 2.078mmol,1.2eq) were added to a 35mL microwave reaction tube, 12mL of solvent 2-butanol was added, and trifluoroacetic acid (709mg, 6.223mmol, 3eq) was added. Microwave, 140 ℃, and reacting for 1 hour, wherein TLC shows that the raw materials are completely reacted. Cooling to room temperature, filtering, adjusting pH of the filtrate to alkaline with 100mL saturated sodium bicarbonate solution, extracting with 10% methanol dichloromethane solution for 3 times, combining organic phases, drying with anhydrous sodium sulfate, filtering, concentrating the filtrate, making sand, and performing column chromatography to obtain compound I-8220 mg.
1H NMR(500MHz,DMSO-d6)δ10.06(s,1H),9.06(s,1H),8.91(s,1H),8.33(s,1H),7.97(s,1H),7.92-7.90(m,2H),7.83(s,1H),7.52(d,J=8Hz,1H),7.38(d,J=8.5Hz,1H),7.27(t,J=8Hz 1H),7.15(d,J=8.5Hz,1H),6.45(dd,J=10.5and 17Hz,1H),6.25(dd,J=2and 17Hz,1H),5.75(m,1H),4.90(s,2H),2.13(s,3H).MS(ESI):m/z 402.1[M+H]+,424.1[M+Na]+.
Example 9: n- (3- ((2- (3- (hydroxymethyl) phenyl) amino) -5- (trifluoromethyl) pyrimidin-4-yl) amino) phenyl)
Figure BDA0001543445930000161
Adding the intermediate 4-1(500mg, 1.459mmol, 1eq) and 5-4(271mg, 1.824mmol,1.25eq) into 12mL of 2-butanol, adding trifluoroacetic acid (500mg, 4.377mmol, 3eq), reacting at 140 ℃ for 1 hour under microwave, separating out white solid, determining the reaction is complete by TLC, pouring the reaction solution into 100mL of saturated aqueous sodium bicarbonate solution, extracting for 3 times (methanol/dichloromethane), drying organically, filtering, concentrating, and performing column chromatography to obtain the compound I-990 mg.
1H NMR(500MHz,DMSO-d6)δ10.16(s,1H),9.63(s,1H),8.74(s,1H),8.37(s,1H),7.77(s,1H),7.57(d,J=8Hz,1H),7.48(d,J=7.5Hz,1H),8.39(s,1H),7.35(t,J=8Hz,1H),7.19(d,J=7.5Hz,1H),7.00(m,1H),6.87(d,J=7.5Hz,1H),6.44(dd,J=10and 17Hz,1H),6.25(dd,J=1.5and 17Hz,1H),5.76(m,1H),5.03(t,J=5.5Hz,1H),4.31(d,J=4.5Hz,2H).MS(ESI):m/z 430.0[M+H]+,452.0[M+Na]+.
Example 10: n- (3- ((2- (1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-7-yl) amino) -5- (trifluoromethyl) pyrimidin-4-yl) phenyl) acrylamide
Figure BDA0001543445930000171
Step (1): dissolving the intermediate 5-4(4.060g, 22.690mmol, 1eq) and 2-1(4.923g, 22.690mmol, 1eq) in 100mL of isopropanol, adding DIPEA (5.865g, 45.379mmol, 2eq) and stirring at room temperature overnight, precipitating white solid, confirming complete reaction by TLC, filtering, concentrating the filtrate, preparing sand, and performing column chromatography to obtain the compound 6-11.140 g.
1H NMR(500MHz,DMSO-d6)δ9.40(s,1H),8.93(s,1H),8.66(s,1H),7.95(d,J=8Hz,1H),7.56(t,J=3Hz,1H),7.25(d,J=7.5Hz,1H),5.04(s,2H).MS(ESI):m/z 330.0[M+H]+.
Step (2): dissolving the intermediate 6-1(300mg, 0.911mmol, 1eq) and 3-1(147.7mg, 0.911mmol, 1eq) in 2-butanol, adding DIPEA (176mg, 1.366mmol, 1.5eq), stirring overnight at room temperature, TLC to confirm the reaction is complete, concentrating the reaction solution, adding 2-butanol to concentrate with DIPEA for 2 times, pulping the residue with 30mL dichloromethane, filtering, and drying to obtain the compound I-10371 mg.
1H NMR(500MHz,DMSO-d6)δ10.91(s,1H),10.36(s,1H),9.47(s,1H),8.68(s,1H),7.86(s,1H),7.70(d,J=8Hz,1H),7.42(t,J=8Hz,1H),7.29(d,J=8Hz,1H),7.01-6.98(m,2H),6.70(d,J=6.5Hz,1H),6.51(dd,J=10and 16.5Hz,1H),6.28(d,J=17Hz,1H),5.78(d,J=10.5Hz,1H),4.43(d,J=13.5Hz,1H),4.10(d,J=13.5Hz,1H).MS(ESI):m/z 456.0[M+H]+,478.0[M+Na]+.
Example 11: n- (3- ((2- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxazol-6-yl) amino) -5- (trifluoromethyl) pyrimidin-4-yl) amino) -4-methoxyphenyl) propenylamide
Figure BDA0001543445930000172
Step (1): adding 8.7g of compound 1-1 into a 250mL three-necked flask under the protection of nitrogen, adding 40mL of dry THF into the flask, adding 4.8mL of triethylamine into the reaction flask, cooling the reaction solution to 0 ℃, dropwise adding 8.65mL of acryloyl chloride, keeping the temperature of the reaction solution at about 0 ℃ in the dropwise adding process, heating to room temperature to react for 3 hours after the dropwise adding is finished, concentrating the reaction solution, washing and extracting residues with 100mL of water and 100mL of ethyl acetate, washing the residues with saturated organic phase sodium chloride, drying, concentrating, stopping concentrating immediately when solid is separated out in the concentrating process, placing the solution in an ice water bath for 15 minutes, filtering, taking a filter cake, and filtering by concentration twice to obtain 8.6g of compound 1-2.
1H-NMR(500M,DMSO):10.33(s,1H),8.32-8.33(m,1H),7.81-7.83(m,1H),7.35-7.37(m,1H),6.26-6.30(m,1H),5.77-5.80(m,1H),3.90(s,3H).HRMS(ESI,[M+H]+)m/z:223.0761.
Step (2): 1.8g of the compound 1-2 was taken and added to a 250mL three-necked flask, 50mL of methanol and 55mL of a saturated aqueous ammonium chloride solution were sequentially added to the flask, stirred at room temperature, and 2.68g of zinc powder was weighed and added to the flask. Oil bath reaction at 60 ℃, TLC monitoring reaction completion, reaction liquid filtration, filtrate concentration to remove methanol, ethyl acetate 10mL × 2 extraction, liquid separation, organic phase drying, concentration, residue silica gel column purification to obtain 1.08g of compound 3-2.
1H-NMR(500M,DMSO):9.69(s,1H),7.02-7.03(m,1H),6.80-6.83(m,1H),6.70-6.71(m,1H),6.37-6.42(dd,J=10Hz,25Hz,1H),6.16-6.20(dd,J=1.9Hz,17Hz,1H),5.64-5.67(dd,J=1.9Hz,10.1Hz,1H),4.72(s,2H),3.72(s,3H).HRMS(ESI,[M+H]+)m/z:193.1003.
And (3): adding intermediate 2-1(12.00g, 55.31mmol,1.0eq), 1, 2-dichloroethane (50mL) and tert-butanol (50mL) into a 500mL three-neck round-bottom flask, adding triethylamine (11.19g, 110.61mmol, 2.0eq) under stirring, dropwise adding a mixed solution of 1, 2-dichloroethane and tert-butanol (50mL +50mL) of intermediate 3-2(10.63g, 55.31mmol,1.0eq) at 0 ℃, keeping the temperature for reaction for 1 hour after dropping, slowly raising the temperature to room temperature for reaction for 3 hours, monitoring by TLC that a raw material point disappears, adding diluted hydrochloric acid to near neutrality after solvent rotary evaporation, extracting and drying EA, and then carrying out high-pressure preparative chromatography to obtain intermediate 4-53.36 g.
1H-NMR(500M,DMSO-d6):10.14(s,1H),9.15(s,1H),8.56(s,1H),7.78(s,1H),7.56-7.58(dd,J1=9Hz,J2=2.5Hz,1H),7.11-7.13(d,J=8.5Hz,1H),6.41--6.46(m,1H),6.22-6.26(dd,J1=17Hz,J2=1.5Hz,1H),5.73-5.75(dd,J1=10Hz,J2=1.5Hz,1H),3.76(s,3H).HR-ESIMS:m/z 373.0743[M+H]+.
And (4): weighing 4-5(270mg, 724umol,1eq) of intermediate and 5-1(130mg, 869mmol, 1.2eq) of intermediate in a 35mL microwave reaction tube, adding 4mL of n-butanol, stirring, adding a catalytic amount of hydrochloric acid solution, reacting for one hour at 110 ℃ by microwave, cooling after reaction, stirring overnight to precipitate a solid, performing suction filtration to obtain a filter cake, adding water into the filter cake, and performing ultrasonic treatment to obtain a compound I-11190 mg.
1H-NMR(500M,DMSO-d6),10.06(s,1H),9.75(s,1H),9.10(s,1H),8.36(s,1H),8.18(br,1H),7.98(br,1H),7.80-7.82(dd,J=8.5Hz,1.5Hz,1H),7.61-7.63(d,J=8.5Hz,1H),7.10-7.12(d,J=8.5Hz,1H),7.05(s,1H),6.40-6.45(m,1H),6.20-6.24(dd,J=17Hz,1.5Hz,1H),5.71-5.74(dd,J=10Hz,1.5Hz,1H),4.88(s,2H),3.78(s,3H),3.39-3.40(t,1H).HR-ESIMS:m/z 486.1568[M+H]+.
Example 12:
Figure BDA0001543445930000191
weighing 4-1(400mg, 1.17mmoL and 1.0eq) of intermediate and 5-5(209mg, 1.28mmoL and 1.1eq) of intermediate in a reaction flask, adding 25mL of 2-butanol, stirring, adding trifluoroacetic acid (399mg, 3.50mmoL and 3.0eq), heating at 110 ℃ in an oil bath under the protection of nitrogen for 3 hours, monitoring the complete reaction of raw materials by TLC, evaporating reaction liquid by rotation, adding water-dichloromethane to residues, stirring by ultrasound, adding insoluble substances, drying filter cakes after suction filtration, stirring by ultrasound by dichloromethane-methanol, drying filter cakes after suction filtration, and obtaining 470mg of compound I-12 in total.
1H-NMR(500M,DMSO-d6)δ:10.14(s,1H),9.51(s,1H),8.67(s,1H),8.33-8.29(m,2H),7.76-7.74(m,2H),7.58-7.53(m,2H),7.31(t,1H),7.20(d,1H),6.86(s,1H),6.44(dd,J=17.0,10.5Hz,1H),6.26(dd,J=17.0,2.0Hz,1H),5.76-5.74(m,1H),4.02(t,2H),2.74(t,2H).HR-ESIMS:m/z 470.1608[M+H]+.
Example 13:
Figure BDA0001543445930000192
weighing intermediate 4-1(400mg, 1.17mmoL, 1.0eq) and intermediate 5-6(227mg, 1.28mmoL, 1.1eq) in a reaction flask, adding 25mL of 2-butanol, stirring, adding trifluoroacetic acid (399mg, 3.50mmoL, 3.0eq), heating at 110 ℃ in an oil bath under the protection of nitrogen for 3 hours, monitoring the reaction completion of raw materials by TLC, evaporating reaction liquid by rotation, adding water-dichloromethane to residues, stirring by ultrasound, adding insoluble substances, filtering, drying filter cakes, stirring by ultrasound with dichloromethane, and drying the filter cakes after filtering to obtain 277mg of compound I-13 in total.
1H-NMR(500M,DMSO-d6)δ:10.25(s,1H),9.68(s,1H),8.92(s,1H),8.68(s,1H),8.35(s,1H),7.80(d,J=7.0Hz,2H),7.58(d,J=8.0Hz,2H),7.32(t,1H),7.20(d,J=7.5Hz,1H),7.05(s,1H),6.47(dd,J=17.0,10.0Hz,1H),6.26(dd,J=17.0,2.0Hz,1H),5.74(dd,J=10.0,2.0Hz,1H),1.39(s,6H).HR-ESIMS:m/z 484.1768[M+H]+.
Example 14:
Figure BDA0001543445930000201
weighing intermediate 4-1(400mg, 1.17mmoL, 1.0eq) and intermediate 5-7(214mg, 1.28mmoL, 1.1eq) in a reaction flask, adding 25mL of 2-butanol, stirring, adding trifluoroacetic acid (399mg, 3.50mmoL, 3.0eq), heating at 110 ℃ for 3 hours under the protection of nitrogen, monitoring the reaction completion of raw materials by TLC, evaporating reaction liquid by spinning, adding water and dichloromethane-methanol (10:1) into residues, extracting twice, combining organic layers, washing with saturated saline, drying, performing suction filtration, concentrating the filtrate to near dryness, and purifying by TLC (petroleum ether-ethyl acetate development) to obtain compound I-14 accounting for 171 mg.
1H-NMR(500M,DMSO-d6)δ:10.03(s,1H),9.24(s,1H),9.16(s,1H),8.51(s,1H),8.31(s,1H),7.73–7.63(m,2H),7.36(d,J=6.1Hz,1H),7.23(t,J=9.3Hz,2H),7.18–7.09(m,1H),6.43(dd,J=17.0,10.1Hz,1H),6.25(dd,J=17.0,1.9Hz,1H),5.74(dd,J=10.1,1.9Hz,1H),4.95(s,2H).HR-ESIMS:m/z 474.1348[M+H]+.
Example 15:
Figure BDA0001543445930000202
mixing intermediate 4-1(500mg,1.50mmol,1.0eq), phenylhydrazine (intermediate 5-8, 320mg,2.95mmol,1.97eq), XphOS (320mg,0.68mmol,0.45eq), (dba)3Pd2(70mg,0.075mmol,0.05eq), potassium carbonate (830mg,6.0mmol,4.0eq) and tert-butanol (20mL) were added to a 35mL microwave tube, nitrogen was bubbled for 1 min, and the reaction was stopped by heating the reactor 50W to 110 ℃ for 30 min. When the temperature of the system is reduced to room temperature, filtering by diatomite to remove insoluble substances, and spin-drying the filtrate. The residue was purified by column chromatography to give 150mg of the compound, followed by dichloromethane: hexane ═ 8 mL: pulping 2mL, and filtering to obtain 120mg of the compound I-15.
1H NMR(500MHz,DMSO-d6)δ10.06(s,1H),9.20(d,J=177.4Hz,1H),8.53-8.18(m,2H),7.90(s,1H),7.75-7.15(m,3H),7.09(t,J=7.2Hz,2H),6.97(d,J=37.2Hz,1H),6.65(s,3H),6.46(dd,J=16.8,10.2Hz,1H),6.28(d,J=16.9Hz,1H),5.77(d,J=10.2Hz,1H).ESIMS:m/z 415.3[M+H]+.
Example 16:
Figure BDA0001543445930000211
intermediate 4-1(500mg,1.45mmol) and intermediate 5-9(447mg,1.74mmol) were charged into a 100mL single neck flask, 2-butanol (12mL) was added, trifluoroacetic acid (0.3mL,4.35mmol) was added with stirring, nitrogen was replaced, and the mixture was heated in an oil bath at 120 ℃ with stirring for 5.5h, and the heating was stopped. After the system cooled to room temperature, 7M NH was added thereto3MeOH, pH 8 adjusted solution. The solvent was spun off and column chromatography gave 340mg of the compound. The compound is prepared by mixing petroleum ether: slurried 2mL of ethyl acetate at 75 ℃ and filtered with suction to give 270mg of Compound I-16.
1H NMR(500MHz,DMSO-d6)δ10.19(s,1H),9.40(s,1H),8.67(s,1H),8.29(s,1H),7.77(s,1H),7.61(d,J=7.0Hz,1H),7.41~7.20(m,3H),7.09(s,1H),6.55(s,2H),6.45(dd,J=17.0,10.0Hz,1H),6.26(dd,J=17.0,1.5Hz,1H),5.75(dd,J=10.0,1.5Hz,1H),3.53(s,4H),0.65(s,4H),0.05(s,6H).ESIMS:m/z 527.0[M+H]+.
Example 17:
Figure BDA0001543445930000212
mixing intermediate 4-1(310mg,0.90mmol), intermediate 5-10(250mg,1.5mmol), XphOS (182mg,0.4mmol), (dba)3Pd2(40mg,0.04mmol) and potassium carbonate (490mg,3.5mmol) were added to a 35mL microwave tube and the solvent tert-butanol (20mL) was added. The reaction was completed with microwave at 115 ℃ for 40 minutes. The reaction solution was filtered with celite, and the filter cake was washed with 10mL of ethyl acetate. The filtrate was spin-dried and column chromatographed to give 160mg of Compound I-17.
1H NMR(500MHz,CDCl3)δ8.32(s,1H),8.04(br,1H),7.52(d,J=8.0Hz,2H),7.47~7.34(m,5H),7.31(t,J=8.0Hz,1H),7.24(d,J=7.9Hz,1H),6.88(s,1H),6.44(d,J=16.8Hz,1H),6.21(dd,J=16.8,10.3Hz,1H),5.77(d,J=10.3Hz,1H),0.24(s,9H).HR-ESIMS:m/z 472.1775[M+H]+.
Example 18:
Figure BDA0001543445930000221
intermediate 4-1(300mg, 875umoL, 1.0eq) and t-butanol 20mL were weighed into a microwave reaction tube, and intermediate 5-11(289mg, 1.40mmoL, 1.6eq), potassium carbonate (484mg, 3.50mmoL, 4.0eq), Xphos (188mg, 394umoL, 0.45eq), (dba) were added sequentially with stirring3Pd2(40mg, 44umoL, 0.05eq), reacting for 30 minutes at 120 ℃ by microwave, monitoring by TLC until the raw materials react completely, carrying out suction filtration on the reaction liquid while the reaction liquid is hot, carrying out sand column chromatography on the filtrate, and eluting by petroleum ether ethyl acetate to obtain 148mg of the compound I-18 in total.
1H-NMR(500M,DMSO-d6)δ:10.22(s,1H),9.71(br,1H),8.86(br 1H),8.36(s,1H),7.78(s,1H),7.68(d,J=8.0Hz,1H),7.52(d,J=7.0Hz,2H),7.34(t,J=8.0Hz,1H),7.15(d,J=7.0Hz,2H),7.09(s,1H),6.43(dd,J=17.0,10.0Hz,1H),6.26(dd,J=17.0,2.0Hz,1H),5.74(dd,J=10.0,2.0Hz,1H),0.90-0.86(m,9H),0.70-0.65(m,6H).HR-ESIMS:m/z 514.2272[M+H]+.
Example 19:
Figure BDA0001543445930000222
weighing 4-1(400mg, 1.17mmoL, 1.0eq) of intermediate and 15mL of 2-butanol in a microwave reaction tube, sequentially adding 5-12(160mg, 1.17mmoL, 1.0eq) of intermediate and trifluoroacetic acid (399mg, 3.50mmoL, 3.0eq) in a stirring manner, reacting at 100 ℃ for 1 hour by microwave reaction, monitoring the disappearance of a raw material point by TLC, cooling a reaction solution, stirring overnight at room temperature, gradually precipitating a solid, filtering, and drying a filter cake to obtain 308mg of a compound I-19 in total.
1H-NMR(500M,DMSO-d6)δ:10.46(br,2H),9.60(br,1H),8.53(s,1H),7.88(br,1H),7.66(d,J=8.0Hz,1H),7.38(t,J=8.0Hz,3H),7.14(d,J=7.5Hz,1H),7.08(s,1H),6.93(d,J=7.5Hz,1H),6.51(dd,J=17.0,10.0Hz,1H),6.25(dd,J=17.0,2.0Hz,1H),5.74(dd,J=10.0,2.0Hz,1H),4.19(s,2H),3.22(s,3H).HR-ESIMS:m/z 444.1631[M+H]+.
Example 20:
Figure BDA0001543445930000231
step (1): methyl 3-aminobenzoate (20.0g, 132.3mmol, 1eq) and DMF (100mL) were weighed in an eggplant-type bottle, dissolved to clear with stirring, triethylamine (40.16g, 396.9mmol, 3.0eq) and benzyl bromide (56.57g, 330.8mmol, 2.5eq) were added in sequence, oil bath was refluxed for 3 hours under nitrogen protection, and TLC was monitored until the starting material spot disappeared. The heating and cooling were stopped, and the reaction mixture was added dropwise to 0 ℃ dilute hydrochloric acid (0.5M, 3L) with mechanical stirring, to precipitate a solid. And (3) carrying out ultrasonic filtration on the solid, carrying out suction filtration, washing a filter cake to be neutral by using water, and drying under reduced pressure to obtain 38.65g of the intermediate 5-13a in total.
1H-NMR(500M,DMSO-d6):7.37-7.18(m,14H),4.75(s,4H),3.75(s,3H).ESIMS:m/z 332.4[M+H]+.
Step (2): intermediate 5-13a (38.5g, 116.17mmol, 1eq) and anhydrous THF (300mL) were weighed into a three-necked round-bottomed flask, the mixture was stirred to dissolve, a THF solution of methylmagnesium bromide (1M, 349mL, 349mmol, 3eq) was slowly added dropwise through a double-ended needle under nitrogen protection, the internal temperature was controlled at about 0 ℃ during the dropwise addition, the internal temperature was maintained at 0 ℃ after the dropwise addition for 3 hours, and the completion of the reaction of the starting materials was monitored by TLC. After the reaction, methyl magnesium bromide was quenched by slowly dropping ammonium chloride solution (4M) at low temperature, most THF was evaporated by rotary evaporation, the residue was added with water, extracted twice with ethyl acetate, organic layers were combined and washed once with saturated brine, and dried over anhydrous sodium sulfate. And carrying out suction filtration on the extract liquor, and then carrying out rotary evaporation to obtain 35.67g of compounds 5-13b in total.
1H-NMR(500M,DMSO-d6):7.34-7.22(m,10H),7.01-6.98(t,J=8Hz,1H),6.88(s,1H),6.71-6.69(d,J=8Hz,1Hz),6.50-6.48(m,1H),4.77(br,1H),4.67(s,4H),1.29(s,6H).ESIMS:m/z 332.2[M+H]+.
And (3): compound 5-13b (27.6g, 8.33mmol, 1eq) and 300mL of methanol were weighed in an eggplant-shaped flask, stirred to dissolve and clear, 10% Pd/C (2.0g) was added, after nitrogen was repeatedly replaced, hydrogen was reacted at room temperature overnight under normal pressure, and TLC monitored that the starting material completely disappeared. After Pd/C is filtered out of the reaction solution, the filtrate is prepared into sand and passes through a silica gel column, and DCM-MeOH is used for eluting to obtain 5.8g of the compound 5-13 in total.
1H-NMR(500M,DMSO-d6):6.94-6.90(t,J=8Hz,1H),6.72-6.71(d,J=1.5Hz,1H),6.60-6.58(d,J=8Hz,1H),6.39-6.37(dd,J=8Hz,1.5Hz,1H),4.88(s,2H),4.74(s,1H),1.37(s,6H)
ESIMS:m/z 152.4[M+H]+.
And (4): intermediate 4-1(450mg, 1.31mmol, 1eq) and compound 5-13(199mg, 1.31mmol, 1eq) were weighed in a 35mL microwave reaction tube, 2-butanol (18mL) was added and stirred to dissolve, the reaction was carried out at 80 ℃ for 1 hour by microwave, the reaction was monitored by TLC for completion, the solvent was evaporated by rotary evaporation, the residue was extracted twice with water and ethyl acetate, the organic phases were combined and washed once with saturated brine and dried over anhydrous sodium sulfate. Processing the extract into sand, passing through silica gel column, eluting with DCM-MeOH, mixing eluates, evaporating to dryness to obtain crude product, pulping the crude product with DCM-MeOH (10:1) under ultrasonic beating, and filtering to obtain compound I-20 (350 mg in total).
1H-NMR(500M,DMSO-d6):10.16(s,1H),9.54(s,1H),8.72(s,1H),8.36(s,1H),7.78(s,1H),7.60-7.55(dd,J=19Hz,8Hz,2H),7.42(s,1H),7.35-7.31(t,J=8Hz,1H),7.19(s,1H),7.01-7.00(d,J=7Hz,1H),6.94(s,1H),6.47-6.42(dd,J=19Hz,8Hz,1H),6.28-6.24(d,J=17Hz,1H),5.77-5.75(m,1H),4.86(s,1H),1.34(s,6H).HR-MS(ESI+):458.1819[M+H]+.
Example 21:
n- (3- ((2- ((1-hydroxy-1, 3-dihydrobenzo [ c ] [1,2] oxaborol-6-yl) oxy) -5- (trifluoromethyl) pyrimidin-4-yl) amino) phenyl) acrylamide
Figure BDA0001543445930000241
Step 1: 2-bromo-5-methoxybenzaldehyde (19.95g, 92.77mmol, 1.0eq), pinacol diboron (35.43g, 139.52mmol, 1.5eq), [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane complex (7.60g, 9.31mmol, 0.10eq), potassium acetate (27.38g, 278.99mmol, 3.0eq) and 1, 4-dioxane (300g) were charged in this order to a 1L reaction flask, and heated at 90 ℃ for 3 hours under nitrogen protection. After the reaction, heating was stopped, the reaction solution was poured into ethyl acetate (1.2L) after cooling in an ice-water bath, stirred and filtered, the filter cake was washed with ethyl acetate (200mL), the filtrate was washed with water (500mL) and saturated brine (300mL) in this order, dried over anhydrous sodium sulfate, filtered, concentrated, purified by a silica gel column, and petroleum ether: ethyl acetate (5:1) gave 27.46g in total of intermediate 5-16 a.
Step 2: adding lithium aluminum hydride (9.72g, 256.13mmol, 3.0eq) into a 1L reaction bottle, cooling to-20 ℃ under the protection of nitrogen, slowly adding tetrahydrofuran (120mL) dropwise with stirring, continuing to cool to-20 ℃ after the dropwise addition is completed, adding a tetrahydrofuran (140mL) solution of an intermediate 5-16a (22.37g, 85.36mmol, 1.0eq) dropwise with stirring, and slowly raising the temperature to room temperature for reaction for 1.5 hours after the dropwise addition is completed. After the reaction, the reaction mixture was cooled to-10 ℃ and slowly added dropwise with ice water (600mL) to quench the reaction, the pH was adjusted to 3 with 6N hydrochloric acid, the mixture was stirred for 20 minutes, extracted with ethyl acetate, the organic phase was washed successively with water (200mL) and saturated brine (100mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to give 14 in total of intermediates 5 to 16 b.00g。1H-NMR(500M,DMSO-d6):9.11(s,1H),7.31(m,1H),7.25(d,J=2.0Hz,1H),7.05(dd,J=2.0and 8.0Hz 1H),4.92(s,2H),3.74(s,3H).MS(ESI):m/z 163.1[M-H]+.
And step 3: dissolving the intermediate 5-16b (2.500g, 15.25mmol, 1.0eq) in dichloromethane (100mL), cooling to-30 ℃, slowly adding dropwise a 1mol/L solution of boron tribromide in dichloromethane (60mL, 60mmol, 3.9eq), after dropwise addition, slowly heating to room temperature for reaction for 3 hours. After the reaction was completed, the reaction mixture was slowly poured into ice water (200mL) to quench the reaction, the phases were separated, the aqueous phase was extracted with methylene chloride, the obtained organic phase was washed successively with water (50mL) and saturated brine (50mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to obtain 2.00g in total of intermediate 5 to 16.
1H-NMR(500M,DMSO-d6):9.24(brs,1H),7.18(d,J=8.0Hz,1H),7.10(d,J=2.5Hz,1H),6.88(dd,J=2.0Hz,8.0Hz 1H),4.87(s,2H).
And 4, step 4: dissolving 2, 4-dichloro-5-trifluoromethylpyrimidine (2.89g, 13.34mmol, 1.0eq) in 1, 2-dichloroethane (25mL) and tert-butanol (25mL), cooling to 0 ℃, adding an ether solution of zinc dichloride (40mL, 40.00mmol, 3.0eq), stirring at 0 ℃ for 20 minutes after the addition is completed, slowly adding a mixed solution of 1, 2-dichloroethane (25mL) and tert-butanol (25mL) of intermediate 5-16(2.00g, 13.34mmol, 1.0eq) and triethylamine (4.05g, 40.02mmol, 3.0eq) in sequence, stopping cooling after the addition is completed, and heating to room temperature for reaction for 16 hours. The reaction solution was poured into chloroform (200mL), washed with water (50mL) and saturated brine (50mL) in this order, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated and purified by a silica gel column, dichloromethane: methanol (20:1) was eluted, combined and concentrated to give intermediate 4-7, a total of 140 mg.
MS(ESI):m/z 331.2[M+H]+.
And 5: the intermediates 4 to 7(140mg, 0.42mmol, 1.0eq) and N- (3-aminophenyl) acrylamide (68mg, 0.42mmol, 1.0eq) were dissolved in sec-butanol (5ml) in this order, N-diisopropylethylamine (110mg, 0.85mmol, 2.0eq) was added, the mixture was reacted at room temperature for 48 hours, and after completion of the reaction, the reaction solution was concentrated and purified by a silica gel column, dichloromethane: eluting with methanol (50:1-20:1), mixing, concentrating, and purifying with high pressure preparative chromatograph (model: k-preplab 100g column: high pressure dynamic axis compression column 50 × 235mm, 10um) to obtain compound I-21 (70mg in total).
1H-NMR(500M,DMSO-d6):10.12(s,1H),9.19(s,1H),9.15(s,1H),8.51(s,1H),7.73(s,1H),7.42-7.37(m,3H),7.27(d,J=2.5Hz,1H),7.13-7.07(m,2H),6.47-6.42(m,1H),6.29-6.25(m,1H),5.76(dd,J=2.0Hz,10.0Hz,1H),5.00(s,2H).MS(ESI):m/z 457.3[M+H]+.
Example 22
Figure BDA0001543445930000251
Step (1): compound 5-13b (500mg, 1.51mmol, 1eq) was weighed into an eggplant-shaped flask, DMF (5mL) was added and stirred to dissolve, sodium hydride (188mg, 3.02mmol, 2eq) was added at 0 ℃ and kept at that temperature for 20min with stirring, methyl iodide (428mg, 3.02mmol, 2eq) was added and heated to 90 ℃ overnight with completion of the oil bath, TLC monitored that the starting material was completely reacted and stopped heating. Cooling, adding small amount of water to quench reaction, evaporating solvent, adding large amount of water, EA extracting for three times, mixing organic layers, washing with water, washing with saturated salt solution, and drying with anhydrous sodium sulfate. Drying the extract, making sand, passing through silica gel column, eluting with PE-EA, mixing eluates, and evaporating to dryness to obtain 280mg of compounds 5-13 c.
1H-NMR(500M,DMSO-d6):7.32-7.23(m,10H),7.06-7.03(m,1H),6.68(s,1H),6.59-6.57(d,J=7Hz,2H),4.70(s,4H),2.74(s,3H),1.29(s,6H).ESI-MS:m/z 346.4[M+H]+.
Step (2): compound 5-13C (270mg, 782. mu. mol,1eq) and 5mL of methanol were weighed into an eggplant-shaped flask, stirred to dissolve, 10% Pd/C (43mg) was added thereto, nitrogen was repeatedly replaced, hydrogen was reacted at room temperature overnight under normal pressure, and TLC monitored that the starting material was completely lost. After Pd/C is filtered out of the reaction solution, the filtrate is evaporated to dryness by rotation to obtain 75mg of the compound 5-14 in total.
1H-NMR(500M,DMSO-d6):6.98-6.96(t,J=7.5Hz,1H),6.62(s,1H),6.52-6.50(d,J=7.5Hz,1H),6.44-6.43(d,J=7Hz,1H),4.98(s,2H),2.95(s,3H),1.38(s,6H).ESI-MS:m/z 166.4[M+H]+.
And (3): weighing compound 4-1(125mg, 365umol, 1eq) and compound 5-14(60mg, 363umol, 1eq) in a 35mL microwave reaction tube, adding 2-butanol (10mL), stirring and dissolving, reacting at 80 ℃ for 2 hours by microwave, monitoring the reaction completion by TLC, evaporating the solvent by rotation, adding water to the residue, extracting twice by ethyl acetate, combining the organic phases, washing with saturated salt water once, and drying with anhydrous sodium sulfate. The extract was purified by TLC (PE: EA) to obtain a total of 100mg of Compound I-24.
1H-NMR(500M,DMSO-d6):10.16(s,1H),9.58(s,1H),8.75(s,1H),8.37(s,1H),7.78(s,1H),7.65-7.64(d,J=7Hz,1H),7.56-7.55(d,J=7.5Hz,1H),7.38-7.31(m,2H),7.18(s,1H),6.98(s,1H),6.91(s,1H),6.46-6.41(dd,J=17Hz,10Hz,1H),6.27-6.23(d,J=17.5Hz,1H),5.76-5.74(d,J=10Hz,1H),2.92(s,3H),1.34(s,6H).HR-SIMS:m/z 472.1964[M+H]+.
Example 23:
Figure BDA0001543445930000261
step (1): dissolving the intermediate 4-1(1g,2.918mmol,1eq) and 4-methyl aminobenzoate (intermediate 5-15, 463mg,3.064mmol,1.05eq) in 50ml of 2-butanol solution, heating to 110 ℃ for reaction for 7h, and separating and purifying by column chromatography after the reaction is finished to obtain the compound I-25a accounting for 100 mg.
1H NMR(500MHz,DMSO-d6)δ10.22(s,1H),10.09(s,1H),8.98(s,1H),8.43(s,1H),7.83(s,1H),7.65-7.60(m,5H),7.41-7.38(m,1H),7.14-7.13(m,1H),6.45-6.39(m,1H),6.26-6.22(m,1H),5.75-5.73(m,1H),3.78(s,3H).MS(ESI+):458.3[M+H]+.
Step (2): intermediate I-25a (500mg,1.09mmol,1eq) and 25ml DCM were added to the reaction flask. And (3) cooling to-10 ℃ under the protection of nitrogen, dropwise adding DIBAL-H (1.82ml of 2.73mmol,2.5eq) after 10 minutes, continuously adding DIBAL-H until a small amount of the raw material remains after TLC (thin layer chromatography) shows that the raw material remains, adding methanol for quenching, and carrying out column chromatography separation and purification to obtain 200mg of the compound I-25 in total.
1H NMR(500MHz,DMSO-d6)δ10.18(s,1H),9.90(s,1H),7.78(s,1H),8.35(s,1H),7.79-7.73(m,1H),7.58-7.56(m,1H),7.48(s,2H),7.37-7.31(m,1H),7.17-7.02(m,3H),6.46-6.41(m,1H),6.27-6.23(m,1H),5.76-5.74(m,1H),4.37-4.34(m,2H),3.18(br,1H).MS(ESI+):428.2[M-H]+.429
Example 24:
Figure BDA0001543445930000271
with reference to the reaction conditions of example 4, a total of 50mg of the product I-26 was obtained.
1H-NMR(500M,DMSO-d6)δ:10.09(s,1H),9.08(s,1H),8.82-8.80(m,2H),7.96-7.89(m,3H),7.53-7.45(m,4H),7.31-7.28(m,1H),6.48-6.42(m,1H),6.27-6.23(m,1H),5.76-5.73(m,1H),4.81(s,2H),3.88(s,3H).HR-ESIMS:m/z 418.1711[M+H]+.
Example 25:
Figure BDA0001543445930000272
the intermediate 4-2(500mg,1.70mmol,1.0eq), the intermediate 5-13(385mg,2.50mmol,1.5eq) and 2-butanol (18mL) were added to a 35mL microwave tube, and the reaction was stopped by heating the reactor 50W to 120 ℃ for 70 minutes. The solvent was dried by evaporation, and the residue was subjected to column chromatography to give the compound I-27 in a total amount of 39 mg.
1H NMR(500MHz,DMSO-d6)δ10.09(s,1H),9.36(s,1H),9.00(s,1H),8.09(d,J=3.0Hz,1H),7.96(s,1H),7.71(d,J=8.1Hz,1H),7.59(d,J=7.9Hz,1H),7.55(s,1H),7.41(d,J=7.9Hz,1H),7.27(t,J=8.1Hz,1H),7.08(t,J=7.9Hz,1H),6.98(d,J=7.6Hz,1H),6.46(dd,J=17.0,10.2Hz,1H),6.26(d,J=16.9Hz,1H),5.76(d,J=10.2Hz,1H),4.85(s,1H),1.37(s,6H).MS(ESI):m/z 430.3[M+Na]+.407
Example 26:
Figure BDA0001543445930000281
mixing intermediate 4-6(500mg,1.60mmol,1.0eq), intermediate 5-13(370mg,2.40mmol,1.5eq), XphOS (340mg,0.72mmol,0.45eq), (dba)3Pd2(58mg,0.064mmol,0.04eq), potassium carbonate (880mg,6.4mmol,4.0eq) and tert-butanol (18mL) were added to a 35mL microwave tube, nitrogen was bubbled for 1 minute, and the reaction was stopped by heating the reactor 50W to 115 ℃ for 3 hours. When the temperature of the system is reduced to room temperature, filtering by diatomite to remove insoluble substances, and spin-drying the filtrate. Column chromatography of the residue gave 82mg of Compound I-28.
1H NMR(500MHz,DMSO-d6)δ10.04(s,1H),8.68(s,2H),8.01(s,1H),7.85(s,1H),7.74(d,J=7.9Hz,1H),7.63(d,J=7.9Hz,1H),7.59(s,1H),7.39(d,J=7.7Hz,1H),7.25(t,J=8.0Hz,1H),7.08(t,J=7.6Hz,1H),6.94(d,J=7.4Hz,1H),6.46(dd,J=16.7,10.2Hz,1H),6.26(d,J=17.0Hz,1H),5.75(d,J=10.1Hz,1H),4.84(s,1H),3.86(s,3H),1.39(s,6H).MS(ESI):m/z 420.4[M+H]+.
Experimental example 1: inhibition of in vitro activity of kinases BTK, BTK (R28H)
1.1BTK inhibitory Activity screening
With kinase buffer (50mM HEPES, 10mM MgCl)22mM DTT, 1mM EGTA, 0.01% Tween 20), 350ng/uL of BTK stock solution is diluted, 6uL of 1.67 X0.134 ng/uL of working solution (final concentration of 0.08ng/uL) is added to each well, different DMSO-dissolved compounds are added to the wells by a nanoliter loading instrument, the final concentration of the compounds is 1000nM-0.244nM, the final concentration of positive drugs is 50nM-0.0122nM, 4-fold gradient is carried out, 7 concentrations are obtained, meanwhile, blank control wells (containing no enzyme) and negative control wells (containing enzyme and adding vehicle DMSO) are arranged, and 2 duplicate wells are arranged. After 30min of reaction of the enzyme with the compound or vehicle, 5 × 250uM ATP (final concentration of 50uM) and 5 × 0.5uM substrate (final concentration of 0.1uM, ULight-poly GT) in kinase buffer were mixed as 1: 1 mixing, adding 4 mu L of the mixture into each hole; after the plate was sealed with a membrane plate and reacted at room temperature for 2 hours, 5. mu.L of 4X 40mM EDTA (final concentration: 10mM) was added to each well for 5 minutes at room temperature, and 5. mu.L of 4X 8nM detection reagent (final concentration: 2nM, Ab) was added to each well and incubated at room temperature for 1 hour; the plate was read with a PE instrument (excitation 620nm, emission 665 nm). Calculating the inhibition ratio, and calculating IC50 value.
1.2BTK (R28H) inhibitory Activity screening
With kinase buffer (50mM HEPES, 10mM MgCl)22mM DTT, 1mM EGTA, 0.01% Tween 20) 200ng/uL of BTK (R28H) stock solution was diluted, 6uL of 1.67 X1.67 ng/uL working solution (final concentration of 1ng/uL) was added to each well, DMSO-dissolved different compounds were added to the wells using a nanoliter loading apparatus to achieve a final concentration of 2000nM to 0.488nM compound and a final concentration of 200nM to 0.0488nM positive drug, 4-fold gradient, total of 7 concentrations, blank control empty (no enzyme) and negative control wells (enzyme-containing, vehicle DMSO) were set, and 2 replicate wells were set. After 30min of reaction of the enzyme with the compound or vehicle, 5 × 500uM ATP (final concentration 100 μ M) prepared in kinase buffer and 5 × 0.5 μ M substrate (final concentration 0.1 μ M, ULight-poly GT) were mixed in a ratio of 1: 1 mixing, adding 4 mu L of the mixture into each hole; after the plate was sealed with a membrane plate and reacted at room temperature for 2 hours, 5. mu.L of 4X 40mM EDTA (final concentration: 10mM) was added to each well for 5 minutes at room temperature, and 5. mu.L of 4X 8nM detection reagent (final concentration: 2nM, Ab) was added to each well and incubated at room temperature for 1 hour; the plate was read with a PE instrument (excitation 620nm, emission 665 nm). The inhibition rate was calculated and the IC50 value was calculated.
Table 1 shows the activity data of the compounds for wild type BTK, mutant BTK (R28H). Activity was characterized by IC50, where "A" indicates IC50 ≦ 5 nM; "B" means 5 < IC50 ≦ 10 nM; "C" means 10 < IC50 ≦ 100 nM; "D" means 100 < IC50 ≦ 1000 nM.
Table 1: inhibitory Activity of Compounds on BTK kinase and mutants thereof
Figure BDA0001543445930000291
CC292 was synthesized with reference to WO2011090760, compound I-182 of example 20.
Experimental example 2: inhibition of in vitro kinase JAK1, JAK2, JAK3 activity
2.1JAK1 inhibitory Activity screening
With kinase buffer (50mM HEPES, 10mM MgCl)22mM DTT, 1mM EGTA, 0.01% Tween 20) 50 ng/. mu.L of JAK1 dilution of the stock solution, 6. mu.L per well (final concentration 0.5 ng/. mu.l), different compounds dissolved in DMSO, were added to the wells, 4-fold dilutions were performed, 7 concentration gradients were made, giving a final concentration of compounds of 1000nM-0.244nM, blank control empty (no enzyme) and negative control wells (enzyme, vehicle DMSO) were set, and 2 replicate wells were set. After 30min of reaction of the enzyme with the compound or vehicle, 5 × 250 μ M ATP (final concentration 50 μ M) and 5 × 0.5 μ M substrate (final concentration 0.1 μ M, μ M Light-poly GT) in kinase buffer were mixed as 1: 1 mixing, adding 4 mu L of the mixture into each hole; after the plate was sealed with a membrane plate and reacted at room temperature for 2 hours, 5. mu.L of 4X 40mM EDTA (final concentration: 10mM) was added to each well for 5 minutes at room temperature, and 5. mu.L of 4X 8nM detection reagent (final concentration: 2nM, Ab) was added to each well and incubated at room temperature for 1 hour; htrf method (excitation 620nm, emission 665 nm). The inhibition rate was calculated and the IC50 value was calculated.
2.2JAK2 inhibitory Activity screening
With kinase buffer (50mM HEPES, 10mM MgCl)22mM DTT, 1mM EGTA, 0.01% Tween 20) 50 ng/. mu.L of JAK2 stock solution was diluted, 6. mu.L (final concentration of 0.005 ng/. mu.L) was added per well, DMSO-dissolved different compounds were added to the wells, 4-fold dilution was performed, 7 concentration gradients were made to give a final compound concentration of 1000nM to 0.244nM, blank control wells (no enzyme) and negative control wells (enzyme-containing, vehicle DMSO) were set, and 2 replicate wells were set. After 30min of reaction of the enzyme with the compound or vehicle, 5 × 100 μ M ATP (final concentration 20 μ M) and 5 × 0.5 μ M substrate (final concentration 0.1 μ M, μ M Light-poly GT) in kinase buffer were mixed as 1: 1 mixing, adding 4 mu L of the mixture into each hole; after the plate was sealed with a membrane plate and reacted at room temperature for 2 hours, 5. mu.L of 4X 40mM EDTA (final concentration: 10mM) was added to each well for 5 minutes at room temperature, and 5. mu.L of 4X 8nM detection reagent (final concentration: 2nM, Ab) was added to each well and incubated at room temperature for 1 hour; htrf method (excitation 620nm, emission 665 nm). The inhibition rate was calculated and the IC50 value was calculated.
2.3JAK3 inhibitory Activity screening
With kinase buffer (50mM HEPES, 10mM MgCl)22mM DTT, 1mM EGTA, 0.01% Tween 20) 50 ng/. mu.L of JAK3 stock solution was diluted, 6. mu.L (final concentration of 0.01 ng/. mu.L) was added per well,DMSO dissolved different compounds were added to the wells, diluted 4-fold, and subjected to 7 concentration gradients to give a final concentration of compounds of 1000nM-0.244nM, while blank control wells (no enzyme) and negative control wells (enzyme-containing, vehicle DMSO) were set, and 2 replicate wells were set. After 30min of reaction of the enzyme with the compound or vehicle, 5 × 100 μ M ATP (final concentration 20 μ M) and 5 × 0.5 μ M substrate (final concentration 0.1 μ M, μ M Light-poly GT) in kinase buffer were mixed as 1: 1 mixing, adding 4 mu L of the mixture into each hole; after the plate was sealed with a membrane, after reacting at room temperature for 2 hours, 5. mu.L of 4X 40mM EDTA (final concentration: 10mM) was added to each well for 5 minutes at room temperature, and 5. mu.L of 4X 8nM detection reagent (final concentration: 2nM, Ab) was added to each well and incubated at room temperature for 1 hour; htrf method (excitation 620nm, emission 665 nm). The inhibition rate was calculated and the IC50 value was calculated.
Table 2 shows the activity data of compounds against wild-type JAK1, JAK2, JAK 3. Activity was characterized by IC50, where "A" indicates IC50 ≦ 5 nM; "B" means 5 < IC50 ≦ 10 nM; "C" means 10 < IC50 ≦ 100 nM; "D" means 100 < IC50 ≦ 1000 nM; "E" means IC50 ≧ 1000 nM.
Table 2: inhibitory Activity of Compounds against JAK1, JAK2, and JAK3 kinases
Figure BDA0001543445930000311
Experimental example 3: activity Studies on Raji, Romas, HEL, Jeko-1, OCI-LY10 cells
3.1 proliferation inhibitory Effect on Raji cells
Taking a bottle of Raji cells in exponential growth phase, collecting the cells, and centrifuging at 1500 rpm for 3min by a low-speed desk centrifuge. The supernatant was discarded and 5mL of complete medium was added using a pipette for cell resuspension. The cells were counted using a cytometer, diluted in complete medium and adjusted to a cell density of 8 x 104 cells/mL. The cells were inoculated in a 96-well plate using a line gun at 100. mu.L/well and incubated in a constant temperature CO2 incubator for 24 hours. The compound was loaded using a nano-liter loading apparatus, CCK-8 was added at 10 μ L/well after 72 hours, and the absorbance was measured at 450nm with an Envision microplate reader after 2 hours, and the inhibition was calculated and the IC50 was calculated.
3.2 inhibition of Romas cell proliferation
A bottle of Romas cells in the exponential growth phase is taken, the cells are collected, and the cells are centrifuged for 3min at 1500 rpm by a low-speed desk centrifuge. The supernatant was discarded and 5mL of complete medium was added using a pipette for cell resuspension. The cells were counted using a cytometer, diluted in complete medium and adjusted to a cell density of 5 x 104 cells/mL. The cells were inoculated in a 96-well plate using a line gun at 100. mu.L/well and incubated in a constant temperature CO2 incubator for 24 hours. The compound was loaded using a nano-liter loading apparatus, CCK-8 was added at 10 μ L/well after 72 hours, and the absorbance was measured at 450nm with an Envision microplate reader after 2 hours, and the inhibition was calculated and the IC50 was calculated.
3.3 inhibition of proliferation of HEL cells
Taking a bottle of HEL cells in exponential growth phase, collecting the cells, and centrifuging at 1500 rpm for 3min by using a low-speed table centrifuge. The supernatant was discarded and 5mL of complete medium was added using a pipette for cell resuspension. The cells were counted using a cytometer, diluted in complete medium and adjusted to a cell density of 7.5 × 104 cells/mL. The cells were inoculated in a 96-well plate using a line gun at 100. mu.L/well and incubated in a constant temperature CO2 incubator for 24 hours. The compound was loaded using a nano-liter loading apparatus, CCK-8 was added at 10 μ L/well after 72 hours, and the absorbance was measured at 450nm with an Envision microplate reader after 2 hours, and the inhibition was calculated and the IC50 was calculated.
3.4 inhibition of Jeko-1 cell proliferation
Taking a bottle of Jeko-1 cells in exponential growth phase, collecting the cells, and centrifuging at 1500 rpm for 3min by a low-speed bench centrifuge. The supernatant was discarded and 5mL of complete medium was added using a pipette for cell resuspension. The cells were counted using a cytometer, diluted in complete medium and adjusted to a cell density of 7.5 × 104 cells/mL. The cells were inoculated in a 96-well plate using a line gun at 100. mu.L/well and incubated in a constant temperature CO2 incubator for 24 hours. The compound was loaded using a nano-liter loading apparatus, CCK-8 was added at 10 μ L/well after 72 hours, and the absorbance was measured at 450nm with an Envision microplate reader after 2 hours, and the inhibition was calculated and the IC50 was calculated.
3.5 inhibition of OCI-LY10 cell proliferation
The diffuse large B-cell lymphoma cells OCI-LY10 used in the experiment are cultured by the institute of medicine and endocrine of Zhejiang university and stored in liquid nitrogen. Cells in good growth state are inoculated in a 96-well plate, after the cells grow adherent to the wall at night, the cells are respectively given with different concentrations of test substances, each concentration is provided with three multiple wells, and the concentration of the test substances is set to be 0.0001-10 mu M. After 72 hours of action, the subject was examined for inhibition of in vitro proliferation of human tumor cell lines by the Celltiter method. The inhibition was calculated from the OD value measured by the microplate reader, and IC50 was calculated.
Table 3 shows the activity data of the compounds on Raji, Romas, HEL, Jeko-1, OCI-LY10 cells. The activity is characterized by IC50, wherein "A" denotes IC50 ≦ 1 μ M; "B" means 1 < IC50 ≦ 10 μ M; "C" means IC50 ≧ 10 μ M.
Table 3: activity data of Compounds on Raji, Romas, HEL, Jeko-1, OCI-LY10 cells
Figure BDA0001543445930000321
Figure BDA0001543445930000331
Note: in all the tables, "-" indicates that no detection was performed.

Claims (16)

1. A compound of formula (I) or a pharmaceutically acceptable salt thereof,
Figure FDA0003077614270000011
wherein the content of the first and second substances,
R1selected from hydrogen, fluorine, chlorine, -CN, C1-4Alkyl, fluoro C1-4Alkyl, chloro C1-4Alkyl, or C1-4An alkoxy group;
R2selected from hydrogen, hydroxy, C1-6Alkoxy radical,Halogen, or cyano;
l is selected from NH, or O;
a is optionally halogen, hydroxy, C1-6Alkyl, halogen substituted C1-6Alkyl, or C1-6Alkoxy-substituted
Figure FDA0003077614270000012
Wherein X-Y is
Figure FDA0003077614270000013
m is 1 or 2, R4Selected from hydroxy, or C1-4An alkoxy group;
or A is
Figure FDA0003077614270000014
Wherein R is3Is selected from
Figure FDA0003077614270000015
2. The compound of claim 1, wherein R1Selected from fluorine, chlorine, C1-4Alkyl, fluoro C1-4Alkyl, or chloro C1-4An alkyl group.
3. The compound of claim 2, wherein R1Selected from fluorine, chlorine, methyl, or trifluoromethyl.
4. A compound according to any one of claims 1 to 3, wherein R2Selected from hydrogen, hydroxy, or C1-4An alkoxy group.
5. The compound of claim 4, wherein R2Selected from hydrogen, or methoxy.
6. The compound of claim 1, wherein a is optionally substituted with halogen, hydroxy, or C1-6Alkyl substituted
Figure FDA0003077614270000016
Wherein X-Y is
Figure FDA0003077614270000017
m is 1 or 2, R4Selected from hydroxy, or C1-4An alkoxy group.
7. The compound of claim 6, wherein A is optionally halogen, or C1-4Alkyl substituted
Figure FDA0003077614270000018
8. The compound of claim 7, wherein a is optionally substituted with fluoro or methyl
Figure FDA0003077614270000019
9. The compound of any one of claims 6-8, wherein X-Y is
Figure FDA0003077614270000021
R4Is a hydroxyl group.
10. The compound of claim 1, wherein a is
Figure FDA0003077614270000022
Figure FDA0003077614270000023
11. The compound of claim 1, wherein a is
Figure FDA0003077614270000024
Wherein R is3Is selected from
Figure FDA0003077614270000025
Figure FDA0003077614270000026
12. The compound of claim 1, wherein the compound of formula (I) is selected from a compound of formula (II) or a pharmaceutically acceptable salt thereof
Figure FDA0003077614270000027
Wherein, X-Y, m, L, R1And R2Is as defined in claim 1.
13. The compound of claim 1, wherein the compound of formula (I) is selected from a compound of formula (III) or a pharmaceutically acceptable salt thereof
Figure FDA0003077614270000028
Wherein, L, R1、R2And R3Is as defined in claim 1.
14. The following compounds or pharmaceutically acceptable salts thereof:
Figure FDA0003077614270000031
Figure FDA0003077614270000041
15. a pharmaceutical composition comprising a compound of claim 1 or a pharmaceutically acceptable salt thereof.
16. Use of the compound of claim 1 or a pharmaceutically acceptable salt thereof or the pharmaceutical composition of claim 15 in the manufacture of a medicament for preventing or treating a BTK-mediated disease.
CN201810020706.2A 2017-01-13 2018-01-10 Bruton's tyrosine kinase inhibitors Active CN108329274B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2017100251997 2017-01-13
CN201710025199 2017-01-13

Publications (2)

Publication Number Publication Date
CN108329274A CN108329274A (en) 2018-07-27
CN108329274B true CN108329274B (en) 2021-09-03

Family

ID=62924704

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201810020706.2A Active CN108329274B (en) 2017-01-13 2018-01-10 Bruton's tyrosine kinase inhibitors

Country Status (1)

Country Link
CN (1) CN108329274B (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020232470A1 (en) 2019-05-13 2020-11-19 Boragen, Inc. Chemical compounds
WO2021003501A1 (en) 2019-07-03 2021-01-07 Boragen, Inc. Chemical compounds
CN114085207B (en) * 2020-10-16 2024-06-25 广州百霆医药科技有限公司 Bruton tyrosine protein kinase inhibitor and application thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014040555A1 (en) * 2012-09-12 2014-03-20 山东亨利医药科技有限责任公司 Nitrogen-containing heteroaromatic ring derivative as tyrosine kinase inhibitor
WO2016100914A1 (en) * 2014-12-18 2016-06-23 Gourlay Steven Treatment of pemphigus
CN106146407A (en) * 2009-12-29 2016-11-23 阿维拉制药公司 Heteroaryl compound and its purposes

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106146407A (en) * 2009-12-29 2016-11-23 阿维拉制药公司 Heteroaryl compound and its purposes
WO2014040555A1 (en) * 2012-09-12 2014-03-20 山东亨利医药科技有限责任公司 Nitrogen-containing heteroaromatic ring derivative as tyrosine kinase inhibitor
WO2016100914A1 (en) * 2014-12-18 2016-06-23 Gourlay Steven Treatment of pemphigus

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
Discovery of Novel Bruton’s Tyrosine Kinase (BTK) Inhibitors Bearing a N,9-Diphenyl‑9H‑purin-2-amine Scaffold;Yang Ge et al.;《ACS Med. Chem. Lett.》;20160921;第7卷(第12期);第1050–1055页 *
Structural optimization of diphenylpyrimidine derivatives (DPPYs) as potent Bruton"s tyrosine kinase (BTK) inhibitors with improved activity toward B leukemia cell lines;Dan Zhao et al.;《EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY》;20161123;第126卷;第444-455页 *

Also Published As

Publication number Publication date
CN108329274A (en) 2018-07-27

Similar Documents

Publication Publication Date Title
ES2683127T3 (en) Useful compositions for treating disorders related to KIT
JP2022058976A (en) Inhibitors of ret
JP6457623B2 (en) 2,4-disubstituted 7H-pyrrolo [2,3-d] pyrimidine derivatives, process for their preparation and use in medicine
WO2021088945A1 (en) Compound as shp2 inhibitor and use thereof
CA2561406C (en) Heterocyclic compound and anti-malignant-tumor agent comprising the same as effective component
RU2734849C2 (en) Aniline pyrimidine derivatives and use thereof
KR20200115583A (en) 2H-indazole derivatives as CDK4 and CDK6 inhibitors and their therapeutic use
CN108329274B (en) Bruton&#39;s tyrosine kinase inhibitors
CN105377835A (en) Protein tyrosine kinase modulators and methods of use
JP7077323B2 (en) Quinazoline compound and its preparation method, use and pharmaceutical composition
CN113825754B (en) Disubstituted sulfonamide selective BCL-2 inhibitors including methyl and trifluoromethyl
JP2024505732A (en) Pyridopyrimidinone derivatives and their production methods and uses
WO2018088780A1 (en) Novel pyrrolopyrimidine derivatives and pharmaceutical composition comprising the same
CN109311891A (en) The crystallization of Pyrrolopyrimidine compounds as JAK inhibitor
CN107266421B (en) Substituted benzimidazole derivatives
CN108101926A (en) Pyrimido five-membered heterocycles containing quinolinone, preparation method and applications
CN109942556A (en) Pyrimidinone compound and its application
CN108341835B (en) Boron-containing compounds as tyrosine kinase inhibitors
CN111848678A (en) Phosphorus-containing thienopyrimidine derivatives
CN113880814B (en) Pyrimidine amine compound and application thereof
CN105541792B (en) Polycyclic class PI3K inhibitor
CN110724137B (en) Thiophene derivative and preparation method and application thereof
CN110776511B (en) Quinoline ring-containing BTK inhibitors
CN111566102B (en) Substituted pyrrolopyridines as activin receptor-like kinase inhibitors
CN114539269A (en) Nitrogen-containing macrocyclic compound, preparation method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant