CN115960098A - Preparation method and application of nitrogen-containing fused ring compounds - Google Patents

Preparation method and application of nitrogen-containing fused ring compounds Download PDF

Info

Publication number
CN115960098A
CN115960098A CN202111053145.4A CN202111053145A CN115960098A CN 115960098 A CN115960098 A CN 115960098A CN 202111053145 A CN202111053145 A CN 202111053145A CN 115960098 A CN115960098 A CN 115960098A
Authority
CN
China
Prior art keywords
group
membered
alkyl
compound
ring
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
CN202111053145.4A
Other languages
Chinese (zh)
Inventor
万惠新
王亚周
查传涛
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.)
Shanghai Lingda Biomedical Co ltd
Original Assignee
Shanghai Lingda Biomedical 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 Lingda Biomedical Co ltd filed Critical Shanghai Lingda Biomedical Co ltd
Publication of CN115960098A publication Critical patent/CN115960098A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4375Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having nitrogen as a ring heteroatom, e.g. quinolizines, naphthyridines, berberine, vincamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • 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
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • 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 preparation method and application of a nitrogen-containing fused ring compound, in particular to a nitrogen-containing fused ring compound shown as a general formula I, or pharmaceutically acceptable salt thereof, or enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof, a preparation method and pharmaceutical application thereof, wherein the definition of each group is described in the specification.

Description

Preparation method and application of nitrogen-containing fused ring compounds
Technical Field
The invention belongs to the field of pharmaceutical chemistry, and particularly relates to a nitrogen-containing fused ring compound which has the activity of inhibiting methionine adenosyltransferase (MAT 2 a) and can be used for preparing a medicament for treating and preventing diseases related to the activity or expression of MAT2a or MTAP.
Background
Methionine Adenosyltransferase (MAT), also known as S-adenosylmethionine synthetase, is a cellular enzyme that catalyzes the synthesis of S-adenosylmethionine (SAM or AdoMet) from methionine and ATP and is considered to be the rate-limiting step of the methionine cycle. SAM is a propylamino donor in polyamine biosynthesis and is the major methyl donor for DNA methylation and it is involved in gene transcription and cell proliferation as well as the production of secondary metabolites. The proliferation and metastasis of tumor cells are abnormally dependent on methionine through a pharmacological method, and the inhibition of methionine circulation can obviously inhibit the proliferation and metastasis of tumor stem cells.
Methyl adenosine-thiophosphorylase (MTAP) is involved in the methionine salvage synthesis pathway, metabolizing methionine adenosine (MTA) to adenine and methionine. MTAP is located on chromosome 9p21, is similar to cancer suppressor gene CDKN2A, and MTAP defect exists in leukemia, glioma, melanoma, lung cancer, ovarian cancer, endometrial cancer, breast cancer and other tumors. Among them, the deletion rate in brain glioma was 41%, the deletion rate in mesothelioma was 31%, and the deletion rate in pancreatic cancer was 26%. S-adenosyl-L-methionine (SAM) is an enzyme cofactor that can participate in the transmethylation reaction and polyamine biosynthesis, and can be generated by the reaction of ATP and L-methionine catalyzed by methionine adenosyltransferase family (MAT) protein. In mammalian tissues, there are two distinct isoenzymes in the MAT gene, encoded by MAT1a and MAT2a, respectively. MAT1a is expressed only in liver tissues of adults, has liver specificity, and its main function is to promote SAM synthesis. MAT2a is expressed in all non-liver tissues and its primary function is to inhibit SAM synthesis. MAT2a is a key enzyme in the adenosylmethionine (SAM) synthesis pathway, and studies have shown that the up-regulation of MAT2a expression is present in a variety of cancer cells and knocking out the MAT2a gene can lead to cancer cell death, with MTAP-deficient tumors being most sensitive. Thus, MAT2a is a potential therapeutic target for MTAP-deficient tumors. The discovery and search of MAT2a inhibitor with novel structure and excellent druggability become a great hot spot for developing MTAP-deficient tumor treatment drugs.
Disclosure of Invention
One of the technical problems to be solved by the invention is to provide a novel MAT2a inhibitor for preparing a tumor treatment medicament.
The scheme for solving the technical problems is as follows:
a nitrogen-containing fused ring compound shown as a general formula I, or pharmaceutically acceptable salt thereof, or enantiomer, diastereoisomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof,
Figure BDA0003251213670000021
in the formula:
R 1 and R 3 Independently selected from 5-12 membered monocyclic or bicyclic aromatic or heteroaryl ring, said aromatic or heteroaryl ring may be substituted with 1-3 different substituents Rn, said Rn is selected from hydrogen, deuterium, halogen, cyano, nitro, amide, sulfonamide, hydroxy, amino, ureido, phosphoryl, alkylphosphoxy, alkylsilyl, C 1 -C 6 Alkyl radical, C 1 -C 6 Alkoxy radical, C 1 -C 6 Haloalkyl, C 1 -C 6 HaloalkoxyBase, C 1 -C 6 Monoalkylamino group, C 1 -C 6 Dialkylamino, alkenyl, alkynyl, 3-8 membered cycloalkyl or heterocycloalkyl, C 1 -C 6 alkyl-S-, C 1 -C 6 alkyl-SO-, C 1 -C 6 alkyl-SO 2 -and the like; or the two Rn can form a 3-12 membered saturated or partially unsaturated or aromatic ring system through a carbon chain or a heteroatom;
R 2 independently selected from hydrogen, deuterium, halogen, cyano, hydroxy, nitro, amino, C 1 -C 12 Alkyl radical, C 1 -C 6 Haloalkyl, C 1 -C 6 alkyl-S-, C 1 -C 6 alkyl-SO-, C 1 -C 6 alkyl-SO 2 -、C 1 -C 6 alkyl-O-, C 1 -C 6 haloalkyl-O-, C 1 -C 6 Monoalkylamino radical, C 1 -C 6 Dialkylamino, 3-12 membered cycloalkylamino or heterocycloalkylamino, 3-12 membered cycloalkyl or heterocycloalkyl, 3-12 membered halocycloalkyl or haloheterocycloalkyl, 3-12 membered cycloalkyl-O-, 3-12 membered halocycloalkyl-O-, 3-12 membered heterocycloalkyl-O-, 5-12 membered aryl or 5-12 membered heteroaryl;
w, X, Y is each independently selected from CR 4 Or N; wherein R is 4 Each independently selected from hydrogen, deuterium, halogen, cyano, hydroxy, nitro, amino, C 1 -C 12 Alkyl radical, C 1 -C 6 Haloalkyl, C 1 -C 6 alkyl-S-, C 1 -C 6 alkyl-SO-, C 1 -C 6 alkyl-SO 2 -、C 1 -C 6 alkyl-O-, C 1 -C 6 haloalkyl-O-, C 1 -C 6 Monoalkylamino group, C 1 -C 6 Dialkylamino, 3-12 membered cycloalkylamino or heterocycloalkylamino, 3-12 membered cycloalkyl or heterocycloalkyl, 3-12 membered halocycloalkyl or haloheterocycloalkyl, 3-12 membered cycloalkyl-O-, 3-12 membered halocycloalkyl-O-, 3-12 membered heterocycloalkyl-O-, 5-12 membered aryl or 5-12 membered heteroaryl; or-X = Y-may be independently selected from-O-or-S-.
Any of the above groupsOne or more hydrogen atoms of (a) may be substituted with a substituent selected from the group consisting of: including but not limited to deuterium, halogen, C 1 -C 8 Alkyl radical, C 3 -C 8 Cycloalkyl, amino, C 1 -C 8 An alkylamino group; wherein said heteroaryl group contains 1 to 3 heteroatoms selected from the group consisting of: n, O, P or S, said heterocycloalkyl comprising 1-3 heteroatoms selected from the group consisting of: n, O, P or S, said ring system comprising a spiro, bridged, fused or partially unsaturated ring system.
In some preferred embodiments, the compound having the general formula (I), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsomer, solvate, polymorph or prodrug thereof, preferably is a compound represented by the general formula (II), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsomer, solvate, polymorph or prodrug thereof:
Figure BDA0003251213670000031
wherein R is 5 、R 6 、R 7 Each independently selected from hydrogen, deuterium, halogen, cyano, hydroxy, nitro, amino, C 1 -C 12 Alkyl radical, C 1 -C 6 Haloalkyl, C 1 -C 6 alkyl-S-, C 1 -C 6 alkyl-SO-, C 1 -C 6 alkyl-SO 2 -、C 1 -C 6 alkyl-O-, C 1 -C 6 haloalkyl-O-, C 1 -C 6 Monoalkylamino group, C 1 -C 6 A dialkylamino, 3-12 membered cycloalkylamino or heterocycloalkylamino, 3-12 membered cycloalkyl or heterocycloalkyl, 3-12 membered halocycloalkyl or haloheterocycloalkyl, 3-12 membered cycloalkyl-O-, 3-12 membered halocycloalkyl-O-, 3-12 membered heterocycloalkyl-O-, 5-12 membered aryl or 5-12 membered heteroaryl; r 1 、R 2 、R 3 The definitions of (a) and (b) are as indicated above.
In some preferred embodiments, it is preferably a compound of formula (III), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsiomer, solvate, polymorph or prodrug thereof:
Figure BDA0003251213670000032
wherein: m 1 Preferably selected from CH or N, M 2 Preferably selected from O, S, NH, etc.;
Figure BDA0003251213670000033
preferably from single or double bonds; r 8 Preferably selected from hydrogen, deuterium, halogen, cyano, C 1 -C 6 Alkyl radical, C 1 -C 6 Haloalkyl, C 1 -C 6 Alkoxy radical, C 1 -C 6 Haloalkoxy, 3-8 membered cycloalkyl or heterocycloalkyl; r 3 Preferably selected from the group consisting of benzene ring, pyridine ring, thiazole ring, imidazole ring, indole ring, indazole ring, indoline group, isoindoline ring, benzofuran group, dihydrofuran group, pyridofuran group, benzimidazolyl group, benzoxazolyl group, benzothiazolyl group, pyridoimidazolyl group, pyridooxazolyl group, pyridothiazolyl group, naphthalene ring, quinoline ring, isoquinoline ring, quinazoline Lin Huanji, benzomorphine group, benzodioxanyl group and the like, and the R is selected from the group consisting of 3 The ring may be substituted with one or more groups selected from: hydrogen, deuterium, halogen, cyano, C 1 -C 6 Alkyl radical, C 1 -C 6 Haloalkyl, C 1 -C 6 Alkoxy radical, C 1 -C 6 Haloalkoxy, 3-8 membered cycloalkyl or heterocycloalkyl; r 2 、R 3 W, X, Y are as defined above.
In some preferred embodiments, R 1 Preferably selected from the group consisting of 5-12 membered monocyclic aryl or heteroaryl rings, said aryl or heteroaryl rings may be substituted with 1-3 substituents Rn, said Rn selected from the group consisting of hydrogen, deuterium, halogen, C 1 -C 6 Alkyl, aryl, heteroaryl, and heteroaryl,C 1 -C 6 Alkoxy radical, C 1 -C 6 Haloalkyl, C 1 -C 6 Haloalkoxy, C 1 -C 6 Monoalkylamino group, C 1 -C 6 A di-alkylamino group; or the two Rn can form a 3-6-membered saturated ring system through a carbon chain or a heteroatom.
In some preferred embodiments, R 1 Preferably selected from 5-6 membered monocyclic aryl or heteroaryl rings, which aryl or heteroaryl rings may be substituted with 1-3 substituents Rn, wherein Rn is selected from halogen, C 1 -C 6 Haloalkyl, C 1 -C 6 A haloalkoxy group.
In some preferred embodiments, R 2 Preferably selected from hydrogen, deuterium, halogen, C 1 -C 6 Alkyl radical, C 1 -C 6 Haloalkyl, C 1 -C 6 alkyl-O-, C 1 -C 6 haloalkyl-O-, C 1 -C 6 Monoalkylamino group, C 1 -C 6 Dialkylamino, C 1 -C 6 Monohaloalkylamino, C 1 -C 6 Bishaloalkylamino, C 1 -C 6 alkyl-S-.
In some preferred embodiments, R 2 Preferably selected from halogen, C 1 -C 6 Alkyl radical, C 1 -C 6 Haloalkyl, C 1 -C 6 alkyl-O-, C 1 -C 6 haloalkyl-O-, -NR a R b 、C 1 -C 6 Monohaloalkylamino, C 1 -C 6 Bishaloalkylamino, C 1 -C 6 alkyl-S-in which R a And R b Each independently selected from hydrogen, deuterium, C 1 -C 6 Alkyl radical, C 1 -C 6 A haloalkyl group.
In some preferred embodiments R 2 Preferably selected from C 1 -C 6 alkyl-O-, -NR a R b Wherein R is a And R b Each independently selected from hydrogen, deuterium, C 1 -C 6 Alkyl radical, C 1 -C 6 A haloalkyl group.
In some preferred embodiments, R 3 Preferably selected from 5-12 membered (e.g. 5,6,7,8,9,10, 11, 12) monocyclic or bicyclic aryl or heteroaryl rings, which aryl or heteroaryl rings may be substituted by 1-3 substituents Rn as defined herein.
In some preferred embodiments, R 3 Preferably selected from phenyl, indazolyl, quinolinyl, benzimidazolyl, benzothiazolyl, which may be substituted with 1-3 substituents Rn as defined herein.
In some preferred embodiments, R 3 Preferably selected from 5-12 membered bicyclic aryl or heteroaryl rings, which may be substituted with 1-3 substituents Rn as described herein.
In some preferred embodiments, R 3 Preferably selected from a 5-12 membered bicyclic aryl or heteroaryl ring, which may be substituted with 2-3 substituents Rn, wherein two substituents Rn may be adjacent and the two adjacent Rn's may form a substituted or unsubstituted 3-6 membered (e.g. 3,4, 5, 6) ring via a carbon chain or heteroatom.
In some preferred embodiments, rn is preferably selected from hydrogen, deuterium, halogen, C 1 -C 6 Alkyl radical, C 1 -C 6 Alkoxy radical, C 1 -C 6 Haloalkyl, C 1 -C 6 Deuterated alkyl, C 1 -C 6 Deuterated alkoxy, alkenyl, alkynyl, C 1 -C 6 alkyl-S-, C 1 -C 6 alkyl-SO-, C 1 -C 6 alkyl-SO 2 -。
In some preferred embodiments, two adjacent Rn may form a 5-6 membered saturated or partially unsaturated or aromatic ring system, preferably a 5-6 membered saturated carbocyclic or saturated heterocyclic ring, through a carbon chain or 1-3 heteroatoms selected from N, O, S, wherein one or more atoms in the saturated carbocyclic or saturated heterocyclic ring may be further oxidized to form = O.
In some preferred embodiments, W, X, Y, R, R2, and R3 are each independently the corresponding group in compounds 1-94, prepared in the examples.
In some preferred embodiments, the compound is any one of compounds 1-94 prepared in the examples, or a pharmaceutically acceptable salt thereof.
A first process for preparing a compound of formula I, said process essentially comprising the steps of a:
a. the compound of the general formula (A) and aryl acetic acid or aryl acetyl chloride or aryl acetate are subjected to dehydration and ring closure reaction under the conditions of acid or base catalysis or condensing agent to generate the compound of the general formula (I);
Figure BDA0003251213670000051
wherein Ra is hydroxyl, chlorine, ester group; rb is hydrogen or alkyl; r is 1 、R 2 、R 3 W, X, Y are as defined above.
A second process for preparing a compound of formula I, said process essentially comprising the following steps b;
b. reacting a compound of formula (B) with an aryl keto acid or aryl keto ester under acid or base catalyzed conditions to produce a compound of formula (I):
Figure BDA0003251213670000052
wherein Rc is hydroxyl or an ester group; r 1 、R 2 、R 3 X, Y are as defined above.
A third process for preparing a compound of formula I, said process essentially comprising the following steps c and d:
c, carrying out ring closure on the compound of the general formula (C) and 2-bromophosphoryl acetate (D) under the catalysis of alkali to generate an intermediate compound of the general formula (E);
d: carrying out coupling reaction on the compound of the general formula (E) and substituted aryl boric acid (or ester), aryl tin reagent or aryl silicon reagent by catalysis of a transition metal complex to generate a compound of the general formula (I);
Figure BDA0003251213670000053
wherein Ra is hydroxyl, chlorine, alkoxy, ester group; rb is hydrogen or alkyl; rd is phosphoryl ester; r 1 、R 2 、R 3 W, X, Y are as defined above.
Preferably, said step is 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, methylene chloride, 1,2-dichloroethane, acetonitrile, N-dimethylformamide, N-dimethylacetamide, dioxane, 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 hexamethyldisilyl, sodium hexamethyldisilyl, lutidine, or combinations thereof.
Preferably, the acid is selected from the group consisting of: hydrochloric acid, sulfuric acid, phosphoric acid, methanesulfonic acid, toluenesulfonic acid, trifluoroacetic acid, formic acid, acetic acid, trifluoromethanesulfonic acid, or combinations 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, palladium dichlorobis (triphenylphosphine) palladium, palladium trifluoroacetate, palladium triphenylphosphine acetate, [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride, bis (trifluorophenylmethylphosphino) 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-phenylmethylphosphine, tricyclohexylphosphine, tri-o-phenylmethylphosphine, or a combination thereof.
The invention provides a class of preferred compounds of general formula (I), including but not limited to the following structures:
Figure BDA0003251213670000061
/>
Figure BDA0003251213670000071
/>
Figure BDA0003251213670000081
and the compound represented by the above general formula (I) does not contain the following structure:
Figure BDA0003251213670000082
another objective of the invention is to provide a medicament for treating or preventing tumor or autoimmune diseases and a composition thereof. The technical scheme for realizing the purpose is as follows:
a pharmaceutical composition for treating or preventing tumor or autoimmune disease, which comprises the nitrogen-containing fused ring compound shown in the general formula (I), or pharmaceutically acceptable salt thereof, or enantiomer, diastereomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof and pharmaceutically acceptable carrier.
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:
the nitrogen-containing fused ring compound shown in the general formula (I) or pharmaceutically acceptable salt thereof, or enantiomer, diastereoisomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof is used for preparing medicaments for treating diseases related to the activity or expression of MAT2a or MTAP protein, in particular medicaments for preventing or treating tumors or autoimmune diseases. The tumor is independently selected from lung cancer, pancreatic cancer, liver cancer, colorectal cancer, bile duct cancer, gallbladder cancer, brain cancer, gastric cancer, leukemia, lymph cancer, melanoma, thyroid cancer, nasopharyngeal cancer, glioma, bladder cancer, astrocytoma, basal cell carcinoma, osteosarcoma, head and neck cancer, chondrosarcoma, ovarian cancer, endometrial cancer, breast cancer, soft tissue sarcoma, mesothelioma and the like; the autoimmune disease is independently selected from the group consisting of thyroiditis, inflammatory enteritis, erythematous pennisetum, fibrosis, muscle weakness, vasculitis, psoriasis, arthritis, scleroderma, dermatitis, and the like.
The invention relates to a compound with the structural characteristics of a general formula (I), which can inhibit the enzyme activity of MAT2a and obviously inhibit the growth of various tumor cells, particularly tumor cells related to MTAP deletion, and is a treatment medicament 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
Through long-term and intensive research, the inventor prepares a nitrogen-containing fused ring compound with a novel structure shown in formula I, and finds that the nitrogen-containing fused ring compound has better activity for inhibiting MAT2a enzyme, and the compound has specific inhibition effect on MAT2a protein under extremely low concentration (which can be as low as less than 100 nM), and has quite excellent cell proliferation inhibition activity related to MTAP deletion. Based on the above findings, the inventors have completed the present invention.
Term(s) for
Unless defined otherwise, all technical and scientific terms 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.
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 terminology used herein in the pertinent description of analytical chemistry, organic synthetic chemistry, and pharmaceutical and medicinal chemistry is 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 2 O-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 does not include carbons that may be present in a substituent of the group.
In addition to the foregoing, when used in the specification and claims of this application, the following terms take the meanings indicated below, unless otherwise specifically indicated.
In this 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" means a-C (= O) -group; "Nitro" means-NO 2 (ii) a "cyano" means-CN; "amino" means-NH 2 (ii) a "substituted amino" refers to an amino group substituted with one or two alkyl, alkylcarbonyl, aralkyl, heteroaralkyl groups as defined below, e.g., monoalkylamino, dialkylamino, alkylamido, aralkylamino, heteroaralkylamino, wherein said substituted amino group may be further substituted with a substituent selected from the group consisting of halogen, C1-C4 alkyl, C1-C4 alkoxy, amino, cyano, alkylamino; "carboxyl" means-COOH.
In this 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 of only 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, 2,2-dimethylpropyl, n-hexyl, heptyl, 2-methylhexyl, 3-methylhexyl, octyl, nonyl, and decyl, and the like.
In this application, the term "alkenyl" as a group or part of another group means a straight or branched hydrocarbon chain group consisting of only carbon 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 attached 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-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 of carbon atoms and hydrogen atoms only, 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 this application, the term "haloalkyl" as a group or part of another group means an alkyl group as described above, e.g. fluoroalkyl, chloroalkyl, substituted with 1 to 3 halogen atoms, the alkyl group including any number of carbon atoms, e.g. methyl, ethyl, propyl, the haloalkyl including but not limited to trifluoromethyl.
In this application, the term "alkoxy" as a group or as part of another group means an alkyl-O-, such as methoxy, ethoxy.
In this application, the term "haloalkoxy" as a group or part of another group means an alkoxy group as described above substituted with 1 to 3 halogen atoms, such as trifluoromethoxy and the like.
In this application, the term "deuterated alkyl" as a group or as part of another group means an alkyl group as described above substituted with 1-3 deuterium atoms.
In the present application, the term "alkylamino" as a group or as part of another group means-alkyl-NH 2 Structural or substituted amino-NR a R b Wherein R is a And R b Each independently hydrogen or an alkyl group as described above. For example, the term "monoalkylamino" refers to a substituted amino-NR a R b Wherein R is a And R b One is hydrogen and the other is alkyl as described above; "Bialkylamino" refers to a substituted amino-NR a R b Wherein R is a And R b Each independently an alkyl group as described above.
As a group or as part of another group in the present applicationIn each case, the term "haloalkylamino" means-alkyl-NH 2 Structural or substituted amino-NR a R b Wherein R is a And R b Each independently hydrogen or haloalkyl as described above. For example, the term "monohaloalkylamino" refers to a substituted amino-NR a R b Wherein R is a And R b One is hydrogen and the other is haloalkyl as described above; "Bihaloalkyl alkylamino" refers to a substituted amino-NR a R b Wherein R is a And R b Each independently a haloalkyl group as described above.
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 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 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, 7,7-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, octa-yl, octa-methyl-5364H-cyclopentadienyl, 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 heterocyclyl group may be a monocyclic, bicyclic, tricyclic, or higher ring system, which may include fused, bridged, or spiro ring systems; wherein the nitrogen, carbon or sulfur atom in the heterocyclic group thereof 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, phthalimido, 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, orthophenanthrene, isoxazolyl, phenoxazinyl, phenothiazinyl, 8652 zft 8652-tetrahydrobenzo [ b ] thienyl, naphthopyridyl, [ 3265 zft 3265 ] triazolo [ 3579-b ] pyridazine, [ 3525-355627 ] triazolo [ 353257 ] triazolo [ 353279-3257 ] triazolo [ 353257 ] pyridazine, 343257-35xzft [ 353257-3282 ] triazolo [ 353257 ] pyridiz [ 355632-b ] triazolo [ 5632 ] pyridiz [ 343282 ] pyrid-3282 a, etc.
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/separating the individual isomers include chiral synthesis from suitable optically pure precursors, or resolution of the racemates (or racemates of salts or derivatives) using, for example, chiral high performance liquid chromatography.
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, formate, acetate, 2,2-dichloroacetate, trifluoroacetate, propionate, hexanoate, octanoate, decanoate, undecenate, 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. 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 the advantages of solubility, histocompatibility, or sustained release in mammalian organisms. Prodrugs include known amino protecting groups and carboxyl protecting groups.
In the present application, "pharmaceutical composition" refers to a formulation of a compound of the present invention with a vehicle generally accepted in the art for delivering 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 a living body, facilitate absorption of the active ingredient, 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;
(iii) Alleviating the disease or condition, i.e., causing the state of the disease or condition to resolve; 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. 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 intermediate compound functional groups may need to be protected by appropriate protecting groups. Such functional groups include hydroxyl, amino, mercapto and carboxylic acid. Suitable hydroxyl protecting groups include trialkylsilyl or diarylalkylsilyl groups (e.g.t-butyldimethylsilyl, t-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 protecting group may also be a polymeric resin.
The main advantages of the invention include:
the compound has excellent methionine adenosyltransferase (MAT 2 a) inhibition effect, and IC50 values are all less than 200nM, and can be less than 10nM at the lowest.
The invention will be further illustrated with reference to the following 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. The experimental procedures, in which specific conditions are not noted in the following examples, are generally carried out under conventional conditions or conditions recommended by the manufacturers. Unless otherwise indicated, percentages and parts are by weight.
Examples preparation
Example 1: 7-methoxy-3- (4-methoxyphenyl) -1-phenyl-1,8-naphthyridin-2 (1H) -one
Figure BDA0003251213670000161
/>
The first step is as follows: under the protection of nitrogen, palladium (Pd) tris-dibenzylideneacetone is added 2 (dba) 3 ) (477mg, 0.520mmol), 4,5-bis (diphenylphosphine) -9,9-dimethylxanthene (Xantphos) (301mg, 0.520mmol) were added in succession to a solution of 2-chloro-6-methoxynicotinaldehyde (890mg, 5.204mmol), aniline (484mg, 5.204mmol) and cesium carbonate (5.09g, 15.613mmol) in 1,4-dioxane (20 mL). The reaction mixture was heated to 120 ℃ and reacted at this temperature for 2 hours. The reaction solution was diluted with Ethyl Acetate (EA) (100 mL), filtered through celite, and the filtrate was concentrated under reduced pressure and column-chromatographed (petroleum ether: ethyl acetate =10: 1) to give an intermediate (1.02 g, crude) as a pale yellow oil. LC-MS (ESI) m/z of 229.0[ 2] M + H] +1 H-NMR(400MHz,DMSO-d 6 )δ10.87(s,1H),9.75(s,1H),8.06(d,J=8.4Hz,1H),7.76(d,J=7.8Hz,2H),7.38(t,J=7.9Hz,2H),7.09(t,J=7.4Hz,1H),6.37(d,J=8.4Hz,1H),3.95(s,3H)。
The second step is that: sodium hydride (NaH) (105mg, 2.628mmol) was added to a solution of the above intermediate (200mg, 0.876mmol) in Tetrahydrofuran (THF) (5 mL) while cooling in an ice-water bath. After the reaction was warmed to room temperature for half an hour, 2- (4-methoxyphenyl) acetyl chloride (178mg, 0.964mmol) was added dropwise. After the reaction mixture was allowed to react at room temperature for 2 hours, the reaction mixture was diluted with ethyl acetate (100 mL), and then washed with water (50 mL) and with an aqueous solution of sodium chloride (50 mL). The organic phase was collected and dried over anhydrous sodium sulfate, concentrated and the crude product isolated as a white solid to give the compound of example 1 (16.8 mg). LC-MS (ESI) m/z:359.2[ 2], [ M + H ]] + .1H NMR(400MHz,DMSO-d 6 )δ8.14(t,J=4.2Hz,2H),7.75-7.64(m,2H),7.54(t,J=7.5Hz,2H),7.49-7.43(m,1H),7.34(dd,J=5.2,3.3Hz,2H),7.04-6.95(m,2H),6.73(d,J=8.4Hz,1H),3.80(s,3H),3.48(s,3H)。
Examples 2 to 5
Synthesized according to the method of example 1 to obtain the compounds of examples 2-5;
Figure BDA0003251213670000171
example 6: 6-methoxy-2- (4-methoxyphenyl) -4-phenylpyridine [2,3-b ] pyrazin-3 (4H) -one
Figure BDA0003251213670000172
The first step is as follows: under the protection of nitrogen, pd 2 (dba) 3 (984mg, 1.08mmol), xantphos (622mg, 1.08mmol) were added to 2-chloro-6-methoxy-3-nitropyridine (4.04g, 21.42mmol), aniline (1.0g, 10.75mmol) and cesium carbonate (Cs) in that order 2 CO 3 ) (10.5g, 32.22mmol) in 1,4-dioxane (50 mL). The reaction mixture was heated to 120 ℃ and the reaction was continued at this temperature for 2 hours. The reaction solution was diluted with ethyl acetate (100 mL), then filtered with celite, and the filtrate was concentrated under reduced pressure and purified by column chromatography (petroleum ether: ethyl acetate = 20). LC-MS (ESI) m/z:246.0[ 2] M + H] +1 H NMR(400MHz,DMSO-d6)δ10.46(s,1H),8.44(d,J=9.1Hz,1H),7.72(d,J=7.7Hz,2H),7.40(t,J=7.9Hz,2H),7.17(t,J=7.4Hz,1H),6.38(d,J=9.1Hz,1H),3.88(s,3H)。
The second step is that: the intermediate (3.65g, 14.88mmol) and palladium on carbon catalyst (365mg, 3.44mmol) were added to methanol (200 mL) under nitrogen. After replacement of hydrogen, the reaction was carried out overnight at room temperature under a hydrogen atmosphere of 1 atm. The reaction solution was filtered through celite, and the filtrate was concentrated and purified by column chromatography (petroleum ether/ethyl acetate volume ratio 20 to 10. LC-MS (ESI) m/z:216.1[ 2], [ M + H ]] +1 H-NMR(400MHz,DMSO-d6)δ7.72(s,1H),7.65(d,J=7.7Hz,2H),7.23(dd,J=8.4,7.5Hz,2H),6.98(d,J=8.1Hz,1H),6.84(t,J=7.3Hz,1H),6.07(d,J=8.1Hz,1H),4.51(s,2H),3.73(s,3H)。
The third step: acetic acid (0.6 mL) was added to a solution of the above intermediate (300mg, 1.395mmol) and ethyl 4-methoxyphenyl-2-acetoacetate (435mg, 2.092mmol) in ethanol (10 mL) at room temperature.After the tube was sealed, the reaction was continued for 2 hours after the reaction was raised to 100 ℃. After completion of the reaction, the reaction solution was diluted with methylene chloride (100 mL), washed with water (50 mL) and then with an aqueous solution of sodium chloride (50 mL). The organic phase was collected, dried over anhydrous sodium sulfate and filtered, and the filtrate was concentrated. Preparative separation by HPLC afforded example 2 (light yellow solid, 68.2 mg). LC-MS (ESI) m/z:360.0[ 2], [ M + H ]]+. 1 H NMR(400MHz,DMSO-d6)δ8.35-8.26(m,2H),8.21(d,J=8.6Hz,1H),7.65-7.54(m,2H),7.50(m,1H),7.43(m,2H),7.12-6.95(m,2H),6.83(d,J=8.6Hz,1H),3.84(s,3H),3.53(s,3H)。
Examples 7 to 10
Synthesized according to the method of example 6 to give the compounds of examples 7-10:
Figure BDA0003251213670000181
examples 11 to 16
Synthesized according to the procedure of example 1 to give the compounds of examples 11 to 16:
Figure BDA0003251213670000182
Figure BDA0003251213670000191
example 17: 7-methoxy-3- (4-methoxyphenyl) -4-methyl-1-phenyl-1,8-naphthyridin-2 (1H) -one
Figure BDA0003251213670000192
The first step is as follows: under the protection of nitrogen, pd 2 (dba) 3 (411mg, 0.449mmol), xantphos (260mg, 0.449mmol) were added sequentially to a solution of methyl 2-chloro-6-methoxy-nicotinate (902mg, 4.49mmol), aniline (418mg, 4.49mmol) and cesium carbonate (4.39g, 13.46mmol) in 1,4-dioxane (20 mL). The reaction mixture is heated toThe reaction was continued at 120 ℃ for 2 hours. The reaction solution was diluted with ethyl acetate (100 mL), then filtered with celite, and the filtrate was concentrated under reduced pressure and purified by column chromatography (petroleum ether: ethyl acetate = 10). LC-MS (ESI) m/z 259.0[ 2] M + H] + .1H NMR(400MHz,DMSO-d6)δ10.37(s,1H),8.13(d,J=8.6Hz,1H),7.71(d,J=7.8Hz,2H),7.35(t,J=7.8Hz,2H),7.05(t,J=7.3Hz,1H),6.27(d,J=8.6Hz,1H),3.91(s,3H),3.85(s,3H)。
The second step is that: lithium bis (trimethylsilyl) amide (3.34mL, 3.34mmol) was added to a solution of the above intermediate (430mg, 1.67mmol) in tetrahydrofuran (20 mL) on an ice bath, and the reaction mixture was reacted at this temperature for 2 hours. Paramethoxybenzeneacetyl chloride (400mg, 2.17mmol) was added to the mixture and the reaction mixture was allowed to react at this temperature for an additional 2 hours. LC-MS detects the completion of the reaction. The reaction mixture was diluted with ethyl acetate (200 mL), the pH was adjusted to neutral with 1N aqueous hydrochloric acid, and the organic phase was washed with aqueous ammonium chloride. The separated organic phase was dried over anhydrous sodium sulfate, the filtered filtrate was concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (petroleum ether: ethyl acetate volume ratio 1:1) to give a pale yellow solid intermediate (250 mg). LC-MS (ESI) m/z:375.0[ m ] +H] + . 1 H NMR(400MHz,DMSO-d6)δ10.29(br.s,1H),8.29(d,J=8.6Hz,1H),7.56-7.46(m,2H),7.45-7.38(m,1H),7.30(m,4H),7.00-6.92(m,2H),6.70(d,J=8.6Hz,1H),3.79(s,3H),3.47(s,3H)。
The third step: trifluoromethanesulfonic anhydride (738mg, 2.619mmol) was added to a pyridine (8 mL) solution of the above intermediate (2458 mg, 0.655mmol) under nitrogen, and the reaction mixture was reacted at 70 ℃ for 16 hours. LCMS check reaction complete. The reaction was diluted with dichloromethane (200 mL) and the organic phase was washed with aqueous ammonium chloride. The separated organic phase was dried over anhydrous sodium sulfate, the filtrate was concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (petroleum ether: ethyl acetate = 10). LCMS (ESI) m/z:508.9[ 2], [ M + H ]] + . 1 H NMR(400MHz,DMSO-D6)δ8.05(d,J=8.7Hz,1H),7.59-7.53(m,2H),7.51-7.45(m,1H),7.42(m,4H),7.07-7.00(m,2H),6.95(d,J=8.7Hz,1H),3.81(s,3H),3.52(s,3H)。
The fourth step: nitrogen gasTetrakis (triphenylphosphine) palladium (Pd (PPh) under protection 3 ) 4 ) (28mg, 0.0247 mmol) was added to the above intermediate (120mg, 0.237mmol), methylboronic acid (142mg, 2.37mmol) and potassium phosphate (151mg, 0.711mmol) in 1,4-dioxane (20 mL). The reaction mixture was reacted at 100 ℃ for 2 hours. Disappearance of the raw material was detected by LC-MS. The reaction solution was diluted with ethyl acetate (200 mL), and the organic phase was washed with aqueous sodium chloride solution. The separated organic phase was dried over anhydrous sodium sulfate, the filtrate was concentrated, and the crude product was isolated by preparative separation to give a white solid product (18 mg). LC-MS (ESI) m/z:373.2[ 2], [ M + H ]] + . 1 H NMR(400MHz,DMSO-d6)δ8.21(d,J=8.7Hz,1H),7.52(m,2H),7.43(m,1H),7.35-7.26(d,J=8.4Hz,2H),7.22(d,J=8.7Hz,2H),6.98(d,J=8.7Hz,2H),6.74(d,J=8.6Hz,1H),3.80(s,3H),3.47(s,3H),2.31(s,3H)。
Example 18:1- (4-Chlorobenzene) -3- (2-methyl-2H-indazol-5-yl) -7- ((2,2,2-trifluoroethyl) amino) -1,8-naphthyridin-2 (1H) -one
Figure BDA0003251213670000201
The first step is as follows: 6-chloro-2- ((4-chlorophenyl) amino) nicotinaldehyde (3.4 g, 12.8mmol), triethyl 2-bromo-2-phosphorylacetate (5.1g, 16.6mmol), DBU (2.9g, 19.2mmol), and lithium chloride (965mg, 23mmol) were dissolved in acetonitrile (200 mL), reacted at room temperature for 4 hours, and refluxed overnight. After the reaction mixture was concentrated, the crude product was purified by silica gel column chromatography (PE/EA = 3:1) to give a yellow solid product (970 mg). LC-MS [ M + H ]] + :m/z 368.9。
The second step is that: the above intermediate (870mg, 2.4 mmol), 2-methyl-5-indazole boronic acid pinacol ester (928mg, 3.6 mmol), dichloro [1,1' -bis (tert-butylphosphino) ferrocenepalladium (78mg, 0.1mmol) and potassium phosphate (1.5g, 7.2mmol) were dissolved in dioxane (40 mL) and water (8 mL) under nitrogen protection, and reacted at 60 ℃ for 2 hours. The reaction was concentrated and purified by silica gel column chromatography (PE/EA = 3:1) to give a yellow solid product (600 mg). LC-MS [ M + H ]] + :m/z 421.0。
The third step: the above intermediate (50mg, 0.1mmol), 2,2,2-trifluoroethylamine (120mg, 1.0mmol), DIEA (46mg, 0.4mmol) was dissolved in N-methylpyrrolidone (2 m)L), reacting for 2 hours at 200 ℃ by microwave. The reaction mixture was diluted with ethyl acetate (30 mL), washed twice with water (10 mL), and the separated organic phase was dried and concentrated. The crude product was prepared by HPLC to give the compound as a white solid (12.84 mg). LC-MS [ M + H ]] + :m/z 484.1。 1 HNMR(400MHz,DMSO-d 6 ):δ8.37(s,1H),8.08(s,2H),7.90(t,J=7.8Hz,2H),7.65-7.41(m,4H),7.39-7.18(m,2H),6.55(d,J=8.5Hz,1H),4.17(s,3H),3.77(m,2H)。
Examples 19 to 39 and 43 to 52
The compound of example 19-39, 43-52 was synthesized according to the procedure of example 18.
Figure BDA0003251213670000211
/>
Figure BDA0003251213670000221
/>
Figure BDA0003251213670000231
/>
Figure BDA0003251213670000241
Example 54:1- (4-chlorobenzene) -3- (3-methoxy-2- (deuterated methyl) -2H-indazol-5-yl) -7- ((2,2,2-trifluoroethyl) amino) -1,8-naphthyridin-2 (1H) -one
Figure BDA0003251213670000242
The first step is as follows: 4-bromo-2- (bromomethyl) -1-nitrobenzene (1.2g, 4.0 mmol) was dissolved in ethanol (50 mL), triethylamine (404.1mg, 4.0 mmol) and deuterated methylamine (2mL, 4.1mmol) were added, and the reaction was heated to 80 ℃ overnight. After the reaction solution was concentrated under reduced pressure, the crude product was purified by silica gel column chromatography (PE: EA = 5:1) to give a white solid compound (201 mg). LC-MS [ M + H ]] + :m/z 248.0/250.0。
The second step: to the above compound (50mg, 0.2mmol) in MeOH/H 2 To the O (9 mL/1 mL) mixed solution was added potassium hydroxide KOH (112mg, 2.0 mmol), and the reaction was carried out overnight at 65 ℃. The reaction solution was concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (PE: EA = 4:1) to give a white solid compound (30 mg). LC-MS [ M + H ]] + :m/z244.1/246.1。
The third step: to the above compound (100mg, 0.41mmol), pinacol diboron ester (117mg, 0.46mmol) and potassium acetate (123.4 mg, 1.26mmol) in 1,4-dioxane (20 mL) under nitrogen protection was added catalyst [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride Pd (dppf) Cl 2 (34.1mg, 0.042mmol) and reacted at 75 ℃ for 4 hours. The reaction solution was concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (PE: EA = 5:1) to give a white solid compound (60 mg). LC-MS [ M + H ]] + :m/z 292.1。
The fourth step: to the above intermediate compound (60mg, 0.2mmol), 3-bromo 7-chloro-1- (4-chlorobenzene) -1,8-naphthyridin-2 (1H) -one (80.0 mg, 0.21mmol) and potassium phosphate (127.2mg, 0.6mmol) in dioxane/H under nitrogen protection 2 Pd (dppf) Cl was added to O (12 mL/2 mL) 2 (14.1mg, 0.02mmol). The reaction solution was heated to 70 ℃ and stirred for 3 hours. The reaction was concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (DCM/MeOH = 10). LC-MS [ M + H ]] + :m/z 454.0/456.0。
The fifth step: the above intermediate compounds (60.0mg, 0.13mmol), 2,2,2-trifluoroethylamine (128.7mg, 1.3mmol) and diisopropylethylenediamine DIEA (46mg, 0.4mmol) were dissolved in NMP (2 mL) and reacted at 200 ℃ for 2 hours with microwave. The crude product was prepared in reverse phase to give the compound as a yellow solid (1.02 mg). LC-MS [ M + H ]] + :m/z 517.1。 1 H-NMR(400MHz,DMSO-d 6 ):δ8.14(s,1H),8.03(s,1H),7.96(dd,J=7.6,1.6Hz,1H),7.90(m,2H),7.52-7.58(m,3H),7.33(d,J=8.4Hz,2H),6.56(m,1H),3.78-3.81(m,2H),3.35(s,3H)。
Examples 55 to 65
Synthesized according to the procedures of examples 34, 53, 54, 66, with methylamine instead of deuterated methylamine, or deuterated methanol instead of methanol, to give the compounds of examples 55-65, 94.
Figure BDA0003251213670000251
/>
Figure BDA0003251213670000261
Example 40:1- (4-Chlorobenzene) -6-fluoro-3- (2-methyl-2H-indazol-5-yl) -7- ((2,2,2-trifluoroethyl) amino) -1,8-naphthyridin-2 (1H) -one
Figure BDA0003251213670000271
The first step is as follows: compound 4-chloroaniline (6.4 g, 50.0mmol) was dissolved in THF (60 mL), lithium hexamethyldisilazide LiHMDS (76mL, 76.0mmol) was added dropwise at-60 ℃ and after 1h of reaction, 2,6-dichloro-5-fluoronicotinic acid (5.0g, 23.8mmol) was added. After the addition, the reaction was allowed to return to room temperature for 4 hours. After quenching with water (10 mL), the pH was adjusted to 2 with 5M aqueous hydrochloric acid and extracted three times with ethyl acetate (50 mL). The combined organic phases were washed with brine, dried and concentrated, and the crude product was slurried with ethyl acetate (30 mL) to give a yellow solid intermediate product (6.1 g). LC-MS [ M + H ]] + :m/z 301.0。
The second step is that: the intermediate product (6.1g, 20.3 mmol) was dissolved in THF (40 mL), and borane-tetrahydrofuran solution (61mL, 61mmol) was added dropwise at 0 ℃ and after completion of the addition, the reaction was allowed to proceed at room temperature for 2 hours. Quenched with methanol (50 mL), stirred for half an hour and concentrated, and the crude product was purified by silica gel column chromatography (PE: EA = 10. LC-MS [ M + H ]] + :m/z 287.1。
The third step: the intermediate product (5.8 g,20.2 mmol) was dissolved in DCM (50 mL) and reacted at 0 ℃ with Dess-Martin reagent (17.2 g,40.4 mmol) for 2 hours at RT. Sequentially adding Na 2 S 2 O 3 (30 mL) and NaHCO 3 Quenched (30 mL) and extracted three times with dichloromethane (50 mL). The combined organic phases were washed with brine, dried and concentrated. The crude product was chromatographed on silica gel (PE: EA = 1)0:1) gave intermediate (2.2 g) as a yellow solid. LC-MS [ M-H ]] - :m/z 283.0。
The fourth step: the above intermediate compound (2.2g, 7.7mmol), ethyl 2-bromo-2- (diethoxyphosphoryl) acetate (3.1g, 10.1mmol), DBU (1.8g, 11.6 mmol) and LiCl (583mg, 13.9mmol) were dissolved in acetonitrile (50 mL) and reacted at room temperature for 4 hours, followed by refluxing overnight. The reaction was directly concentrated, and the resulting crude product was purified by silica gel column chromatography (PE: EA = 2:1) to give a yellow solid intermediate product (300 mg). LC-MS [ M + H ]] + :m/z 386.9
The fifth step: the above intermediate compound (700mg, 1.8mmol) was dissolved in DMSO (4 mL), and potassium fluoride (526mg, 9.1mmol) and 2,2,2-trifluoroethylamine (2 mL) were added and reacted at 150 ℃ for 1.5 hours by microwave. Water (10 mL) was added for dilution, extraction was carried out three times with ethyl acetate (50 mL), and the combined organic phases were washed with brine and dried. After concentration under reduced pressure, the resulting crude product was purified by silica gel column chromatography (PE: EA = 2:1) to give a yellow solid product (500 mg). LC-MS [ M + H ]] + :m/z 450.0。
And a sixth step: the above intermediate product (60mg, 0.13mmol), 2-methyl-5-indazole boronic acid pinacol ester (36mg, 0.14mmol), dichloro [1,1' -bis (tert-butylphosphine) ferrocene palladium (8mg, 0.01mmol), potassium phosphate (85mg, 0.39mmol) was dissolved in dioxane/H 2 O (10 mL/2 mL) at 60 ℃ for 1h under the protection of nitrogen. Concentration and reverse phase gave the title product as a yellow solid (23 mg). LC-MS [ M + H ]] + :m/z 502.0。 1 H NMR(400MHz,DMSO)δ8.38(s,1H),8.08-8.05(m,3H),7.94(d,J=10.8Hz,1H),7.69-7.44(m,4H),7.33(d,J=8.8Hz,2H),4.18(s,3H),3.88-3.61(m,2H)。
Synthesized according to the procedure of example 40, examples 41-42, 53, 66-67 were obtained.
Figure BDA0003251213670000281
Example 68:1- (4-Chlorobenzene) -3- (3,4-dihydro-2H- [1,3] oxazine [3,2-b ] indazol-9-yl) -7- ((2,2,2-trifluoroethyl) amino) -1,8-naphthyridin-2 (1H) -one
Figure BDA0003251213670000282
The first step is as follows: 5-bromo-2-nitrobenzaldehyde (1.0 g, 2mmol) and 3-amino-1-propanol (625mg, 8.7mmol) were dissolved in ethanol (20 mL), tetraisopropyl titanate (4.9 g, 17.2mmol) was added, and after reaction at room temperature overnight, sodium borohydride (2454mg, 6.5mmol) was added, and the reaction was continued at room temperature for 6 hours. The reaction was quenched by addition of ammonia (6 mL), the reaction was filtered, the filtrate was dried and concentrated, and the crude product was purified by silica gel column chromatography (pure EtOAc elution) to give a yellow solid (780 mg). LC-MS [ M + H ]] + :m/z 289.0。
The second step is that: KOH (607mg, 10.8mmol) was added to a mixed solution of the above-mentioned compound (780mg, 2.7mmol) in t-butanol and water (15 mL/5 mL), and the reaction was carried out at 85 ℃ for 5 hours. Diluted with water (45 mL), extracted three times with ethyl acetate (50 mL), and the combined organic phases were MgSO 4 After drying, filtration and concentration of the organic phase, the crude product was purified by silica gel column chromatography (pure EtOAc elution) to give a yellow solid (540 mg). LC-MS [ M + H ]] + :m/z 252.9。
The third step: under nitrogen protection, the above intermediate compound (540mg, 2.1mmol), pinacol diboron ester (707mg, 2.7mmol) and potassium acetate KOAc (617mg, 6.3mmol) were dissolved in 1,4-dioxane (10 mL), and Pd (dppf) Cl as a catalyst was added 2 (146mg, 0.2mmol) and reacted at 90 ℃ overnight. After cooling to room temperature, the reaction mixture was filtered, concentrated under reduced pressure, and the resulting crude product was purified by silica gel column chromatography (100% EA) to give a yellow crude product (520 mg). LC-MS [ M + H ]] + :m/z 301.1。
The fourth step: to the above intermediate compound (123mg, 0.41mmol), 3-bromo 7-chloro-1- (4-chlorobenzene) -1,8-naphthyridin-2 (1H) -one (150mg, 0.41mmol) and potassium phosphate (260mg, 1.31mmol) in dioxane/H under nitrogen protection 2 To O (3 mL/0.5 mL) was added Pd (dppf) Cl 2 (26mg, 0.04mmol). The reaction was heated to 70 ℃ and stirred for 2 hours. The reaction solution was concentrated under reduced pressure, and the crude product was purified by silica gel column chromatography (pure ethyl acetate elution) to give a yellow solid compound (84 mg). LC-MS [ M + H ]] + :m/z 463.0。
The fifth step: the above intermediate compound (84 m)g,0.18 mmol), 2,2,2-trifluoroethylamine (1 mL), potassium fluoride KF (54mg, 0.90mmol) was dissolved in DMSO (2 mL) and reacted at 150 ℃ for 4h with microwave. The crude product was prepared in reverse phase to give the compound as a yellow solid (22 mg). LC-MS [ M + H ]] + :m/z 526.0。 1 H NMR(400MHz,DMSO-d 6 ):δ8.08(s,1H),7.94(s,1H),7.90-7.85(m,2H),7.59-7.55(m,3H),7.37(d,J=9.2Hz,1H),7.35-7.30(m,2H),6.55(d,J=8.4Hz,1H),4.53(t,J=5.2Hz,2H),4.39(t,J=6.0Hz,2H),3.80-3.73(m,2H),2.37-2.31(m,2H)。
The following indazolocyclic boronic acid (or boronic acid) and bromo-pyrimidopyridinone intermediates were synthesized with reference to example 68 and the literature reported synthetic methods:
Figure BDA0003251213670000291
Figure BDA0003251213670000301
examples 69 to 93
The compounds of examples 69-93 were synthesized according to the procedure for example 68 substituting 3,4-dihydro-2H- [1,3] oxazine [3,2-b ] indazole-9-boronic acid pinacol ester with a different indazolo-cyclic boronic ester (or boronic acid) intermediate.
Figure BDA0003251213670000302
/>
Figure BDA0003251213670000311
/>
Figure BDA0003251213670000321
Figure BDA0003251213670000331
Test example 1 MAT2a enzyme inhibitory Activity test
The compounds of the examples were tested for enzyme inhibitory activity against MAT2a using Colorimetric Assay, the following procedure: 1) Using standard reaction buffer (Tris, pH 8.0,50mM KCl,15mM MgCl) 2 300uM edta,0.005% w/v bovine serum albumin) were allocated 10 compound concentration gradients: test compound concentrations were 10uM starting, 3-fold dilution, 10 concentrations, single well assay. The 10 different concentration solutions were diluted in a 384 well plate in steps to 100 fold final concentration. 250nL were then transferred to 384 reaction plates using Echo550 for use. 100% DMSO-D6 in negative control wells and positive control wells, respectively, plus 250 nL. 2) The enzyme solution was prepared with standard reaction buffer at 1.67 fold final concentration. 3) Adding 15 mu L of enzyme solution with 1.67 times of final concentration into the compound hole and the positive control hole respectively; add 15. Mu.L of standard reaction buffer to the negative control wells. 4) The mixture was centrifuged at 1000rpm for 60 seconds, shaken and mixed, and then incubated for 15 minutes. 5) A substrate mixture solution was prepared with standard reaction buffer at 2.5 times the final concentration. 6) Then, 10. Mu.L of a substrate mixture solution of 2.5 times the final concentration was added to initiate the reaction. 7) The 384 well plates were centrifuged at 1000rpm for 60 seconds, shaken and mixed and then incubated for 150 minutes. 8) The reaction was stopped by adding 50. Mu.L of Biomol, centrifuged at 1000rpm for 60 seconds, and incubated for 15 minutes. The OD620 is read and the data is processed. 9) And data analysis: calculation formula% Inhibition = (OD) 620 _max–OD 620 _sample)/(OD 620 _max–OD 620 Min) x 100; wherein: OD 620 Sample is the sample well absorbance; OD 620 Min: absorbance of negative control wells, representing readings from wells without enzyme; OD 620 Max: absorbance of the positive control wells represents the reading of wells without compound inhibition. 10 ) fitting dose-response curves: the log value of the concentration is taken as an X axis, the percent inhibition is taken as a Y axis, and the analysis software GraphPad Prism5 log (inhibitor) vs. response-Variable slope is adopted to fit a dose-effect curve, so as to obtain the IC of each compound on the enzyme activity 50 The value is obtained. (A represents IC) 50 <100nM, B represents 100 nM. Ltoreq.IC 50 <500nM, C stands for IC 50 ≥500nM)。
As a result: most of the compounds of the examples of the present invention have the advantages ofHigh MAT2a inhibitory Activity, most of example Compound IC 50 Less than 200nM, IC of some embodiments 50 Even less than 10nM.
Figure BDA0003251213670000332
/>
Figure BDA0003251213670000341
Test example 2: example Compound Pair HCT-116 wt And HCT-116 MTAP- Inhibition of cell proliferation.
1. The experimental reagent:
Figure BDA0003251213670000342
2. cell line:
cell lines Type of culture Source Culture medium
HCT116MTAP Wall-mounted type Horizon RPMI-1640+10%FBS
HCT116wt Wall-mounted type Horizon RPMI-1640+10%FBS
3. The testing steps are as follows: 1) Taking HCT-116 in logarithmic growth phase wt /HCT116 MTAP- Cells (horizons) were seeded at an appropriate density into 96-well culture plates at 80. Mu.L per well, and after overnight incubation, compounds at different concentrations were added for 4hr, and a solvent control group (negative control) was set. 2) After the compound acted on the cells for 120hr, the effect of the compound on cell proliferation was detected by using a CTG cell counting kit, 40. Mu.L of CTG reagent was added to each well, and the mixture was placed in an incubator at 37 ℃ for 60min and then read by using a Multilabel Reader microplate Reader from PerkinElmer. 3) The inhibition rate (%) of the compound on tumor cell growth was calculated by the following formula (%) = (OD negative control well-OD administration well)/OD negative control well × 100%. IC (integrated circuit) 50 The values were determined by four parameter regression using a microplate reader random plus software GraphPad Prism 5.
As a result: most of the compounds of the examples of this invention are as in examples 11, 12, 14, 18, 19, 20, 22, 23, 24, 25, 26, 27, 28, 29, 34, 35, 37, 38, 40, 43-62 to HCT-116 MTAP- Cell proliferation inhibitory IC 50 Less than 1uM, e.g., IC of inhibitory activity of examples 18, 34, 37, 38, 39, 40, 42, 43, 44, 48, 49, 50, 53-78, etc 50 More preferably less than 100nM; and all example compounds of the invention for HCT-116 wt Cell proliferation inhibitory Activity IC 50 All greater than 5uM, some of the example compounds as in examples 12, 14, 18, 20, 22, 23, 34, 37, 40, 46, 47, 50, 53, 55-62, 68 to HCT-116 wt Cell proliferation inhibitory Activity IC 50 More than 10uM, showing higher cell selectivity.
Test example 3: ADMET testing of the example Compounds
(1) Metabolic stability test: the system is 150 mu L liver microsome (final concentration 0.5 mg/mL) for metabolic stability incubation, the system contains NADPH (final concentration 1 mM), 1 mu M test compound and positive control midazolam or negative control atenolol, the reaction is stopped by acetonitrile containing tinidazole at 0min, 5min, 10min, 20min and 30min respectively, the reaction is vortexed for 10min, and the reaction is centrifuged for 10min at 15000rmp, and 50 mu L supernatant is sampled in a 96-well plate. The metabolic stability of the compounds was calculated by determining the relative decrease of the bulk drug.
As a result: the compound of the embodiment of the invention has high stability to liver microsomes of various genera (rat, mouse, dog, monkey and human), and the half-life period is more than 20min, such as the compounds of the embodiment 18, 34, 40, 54, 58, 59, 74 and the like.
Figure BDA0003251213670000351
Test example 4: example Compounds pharmacokinetic parameters in mice
6 male SPF-grade Balb c mice (Shanghai Spill-Bikeka experimental animals) were divided into two groups, and the tested compounds were formulated into appropriate solutions or suspensions; one group was administered intravenously (1 mg/kg) and one group was administered orally (5 mg/kg). Blood is collected by jugular venipuncture, each sample is collected by about 0.2 mL/time point, heparin sodium is anticoagulated, and the blood collection time points are as follows: pre-and 5, 15 and 30min,1, 2,4, 6, 8 and 24h after administration; blood samples were collected and placed on ice, plasma was centrifuged (centrifugation conditions: 8000 rpm, 6 min, 2-8 ℃) and collected plasma was stored at-80 ℃ before analysis. Plasma samples were analyzed by LC-MS/MS.
According to the data of the blood concentration of the drug, pharmacokinetic calculation software WinNonlin5.2 non-atrioventricular model is used for respectively calculating the pharmacokinetic parameters AUC of the test sample 0-t 、AUC 0-∞ 、MRT 0-∞ 、C max 、T max 、T 1/2 And V d Isoparameters and their mean and standard deviation. In addition, the bioavailability (F) will be calculated by the following formula.
Figure BDA0003251213670000361
For concentration under quantificationThe lower limit of the sample, when subjected to pharmacokinetic parameter calculations, reached C max The previously sampled samples should be calculated to zero when C is reached max Samples from later sampling points should be calculated as not quantifiable (BLQ).
Figure BDA0003251213670000362
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 (10)

1. A nitrogen-containing fused ring compound shown as a general formula I, or pharmaceutically acceptable salt thereof, or enantiomer, diastereoisomer, tautomer, torsional isomer, solvate, polymorph or prodrug thereof,
Figure FDA0003251213660000011
in the formula: r 1 And R 3 Each independently selected from 5-12 membered monocyclic or bicyclic aromatic or heteroaryl ring, said aromatic or heteroaromatic ring being substituted with 1-3 different substituents Rn, said Rn being selected from hydrogen, deuterium, halogen, cyano, nitro, amide, sulfonamide, hydroxy, amino, ureido, phosphoryl, alkylphosphoxy, alkylsilyl, C 1 -C 6 Alkyl radical, C 1 -C 6 Alkoxy, haloalkyl, haloalkoxy, C 1 -C 6 Monoalkylamino group, C 1 -C 6 Dialkylamino, alkenyl, alkynyl, 3-8 membered cycloalkyl or heterocycloalkyl, C 1 -C 6 alkyl-S-, C 1 -C 6 alkyl-SO-, C 1 -C 6 alkyl-SO 2 -and the like; orThe two Rn may form a 3-12 membered saturated or partially unsaturated or aromatic ring system via a carbon chain or a heteroatom;
R 2 independently selected from hydrogen, deuterium, halogen, cyano, hydroxy, nitro, amino, C 1 -C 12 Alkyl radical, C 1 -C 6 Haloalkyl, C 1 -C 6 alkyl-S-, C 1 -C 6 alkyl-SO-, C 1 -C 6 alkyl-SO 2 -、C 1 -C 6 alkyl-O-, C 1 -C 6 haloalkyl-O-, C 1 -C 6 Monoalkylamino radical, C 1 -C 6 Dialkylamino, 3-12 membered cycloalkylamino or heterocycloalkylamino, 3-12 membered cycloalkyl or heterocycloalkyl, 3-12 membered halocycloalkyl or haloheterocycloalkyl, 3-12 membered cycloalkyl-O-, 3-12 membered halocycloalkyl-O-, 3-12 membered heterocycloalkyl-O-, 5-12 membered aryl or 5-12 membered heteroaryl;
w, X, Y is each independently selected from CR 4 Or N; wherein R is 4 Each independently selected from hydrogen, deuterium, halogen, cyano, hydroxy, nitro, amino, C 1 -C 12 Alkyl radical, C 1 -C 6 Haloalkyl, C 1 -C 6 alkyl-S-, C 1 -C 6 alkyl-SO-, C 1 -C 6 alkyl-SO 2 -、C 1 -C 6 alkyl-O-, C 1 -C 6 haloalkyl-O-, C 1 -C 6 Monoalkylamino radical, C 1 -C 6 A dialkylamino, 3-12 membered cycloalkylamino or heterocycloalkylamino, 3-12 membered cycloalkyl or heterocycloalkyl, 3-12 membered halocycloalkyl or haloheterocycloalkyl, 3-12 membered cycloalkyl-O-, 3-12 membered halocycloalkyl-O-, 3-12 membered heterocycloalkyl-O-, 5-12 membered aryl or 5-12 membered heteroaryl; or-X = Y-may be independently selected from-O-or-S-;
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, C 1 -C 8 An alkyl group; wherein said heteroaryl group contains 1 to 3 heteroatoms selected from the group consisting of: n, O, P or S, said heterocycloalkyl comprising 1-3 heteroatoms selected from the group consisting of: n, O, POr S, wherein the ring system comprises saturated or partially unsaturated ring systems such as spiro rings, bridged rings, fused rings and the like.
2. A compound according to claim 1, preferably represented by the general formula (II), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, atropisomer, solvate, polymorph or prodrug thereof:
Figure FDA0003251213660000012
wherein R is 5 、R 6 、R 7 Each independently selected from hydrogen, deuterium, halogen, cyano, hydroxy, nitro, amino, C 1 -C 12 Alkyl radical, C 1 -C 6 Haloalkyl, C 1 -C 6 alkyl-S-, C 1 -C 6 alkyl-SO-, C 1 -C 6 alkyl-SO 2 -、C 1 -C 6 alkyl-O-, C 1 -C 6 haloalkyl-O-, C 1 -C 6 Monoalkylamino group, C 1 -C 6 Dialkylamino, 3-12 membered cycloalkylamino or heterocycloalkylamino, 3-12 membered cycloalkyl or heterocycloalkyl, 3-12 membered halocycloalkyl or haloheterocycloalkyl, 3-12 membered cycloalkyl-O-, 3-12 membered halocycloalkyl-O-, 3-12 membered heterocycloalkyl-O-, 5-12 membered aryl or 5-12 membered heteroaryl; r is 1 、R 2 、R 3 As defined in claim 1.
3. The compound of claim 1, preferably represented by the general formula (III), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, atropisomer, solvate, polymorph or prodrug thereof:
Figure FDA0003251213660000021
wherein: m 1 Preferably selected from CH or N, M 2 Preferably selected from O, S, NH, etc.;
Figure FDA0003251213660000022
preferably from a single or double bond; r is 8 Preferably selected from hydrogen, deuterium, halogen, cyano, C 1 -C 6 Alkyl radical, C 1 -C 6 Haloalkyl, C 1 -C 6 Alkoxy radical, C 1 -C 6 Haloalkoxy, 3-8 membered cycloalkyl or heterocycloalkyl; r 3 Preferably selected from the group consisting of benzene ring, pyridine ring, thiazole ring, imidazole ring, indole ring, indazole ring, indoline group, isoindoline ring, benzofuran group, dihydrofuran group, pyridofuran group, benzimidazolyl group, benzoxazolyl group, benzothiazolyl group, pyridoimidazolyl group, pyridooxazolyl group, pyridothiazolyl group, naphthalene ring, quinoline ring, isoquinoline ring, quinazoline Lin Huanji, benzomorphine group, benzodioxanyl group and the like, and the R is selected from the group consisting of 3 The ring may be substituted with one or more groups selected from: hydrogen, deuterium, halogen, cyano, C 1 -C 6 Alkyl radical, C 1 -C 6 Haloalkyl, C 1 -C 6 Alkoxy radical, C 1 -C 6 Haloalkoxy, 3-8 membered cycloalkyl or heterocycloalkyl; r is 2 、R 3 W, X, Y are as defined in claim 1.
4. The compound of claim 1, wherein R is 2 Selected from halogen, C 1 -C 6 Alkyl radical, C 1 -C 6 Haloalkyl, C 1 -C 6 alkyl-O-, C 1 -C 6 haloalkyl-O-, -NR a R b 、C 1 -C 6 Monohaloalkylamino, C 1 -C 6 Bishaloalkylamino, C 1 -C 6 alkyl-S-in which R a And R b Each independently selected from hydrogen, deuterium, C 1 -C 6 Alkyl radical, C 1 -C 6 A haloalkyl group.
5. The compound of claim 1, wherein R is 3 Selected from a 5-12 membered bicyclic aryl or heteroaryl ring, which may be substituted with 2-3 substituents Rn, wherein two substituents Rn are adjacent and the two adjacent Rn may form a substituted or unsubstituted 3-6 membered (e.g., 3,4, 5, 6) ring through a carbon chain or heteroatom.
6. The compound of claim 1, wherein W, X, Y, R, R2, and R3 are each independently the corresponding group in compounds 1-94 prepared in the examples.
7. The compound of any one of claims 1-3, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsiomer, solvate, polymorph, or prodrug thereof, wherein the compound has the structure:
Figure FDA0003251213660000031
/>
Figure FDA0003251213660000041
Figure FDA0003251213660000042
and the compound represented by the above general formula (I) does not contain the following structure:
Figure FDA0003251213660000043
8. a process for the preparation of a compound of formula I, said process essentially comprising the following steps c and d:
c, carrying out ring closure on the compound of the general formula (C) and 2-bromo-phosphoryl acetate (D) under the catalysis of alkali to generate an intermediate compound of the general formula (E);
d: reacting the compound of the general formula (E) with substituted aryl boric acid (or ester), aryl tin reagent or aryl silicon reagent by a coupling reaction catalyzed by a transition metal complex to generate the compound of the general formula (I);
Figure FDA0003251213660000051
wherein Ra is hydroxyl, chlorine, alkoxy, ester group; rb is hydrogen or alkyl; rd is phosphoryl ester; r 1 、R 2 、R 3 W, X, Y are as defined in claim 1.
9. A pharmaceutical composition comprising a compound of formula I as claimed in any one of claims 1 to 3 or 7, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsionade, 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.
10. Use of a compound of formula I according to any one of claims 1 to 3 or 7, or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, torsiomer, solvate, polymorph or prodrug thereof, or a pharmaceutical composition according to claim 9, for the preparation of a medicament for the treatment or prophylaxis of a disease associated with MAT2a or MTAP protein activity or expression, in particular of a tumor or an autoimmune disease; the tumor is independently selected from lung cancer, pancreatic cancer, liver cancer, colorectal cancer, bile duct cancer, gallbladder cancer, brain cancer, gastric cancer, leukemia, lymph cancer, melanoma, thyroid cancer, nasopharyngeal cancer, glioma, bladder cancer, astrocytoma, basal cell carcinoma, osteosarcoma, head and neck cancer, chondrosarcoma, ovarian cancer, endometrial cancer, breast cancer, soft tissue sarcoma, mesothelioma and the like; the autoimmune disease is independently selected from the group consisting of thyroiditis, inflammatory enteritis, lupus erythematosus, fibrosis, muscle weakness, vasculitis, psoriasis, arthritis, scleroderma, dermatitis, and the like.
CN202111053145.4A 2020-09-11 2021-09-07 Preparation method and application of nitrogen-containing fused ring compounds Pending CN115960098A (en)

Applications Claiming Priority (10)

Application Number Priority Date Filing Date Title
CN2020109562781 2020-09-11
CN202010956278 2020-09-11
CN202011174227X 2020-10-28
CN202011174227 2020-10-28
CN202011511129 2020-12-19
CN2020115111290 2020-12-19
CN2021100275269 2021-01-10
CN202110027526 2021-01-10
CN2021101854894 2021-02-10
CN202110185489 2021-02-10

Publications (1)

Publication Number Publication Date
CN115960098A true CN115960098A (en) 2023-04-14

Family

ID=80632077

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111053145.4A Pending CN115960098A (en) 2020-09-11 2021-09-07 Preparation method and application of nitrogen-containing fused ring compounds

Country Status (2)

Country Link
CN (1) CN115960098A (en)
WO (1) WO2022052924A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023169554A1 (en) * 2022-03-11 2023-09-14 赛诺哈勃药业(成都)有限公司 Methionine adenosine transferase inhibitor, preparation method therefor and use thereof

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BR0113628A (en) * 2000-08-31 2003-07-01 Hoffmann La Roche 7-oxopyridopyrimidines as inhibitors of cell proliferation
JP4146721B2 (en) * 2000-08-31 2008-09-10 エフ.ホフマン−ラ ロシュ アーゲー 7-Oxo-pyridopyrimidines
MA52232A (en) * 2018-03-30 2021-02-17 Agios Pharmaceuticals Inc MAT2A HETERBICYCLIC INHIBITORS AND THEIR METHODS OF USE FOR THE TREATMENT OF CANCER
US20220144820A1 (en) * 2018-12-27 2022-05-12 Les Laboratoires Servier Sas Aza-heterobicyclic inhibitors of mat2a and methods of use for treating cancer
KR20220050832A (en) * 2018-12-27 2022-04-25 르 라보레또레 쎄르비에르 AZA-heteroid disease inhibitor of Mat2A and method of use for cancer treatment
PE20220387A1 (en) * 2019-05-31 2022-03-18 Servier Lab HETEROBICYCLIC MAT2A INHIBITORS AND METHODS OF USE FOR THE TREATMENT OF CANCER
WO2021139775A1 (en) * 2020-01-10 2021-07-15 江苏先声药业有限公司 Pyridone compound and application

Also Published As

Publication number Publication date
WO2022052924A1 (en) 2022-03-17

Similar Documents

Publication Publication Date Title
CN111704611B (en) Aryl spiro SHP2 inhibitor compound, preparation method and application
CN111153901B (en) Nitrogen-containing fused heterocyclic SHP2 inhibitor compound, preparation method and application
CN112300194B (en) Condensed ring pyridone compounds, preparation method and application
CN112142735B (en) Condensed cyanopyridine compound, preparation method and application
AU2008250328A1 (en) Substituted imidazopyridazines as PI3K lipid kinase inhibitors
AU2012310168B2 (en) 6 - substituted 3 - (quinolin- 6 - ylthio) - [1,2,4] triazolo [4, 3 -a] pyradines as tyrosine kinase
CN109721600B (en) Nitrogen-containing fused ring compounds and preparation method and application thereof
CN116406271A (en) Bicyclic compounds
CN114685487B (en) Pyrimidine heterocyclic compounds, preparation method and application
CN110964012B (en) Fused heterocyclic biaryl benzyl alcohol compound, preparation method and application
WO2021139775A1 (en) Pyridone compound and application
CN112094269B (en) Saturated six-membered ring heterocyclic compound, preparation method and application
CN113527299B (en) Nitrogen-containing condensed ring compound, preparation method and application
CN110857300B (en) Condensed ring triazole compounds, preparation method and application
CN114127063B (en) Pyrimidine five-membered heterocyclic compounds and application thereof as mutant IDH2 inhibitor
CN110950876A (en) Furanolactam compounds, preparation method and application
CN116635371A (en) Polycyclic pyridazinone derivative serving as SOS1 inhibitor, and preparation method and application thereof
CN112300173B (en) Nitrogen-containing polycyclic compounds, preparation method and application
CN112778336B (en) Nitrogen-containing condensed ring STING regulator compound, preparation method and application
CN115960098A (en) Preparation method and application of nitrogen-containing fused ring compounds
CN116323562A (en) Compounds with kinase inhibitory activity
JP2023549418A (en) Spirocyclic compounds
CN111057048B (en) Aminopyrazine/pyridine compound, preparation method and application
WO2022063128A1 (en) Aromatic ring or arylheterocyclic pyridone compound, pharmaceutical compositions, and use thereof
CN114907350A (en) Nitrogen-containing condensed ring compounds, preparation method and application

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