CN111057048A - Aminopyrazine/pyridine compound, preparation method and application - Google Patents

Aminopyrazine/pyridine compound, preparation method and application Download PDF

Info

Publication number
CN111057048A
CN111057048A CN202010067960.5A CN202010067960A CN111057048A CN 111057048 A CN111057048 A CN 111057048A CN 202010067960 A CN202010067960 A CN 202010067960A CN 111057048 A CN111057048 A CN 111057048A
Authority
CN
China
Prior art keywords
membered
group
alkyl
compound
cancer
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.)
Granted
Application number
CN202010067960.5A
Other languages
Chinese (zh)
Other versions
CN111057048B (en
Inventor
万惠新
潘建峰
马金贵
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.)
Rudong Lingda Biomedical Technology Co ltd
Original Assignee
Rudong Lingda Biomedical 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 Rudong Lingda Biomedical Technology Co ltd filed Critical Rudong Lingda Biomedical Technology Co ltd
Publication of CN111057048A publication Critical patent/CN111057048A/en
Application granted granted Critical
Publication of CN111057048B publication Critical patent/CN111057048B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/14Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing three or more hetero rings
    • 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
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers

Abstract

The invention discloses an aminopyrazine/pyridine compound, a preparation method and application thereof, in particular to a pyrazine/pyridine amine compound shown as a general formula I, or pharmaceutically acceptable salt thereof, or enantiomer, diastereoisomer, tautomer, solvate, polymorph or prodrug thereof, a preparation method and pharmaceutical application thereof, wherein the definition of each group is described in the specification.

Description

Aminopyrazine/pyridine compound, preparation method and application
Technical Field
The invention belongs to the field of pharmaceutical chemistry, and particularly relates to aminopyrazine/pyridine Ras-Raf-MEK-ERK signal pathway inhibitor compounds with antitumor activity, a preparation method and application.
Background
Extracellular signal-regulated kinases (ERKs) are a class of serine/threonine protein kinases found in the 90's of the 20 th century, and are one of the important subfamilies of the mitogen-activated protein kinase MAPKs family. Activated ERK can transmit extracellular signals to the nucleus, promote phosphorylation of cytoplasmic target proteins or regulate the activity of other protein kinases, thereby regulating gene expression. Ras-Raf-MEK-ERK signaling is central to the signaling network involved in regulating cell growth, development and differentiation, and therefore ERK has a variety of biological effects in regulating cell proliferation, differentiation, migration, invasion and apoptosis.
The Ras/Raf/MEK/ERK pathway is a main signal pathway related to the ERK function, becomes a hot spot for the development of cancer-targeted drugs, and in recent years, a plurality of drugs developed aiming at node proteins on the signal pathway are successfully marketed. For example, specific B-Raf inhibitors Vemurafenib and dabrafenib, which are used for treating B-RafV600E mutant non-small cell lung cancer, were marketed in 2011 and 2013, respectively, for the treatment of melanoma, which received FDA-breakthrough drug eligibility. The MEK1/2 inhibitor trametinib was also marketed in 2013 for the treatment of melanoma. However, inhibition of these upstream pathway nodes has its limitations, tumors can rapidly develop resistance to B-Raf and MEK inhibitors, and Ras protein mutations are also found in numerous tumors, such as colorectal, pancreatic, lung, etc. The above mechanisms of drug resistance generation include point mutations, changes in protein multimeric forms, changes in protein peptide chain length, and the like, which are extremely challenging for the development of therapeutic drugs for next generation Ras-Raf-MEK resistance. However, the ERK is used as a downstream key node of the pathway, so that the occurrence of drug-resistant mutation is not discovered at present, and the ERK targeting drug can greatly improve the treatment of patients with drug resistance to upstream target inhibitors, so that the ERK targeting drug is a promising anti-cancer drug research and development field.
Although a number of ERK inhibitors have been introduced into clinical studies in the early days, such as GDC0994, SCH772984, etc., these compounds have either been too toxic or poorly druggable to terminate clinical studies. Therefore, the discovery and search of novel ERK inhibitor compounds with high selectivity, high activity and high druggability become a major hot spot at present.
Disclosure of Invention
One of the technical problems to be solved by the invention is to provide a novel specific ERK kinase inhibitor for preparing a tumor treatment medicament.
The scheme for solving the technical problems is as follows:
an aminopyrazine/pyridine compound having a general formula I, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, solvate, polymorph or prodrug thereof,
Figure BDA0002376519750000021
in the formula:
r1 is independently selected from hydrogen, halogen, C1-C6 alkyl, C1-C6 alkoxy, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl;
r2 is independently selected from hydrogen, halogen, cyano, carbonyl, ester, amide, sulfonamide, sulfone, sulfoxide, sulfinyl, amino or substituted amino, C1-C10 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C1-C10 alkoxy, 3-10 membered saturated or partially unsaturated cycloalkyl or heterocycloalkyl, 5-10 membered aryl or heteroaryl;
m is independently selected from N, CR; r is independently selected from hydrogen, halogen, C1-C6 alkyl, C1-C6 alkoxy, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl;
r3 is independently selected from hydrogen, halogen, hydroxy, cyano, carbonyl, C1-C5 alkyl, C1-C5 alkoxy, amino, C1-C5 alkylamino;
l is independently selected from the group consisting of a bond, acyl, sulfonyl, alkylene of C1-C4, alkenylene of C2-C4, alkynylene of C2-C4, 3-8 membered cycloalkyl, 4-8 membered heterocycloalkyl;
r4 is independently selected from C1-C6 alkyl, 3-8 membered cycloalkyl, 4-8 membered saturated or partially unsaturated heterocycloalkyl, 5-8 membered aryl or heteroaryl; and the above R4 may be substituted with 1 to 3 of any of the following groups: deuterium, halogen, hydroxyl, amino, cyano, sulfone, sulfoxide, ester, amide, C1-C4 alkyl, C1-C4 alkoxy, C1-C4 thio, 3-8 membered cycloalkyl, 4-8 membered heterocycloalkyl;
r5, R6 are independently selected from hydrogen, halogen, C1-C6 alkyl, C1-C6 alkoxy, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl; or a 3-6 membered saturated or partially unsaturated carbocyclic or heterocyclic ring is formed between R5 and R6 through a carbon chain or heteroatom;
ar is independently selected from a 5-10 membered aromatic ring or aromatic heterocycle;
z is independently selected from hydrogen, oxygen;
m and n are independently selected from 0, 1 and 2;
one or more hydrogen atoms on any of the above groups may be substituted with a substituent selected from the group consisting of: including but not limited to deuterium, halogen, hydroxyl, amino or substituted amino, cyano, sulfone or sulfoxide, urea, sulfonylurea, C1-C8 alkyl, 3-8 membered cycloalkyl, C1-C8 alkoxy, C1-C8 alkylamino or cycloalkylamino, alkenyl, alkynyl, acyl or sulfonyl or sulfinyl, 5-to 8-membered aryl or heteroaryl, 4-to 8-membered cycloalkyl or heterocycloalkyl; wherein said heteroaryl group contains 1 to 3 heteroatoms selected from the group consisting of: n, O, P or S, the heterocycloalkyl group containing 1 to 3 heteroatoms selected from the group consisting of: n, O, P or S, including but not limited to saturated or partially unsaturated or unsaturated monocyclic, fused, bridged, spiro, and the like ring systems.
In a further embodiment, a compound having the general formula (I), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, solvate, polymorph, or prodrug thereof, characterized by:
Figure BDA0002376519750000031
r1 is preferably selected from hydrogen, deuterium, halogen, C1-C6 alkyl;
r2 is preferably selected from hydrogen, deuterium, C1-C8 alkyl, C1-C8 alkoxy, C1-C8 alkylamino, saturated or partially unsaturated 3-to 8-membered cycloalkyl, 3-to 8-membered cycloalkyloxy, 4-to 8-membered cycloalkylamino, 4-to 8-membered heterocycloalkylamino, saturated or partially unsaturated 3-to 8-membered heterocycloalkyl, 3-to 8-membered heterocycloalkoxy, 5-to 10-membered arylamine or heteroarylamine, 5-to 10-membered aryl or 5-to 10-membered heteroaryl; further preferably selected from: substituted or substituted cyclohexane, cyclohexenyl, tetrahydropyranyl, dihydropyranyl, piperidinyl, morpholinyl, piperazinyl, pyrazolyl, imidazolyl, isoxazolyl, phenyl, pyridinyl, isopropyl, cyclopentyl, cyclopropyl, oxetanyl, azetidinyl, thiocyclohexyl and the like; and the above groups may be substituted with one or more substituents preferably selected from the group consisting of: halogen, deuterium, cyano, hydroxy, amino, C1-C3 alkyl, C1-C3 alkylhydroxy, C1-C3 alkoxy, C1-C3 alkylamino, C1-C3 haloalkyl, C1-C3 haloalkoxy, oxo, acyl, sulfonyl, sulfone, etc.;
m, M1, M2, M3 are preferably from N, CR 3; m4, M5 are preferably selected from N, CR3, O, S, etc.; wherein R3 is preferably selected from H, F, methyl, CN;
m is preferably from 0 to 2;
l is preferably selected from acyl, sulfonyl, substituted or unsubstituted alkylene; and one or more hydrogens on the above alkylene group may be substituted with a substituent preferably selected from the group consisting of: deuterium, fluorine, hydroxyl, methyl, ethyl, cyclopropyl, alkoxy of C1-C2, alkylamino of C1-C2, alkylhydroxyl of C1-C2, alkoxyalkyl of C1-C2, alkylaminoalkyl of C1-C2, etc.;
r4 is preferably selected from phenyl or pyridyl, and the above groups may be substituted with one or more substituents selected from the group consisting of: halogen, cyano, C1-C4 alkyl, C1-C4 hydroxyalkyl, C1-C4 aminoalkyl, C1-C4 alkoxy, C1-C4 haloalkyl, C1-C4 haloalkoxy, C1-C4 sulfone or sulfoxide, ester, amide, sulfonamide, etc.;
r5 and R6 are preferably selected from hydrogen, fluorine, methyl, hydroxymethyl, methoxymethyl and the like.
In a further preferred mode, the compound having the general formula (1), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, solvate, polymorph or prodrug thereof, is preferably as follows in general formula (III):
Figure BDA0002376519750000041
wherein R7 is preferably selected from hydrogen, deuterium, fluoro, methyl, ethyl, hydroxymethyl, methoxymethyl, aminomethyl, methylaminomethyl, and the like, and R1, R2, R3, R4, R5, R6, M, M1, M2, M3, M4, M, and the like are as defined above.
A process for preparing a compound of formula I, said process comprising steps a-c:
a) carrying out coupling reaction on the compound with the intermediate general formula (A1) or (A2) and a raw material of dibromoaminopyrazine or dibromoaminopyridine under the catalysis of transition metal to convert the compound into the compound with the intermediate general formula (B1) or (B2); and
b) carrying out cross-coupling reaction on the intermediate compound with the general formula (B1) or (B2) and a raw material R2-FG1 under the reaction condition of the presence of a transition metal catalyst to obtain a compound with a general formula (I) or an intermediate compound with a general formula (C);
c) reacting the compound in the general formula (C) with a raw material compound R4L-FG2 under the condition of base catalysis to generate a compound in the general formula (I);
Figure BDA0002376519750000042
in the formulae, FG1 represents boric acid, boric acid ester, organic tin, organic zinc and other groups, FG2 represents carboxylic acid, acyl chloride, sulfonyl chloride and other groups, and the definition of each other group is as described above;
preferably, said steps a), b), c) are each carried out in a solvent, and said solvent is selected from the group consisting of: water, methanol, ethanol, isopropanol, butanol, ethylene glycol methyl ether, N-methyl pyrrolidone, dimethyl sulfoxide, tetrahydrofuran, toluene, dichloromethane, 1, 2-dichloroethane, acetonitrile, N-dimethylformamide, N-dimethylacetamide, dioxane, or a combination thereof.
Preferably, the transition metal catalyst is selected from the group consisting of: tris (dibenzylideneacetone) dipalladium (Pd)2(dba)3) Tetrakis (triphenylphosphine) palladium (Pd (PPh)3)4) Palladium acetate, palladium chloride, dichlorobis (triphenylphosphine) palladium, palladium trifluoroacetate, triphenylphosphine palladium acetate, [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride, bis (tri-o-phenylphosphino) palladium dichloride, 1, 2-bis (diphenylphosphino) ethane palladium dichloride, or a combination thereof; the catalyst ligand is selected from the group consisting of: tri-tert-butylphosphine, tri-tert-butylphosphine tetrafluoroborate, tri-n-butylphosphine, triphenylphosphine, tri-p-benzylphosphine, tricyclohexylphosphine, tri-o-phenylphosphine, or a combination thereof.
Preferably, the inorganic base is selected from the group consisting of: sodium hydride, potassium hydroxide, sodium acetate, potassium tert-butoxide, sodium tert-butoxide, potassium fluoride, cesium fluoride, potassium phosphate, potassium carbonate, potassium bicarbonate, sodium carbonate, sodium bicarbonate, or combinations thereof; the organic base is selected from the group consisting of: pyridine, triethylamine, N, N-diisopropylethylamine, 1, 8-diazabicyclo [5.4.0] undec-7-ene (DBU), lithium hexamethyldisilazide, sodium hexamethyldisilazide, lutidine, or a combination thereof.
The invention provides a class of preferred compounds of formula (I) including, but not limited to, the following structures:
Figure BDA0002376519750000051
Figure BDA0002376519750000061
Figure BDA0002376519750000071
the invention also aims to provide a medicament for treating or preventing tumors and a composition thereof. The technical scheme for realizing the purpose is as follows:
a pharmaceutical composition for treating tumor comprises aminopyrazine/pyridine compounds shown in the general formula (I), or pharmaceutically acceptable salts thereof, or enantiomers, diastereomers, tautomers, solvates, polymorphs or prodrugs thereof, and pharmaceutically acceptable carriers.
Another object of the present invention is to provide a use of the above compound. The technical scheme for realizing the purpose is as follows:
application of the aminopyrazine/pyridine compound shown in the general formula (I) or pharmaceutically acceptable salt thereof, or enantiomer, diastereoisomer, tautomer, solvate, polymorph or prodrug thereof in preparation of drugs for preventing or treating tumors.
The tumor includes but is not limited to any one of non-small cell lung cancer, melanoma, lung adenocarcinoma, lung squamous carcinoma, breast cancer, prostate cancer, liver cancer, pancreatic cancer, skin cancer, stomach cancer, intestinal cancer, bile duct cancer, brain cancer, leukemia, lymph cancer, nasopharyngeal carcinoma, etc.
The invention relates to a compound with the structural characteristics of a general formula (I), which can inhibit a plurality of tumor cells, particularly can efficiently kill tumors related to Ras-Raf-MEK-ERK signal channel abnormity, and is a therapeutic drug with a brand-new action mechanism.
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) may be combined with each other to form new or preferred embodiments. The space is not described herein in a repeated fashion.
Detailed Description
The inventor has made long-term and intensive studies to prepare a compound with a novel structure shown in formula I and found that the compound has better ERK kinase inhibitory activity, and the compound has specific inhibitory action on ERK kinase at very low concentration (which can be as low as less than or equal to 10nmol/L), and has excellent cell proliferation inhibitory activity on Ras-Raf-MEK-ERK, so that the compound can be used for treating related diseases such as tumors caused by Ras-Raf-MEK-ERK kinase mutation or abnormal expression amount. Based on the above findings, the inventors have completed the present invention.
Term(s) for
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 the claimed subject matter belongs. All patents, patent applications, and publications cited herein are incorporated by reference in their entirety unless otherwise indicated.
It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the subject matter claimed. In this application, the use of the singular also includes the plural unless specifically stated otherwise. It must be noted that, as used in this specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. It should also be noted that the use of "or", "or" means "and/or" unless stated otherwise. Furthermore, the term "comprising" as well as other forms, such as "includes," "including," and "containing," are not limiting.
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, -CH 2O-is equivalent to-OCH 2-.
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. For example, C1-6 alkyl refers to an alkyl group as defined below having a total of 1 to 6 carbon atoms. 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" means fluorine, chlorine, bromine or iodine; "hydroxy" means an-OH group; "hydroxyalkyl" refers to an alkyl group as defined below substituted with a hydroxyl (-OH) group; "carbonyl" refers to a-C (═ O) -group; "nitro" means-NO2(ii) a "cyano" means-CN; "amino" means-NH2(ii) a "substituted amino" means an alkyl, alkylcarbonyl, aralkyl, heteroaryl group, as defined below, substituted with one or twoAlkyl-substituted amino groups, for example, monoalkylamino, dialkylamino, alkylamido, aralkylamino, heteroaralkylamino; "carboxyl" means-COOH.
In the present application, the term "alkyl", as a group or as part of another group (e.g. as used in groups such as halogen-substituted alkyl), means a straight or branched hydrocarbon chain group consisting only of carbon and hydrogen atoms, containing no unsaturated bonds, having, for example, from 1 to 12 (preferably from 1 to 8, more preferably from 1 to 6) carbon atoms and being attached to the rest of the molecule by single bonds. Examples of alkyl groups include, but are 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, decyl, and the like.
In the present application, the term "alkenyl" as a group or part of another group means a straight or branched hydrocarbon chain group consisting of only carbon atoms and hydrogen atoms, containing at least one double bond, having, for example, 2 to 14 (preferably 2 to 10, more preferably 2 to 6) carbon atoms, and being connected to the rest of the molecule by a single bond, 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 "alkynyl" as a group or part of another group means a straight or branched hydrocarbon chain group consisting solely of carbon and hydrogen atoms, containing at least one triple bond and optionally one or more double bonds, having for example 2 to 14 (preferably 2 to 10, more preferably 2 to 6) carbon atoms and being connected to the rest of the molecule by single bonds, such as but not limited to ethynyl, prop-1-ynyl, but-1-ynyl, pent-1-en-4-ynyl and the like.
In the present application, the term "cycloalkyl" as a group or part of another group means a stable non-aromatic monocyclic or polycyclic hydrocarbon group consisting of only carbon atoms and hydrogen atoms, which may include fused, bridged or spiro ring systems, having 3 to 15 carbon atoms, preferably having 3 to 10 carbon atoms, more preferably having 3 to 8 carbon atoms, and which is saturated or unsaturated and may be attached to the rest of the molecule by a single bond via any suitable carbon atom. Unless otherwise specifically indicated in the specification, carbon atoms in cycloalkyl groups may be optionally oxidized. Examples of cycloalkyl 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-5H-benzocycloheptenyl, 5,6,7,8,9, 10-hexahydro-benzocyclooctenyl, fluorenyl, bicyclo [2.2.1] heptyl, 7-dimethyl-bicyclo [2.2.1] heptyl, bicyclo [2.2.1] heptenyl, bicyclo [2.2.2] octyl, bicyclo [3.1.1] heptyl, bicyclo [3.2.1] octyl, bicyclo [2.2.2] octenyl, Bicyclo [3.2.1] octenyl, adamantyl, octahydro-4, 7-methylene-1H-indenyl, octahydro-2, 5-methylene-pentalenyl and the like.
In this application, the term "heterocyclyl" as a group or part of another group 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 the specification, a heterocyclic group may be a monocyclic, bicyclic, tricyclic or higher ring system, which may include fused ring systems, bridged ring systems or spiro ring systems; wherein the nitrogen, carbon or sulfur atom in the heterocyclic group may be optionally oxidized; the nitrogen atoms may optionally be quaternized; and the heterocyclic group may be partially or fully saturated. The heterocyclic group may be attached to the rest of the molecule via a carbon atom or a heteroatom and by a single bond. In heterocyclic groups containing fused rings, one or more of the rings may be aryl or heteroaryl as defined below, provided that the point of attachment to the rest of the molecule is a non-aromatic ring atom. For the purposes of the present invention, heterocyclyl is preferably a stable 4-to 11-membered non-aromatic monocyclic, bicyclic, bridged or spiro group containing 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur, more preferably a stable 4-to 8-membered non-aromatic monocyclic, bicyclic, bridged or spiro group containing 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur. Examples of heterocyclyl groups include, but are not limited to: pyrrolidinyl, morpholinyl, piperazinyl, homopiperazinyl, piperidinyl, thiomorpholinyl, 2, 7-diaza-spiro [3.5] nonan-7-yl, 2-oxa-6-aza-spiro [3.3] heptan-6-yl, 2, 5-diaza-bicyclo [2.2.1] heptan-2-yl, azetidinyl, pyranyl, tetrahydropyranyl, thiopyranyl, tetrahydrofuranyl, oxazinyl, dioxolanyl, tetrahydroisoquinolinyl, decahydroisoquinolinyl, imidazolinyl, imidazolidinyl, quinolizinyl, thiazolidinyl, isothiazolidinyl, isoxazolidinyl, indolinyl, octahydroindolyl, octahydroisoindolyl, pyrrolidinyl, pyrazolidinyl, phthalimidyl, and the like.
In this application, the term "aryl" as a group or as part of another group means a conjugated hydrocarbon ring system group having 6 to 18 carbon atoms, preferably having 6 to 10 carbon atoms. For the purposes of the present invention, an aryl group may be a monocyclic, bicyclic, tricyclic or higher polycyclic ring system and may also be fused to a cycloalkyl or heterocyclic group 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 a group or part of another group means a 5-to 16-membered conjugated ring system group having 1 to 15 carbon atoms (preferably having 1 to 10 carbon atoms) and 1 to 6 heteroatoms selected from nitrogen, oxygen and sulfur in the ring. Unless otherwise specifically indicated in the specification, a heteroaryl group may be a monocyclic, bicyclic, tricyclic or higher ring system, and may also be fused to a cycloalkyl or heterocyclic group as defined above, provided that the heteroaryl group is attached to the rest of the molecule by a single bond via an atom on the aromatic ring. The nitrogen, carbon or sulfur atoms in the heteroaryl group may be optionally oxidized; the nitrogen atoms may optionally be quaternized. For the purposes of the present invention, heteroaryl is preferably a stable 5-to 12-membered aromatic group containing 1 to 5 heteroatoms selected from nitrogen, oxygen and sulfur, more preferably a stable 5-to 10-membered aromatic group containing 1 to 4 heteroatoms selected from nitrogen, oxygen and sulfur or a 5-to 6-membered aromatic group containing 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur. Examples of heteroaryl groups include, but are not limited to, thienyl, imidazolyl, pyrazolyl, thiazolyl, oxazolyl, oxadiazolyl, isoxazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, benzimidazolyl, benzopyrazolyl, indolyl, furyl, pyrrolyl, triazolyl, tetrazolyl, triazinyl, indolizinyl, isoindolyl, indazolyl, isoindolyl, purinyl, quinolyl, isoquinolyl, diazonaphthyl, naphthyridinyl, quinoxalinyl, pteridinyl, carbazolyl, carbolinyl, phenanthridinyl, phenanthrolinyl, acridinyl, phenazinyl, isothiazolyl, benzothiazolyl, benzothienyl, oxazolyl, cinnolinyl, quinazolinyl, thiophenyl, indolizinyl, orthophenanthrolidinyl, isoxazolyl, phenoxazinyl, phenothiazinyl, 4,5,6, 7-tetrahydrobenzo [ b ] thienyl, naphthopyridyl, pyridinyl, and the like, [1,2,4] triazolo [4,3-b ] pyridazine, [1,2,4] triazolo [4,3-a ] pyrazine, [1,2,4] triazolo [4,3-c ] pyrimidine, [1,2,4] triazolo [4,3-a ] pyridine, imidazo [1,2-b ] pyridazine, imidazo [1,2-a ] pyrazine and the like.
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, "optional" 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 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.
"stereoisomers" refers to compounds that consist of the same atoms, are bonded by the same bonds, but have different three-dimensional structures. The present invention is intended to cover various stereoisomers and mixtures thereof.
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 one atom of a molecule to another atom of the same molecule. All tautomeric forms of the compounds of the invention are also intended to be included within the scope of the invention.
The compounds of the present invention or pharmaceutically acceptable salts thereof may contain one or more chiral carbon atoms and may therefore give rise to enantiomers, diastereomers and other stereoisomeric forms. Each chiral carbon atom may be defined as (R) -or (S) -, based on stereochemistry. The present invention is intended to include all possible isomers, as well as racemates and optically pure forms thereof. The compounds of the invention may be prepared by selecting as starting materials or intermediates racemates, diastereomers or enantiomers. Optically active isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, e.g., crystallization and chiral chromatography.
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 separation, Methods and Protocols, Methods in Molecular Biology, Vol.243, 2004; A.M.Stalcup, Chiral separation, Annu.Rev.Anal.Chem.3: 341. 63, 2010; Fumis et al (Eds.), GEL' S CYENCLOP EDIA OFACTICA ORGANIC CHEMISTRY 5. ED., Longmanetic and scientific Ltd., EsX, 1991, Res.816; Help.1990. 23,128. E.32.
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 with inorganic or organic acids which 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; organic acid salts include, but are not limited to, formates, acetates, 2-dichloroacetates, trifluoroacetates, propionates, caproates, caprylates, caprates, undecylenates, glycolates, gluconates, lactates, sebacates, adipates, glutarates, malonates, oxalates, maleates, succinates, fumarates, tartrates, citrates, palmitates, stearates, oleates, cinnamates, laurates, malates, glutamates, pyroglutamates, aspartates, benzoates, methanesulfonates, benzenesulfonates, p-toluenesulfonates, alginates, ascorbates, salicylates, 4-aminosalicylates, napadisylates, and the like. These salts can be prepared by methods known in the art.
"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.
"polymorph" refers to different solid crystalline phases of certain compounds of the present invention in the solid state due to the presence of two or more different molecular arrangements. Certain compounds of the present invention may exist in more than one crystalline form and the present invention is intended to include the various crystalline forms and mixtures thereof.
Typically, crystallization will result in solvates of the compounds of the invention. The term "solvate" as used herein refers to an aggregate comprising one or more molecules of the compound of the present invention and one or more solvent molecules. The solvent may be water, in which case the solvate is a hydrate. Alternatively, the solvent may be an organic solvent. Thus, the compounds of the present invention may exist as hydrates, including monohydrates, dihydrate, hemihydrate, sesquihydrates, trihydrate, tetrahydrate, and the like, as well as the corresponding solvated forms. The compounds of the invention may form true solvates, but in some cases it is also possible to retain only adventitious water or a mixture of water plus a portion of adventitious solvent. The compounds of the invention may be reacted in a solvent or precipitated or crystallized from a solvent. Solvates of the compounds of the invention are also included within the scope of the invention.
The invention also includes prodrugs of the above compounds. In the present application, the term "prodrug" denotes a compound that can be converted under physiological conditions or by solvolysis to the biologically active compound of the invention. Thus, the term "prodrug" refers to a pharmaceutically acceptable metabolic precursor of a compound of the invention. Prodrugs may not be active when administered to a subject in need thereof, but are converted in vivo to the active compounds of the invention. Prodrugs are generally rapidly converted in vivo to yield the parent compound of the invention, for example, by hydrolysis in blood. Prodrug compounds generally provide solubility, histocompatibility, or sustained release advantages in mammalian organisms. Prodrugs include known amino protecting groups and carboxyl protecting groups. Specific methods for preparing prodrugs can be found in Saulnier, M.G., et al, bioorg.Med.chem.Lett.1994,4, 1985-1990; greenwald, r.b., et al, j.med.chem.2000,43,475.
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 carrier" 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 approved by the relevant governmental regulatory agency for human or livestock use.
The "tumor" and "diseases related to abnormal cell proliferation" include, but are not limited to, leukemia, gastrointestinal stromal tumor, 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, and the like.
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;
(ii i) alleviating, i.e., causing regression of the state of, the disease or condition; or
(iv) Alleviating the symptoms caused by the disease or disorder.
The terms "effective amount," "therapeutically effective amount," or "pharmaceutically effective amount" as used herein, refer to an amount of at least one agent or compound that is sufficient to alleviate one or more symptoms of the disease or disorder being treated to some extent after administration. The result may be a reduction and/or alleviation of signs, symptoms, or causes, or any other desired change in a biological system. For example, an "effective amount" for treatment is the amount of a composition comprising a compound disclosed herein that is clinically necessary to provide a significant remission effect of the condition. An effective amount suitable in any individual case can be determined using techniques such as a dose escalation assay.
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 terms "drug combination", "administering other treatment", "administering other therapeutic agent" and the like as used herein refer to a drug treatment obtained by mixing or combining more than one active ingredient, including fixed and unfixed combinations of active ingredients. The term "fixed combination" refers to the simultaneous administration of at least one compound described herein and at least one co-agent to a patient in the form of a single entity or a single dosage form. The term "non-fixed combination" refers to the simultaneous administration, concomitant administration, or sequential administration at variable intervals of at least one compound described herein and at least one synergistic formulation to a patient as separate entities. These also apply to cocktail therapy, for example the administration of three or more active ingredients.
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.
The invention will be further illustrated with reference to specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. Experimental procedures without specific conditions noted in the following examples, generally according to conventional conditions, or according to conditions recommended by the manufacturer. Percentages and parts are by weight unless otherwise indicated.
The first preparation method of the intermediate comprises the following steps: preparation of aromatic cyclic lactams
Figure BDA0002376519750000161
Referring to the synthetic route and method of patent WO2017004383A1, 5-bromo (iodo) -isoindolin-1-one intermediates 1-A-1-C are prepared.
Figure BDA0002376519750000162
And a second intermediate preparation method comprises the following steps: synthesis of pyrazine/pyridinamine compounds
Intermediate 2-A: 3-bromo-5- (tetrahydro-2H-pyran-4-yl) pyrazin-2-amine
Figure BDA0002376519750000171
The first step is as follows: 5-Bromopyrazin-2-amine (600mg, 3.5mmol), (3, 6-dihydro-2H-pyran-4-yl) pinacol boronate (798mg, 3.8mmol), sodium carbonate (720mg, 6.8mmol) and Pd (dppf) Cl2DCM (277mg, 0.34mmol) was dissolved in ethylene glycol dimethyl ether and water (30mL/10mL) and heated to 90 degrees under nitrogen overnight reaction. The reaction solution was cooled to room temperature, filtered, concentrated under reduced pressure, and the residue was purified by column chromatography to give 5- (3, 6-dihydro-2H-pyran-4-yl) pyrazin-2-amine (300mg, white solid). LC-MS ESI [ M + H ]]+=178.1。
The second step is that: 5- (3, 6-dihydro-2H-pyran-4-yl) -pyrazin-2-amine (300mg, 1.7mmol) was dissolved in methanol (20mL), 10% palladium on carbon (120mg) was added, and the mixture was replaced with hydrogen and reacted at 30 ℃ overnight. The reaction solution was filtered, concentrated under reduced pressure, and purified by column chromatography to give 5- (tetrahydro-2H-pyran-4-yl) pyrazin-2-amine (280mg, white solid). LC-MS ESI [ M + H ]]+=180.1。
The third step: 5- (tetrahydro-2H-pyran-4-yl) pyrazin-2-amine (280mg, 1.56mmol) was dissolved in acetonitrile (20mL), NBS (300mg, 1.68mmol) was added, and the mixture was stirred at room temperature for 1 hour. The reaction mixture was concentrated under reduced pressure in a liquid-cooled manner, and purified by column chromatography to give intermediate 2-A (400mg, yellow solid). LC-MS ESI [ M + H ]]+=258.2/260.2;1H-NMR(DMSO-d6,400MHz)7.90(s,1H),6.50(s,2H),3.90-3.93(m,2H),3.36-3.43(m,2H),2.75-2.79(m,1H),1.62-1.72(m,4H)。
Referring to the synthetic route and the method of patent WO2015066188A1, pyrazine/pyridinamine intermediate compounds 2-B to 2-D are prepared.
Figure BDA0002376519750000172
And a third intermediate preparation method: preparation of 2-bromo-6, 7-dihydroimidazo [1,2-a ] pyrazin-8 (5H) -one
Figure BDA0002376519750000173
Referring to the synthetic route and the method of patent WO2017080979A1, 2-bromo-6, 7-dihydroimidazo [1,2-a ] pyrazine-8 (5H) -ketone intermediates 3-A-3-C are prepared.
Figure BDA0002376519750000181
The preparation method of the intermediate is four: preparation of 2-bromo-6, 7-dihydropyrazolo [1,5-a ] pyrazin-4 (5H) -one
Figure BDA0002376519750000182
Referring to the synthetic route and method of patent WO2017033966A1, 2-bromo-6, 7-dihydropyrazolo [1,5-a ] pyrazin-4 (5H) -one intermediates 4-A-4-C are prepared.
Figure BDA0002376519750000183
The general preparation method comprises the following steps: preparation of pyrazine/pyridinamine lactam compounds
Figure BDA0002376519750000184
The first step is as follows: dissolving the aromatic ring lactam intermediate (1eq.) in anhydrous dioxane, and adding the boric acid pinacol ester (2eq.), anhydrous potassium carbonate powder (2.5eq.) and Pd (dppf) Cl in sequence2(0.1eq.) and reflux with heating under nitrogen for 2 hours. TLC monitored the reaction was complete, cooled to room temperature and used directly in the next reaction.
The second step is that: cooling the reaction solution to room temperature, and rapidly adding 2-amino-3-bromopyrazine intermediate (1.5eq.) or 2-amino-3-bromopyridine intermediate (1.25eq.) and Pd (PPh)3)4(0.1eq.) and heating was continued for 2 hours. After the reaction was completed, the mixture was filtered through celite, washed with ethyl acetate, and the filtrate was concentrated. Diluting the residue with dichloromethane, washing the organic phase with water, saturated sodium bicarbonate solution and saturated brine successively, drying with anhydrous sodium sulfate, filtering, concentrating under reduced pressure, separating and purifying the residue with preparative HPLC to obtain the target compound, and confirming the structure by nuclear magnetism and mass spectrometry.
The general preparation method II comprises the following steps: preparation of pyrazine/pyridinamine lactam compounds
Figure BDA0002376519750000191
The first step is as follows: dissolving the aromatic ring lactam intermediate (1eq.) in anhydrous dioxane, and adding the boric acid pinacol ester (2eq.), anhydrous potassium carbonate powder (2.5eq.) and Pd (dppf) Cl in sequence2(0.1eq.) and reflux with heating under nitrogen for 2 hours. TLC monitored the reaction was complete, cooled to room temperature and used directly in the next reaction.
The second step is that: cooling the reaction solution to room temperature, and rapidly adding 2-amino-3, 5-dibromopyrazine compound (1.5eq.) or 2-amino-3, 5-dibromopyridine compound (1.25eq.) and Pd (PPh)3)4(0.1eq.) and heating was continued for 2 hours. After the reaction was completed, the mixture was filtered through celite, washed with ethyl acetate, and the filtrate was concentrated. The residue is diluted by dichloromethane, the organic phase is washed by water, saturated sodium bicarbonate solution and saturated brine in turn, dried by anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and the residue is separated and purified by column chromatography to obtain the target product.
The third step: dissolving the second-step product (1eq.) in anhydrous dioxane, and sequentially adding boric acid or other metal active raw materials (2eq.) of R2, anhydrous potassium carbonate powder (2.5eq.) and Pd (PPh)3)4(0.1eq.) and reflux with heating under nitrogen for 2 hours. After the reaction was completed, the mixture was filtered through celite, washed with ethyl acetate, and the filtrate was concentrated.Diluting the residue with dichloromethane, washing the organic phase with water, saturated sodium bicarbonate solution and saturated brine successively, drying with anhydrous sodium sulfate, filtering, concentrating under reduced pressure, separating and purifying the residue with preparative HPLC to obtain the target compound, and confirming the structure by nuclear magnetism and mass spectrometry.
The general preparation method comprises the following steps: preparation of pyrazine/pyridinamine lactam compounds
Figure BDA0002376519750000201
The first step is as follows: dissolving aromatic ring lactam compound (1eq.) in anhydrous dioxane, and sequentially adding boric acid pinacol ester (2eq.), anhydrous potassium carbonate powder (2.5eq.) and Pd (dppf) Cl2(0.1eq.) and reflux with heating under nitrogen for 2 hours. TLC monitored the reaction was complete, cooled to room temperature and used directly in the next reaction.
The second step is that: cooling the reaction solution to room temperature, and rapidly adding 2-amino-3-bromopyrazine intermediate (1.5eq.) or 2-amino-3-bromopyridine intermediate (1.25eq.) and Pd (PPh)3)4(0.1eq.) and heating was continued for 2 hours. After the reaction was completed, the mixture was filtered through celite, washed with ethyl acetate, and the filtrate was concentrated. The residue is diluted by dichloromethane, the organic phase is washed by water, saturated sodium bicarbonate solution and saturated brine in turn, dried by anhydrous sodium sulfate, filtered, concentrated under reduced pressure, and the residue is separated and purified by column chromatography to obtain the target product.
The third step: the second-step product (1eq.) was dissolved in anhydrous DMF, and anhydrous potassium carbonate powder (2.2eq.) and aryl benzyl halide reagent (1eq.) were added in that order and stirred at room temperature overnight. After the reaction is finished, dichloromethane and water are used for dilution, the organic phase is washed by water, saturated sodium bicarbonate solution and saturated saline solution in turn, anhydrous sodium sulfate is dried, the filtration and the decompression concentration are carried out, the remainder is separated and purified by preparative HPLC to obtain the target compound, and the structure is confirmed by adopting nuclear magnetism and mass spectrum.
Examples
The following compounds of examples were prepared by using intermediates 1 to 4 and other commercial reagents as starting materials and using the synthesis methods of example general preparation method one, example general preparation method two and example general preparation method three, respectively.
Example 1: 5- (3-amino-6- (tetrahydro-2H-pyran-4-yl) pyrazin-2-yl) -2- (3-chlorobenzyl) isoindolin-1-one
Figure BDA0002376519750000202
The first step is as follows: (1-Oxoisoindolin-5-yl) pinacolboronic acid ester (100mg, 0.39mmol), 1- (bromomethyl) -3-chlorobenzene (121.3mg, 0.59mmol) and potassium tert-butoxide (87mg, 0.78mmol) were dissolved in DMF (10mL) and heated to 30 ℃ for reaction overnight. The reaction mixture was concentrated under reduced pressure, and the residue was purified by column chromatography to give (2- (3-chlorobenzyl) -1-oxoisoindolin-5-yl) pinacol boronic acid ester (50mg, white solid). LC-MS ESI [ M + H ]]+=384.1。
The second step is that: (2- (3-chlorobenzyl) -1-oxoisoindolin-5-yl) pinacol boronic acid ester (50mg, 0.13mmol), 3-bromo-5- (tetrahydro-2H-pyran-4-yl) pyrazin-2-amine (34mg, 0.13mmol), cesium carbonate (84mg, 0.26mmol) and Pd (PPh)3)4(15mg, 0.013mmol) was dissolved in dioxane and water (10mL/2mL) and heated to 90 ℃ under nitrogen for overnight reaction. The reaction was cooled to room temperature, filtered, concentrated under reduced pressure, and the residue was purified by column chromatography to give 5- (3-amino-6- (tetrahydro-2H-pyran-4-yl) pyrazin-2-yl) -2- (3-chlorobenzyl) isoindolin-1-one (8.2mg, white solid). LC-MS ESI [ M + H ]]+=435.2;1H-NMR(CDCl3,400MHz)8.06(d,J=7.6Hz,1H),7.85(d,J=8.0Hz,1H),7.76(d,J=10.8Hz,2H),7.20-7.37(m,4H),6.00(br s,2H),4.82(s,2H),4.38(s,2H),4.08-4.11(m,2H),3.51-3.57(m,2H),2.93-2.95(m,1H),1.70-2.03(m,4H)。
Example 2: (R) -2- (3-amino-6- (tetrahydro-2H-pyran-4-yl) pyrazin-2-yl) -6- (methoxymethyl) -5- ((6-methylpyridin-2-yl) methyl) -6, 7-dihydropyrazolo [1,5-a ] pyrazin-4 (5H) -one
Figure BDA0002376519750000211
The first step is as follows: reacting (R) -2-bromo-6- (methyl)Oxymethyl) -6, 7-dihydropyrazolo [1,5-a]Pyrazine-4 (5H) -one (780mg, 3.0mmol) was dissolved in THF (15mL), cooled to 0 ℃ under nitrogen, 60% NaH (144mg,3.6mmol) was added, stirred for 30min, 2- (bromomethyl) -6-methylpyridine (670mg, 3.6mmol) was added dropwise, and stirred at room temperature overnight. Saturated NH is poured into the reaction solution4Extracting with ethyl acetate in Cl aqueous solution, and anhydrous MgSO4Drying, filtering, concentrating under reduced pressure, and purifying the residue by column chromatography to obtain (R) -2-bromo-6- (methoxymethyl) -5- ((6-methylpyridin-2-yl) methyl) -6, 7-dihydropyrazolo [1,5-a]Pyrazin-4 (5H) -one (270mg, colourless oil). LC-MS ESI [ M + H ]]+=365.1;1H-NMR(CDCl3,400MHz)7.46(t,J=7.6Hz,1H),7.01(d,J=7.6Hz,1H),6.79(s,1H),6.49(d,J=8.0Hz,1H),5.89-5.98(q,2H),4.32-4.41(m,2H),4.19-4.22(m,1H),3.49-3.52(m,1H),3.27-3.31(m,4H),2.53(s,3H)。
The second step is that: reacting (R) -2-bromo-6- (methoxymethyl) -5- ((6-methylpyridin-2-yl) methyl) -6, 7-dihydropyrazolo [1, 5-a)]Pyrazin-4 (5H) -one (200mg, 0.55mmol) and pinacol diborane (207mg, 0.81mmol) were dissolved in 1, 4-dioxane (10mL) and KOAc (159mg,1.62mmol), dppf (15mg, 0.027mmol) and Pd (dppf) Cl were added under nitrogen2.CH2Cl2(22mg, 0.027mmol), replaced with nitrogen three times, and heated to 80 ℃ for reaction overnight. Cooling the reaction solution to room temperature, filtering, concentrating under reduced pressure, and purifying the residue by column chromatography to obtain (R) - (6- (methoxymethyl) -5- ((6-methylpyridin-2-yl) methyl) -4-oxo-4, 5,6, 7-tetrahydropyrazolo [1, 5-a)]Pyrazine-2-yl) pinacol borate (137mg, white solid). LC-MS ESI [ M + H ]]+=413.1。
The third step: mixing (R) - (6- (methoxymethyl) -5- ((6-methylpyridin-2-yl) methyl) -4-oxo-4, 5,6, 7-tetrahydropyrazolo [1, 5-a)]Pyrazine-2-yl) pinacol borate (112mg, 0.27mmol), 3-bromo-5- (tetrahydro-2H-pyran-4-yl) pyrazine-2-amine (70mg, 0.27mmol), cesium carbonate (176mg, 0.54mmol) and Pd (PPh)3)4(31mg, 0.027mmol) was dissolved in dioxane and water (10mL/2mL) and heated to 90 ℃ under nitrogen for overnight reaction. Cooling the reaction solution to room temperature, filtering, concentrating under reduced pressure, and purifying the residue by column chromatography to obtain (R) -2- (3-amino-6- (tetrahydro-2H-pyrazine)Pyran-4-yl) pyrazin-2-yl) -6- (methoxymethyl) -5- ((6-methylpyridin-2-yl) methyl) -6, 7-dihydropyrazolo [1,5-a]Pyrazin-4 (5H) -one (8.1mg, white solid). LC-MS ESI [ M + H ]]+=464.3;1H-NMR(CDCl3,400MHz)7.82(s,1H),7.60(d,J=3.2Hz,1H),7.44(t,J=8.0Hz,1H),7.02(d,J=8.0Hz,1H),6.48(d,J=8.0Hz,1H),6.20(br s,2H),5.98-6.10(m,2H),4.38-4.46(m,2H),4.26(d,J=7.2Hz,1H),4.02-4.12(m,2H),3.49-3.59(m,3H),3.31-3.35(m,4H),2.86-2.88(m,1H),2.56(s,3H),1.94-1.97(m,2H),1.82-1.86(m,2H)。
Example 3: 5- (3-amino-6- (tetrahydro-2H-pyran-4-yl) pyrazin-2-yl) -1- (2- (3-chlorophenyl) acetyl) indoline-7-carbonitrile
Figure BDA0002376519750000221
The first step is as follows: (7-Cyanoindolin-5-yl) pinacolboronic acid ester (100mg, 0.37mmol) and 2- (3-chlorophenyl) acetic acid (75mg, 0.44mmol) were dissolved in pyridine, and 2 drops of phosphorus oxychloride were added dropwise and stirred at room temperature overnight. The reaction mixture was concentrated under reduced pressure, and the residue was purified by column chromatography to give (1- (2- (3-chlorophenyl) acetyl) -7-cyanoindolin-5-yl) boronic acid (35mg, white solid). LC-MS ESI [ M + H ]]+=341.0。
The second step is that: (1- (2- (3-chlorophenyl) acetyl) -7-cyanoindolin-5-yl) boronic acid (35mg, 0.10mmol), 3-bromo-5- (tetrahydro-2H-pyran-4-yl) pyrazin-2-amine (26mg, 0.10mmol), sodium carbonate (21mg, 0.20mmol) and Pd (PPh)3)4(3mg, 0.002mmol) was dissolved in dioxane and water (10mL/2mL) and heated to 90 ℃ under nitrogen for overnight reaction. The reaction solution was cooled to room temperature, filtered, concentrated under reduced pressure, and purified by column chromatography to give 5- (3-amino-6- (tetrahydro-2H-pyran-4-yl) pyrazin-2-yl) -1- (2- (3-chlorophenyl) acetyl) indoline-7-carbonitrile (9.5mg, white solid). LC-MS ESI [ M + H ]]+=474.2;1H-NMR(DMSO-d6,400MHz)8.11(s,1H),7.86(s,1H),7.63(s,1H),7.47(t,J=7.6Hz,1H),7.38(d,J=7.6Hz,1H),7.30(s,1H),7.25(d,J=7.6Hz,1H),6.17(br s,2H),5.97(s,2H),4.24(t,J=8.0Hz,2H),3.93-3.96(m,2H),3.37-3.47(m,4H),2.80-2.90(m,1H),1.74-1.80(m,4H)。
Example 4: (S) -5- (3-amino-6- (tetrahydro-2H-pyran-4-yl) pyrazin-2-yl) -2- (1- (3-chlorophenyl) -2-hydroxyethyl) isoindolin-1-one
Figure BDA0002376519750000231
The first step is as follows: methyl 4-bromo-2-methylbenzoate (1.0g, 4.39mmol) was dissolved in CCl4NBS (850mg, 4.78mmol) and AIBN (59mg, 0.36mmol) were added to (15mL) under nitrogen, heated to 80 ℃ and stirred overnight. The reaction solution was cooled to room temperature, concentrated under reduced pressure, and the residue was purified by column chromatography to give methyl 4-bromo-2- (bromomethyl) benzoate (1.0g, white solid). LC-MS ESI [ M + H ]]+=307.1。
The second step is that: methyl 4-bromo-2- (bromomethyl) benzoate (300mg, 0.98mmol) and (S) -2-amino-2- (3-chlorophenyl) ethyl-1-ol (168mg, 0.98mmol) were dissolved in acetonitrile (10mL), DIEA (0.8mL,4.84mmol) was added under nitrogen, heated to 70 ℃ and stirred overnight. The reaction mixture was cooled to room temperature, concentrated under reduced pressure, and the residue was purified by column chromatography to give (S) -5-bromo-2- (1- (3-chlorophenyl) -2-hydroxyethyl) isoindolin-1-one (160mg, white solid). LC-MS ESI [ M + H ]]+=365.9。
The third step: (S) -5-bromo-2- (1- (3-chlorophenyl) -2-hydroxyethyl) isoindolin-1-one (160mg, 0.44mmol) and pinacol diborane (223mg, 0.88mmol) were dissolved in 1, 4-dioxane (6mL), and KOAc (129mg,1.32mmol), dppf (49mg, 0.088mmol) and Pd (dppf) Cl were added under nitrogen protection2.CH2Cl2(36mg, 0.044mmol), replaced with nitrogen three times, and heated to 100 ℃ for reaction overnight. The reaction mixture was cooled to room temperature, filtered, concentrated under reduced pressure, and the residue was purified by column chromatography to give (S) - (2- (1- (3-chlorophenyl) -2-hydroxyethyl) -1-oxoisoindolin-5-yl) pinacol borate (56.6mg, white solid). LC-MS ESI [ M + H ]]+=414.5。
The fourth step: (S) - (2- (1- (3-chlorophenyl) -2-hydroxyethyl) -1-oxoisoindolin-5-yl) pinacol borate (56.6mg, 0.14mmol), 3-bromo-5- (tetrahydro-2H-pyran-4-yl) pyrazin-2-amine (35mg, 0.14mmol), sodium carbonate (44mg, 0.41 mmol), sodium chloride (sodium chloride, and sodium chloridemmol) and Pd (PPh)3)4(8mg, 0.007mmol) was dissolved in dioxane and water (6mL/1mL) and heated to 80 ℃ under nitrogen for reaction overnight. The reaction mixture was cooled to room temperature, filtered, concentrated under reduced pressure, and the residue was purified by column chromatography to give (S) -5- (3-amino-6- (tetrahydro-2H-pyran-4-yl) pyrazin-2-yl) -2- (1- (3-chlorophenyl) -2-hydroxyethyl) isoindolin-1-one (40mg, white solid). LC-MS ESI [ M + H ]]+=465.3;1H-NMR(CD3OD,400MHz)7.85-7.93(m,4H),7.43(s,1H),7.30-7.39(m,3H),5.50-5.53(m,1H),4.76(d,J=17.6Hz,1H),4.47(d,J=17.6Hz,1H),4.14-4.26(m,2H),4.04(dd,J=2.4,11.6Hz,2H),3.53-3.59(td,J=2.4,11.6Hz,2H),2.88-2.94(m,1H),1.80-1.95(m,4H)。
Example 5: (S) -6- (3-amino-6- (tetrahydro-2H-pyran-4-yl) pyrazin-2-yl) -2- (1- (3-chlorophenyl) -2-hydroxyethyl) isoindolin-1-one
Figure BDA0002376519750000241
The target compound was prepared from methyl 5-bromo-2-methylbenzoate by the same synthetic method as in example 4. LC-MS ESI [ M + H ]]+=465.3;1H-NMR(CD3OD,400MHz)8.17(s,1H),7.97(dd,J=1.2,8.0Hz,1H),7.86(s,1H),7.68(d,J=7.6Hz,1H),7.31-7.43(m,4H),5.48-5.53(m,1H),4.76(d,J=18.0Hz,1H),4.45(d,J=18.0Hz,1H),4.13-4.26(m,2H),4.04(dd,J=2.4,11.6Hz,2H),3.56(td,J=2.0,11.6Hz,2H),2.88-2.95(m,1H),1.81-1.95(m,4H)。
Example 6: (R) -2- (6- (3-amino-6- (tetrahydro-2H-pyran-4-yl) pyrazin-2-yl) -1-oxoisoindolin-2-yl) -N- ((S) -1- (3-chlorophenyl) -2-hydroxyethyl) propionamide
Figure BDA0002376519750000242
The first step is as follows: methyl 5-bromo-2-methylbenzoate (1.0g, 4.39mmol) was dissolved in CCl4NBS (1.1g, 6.18mmol) and AIBN (108mg, 0.66mmol) were added to (15mL) under nitrogen, heated to 80 ℃ and stirred overnight. Cooling the reaction liquid to room temperature, decompressing and concentrating, and carrying out column chromatography purification on the residuesThis gave methyl 5-bromo-2- (bromomethyl) benzoate (1.3g, white solid). LC-MS ESI [ M + H ]]+=307.1。
The second step is that: methyl 5-bromo-2- (bromomethyl) benzoate (500mg, 1.63mmol) and D-alanine tert-butyl ester hydrochloride (297mg, 1.63mmol) were dissolved in acetonitrile (10mL), TEA (1.5mL, 10.8mmol) was added under nitrogen, heated to 70 ℃ and stirred overnight. The reaction mixture was cooled to room temperature, concentrated under reduced pressure, and the residue was purified by column chromatography to give tert-butyl (R) -2- (6-bromo-1-oxoisoindolin-2-yl) propionate (270mg, white solid). LC-MS ESI [ M + H ]]+=340.0。
The third step: (R) -tert-butyl 2- (6-bromo-1-oxoisoindolin-2-yl) propionate (160mg, 0.47mmol) and pinacol diborane (240mg, 0.94mmol) were dissolved in 1, 4-dioxane (5mL), and KOAc (139mg,1.42mmol), dppf (52mg, 0.09mmol) and Pd (dppf) Cl were added under nitrogen protection2.CH2Cl2(39mg, 0.05mmol), replaced with nitrogen three times, and heated to 100 ℃ for reaction overnight. The reaction mixture was cooled to room temperature, filtered, concentrated under reduced pressure, and the residue was purified by column chromatography to give (R) - (2- (1- (tert-butoxy) -1-oxopropyl-2-yl) -3-oxoisoindolin-5-yl) pinacol borate (119.4mg, white solid). LC-MS ESI [ M + H ]]+=388.2。
The fourth step: (R) - (2- (1- (tert-butoxy) -1-oxopropyl-2-yl) -3-oxoisoindolin-5-yl) pinacol borate (119.4mg, 0.31mmol), 3-bromo-5- (tetrahydro-2H-pyran-4-yl) pyrazin-2-amine (79.3mg, 0.31mmol), sodium carbonate (98.1mg,0.93mmol) and Pd (PPh)3)4(17.8mg, 0.016mmol) was dissolved in dioxane and water (10mL/1.5mL) and heated to 80 ℃ under nitrogen for reaction overnight. The reaction was cooled to room temperature, filtered, concentrated under reduced pressure, and the residue was purified by column chromatography to give tert-butyl (R) -2- (6- (3-amino-6- (tetrahydro-2H-pyran-4-yl) pyrazin-2-yl) -1-oxoisoindolin-2-yl) propionate (95.1mg, white solid). LC-MS ESI [ M + H ]]+=439.2。
The fifth step: tert-butyl (R) -2- (6- (3-amino-6- (tetrahydro-2H-pyran-4-yl) pyrazin-2-yl) -1-oxoisoindolin-2-yl) propanoate (95.1mg, 0.22mmol) was dissolved in anhydrous dichloromethane (2mL) and TFA (1. sup. th-e) was added dropwisemL) was added, and the reaction was carried out at room temperature for 1 hour. Concentrating under reduced pressure, dissolving the residue in methanol (15mL), adding K2CO3(500mg) and water (2mL) under reflux for 30min, concentrated under reduced pressure, and purified by preparative chromatography to give (R) -2- (6- (3-amino-6- (tetrahydro-2H-pyran-4-yl) pyrazin-2-yl) -1-oxoisoindolin-2-yl) propionic acid (123.2mg, crude, white solid). LC-MS ESI [ M + H ]]+=383.2。
And a sixth step: (R) -2- (6- (3-amino-6- (tetrahydro-2H-pyran-4-yl) pyrazin-2-yl) -1-oxoisoindolin-2-yl) propionic acid (123.2mg, crude) was dissolved in DMF (5mL), HATU (245mg, 0.64mmol) and TEA (130.3mg, 1.29mmol) were added, and after stirring for 5 minutes, (S) -2-amino-2- (3-chlorophenyl) ethyl-1-ol (55mg, 0.32mmol) was added and the reaction was allowed to proceed at room temperature overnight. Diluting with ethyl acetate, washing with a saturated ammonium chloride solution, drying, concentrating under reduced pressure, and purifying by column chromatography to give (R) -2- (6- (3-amino-6- (tetrahydro-2H-pyran-4-yl) pyrazin-2-yl) -1-oxoisoindolin-2-yl) -N- ((S) -1- (3-chlorophenyl) -2-hydroxyethyl) propionamide (35mg, white solid). LC-MS ESI [ M + H ]]+=536.2;1H-NMR(DMSO_d6,400MHz)8.65(d,J=7.2Hz,1H),8.02(s,1H),7.97(d,J=8.0Hz,1H),7.90(d,J=2.4Hz,1H),7.72(dd,J=2.0,8.8Hz,1H),7.28-7.39(m,4H),5.02-5.04(m,1H),4.55-4.84(m,3H),3.93-3.96(m,2H),3.56(d,J=4.4Hz,2H),3.41-3.45(m,2H),2.83-2.92(m,1H),1.76(br s,4H),1.44(d,J=5.6Hz,3H)。
Example 7: 5- (3-amino-6- (tetrahydro-2H-pyran-4-yl) pyrazin-2-yl) -2- (3-chloro-2- (hydroxymethyl) benzyl) isoindolin-1-one
Figure BDA0002376519750000251
The target compound was prepared from (2- (aminomethyl) -6-chlorophenyl) methanol by the same synthesis method as in example 4. LC-MS ESI [ M + H ]]+=465.2;1H-NMR(CD3OD,400MHz)7.85-7.93(m,4H),7.23-7.36(m,3H),4.82(s,2H),4.76(s,2H),4.61(s,2H),4.05(dd,J=2.4,11.6Hz,2H),3.54-3.59(td,J=2.4,11.6Hz,2H),2.88-2.94(m,1H),1.81-1.95(m,4H)。
Example 8: (S) -5- (3-amino-6- (tetrahydro-2H-pyran-4-yl) pyrazin-2-yl) -2- (1- (3-bromo-5-fluorophenyl) -2-hydroxyethyl) isoindolin-1-one
Figure BDA0002376519750000261
The target compound was prepared by the same synthetic method as example 4. LC-MS ESI (M + H) 527.1;1H-NMR(CD3OD,400MHz)7.85-7.93(m,4H),7.52-7.58(m,2H),6.88(d,J=8.4Hz,1H),5.50-5.53(m,1H),4.76(d,J=17.6Hz,1H),4.47(d,J=17.6Hz,1H),4.14-4.26(m,2H),4.04(dd,J=2.4,11.6Hz,2H),3.53-3.59(td,J=2.4,11.6Hz,2H),2.88-2.94(m,1H),1.80-1.95(m,4H)。
example 9: (S) -5- (3-amino-6- (pyridin-4-yl) pyrazin-2-yl) -2- (1- (3-bromo-5-fluorophenyl) -2-hydroxyethyl) isoindolin-1-one
Figure BDA0002376519750000262
The target compound was prepared by the same synthetic method as example 4. LC-MS ESI (M + H) 520.1;1H-NMR(CD3OD,400MHz)8.79(d,2H),8.55(d,2H),7.85-7.93(m,4H),7.52-7.58(m,2H),6.88(d,J=8.4Hz,1H),5.50-5.53(m,1H),4.76(d,J=17.6Hz,1H),4.47(d,J=17.6Hz,1H),4.14-4.26(m,2H)。
example 10: (S) -5- (3-amino-6-trans- (4-hydroxycyclohexyl) pyrazin-2-yl) -2- (1- (3-bromo-5-fluorophenyl) -2-hydroxyethyl) isoindolin-1-one
Figure BDA0002376519750000263
The target compound was prepared by the same synthetic method as example 4. LC-MS ESI (M + H) 541.1;1H-NMR(CD3OD,400MHz)7.85-7.93(m,4H),7.52-7.58(m,2H),6.88(d,J=8.4Hz,1H),5.50-5.53(m,1H),4.76(d,J=17.6Hz,1H),4.47(d,J=17.6Hz,1H),4.14-4.30(m,3H),1.66-1.92(m,3H),1.39-1.57(m,2H),1.13-1.33(m,4H)。
example 11: (S) -5- (2-amino-5- (morphininyl) pyridin-3-yl) -2- (1- (3, 5-difluorophenyl) -2-hydroxyethyl) isoindolin-1-one
Figure BDA0002376519750000271
The target compound was prepared by the same synthetic method as example 4. LC-MS ESI (M + H) 467.2;1H-NMR(CD3OD,400MHz)7.75(d,J=2.3Hz,1H),7.30-7.40(m,2H),7.34(s,1H),6.60-6.74(m,4H),5.49-5.54(m,1H),4.78(d,J=17.6Hz,1H),4.50(d,J=17.6Hz,1H),4.13-4.25(m,2H),3.74-3.91(m,4H),2.90-3.07(m,4H)。
example 12: (R) -2- (3-amino-6- (tetrahydro-2H-pyran-4-yl) pyrazin-2-yl) -6- (methoxymethyl) -7- ((6-methylpyridin-2-yl) methyl) -6, 7-dihydroimidazo [1,2-a]Pyrazin-8 (5H) -ones
Figure BDA0002376519750000272
The target compound was prepared by the same synthetic method as example 2. LC-MS ESI (M + H) 464.2;1H-NMR(CDCl3,400MHz)8.02(s,1H),7.83(s,1H),7.44(t,J=8.0Hz,1H),7.02(d,J=8.0Hz,1H),6.48(d,J=8.0Hz,1H),6.20(brs,2H),5.98-6.10(m,2H),4.38-4.46(m,2H),4.26(d,J=7.2Hz,1H),4.02-4.12(m,2H),3.49-3.59(m,3H),3.31-3.35(m,4H),2.85-2.88(m,1H),2.56(s,3H),1.94-1.97(m,2H),1.83-1.86(m,2H)。
test example 1 determination of the inhibitory activity of the compounds of the present invention against ERK1 and ERK2 kinases (1) formulation of 1 XKinasebuffer; (2) preparation of compound concentration gradient: test compounds were tested at 10uM starting concentration, 3-fold diluted 10 concentrations, duplicate wells tested, and serially diluted in 96-well plates to 100-fold final concentration of 10 different concentration solutions. Then further diluting each concentration of compound with 1 x Kinasebuffer to an intermediate diluted solution of 5 times the final concentration; (3) respectively adding 5 mu L of the prepared compound solution into compound holes of a 384-hole plate, and testing each concentration single hole; adding 5 mu L of 5% DMSO into the negative control well and the positive control well respectively; (4) preparing a Kinase solution with 2.5 times of final concentration by using 1 XKinase buffer; (5) add 10. mu.L of 2.5 fold final concentration kinase solution to the compound well and positive control well, respectively; add 10. mu.L of 1 XKinase buffer to the negative control wells; (6) centrifuging at 1000rpm for 30 s, shaking, mixing uniformly, and incubating at room temperature for 10 min; (7) preparing a mixed solution of ATP and Kinase substrate22 at a final concentration of 2.5 times with 1 XKinasebuffer; (8) adding 10 μ L of a mixed solution of ATP and substrate at 2.5 times final concentration to initiate reaction; (9) centrifuging a 384-well plate at 1000rpm for 30 seconds, shaking and uniformly mixing, and then respectively incubating at 28 ℃ for corresponding time; (10) adding 30 mu L of termination detection solution to stop kinase reaction, centrifuging at 1000rpm for 30 seconds, and uniformly mixing by oscillation; (11) the conversion rate was read using Caliper EZ Reader ii, the log of the concentration was taken as the X-axis and the percent inhibition was taken as the Y-axis, and the log (inhibitor) vs. response-Variable slope of the analytical software GraphPad Prism5 was used to fit the dose-effect curves, thus obtaining the IC50 values of each compound for enzyme activity.
2. As a result, the present invention provides examples 1 to 12, which have IC of inhibitory activity against ERK250Value mean IC50Less than 500nM, inhibitory Activity of some examples even IC50Less than 10nM, showing strong inhibitory activity.
Test example 2: test of Effect of Compounds of the present invention on Ras-Raf-MEK-ERK-mediated proliferation Capacity of tumor cells
1. The test method comprises the following steps: colo-205 cells (ATCC) in logarithmic growth phase were inoculated into 96-well culture plates at an appropriate density, 90. mu.L per well, incubated overnight, and then a solvent control group (negative control) was set by adding compounds at different concentrations for 72 hr. After the compound acts on cells for 72 hours, the influence of the compound on cell proliferation is detected by a CCK-8 cell counting kit (Dojindo), 10 mu L of CCK-8 reagent is added into each hole, the hole is placed in a 370C incubator for 2 to 4 hours, a SpectraMax 190 reading is carried out by a full-wavelength microplate reader, and the measurement wavelength is 450 nm. The inhibition rate (%) of the compound on the growth of tumor cells was calculated by the following formula (OD negative control well-OD administration well)/OD negative control well × 100%. The IC50 value was determined by four parameter regression using microplate reader random accessory software.
2. As a result, the present invention provides proliferation inhibitory activity, IC, on Colo-205 cells in some of examples 1 to 1250Values of less than 1000nM, IC of inhibitory Activity of some of the example Compounds50Values even less than 200nM show strong cell proliferation inhibitory activity.
All documents referred to herein are incorporated by reference into this application as if each were individually incorporated by reference. Furthermore, it should be understood that various changes and modifications of the present invention can be made by those skilled in the art after reading the above teachings of the present invention, and these equivalents also fall within the scope of the present invention as defined by the appended claims.

Claims (6)

1. Pyrazine/pyridinamines compound shown as general formula I, or pharmaceutically acceptable salt thereof, or enantiomer, diastereoisomer, tautomer, solvate, polymorph or prodrug thereof,
Figure FDA0002376519740000011
in the formula:
r1 is independently selected from hydrogen, halogen, C1-C6 alkyl, C1-C6 alkoxy, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl;
r2 is independently selected from hydrogen, halogen, cyano, carbonyl, ester, amide, sulfonamide, sulfone, sulfoxide, sulfinyl, amino or substituted amino, C1-C10 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C1-C10 alkoxy, 3-10 membered saturated or partially unsaturated cycloalkyl or heterocycloalkyl, 5-10 membered aryl or heteroaryl;
m is independently selected from N, CR; r is independently selected from hydrogen, halogen, C1-C6 alkyl, C1-C6 alkoxy, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl;
r3 is independently selected from hydrogen, halogen, hydroxy, cyano, carbonyl, C1-C5 alkyl, C1-C5 alkoxy, amino, C1-C5 alkylamino;
l is independently selected from the group consisting of a bond, acyl, sulfonyl, alkylene of C1-C4, alkenylene of C2-C4, alkynylene of C2-C4, 3-8 membered cycloalkyl, 4-8 membered heterocycloalkyl;
r4 is independently selected from C1-C6 alkyl, 3-8 membered cycloalkyl, 4-8 membered saturated or partially unsaturated heterocycloalkyl, 5-8 membered aryl or heteroaryl; and the above R4 may be substituted with 1 to 3 of any of the following groups: deuterium, halogen, hydroxyl, amino, cyano, sulfone, sulfoxide, ester, amide, C1-C4 alkyl, C1-C4 alkoxy, C1-C4 thio, 3-8 membered cycloalkyl, 4-8 membered heterocycloalkyl;
r5, R6 are independently selected from hydrogen, halogen, C1-C6 alkyl, C1-C6 alkoxy, 3-8 membered cycloalkyl, 3-8 membered heterocycloalkyl; or a 3-6 membered saturated or partially unsaturated carbocyclic or heterocyclic ring is formed between R5 and R6 through a carbon chain or heteroatom;
ar is independently selected from a 5-10 membered aromatic ring or aromatic heterocycle;
z is independently selected from hydrogen, oxygen;
m and n are independently selected from 0, 1 and 2;
one or more hydrogen atoms on any of the above groups may be substituted with a substituent selected from the group consisting of: including but not limited to deuterium, halogen, hydroxyl, amino or substituted amino, cyano, sulfone or sulfoxide, urea, sulfonylurea, C1-C8 alkyl, 3-8 membered cycloalkyl, C1-C8 alkoxy, C1-C8 alkylamino or cycloalkylamino, alkenyl, alkynyl, acyl or sulfonyl or sulfinyl, 5-to 8-membered aryl or heteroaryl, 4-to 8-membered cycloalkyl or heterocycloalkyl; wherein said heteroaryl group contains 1 to 3 heteroatoms selected from the group consisting of: n, O, P or S, the heterocycloalkyl group containing 1 to 3 heteroatoms selected from the group consisting of: n, O, P or S, including but not limited to saturated or partially unsaturated or unsaturated monocyclic, fused, bridged, spiro, and the like ring systems.
2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, solvate, polymorph or prodrug thereof, wherein the compound is selected from the group consisting of formula (II):
Figure FDA0002376519740000021
r1 is preferably selected from hydrogen, deuterium, halogen, C1-C6 alkyl;
r2 is preferably selected from hydrogen, deuterium, C1-C8 alkyl, C1-C8 alkoxy, C1-C8 alkylamino, saturated or partially unsaturated 3-to 8-membered cycloalkyl, 3-to 8-membered cycloalkyloxy, 4-to 8-membered cycloalkylamino, 4-to 8-membered heterocycloalkylamino, saturated or partially unsaturated 3-to 8-membered heterocycloalkyl, 3-to 8-membered heterocycloalkoxy, 5-to 10-membered arylamine or heteroarylamine, 5-to 10-membered aryl or 5-to 10-membered heteroaryl; further preferably selected from: substituted or substituted cyclohexane, cyclohexenyl, tetrahydropyranyl, dihydropyranyl, piperidinyl, morpholinyl, piperazinyl, pyrazolyl, imidazolyl, isoxazolyl, phenyl, pyridinyl, isopropyl, cyclopentyl, cyclopropyl, oxetanyl, azetidinyl, thiocyclohexyl and the like; and the above groups may be substituted with one or more substituents preferably selected from the group consisting of: halogen, deuterium, cyano, hydroxy, amino, C1-C3 alkyl, C1-C3 alkylhydroxy, C1-C3 alkoxy, C1-C3 alkylamino, C1-C3 haloalkyl, C1-C3 haloalkoxy, oxo, acyl, sulfonyl, sulfone, etc.;
m, M1, M2, M3 are preferably from N, CR 3; m4, M5 are preferably selected from N, CR3, O, S, etc.; wherein R3 is preferably selected from H, F, methyl, CN;
m is preferably from 0 to 2;
l is preferably selected from acyl, sulfonyl, substituted or unsubstituted alkylene; and one or more hydrogens on the above alkylene group may be substituted with a substituent preferably selected from the group consisting of: deuterium, fluorine, hydroxyl, methyl, ethyl, cyclopropyl, alkoxy of C1-C2, alkylamino of C1-C2, alkylhydroxyl of C1-C2, alkoxyalkyl of C1-C2, alkylaminoalkyl of C1-C2, etc.;
r4 is preferably selected from phenyl or pyridyl, and the above groups may be substituted with one or more substituents selected from the group consisting of: halogen, cyano, C1-C4 alkyl, C1-C4 hydroxyalkyl, C1-C4 aminoalkyl, C1-C4 alkoxy, C1-C4 haloalkyl, C1-C4 haloalkoxy, C1-C4 sulfone or sulfoxide, ester, amide, sulfonamide, etc.;
r5 and R6 are preferably selected from hydrogen, fluorine, methyl, hydroxymethyl, methoxymethyl and the like.
3. A compound according to claims 1,2, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, solvate, polymorph or prodrug thereof, preferably according to the following general formula (III):
Figure FDA0002376519740000031
wherein R7 is preferably selected from hydrogen, deuterium, fluoro, methyl, ethyl, hydroxymethyl, methoxymethyl, aminomethyl, methylaminomethyl, and the like, and R1, R2, R3, R4, R5, R6, M, M1, M2, M3, M4, M, and the like are as defined above.
4. The compound of claims 1,2,3, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, solvate, polymorph, or prodrug thereof, wherein the compound has the structure:
Figure FDA0002376519740000032
Figure FDA0002376519740000041
Figure FDA0002376519740000051
Figure FDA0002376519740000061
5. use of a compound of formula I according to claims 1,2,3,4 or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, solvate, polymorph or prodrug thereof, for the preparation of a medicament, in particular a tumor treatment medicament, for the treatment of a disease associated with a kinase activity or an expression amount of a protein kinase, in particular of the Ras/Raf/MEK/ERK pathway; the tumor is independently selected from non-small cell lung cancer, lung adenocarcinoma, lung squamous carcinoma, breast cancer, prostatic cancer, liver cancer, skin cancer, gastric cancer, intestinal cancer, cholangiocarcinoma, brain cancer, leukemia, lymph cancer, nasopharyngeal cancer, pancreatic cancer, etc.
6. A pharmaceutical composition comprising a compound of formula I as defined in any one of claims 1,2,3,4, 5 or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, solvate, polymorph or prodrug thereof, wherein the pharmaceutical composition comprises:
(i) an effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, solvate, polymorph, or prodrug thereof; and
(ii) a pharmaceutically acceptable carrier.
CN202010067960.5A 2019-01-30 2020-01-20 Aminopyrazine/pyridine compound, preparation method and application Active CN111057048B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201910089203 2019-01-30
CN2019100892035 2019-01-30

Publications (2)

Publication Number Publication Date
CN111057048A true CN111057048A (en) 2020-04-24
CN111057048B CN111057048B (en) 2022-03-18

Family

ID=70306885

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010067960.5A Active CN111057048B (en) 2019-01-30 2020-01-20 Aminopyrazine/pyridine compound, preparation method and application

Country Status (1)

Country Link
CN (1) CN111057048B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022226665A1 (en) * 2021-04-30 2022-11-03 Ontario Institute For Cancer Research (Oicr) Halo-substituted amino aza-heteroaryl compounds as inhibitors of the haematopoietic progenitor kinase 1 (hpk1)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011163330A1 (en) * 2010-06-24 2011-12-29 Merck Sharp & Dohme Corp. Novel heterocyclic compounds as erk inhibitors
CN105050598A (en) * 2012-09-28 2015-11-11 默沙东公司 Novel compounds that are erk inhibitors
WO2016100050A1 (en) * 2014-12-15 2016-06-23 Merck Sharp & Dohme Corp. Erk inhibitors
CN106928216A (en) * 2015-12-31 2017-07-07 中国科学院上海药物研究所 Compound, Preparation Method And The Use with ERK kinase inhibiting activities
CN108617166A (en) * 2015-10-21 2018-10-02 大冢制药株式会社 Kinases inhibitor benzolactam compounds

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011163330A1 (en) * 2010-06-24 2011-12-29 Merck Sharp & Dohme Corp. Novel heterocyclic compounds as erk inhibitors
CN105050598A (en) * 2012-09-28 2015-11-11 默沙东公司 Novel compounds that are erk inhibitors
WO2016100050A1 (en) * 2014-12-15 2016-06-23 Merck Sharp & Dohme Corp. Erk inhibitors
CN108617166A (en) * 2015-10-21 2018-10-02 大冢制药株式会社 Kinases inhibitor benzolactam compounds
CN106928216A (en) * 2015-12-31 2017-07-07 中国科学院上海药物研究所 Compound, Preparation Method And The Use with ERK kinase inhibiting activities

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
DEZHONG JI ET AL.: ""Discovery of potent, orally bioavailable ERK1/2 inhibitors with isoindolin-1-one structure by structure-based drug design"", 《EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY》 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022226665A1 (en) * 2021-04-30 2022-11-03 Ontario Institute For Cancer Research (Oicr) Halo-substituted amino aza-heteroaryl compounds as inhibitors of the haematopoietic progenitor kinase 1 (hpk1)

Also Published As

Publication number Publication date
CN111057048B (en) 2022-03-18

Similar Documents

Publication Publication Date Title
CN112300194B (en) Condensed ring pyridone compounds, preparation method and application
CN111153901B (en) Nitrogen-containing fused heterocyclic SHP2 inhibitor compound, preparation method and application
CN112142735B (en) Condensed cyanopyridine compound, preparation method and application
CN110950876B (en) Furanolactam compounds, preparation method and application
CN112457326B (en) Aromatic heterocyclic lactam compound, preparation method and application
WO2016173557A1 (en) Compound having kinase inhibition activity, and preparation method and uses
CN110964012B (en) Fused heterocyclic biaryl benzyl alcohol compound, preparation method and application
CN109721600B (en) Nitrogen-containing fused ring compounds and preparation method and application thereof
WO2016192630A1 (en) Compound having kinase inhibiting activity, method of preparing same, and use of same
CN113061132A (en) Condensed ring lactam compound, preparation method and application
CN111518100A (en) Cyclopropenoarylbenzofuran substituted nitrogen heteroaryl compound and application thereof
CN112300196A (en) Piperidine condensed ring compound, preparation method and application
CN110655520A (en) Pyrimido-cyclic compounds, process for their preparation and their use
CN113527299A (en) Nitrogen-containing condensed ring compounds, preparation method and application
CN112094269B (en) Saturated six-membered ring heterocyclic compound, preparation method and application
CN112300173B (en) Nitrogen-containing polycyclic compounds, preparation method and application
CN109790160B (en) Pyrido five-membered aromatic ring compound, preparation method and application thereof
CN112778336B (en) Nitrogen-containing condensed ring STING regulator compound, preparation method and application
CN111057048B (en) Aminopyrazine/pyridine compound, preparation method and application
CN114907350B (en) Nitrogen-containing condensed ring compound, preparation method and application
CN114805361A (en) Amino-substituted aromatic heterocyclic pyrazole compounds, preparation method and application
CN115960098A (en) Preparation method and application of nitrogen-containing fused ring compounds
CN114075196B (en) Aromatic ring lactam compound, preparation method and application thereof
CN111763217B (en) Thieno-nitrogen heterocyclic compounds, preparation method and application
CN114685487A (en) Pyrimidine heterocyclic compound, preparation method and application

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
CB02 Change of applicant information
CB02 Change of applicant information

Address after: 305, 3rd floor, 866 HaLei Road, Pudong New Area, Shanghai 200120

Applicant after: Shanghai Lingda biomedical Co., Ltd

Address before: No.888 Zhujiang Road, juegang Town, Rudong County, Nantong City, Jiangsu Province

Applicant before: RUDONG LINGDA BIOMEDICAL TECHNOLOGY Co.,Ltd.

SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant