CN110734436A - Pyrimidine or pyrazine ring compounds and application thereof - Google Patents

Pyrimidine or pyrazine ring compounds and application thereof Download PDF

Info

Publication number
CN110734436A
CN110734436A CN201810776810.4A CN201810776810A CN110734436A CN 110734436 A CN110734436 A CN 110734436A CN 201810776810 A CN201810776810 A CN 201810776810A CN 110734436 A CN110734436 A CN 110734436A
Authority
CN
China
Prior art keywords
cancer
substituted
independently
pyrimidine
pharmaceutically acceptable
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN201810776810.4A
Other languages
Chinese (zh)
Inventor
邹斌
马世超
王翔
张忠国
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.)
Shanghai Qing Yu Medical Science And Technology Co Ltd
Original Assignee
Shanghai Qing Yu Medical Science And Technology 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 Shanghai Qing Yu Medical Science And Technology Co Ltd filed Critical Shanghai Qing Yu Medical Science And Technology Co Ltd
Priority to CN201810776810.4A priority Critical patent/CN110734436A/en
Publication of CN110734436A publication Critical patent/CN110734436A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains two hetero rings
    • C07D513/04Ortho-condensed systems
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Abstract

The invention discloses a pyrimidine or pyrazine five-membered heterocyclic compound, a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer or a solvate thereof. The invention also provides a preparation method of the compound, a composition containing the compound and the compoundThe use of a compound for the preparation of a medicament for the treatment of a disease or disorder associated with the mechanism of action of the EED protein and/or PRC2 protein complex.

Description

Pyrimidine or pyrazine ring compounds and application thereof
Technical Field
The invention discloses a pyrimidine or pyrazine ring compound, a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer or a solvate thereof or an isotope labeling compound thereof. The invention also provides a preparation method of the compound and an intermediate thereof, a composition containing the compound and application of the compound as a medicament for treating diseases related to the action mechanism of the EED protein and/or the PRC2 protein complex.
Background
PcG (Polycomb group) protein is important chromatin modifying enzyme, it regulates the transcription of gene by modification of chromatin, thus has important effect on the growth, differentiation and long-term cell memory of stem cells, in mammalian cells, PcG protein is mainly divided into two classes of transcription inhibition complexes, PRC1(Polycomb regenerative Complex 1) and PRC2(Polycomb regenerative Complex 2), respectively, wherein PRC2 inhibits the expression of related gene by methylation modification of 27 th lysine (H3K27) of histone 3 in chromatin, PRC2 protein Complex is mainly composed of EZH2(Enhancer of Ze Homolog 2) (or its homologous protein EZH1), EED emyonecotrope (pancreatic cancer) and pancreatic cancer protein III, which are important for the development of pancreatic cancer cell expressing gene, such as pancreatic cancer receptor kinase, 2, pancreatic cancer receptor kinase, lung cancer receptor kinase-2, lung cancer receptor tyrosine kinase, pancreatic cancer receptor tyrosine kinase, etc. pancreatic cancer receptor tyrosine kinase, etc. 2, pancreatic cancer receptor tyrosine kinase, as a pancreatic cancer receptor tyrosine kinase, etc., pancreatic cancer receptor tyrosine kinase, as a pancreatic cancer receptor tyrosine kinase, a pancreatic cancer receptor-trypsin.
The current EZH2 inhibitors entering the clinic are EPZ-6438 (episome, second stage of clinic), GSK2816126(GSK, stage ), CPI-1205(Constellation, stage ) and so on, although EZH2 inhibitors have been developed into the clinical research stage, these inhibitors all contain pharmacophores of 2-pyridones in common, and, in clinical treatment with the existing EZH2 inhibitors, secondary mutations have begun to appear, EED inhibitors have allosteric inhibitory effects on EZH2 enzyme function, which may achieve the same or similar biological functions as EZH2, EED inhibitors on are very good at overcoming EZH2, EED inhibitors on may be combined with EZH2 inhibitors to achieve better synergistic effects.
Disclosure of Invention
The pyrimidine or pyrazine ring compound provided by the invention is an brand-new EED inhibitor, shows good inhibitory activity to tumor cells, has broad drug development prospect, and is simple in preparation method, mild in reaction condition, high in yield and purity, simple in post-treatment, green and environment-friendly, and beneficial to industrial production.
the present invention provides a pyrimidine or pyrazine ring compound represented by the formula (I), a pharmaceutically acceptable salt, hydrate, prodrug, stereoisomer or solvate thereof,
Figure BDA0001732766820000031
wherein
X1Independently is C or N;
X2independently is C or N;
X3independently is C or N;
X4independently is C, N or S;
z is independently C or O;
wherein
Figure BDA0001732766820000032
Is a single or double bond;
m is independently 0 or 1;
n is independently 0 or 1;
p is independently 0 or 1;
R1independently hydrogen or halogen;
R2independently of one another hydrogen, halogen, cyano, C1-8Alkyl radical, C1-8Haloalkyl, R2aSubstituted or unsubstituted C3-8Cycloalkyl radical, R2bSubstituted or unsubstituted C3-8Heterocyclic radical, R2cSubstituted or unsubstituted alkenyl, R2bSubstituted or unsubstituted C5-8Cycloalkenyl radical, R2bSubstituted or unsubstituted C5-8Heterocycloalkenyl, R2dSubstituted or unsubstituted amino, R2dSubstituted or unsubstituted oxy, R2dSubstituted or unsubstituted amide, 0-3R2eSubstituted C6-10Aryl or 0-3R2eSubstituted radicals containing C1-20Carbon atom and 1-4N, NR2e1O or S (O)0-2Heteroaromatic rings of hetero atoms;
R2aand R2bIndependently amino, amino protected by a protecting group, monofluorine or polyfluoro;
R2cindependently is C1-4Alkyl, ester group;
R2dindependently is C1-20Alkyl, benzyl, substituted or unsubstituted C6-10Aryl, substituted or unsubstituted, containing C1-20Carbon atom and 1-4N, NR2e1O or S (O)0-2Heteroaromatic rings of hetero atoms;
R2eindependently is halogen, -C (═ O) NR2e1R2e2、-S(=O)2R2e3、N R2e1R2e2Containing C1-20Carbon atom and 1-4 NR2e1O or S (O)0-2Hetero-atomic heterocyclic rings; wherein R is2e1Independently is hydrogen or C1-4An alkyl group; r2e2Independently is hydrogen or C1-4An alkyl group; r2e3Is C1-4An alkyl group.
In another preferred embodiment, when Z is absent,is a single or double bond; m and n are 0; p is 1;
in another preferred embodiment of , when Z is C,
Figure BDA0001732766820000042
is a single bond; m is 1; n is 0; p is 1;
in another preferred embodiment of , when Z is O,
Figure BDA0001732766820000043
m is 1, n and p are of 0 and of 1;
in another preferred embodiment of , X1When is C, X2Is N, X3Is C, X4Is N;
in another preferred embodiment of , X1Is N, X2When is N, X3Is C, X4Is C;
in another preferred embodiment of , X1Is N, X2When is C, X3Is N, X4Is C;
in another preferred embodiment of , X1Is N, X2When is C, X3Is C, X4Is S;
in another preferred embodiment of , R1Selected from H, F.
In another preferred embodiment of , R2Is halogen, cyano, C1-8Alkyl radical, C1-8Haloalkyl or has the structure:
Figure BDA0001732766820000051
wherein j is 0, 1,2 or 3, k is 0, 1,2,3 or 4, l is 0, 1 or 2, V is C, N or O, and at most two N or O are simultaneously present in the same ring , and ring A contains 1-3 heteroatoms of substituted or unsubstituted C5-10A heteroaryl group; ring B is selected from substituted or unsubstituted C containing 1-4 heteroatoms5-10Heteroaryl, wherein the heteroatom is selected from N, O or S; q is N, O or S.
In another preferred embodiment, ring A is a six membered heteroaryl group containing 1-3 heteroatoms.
In another preferred embodiment, ring B is a five membered heterocyclic group containing 1-4 heteroatoms.
In another preferred embodiment, the compound is selected from any of the following compounds:
Figure BDA0001732766820000052
wherein m, n, X1、X2、X3、X4、R1、R2And
Figure BDA0001732766820000053
is as defined above.
In another preferred embodiment, the compound is selected from any of the following compounds:
Figure BDA0001732766820000054
Figure BDA0001732766820000061
Figure BDA0001732766820000071
Figure BDA0001732766820000081
Figure BDA0001732766820000101
the invention also provides an isotope label of the pyrimidine or pyrazine ring compound, pharmaceutically acceptable salt, hydrate, prodrug, stereoisomer or solvate thereof shown in the formula (I)The compound is noted. The atom capable of being labeled by an isotope in the compound represented by the general formula (I) includes, but is not limited to, hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine, chlorine, iodine and the like. Each of which can be isotopically substituted2H、3H、11C、13C、14C、15N、18F、31P、32P、35S、36Cl and125i, and the like.
The invention also provides a preparation method of the pyrimidone compound shown in the formula (I) and an intermediate thereof, which comprises the following steps:
the present invention provides a process for producing a pyrimidone compound represented by formula (I), comprising the steps of:
wherein W represents halogen, preferably Br, I; x1、X2、X3、X4、R1、R2Z, m, n and p are as defined above.
In another preferred embodiment of , the process for preparing compound I-A comprises the steps of:
and (3) carrying out coupling on the halogenated intermediate A and boric acid, boric acid ester or borate under the coupling condition to generate the compound I-A.
Figure BDA0001732766820000121
Wherein, W, R1、R2、X1、X2、X3、X4、Z、j、k、m、n、p、R2b、R2c、R2eIs as defined above.
In another preferred embodiment of , the process for preparing compound I-B comprises the steps of:
under the condition that the halogenated intermediate A is coupled with zinc cyanide, cyano compounds I-B are generated.
The reaction equation is as follows:
Figure BDA0001732766820000122
wherein, W, R1、R2、X1、X2、X3、X4Z, m, n and p are as defined above.
In another preferred embodiment of , the process for preparing compounds I-C comprises the steps of:
the halogenated intermediate A is coupled with substituted amine or mercaptan under the condition of coupling to generate the compound I-C.
The reaction equation is as follows:
Figure BDA0001732766820000131
wherein, W, R1、R2、X1、X2、X3、X4Z, m, N and p are as defined above and Q is N or S.
The invention also provides a preparation method of compounds A, which comprises the following steps:
and substituting the chloropyrimidine or chloropyrazine intermediate B by the intermediate amine C under an alkaline condition to obtain a halogenated pyrimidine or pyrazino ring intermediate A.
The reaction equation is as follows:
Figure BDA0001732766820000132
wherein, W, R1、X1、X2、X3、X4Z, m, n and p are as defined above.
The invention also provides a preparation method of compounds B1, which comprises the following steps:
dibromo pyrazine B1-a reacts with hydrazine hydrate to obtain substituted hydrazine B1-B, and B1-B is subjected to ring closing in trimethyl orthoformate to obtain an intermediate B1.
The reaction equation is as follows:
Figure BDA0001732766820000133
the invention also provides a preparation method of the compound B2, which comprises the following steps:
chloro-bromo-pyrimidine B2-a is condensed with N, N-dimethylformamide dimethyl acetal to obtain B2-B, B2-B is reacted with hydroxylamine to obtain B2-c, and B2-c is subjected to ring closure in polyphosphoric acid to obtain an intermediate B2.
The reaction equation is as follows:
Figure BDA0001732766820000141
the invention also provides a preparation method of the compound B3, which comprises the following steps:
bromination of diaminopyridine B3-a gives intermediate B3-B, and reflux of B3-B in thionyl chloride gives intermediate B3.
The reaction equation is as follows:
Figure BDA0001732766820000142
the present invention also provides compounds C1 or C2,
Figure BDA0001732766820000143
wherein R is1Is as defined above.
The invention also provides a preparation method of C1 or C2, which comprises the following steps:
m-bromophenol C1-a is subjected to intramolecular condensation after the substitution of phenolic hydroxyl to obtain acetal C1-b to obtain intermediate C1-C, and it is noted that when R is1When hydrogen is used, a by-product C1-d generated in the condensation process can be converted into cyano through the next step and then separated and removed by a column chromatography method, bromo-benzofuran C1-C is cyanated to obtain an intermediate C1-e, and a compound C1-e is subjected to cyano reduction to obtain amine C2.
Hydrogenation reduction of amine C2 affords amine C1. Alternatively, the compound C1-e can be obtained by hydrogenating and reducing furan and cyano while protecting the resulting amino group with Boc to give C1-f; finally, compound C1-f was deprotected to afford amine C1.
Figure BDA0001732766820000151
Wherein R is1Is as defined above.
The present invention also provides a process for the preparation of compound C3,
Figure BDA0001732766820000152
wherein R is1Is as defined above.
The invention also provides a preparation method of the compound C3, which comprises the following steps:
the m-bromophenol C1-a is substituted by propargyl through substituted phenolic hydroxyl to obtain a compound C3-a; rearrangement of compound C3-a at elevated temperature gives intermediate C3-b, analogous when R is1When hydrogen, a rearrangement by-product C3-C is formed; after obtaining the compound C3-b, the amine C3 can be obtained by a method similar to that for the synthesis of C1.
The reaction equation is as follows:
Figure BDA0001732766820000161
wherein R is1Is as defined above.
The present invention also provides a method of making a C4,
Figure BDA0001732766820000162
wherein n is 1 or 2; r1As described above.
The invention also provides a preparation method of the compound C4, which comprises the following steps:
demethylating the C4-a to obtain o-diphenol C4-b; C4-C is obtained by the naphthenic alkylation of o-diphenol C4-b, and aldehyde group substitution is introduced to obtain an intermediate C4-d; condensing aldehyde C4-d with hydroxylamine to obtain formaldehyde oxime C4-e; reducing formaldoxime and simultaneously carrying out Boc protection to obtain C4-f; finally, deprotection affords the amine C4.
The reaction equation is as follows:
Figure BDA0001732766820000171
wherein n is 1 or 2; r1As described above.
The inert solvent involved in the present invention is selected from: dichloromethane, chloroform, 1, 2-dichloroethane, dioxane, DMF, acetonitrile, DMSO, NMP, THF, or a combination thereof.
The base to which the present invention relates includes organic bases and inorganic bases.
The organic base to which the present invention relates is selected from: TEA, DIPEA, or a combination thereof.
The inorganic base involved in the invention is selected from: sodium hydride, potassium carbonate, sodium carbonate, cesium carbonate, potassium tert-butoxide, sodium tert-butoxide, LiHMDS, LDA, butyllithium or combinations thereof.
Isotopically labeled compounds of the compounds represented by general formula (I) of the present invention can be prepared by a synthetic method similar to that for unlabeled compounds, except that the starting materials and/or reagents which are not labeled are replaced with isotopically labeled starting materials and/or reagents.
The invention also provides pharmaceutical compositions, which comprise compounds shown in the general formula (I), pharmaceutically acceptable salts, hydrates, prodrugs, stereoisomers or solvates thereof, or isotopically labeled compounds of the compounds shown in the general formula (I), pharmaceutically acceptable salts, hydrates, prodrugs, stereoisomers or solvates thereof, and pharmaceutically acceptable auxiliary materials.
The invention also provides application of the pyrimidone compound shown in the general formula (I), pharmaceutically acceptable salts thereof or solvates thereof in preparing a medicament for preventing and/or treating cancers related to the action mechanism of the EED protein and/or the PRC2 protein compound. Preferably, the cancer includes, but is not limited to, diffuse large B-cell lymphoma, follicular lymphoma; or preferably, the cancer includes, but is not limited to, lymphoma, leukemia, multiple myeloma, mesothelioma, gastric cancer, malignant rhabdoid tumor, liver cancer, prostate cancer, breast cancer, brain tumor, cervical cancer, colon cancer, melanoma, endometrial cancer, esophageal cancer, head and neck cancer, lung cancer, nasopharyngeal cancer, ovarian cancer, pancreatic cancer, kidney cancer, rectal cancer, thyroid cancer, parathyroid tumor, uterine tumor, soft tissue sarcoma, and the like, and the brain tumor includes, but is not limited to, neuroblastoma, glioma, glioblastoma, and astrocytoma.
The invention also provides pharmaceutical preparations comprising said pyrimidone compound of formula (I), a pharmaceutically acceptable salt, hydrate, prodrug, stereoisomer or solvate thereof, or an isotopically labeled compound of formula (I), a pharmaceutically acceptable salt, hydrate, prodrug, stereoisomer or solvate thereof, which can be administered in a suitable manner, e.g., as a suspension, syrup, emulsion, solution, etc., in the form of tablets, capsules (e.g., sustained or timed release capsules), pills, powders, granules (e.g., small particles), elixirs, tinctures, suspensions (e.g., nanosuspensions, microsuspensions) and spray-dried dispersions, and can be administered orally, sublingually, by inhalation including subcutaneous injection, intravenous injection, intramuscular injection, intrasternal injection, nasal administration (e.g., nasal membrane inhalation), topical administration (e.g., creams and ointments), rectal administration (e.g., suppositories), and the like.
The invention also provides the medicine preparation which can be formulated into proper dosage so as to be convenient and control the dosage of the medicine. The dosage regimen of the compounds disclosed herein will vary with such factors as the pharmacodynamics and mode of administration, the subject, sex, age, health, weight, condition, other concurrent conditions, frequency of administration, liver and kidney function, and the effect desired, etc. The compounds disclosed herein may be administered in a single dose per day, or may be administered in a total dose divided into multiple doses (e.g., two to four times per day).
The invention also provides a product of the pyrimidone compound shown in the general formula (I), pharmaceutically acceptable salt thereof or solvate thereof and other medicaments which are selected from anticancer medicaments, tumor immunity medicaments, antiallergic medicaments, antiemetic medicaments, analgesic medicaments, cytoprotective medicaments and the like, wherein has better effect when used together.
It is to be understood that within the scope of the present invention, the above-described features of the present invention and those specifically described below (e.g., in the examples) can be combined with one another to form new or preferred embodiments.
Description of the terms
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
As used herein, the term "about" when used in reference to a specifically recited value means that the value may vary by no more than 1% from the recited value. For example, as used herein, the expression "about 100" includes 99 and 101 and all values in between (e.g., 99.1, 99.2, 99.3, 99.4, etc.).
As used herein, the term "comprising" or "includes" can be open, semi-closed, and closed. In other words, the term also includes "consisting essentially of …," or "consisting of ….
Radical definition
Definitions for the terms of the standardization industry can be found in the literature references including Carey and Sundberg "ADVANCED ORGANIC CHEMISTRY 4. THED." vols. A (2000) and B (2001), Plenum Press, New York). Unless otherwise indicated, conventional methods within the skill of the art are employed, such as mass spectrometry, NMR, IR and UV/VIS spectroscopy, and pharmacological methods. Unless a specific definition is set forth, the terms used herein in the pertinent description of analytical chemistry, organic synthetic chemistry, and pharmaceutical chemistry are known in the art. Standard techniques can be used in chemical synthesis, chemical analysis, pharmaceutical preparation, formulation and delivery, and treatment of patients. For example, the reaction and purification can be carried out using the instructions of the kit from the manufacturer, or according to the methods known in the art or the instructions of the present invention. The techniques and methods described above can generally be practiced according to conventional methods well known in the art, as described in various general and more specific documents referred to and discussed in this specification. In the present specification, groups and substituents thereof may be selected by one skilled in the art to provide stable moieties and compounds.
When a substituent is described by a general formula written from left to right, the substituent also includes chemically equivalent substituents obtained when the formula is written from right to left. For example, -CH2O-is equivalent to-OCH2-。
The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. All documents, or portions of documents, cited in this application, including but not limited to patents, patent applications, articles, books, operating manuals, and treatises, are hereby incorporated by reference in their entirety.
Certain chemical groups defined herein are preceded by a shorthand notation to indicate the total number of carbon atoms present in the group. E.g. C1-6Alkyl refers to an alkyl group as defined below having a total of 1 to 6 carbon atoms (integers within the range are indicated, such as 0, 1,2,3,4, 5, 6). The total number of carbon atoms in the shorthand notation excludes carbons that may be present in a substituent of the group.
In addition to the foregoing, the following terms, when used in the specification and claims of this application, have the meanings indicated below, unless otherwise specifically indicated.
In the present application, the term "halogen" refers to fluorine, chlorine, bromine or iodine.
"hydroxy" means an-OH group.
"hydroxyalkyl" refers to an alkyl group as defined below substituted with a hydroxyl group (-OH).
"carbonyl" refers to a-C (═ O) -group.
"cyano" means-CN.
"amino" means-NH2
"substituted amino" refers to an amino group substituted with or two alkyl, alkylcarbonyl, aralkyl, heteroaralkyl groups as defined below, for example, monoalkylamino, dialkylamino, alkylamido, aralkylamino, heteroaralkylamino.
"carboxyl" means-COOH.
In this application, the term "alkyl" as a group or as part of another group (as used in, for example, halo-substituted alkyl groups and the like) refers to a fully saturated straight or branched hydrocarbon chain radical consisting only of carbon and hydrogen atoms, having, for example, 1 to 12 (preferably 1 to 8, more preferably 1 to 6) carbon atoms, and attached to the remainder of the molecule by a single bond, including, for example, but not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 2-methylbutyl, 2-dimethylpropyl, n-hexyl, heptyl, 2-methylhexyl, 3-methylhexyl, octyl, nonyl, and decyl, and the like.
In this application, the term "alkenyl" as the part of a group or other group means a straight or branched hydrocarbon chain group consisting only of carbon and hydrogen atoms, containing at least double bonds, having, for example, 2 to 14 (preferably 2 to 10, more preferably 2 to 6) carbon atoms, and attached to the rest of the molecule by single bonds, such as, but not limited to, vinyl, propenyl, allyl, but-1-enyl, but-2-enyl, pent-1, 4-dienyl, and the like.
In the present application, the term "cycloalkyl" as the part of a group or other group means a stable non-aromatic mono-or polycyclic hydrocarbon group consisting of only carbon and hydrogen atoms, which may include fused, bridged or spiro ring systems, having 3 to 15 carbon atoms, preferably 3 to 10 carbon atoms, more preferably 3 to 8 carbon atoms, and which is saturated or unsaturated and may be attached to the rest of the molecule via any suitable carbon atom by a single bond unless otherwise specifically indicated in the specification, examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cyclooctyl, 1H-indenyl, 2, 3-indanyl, 1,2,3, 4-tetrahydro-naphthyl, 5,6,7, 8-tetrahydro-naphthyl, 8, 9-dihydro-7H-benzocyclohepten-6-yl, 6,7,8, 9-tetrahydro-7H-benzocyclohepten-6-yl, 5H-octahydro-2, 2-5-cyclooctenyl, 2-bicyclo [ 1H-indenyl ] octa-2.7H-7-5-bicyclo [ 1.2.7.7.2.7.7.7-H-cyclooctenyl, 2.2.7-2.7H-octahydrobicyclo [ 1.7.7.7.7-6-octahydro-6-1.7-1H-1.7-octahydro-6-1.7-1H-octahydro-6-1.7-1-6-1H-octahydro-1-1.7-2.7-octahydro-cyclooctenyl, 2.7-2-octahydro-2-.
In the present application, the term "heterocyclyl" as the moiety or otherwise means a stable 3-to 20-membered non-aromatic cyclic group consisting of 2 to 14 carbon atoms and 1 to 6 heteroatoms selected from nitrogen, phosphorus, oxygen and sulfur unless otherwise specifically indicated in this specification, heterocyclyl groups may be monocyclic, bicyclic, tricyclic or higher ring systems which may include fused, bridged or spiro ring systems, the nitrogen, carbon or sulfur atoms in the heterocyclyl group may optionally be oxidized, the nitrogen atom may optionally be quaternized, and the heterocyclyl group may be partially or fully saturated, heterocyclyl groups may be linked to the remainder of the molecule via a carbon or heteroatom and by a single bond, in heterocyclyl groups containing fused rings, or more rings may be aryl or heteroaryl groups as defined below, provided that the point of attachment to the remainder of the molecule is a non-aromatic ring, for the purpose of the present invention, the heterocyclyl group is preferably a stable 4-to 11-membered, 4-membered, 3-membered, 7, 5-membered, 5, 3, 7, 5,6, 5, 3, 7, 5, 3, 7, 5, 3,5, or more membered, 3, 7, 5, or more membered, 5, or more, 5, or more, 7, 5, 7, or more, 7, three.
For purposes of this invention, aryl groups may be monocyclic, bicyclic, tricyclic or higher ring systems, and may also be fused to cycloalkyl or heterocyclyl groups as defined above, provided that the aryl group is attached to the remainder of the molecule by a single bond via an atom on the aromatic ring, examples of aryl groups include, but are not limited to, phenyl, naphthyl, anthracenyl, phenanthrenyl, fluorenyl, 2, 3-dihydro-1H-isoindolyl, 2-benzoxazolinone, 2H-1, 4-benzoxazin-3 (4H) -one-7-yl, and the like.
In the present application, the term "arylalkyl" refers to an alkyl group as defined above substituted with an aryl group as defined above.
In this application, the term "heteroaryl" as the moiety or otherwise is intended to mean a 5-to 16-membered conjugated ring system having from 1 to 15 carbon atoms (preferably having from 1 to 10 carbon atoms) and from 1 to 6 heteroatoms selected from nitrogen, oxygen and sulfur in the ring unless otherwise specifically indicated in this specification, the heteroaryl may be a monocyclic, bicyclic, tricyclic or more polycyclic ring system and may also be fused to a cycloalkyl or heterocyclic group as defined above, provided that the heteroaryl is attached to the remainder of the molecule via a single bond via an atom on the aromatic ring, the nitrogen, carbon or sulfur atom in the heteroaryl may optionally be oxidized, the nitrogen atom may optionally be quaternized for the purposes of the present invention, the heteroaryl is preferably a stable 5-to 12-membered aromatic group containing from 1 to 5 heteroatoms selected from nitrogen, oxygen and sulfur, more preferably a stable 5-to 10-membered aromatic group containing from 1 to 4 heteroatoms selected from nitrogen, oxygen and sulfur, or a stable 5-to 10-membered aromatic group containing from 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur, or a 1 to 3-membered heteroaromatic group selected from nitrogen, oxygen and sulfur, 2,3,4, 2,4, 1 to 3,2, 4,3, 1,4, 3,1, 3,4, 3,4, 3, or more of the groups selected from the group consisting of a, 1,3, 4,3, 1,4, 3,1, 4,3, 1,3, 1,4, 3,1, 3,4, three.
In the present application, the term "heteroarylalkyl" refers to an alkyl group as defined above substituted with a heteroaryl group as defined above.
In this application, "optionally" or "optionally" means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where the event or circumstance occurs and instances where it does not. For example, "optionally substituted aryl" means that the aryl group is substituted or unsubstituted, and the description includes both substituted and unsubstituted aryl groups. The "optionally" substituents described in the claims and the description section of the present invention are selected from alkyl, alkenyl, alkynyl, halogen, haloalkyl, haloalkenyl, haloalkynyl, cyano, nitro, optionally substituted aryl, optionally substituted heteroaryl, optionally substituted cycloalkyl, optionally substituted heterocycloalkyl.
The terms "moiety," "structural moiety," "chemical moiety," "group," "chemical group" as used herein refer to a specific fragment or functional group in a molecule. Chemical moieties are generally considered to be chemical entities that are embedded in or attached to a molecule.
When the compounds of the present invention contain olefinic double bonds, the compounds of the present invention are intended to include both E-and Z-geometric isomers unless otherwise specified.
"tautomer" refers to an isomer formed by the transfer of a proton from atoms of a molecule to another atoms of the same molecule all tautomeric forms of the compounds of the invention are intended to be encompassed within the scope of the invention.
The compounds of the present invention, or pharmaceutically acceptable salts thereof, may contain or more chiral carbon atoms and may therefore give rise to enantiomers, diastereomers, and other stereoisomeric forms each of which may be defined as (R) -or (S) -based on stereochemistry.
Conventional techniques for preparing/isolating individual isomers include Chiral synthesis from suitable optically pure precursors, or resolution of racemates (or racemates of salts or derivatives) using, for example, Chiral high performance liquid chromatography, see, for example, GeraldG ü bitz and Martin G.Schmid (Eds.), Chiral Separations, Methods and Protocols, Methods in Molecular Biology, Vol.243, 2004; A.M.Stalcup, Chiral Separations, Annu.Rev.Anal.Chem.3: 341. 63, 2010; Fumis et al (Eds.), GEL' S CYENCLOP EDIA PRACTICA CHEMICAL TRY 5. CHEMISTH., TechtH., Longman Scientific and chemical Ltd., EsX, 1991, 809. 1990. 23,128.
In the present application, the term "pharmaceutically acceptable salts" includes pharmaceutically acceptable acid addition salts and pharmaceutically acceptable base addition salts.
"pharmaceutically acceptable acid addition salts" refers to salts formed with inorganic or organic acids that retain the biological effectiveness of the free base without other side effects, inorganic acid salts include, but are not limited to, hydrochloride, hydrobromide, sulfate, nitrate, phosphate and the like, and organic acid salts include, but are not limited to, formate, acetate, 2-dichloroacetate, trifluoroacetate, propionate, hexanoate, octanoate, decanoate, deca -enoate, glycolate, gluconate, lactate, sebacate, adipate, glutarate, malonate, oxalate, maleate, succinate, fumarate, tartrate, citrate, palmitate, stearate, oleate, cinnamate, laurate, malate, glutamate, pyroglutamate, aspartate, benzoate, methanesulfonate, benzenesulfonate, p-toluenesulfonate, alginate, ascorbate, salicylate, 4-aminosalicylate, napadisylate and the like.
"pharmaceutically acceptable base addition salts" refers to salts with inorganic or organic bases which maintain the biological effectiveness of the free acid without other side effects. Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like. Preferred inorganic salts are ammonium, sodium, potassium, calcium and magnesium salts. Salts derived from organic bases include, but are not limited to, the following: primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, diethanolamine, triethanolamine, dimethylethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purine, piperazine, piperidine, N-ethylpiperidine, polyamine resins, and the like. Preferred organic bases include isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine. These salts can be prepared by methods known in the art.
In the present application, a "pharmaceutical composition" refers to a formulation of a compound of the present invention with a vehicle generally accepted in the art for delivery of biologically active compounds to a mammal (e.g., a human). The medium includes a pharmaceutically acceptable carrier. The purpose of the pharmaceutical composition is to facilitate administration to an organism, facilitate absorption of active ingredients and exert biological activity.
The term "pharmaceutically acceptable" as used herein refers to a substance (e.g., carrier or diluent) that does not affect the biological activity or properties of the compounds of the present invention and is relatively non-toxic, i.e., the substance can be administered to an individual without causing an adverse biological response or interacting in an adverse manner with any of the components contained in the composition.
As used herein, a "pharmaceutically acceptable excipient" includes, but is not limited to, any adjuvant, carrier, excipient, glidant, sweetener, diluent, preservative, dye/colorant, flavoring agent, surfactant, wetting agent, dispersing agent, suspending agent, stabilizing agent, isotonic agent, solvent, or emulsifying agent that is permitted by the relevant government regulatory authority to be acceptable for use in humans or livestock.
The "tumor" of the present invention includes, but is not limited to, brain tumors including neuroblastoma, glioma, glioblastoma and astrocytoma, sarcoma, melanoma, articular chondroma, cholangioma, leukemia, gastrointestinal stromal tumor, diffuse large B-cell lymphoma, lymphoid cancer such as follicular lymphoma, histiocytic lymphoma, non-small cell lung cancer, pancreatic cancer, squamous cell lung cancer, lung adenocarcinoma, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell cancer, cervical cancer, ovarian cancer, intestinal cancer, nasopharyngeal cancer, brain cancer, bone cancer, esophageal cancer, melanoma, renal cancer, oral cancer, multiple myeloma, mesothelioma, malignant rhabdoid tumor, endometrial cancer, head and neck cancer, thyroid cancer, parathyroid tumor, uterine tumor, and soft tissue sarcoma.
The terms "preventing," "prevention," and "prevention" as used herein include reducing the likelihood of occurrence or worsening of a disease or disorder in a patient.
As used herein, the term "treatment" and other similar synonyms include the following meanings:
(i) preventing the occurrence of a disease or condition in a mammal, particularly when such mammal is susceptible to the disease or condition, but has not been diagnosed as having the disease or condition;
(ii) inhibiting the disease or disorder, i.e., arresting its development;
(iii) alleviating the disease or condition, i.e., causing regression of the state of the disease or condition; or
(iv) Alleviating the symptoms caused by the disease or disorder.
The term "effective amount," "therapeutically effective amount," or "pharmaceutically effective amount" as used herein refers to an amount of at least agents or compounds that is sufficient to alleviate to some extent the or more symptoms of the disease or disorder being treated upon administration.
The terms "administering," "administration," "administering," and the like as used herein refer to a method capable of delivering a compound or composition to a desired site for biological action. These methods include, but are not limited to, oral routes, via the duodenal route, parenteral injection (including intravenous, subcutaneous, intraperitoneal, intramuscular, intraarterial injection or infusion), topical administration, and rectal administration. Administration techniques useful for the compounds and methods described herein are well known to those skilled in the art, for example, in Goodman and Gilman, the pharmacological Basis of Therapeutics, current ed.; pergamon; and Remington's, Pharmaceutical Sciences (current edition), Mack Publishing Co., Easton, Pa. In preferred embodiments, the compounds and compositions discussed herein are administered orally.
The term "fixed combination" refers to the simultaneous administration to a patient of at least compounds described herein and at least synergistic agents in the form of a single entity or a single dosage form.
It will also be appreciated by those skilled in the art that in the processes described below, the functional groups of the intermediate compounds may need to be protected by suitable protecting groups. Such functional groups include hydroxyl, amino, mercapto and carboxylic acid. Suitable hydroxy protecting groups include trialkylsilyl or diarylalkylsilyl groups (e.g.tert-butyldimethylsilyl, tert-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like. Suitable protecting groups for amino, amidino and guanidino include t-butyloxycarbonyl, benzyloxycarbonyl and the like. Suitable thiol protecting groups include-C (O) -R "(where R" is alkyl, aryl or aralkyl), p-methoxybenzyl, trityl and the like. Suitable carboxyl protecting groups include alkyl, aryl or aralkyl esters.
Protecting groups may be introduced and removed according to standard techniques known to those skilled in the art and as described herein. The use of protecting Groups is described in detail in Greene, T.W. and P.G.M.Wuts, Protective Groups in organic Synthesis, (1999),4th Ed., Wiley. The protecting group may also be a polymeric resin.
Detailed Description
The invention is further illustrated by the following examples , but is not intended to be limited thereby within the scope of the examples.
The starting materials used in the following examples are commercially available from chemical vendors such as Aldrich, TCI, Alfa Aesar, Bidey, Annelgie, etc., or can be synthesized by known methods.
In the following examples, the ice bath is-5 ℃ to 0 ℃, the room temperature is 10 ℃ to 30 ℃, the reflux temperature is the solvent reflux temperature under normal pressure, the reaction overnight is the time of 8 to 15 hours, and the specific operation temperature is not limited in the following examples, and all the operations are performed at room temperature.
In the following examples, the separation and purification of intermediates and final products are by normal phase or reverse phase chromatographic column separation or other suitable methods. The normal phase flash chromatographic column uses ethyl acetate and n-hexane or methanol and dichloromethane and the like as mobile phases. Reverse phase preparative High Pressure Liquid Chromatography (HPLC) was carried out using a C18 column and UV 214nm and 254nm detection, with mobile phases A (water and 0.1% by volume formic acid), B (acetonitrile) or mobile phases A (water and 0.1% by mass ammonium bicarbonate), B (acetonitrile).
In each example:
LCMS apparatus: pump Agilent 1260 UV detector: agilent 1260 DAD MassSpectrometer API 3000
A chromatographic column: waters sunfire C18, 4.6X 50mm,5um
Mobile phase: A-H2O (0.1% HCOOH); b-acetonitrile
NMR instrument: bruker Ascend 400M (1H NMR:400MHz;13C NMR:100MHz)。
Example 1: 5-bromo-8-chloro- [1,2,4] triazolo [4,3-a ] pyrazine (B1)
Step Synthesis of 5-bromo-2-chloro-3-hydrazinopyrazine (B1-B):
Figure BDA0001732766820000291
b1-a (2g,7.34mmol) and ethanol (20mL) were added to a 50mL single-neck flask, hydrazine hydrate (1.72g,53.65mmol) was slowly added dropwise in an ice bath, and the suspension was stirred at 40 ℃ for 3 hours. After the reaction was completed, the reaction mixture was cooled to room temperature to precipitate a pale yellow solid, which was collected by filtration, and the filter cake was washed with ethanol (5mL) and dried to obtain product B1-B as a pale yellow solid (1g, yield 60%).
1H NMR(400MHz,CDCl3)δ7.76(s,1H),6.55(d,J=0.5Hz,1H),3.97(s,2H)ppm。
Step two, synthesizing 5-bromo-8-chloro- [1,2,4] triazolo [4,3-a ] pyrazine (B1):
Figure BDA0001732766820000292
a50 mL single-neck flask was charged with B1-B (1g, 4.48mmol) and trimethyl orthoformate (20mL), and the temperature was raised to 120 ℃ to react for 10 hours. After the reaction was completed, the temperature was reduced to room temperature, trimethyl orthoformate was removed by rotary evaporation, and the concentrate was purified by a silica gel column (PE: EA ═ 20:1) to give 5-bromo-8-chloro- [1,2,4] triazolo [4,3-a ] pyrazine B1(700mg, yield 67%) as a yellow solid product.
1H NMR(400MHz,CDCl3)δ9.09(s,1H),7.85(s,1H)ppm;LCMS:m/z 232.9[M+H]+
Example 2: 5-chloro-8-bromo- [1,2,4] triazolo [1,5-c ] pyrimidine (B2)
Step Synthesis of N' - (5-bromo-2-chloropyrimidin-4-yl) -N, N-dimethylformamidine (B2-B)
In a 500mL single-necked flask, B2-a (40g, 192mmol), N, N-dimethylformamide dimethyl acetal (30,250mmol) and toluene (200mL) were sequentially added, and the mixture was heated to 120 ℃ to react for 3 hours. After the reaction was complete, the toluene was removed under reduced pressure, the solid was collected by filtration, and the filter cake was washed with 200mL of dichloromethane and dried to give the product N' - (5-bromo-2-chloropyrimidin-4-yl) -N, N-dimethylformamidine B2-B as a white solid (42g, 82% yield).
LCMS:m/z 269.2[M+H]+
Step two Synthesis of N' - (5-bromo-2-chloropyrimidin-4-yl) -N-hydroxyformamidine (B2-c)
Figure BDA0001732766820000302
In a dry 500mL single-neck flask were added B2-B (42g,160mmol), methanol (250mL), hydroxylamine hydrochloride (14g,46mmol) in that order. The reaction was stirred at room temperature for 3 hours. After the reaction was complete, the methanol was removed under reduced pressure, collected by filtration, and the filter cake was washed with 100mL of dichloromethane and dried to give the product N' - (5-bromo-2-chloropyrimidin-4-yl) -N-hydroxyformamidine (B2-c) (40g, 84% yield) as a white solid.
1H NMR(400MHz,DMSO-d6)δ11.28(s,1H),8.67(s,1H),8.50(dd,J=9.2,6.1Hz,1H),7.88(d,J=8.2Hz,1H)ppm;LCMS:m/z 250.9[M+H]+
Step three, synthesizing 8-bromo- [1,2,4] triazolo [1,5-c ] pyrimidine-5-hydroxy (B2-d)
Figure BDA0001732766820000303
To a dry 250mL single neck flask was added 20mL of polyphosphoric acid, B2-c (7.5g,29.6mmol) in that order. The reaction temperature was raised to 120 ℃ and the reaction was stirred overnight. After completion of the reaction, 150m of ice water was added to the reaction system, the pH was adjusted to about 8 with a 2N aqueous solution of sodium hydroxide, the reaction solution was extracted three times with N-butanol (150mL), the organic phases were combined, and the organic phase was concentrated under reduced pressure to give 8-bromo- [1,2,4] triazolo [1,5-c ] pyrimidine-5-hydroxy (B2-d) (3.5g, yield: 50%).
1H NMR(400MHz,DMSO-d6)δ12.42(s,1H),8.45(s,1H),7.96(s,1H)ppm;LCMS:m/z215.0[M+H]+
Step four, synthesis of 8-bromo-5-chloro- [1,2,4] triazolo [1,5-c ] pyrimidine (B2)
Figure BDA0001732766820000311
10mL of phosphorus oxychloride and B2-d (3.5g,16.28mmol) were added sequentially in a dry 50mL single neck bottle. The reaction temperature was raised to 110 ℃ and stirred for 12 hours. After the reaction is finished, removing phosphorus oxychloride under reduced pressure. The obtained solid was purified by column chromatography (ethyl acetate: petroleum ether ═ 1:10) to give 8-bromo-5-chloro- [1,2,4] triazolo [1,5-c ] pyrimidine (B2) (0.8g, yield: 21%) as a yellow solid.
1H NMR(400MHz,CDCl3)δ8.53(s,1H),8.32(s,1H)ppm;LCMS:m/z 232.9[M+H]+
Using the procedure described above, the intermediate 5-bromo-8-chloro- [1,2,4] triazolo [1,5-a ] pyrazine B3 was obtained using similar starting materials
Figure BDA0001732766820000312
LCMS:m/z 232.9[M+H]+
Example 3: 7-bromo-4-chloro- [1,2,5] thiadiazolo [3,4-c ] pyridine (B3)
Step Synthesis of 2, 5-dibromopyridine-3, 4-diamine (B3-B)
Figure BDA0001732766820000321
To a dry 250mL three-necked flask were added B3-a (4.0g,36.7mmol) and 48% hydrobromic acid (80mL) in that order. Liquid bromine (7.0g,44mmol) was added dropwise with stirring at room temperature. The reaction solution was stirred at 110 ℃ for 5 hours. After the reaction was completed, it was cooled to room temperature, and filtered to obtain a brown solid, which was washed with a small amount of cold water. The solid was dissolved in ethyl acetate (200mL) and washed with saturated sodium bicarbonate (100mL) and saturated brine (100 mL). The ethyl acetate layer was dried over anhydrous sodium sulfate, filtered, and concentrated to give B3-B (6.6g, yield 67.5%) as a brown solid.
Step two: synthesis of 7-bromo-4-chloro- [1,2,5] thiadiazolo [3,4-c ] pyridine (B3)
Figure BDA0001732766820000322
10mL of thionyl chloride and B3-B (750mg,2.8mmol) were added sequentially in a dry 50mL one-neck flask, the reaction was stirred at 100 ℃ for 5 hours and then cooled to room temperature, the excess thionyl chloride was removed by spin-drying, the residue was dissolved in ethyl acetate (200mL), washed with saturated brine (200mL), dried over anhydrous sodium sulfate, filtered and concentrated to give crude B3(4.64g, 100%) which was used directly in the next step.
1H NMR(400MHz,CDCl3)δ8.48(s,1H)
Example 4: synthesis of intermediates (2, 3-dihydrobenzofuran-4-yl) methylamine (C1-1) and benzofuran-4-ylmethylamine (C2-1)
Step intermediate bromo-3- (2, 2-diethoxyethoxy) benzene (C1-1b)
In a 500mL single-neck flask were added m-bromophenol C1-1a (13g,75.14mmol) and N, N-dimethylformamide (150mL) in that order. At 0 deg.C, sodium hydride (1.98g, 82.66mmol) was added and stirred for half an hour. Then, 2-bromo-1, 1-diethoxyethane (16.29g, 82.66mmol) was added to the reaction solution, and the mixture was heated to 120 ℃ and stirred overnight. After cooling to room temperature, the reaction was quenched with 100mL of water and extracted with ethyl acetate (500mL X2). The combined organic phases were washed twice with brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give 21g of product C1-1 b.
1H NMR(400MHz,CDCl3)δ7.16-7.06(m,3H),6.88-6.83(m,1H),4.81(t,J=5.2Hz,1H),3.98(d,J=5.2Hz,2H),3.80-3.71(m,2H),3.67-3.59(m,2H),1.26-1.23(m,6H)ppm;LCMS:m/z 289.0[M+H]+
Step two: intermediate 4-bromobenzofuran (C1-1C)
In a 500mL single-neck flask were added bromine-3- (2, 2-diethoxyethoxy) benzene C1-1b (21g, 72.6mmol), 150mL toluene, and polyphosphoric acid (10.68g, 108.93mmol) in that order. The reaction solution was heated at 95 ℃ and stirred for 4 hours. After cooling to room temperature, the reaction solution was quenched with 1L of ice water and extracted with ethyl acetate (500mL X3). The combined organic phases were washed twice with brine, dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a total of 5g of crude C1-1C and C1-1d mixture.
Step three: intermediate benzofuran-4-carbonitrile (C1-1e)
Figure BDA0001732766820000341
In a 100mL three-necked flask was added a mixture of C1-1C and C1-1d (5g, 25.1mmol), zinc cyanide (4.2g, 37.7mmol), tetrakistriphenylphosphine palladium (2.9g,2.5mmol) and anhydrous N, N-dimethylformamide (50mL) in that order, the reaction mixture was heated under nitrogen at 100 ℃ with stirring for 18 hours, after cooling to room temperature, 200mL water was added, extracted with ethyl acetate (200mL X3), the combined organic phases were washed twice with brine, dried over anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and the residue was purified on silica gel (petroleum ether: ethyl acetate 100: 1) to give benzofuran-4-carbonitrile C1-1e (1.1g, 30% yield) as a pale yellow solid, the conversion product of C1-1d was isolated at this step .
1H NMR(400MHz,DMSO-d6)δ8.31(d,J=2.2Hz,1H),8.03(d,J=8.4Hz,1H),7.84-7.71(m,1H),7.58-7.46(m,1H),7.20(dd,J=2.2,1.0Hz,1H)ppm。
Step four: intermediate benzofuran-4-ylmethylamine (C2-1)
Figure BDA0001732766820000342
In a single-neck flask, benzofuran-4-carbonitrile C1-1e (300mg, 2.09mmol), methanol (50mL) and nickel chloride (543mg,4.19mmol) were added, and sodium borohydride (159mg, 4.19mmol) was slowly added to react at room temperature for 2 hours. The mixture was filtered through celite, washed with methanol, and the filtrate was concentrated under reduced pressure to give the crude product. The crude product was purified on silica gel (dichloromethane: methanol ═ 10:1) to give benzofuran-4-ylmethylamine C2-1(50mg, 16.2% yield) as a yellow solid.
1H NMR(400MHz,DMSO-d6)δ9.20-7.80(m,3H),7.60(d,J=8.2Hz,1H),7.44(d,J=7.3Hz,1H),7.35(t,J=7.8Hz,1H),7.28(dt,J=9.6,4.8Hz,1H),4.23(s,2H)ppm;LCMS:m/z148.1[M+H]+
Step five: intermediate tert-butyl ((2, 3-dihydrobenzofuran-4-yl) methyl) carbamate (C1-1f)
Figure BDA0001732766820000351
In a 100mL single neck flask was added in the order benzofuran-4-carbonitrile C1-1e (1.1g, 7.68mmol), BOC anhydride (2.5g, 11.52mmol), methanol (50mL) and 10% palladium on carbon (2g, 50% water). The reaction mixture was sparged with hydrogen for 5 minutes, purged three times with a hydrogen balloon and stirred at 60 ℃ for 18 hours under a hydrogen balloon. The mixture was filtered through celite, washed with methanol (50mL X2), and the filtrate was concentrated under reduced pressure to give compound C1-1f (1.5g, 79% yield).
LCMS:m/z 195.1[M+H-tBu]+
Step six: intermediate (2, 3-dihydrobenzofuran-4-yl) methylamine (C1-1)
Figure BDA0001732766820000352
In a 50mL single-necked flask, ((2, 3-dihydrobenzofuran-4-yl) methyl) carbamic acid tert-butyl ester C1-1f (1.3g, 5.21mmol) and 15mL HCl/dioxane (4M) were added in this order, and the mixture was stirred at room temperature for 4 hours. After the reaction was complete, the mixture was concentrated under reduced pressure. The residue was diluted with a mixed solvent (methanol: acetonitrile ═ 1:10, 50mL), and then potassium carbonate (2g, 14.6mmol) was added. The mixture was heated at 60 ℃ and stirred for 3 hours, cooled to room temperature, filtered, and the filtrate was concentrated under reduced pressure to give compound C1-1(700mg, 90% yield) as a white solid.
1H NMR(400MHz,CD3OD)δ7.15(t,J=7.8Hz,1H),6.89(d,J=7.7Hz,1H),6.72(d,J=8.0Hz,1H),4.59(t,J=8.7Hz,2H),3.93(s,2H),3.27(t,J=8.7Hz,2H)ppm;LCMS:m/z150.1[M+H]+.
Using the procedure described above, the following intermediates C2-2 and C1-2 were obtained using similar starting materials.
Figure BDA0001732766820000361
Example 5: intermediate (6-fluorobenzchroman-5-yl) methylamine (C3-1)
Step intermediate 2-bromo-1-fluoro-4- (prop-2-yn-1-yloxy) benzene (C3-1b)
Figure BDA0001732766820000362
In a dry 2L single neck flask, 3-bromo-4-fluorophenol C3-1a (87g, 455.5mmol) was charged at room temperature, dry N, N-dimethylformamide (800mL) was purged with nitrogen three times, 60% sodium hydride (20g,501mmol) was added in portions over 30 minutes under an ice water bath, the reaction solution was stirred at room temperature for 15 minutes and then cooled with an ice water bath, chloropropyne (50.9g,683.3mmol) was then added, the reaction solution was stirred at room temperature for 18 hours, after completion of the reaction, water (1L) was added, extraction was performed three times with ethyl acetate (1L), the organic phase was washed with a saturated aqueous sodium chloride solution (500mL) for times, the organic phase was dried with sodium sulfate, filtered, and distilled under reduced pressure to give a crude product, and the product was purified by a silica gel column (petroleum ether: ethyl acetate: 20:1) to give 2-bromo-1-fluoro-4- (prop-2-yn-1-yloxy) benzene C3-1b (80g, 76.7% yield) as a yellow oily liquid.
1H NMR(400MHz,CDCl3)δ7.17(dd,J=5.5,3.0Hz,1H),7.05(dd,J=9.0,8.1Hz,1H),6.89(ddd,J=9.1,3.7,3.2Hz,1H),4.66(d,J=2.4Hz,2H),2.54(t,J=2.4Hz,1H)ppm。
Step two: intermediate 5-bromo-6-fluoro-2H-chromene (C3-1C)
Figure BDA0001732766820000371
2-bromo-1-fluoro-4- (prop-2-yn-1-yloxy) benzene C3-1b (2g,8.7mmol) and N, N-diethylaniline (15mL) were added to a dry 20mL microwave tube, the reaction was heated to 250 ℃ in a microwave synthesizer and reacted for 2.5 hours, after completion of the reaction, diluted with ethyl acetate (100mL), washed with 2N hydrochloric acid to remove N, N-diethylaniline, the organic phase was washed times with saturated aqueous sodium chloride (50mL), dried over sodium sulfate, filtered, and distilled under reduced pressure to give a crude product, which was purified by silica gel column (petroleum ether: ethyl acetate ═ 20:1) to give a brown oily mixture of C3-1C and C3-1d (1.88g, 94% yield).
Step three: intermediate 6-fluoro-2H-chromene-5-carbonitrile (C3-1e)
In a dry 500mL three necked flask was added in the order C3-1C and C3-1d brown oil mixture (20.2g,88.2mmol), zinc cyanide (13.5g,115mmol), anhydrous N, N dimethylformamide (250mL) and tetrakis triphenylphosphine palladium (10.2g,8.8 mmol). the reaction was stirred at 110 ℃ under nitrogen protection for 18H after completion of the reaction, cooled to room temperature, 1L of water was added, extracted three times with ethyl acetate (500 mL). the organic phase was washed times with saturated aqueous sodium chloride (500mL), dried over sodium sulfate, filtered, and distilled under reduced pressure to give a crude product which was purified by silica gel column (petroleum ether: ethyl acetate ═ 50: 1-10: 1) to give 6-fluoro-2H-chromene-5-carbonitrile C3-1e (8.12g, 52.6% yield) of C3-1d which was isolated.
1H NMR(400MHz,CDCl3)δ7.02-6.85(m,2H),6.70(dt,J=10.0,1.8Hz,1H),6.07(dt,J=10.0,3.6Hz,1H),4.88(dd,J=3.6,2.0Hz,2H)ppm;LC-MS:m/z 176.1[M+H]+
Step four: intermediate tert-butyl ((6-fluorobenzo-dihydropyran-5-yl) methyl) carbamate (C3-1f)
Figure BDA0001732766820000381
6-fluoro-2H-chromene-5-carbonitrile C3-1e (4.6g,26.3mmol), BOC anhydride (7.45g,34.1mmol), methanol (200mL), and 10% palladium on carbon (1.6g, 50%) were added sequentially in a 500mL single-neck flask, the reaction was bubbled with hydrogen for 5 minutes, then purged with hydrogen balloon three times, heated to 60 ℃ in hydrogen and stirred overnight, after completion of the reaction, cooled to room temperature, the reaction was filtered, and distilled under reduced pressure to give the purer product C3-1f (7.4g, 100%) which was used directly in the next steps.
1H NMR(400MHz,CDCl3)δ6.81(t,J=9.1Hz,1H),6.70(dd,J=9.0,4.9Hz,1H),4.75(s,1H),4.32(d,J=5.2Hz,2H),4.18-4.05(m,2H),3.49(d,J=1.8Hz,2H),2.86(t,J=6.4Hz,2H),2.10-1.91(m,2H),1.45(d,J=8.0Hz,9H)ppm;LC-MS:m/z226.1[M-tBu+H]+
Step five: intermediate (6-fluorobenzchroman-5-yl) methylamine (C3-1)
Figure BDA0001732766820000382
A50 mL one-neck flask was charged with C3-1f (1.0g, 3.55mmol) and 10mL of HCl/dioxane solution (4M) in this order, and stirred at room temperature for 4 hours. After the reaction was complete, the mixture was concentrated under reduced pressure to give compound C3-1(612mg, 95% yield) as a white solid.
1H NMR(400MHz,CD3OD)δ6.98(t,J=9.2Hz,1H),6.86(dd,J=9.1,5.1Hz,1H),4.28-4.07(m,4H),2.89(t,J=6.4Hz,2H),2.14-2.01(m,2H)ppm;LC-MS:m/z182.1[M+H]+
Example 6: intermediate (5-fluorobenzo [ d ] [1,3] dioxol-4-yl) methylamine (C4-1)
Step intermediate 4-fluorobenzene-1, 2-diol (C4-1b)
Figure BDA0001732766820000391
4-fluoro-1, 2-dimethoxybenzene C4-1a (10g, 64.0mmol) was dissolved in anhydrous dichloromethane (120mL) at-78 deg.C, boron tribromide (1 molar solution in dichloromethane) was slowly added to the reaction mixture, the reaction mixture was allowed to warm to room temperature and stirring was continued for 16 hours, the reaction mixture was poured into crushed ice (200g) and extracted with ethyl acetate (3X 50mL), the organic phase was washed times with saturated aqueous sodium chloride (100mL), dried over sodium sulfate, filtered, and distilled under reduced pressure to give 4-fluorobenzene-1, 2-diol C4-1b (8.0g, 100%) as a colorless liquid.
1H NMR(400MHz,CDCl3):δ6.80-6.77(m,1H),6.65-6.63(m,1H),6.50-6.46(m,1H),6.25(s,1H),5.81(s,1H)ppm.
Step two: intermediate 5-fluorobenzo [ d ] [1,3] dioxole (C4-1C)
Figure BDA0001732766820000392
4-fluorobenzene-1, 2-diol C4-1b (8.0g, 62.5mmol) was dissolved in DMF (25.0mL), and cesium carbonate (30.4g, 93.75mmol) was added slowly at room temperature and stirred at the same temperature for 10 min. Chlorobromomethane (5.5mL, 93.75mmol) was added portionwise to the reaction mixture over 3 minutes at the same temperature. The resulting reaction mixture was stirred at 120 ℃ for 1 hour. The reaction mixture was cooled to room temperature and poured into crushed ice (200mL) and stirred for 15 minutes, then extracted with ether (3 × 50 mL). The combined organic phases were washed with brine (200mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The crude product was purified by silica gel column (petroleum ether: ethyl acetate ═ 20:1) to give 5-fluorobenzo [ d ] [1,3] dioxole C4-1C (4.0g, crude) as a colorless liquid.
GC-MS:m/z 139.1[M+H]+.
Step three: intermediate 5-fluorobenzo [ d ] [1,3] dioxol-4-carbaldehyde (C4-1d)
Figure BDA0001732766820000401
5-Fluorobenzo [ d ] [1,3] dioxole C4-1C (4.0g, 28.57mmol) was dissolved in anhydrous THF (50 mL). The reaction was cooled to-78 deg.C, lithium diisopropylamide (2M in THF, 28.57mL, 57.14mmol) was added over 15 minutes under nitrogen, and stirring continued at the same temperature for 1 hour. DMF (4.17mL, 57.14mmol) was added to the reaction and the reaction mixture was allowed to warm slowly to room temperature and stirred for 16 h. After completion of the reaction, the reaction mixture was poured into crushed ice (200mL), stirred for 15 minutes, and extracted with ethyl acetate (2X 50 mL). The combined organic phases were washed with brine (200mL), dried over anhydrous sodium sulfate and concentrated under reduced pressure. The crude product was purified by silica gel column (petroleum ether: ethyl acetate ═ 20:1) to give 5-fluorobenzo [ d ] [1,3] dioxol-4-one C4-1d (1.5g, 76% yield over two steps) as a yellow solid.
1H NMR(400MHz,CDCl3):δ10.26(s,1H),6.92(dd,J=8.5,4.0Hz,1H),6.60(dd,J=10.5,9.0Hz,1H),6.16(s,2H)ppm.
Step four: intermediate 5-fluorobenzo [ d ] [1,3] dioxol-4-carbaldehyde oxime (C4-1e)
Figure BDA0001732766820000402
5-Fluorobenzo [ d ] [1,3] dioxole-4-carbaldehyde C4-1d (1.5g, 8.92mmol) was dissolved in ethanol (10mL), and sodium hydroxide (535mg, 13.38mmol) dissolved in ethanol (10mL), water (8.0mL) and hydroxylamine hydrochloride (727mg, 10.70mmol) were added. The reaction solution was stirred at room temperature for 30 minutes. After the reaction was complete, the reaction was poured into cold water. The solid precipitate was collected by filtration, washed with water (100mL) and dried to give 5-fluorobenzo [ d ] [1,3] dioxole-4-carbaldehyde oxime C4-1e (1.2g, 75% yield) as a pale yellow solid.
1H NMR(400MHz,CDCl3):δ9.20(s,1H),8.30(s,1H),6.76-6.73(m,1H),6.60-6.56(m,1H),6.61(s,2H)ppm.
Step five: intermediate tert-butyl ((5-fluorobenzo [ d ] [1,3] dioxol-4-yl) methyl) carbamate (C4-1f)
Figure BDA0001732766820000411
At 0 ℃ to 5-fluorobenzo [ d][1,3]To a solution of dioxole-4-formaldoxime C4-1e (1.2g, 6.55mmol) in MeOH (10mL) was added nickel chloride hexahydrate (1.86g, 7.86 mmol). After stirring for 5 min, sodium borohydride (2.48g, 65.5mmol) was added in portions over 3 min, followed by (Boc)2O (1.71g, 7.86 mmol). The reaction was stirred at room temperature for an additional 30 minutes, then diluted with ethyl acetate (50mL) and filtered through celite. The crude product obtained by concentrating the filtrate under reduced pressure was purified by silica gel column (petroleum ether: ethyl acetate ═ 20:1) to give ((5-fluorobenzo [ d)][1,3]Dioxol-4-yl) methyl) carbamic acid tert-butyl ester C4-1f (1.2g, 68% yield) as an off-white solid.
1H NMR(400MHz,CDCl3):δ6.65-6.63(m,1H),6.52-6.49(m,1H),6.01(s,2H),4.93(bs,1H),4.35(s,2H),1.44(s,9H)ppm.
Step six: intermediate (5-fluorobenzo [ d ] [1,3] dioxol-4-yl) methylamine (C4-1)
Figure BDA0001732766820000421
Tert-butyl ((5-fluorobenzo [ d ] [1,3] dioxol-4-yl) methyl) carbamate C4-1f (1.2g, 10.11mmol) was dissolved in dichloromethane (15mL) and 4M HCl in dioxane (15mL, 60.6mmol) was added. After stirring at room temperature for 30 minutes, the volatile matter was removed by distillation under the reduced pressure. The crude product was slurried with ethyl acetate (10mL) and filtered to give (5-fluorobenzo [ d ] [1,3] dioxol-4-yl) methylamine hydrochloride C4-1(800mg, quantitative) as a white solid.
1H NMR(400MHz,CDCl3):δ8.46(s,D2O exchangeable,3H),6.98-6.95(m,2H),6.75(t,J=2.0Hz,2H),6.13(s,2H),3.96(s,2H)ppm;LC-MS:m/z 170.1[M+H]+.
Using the procedure described above, the following compounds were obtained using similar starting materials
Example 7: intermediate (6-fluoro-2, 3-dihydrobenzo [ b ] [1,4] dioxin-5-yl) methylamine (C4-2).
Figure BDA0001732766820000422
1H NMR(400MHz,DMSO-d6):δ6.89(dd,J=9.0,5.5Hz,1H),6.73(t,J=9.0Hz,1H),4.33-4.31(m,2H),4.24-4.22(m,2H),3.86(d,J=1.0Hz,2H)ppm;LC-MS:m/z184.1[M+H]+.
Example 8: 5-bromo-N- ((2, 3-dihydrobenzofuran-4-yl) methyl) - [1,2,4] triazolo [4,3-a ] pyrazin-8-amine (A-1)
Figure BDA0001732766820000431
A25 mL round bottom flask was charged with B1(98mg,0.42mmol), C1-1(100mg,0.38mmol), ethanol (8mL) and triethylamine (115mg, 1.14mmol) in that order. The reaction was stirred at 90 ℃ for 2 hours, then cooled to room temperature, and the volatiles were removed by distillation under reduced pressure. The crude product was purified on a silica gel column (dichloromethane: methanol ═ 30:1) to give 5-bromo-N- ((2, 3-dihydrobenzofuran-4-yl) methyl) - [1,2,4] triazolo [4,3-a ] pyrazin-8-amine a-1(70mg, 53% yield).
Using the above procedure, the following intermediates A-2, A-3, A-4, A-5, A-6, A-7, A-8, A-9 and A-10 were obtained using similar starting materials.
Figure BDA0001732766820000441
Figure BDA0001732766820000451
Example 9: n- ((2, 3-dihydrobenzofuran-4-yl) methyl) -5- (4- (methylsulfonyl) phenyl) - [1,2,4] triazolo [4,3-a ] pyrazin-8-amine
Figure BDA0001732766820000461
Intermediate A-1(30mg,0.086mmol), 1, 4-dioxane (2mL), purified water (0.5mL), 4- (methylsulfonyl) phenyl) boronic acid (21mg, 0.10mmol), [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium (6mg, 0.008mmol) and potassium carbonate (24mg, 0.17mmol) were added sequentially at room temperature in a 20mL stopcock. And sealing the tube and heating to 80 ℃ under the nitrogen atmosphere, and reacting for 6 hours. After completion of the reaction, 20mL of water was added to the reaction solution and extracted with ethyl acetate (30 mL. times.3). The organic phase was washed successively with water (20 mL. times.1), and saturated brine (20 mL. times.1). The organic phase was collected, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure and dissolved in methanol (3mL) and purified by high performance liquid chromatography to give 2- (((2, 3-dihydrobenzofuran-4-yl) methyl) amino) -3-methyl-5- (4- (methylsulfonyl) phenyl) pyrimidin-4 (3H) -one (12mg, yield: 32.9%) as a white solid.
1H NMR(400MHz,MeOD)δ9.13(s,1H),8.15(d,J=8.3Hz,2H),7.89(d,J=8.2Hz,2H),7.49(s,1H),7.12(t,J=7.7Hz,1H),6.94(d,J=7.6Hz,1H),6.72(d,J=8.0Hz,1H),4.81(s,2H),4.62(t,J=8.7Hz,2H),3.31(d,J=8.8Hz,2H),3.20(s,3H)ppm;LCMS:m/z 421[M+H]+
Using the above method, the following compounds can be synthesized:
example 10: n- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) -5- (4- (methylsulfonyl) phenyl) - [1,2,4] triazolo [4,3-a ] pyrazin-8-amine
1H NMR(400MHz,MeOD)δ9.13(s,1H),8.15(d,J=8.3Hz,2H),7.89(d,J=8.2Hz,2H),7.49(s,1H),7.12(t,J=7.7Hz,1H),6.94(d,J=7.6Hz,1H),6.72(d,J=8.0Hz,1H),4.81(s,2H),4.62(t,J=8.7Hz,2H),3.31(d,J=8.8Hz,2H),3.20(s,3H)ppm;LCMS:m/z 421[M+H]+
Example 11: n- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) -5- (4- (methylsulfonyl) phenyl) - [1,2,4] triazolo [4,3-a ] pyrazin-8-amine
Figure BDA0001732766820000471
1H NMR(400MHz,CDCl3)δ8.71(dd,J=4.8,1.5Hz,1H),8.32(s,1H),7.68(dd,J=7.7,1.5Hz,1H),7.37-7.29(m,2H),6.87(t,J=9.3Hz,1H),6.74-6.60(m,2H),4.84(d,J=5.5Hz,2H),4.63(t,J=8.7Hz,2H),3.40(t,J=8.7Hz,2H),2.44(s,3H)ppm;LCMS:m/z377.1[M+H]+
Example 12: n- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) -5- (4-methylpyridin-3-yl) - [1,2,4] triazolo [4,3-a ] pyrazin-8-amine
1H NMR(400MHz,CDCl3)δ8.66(d,J=4.6Hz,1H),8.59(s,1H),8.30(s,1H),7.39-7.29(m,2H),6.86(t,J=9.3Hz,1H),6.68(d,J=8.6Hz,2H),4.85(d,J=4.3Hz,2H),4.63(t,J=8.5Hz,2H),3.41(t,J=8.4Hz,2H),2.23(s,3H)ppm;LCMS:m/z 377.1[M+H]+
Example 13: n- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) -5- (2-fluoropyridin-4-yl) - [1,2,4] triazolo [4,3-a ] pyrazin-8-amine
Figure BDA0001732766820000473
1H NMR(400MHz,DMSO-d6)δ9.53(s,1H),9.07(s,1H),8.36(d,J=5.1Hz,1H),7.74-7.63(m,2H),7.55(s,1H),6.89(t,J=9.4Hz,1H),6.64(dd,J=8.5,3.6Hz,1H),4.73(d,J=4.2Hz,2H),4.51(t,J=8.7Hz,2H),3.30(d,J=8.7Hz,2H)ppm;LCMS:m/z 380.8[M+H]+
Example 14: n- ((6-Fluorochroman-5-yl) methyl) -5- (4-methylpyridin-3-yl) - [1,2,4] triazolo [4,3-a ] pyrazin-8-amine
1H NMR(400MHz,CDCl3)δ8.67(d,J=5.0Hz,1H),8.60(s,1H),8.30(s,1H),7.42-7.31(m,2H),6.90(t,J=9.0Hz,1H),6.79(dd,J=9.0,4.9Hz,1H),6.36(s,1H),4.85(d,J=4.2Hz,2H),4.22-4.11(m,2H),2.95(t,J=6.5Hz,2H),2.24(s,3H),2.04(dd,J=11.2,5.7Hz,2H)ppm;LCMS:m/z 391.1[M+H]+
Example 15: n- ((6-Fluorochroman-5-yl) methyl) -5- (2-methylpyridin-3-yl) - [1,2,4] triazolo [4,3-a ] pyrazin-8-amine
Figure BDA0001732766820000482
1H NMR(400MHz,CDCl3)δ8.72(dd,J=4.9,1.6Hz,1H),8.32(s,1H),7.69(dd,J=7.7,1.6Hz,1H),7.40-7.29(m,2H),6.90(t,J=9.1Hz,1H),6.79(dd,J=9.0,4.9Hz,1H),6.36(s,1H),4.84(d,J=4.4Hz,2H),4.20-4.08(m,2H),2.95(t,J=6.6Hz,2H),2.46(s,3H),2.04(dd,J=11.1,5.8Hz,2H)ppm;LCMS:m/z 391.1[M+H]+
Example 16: 4- (8- (((6-fluorochroman-5-yl) methyl) amino) - [1,2,4] triazolo [4,3-a ] pyrazin-5-yl) benzamide
Figure BDA0001732766820000483
1H NMR(400MHz,DMSO-d6)δ9.34(s,1H),8.53(s,1H),8.12(s,1H),8.04(d,J=7.8Hz,2H),7.79(d,J=7.8Hz,2H),7.49(s,2H),6.93(t,J=9.0Hz,1H),6.78-6.66(m,1H),4.72(s,2H),4.06(s,2H),2.91(s,2H),1.92(s,2H)ppm;LCMS:m/z 418.8[M+H]+
Example 17: tert-butyl (4- (8- (((6-fluorochroman-5-yl) methyl) amino) - [1,2,4] triazolo [4,3-a ] pyrazin-5-yl) cyclohex-3-en-1-yl) carbamate
Figure BDA0001732766820000491
1H NMR(400MHz,MeOD)δ9.15(s,1H),7.64-7.33(m,1H),7.16(s,1H),6.76(t,J=9.2Hz,1H),6.62(dd,J=9.0,4.8Hz,1H),6.06(s,1H),4.63(d,J=8.6Hz,2H),4.06-3.94(m,2H),3.68(d,J=4.5Hz,1H),2.81(t,J=6.5Hz,2H),2.46(dd,J=28.2,16.9Hz,3H),2.08(dd,J=15.8,8.0Hz,1H),1.99-1.80(m,3H),1.74-1.61(m,1H),1.37(s,9H)ppm;LCMS:m/z 494.8[M+H]+
Example 18: 5- (4-aminocyclohex-1-en-1-yl) -N- ((6-fluorochroman-5-yl) methyl) - [1,2,4] triazolo [4,3-a ] pyrazin-8-amine
Figure BDA0001732766820000492
1H NMR(400MHz,MeOD)δ9.29(d,J=3.4Hz,1H),7.31(s,1H),6.90(t,J=9.2Hz,1H),6.76(dd,J=9.0,4.9Hz,1H),6.22(s,1H),4.79(s,2H),4.18-4.07(m,2H),3.60(s,1H),2.94(t,J=6.5Hz,2H),2.83-2.56(m,3H),2.43(d,J=7.0Hz,1H),2.26(d,J=9.9Hz,1H),2.08-1.92(m,3H)ppm;LCMS:m/z 395.2[M+H]+
Example 19: n- ((5-fluorobenzofuran-4-yl) methyl) -5- (2-methylpyridin-3-yl) - [1,2,4] triazolo [4,3-a ] pyrazin-8-amine
1H NMR(400MHz,DMSO-d6)δ8.98(t,J=5.6Hz,1H),8.91(s,1H),8.61(d,J=4.2Hz,1H),8.02(d,J=1.6Hz,1H),7.83(d,J=7.3Hz,1H),7.53(dd,J=8.7,3.7Hz,1H),7.38(dd,J=7.4,5.0Hz,1H),7.29(d,J=12.9Hz,2H),7.16(t,J=9.6Hz,1H),4.98(d,J=4.7Hz,2H),2.32(s,3H)ppm;LCMS:m/z 375.1[M+H]+
Example 20: n- ((5-fluorobenzofuran-4-yl) methyl) -5- (4- (methylsulfonyl) phenyl) - [1,2,4] triazolo [4,3-a ] pyrazin-8-amine
Figure BDA0001732766820000501
1H NMR(400MHz,DMSO-d6)δ9.39(s,1H),9.15(s,1H),8.08-7.89(m,4H),7.53(dd,J=10.8,5.3Hz,2H),7.28(s,1H),7.16(t,J=9.7Hz,1H),4.99(d,J=5.5Hz,2H),3.29(s,3H)ppm;LCMS:m/z 438.1[M+H]+
Example 21: n- ((6-fluoro-2, 3-dihydrobenzo [ b ] [1,4] dioxin-5-yl) methyl) -5- (4- (methylsulfonyl) phenyl) - [1,2,4] triazolo [4,3-a ] pyrazin-8-amine
Figure BDA0001732766820000502
1H NMR(400MHz,DMSO-d6)δ9.39(s,1H),8.36(t,J=4.9Hz,1H),8.07(d,J=8.3Hz,2H),7.99(d,J=8.3Hz,2H),7.56(s,1H),6.84(dd,J=9.0,5.5Hz,1H),6.68(t,J=9.1Hz,1H),4.74(d,J=4.6Hz,2H),4.32(d,J=4.0Hz,2H),4.24(d,J=3.8Hz,2H),3.30(s,3H)ppm;LCMS:m/z 456.1[M+H]+
Example 22: n- ((6-fluoro-2, 3-dihydrobenzo [ b ] [1,4] dioxin-5-yl) methyl) -5- (2-methylpyridin-3-yl) - [1,2,4] triazolo [4,3-a ] pyrazin-8-amine
Figure BDA0001732766820000503
1H NMR(400MHz,DMSO-d6)δ8.91(s,1H),8.63(d,J=3.8Hz,1H),8.15(t,J=5.0Hz,1H),7.86(d,J=6.7Hz,1H),7.40(dd,J=7.6,4.9Hz,1H),7.33(s,1H),6.85(dd,J=9.0,5.5Hz,1H),6.69(t,J=9.2Hz,1H),4.70(dd,J=23.0,4.5Hz,2H),4.32(d,J=10.6,6.2Hz,2H),4.24(d,J=4.3Hz,2H),2.34(s,3H)ppm;LCMS:m/z 393.1[M+H]+
Example 23: n- ((5-fluorobenzo [ d ] [1,3] dioxazol-4-yl) methyl) -5- (2-methylpyridin-3-yl) - [1,2,4] triazolo [4,3-a ] pyrazin-8-amine
Figure BDA0001732766820000511
1H NMR(400MHz,DMSO-d6)δ8.91(s,1H),8.63(d,J=4.5Hz,1H),8.54(t,J=5.2Hz,1H),7.86(d,J=7.5Hz,1H),7.40(dd,J=7.6,4.9Hz,1H),7.30(s,1H),6.84(dd,J=8.5,4.2Hz,1H),6.72-6.58(m,1H),6.03(d,J=13.0Hz,2H),4.70(dd,J=21.2,5.7Hz,2H),2.33(s,3H)ppm;LCMS:m/z 379.1[M+H]+
Example 24: n- ((5-Fluorobenzo [ d ] [1,3] dioxazol-4-yl) methyl) -5- (4- (methylsulfonyl) phenyl) - [1,2,4] triazolo [4,3-a ] pyrazin-8-amine
Figure BDA0001732766820000512
1H NMR(400MHz,DMSO-d6)δ9.40(s,1H),8.72(d,J=5.3Hz,1H),8.06(d,J=8.3Hz,2H),7.99(d,J=8.4Hz,2H),7.53(s,1H),6.83(dd,J=8.5,4.2Hz,1H),6.65(dd,J=10.7,8.6Hz,1H),6.04(s,2H),4.74(d,J=4.9Hz,2H),3.30(s,3H)ppm;LCMS:m/z 442.1[M+H]+
Example 25: 5-cyclopropyl-N- ((6-fluorochroman-5-yl) methyl) - [1,2,4] triazolo [4,3-a ] pyrazin-8-amine
Figure BDA0001732766820000513
1H NMR(400MHz,DMSO-d6)δ9.39(s,1H),8.09(s,1H),7.12(s,1H),6.90(t,J=9.1Hz,1H),6.70(d,J=4.6Hz,1H),4.63(s,2H),4.05(s,2H),2.87(s,2H),2.04(s,1H),1.89(s,2H),0.97(d,J=7.0Hz,2H),0.74(s,2H)ppm;LCMS:m/z 340.1[M+H]+
Example 26: n- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) -5- (4- (methylsulfonyl) phenyl) - [1,2,4] triazolo [1,5-a ] pyrazin-8-amine
Figure BDA0001732766820000521
1H NMR(400MHz,DMSO-d6)δ8.67(s,1H),8.62(s,1H),8.25(s,2H),8.05(s,2H),7.99(s,1H),6.89(s,1H),6.64(s,1H),4.75(s,2H),4.51(s,2H),3.32(s,2H),3.28(s,3H)ppm;LCMS:m/z 440.1[M+H]+
Example 27: n- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) -5- (2-methylpyridin-3-yl) - [1,2,4] triazolo [1,5-a ] pyrazin-8-amine
1H NMR(400MHz,CDCl3)δ8.66(dd,J=4.9,1.6Hz,1H),8.25(s,1H),7.71(dd,J=7.7,1.7Hz,1H),7.57(s,1H),7.29(dd,J=7.7,5.0Hz,1H),6.86(t,J=9.4Hz,1H),6.68(dd,J=8.7,3.9Hz,1H),6.28(t,J=5.4Hz,1H),4.83(d,J=5.7Hz,2H),4.63(t,J=8.7Hz,2H),3.41(t,J=8.7Hz,2H),2.44(s,3H)ppm;LCMS:m/z 377.1[M+H]+
Example 28: n- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) -5- (4-methylpyridin-3-yl) - [1,2,4] triazolo [1,5-a ] pyrazin-8-amine
1H NMR(400MHz,CDCl3)δ8.61(dd,J=4.9,1.6Hz,1H),8.57(s,1H),8.24(s,1H),7.59(s,1H),7.30(dd,J=7.7,5.0Hz,1H),6.87(t,J=9.4Hz,1H),6.68(dd,J=8.7,3.9Hz,1H),6.33(s,1H),4.83(d,J=5.7Hz,2H),4.63(t,J=8.7Hz,2H),3.41(t,J=8.7Hz,2H),2.22(s,3H)ppm;LCMS:m/z 377.1[M+H]+
Example 29: n- ((6-Fluorochroman-5-yl) methyl) -5- (2-methylpyridin-3-yl) - [1,2,4] triazolo [1,5-a ] pyrazin-8-amine
1H NMR(400MHz,MeOD)δ8.46(d,J=3.9Hz,1H),8.22(s,1H),7.77(d,J=7.3Hz,1H),7.52(s,1H),7.35-7.24(m,1H),6.77(t,J=9.0Hz,1H),6.63(dd,J=8.6,4.7Hz,1H),4.70(d,2H),4.00(t,J=4.6Hz,2H),3.21(s,3H),2.86(t,J=6.1Hz,2H),2.27(s,3H),1.88(t,J=4.9Hz,2H)ppm;LCMS:m/z 391.1[M+H]+
Example 30: n- ((6-Fluorochroman-5-yl) methyl) -5- (4-methylpyridin-3-yl) - [1,2,4] triazolo [1,5-a ] pyrazin-8-amine
Figure BDA0001732766820000532
1H NMR(400MHz,DMSO-d6)δ8.98(s,1H),8.90(d,J=5.4Hz,1H),8.64(s,1H),8.55(s,1H),8.05(d,J=5.6Hz,1H),7.82(s,1H),6.94(t,J=9.2Hz,1H),6.76-6.72(m,1H),4.73(d,J=4.1Hz,2H),4.12-4.02(m,2H),2.93(t,J=6.3Hz,2H),2.43(s,3H),1.99-1.87(m,2H)ppm;LCMS:m/z 391.1[M+H]+
Example 31: n- ((6-Fluorochroman-5-yl) methyl) -5- (pyrimidin-5-yl) - [1,2,4] triazolo [1,5-a ] pyrazin-8-amine
Figure BDA0001732766820000533
1H NMR(400MHz,DMSO-d6)δ9.38(s,2H),9.23(s,1H),8.62(s,1H),8.53(t,J=4.9Hz,1H),8.07(s,1H),6.92(t,J=9.2Hz,1H),6.72(dd,J=8.9,4.8Hz,1H),4.72(d,J=4.7Hz,2H),4.12-4.00(m,2H),2.90(t,J=6.4Hz,2H),1.98-1.85(m,2H)ppm;LCMS:m/z377.8[M+H]+
Example 32: n- ((6-Fluorochroman-5-yl) methyl) -5- (prop-1-en-2-yl) - [1,2,4] triazolo [1,5-a ] pyrazin-8-amine
Figure BDA0001732766820000541
1H NMR(400MHz,DMSO-d6)δ8.58(s,1H),8.20(t,J=5.2Hz,1H),7.71(s,1H),6.91(t,J=9.2Hz,1H),6.70(dd,J=9.0,4.8Hz,1H),6.32(s,1H),5.40(s,1H),4.68(d,J=4.9Hz,2H),4.10-3.99(m,2H),2.87(t,J=6.5Hz,2H),2.22(s,3H),1.97-1.81(m,2H)ppm;LCMS:m/z 339.9[M+H]+
Example 33: n- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) -5- (4-fluorophenyl) - [1,2,4] triazolo [1,5-a ] pyrazin-8-amine
1H NMR(400MHz,DMSO-d6)δ8.58(s,1H),8.47(t,J=5.8Hz,1H),8.02-7.92(m,2H),7.80(s,1H),7.36(t,J=8.9Hz,2H),6.93-6.84(m,1H),6.63(dd,J=8.6,3.8Hz,1H),4.71(d,J=5.7Hz,2H),4.51(t,J=8.7Hz,2H),3.29(d,J=8.7Hz,2H)ppm;LCMS:m/z 379.8[M+H]+
Example 34: n- ((6-Fluorochroman-5-yl) methyl) -5-methyl- [1,2,4] triazolo [1,5-a ] pyrazin-8-amine
Figure BDA0001732766820000543
1H NMR(400MHz,DMSO-d6)δ8.51(s,1H),7.78(t,J=5.1Hz,1H),7.47(s,1H),6.90(t,J=9.2Hz,1H),6.70(dd,J=9.0,4.8Hz,1H),4.63(d,J=4.8Hz,2H),4.09-3.99(m,2H),2.86(t,J=6.5Hz,2H),2.50-2.48(m,3H),1.95-1.81(m,2H)ppm;LCMS:m/z 313.9[M+H]+
Example 35: n- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) -5- (1-methyl-1, 2,3, 6-tetrahydropyridin-4-yl) - [1,2,4] triazolo [1,5-a ] pyrazin-8-amine
Figure BDA0001732766820000551
1H NMR(400MHz,DMSO-d6)δ8.57(s,1H),8.36(t,J=5.8Hz,1H),7.60(s,1H),6.99(t,J=3.3Hz,1H),6.92-6.80(m,1H),6.62(dd,J=8.6,3.8Hz,1H),4.67(d,J=5.7Hz,2H),4.49(t,J=8.8Hz,2H),3.26(t,J=8.7Hz,2H),3.13(d,J=2.6Hz,2H),2.62(s,4H),2.31(s,3H)ppm;LCMS:m/z 380.9[M+H]+
Example 36: n- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) -5- (1,2,3, 6-tetrahydropyridin-4-yl) - [1,2,4] triazolo [1,5-a ] pyrazin-8-amine
Figure BDA0001732766820000552
1H NMR(400MHz,DMSO-d6)δ8.59(s,1H),8.43(t,J=5.7Hz,1H),8.24(s,1H),7.63(s,1H),7.01(s,1H),6.92-6.83(m,1H),6.63(dd,J=8.6,3.8Hz,1H),4.68(d,J=5.7Hz,2H),4.49(t,J=8.7Hz,2H),3.63(s,2H),3.28(d,J=8.8Hz,2H),3.12(t,J=5.5Hz,2H),2.62(s,2H)ppm;LCMS:m/z 366.9[M+H]+
Example 37: n- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) -8- (4- (methylsulfonyl) phenyl) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-amine
Figure BDA0001732766820000553
1H NMR(400MHz,DMSO-d6)δ8.34(s,1H),8.26(s,1H),8.18(d,J=8.4Hz,2H),8.05(d,J=8.4Hz,2H),6.87(t,J=8.8Hz,1H),6.70-6.67(m,2H),4.89(d,J=6.0Hz,2H),4.63(t,J=8.8Hz,2H),3.42(t,J=8.4Hz,2H)ppm;LCMS:m/z 440.1[M+H]+
Example 38: n- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) -8- (2-methylpyridin-3-yl) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-amine
1H NMR(400MHz,CDCl3)δ8.58(d,J=4.5Hz,1H),8.27(s,1H),7.89(s,1H),7.68(d,J=7.5Hz,1H),7.24(d,J=7.2Hz,1H),6.88(t,J=9.2Hz,1H),6.69(dd,J=8.5,3.8Hz,1H),6.58(s,1H),4.87(d,J=5.9Hz,2H),4.64(t,J=8.7Hz,2H),3.43(t,J=8.7Hz,2H),2.53(s,3H)ppm;LCMS:m/z 377.1[M+H]+
Example 39: n- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) -8- (4-methylpyridin-3-yl) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-amine
Figure BDA0001732766820000562
1H NMR(400MHz,CDCl3)δ8.53(s,2H),8.26(s,1H),7.89(s,1H),7.27(s,1H),6.87(t,J=9.4Hz,1H),6.69(dd,J=8.6,3.9Hz,1H),6.58(s,1H),4.88(d,J=6.0Hz,2H),4.64(t,J=8.7Hz,2H),3.44(t,J=8.7Hz,2H),2.29(s,3H)ppm;LCMS:m/z 377.1[M+H]+
Example 40: n- ((6-Fluorochroman-5-yl) methyl) -8- (2-methylpyridin-3-yl) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-amine
Figure BDA0001732766820000563
1H NMR(400MHz,CDCl3)δ8.58(dd,J=4.8,1.6Hz,1H),8.26(s,1H),7.91(s,1H),7.69(dd,J=7.6,1.5Hz,1H),7.25-7.20(m,1H),6.91(t,J=9.1Hz,1H),6.80(dd,J=9.0,5.0Hz,1H),4.89(dd,J=5.5,1.3Hz,2H),4.21-4.10(m,2H),2.99(t,J=6.5Hz,2H),2.54(s,3H),2.05(p,J=6.4Hz,2H)ppm;LCMS:m/z 391.1[M+H]+
Example 41: n- ((6-Fluorochroman-5-yl) methyl) -8- (2-methylpyridin-3-yl) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-amine
Figure BDA0001732766820000571
1H NMR(400MHz,CDCl3)δ8.53(t,J=2.4Hz,2H),8.25(s,1H),7.92(s,1H),7.27(s,1H),7.26-7.25(m,1H),6.91(s,1H),6.81(d,J=5.0Hz,1H),6.39(t,J=5.3Hz,1H),4.90(dd,J=5.5,1.4Hz,2H),4.22-4.09(m,2H),3.00(t,J=6.5Hz,2H),2.31(s,3H),2.12-1.98(m,2H)ppm;LCMS:m/z 391.1[M+H]+
Example 42: 8- (2, 4-difluorophenyl) -N- ((6-fluorochroman-5-yl) methyl) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-amine
Figure BDA0001732766820000572
1H NMR(400MHz,CDCl3)δ8.27(s,1H),8.14(d,J=1.5Hz,1H),7.82(td,J=8.5,6.5Hz,1H),7.00(tdd,J=11.1,8.7,2.5Hz,2H),6.90(t,J=9.1Hz,1H),6.78(dd,J=9.0,5.0Hz,1H),6.38(t,J=5.2Hz,1H),4.89(dd,J=5.5,1.4Hz,2H),4.17-4.12(m,2H),2.97(t,J=6.6Hz,2H),2.03(dd,J=10.2,4.9Hz,2H)ppm;LCMS:m/z 411.8[M+H]+
Example 43: n- ((6-Fluorochroman-5-yl) methyl) -8- (2-fluorophenyl) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-amine
Figure BDA0001732766820000573
1H NMR(400MHz,DMSO-d6)δ8.65(s,1H),8.55(s,1H),8.10(s,1H),7.81(t,J=7.7Hz,1H),7.46(dd,J=13.0,6.0Hz,1H),7.33(dd,J=16.5,8.7Hz,2H),6.93(t,J=9.3Hz,1H),6.73(dd,J=9.0,4.8Hz,1H),4.76(d,J=4.7Hz,2H),4.10-4.03(m,2H),2.92(t,J=6.4Hz,2H),1.97-1.82(m,2H)ppm;LCMS:m/z 393.8[M+H]+
Example 44: n- ((6-Fluorochroman-5-yl) methyl) -8- (4- (trifluoromethyl) phenyl) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-amine
Figure BDA0001732766820000581
1H NMR(400MHz,CDCl3)δ8.32(s,1H),8.24(s,1H),8.08(d,J=8.2Hz,2H),7.74(d,J=8.3Hz,2H),6.90(t,J=9.1Hz,1H),6.79(dd,J=9.0,5.0Hz,1H),6.45(t,J=5.3Hz,1H),4.90(dd,J=5.5,1.2Hz,2H),4.17-4.12(m,2H),2.97(t,J=6.6Hz,2H),2.08-2.02(m,2H)ppm;LCMS:m/z 443.8[M+H]+
Example 45: 8- (cyclohex-1-en-1-yl) -N- ((6-fluorochroman-5-yl) methyl) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-amine
1H NMR(400MHz,CDCl3)δ8.24(s,1H),7.90(s,1H),6.94(t,J=3.9Hz,1H),6.92-6.83(m,1H),6.76(dd,J=9.0,4.9Hz,1H),6.19(t,J=5.2Hz,1H),4.82(dd,J=5.5,1.2Hz,2H),4.18-4.06(m,2H),2.93(t,J=6.6Hz,2H),2.57-2.45(m,2H),2.31(dd,J=6.1,2.4Hz,2H),2.01(dq,J=12.9,6.4Hz,2H),1.89-1.80(m,2H),1.76-1.65(m,2H)ppm;LCMS:m/z379.9[M+H]+
Example 46: n- ((6-Fluorochroman-5-yl) methyl) -8- (3-fluorophenyl) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-amine
Figure BDA0001732766820000583
1H NMR(400MHz,CDCl3)δ8.31(s,1H),8.20(s,1H),7.78-7.68(m,2H),7.46(dt,J=14.1,7.1Hz,1H),7.07(td,J=8.4,2.5Hz,1H),6.95-6.86(m,1H),6.78(dd,J=9.0,5.0Hz,1H),6.38(t,J=5.1Hz,1H),4.89(dd,J=5.5,1.4Hz,2H),4.19-4.12(m,2H),2.97(t,J=6.6Hz,2H),2.09-1.99(m,2H)ppm;LCMS:m/z 393.8[M+H]+Example 47: 8- (2, 3-dichlorophenyl) -N- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) - [1,2,4]Triazolo [1,5-c]Pyrimidin-5-amines
Figure BDA0001732766820000591
1H NMR(400MHz,CDCl3)δ8.26(s,1H),8.00(s,1H),7.54(dd,J=8.0,1.3Hz,1H),7.42(dd,J=7.6,1.3Hz,1H),7.31(t,J=7.8Hz,1H),6.87(t,J=9.4Hz,1H),6.69(dd,J=8.6,3.9Hz,1H),6.60(t,J=5.7Hz,1H),4.88(d,J=5.9Hz,2H),4.63(t,J=8.7Hz,2H),3.43(t,J=8.7Hz,2H)ppm;LCMS:m/z 429.7[M+H]+
Example 48: n- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) -8- (4-methoxyphenyl) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-amine
Figure BDA0001732766820000592
1H NMR(400MHz,CDCl3)δ8.30(s,1H),8.10(s,1H),7.86(d,J=8.8Hz,2H),7.02(t,J=8.6Hz,2H),6.91-6.82(m,1H),6.67(dd,J=8.6,3.9Hz,1H),6.47(d,J=6.1Hz,1H),4.86(d,J=6.0Hz,2H),4.62(t,J=8.7Hz,2H),3.86(s,3H),3.41(t,J=8.7Hz,2H)ppm;LCMS:m/z 391.8[M+H]+
Example 49: 8- (3, 5-Dimethylisothiazol-4-yl) -N- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-amine
Figure BDA0001732766820000593
1H NMR(400MHz,CDCl3)δ8.27(s,1H),7.81(s,1H),6.90-6.84(m,1H),6.69(dd,J=8.7,3.9Hz,1H),6.58(s,1H),4.85(d,J=5.9Hz,2H),4.64(t,J=8.7Hz,2H),3.43(t,J=8.7Hz,2H),2.41(s,3H),2.27(s,3H)ppm;LCMS:m/z 380.8[M+H]+
Example 50: n- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) -8-vinyl- [1,2,4] triazolo [1,5-c ] pyrimidin-5-amine
Figure BDA0001732766820000601
1H NMR(400MHz,CDCl3)δ8.30(s,1H),7.89(s,1H),6.85(t,J=9.4Hz,1H),6.75(dd,J=17.6,11.3Hz,1H),6.66(dd,J=8.6,3.9Hz,1H),6.56-6.43(m,2H),5.49(d,J=11.3Hz,1H),4.83(d,J=6.0Hz,2H),4.61(t,J=8.7Hz,2H),3.37(t,J=8.7Hz,2H)ppm;LCMS:m/z 311.9[M+H]+
Example 51: 8- (4, 4-Difluorocyclohex-1-en-1-yl) -N- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) - [1,2,4] triazolo [1,5-c ] pyrimidin-5-amine
Figure BDA0001732766820000602
1H NMR(400MHz,CDCl3)δ8.26(s,1H),7.90(s,1H),6.84(dd,J=11.8,6.7Hz,2H),6.66(dd,J=8.7,3.9Hz,1H),6.48(t,J=5.7Hz,1H),4.82(d,J=6.0Hz,2H),4.61(t,J=8.7Hz,2H),3.38(t,J=8.7Hz,2H),2.82(dd,J=19.1,10.5Hz,4H),2.23(ddd,J=20.4,13.5,6.6Hz,2H)ppm;LCMS:m/z 401.8[M+H]+
Example 52N- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) -7- (4- (methylsulfonyl) phenyl) - [1,2,5] thiadiazolo [3,4-c ] pyridin-4-amine
Figure BDA0001732766820000603
1H NMR(400MHz,CDCl3)δ8.24(s,1H),8.05(d,J=8.5Hz,2H),7.97(d,J=8.5Hz,2H),6.85–6.71(m,1H),6.59(dd,J=8.7,3.9Hz,1H),6.46(s,1H),4.84(d,J=5.8Hz,2H),4.55(t,J=8.7Hz,2H),3.35(t,J=8.7Hz,2H),3.03(s,3H)ppm;LCMS:m/z 457.1[M+H]+
Example 53N8- ((6-fluoro-2, 3-dihydrobenzo [ b ]][1,4]Dioxin-5-yl) methyl) -N5- (pyridin-2-yl) - [1,2,4]Triazolo [4,3-a]Pyrazine-5, 8-diamines
Figure BDA0001732766820000611
Adding 5-bromo-N- ((6-fluoro-2, 3-dihydrobenzo [ b ] in sequence into a 25mL single-mouth bottle][1,4]Dioxin-5-yl) methyl) - [1,2,4]Triazolo [4,3-a]Pyrazine-8-amine (80mg,0.21mmol), pyridin-2-amine (30mg,0.32mmol), Pd2(dba)3(38mg,0.042mmol), Brett-Phos (17mg,0.032mmol) and cesium carbonate (137mg,0.42mmol), 1, 4-dioxane (4mL) is added, the reaction solution is stirred at 100 ℃ in a nitrogen atmosphere overnight, after the reaction is finished, ethyl acetate (8mL) is added to the reaction solution for dilution, water (2mL x 2) is added for washing for 2 times, the organic phase is dried and concentrated, and the crude product is prepared by liquid chromatography to obtain an off-white solid product N8- ((6-fluoro-2, 3-dihydrobenzo [ b ] b][1,4]Dioxin-5-yl) methyl) -N5- (pyridin-2-yl) - [1,2,4]Triazolo [4,3-a]Pyrazine-5, 8-diamine (6mg, 7% yield).
1H NMR(400MHz,CDCl3)δ8.55(s,1H),8.17(d,J=4.8Hz,1H),7.52(t,J=7.3Hz,1H),7.42(s,1H),6.86–6.74(m,2H),6.62(t,J=8.9Hz,1H),6.56(s,1H),6.44(d,J=8.5Hz,2H),4.87(d,J=5.5Hz,2H),4.42–4.31(m,2H),4.29–4.16(m,2H)ppm;LCMS:m/z394.1[M+H]+
Example 54: n- ((6-fluorobenzo dihydropyran-5-yl) methyl) -5- (phenylthio) - [1,2,4] triazolo [4,3-a ] pyrazin-8-amine
Figure BDA0001732766820000621
To a microwave tube were added 5-bromo-N- ((6-fluorobenzohydropyran-5-yl) methyl) - [1,2,4] triazolo [4,3-a ] pyrazin-8-amine (50mg,132umol), sodium thiophenol (34.94mg,264umol), dibenzylideneacetone dipalladium (36.32mg,39.66 umol), 4, 5-bis diphenylphosphino-99-dimethylxanthene (22.95mg,39.66umol), N-ethyl-N-isopropylpropan-2-amine (34.17,264umol) and 1, 4-dioxane (4mL) in this order. The reaction temperature was raised to 100 ℃ and stirred for 2 hours. After the completion of the reaction, the reaction mixture was filtered, and the reaction mixture was spin-dried and separated by liquid chromatography to give N- ((6-fluorobenzchroman-5-yl) methyl) -5- (phenylthio) - [1,2,4] triazolo [4,3-a ] pyrazin-8-amine (16mg, yield: 30%) as a white solid.
1H NMR(400MHz,DMSO-d6)δ9.12(s,1H),8.88(s,1H),7.78(s,1H),7.32(d,J=4.1Hz,4H),7.26(dd,J=9.0,4.2Hz,1H),6.92(t,J=9.2Hz,1H),6.72(dd,J=9.0,4.8Hz,1H),4.70(d,J=3.6Hz,2H),4.12–3.98(m,2H),2.89(t,J=6.4Hz,2H),1.97–1.83(m,2H)ppm;LCMS:408.1[M+H]+
Using the above method, the following compounds can be synthesized:
example 55: 5- ((2, 3-dichloropyridin-4-yl) thio) -N- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) - [1,2,4] triazolo [4,3-a ] pyrazin-8-amine
Figure BDA0001732766820000622
1H NMR(400MHz,DMSO-d6)δ9.36(s,1H),9.24(s,1H),8.04(d,J=5.3Hz,1H),7.80(s,1H),6.89(dd,J=17.1,7.2Hz,2H),6.66(dd,J=8.6,3.8Hz,1H),4.73(d,J=5.0Hz,2H),4.53(t,J=8.7Hz,2H),3.31–3.29(m,2H)ppm;LCMS:m/z 463.0[M+H]+
Example 56: n- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) -5- (1,2,3, 6-tetrahydropyridin-4-yl) - [1,2,4] triazolo [1,5-a ] pyrazin-8-amine
Figure BDA0001732766820000631
To a dry 25mL single neck flask were added (8- (((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) amino) - [1,2,4] triazolo [1,5-a ] pyrazin-5-yl) -3, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester (50mg,107umol) and 4mol/L hydrochloric acid-1, 4-dioxane solution (2mL) in that order. The reaction was allowed to react at room temperature and stirred for half an hour. After the completion of the reaction, the organic solvent was spin-dried, and separated by liquid chromatography to give N- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) -5- (1,2,3, 6-tetrahydropyridin-4-yl) - [1,2,4] triazolo [1,5-a ] pyrazin-8-amine (10mg, yield: 26%) as a white solid.
1H NMR(400MHz,DMSO-d6)δ8.59(s,1H),8.43(t,J=5.7Hz,1H),8.24(s,1H),7.63(s,1H),7.01(s,1H),6.92–6.83(m,1H),6.63(dd,J=8.6,3.8Hz,1H),4.68(d,J=5.7Hz,2H),4.49(t,J=8.7Hz,2H),3.63(s,2H),3.28(d,J=8.8Hz,2H),3.12(t,J=5.5Hz,2H),2.62(s,2H)ppm;LCMS:m/z 366.9[M+H]+
Using the above method, the following compounds can be synthesized:
example 57: 5- (4-aminocyclohex-1-en-1-yl) -N- ((6-fluorochroman-5-yl) methyl) - [1,2,4] triazolo [4,3-a ] pyrazin-8-amine
Figure BDA0001732766820000632
1H NMR(400MHz,MeOD)δ9.29(d,J=3.4Hz,1H),7.31(s,1H),6.90(t,J=9.2Hz,1H),6.76(dd,J=9.0,4.9Hz,1H),6.22(s,1H),4.79(s,2H),4.18–4.07(m,2H),3.60(s,1H),2.94(t,J=6.5Hz,2H),2.83–2.56(m,3H),2.43(d,J=7.0Hz,1H),2.26(d,J=9.9Hz,1H),2.08–1.92(m,3H)ppm;LCMS:m/z 395.2[M+H]+
Example 58: 8- (((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) amino) - [1,2,4] triazolo [4,3-a ] pyrazine-5-carbonitrile
Figure BDA0001732766820000641
Respectively adding 5-bromo-N- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) - [1,2,4] triazolo [4,3-a ] pyrazine-8-amine A-2(200mg,0.64mmol), zinc cyanide (114mg,0.97mmol), palladium (112mg,0.097mmol) and DMF (6mL) into a 25mL single-neck flask, reacting for 16h, cooling to room temperature after the reaction is finished, adding ethyl acetate (20mL) for dilution and dissolution, washing 3 times with saturated saline (5mL x 3), drying an organic phase with anhydrous sodium sulfate, concentrating under reduced pressure, purifying a crude product with column chromatography to obtain a light yellow solid product 8- ((5-fluoro-2, 3-dihydrobenzofuran-4-yl) methyl) amino) - [1,2,4] triazolo [4,3-a ] pyrazine-5-carbonitrile (160mg, yield 80%).
1H NMR(400MHz,DMSO-d6)δ7.34(s,1H),6.36(s,1H),5.30(t,J=10.5Hz,1H),5.13(dd,J=8.5,3.8Hz,1H),3.31(d,J=5.8Hz,2H),3.06(t,J=8.7Hz,2H),1.78(t,J=8.8Hz,2H)ppm;LCMS:m/z 311.1[M+H]+
Pharmacology and uses
EED, , which is the major component of the PRC2 protein complex, although it does not have enzymatic activity, has important role in the overall function of PRC 2. the role of EED on PRC2 is embodied in two aspects, 1) EED binds directly to trimethylated H3K27Me3, which allows the localization of the PCR2 complex to the chromatin to be modified, 2) EED has a large allosteric promoting effect on the enzymatic function of EZH 2. therefore, the development of target compounds as allosteric EED provides a new strategy for inhibiting the enzymatic activity of EZH2, and such inhibitors have advantages over inhibitors of the enzymatic site of EZH2, such as when a patient is resistant to the enzyme inhibitor of EZH2, EED inhibitors can also act to inhibit the enzymatic activity of EZH 2.
The biological function of the disclosed compounds is demonstrated in biochemical and cellular level assays. For example, in biochemical assays, the compounds disclosed herein are capable of strong competitive binding (IC) to the H3K27Me3 polypeptide bound to EED proteins50Can reach<10nM) at the cellular level, the compounds disclosed herein can inhibit not only the methylation level of histone H3K27 but also the proliferation of cancer cells by this effect.
Evaluation of the Effect of Compounds in blocking the binding of EED to H3K27me3 by the AlphaScreen (a-Screen) method
First, compound solutions were prepared in different concentration gradients, compound powders were dissolved in DMSO to give a stock solution, 1.5. mu.l of compound stock solution was mixed with 198.5. mu.l of reaction buffer (25mM HEPES (pH 8.0), 50mM NaCl, 0.015% Tween 20, 0.5% BSA) and diluted with 3-fold gradients in the above buffer containing 0.75% DMSO, setting 9 different test concentrations with compounds, 5. mu.l of different concentration gradients were applied to ProxiPlate-384Plus, White assay plate (PerkinElmer, 6008280), each concentration gradient set in 2 replicates.
And then a binding blocking reaction is performed. The full-length EED protein with His6 tag (amino acids 441) was diluted to 60nM and the biotin-tagged polypeptide fragment H3K27me3 (amino acids 19-33) (Biotinylated-H3K27me3) to 75nM with the above buffer. Mu.l of the 75nM polypeptide fragment and 5. mu.l of 60nM protein were transferred to each test well containing the compound, the test plate was sealed with a membrane, and incubated for 30 minutes at room temperature.
And finally, detecting by using an AlphaScreen method. Just before use, the nickel chelate acceptor beads and streptavidin donor beads were mixed in the above reaction buffer at a ratio of 1:1 (Perkin Elmer, product No. 6760619M), and 5. mu.l of the above premixed test solution was added to each well, with a final concentration of 5. mu.g/mL of both donor and acceptor beads. The plate was sealed with tin foil and left at room temperature in the dark for 1 hour. The signal was read using the AlphaScreen detector on Spectra max i 3. AlphaScreen signals were normalized to the readings obtained for the positive control (maximum signal control) and the negative control (minimum signal control) to give inhibition for different concentrations of compound, after which non-linear regression analysis was performed using GraphPad Prism 5, and inhibition curves were developed by the Y ═ Bottom + (Top-Bottom)/(1+10^ ((LogIC50-X) × HillSlope)) dose response equation, giving IC50 values for each compound.
To exclude false positives due to interference of the compound with the AlphaScreen detection System, the compound was diluted in the same way, and the biotin-labeled polypeptide Biotinylated- (His) was used6Instead of EED and the polypeptide H3K27me3 in the test system, after an equivalent incubation time, the signal values were read on Spectra max i 3. The data was processed in the same way.
Results
The following table shows the IC of some of the compounds of the invention50The value is obtained.
Letter A stands for IC50Less than 100 nM;
letter B stands for IC50100nM to 1000 nM;
letter C stands for IC50Is more than 1000nM
Figure BDA0001732766820000661
Figure BDA0001732766820000671
Figure BDA0001732766820000681
ELISA (H3K27 trimethylation) analysis
Representative compounds of the disclosure were diluted in DMSO in 3-fold gradients, with 10 concentration gradients detected per compound, with the highest assay concentration being 10 μ M. Compound was diluted 200-fold into G401 cells cultured in 96-well plates (final DMSO concentration 0.5%). The ELISA method detects the trimethylation level of the histone H3K27 after 72 hours of culture of the administration cells.
Histone extraction Compound-treated cells in 96-well plates were treated with 1 XPPBS (10 XPPBS buffer (80g NaCl (Sigma, product No. S3014),2gKCl (Sigma, product No. 60128),14.4g Na2HP04(Sigma, product number S5136),2.4gKH2P04(Sigma, product No. P9791) was washed three times in 1L water, pH to 7.4), 100. mu. L0.4N HCl was added to each well, and the cells were lysed by placing at 4 ℃ with gentle shaking for 2 hours. The cell lysate was then neutralized with 80. mu.L of neutralization buffer (0.5M disodium hydrogen phosphate, pH 12.5,2.5mM DTT; 1% cocktail (Sigma, product No. P8340) (cell lysate was mixed well with neutralization buffer).
ELISA detection method comprises transferring Cell lysate in parallel to 2 384-well assay plates (PerkinElmer, OptiPlate-384HB, product No. 6007290), plates for detecting the level of H3K27 trimethylation, plates for determining the level of H3, PBS adjusted to a final volume of 50. mu.L/well, coating overnight at 4 deg.C, discarding the well solutions, washing 5 times with TBST buffer (L xTBS (10x TBS:24.2g Tris (Sigma, product No. T6066),80g NaCl (Sigma, product No. S3014) to 1L water, HCl adjusted to pH 7.6), 0.1% Tween-20), draining water on absorbent paper, adding 70. mu.L of blocking buffer (TBST, 5% blocking buffer) to the coated wells, discarding 1 hour at room temperature, adding blocking buffer, adding 2-grade antibody (30. mu.L/well), blocking buffer 23-grade blocking antibody (TBST, 5% blocking buffer), diluting with 200. mu.7. mu.1. mu.L of blocking buffer, diluting with 200. mu.7. mu.20. mu.L of the sample, 2000. mu.L of the primary antibody, diluting with 200. mu.20 of primary antibody, adding the primary antibody, diluting with primary antibody, diluting the secondary antibody (TBS) to the secondary antibody, diluting with TBS + 2000. medium of the secondary antibody, diluting with the secondary antibody, diluting the secondary antibody (TBS + 2000) after dilution of the primary antibody), diluting the primary antibody, diluting the secondary antibody after adding the secondary antibody after diluting the primary antibody, diluting with the secondary antibody, diluting the secondary antibody after diluting the secondary antibody, diluting buffer after diluting the secondary antibody (TBS 2-wash medium after diluting the secondary antibody, diluting with the primary antibody, diluting the secondary antibody after diluting the secondary antibodyPercent inhibition of substance. Data were fit to dose response curves using the GraphPad prisim5 program to obtain the IC of the test compound50The value is obtained. Results
The following table shows the IC of some of the compounds of the invention50The value is obtained.
Letter A stands for IC50Less than 100 nM;
letter B stands for IC50100nM to 1000 nM;
letter C stands for IC50Is more than 1000nM
Figure BDA0001732766820000701
Figure BDA0001732766820000711
Cell proliferation assay
Human B-cell non-Hodgkin lymphoma cells, KARPAS-422S, were cultured in culture flasks using standard cell culture conditions. The medium was 15% fetal bovine serum (FBS, Invitrogen, product No. 10099-141), 1% penicillin/streptomycin solution (P/S) RPMI-1640(Invitrogen, product No. 11875), and the flasks were cultured in a sterile incubator at 37 ℃ and 95% relative humidity, 5% CO 2. To examine the effect of PRC2 inhibitors on cell proliferation, cells in exponential growth phase were taken at 1X104The density of cells/well was seeded into 96-well plates (Corning, product No. 3904) and 100 μ L of medium was added per well. Subsequently, different concentrations of compounds disclosed herein were added to the seeded wells (9 concentration gradients were set for each compound, with the highest assay concentration being 10 μ M, 3-fold gradient dilutions), 2 replicates were set for each treatment concentration, and a final DMSO concentration of 0.5%. Then, the number of viable cells was measured every 3 to 4 days by using Vi-CELL (Beckman Coulter). The cells counted each time were in equal density (1X 10)4Individual cells/well) were seeded into new 96-well plates, supplemented with fresh medium to 100 μ L, while adding different concentrations of compounds. After culturing to day 13, 100. mu.L of CellTiter-glo (CTG) (Promega, product No. G7573) was added to each well, and the mixture was left to stand at room temperature in the dark for 10 days20 minutes, the luminescence signal was read using Molecular Devices, SpectraMaxi 3X. The IC of the test compound was obtained by fitting the data to a dose response curve using GraphPad prism 550The value is obtained.
Results
The following table shows the IC of some of the compounds of the invention50The value is obtained.
Letter A stands for IC50Less than 100 nM;
letter B stands for IC50100nM to 1000 nM;
letter C stands for IC50Is more than 1000nM
The disclosed compounds are useful for treating cancers associated with the mechanism of action of the EED protein and/or PRC2 protein complex, including but not limited to, lymphomas including diffuse large B-cell lymphoma, follicular lymphoma and the like, leukemia, multiple myeloma, mesothelioma, gastric cancer, malignant rhabdoid tumor, liver cancer, prostate cancer, breast cancer, brain tumors including neuroblastoma, glioma, glioblastoma and astrocytoma, cervical cancer, colon cancer, melanoma, endometrial cancer, esophageal cancer, head and neck cancer, lung cancer, nasopharyngeal cancer, ovarian cancer, pancreatic cancer, renal cancer, rectal cancer, thyroid cancer, parathyroid tumor, uterine tumor, and soft tissue sarcoma, and the like.

Claims (16)

1, pyrimidine or pyrazine five-membered heterocyclic compounds shown in the general formula (I), pharmaceutically acceptable salts, hydrates, prodrugs, stereoisomers or solvates thereof,
wherein, X1Independently is C or N;
X2independently is C or N;
X3independently is C or N;
X4independently C, N, S;
z is independently C or O;
wherein
Figure FDA0001732766810000012
Is a single or double bond;
m is independently 0 or 1;
n is independently 0 or 1;
p is independently 0 or 1;
R1independently hydrogen or halogen;
R2independently of one another hydrogen, halogen, cyano, C1-8Alkyl radical, C1-8Haloalkyl, R2aSubstituted or unsubstituted C3-8Cycloalkyl radical, R2bSubstituted or unsubstituted C3-8Heterocyclic radical, R2cSubstituted or unsubstituted alkenyl, R2bSubstituted or unsubstituted C5-8Cycloalkenyl radical, R2bSubstituted or unsubstituted C5-8Heterocycloalkenyl, R2dSubstituted or unsubstituted amino, R2dSubstituted or unsubstituted oxy, R2dSubstituted or unsubstituted amide, 0-3R2eSubstituted C6-10Aryl or 0-3R2eSubstituted radicals containing C1-20Carbon atom and 1-4N, NR2e1O or S (O)0-2Heteroaromatic rings of hetero atoms;
R2aand R2bIndependently amino, amino protected by a protecting group, monofluorine or polyfluoro;
R2cindependently is C1-4Alkyl, ester group;
R2dindependently is C1-20Alkyl, benzyl, substituted or unsubstituted C6-10Aryl, substituted or unsubstituted, containing C1-20Carbon atom and 1-4N, NR2e1O or S (O)0-2A heteroaromatic ring of heteroatoms;
R2eindependently is halogen, -C (═ O) NR2e1R2e2、-S(=O)2R2e3、N R2e1R2e2Containing C1-20Carbon atom and 1-4 NR2e1O or S (O)0-2A heterocyclic ring of a heteroatom; wherein R is2e1Independently is hydrogen or C1-4An alkyl group; r2e2Independently is hydrogen or C1-4An alkyl group; r2e3Is C1-4An alkyl group.
2. The pyrimidone compound of claim 1, or a pharmaceutically acceptable salt, hydrate, prodrug, stereoisomer, or solvate thereof, wherein:
when Z is not present, the compound is,
Figure FDA0001732766810000021
is a single or double bond; m and n are 0; p is 1; or
When the Z is C, the compound has the structure of,
Figure FDA0001732766810000022
is a single bond; m is 1; n is 0; p is 1;
when the Z is O, the reaction is carried out,
Figure FDA0001732766810000023
is a single bond, m is 1, and n and p are of 0 and of 1.
3. The pyrimidone compound, the pharmaceutically acceptable salt, the hydrate, the prodrug, the stereoisomer, or the solvate thereof, according to claim 1, wherein:
Figure FDA0001732766810000024
when X is present1When is C, X2Is N, X3Is C, X4Is N; or
When X is present1Is N, X2When is N, X3Is C, X4Is C; or
When X is present1Is N, X2When is C, X3Is N, X4Is C; or
When X is present1Is N, X2When is C, X3Is C, X4Is S.
4. The pyrimidine or pyrazino five-membered heterocyclic compound according to claim 1, a pharmaceutically acceptable salt thereof, a hydrate, a prodrug, a stereoisomer, or a solvate thereof, wherein
R1Is H or F.
5. The pyrimidine or pyrazino five-membered heterocyclic compound according to claim 1, a pharmaceutically acceptable salt thereof, a hydrate, a prodrug, a stereoisomer, or a solvate thereof, wherein R is2Is halogen, cyano, C1-8Alkyl radical, C1-8Haloalkyl or a structure selected from:
Figure FDA0001732766810000031
wherein j is 0, 1,2 or 3, k is 0, 1,2,3 or 4, l is 0, 1 or 2, V is C, N or O, and at most two N or O are simultaneously present in the same ring , and ring A contains 1-3 heteroatoms of substituted or unsubstituted C5-10A heteroaryl group; ring B is selected from substituted or unsubstituted C containing 1-4 heteroatoms5-10Heteroaryl, wherein the heteroatom is selected from N, O or S; q is N, O or S.
6. The pyrimidine or pyrazino five-membered heterocyclic compound according to claim 1, a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer, or a solvate thereof, wherein said general formula (I) is selected from any of the following compounds:
Figure FDA0001732766810000041
wherein m, n, X1、X2、X3、X4、R1、R2And
Figure FDA0001732766810000043
is as defined in claim 1.
7. The pyrimidine or pyrazino five-membered heterocyclic compound of the general formula (I), which is selected from the group consisting of any compounds below, a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer, or a solvate thereof, according to claim 1:
Figure FDA0001732766810000042
Figure FDA0001732766810000051
or a pharmaceutically acceptable salt thereof.
8, isotopically-labeled compounds of pyrimidine or pyrazino five-membered heterocyclic compound represented by the general formula (I), or a pharmaceutically acceptable salt, hydrate, prodrug, stereoisomer or solvate thereof, according to any one of claims 1 to 7, wherein the isotope is selected from the group consisting of2H、3H、11C、13C、14C、15N、18F、31P、32P、35S、36Cl and125I。
9, A preparation method of pyrimidine or pyrazine five-membered heterocyclic compound shown in formula (I), which comprises the following steps:
the halogenated intermediate compound A is subjected to coupling reaction to obtain a formula (I), and the reaction equation is as follows:
Figure FDA0001732766810000062
wherein W represents halogen, preferably Br; r1、R2、X1、X2、X3、X4Z, m, n, p and
Figure FDA0001732766810000063
is as defined in claim 1.
A process for the preparation of intermediate compounds a comprising the steps of:
the chloro pyrimidine or pyrazine ring intermediate B is substituted by intermediate amine C under alkaline condition to obtain halogenated pyrimidine or pyrazine ring intermediate A, and the reaction equation is as follows:
wherein R is1、W、X1、X2、X3、X4Z, m, n, p and
Figure FDA0001732766810000074
is as defined in claim 9.
11, intermediates for the preparation of a pyrimidine or pyrazino five-membered heterocyclic compound represented by the general formula (I) according to any of claims 1 to 7, selected from the group consisting of:
Figure FDA0001732766810000072
wherein R is1、W、X1、X2、X3、X4Z, m, n, p and
Figure FDA0001732766810000073
is as defined in claim 9.
Use of pyrimidine or pyrazino five-membered heterocyclic compounds according to any of claims 1 to 7, a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer or a solvate thereof, or an isotopically labeled compound according to claim 8, for the preparation of a medicament for the prevention and/or treatment of cancer associated with the mechanism of action of the EED protein and/or the PRC2 protein complex.
the use according to claim 12, wherein said cancer is selected from diffuse large B-cell lymphoma, follicular lymphoma.
14, the use according to claim 12, wherein the cancer is selected from the group consisting of lymphoma, leukemia, multiple myeloma, mesothelioma, gastric cancer, malignant rhabdoid tumor, liver cancer, prostate cancer, breast cancer, brain tumor, cervical cancer, colon cancer, melanoma, endometrial cancer, esophageal cancer, head and neck cancer, lung cancer, nasopharyngeal cancer, ovarian cancer, pancreatic cancer, renal cancer, rectal cancer, thyroid cancer, parathyroid tumor, uterine tumor and soft tissue sarcoma.
15, pharmaceutical composition comprising the pyrimidine or pyrazino five-membered heterocyclic compound according to any of claims 1-7, a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer, or a solvate thereof, or the isotopically labeled compound of claim 8, and a pharmaceutically acceptable excipient.
16, A product of the pyrimidine or pyrazino five-membered heterocyclic compound according to any of claims 1 to 7, a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer or a solvate thereof, or an isotopically labeled compound according to claim 8, in combination with another drug selected from the group consisting of an anticancer drug, a tumor immune drug, an antiallergic drug, an antiemetic drug, an analgesic drug, and a cytoprotective drug.
CN201810776810.4A 2018-07-19 2018-07-19 Pyrimidine or pyrazine ring compounds and application thereof Pending CN110734436A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201810776810.4A CN110734436A (en) 2018-07-19 2018-07-19 Pyrimidine or pyrazine ring compounds and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201810776810.4A CN110734436A (en) 2018-07-19 2018-07-19 Pyrimidine or pyrazine ring compounds and application thereof

Publications (1)

Publication Number Publication Date
CN110734436A true CN110734436A (en) 2020-01-31

Family

ID=69234231

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201810776810.4A Pending CN110734436A (en) 2018-07-19 2018-07-19 Pyrimidine or pyrazine ring compounds and application thereof

Country Status (1)

Country Link
CN (1) CN110734436A (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021032004A1 (en) * 2019-08-22 2021-02-25 上海青煜医药科技有限公司 Azaheteroaryl compound and application thereof
CN113636993A (en) * 2020-05-11 2021-11-12 苏州亚盛药业有限公司 Process for producing (5-fluoro-2, 3-dihydrobenzofuran-4-yl) methylamine or salt thereof, and intermediate therefor

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016103155A1 (en) * 2014-12-23 2016-06-30 Novartis Ag Triazolopyrimidine compounds and uses thereof
CN109790166A (en) * 2016-06-20 2019-05-21 诺华股份有限公司 Imidazopyridine is used for treating cancer
CN109906224A (en) * 2016-06-20 2019-06-18 诺华股份有限公司 Triazole pyridine compounds and its application

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016103155A1 (en) * 2014-12-23 2016-06-30 Novartis Ag Triazolopyrimidine compounds and uses thereof
CN107108637A (en) * 2014-12-23 2017-08-29 诺华股份有限公司 Triazolopyrimidine compound and application thereof
CN109790166A (en) * 2016-06-20 2019-05-21 诺华股份有限公司 Imidazopyridine is used for treating cancer
CN109906224A (en) * 2016-06-20 2019-06-18 诺华股份有限公司 Triazole pyridine compounds and its application

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021032004A1 (en) * 2019-08-22 2021-02-25 上海青煜医药科技有限公司 Azaheteroaryl compound and application thereof
CN112409385A (en) * 2019-08-22 2021-02-26 上海青煜医药科技有限公司 Azaaryl compounds and uses thereof
CN113636993A (en) * 2020-05-11 2021-11-12 苏州亚盛药业有限公司 Process for producing (5-fluoro-2, 3-dihydrobenzofuran-4-yl) methylamine or salt thereof, and intermediate therefor
WO2021228034A1 (en) * 2020-05-11 2021-11-18 Ascentage Pharma (Suzhou) Co., Ltd. Preparation method of (5-fluoro-2, 3-dihydrobenzofuran-4-yl) methanamine or its salt, and intermediates thereof

Similar Documents

Publication Publication Date Title
CN111153901B (en) Nitrogen-containing fused heterocyclic SHP2 inhibitor compound, preparation method and application
CN110156786B (en) Pyrimido-cyclic compounds, process for their preparation and their use
TWI704151B (en) Erk inhibitors
CN106928219B (en) Nitrogen-containing fused heterocyclic compound, preparation method, intermediate, composition and application
CN110563722A (en) pyridine or pyridazine ring compound and application thereof
CN112142735A (en) Condensed cyanopyridine compound, preparation method and application
TW201538494A (en) 2,4-disubstituted phenylene-1,5-diamine derivatives and applications thereof, and pharmaceutical compositions and pharmaceutically acceptable compositions prepared therefrom
WO2012098068A1 (en) Pyrazolo pyrimidines as dyrk1a and dyrk1b inhibitors
CN113135910A (en) Pyrimidine-4 (3H) -ketone heterocyclic compound, preparation method and pharmaceutical application thereof
WO2009010488A1 (en) Heterocyclic compounds useful as mk2 inhibitors
CN113061132B (en) Condensed ring lactam compound, preparation method and application
CN111518100A (en) Cyclopropenoarylbenzofuran substituted nitrogen heteroaryl compound and application thereof
WO2019120276A1 (en) Pyrimidone compound and application thereof
CN114685487A (en) Pyrimidine heterocyclic compound, preparation method and application
JP2015508775A5 (en)
JP2022523477A (en) A pharmaceutical composition for preventing or treating a pyrrolopyrimidine derivative and a protein kinase-related disease containing the derivative as an active ingredient.
TW202204351A (en) Compounds having a macrocyclic structure and uses thereof
CN110655520A (en) Pyrimido-cyclic compounds, process for their preparation and their use
CN110734436A (en) Pyrimidine or pyrazine ring compounds and application thereof
CN114805311A (en) Spirocyclic indenes
CN114437116A (en) Heterocyclic compound and preparation method, pharmaceutical composition and application thereof
CN112300173B (en) Nitrogen-containing polycyclic compounds, preparation method and application
JP2024516194A (en) Compounds as PD1/PD-L1 inhibitors and methods thereof
CN114907350A (en) Nitrogen-containing condensed ring compounds, preparation method and application
CN108727368B (en) Nitrogen-containing fused heterocyclic compound, preparation method, intermediate, composition 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
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20200131

WD01 Invention patent application deemed withdrawn after publication