CN114163457A - Pyrimido five-membered nitrogen heterocyclic compound and use thereof - Google Patents

Pyrimido five-membered nitrogen heterocyclic compound and use thereof Download PDF

Info

Publication number
CN114163457A
CN114163457A CN202010954235.XA CN202010954235A CN114163457A CN 114163457 A CN114163457 A CN 114163457A CN 202010954235 A CN202010954235 A CN 202010954235A CN 114163457 A CN114163457 A CN 114163457A
Authority
CN
China
Prior art keywords
compound
ring
pharmaceutically acceptable
independently selected
acceptable form
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
CN202010954235.XA
Other languages
Chinese (zh)
Inventor
袁建栋
方华祥
黄仰青
顾家宁
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.)
Ganjiang New Area Borui Innovative Medicine Co ltd
Original Assignee
Ganjiang New Area Borui Innovative Medicine 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 Ganjiang New Area Borui Innovative Medicine Co ltd filed Critical Ganjiang New Area Borui Innovative Medicine Co ltd
Priority to CN202010954235.XA priority Critical patent/CN114163457A/en
Publication of CN114163457A publication Critical patent/CN114163457A/en
Pending legal-status Critical Current

Links

Classifications

    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia

Abstract

The invention relates to a pyrimido five-membered nitrogen heterocyclic compound and application thereof. In particular, the invention relates to a pyrimido five-membered nitrogen heterocyclic derivative shown in a general formula (I), a preparation method thereof, a pharmaceutical composition containing the derivative, and an application of the derivative as an SHP2 inhibitor for preventing and/or treating tumors or cancers, wherein each substituent in the general formula (I) is defined as the same as that in the specification.

Description

Pyrimido five-membered nitrogen heterocyclic compound and use thereof
Technical Field
The invention discloses a pyrimido five-membered nitrogen heterocyclic compound, a pharmaceutically acceptable salt, a hydrate, a prodrug, a stereoisomer, a solvate or an isotope labeling compound thereof. The invention also provides a preparation method of the compound and an intermediate compound thereof, a composition containing the compound and application of the compound in preparing a medicament for preventing and/or treating diseases or symptoms related to the abnormal activity of SHP 2.
Background
The tyrosine phosphatase SHP2 consists of two N-terminal Src homology 2 domains (N-SH2 and C-SH2) and a protein tyrosine phosphatase catalytic domain (PTP). In the basal state, N-SH2 can be combined with PTP to form a ring structure, so that the combination of PTP and substrate is blocked, and the enzyme catalytic activity is inhibited; when tyrosine of the upstream receptor protein is phosphorylated, NSH2 binds to it, and the PTP catalytic domain is released to exert phosphatase activity.
At the cellular level, SHP2 is involved in multiple tumor cell signaling pathways, such as RTK/Ras/MAPK, JAK/STAT, and PB3K/Akt, among others, through a functional role downstream of the cytoplasm of many receptor tyrosine kinases. Through the regulation of these kinases and signaling pathways, SHP2 is closely related to many important vital cell activities, such as cell proliferation, migration, differentiation, death, cytokine regulation, tumorigenesis, etc.
At the same time, SHP2 is also involved in apoptosis receptor 1(PD1) mediated immune system suppression. After binding of PD-1 to PD-L1 in T cells, large amounts of SHP2 could be recruited in the cells. SHP2 is capable of dephosphorylating an antigen receptor pathway protein within T cells, thereby inhibiting activation of T cells. Thus, inhibition of the activity of SHP2 could reverse immunosuppression in the tumor microenvironment.
As an important class of cell signaling factors, SHP-2 mutations are closely associated with a variety of diseases. The research finds that: SHP-2 mutations are found in neuroblastoma, AML (4%), breast cancer, NSCLC (10%), lung adenocarcinoma (30%), esophageal cancer, head and neck tumors, melanoma, and gastric cancer.
At present, a plurality of allosteric inhibitors of SHP-2 enter clinical research stages, such as TNO-155 developed by Novartis company, RMC-4630 developed by Revolition Medicine company, JAB-3068 developed by Beijing plus Corse, and the like. However, no SHP-2 inhibitor has been developed and marketed for the treatment of Noonan syndrome, leopard syndrome, leukemia, neuroblastoma, melanoma, breast cancer, esophageal cancer, head and neck tumors, lung cancer and colon cancer. Therefore, the development of a SHIP-2 inhibitor drug with good drug forming property is urgently needed.
Disclosure of Invention
The pyrimido five-membered nitrogen heterocyclic compound provided by the invention is a brand-new SHP2 inhibitor, shows good inhibitory activity on tumor cells, has good pharmacy and has wide drug development prospect. And the preparation method of the compound is simple and is beneficial to industrial production.
According to a first aspect of the present invention, there is provided a compound represented by formula I, or a pharmaceutically acceptable salt thereof, or a stereoisomer thereof, or a tautomer thereof, or a hydrate thereof, or a solvate thereof, or a metabolite thereof, or a prodrug thereof,
Figure BDA0002678062020000021
wherein the content of the first and second substances,
X1、X2or X3Independently selected from CR5Or N, and at least one of which is N;
X4or X5Independently selected from C or N;
R1、R2、R3and R4Independently selected from H, halogen, hydroxy, amino, cyano, C2-C8Alkenyl radical, C2-C8Alkynyl, C1-C8Aldehyde group, C1-C8Alkyl radical, C1-C8Heteroalkyl group, C3-C8Cycloalkyl radical, C3-C8Heterocycloalkyl radical, C1-C8Alkoxy or C1-C3Haloalkoxy, said C1-C8Alkyl radical, C1-C8Heteroalkyl group, C3-C8Cycloalkyl radical, C3-C8Heterocycloalkyl radical, C1-C8Alkoxy or C1-C3Haloalkoxy is optionally substituted with one or more R5Substituted when by more than one R5When substituted, R5May be the same or different;
R5selected from H, halogen, hydroxy, amino, cyano, C1-C3Alkyl radical, C1-C3Heteroalkyl group, C3-C8Cycloalkyl radical, C3-C8Heterocycloalkyl radical, C1-C3Alkoxy or C1-C3A haloalkoxy group;
n is selected from 0, 1,2,3,4 and 5;
m is selected from 0, 1,2,3,4 and 5;
ring A is independently selected from C3-C8Cycloalkyl radical, C6-C12Spiro cycloalkyl, C3-C8Heterocycloalkyl radical, C6-C10Aryl or C5-C12A heteroaryl group;
ring B is independently selected from C3-C8Cycloalkyl radical, C3-C8Heterocycloalkyl radical, C6-C10Aryl or C5-C12A heteroaryl group;
the heteroatoms or heteroatom groups contained in the heteroalkyl, heterocycloalkyl, heteroaryl groups are each independently selected from-C (═ O) N (R)5)-、-N(R5)-、-NH-、-N=、-O-、-S-、-C(=O)O-、-C(=O)-、-C(=S)-、-S(=O)-、-S(=O)2-and-N (R)5)C(=O)N(R5) -; the number of heteroatoms or groups of heteroatoms is independently selected from 1,2 and 3, respectively.
In one preferred embodiment, in the pyrimido five-membered nitrogen heterocyclic compound structure shown in formula (I) in the invention, when X is1And X3When is N, X2Is C.
In one preferred embodiment, in the pyrimido five-membered nitrogen heterocyclic compound structure shown in formula (I) in the invention, when X is2And X3When is N, X1Is C.
In one preferred embodiment, the pyrimidine shown in formula (I) isIn the structure of the pyrido-five-membered nitrogen heterocyclic compound, when X is1And X2When is C, X3Is N.
In one preferred embodiment, in the pyrimido five-membered nitrogen heterocyclic compound structure shown in formula (I) in the invention, when X is4When N, ring A is selected from a pyrrole ring, a spirocyclic pyrrole ring, a piperidine ring, a spirocyclic piperidine ring or a dihydropyrazole ring.
In one preferred embodiment, in the pyrimido five-membered nitrogen heterocyclic compound structure shown in formula (I) in the invention, when X is4When N, the ring A is independently selected from
Figure BDA0002678062020000041
Figure BDA0002678062020000042
In one preferred embodiment, in the pyrimido five-membered nitrogen heterocyclic compound structure shown in formula (I) in the invention, when X is4When C is used, the ring A is selected from a benzene ring, a pyridine ring, a pyrimidine ring, a pyrazine ring or a pyridazine ring.
In one preferred embodiment, in the pyrimido five-membered nitrogen heterocyclic compound structure shown in formula (I) in the invention, when X is4When C, the ring A is independently selected from
Figure BDA0002678062020000043
Figure BDA0002678062020000044
In one preferred embodiment, in the pyrimido five-membered nitrogen heterocyclic compound structure shown in formula (I) in the invention, when X is5When N, ring B is independently a pyrrole ring, a piperidine ring, a piperazine ring, or a morpholine ring.
In one preferred embodiment, in the pyrimido five-membered nitrogen heterocyclic compound structure shown in formula (I) in the invention, when X is5When N is present, ring B is independently
Figure BDA0002678062020000045
In one preferred embodiment, in the pyrimido five-membered nitrogen heterocyclic compound structure shown in formula (I) in the invention, when X is5When C, ring B is independently a benzene ring, a pyridine ring, a pyrimidine ring, a pyrazine ring or a pyridazine ring.
In one preferred embodiment, in the pyrimido five-membered nitrogen heterocyclic compound structure shown in formula (I) in the invention, when X is5When C, ring B is independently selected from
Figure BDA0002678062020000051
Figure BDA0002678062020000052
In one preferred embodiment, in the pyrimido five-membered nitrogen heterocyclic compound structure shown in formula (I) in the invention, R1、R2、R3And R4Selected from H, halogen, hydroxy, amino, cyano, C2-C4Alkenyl radical, C1-C4Alkyl radical, C1-C4Heteroalkyl group, C3-C6Cycloalkyl radical, C3-C6Heterocycloalkyl radical, C1-C4Alkoxy or C1-C3Haloalkoxy, said C1-C4Alkyl radical, C1-C4Heteroalkyl group, C3-C6Cycloalkyl radical, C3-C6Heterocycloalkyl radical, C1-C4Alkoxy or C1-C3Haloalkoxy is optionally substituted with one or more R5Substituted when by more than one R5When substituted, R5May be the same or different; wherein R is5Selected from H, F, Cl, Br, hydroxyl, amino, cyano, methyl, ethyl, cyclopropyl, methoxy, ethoxy, trifluoromethoxy.
In one preferred embodiment, in the pyrimido five-membered nitrogen heterocyclic compound structure shown in formula (I) in the invention, R1、R2、R3And R4Selected from H, halogen, hydroxyl, amino, cyano, methyl, ethyl, isopropyl, cyclopropylVinyl, methoxy, ethoxy, isopropoxy, trifluoromethyl, difluoromethyl, trifluoromethoxy, -OCH2CHF2、-N(CH3)2、-NH(CH3) or-OCH2CF3
In some embodiments, the compound of formula (I) of the present invention is selected from compounds of formula (II), formula (III), or formula (IV):
Figure BDA0002678062020000053
Figure BDA0002678062020000061
wherein R is1、R2、R3、R4、X4、X5Ring a and ring B are as defined in claim 1, m is selected from 0, 1,2,3,4 and 5, and n is selected from 0, 1,2,3,4 and 5.
In some embodiments, the compound of formula (I) according to the present invention is selected from the compounds of formulae (II-1) to (IV-3) below:
Figure BDA0002678062020000062
Figure BDA0002678062020000071
wherein R is1、R2、R3、R4、X4、X5Ring a and ring B are as defined in claim 1, m is selected from 0, 1,2,3,4 and 5, and n is selected from 0, 1,2,3,4 and 5.
According to a specific embodiment of the present invention, the compound represented by formula I according to the present invention is any one of the following compounds:
Figure BDA0002678062020000072
in the present invention, the groups and substituents thereof in the compound of formula I can be selected by those skilled in the art to provide stable compounds of formula I, or pharmaceutically acceptable salts thereof, or stereoisomers thereof, or tautomers thereof, or hydrates thereof, or solvates thereof, or metabolites thereof, or prodrugs thereof, including but not limited to I-1 to I-6 described in the examples of the present invention.
The reaction solvent used in each reaction step described in the present invention is not particularly limited, and any solvent that can dissolve the starting materials to some extent and does not inhibit the reaction is included in the present invention. Further, many equivalents, substitutions, or equivalents in the art to which this invention pertains, as well as different proportions of solvents, solvent combinations, and solvent combinations described herein, are deemed to be encompassed by the present invention.
According to a second aspect of the present invention, the present invention provides a pharmaceutical composition comprising an effective dose of a compound represented by formula I, or a pharmaceutically acceptable salt thereof, or a stereoisomer thereof, or a tautomer thereof, or a hydrate thereof, or a solvate thereof, or a metabolite thereof, or a prodrug thereof, and at least one pharmaceutical excipient.
The pharmaceutical excipients can be those widely used in the field of pharmaceutical production. The excipients are used primarily to provide a safe, stable and functional pharmaceutical composition and may also provide methods for dissolving the active ingredient at a desired rate or for promoting the effective absorption of the active ingredient after administration of the composition by a subject. The pharmaceutical excipients may be inert fillers or provide a function such as stabilizing the overall pH of the composition or preventing degradation of the active ingredients of the composition. The pharmaceutical excipients may include one or more of the following excipients: binders, suspending agents, emulsifiers, diluents, fillers, granulating agents, adhesives, disintegrating agents, lubricants, antiadherents, glidants, wetting agents, gelling agents, absorption delaying agents, dissolution inhibitors, reinforcing agents, adsorbents, buffering agents, chelating agents, preservatives, colorants, flavoring agents and sweeteners.
The pharmaceutical compositions of the present invention may be prepared according to the disclosure using any method known to those skilled in the art. For example, conventional mixing, dissolving, granulating, emulsifying, levigating, encapsulating, entrapping or lyophilizing processes.
The pharmaceutical compositions of the present invention may be administered in any form, including injection (intravenous), mucosal, oral (solid and liquid formulations), inhalation, ocular, rectal, topical or parenteral (infusion, injection, implant, subcutaneous, intravenous, intraarterial, intramuscular) administration. The pharmaceutical compositions of the present invention may also be in a controlled release or delayed release dosage form (e.g., liposomes or microspheres). Examples of solid oral formulations include, but are not limited to, powders, capsules, caplets, soft capsules, and tablets. Examples of liquid formulations for oral or mucosal administration include, but are not limited to, suspensions, emulsions, elixirs and solutions. Examples of topical formulations include, but are not limited to, emulsions, gels, ointments, creams, patches, pastes, foams, lotions, drops or serum formulations. Examples of formulations for parenteral administration include, but are not limited to, solutions for injection, dry preparations which can be dissolved or suspended in a pharmaceutically acceptable carrier, suspensions for injection, and emulsions for injection. Examples of other suitable formulations of the pharmaceutical composition include, but are not limited to, eye drops and other ophthalmic formulations; aerosol: such as nasal sprays or inhalants; liquid dosage forms suitable for parenteral administration; suppositories and lozenges.
Oral administration of the compounds of the invention is preferred. Intravenous administration of the compounds of the invention is also preferred. Depending on the circumstances, other application routes may be applied or even preferred. For example, transdermal administration may be highly desirable for patients who are forgetful or whose oral medications are irritable. In particular cases, the compounds of the invention may also be administered by transdermal, intramuscular, intranasal or intrarectal routes. The route of administration may vary in any manner, limited by the physical nature of the drug, the convenience of the patient and caregiver, and other relevant circumstances.
According to a third aspect of the present invention, the present invention provides a use of a compound represented by formula I, or a pharmaceutically acceptable salt thereof, or a stereoisomer thereof, or a tautomer thereof, or a pharmaceutical composition thereof, in the preparation of a medicament for the treatment and/or prevention of a disease caused by an abnormal mutation in SHP 2. The compound provided by the invention can be used for treating and/or preventing one or more than two diseases caused by SHP2 abnormal mutation, and has good clinical application and medical application.
According to a fourth aspect of the present invention, the present invention provides a compound represented by formula I, or a pharmaceutically acceptable salt thereof, or a stereoisomer thereof, or a tautomer thereof, or a hydrate thereof, or a solvate thereof, or a metabolite thereof, or a prodrug thereof, or a pharmaceutical composition thereof, for use in preparing a SHP2 inhibitor drug. The compound provided by the invention has excellent SHP2 enzyme activity and cell proliferation inhibition activity, can be effectively used as a SHP2 inhibitor and is used as a therapeutic drug of a SHP2 inhibitor.
According to a fifth aspect of the present invention, the present invention provides a compound represented by formula I, or a pharmaceutically acceptable salt thereof, or a stereoisomer thereof, or a tautomer thereof, or a hydrate thereof, or a solvate thereof, or a metabolite thereof, or a prodrug thereof, or a pharmaceutical composition thereof for use in the preparation of a medicament for the treatment and/or prevention of cancer. The compound provided by the invention can be used for preparing a medicament for treating and/or preventing cancers, wherein the cancers comprise but are not limited to Noonan syndrome, leopard syndrome, juvenile myelomonocytic leukemia, neuroblastoma, melanoma, acute myelogenous leukemia, breast cancer, esophageal cancer, lung cancer, colon cancer, gastric cancer, anaplastic large-cell lymphoma, glioblastoma, non-small cell lung cancer or head and neck tumors.
The invention has the following advantages:
1. the pyrimidine five-membered nitrogen heterocyclic compound disclosed by the invention is a novel allosteric inhibitor, and can be combined with a non-catalytic region of SHP2 to lock a basic state with weak activity of SHP2, so that the purpose of inhibiting the activity of the pyrimidine five-membered nitrogen heterocyclic compound is achieved. The pyrimido five-membered nitrogen heterocyclic compound disclosed by the invention overcomes the defects of poor general selectivity and drug-forming property and the like of a PTP catalytic region inhibitor, shows good biological activity and drug-forming property, and has good drug development prospect.
2. Compared with the compounds RMC4550 and TNO155 with the disclosed structures, the compound shows more excellent activity and PK property in an SHP2 enzyme activity inhibition experiment, a phosphorylated protein kinase (p-ERK) cell experiment, an NCI-H358 cell, MV-4-11 cell proliferation inhibition experiment and other evaluation systems under the same conditions.
Terms and definitions
Unless stated to the contrary, the following terms used in the specification and claims have the following meanings.
The term "alkyl" refers to a saturated aliphatic hydrocarbon group, e.g., straight and branched chain groups comprising 1 to 20 carbon atoms, e.g., straight and branched chain groups which may be 1 to 18 carbon atoms, 1 to 12 carbon atoms, 1 to 8 carbon atoms, 1 to 6 carbon atoms, or 1 to 4 carbon atoms. In this context "alkyl" may be a monovalent, divalent or trivalent radical. Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 2-trimethylpropyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 2-dimethylbutyl, 1, 3-dimethylbutyl, 2-ethylbutyl, and the various branched chain isomers thereof, and the like. Non-limiting examples also include methylene, methine, ethylene, ethylidene, propylidene, butylidene, and various branched chain isomers thereof. Alkyl groups may be optionally substituted or unsubstituted.
The term "cycloalkyl" refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent, for example, which includes from 3 to 12 ring atoms, which may be, for example, from 3 to 12, from 3 to 10, or from 3 to 6 ring atoms, or may be a 3,4, 5, 6 membered ring. Non-limiting examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cycloheptatrienyl, cyclooctyl, and the like. The cyclic group may be optionally substituted or unsubstituted.
The term "spirocycloalkyl" refers to a cycloalkyl group in which two rings share a single ring atom. The common ring atom is also referred to as a spiro atom, most commonly referred to as a quaternary carbon (spiro carbon).
The term "heteroalkyl" refers to a stable straight or branched chain consisting of the stated number of carbon atoms and one or more heteroatoms selected from the group consisting of O, N and S, and wherein the nitrogen and sulfur atoms are optionally oxidized, and the nitrogen atoms are optionally quaternized. Examples include, but are not limited to-CH2-CH2-O-CH3、-CH2-CH2-NH-CH3、-CH2-CH2-N(CH3)-CH3、-CH2-S-CH2-CH3、-CH2-CH2-S(O)-CH3、-CH2-CH2-S(O)2-CH3or-CH2-CH=N-OCH3. Up to two heteroatoms may be consecutive, e.g. -CH2-NH-O-CH3. In certain embodiments, the heteroalkyl is optionally substituted as described elsewhere herein.
The term "heterocycloalkyl" refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent, for example, which includes from 3 to 20 ring atoms, which may be, for example, from 3 to 16, from 3 to 12, from 3 to 10, or from 3 to 6 ring atoms, wherein one or more ring atoms is selected from nitrogen, oxygen, or S (O)m(wherein m is 0, 1, or 2) but does not include the ring moiety of-O-O-, -O-S-, or-S-S-, and the remaining ring atoms are carbon. In some embodiments it is preferred to include 3 to 12 ring atoms of which 1-4 are heteroatoms, more preferably the heterocycloalkyl ring comprises 3 to 10 ring atoms, most preferably a 5 or 6 membered ring of which 1-4 are heteroatoms, more preferably 1-3 are heteroatoms, most preferably 1-2 are heteroatoms. Non-limiting examples of monocyclic heterocycloalkyl include pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, and the like. Non-limiting examples of polycyclic heterocycloalkyl groups include spiro, fused, or bridged heterocycloalkyl groups.
The term "alkenyl" refers to a straight or branched chain unsaturated aliphatic hydrocarbon group having at least one double bond, consisting of carbon atoms and hydrogen atoms. Non-limiting examples of alkenyl groups include, but are not limited to, ethenyl, 1-propenyl, 2-propenyl, 1-butenyl, isobutenyl, 1, 3-butadienyl, and the like.
The term "alkynyl" refers to a straight or branched chain unsaturated aliphatic hydrocarbon group having at least one triple bond composed of carbon atoms and hydrogen atoms. Non-limiting examples of alkynyl groups include, but are not limited to, ethynyl (-C ≡ CH), 1-propynyl (-C ≡ C-CH)3) 2-propynyl (-CH)2-C.ident.CH), 1, 3-butadiynyl (-C.ident.C-C.ident.CH), and the like.
The term "alkoxy" refers to-O-alkyl.
The term "haloalkoxy" denotes an alkoxy group substituted with one or more halogen atoms, which may be the same or different, such as trifluoromethoxy and the like.
The term "aryl" refers to an all-carbon monocyclic or fused polycyclic aromatic ring group having a conjugated pi-electron system. For example, the aryl group can have 6 to 20 carbon atoms, 6 to 14 carbon atoms, or 6 to 12 carbon atoms. Non-limiting examples of aryl groups include, but are not limited to, phenyl, naphthyl, anthracenyl, and 1,2,3, 4-tetrahydronaphthalene, and the like.
The term "heteroaryl" refers to the group remaining after 1 hydrogen atom has been removed from the "heteroaromatic ring" molecule, which may be unsubstituted or substituted, including but not limited to alkyl, alkyloxy, aryl, aralkyl, amino, halo, hydroxy, cyano, nitro, carbonyl, and heteroalicyclic. Non-limiting examples of unsubstituted heteroaryl groups include, but are not limited to, pyrrolyl, furanyl, thienyl, imidazolyl, oxazolyl, pyrazolyl, pyridyl, pyrimidinyl, pyrazinyl, quinolinyl, isoquinolinyl, tetrazolyl, triazinyl.
The term "halogen" refers to fluorine, chlorine, bromine and iodine, with fluorine, chlorine and bromine being preferred in some embodiments.
The term "amino" refers to the group-NH2The radicals, -NH (alkyl) and-N (alkyl)2Specific examples of amino groups include, but are not limited to, -NH2、-NHCH3、-NHCH(CH3)2、-N(CH3)2、-NHC2H5、-N(CH3)C2H5And the like.
The term "hydroxy" refers to an-OH group.
The term "cyano" refers to the group — CN.
The terms "optional" or "optionally" mean that the subsequently described event or circumstance may or may not occur. For example: "heterocyclic group optionally substituted with alkyl" means that alkyl may or may not be present, that is, includes the case where the heterocyclic group is substituted with alkyl and the case where the heterocyclic group is not substituted with alkyl.
"substituted" means that one or more hydrogen atoms in a group are replaced with a substituent. For example, up to 5, more preferably 1 to 3 hydrogen atoms in the group are independently substituted with a corresponding number of substituents.
"pharmaceutically acceptable salts" refers to salts of the compounds of the present invention, prepared from the compounds of the present invention found to have particular substituents, with relatively nontoxic acids or bases. When compounds of the present invention contain relatively acidic functional groups, base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of a base in neat solution or in a suitable inert solvent. Pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic ammonia or magnesium salts or similar salts. When compounds of the present invention contain relatively basic functional groups, acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of acid in neat solution or in a suitable inert solvent. Examples of pharmaceutically acceptable acid addition salts include inorganic acid salts including, for example, hydrochloric acid, hydrobromic acid, nitric acid, carbonic acid, bicarbonate, phosphoric acid, monohydrogen phosphate, dihydrogen phosphate, sulfuric acid, hydrogen sulfate, hydroiodic acid, phosphorous acid, and the like; and salts of organic acids including acids such as acetic, propionic, isobutyric, maleic, malonic, benzoic, succinic, suberic, fumaric, lactic, mandelic, phthalic, benzenesulfonic, p-toluenesulfonic, citric, tartaric, methanesulfonic, and the like; also included are Salts of amino acids (e.g., arginine, etc.), and Salts of organic acids such as glucuronic acid (see Berge et al, "Pharmaceutical Salts", Journal of Pharmaceutical Science 66:1-19 (1977)). Certain specific compounds of the invention contain both basic and acidic functionalities and can thus be converted to any base or acid addition salt. Preferably, the neutral form of the compound is regenerated by contacting the salt with a base or acid and isolating the parent compound in a conventional manner. The parent form of the compound differs from the various salt forms by certain physical properties, such as solubility in polar solvents. According to an embodiment of the present invention, preferably the pharmaceutically acceptable salt of the compound of formula I of the present invention is a hydrochloride, hydrobromide, phosphate, or sulfate salt, most preferably a hydrochloride salt.
The term "solvate" refers to a physical association of a compound of the present invention with one or more solvent molecules. The physical association includes various degrees of ionic and covalent bonding, including hydrogen bonding. "solvates" includes both solution phase and isolatable solvates. Non-limiting examples of solvates include ethanolates, methanolates, and the like. A "hydrate" is where the solvent molecule is H2A solvate of O.
"pharmaceutical composition" means a mixture containing one or more compounds, or pharmaceutically acceptable salts thereof, or stereoisomers thereof, or tautomers thereof, and other chemical components, as well as other components, such as pharmaceutically acceptable excipients. The purpose of the pharmaceutical composition is to facilitate administration to an organism, facilitate absorption of the active ingredient and exert biological activity.
Any formula or structure given herein is also intended to represent an unlabeled form of the compound as well as an isotopically labeled form. Isotopically-labeled compounds have the structure shown by the formulae given herein, except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Examples of isotopes that can be incorporated into compounds of the present disclosure include isotopes of hydrogen, carbon, nitrogen, oxygen, fluorine, and chlorine, such as, but not limited to2H (deuterium, D),3H (tritium),11C、13C、14C、15N、18F、35S、36Cl and125I. various isotopically-labeled compounds of the present disclosure, e.g., incorporation of3H、13C and14c, and the like. Such isotopically labeled compounds can be used in metabolic studies, reaction kinetic studies, detection or imaging techniques such as Positron Emission Tomography (PET) or Single Photon Emission Computed Tomography (SPECT) including drug or substrate tissue distribution analysis, or in the radiation treatment of patients. Isotopically labeled compounds of the present disclosure and prodrugs thereof can generally be prepared by carrying out the procedures disclosed in the schemes or in the examples and preparations described below, by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent.
The compounds of the present invention may be in the form of prodrug compounds. By "prodrug compound" is meant a derivative which is converted into a compound according to the present invention by a reaction with an enzyme or gastric acid or the like under physiological conditions in an organism, for example by oxidation, reduction or hydrolysis or the like, each of which reactions is carried out enzymatically. Examples of prodrugs are compounds wherein an amino group in a compound of the invention is acylated, alkylated or phosphorylated to form, for example, eicosanoylamino, alanylamino, pivaloyloxymethylamino, or wherein a hydroxyl group is acylated, alkylated, phosphorylated or converted to a boronic ester, for example, acetyloxy, palmitoyloxy, pivaloyloxy, succinyloxy, fumaroyloxy, alanyloxy, or wherein a carboxyl group is esterified or amidated. These compounds can be produced from the compounds of the present invention according to known methods. Other examples of prodrugs are compounds wherein, for example, the carboxylic acid ester in the compounds of the invention is converted to alkyl-, aryl-, choline-, amino, acyloxymethyl ester, linolenoyl ester.
In the case where tautomerism of the compounds of the invention or their prodrugs, such as, for example, keto-enol tautomerism, occurs, the individual forms, such as, for example, the keto and enol forms and mixtures thereof in any ratio, are each within the scope of the invention. The same applies to stereoisomers, such as, for example, enantiomers, cis/trans isomers, and conformational isomers, and the like.
If desired, isomers may be separated by methods well known in the art, for example by liquid chromatography. The same applies to enantiomers by using, for example, a chiral stationary phase. Furthermore, enantiomers can be separated by: they are converted into diastereomers, i.e. coupled with enantiomerically pure auxiliary compounds, the resulting diastereomers are subsequently separated and the auxiliary residues are cleaved. Alternatively, any enantiomer of a compound of the invention may be obtained from stereoselective synthesis using optically pure starting materials.
The term "treating" means administering a compound or formulation of the invention to prevent, ameliorate or eliminate a disease or one or more symptoms associated with the disease, and includes:
(i) preventing the occurrence of a disease or condition in a mammal, particularly when such mammal is susceptible to the disease condition, but has not yet been diagnosed as having the disease condition;
(ii) inhibiting the disease or disease state, i.e., arresting its development;
(iii) alleviating the disease or condition, i.e., causing regression of the disease or condition.
The term "effective amount" means an amount of a compound of the invention that (i) treats or prevents a particular disease, condition, or disorder, (ii) alleviates, ameliorates, or eliminates one or more symptoms of a particular disease, condition, or disorder, or (iii) prevents or delays the onset of one or more symptoms of a particular disease, condition, or disorder described herein. The amount of a compound of the present invention that constitutes an "effective dose" varies depending on the compound, the disease state and its severity, the mode of administration, and the age of the mammal to be treated, but can be routinely determined by those skilled in the art in light of their own knowledge and this disclosure.
Additional aspects and advantages of the invention will be set forth in part in the description which follows and, in part, will be obvious from the description, or may be learned by practice of the invention.
Figure BDA0002678062020000161
The specific reaction conditions are described below:
the compound A and the compound B are reacted by heating in a solvent such as DMF, DMSO, NMP or the like under an alkaline condition such as potassium carbonate, DIEA, cesium carbonate, sodium tert-butoxide or the like to obtain a compound C. The compound C and Boc anhydride are catalyzed by DMAP in dichloromethane to obtain a compound D, and the compound D and the compound E are subjected to Pd2(dba)3Using Xantphos as ligand, adding organic or inorganic base such as DIEA, cesium carbonate, etc. to obtain compound F, and deprotecting compound F with TFA or ethyl acetate hydrochloride to obtain the compound represented by formula I in free or hydrochloride form.
Detailed Description
The preparation of the compound of formula I, or a pharmaceutically acceptable salt, a stereoisomer, a tautomer, a hydrate, a solvate, a metabolite, or a prodrug thereof according to the present invention can be accomplished by the following exemplary methods and relevant publications used by those skilled in the art, but the scope of the present invention is not limited by these examples.
The structure of the compounds of the invention is determined by Nuclear Magnetic Resonance (NMR) or Mass Spectrometry (MS). NMR was measured using a Bruker AVANCE-400 or Varian Oxford-300 nuclear magnetic spectrometer using deuterated dimethyl sulfoxide (DMSO-d) as the solvent6) Deuterated chloroform (CDC 1)3) Deuterated methanol (CD)3OD), internal standard Tetramethylsilane (TMS), chemical shift is 10-6(ppm) is given as a unit.
MS was measured using an Agilent SQD (ESI) mass spectrometer (manufacturer: Agilent, model: 6110) or Shimadzu SQD (ESI) mass spectrometer (manufacturer: Shimadzu, model: 2020).
HPLC measurements were performed using an Agilent 1200DAD high pressure liquid chromatograph (Sunfirc C18, 150X4.6mm, 5wn, column) and Waters 2695-2996 high pressure liquid chromatograph (Gimini C18, 150X4.6mm, 5ym column).
The thin layer chromatography silica gel plate is Qingdao sea GF254 silica gel plate, the specification of the silica gel plate used by Thin Layer Chromatography (TLC) is 0.15mm-0.2mm, and the specification of the thin layer chromatography separation and purification product is 0.4mm-0.5 mm.
Column chromatography generally uses Qingdao ocean 200-mesh and 300-mesh silica gel as a carrier.
Known starting materials of the present invention can be synthesized by or according to methods known in the art, or purchased from companies such as Shao Yuan chemical technology (Accela ChemBio Inc), Beijing coupled Chemicals, and the like.
In the examples, the reaction was carried out under an argon atmosphere or a nitrogen atmosphere unless otherwise specified. The hydrogenation reaction was usually evacuated and charged with hydrogen and repeated 3 times.
In the examples, the reaction temperature was room temperature and the temperature range was 5 ℃ to 35 ℃ unless otherwise specified.
The progress of the reaction in the examples was monitored by Thin Layer Chromatography (TLC) using a system of developing reagents, A: dichloromethane and methanol systems; b: petroleum ether and ethyl acetate, the volume ratio of the solvent is adjusted according to the polarity of the compound.
The system of eluents for column chromatography and developing agents for thin layer chromatography used for purifying compounds include a: dichloromethane and methanol systems; b: the volume ratio of the solvent in the petroleum ether and ethyl acetate system is adjusted according to different polarities of the compounds, and a small amount of triethylamine, an acidic or basic reagent and the like can be added for adjustment.
The present invention is described in detail below by way of examples, but is not meant to be limited to any of the disadvantages of the present invention. The compounds of the present invention may be prepared by a variety of synthetic methods well known to those skilled in the art, including the specific embodiments listed below, embodiments formed by combinations thereof with other chemical synthetic methods, and equivalents thereof known to those skilled in the art, with preferred embodiments including, but not limited to, examples of the present invention. It will be apparent to those skilled in the art that various changes and modifications can be made in the specific embodiments of the invention without departing from the spirit and scope of the invention. The following synthetic schemes describe the steps for preparing the compounds disclosed herein. Unless otherwise indicated, each substituent has the definition as described herein.
And (3) synthesis of an intermediate:
synthesis of intermediate Q1
Figure BDA0002678062020000181
The first step is as follows: synthesis of Q1-2
Adding compound Q1-1(100g, 495mmol) into DME (1000ml), cooling to-10 ℃, adding isobutyl chloroformate (67.6g, 495mmol) and 4-methylmorpholine (50g,495mmol), reacting at room temperature for 5 hours, filtering after TLC reaction is finished, washing the solid with DME (250ml), adding the filtrate into a 2L three-necked flask, adding sodium borohydride (37.6g, 990mmol) to treat the filtrate, stirring at room temperature for 30min, slowly adding methanol (250ml), continuing to react at room temperature for 3 hours, completing the TLC reaction, spin-drying the reaction liquid, adding water (1500ml), extracting the aqueous phase with DCM (300ml 3), combining the organic phases, washing the organic phase with water and saturated sodium chloride again, drying the organic phase, and spin-drying to obtain compound Q1-2(86.5g, white solid) with 93 percent yield. Used directly in the next step.
The second step is that: synthesis of Q1-3
Compound Q1-2(25g, 133mmol)) was added to DCM (250ml), TEA (26.9g, 266mmol) was added, and MsCl (18.3g, 156mmol) was added dropwise under ice bath, followed by reaction at room temperature for 1 h. TLC showed the reaction was complete, the reaction was diluted with DCM, washed with water and saturated sodium chloride, the organic phase was dried, concentrated and concentrated to dryness, and the residue obtained from the concentration was chromatographed on silica gel (eluent petroleum ether: ethyl acetate: 3: 1-1: 1) to give compound Q1-3(33.6g, colorless clear liquid) in 95% yield.
MS m/z(ESI):266[M+1]。
The third step: synthesis of Q1-4
Adding N-Boc-4-cyanopiperidine (28g, 130mmol) into THF (300ml), cooling to-78 deg.C, slowly dropwise adding 2.0M LDA (75ml, 150mmol), continuing to keep at-78 deg.C after dropwise adding, reacting for 1.5h, then dropwise adding THF solution (150ml) of compound Q1-3(26.6, 100mmol) obtained in the previous step, continuing to keep at-78 deg.C after dropwise adding, reacting for 3h, after TLC shows that the reaction is finished, dropwise adding saturated ammonium chloride solution (50ml), quenching, adding saturated sodium chloride aqueous solution (500ml), separating out organic phase, extracting aqueous phase with ethyl acetate (150 ml. times.3), combining organic phases, drying with anhydrous sodium sulfate, filtering, concentrating to obtain residue, passing through silica gel chromatographic column (petroleum ether: ethyl acetate: 10: 1-1: 1) to obtain compound Q1-4(25.7g, white solid), yield 67.6%.
MS m/z(ESI):380[M+1]。
The fourth step: synthesis of Q1-5
The compound Q1-4(25g, 65.8mmol) obtained in the previous step was added to a mixed solvent of DMA (200ml) and water (20ml), and triethylamine (33.2g, 329mmol) and a catalyst of dichloro-di-tert-butyl- (4-dimethylaminophenyl) phosphonium palladium (II) (4.6g, 6.6mmol, CAS:887919-35-9) were further added, and the reaction was carried out at 130 ℃ for 4 hours under nitrogen protection. After TLC showed the reaction was complete, the reaction was cooled and diluted with water (800mL), the aqueous phase was extracted with ethyl acetate (200mL × 3), the organic phases were combined, washed with saturated brine (200mL × 2), dried over anhydrous sodium sulfate, filtered and the residue obtained by spin-dry concentration was subjected to silica gel chromatography (eluent petroleum ether: ethyl acetate ═ 3:1 to 1:1) to give compound Q1-5(14.6g, white solid) in 73% yield.
MS m/z(ESI):303[M+1]。
The fifth step: synthesis of Q1-6
Compound Q1-5(10g, 33mol) obtained in the previous step was added to tetraethyl titanate (100mL), followed by addition of (R) - (+) -tert-butylsulfinamide (4.8g, 40mml), reaction was carried out at 90 ℃ for 3 hours, TLC showed completion of the reaction, cooling to room temperature, the reaction solution was slowly added to ice water (500mL), the resulting aqueous phase was extracted with dichloromethane (150mL × 3), the organic phases were combined, the organic phase was washed with saturated brine (100mL × 2), the organic phase was dried over anhydrous sodium sulfate, filtered, and the residue obtained by rotary drying and concentration was subjected to silica gel chromatography (eluent petroleum ether: ethyl acetate ═ 3:1 to 1:1) to obtain compound Q1-6(12g, yellow solid) with a yield of 89.6%.
MS m/z(ESI):406[M+1]。
And a sixth step: synthesis of Q1-7
Adding the compound Q1-6(10g, 24.6mmol) obtained in the previous step into tetrahydrofuran (100mL), cooling to-78 ℃, slowly dropwise adding DIBAL-H (30mL, 30mmol, 1M toluene solution), then continuing to react at-78 ℃ for 0.5H, after TLC shows that the reaction is finished, adding saturated Rochelle salt solution (300mL) at-50 ℃ to quench the reaction, stirring at room temperature for 30min, extracting the obtained water phase with ethyl acetate (150mL x 3), combining the organic phases, washing the organic phases with saturated saline (100mL x 2), drying the organic phases with anhydrous sodium sulfate, filtering, and carrying out spin-drying concentration on the obtained residue through a silica gel chromatographic column (eluent petroleum ether: ethyl acetate ═ 3: 1-1: 1) to obtain the compound Q1-7(8.6g, white solid) with the yield of 85.3%.
MS m/z(ESI):408[M+1]。
The seventh step: synthesis of intermediate Q1
Compound Q1-7(5g, 12.2mmol) obtained in the previous step was added to ethyl acetate (50ml), 4M HCl/ethyl acetate solution (25ml) was added and the reaction was allowed to proceed at room temperature for 1h, after TLC showed completion of the reaction, it was filtered, the solid was washed with ethyl acetate and dried to give the hydrochloride of compound Q1 (3.2g, white solid) in 94% yield.
MS m/z(ESI):204[M+1]。
Synthesis of intermediate Q2
Figure BDA0002678062020000211
The first step is as follows: synthesis of Q2-2
5-bromo-2, 4-dichloropyrimidine (5.0g, 21.94mmol) was added to a solution of ethanol (100mL), followed by triethylamine (3.7mL, 26.33mmol) and 2, 2-dimethoxyethylamine (2.53mL, 24.14mmol), and the reaction mixture was stirred at room temperature for 24 h. After completion of the reaction, the reaction mixture was concentrated under reduced pressure. 200mL of water was added, the resulting aqueous phase was extracted with ethyl acetate (100mL × 3), the organic phases were combined, the organic phase was washed with saturated brine (70mL × 2), the organic phase was dried over anhydrous sodium sulfate, filtered, and the residue obtained by spin-dry concentration was subjected to silica gel chromatography (eluent petroleum ether: ethyl acetate ═ 3:1 to 1:1) to obtain compound Q2-2(5.4g, white solid) in 83% yield.
MS m/z(ESI):296[M+1]。
The second step is that: synthesis of Q2-3
Compound Q2-2(5.4g, 18.2mmol) obtained in the previous step was dissolved in concentrated sulfuric acid (50mL) and then warmed to 75 ℃. After 2h of reaction, TLC showed the reaction was complete, cooled to room temperature, the reaction was poured slowly into ice water, then slowly basified with 5M aqueous sodium hydroxide to pH6 to precipitate a large amount of solid, filtered, the filter cake washed with water and the solid dried to give compound Q2-3(3.2g, grey solid) in 82.5% yield.
MS m/z(ESI):214[M+1]。
The third step: synthesis of intermediate Q2
To compound Q2-3(3.2g, 15mmol) was added phosphorus oxychloride (50mL), followed by DIEA (5.8g, 45mmol), and the mixture was heated to 115 ℃. After 3h of reaction, TLC indicated the end of the reaction, the mixture was concentrated in vacuo and the residue was diluted with EtOAc (100ml) and slowly adjusted to pH 7 with saturated sodium bicarbonate solution. The organic phase was separated, the aqueous phase was extracted with ethyl acetate, the combined organic phases were washed with saturated brine (70mL × 2), the organic phase was dried over anhydrous sodium sulfate, filtered, and the residue obtained by spin-drying and concentration was subjected to flash silica gel chromatography (eluent petroleum ether: ethyl acetate: 3:1 to 1:1) to obtain compound Q2(2.7g, white solid) in 78% yield.
MS m/z(ESI):232[M+1]。
Synthesis of intermediate Q3
Figure BDA0002678062020000221
The first step is as follows: synthesis of Q3-2
5-iodo-2, 4-dichloropyrimidine (16.3g, 59.2mmol) and hydrazine hydrate (8.8mL, 181mmol) were added to anhydrous ethanol (300mL), followed by heating under reflux for 12 hours. TLC showed the reaction was complete and cooled to 0 c and a large amount of solid precipitated, filtered, and the filter cake was washed with ice ethanol (100mL) and dried to give Q3-2(15.1g, yellow solid) in 94.3% yield, which was used in the next step without further purification.
MS m/z(ESI):271[M+1]。
The second step is that: synthesis of intermediate Q3
Compound Q3-2(15g, 55.3mmol) obtained in the previous step was added to trimethyl orthoformate (100mL) and heated under reflux for 12 hours, TLC showed the reaction was complete, cooled to 0 deg.C, and a large amount of solid precipitated, filtered, and the filter cake was washed with ice 50% ethanol (50mL) and dried to give compound Q3(11.3g, yellow solid) in 88% yield, which was used in the next step without further purification.
MS m/z(ESI):281[M+1]。
Synthesis of intermediate Q4
Figure BDA0002678062020000231
The first step is as follows: synthesis of Compound Q4-2
Compound Q4-1(40g, 192mmol) was added to N, N-dimethylformamide dimethyl acetal (200ml), and the temperature was raised to 120 ℃ for reaction for 3 hours. TLC showed the reaction was complete and most of the solvent was removed under reduced pressure, then water (200ml) was added and a large amount of solid precipitated, the solid was collected by filtration, the filter cake was washed with dichloromethane and dried to give compound Q4-2(42g, white solid) in 82% yield, which was used in the next step without further purification.
MS m/z(ESI):263[M+1]。
The second step is that: synthesis of Compound Q4-3
Compound Q4-2(42g, 160mmol) obtained in the previous step was added to methanol (250mL), followed by hydroxylamine hydrochloride (13.4g, 192 mmol). The reaction was stirred at room temperature for 3 hours and after TLC showed completion of the reaction, the reaction was cooled to 0 ℃ and a large amount of solid precipitated, filtered and the filter cake was washed with 100mL of ice methanol and the solid dried to give compound Q4-3(33.6g, white solid) in 84% yield which was used in the next step without further purification.
MS m/z(ESI):251[M+1]。
The third step: synthesis of Compound Q4-4
Compound Q4-3(7.5g, 29.6mmol) obtained in the previous step was added to polyphosphoric acid (50mL), the reaction temperature was raised to 120 ℃, the reaction was stirred for 5 hours, after TLC showed the end of the reaction, 150mL of ice water was added to the reaction system, pH was adjusted to about 8 with 2N aqueous sodium hydroxide solution, then the obtained aqueous phase was extracted with N-butanol (150mL x 3), the organic phases were combined, the combined organic phase was washed with saturated brine (70mL x 2), the organic phase was dried with anhydrous sodium sulfate, filtered, spin-dried and concentrated to obtain compound Q4-4(3.5g, pale yellow solid) with 50% yield.
MS m/z(ESI):215[M+1]。
The fourth step: synthesis of intermediate Q4
To compound Q4-4(3.2g, 15mmol) was added phosphorus oxychloride (50mL), followed by DIEA (5.8g, 45mmol), and the mixture was heated to 115 ℃. After 3h of reaction, TLC indicated the end of the reaction, the mixture was concentrated in vacuo and the residue was diluted with EtOAc (100ml) and slowly adjusted to pH 7 with saturated sodium bicarbonate solution. The organic phase was separated, the aqueous phase was extracted with ethyl acetate, the combined organic phases were washed with saturated brine (70mL × 2), the organic phase was dried over anhydrous sodium sulfate, filtered, and the residue obtained by spin-drying and concentration was subjected to flash silica gel chromatography (eluent petroleum ether: ethyl acetate: 3:1 to 1:1) to obtain compound Q4(2.5g, white solid) with a yield of 71.5%.
MS m/z(ESI):233[M+1]。
Synthesis of intermediate Q5
Figure BDA0002678062020000251
The first step is as follows: synthesis of Compound Q5-2
Compound Q5-1(10g, 88.4mmol) was added to a THF solution (100ml), followed by the addition of butyl 2-hydroxymethylpyrrolidine-1-carboxylate (21.4g, 106.1mmol, CAS:170491-63-1) and triphenylphosphine (34,8g, 132.6mmol) in that order, the reaction was cooled to below 5 ℃ in an ice-water bath, and DEAD (23.1g, 132.6mmol) was slowly added dropwise. After completion of the dropwise addition and stirring at room temperature for 16 hours, TLC showed the reaction to be complete, the reaction mixture was concentrated under reduced pressure, and the resulting residue was concentrated by spin drying through flash silica gel chromatography (eluent petroleum ether: ethyl acetate 10:1) to give compound Q5-2(25g, pale yellow oil) in 95.4% yield.
MS m/z(ESI):297[M+1]。
The second step is that: synthesis of Compound Q5-3
Compound Q5-2(5g, 16.8mmol) obtained in the previous step was added to ethyl acetate (50ml), 4M HCl/ethyl acetate solution (25ml) was added, the reaction was carried out at room temperature for 1h, TLC showed completion of the reaction, filtration was carried out, the solid was washed with ethyl acetate, and drying was carried out to obtain hydrochloride of compound Q5-3 (4.4g, white solid) with a yield of 96.8%.
MS m/z(ESI):197[M+1]。
The third step: synthesis of Compound Q5-4
The hydrochloride of compound Q5-3 (2.20g, 8.17mmol) obtained in the previous step was added to ethanol (50mL), followed by addition of potassium carbonate (5.64g, 40.87), and after stirring at room temperature for half an hour, the mixture was heated to 65 ℃ and stirred for 12 hours. TLC showed the reaction was complete and the mixture was filtered and the filtrate was concentrated under reduced pressure and the resulting residue was concentrated by spin drying on flash silica chromatography (eluent petroleum ether: ethyl acetate 10:1) to give compound Q5-4(1.22g, colorless oil) in 84.7% yield.
MS m/z(ESI):177[M+1]。
The fourth step: synthesis of Compound Q5-5
Compound Q5-4(1.2g, 6.8mmol) obtained in the previous step was dissolved in THF (20ml) and then cooled to-78 ℃ under N2Under the protection of atmosphere, n-butyllithium solution (5.4mL, 13.6mmol, 2.5M hexane solution) was slowly added dropwise, and after the addition, the temperature was naturally raised to 0 ℃ and stirred for 1.5 h. Then the reaction solution was cooled again to-78 deg.C, andto the above mixture was added dropwise a solution of elemental iodine (2g, 8.16mmol) in THF (10 mL). After the addition, the reaction mixture was allowed to warm to room temperature and stirred at room temperature for 2 hours. TLC shows that after the reaction is finished, saturated NH is added into the reaction liquid4Aqueous Cl (100mL), the resulting aqueous phase was extracted with ethyl acetate (100mL × 3), the organic phases were combined, the organic phase was washed with saturated brine (70mL × 2), the organic phase was dried over anhydrous sodium sulfate, filtered, and the resulting residue was concentrated by spin-drying through a silica gel column chromatography (eluent petroleum ether: ethyl acetate ═ 3:1 to 1:1) to give compound Q5-5(1.5g, yellow solid) in 72.8% yield.
MS m/z(ESI):303[M+1]。
The fifth step: synthesis of Compound Q5-6
The compound Q5-5(303mg, 1mmol) obtained in the previous step was dissolved in dioxane (5ml), and methyl 3-mercaptopropionate (180mg, 1.5mmol) and Pd were added2(dba)3(22.73mg, 0.025mmol, 0.05 equiv.) and Xantphos (14.36mg, 0.025mmol) and DIEA (387mg, 3 mmol). After stirring for 1 hour at 90 ℃ under nitrogen, the reaction mixture was concentrated under reduced pressure. The residue obtained by spin-dry concentration was subjected to silica gel chromatography (eluent petroleum ether: ethyl acetate: 3:1 to 1:1) to obtain compound Q5-6(239mg, pale yellow solid) in 81% yield.
MS m/z(ESI):295[M+1]。
And a sixth step: synthesis of intermediate Q5
Compound Q5-6(250mg, 0.85mmol) obtained in the previous step was dissolved in THF (2ml) and then cooled to 0 ℃. Sodium tert-butoxide (96mg, 1mmol) was added and after stirring at 0 ℃ for 0.5h, TLC showed the reaction was complete, the reaction mixture was diluted with PE to precipitate a large amount of solid, which was collected by filtration and washed with ethyl acetate to give an intermediate (164mg, pale yellow solid) in 84% yield.
MS m/z(ESI):209[M+1]。
Referring to the synthesis of intermediate Q5, the following intermediates were synthesized using commercially available starting materials in place of butyl 2-hydroxymethylpyrrolidine-1-carboxylate:
Figure BDA0002678062020000271
example 1: preparation of Compound represented by formula I-1
Figure BDA0002678062020000272
The synthetic route is as follows:
Figure BDA0002678062020000281
the first step is as follows: synthesis of Compound 1A
Compound Q2(220mg, 0.95mmol), Q1(267mg, 0.95mmol) and N, N-diisopropylethylamine (0.47mL, 2.84mmol) were dissolved in 5mL of dimethyl sulfoxide and reacted at 90 ℃ for 1.5 hours. After the reaction was completed, 20mL of ethyl acetate and 40mL of water were added, extraction was performed with ethyl acetate (20mL × 3), the organic phases were combined, washed with a saturated sodium oxide solution (100mL), dried over anhydrous sodium sulfate, filtered, the filtrate was collected, the filtrate was concentrated under reduced pressure, and the residue was subjected to silica gel chromatography (eluent dichloromethane: methanol: 50:1 to 10:1) to obtain compound 1A (244mg, pale yellow solid) in 64.3% yield.
MS m/z(ESI):399[M+1]。
The second step is that: synthesis of Compound 1B
Compound 1A (240mg, 0.6mmol) obtained in the previous step was added to dichloromethane (5ml), followed by DIEA (154mg, 1.2mmol) and (Boc)2(262mg, 1.2mmol), stirred at rt for 12h, TLC showed the reaction was complete, the reaction was diluted with 10ml dichloromethane, the organic phase was washed with saturated aqueous sodium chloride solution, dried over anhydrous sodium sulfate, filtered, the filtrate was collected, the filtrate was concentrated under reduced pressure, and the residue was chromatographed on silica gel (eluent dichloromethane: methanol 50: 1-10: 1) to give compound 1B (266mg, pale yellow solid) in 89% yield.
MS m/z(ESI):499[M+1]。
The third step: synthesis of Compound 1C
Will be at the topCompound 1B (250mg, 0.5mmol) obtained in one step was dissolved in dioxane (5ml), and intermediate Q5(230mg, 1mmol) and Pd were added2(dba)3(22.73mg, 0.025mmol) and Xantphos (14.36mg, 0.025mmol) and DIEA (190mg, 1.5 mmol). After stirring for 1 hour at 90 ℃ under nitrogen, the reaction mixture was concentrated under reduced pressure. The residue obtained from spin-dry concentration was chromatographed on silica gel (eluent dichloromethane: methanol 50: 1-10: 1) to give compound 1C (241mg, pale yellow solid) in 77% yield.
MS m/z(ESI):627[M+1]。
The fourth step: synthesis of Compound I-1
Compound 1C (200mg, 0.32mmol) obtained in the previous step was added to ethyl acetate (5ml), 4M HCl/ethyl acetate solution (2ml) was added and reacted at room temperature for 1h, TLC showed completion of the reaction, filtration was carried out, the solid was washed with ethyl acetate and dried to give the hydrochloride of compound I-1 (157mg, yellow solid) in 87% yield.
MS m/z(ESI):527[M+1]。
HNMR:(400MHz,DMSO)8.55-8.54(m,1H),8.48(brs,2H),8.14(s,1H),8.03-7.94(m,1H),7.92(s,1H),7.84(s,1H),7.37-7.34(m,2H),5.92-5.90(m,1H),4.75-4.72(m,1H),4.54(brs,1H),4.04-3.93(m,2H),3.77-3.75(m,2H),3.66-3.57(m,5H).3.25-3.22(m,2H),3.16-3.11(m,1H),2.17-1.93(m,5H),1.74-1.52(m,3H)。
Example 2: preparation of the Compound represented by the formula I-2
Figure BDA0002678062020000291
The synthetic procedure of example 2 referring to example 1, wherein the third step replaces compound Q5 with compound Q6, compound I-2 was synthesized.
MS m/z(ESI):527[M+1]。
HNMR:(400MHz,DMSO-d6)9.39(s,1H),8.56-8.55(m,1H),8.54(brs,2H),7.95(s,1H),7.93-7.91(m,1H),7.40-7.34(m,2H),5.92-5.90(m,1H),4.67-4.65(m,1H),4.64(brs,1H),4.52-4.10(m,2H),3.70-3.68(m,1H),3.61-3.43(m,6H),3.16-3.12(m,1H),2.15-2.12(m,1H),2.03-1.93(m,4H),1.71-1.53(m,3H)。
Example 3: preparation of Compound represented by formula I-3
Figure BDA0002678062020000301
The synthetic procedure of example 3 referring to example 1, wherein the third step replaces compound Q5 with compound Q7, compound I-3 was synthesized.
MS m/z(ESI):527[M+1]。
HNMR:(400MHz,DMSO-d6)9.39(s,1H),8.56-8.55(m,1H),8.54(brs,2H),7.95(s,1H),7.93-7.91(m,1H),7.40-7.34(m,2H),5.92-5.90(m,1H),4.67-4.65(m,1H),4.64(brs,1H),4.52-4.10(m,2H),3.70-3.68(m,1H),3.61-3.43(m,6H),3.16-3.12(m,1H),2.15-2.12(m,1H),2.03-1.93(m,4H),1.71-1.53(m,3H)。
Example 4: preparation of Compound represented by formula I-4
Figure BDA0002678062020000311
The synthetic procedure of example 4 referring to example 1, wherein compound I-4 was synthesized by substituting compound Q2 with compound Q3 in the first step and compound Q5 with compound Q7 in the third step.
MS m/z(ESI):528[M+1]。
HNMR:(400MHz,DMSO)9.39(s,1H),8.56-8.55(m,1H),8.54(brs,2H),7.95(s,1H),7.93-7.91(m,1H),7.40-7.34(m,2H),5.92-5.90(m,1H),4.67-4.65(m,1H),4.64(brs,1H),4.52-4.10(m,2H),3.70-3.68(m,1H),3.61-3.43(m,6H),3.16-3.12(m,1H),2.15-2.12(m,1H),2.03-1.93(m,4H),1.71-1.53(m,3H)。
Example 5: preparation of Compound represented by the formula I-5
Figure BDA0002678062020000312
The synthetic procedure of example 5 referring to example 1, wherein compound Q5 is replaced with compound Q8 in the third step, compound I-5 is synthesized.
MS m/z(ESI):528[M+1]。
HNMR:(400MHz,DMSO)8.57-8.56(m,1H),8.44(brs,3H),8.12(s,1H),7.96-7.94(m,1H),7.89-7.88(m,1H),7.69-7.68(m,1H),7.39-7.35(m,2H),6.05(d,J=6.4MHz,1H),4.82-4.78(m,1H),4.55-4.54(m,1H),4.11-3.96(m,5H),3.78-3.73(m,3H),2.03-1.93(m,4H).1.77-1.72(m,2H),1.64(m,1H),0.77-0.76(m,2H),0.70-0.67(m,2H)。
Example 6: preparation of Compound represented by formula I-6
Figure BDA0002678062020000321
The synthetic procedure of example 6 referring to example 1, wherein compound I-6 was synthesized by substituting compound Q2 with compound Q4 in the first step and compound Q5 with compound Q7 in the third step.
MS m/z(ESI):528[M+1]。
HNMR:(400MHz,DMSO-d6)8.73(brs,3H),8.14(s,1H),8.56-8.55(m,2H),8.27(s,1H),8,06-8.05(m,1H),7.40-7.38(m,1H),7.33-7.31(m,1H),6.21-6.20(m,1H),5.07-4.98(m,2H),4.85-4.83(m,1H),4.52(m,1H),3.73-3.49(m,7H),2.17(m,2H).2.03-1.99(m,4H),1.74-1.43(m,4H)。
Effect example 1: inhibition of cell proliferation assay
Experimental materials:
RPMI-1640 medium, penicillin/streptomycin antibiotics from Verben, fetal bovine serum from Biosera. 3D CellTiter-Glo (cell viability chemiluminescence detection reagent) reagents were purchased from Promega. Staurosporine was purchased from the pottery biochemistry. The NCI-H358 cell line or MV-4-11 cell line was purchased from Wuhan Poncide Life technologies, Inc. Nivo multi-label analyzer (PerkinElmer).
The experimental method comprises the following steps:
NCI-H358 cells or MV-4-11 cells were seeded in 96-well ultra-low adsorption U-plates in 80. mu.L cell suspension per well containing 2000 NCI-H358 cells. The cell plates were placed in a carbon dioxide incubator overnight.
The test compounds were diluted 3-fold with a calandria to the 9 th concentration, i.e. from 600 μ M to 100nM, setting up a duplicate well experiment. Add 78. mu.L of medium to the intermediate plate, transfer 2. mu.L of each well of the gradient dilution compound to the intermediate plate according to the corresponding position, mix well and transfer 20. mu.L of each well to the cell plate. The concentration of compound transferred to the cell plate ranged from 3. mu.M to 0.5 nM. The cell plates were placed in a carbon dioxide incubator for 5 days.
On the day of reading, 100. mu.L of cell viability chemiluminescence detection reagent was added to the cell plate per well and the luminescence signal was stabilized by incubation for 10 minutes at room temperature. Reading with a multi-label analyzer.
Max holes: DMSO solvent wells
Min hole: staurosporine treated wells
And (3) data analysis:
the original data was converted to inhibition rate, IC, using the equation (Sample-Min)/(Max-Min) × 100%50The values of (A) can be obtained by curve fitting of four parameters (obtained in the GraphPad Prism "log (inhibitor)" vs. response- -Variable slope "mode). Table 1 provides the inhibitory activity of the compounds of the present invention on proliferation of NCI-H358 cells or MV-4-11 cells.
Table 1: inhibition of cell proliferation Activity data (IC) of Compounds of the examples of the invention50)
Figure BDA0002678062020000331
From the experimental results in table 1 we can see that: the compounds of the examples shown in the present invention have good activity in inhibiting NCI-H358 or MV-4-11 cell proliferation. The activity of part of compounds is far more than that of a reference substance, and the compounds show extremely important anti-tumor prospect.
Effect example 2: drug metabolism test
1) Using the compounds of the invention prepared in the above examples, the oral drug was formulated as a 0.3mg/mL clear solution (2% DMSO + 30% PEG300+ 2% Tween 80+ 66% H)2O), intravenous drug was formulated as a 0.2mg/mL clear solution (2% DMSO + 30% PEG300+ 2% Tween 80+ 66% H)2O)。
2) Male CD-1 mice, 3 each per group, weighing 27-28g, were provided by Shanghai Si Laike laboratory animal responsibility Co., Ltd. The test mice are given an environmental adaptation period of 2-4 days before the experiment, are fasted for 8-12h before the administration, are fed with water after the administration for 2h, and are fed with food after 4 h.
3) After the mice are fasted but can drink water freely for 12 hours, blank plasma at 0 moment is adopted;
4) taking the mice in the step 2), and orally taking (PO) the compound to be detected for 3 mg/kg; intravenous (IV) administration of 1mg/kg of test compound;
5) continuously taking blood from fundus venous plexus 5min, 15min, 30min, 1h, 2h, 4h, 8h, 10h and 24h after oral administration, placing in an EP tube distributed with heparin, centrifuging at 8000rpm/min for 5min, taking upper layer plasma, freezing at-20 deg.C, and analyzing by LC-MS/MS;
6) calculating pharmacokinetic parameters by adopting WinNonlin software according to the blood concentration-time data obtained in the step 5), wherein the specific data are shown in a table 2.
Table 2: pharmacokinetic data for the Compounds of the examples of the invention
Figure BDA0002678062020000351
The pharmacokinetic experimental data are shown in table 2, and the results show that after the compound is orally or intravenously administered to a mouse, the compound has very high exposure amount, very good half-life period, tissue distribution, area under the curve and bioavailability in animal plasma, and part of the compound is superior to a reference product TNO-155, so that the compound has good clinical application prospect.
In the description herein, references to the description of the term "one embodiment," "some embodiments," "an example," "a specific example" or "some examples" or the like are intended to mean that a particular feature, structure, material, or characteristic described in connection with the embodiment or example is included in at least one embodiment or example of the invention. In this specification, the schematic representations of the terms used above do not necessarily refer to the same embodiment or example. Furthermore, the particular features, structures, materials, or characteristics described may be combined in any suitable manner in any one or more embodiments or examples.
Although embodiments of the present invention have been shown and described above, it is understood that the above embodiments are exemplary and should not be construed as limiting the present invention, and that variations, modifications, substitutions and alterations can be made in the above embodiments by those of ordinary skill in the art without departing from the principle and spirit of the present invention.

Claims (17)

1. A compound of formula I, or a pharmaceutically acceptable form thereof, which is a pharmaceutically acceptable salt, or a stereoisomer, or a tautomer, or a hydrate, or a solvate, or a metabolite, or a prodrug;
Figure FDA0002678062010000011
wherein the content of the first and second substances,
X1、X2or X3Independently selected from CR5Or N, and X1、X2Or X3At least one of which is N;
X4or X5Independently selected from C or N;
R1、R2、R3and R4Independently selected from H, halogen, hydroxy, amino, cyano, C2-C8Alkenyl radical, C2-C8Alkynyl, C1-C8Aldehyde group, C1-C8Alkyl radical, C1-C8Heteroalkyl group, C3-C8Cycloalkyl radical, C3-C8Heterocycloalkyl radical, C1-C8Alkoxy or C1-C3Haloalkoxy, said C1-C8Alkyl radical, C1-C8Heteroalkyl group, C3-C8Cycloalkyl radical, C3-C8Heterocycloalkyl radical, C1-C8Alkoxy or C1-C3Haloalkoxy is optionally substituted with one or more R5Substituted when by more than one R5When substituted, R5May be the same or different;
R5selected from H, halogen, hydroxy, amino, cyano, C1-C3Alkyl radical, C1-C3Heteroalkyl group, C3-C8Cycloalkyl radical, C3-C8Heterocycloalkyl radical, C1-C3Alkoxy or C1-C3A haloalkoxy group;
n is selected from 0, 1,2,3,4 and 5;
m is selected from 0, 1,2,3,4 and 5;
ring A is independently selected from C3-C8Cycloalkyl radical, C6-C12Spiro cycloalkyl, C3-C8Heterocycloalkyl radical, C6-C10Aryl or C5-C12A heteroaryl group;
ring B is independently selected from C3-C8Cycloalkyl radical, C3-C8Heterocycloalkyl radical, C6-C10Aryl or C5-C12A heteroaryl group;
the heteroatoms or heteroatom groups contained in the heteroalkyl, heterocycloalkyl, heteroaryl groups are each independently selected from-C (═ O) N (R)5)-、-N(R5)-、-NH-、-N=、-O-、-S-、-C(=O)O-、-C(=O)-、-C(=S)-、-S(=O)-、-S(=O)2-and-N (R)5)C(=O)N(R5) The number of heteroatoms or groups of heteroatoms is independently selected from 1,2 and 3, respectively.
2. A compound, or pharmaceutically acceptable form thereof, according to claim 1, wherein when X is4When N, said ring A is independently selected from the group consisting of a pyrrole ring, a cycloalkyl spiropyrrole ring, a piperidine ring, a cycloalkyl spiropiperidine ring or a dihydropyrazole ring.
3. A compound, or pharmaceutically acceptable form thereof, according to claim 2, wherein when X is4When N, the ring A is independently selected from
Figure FDA0002678062010000021
Figure FDA0002678062010000022
4. A compound, or pharmaceutically acceptable form thereof, according to claim 1, wherein when X is4When C, the ring A is independently selected from a benzene ring, a pyridine ring, a pyrimidine ring, a pyrazine ring or a pyridazine ring.
5. The compound, or pharmaceutically acceptable form thereof, according to claim 4, wherein when X is4When C, the ring A is independently selected from
Figure FDA0002678062010000023
Figure FDA0002678062010000024
6. A compound, or pharmaceutically acceptable form thereof, according to any one of claims 1 to 5, wherein when X is5When N, the ring B is independently selected from a pyrrole ring, a piperidine ring, a piperazine ring, or a morpholine ring.
7. The compound, or pharmaceutically acceptable form thereof, according to claim 6, wherein when X is5When N, the ring B is independently selected from
Figure FDA0002678062010000031
8. A compound, or pharmaceutically acceptable form thereof, according to any one of claims 1 to 5, wherein when X is5When C, the ring B is independently selected from a benzene ring, a pyridine ring, a pyrimidine ring, a pyrazine ring or a pyridazine ring.
9. The compound, or pharmaceutically acceptable form thereof, according to claim 8, wherein when X is5When C, the ring B is independently selected from
Figure FDA0002678062010000032
Figure FDA0002678062010000033
10. A compound, or pharmaceutically acceptable form thereof, according to any one of claims 1 to 9, wherein R is1、R2、R3And R4Each independently selected from H, halogen, hydroxy, amino, cyano, C2-C4Alkenyl radical, C1-C4Alkyl radical, C1-C4Heteroalkyl group, C3-C6Cycloalkyl radical, C3-C6Heterocycloalkyl radical, C1-C4Alkoxy or C1-C3Haloalkoxy, said C1-C4Alkyl radical, C1-C4Heteroalkyl group, C3-C6Cycloalkyl radical, C3-C6Heterocycloalkyl radical, C1-C4Alkoxy or C1-C3Haloalkoxy is optionally substituted with one or more R5Substituted when by more than one R5When substituted, R5May be the same or different;
R5selected from H, F, Cl, Br, hydroxyl, amino, cyano, methyl, ethyl, cyclopropyl, methoxy, ethoxy, trifluoromethoxy.
11. A compound, or pharmaceutically acceptable form thereof, according to claim 10, wherein R is1、R2、R3And R4Each independently selected from H, halogen, hydroxy, amino, cyano, methyl, ethyl, isopropyl, cyclopropyl, vinyl, methoxy, ethoxy, isopropoxy, trifluoromethyl, difluoromethyl, trifluoromethoxy, trifluoromethyl, and mixtures thereof,-OCH2CHF2、-N(CH3)2、-NH(CH3) or-OCH2CF3
12. The compound of claim 1, or a pharmaceutically acceptable form thereof, wherein the compound of formula I is selected from the group consisting of compounds of formula (II), formula (III), or formula (IV):
Figure FDA0002678062010000041
wherein R is1、R2、R3、R4、X4、X5Ring a and ring B are as defined in claim 1, m is selected from 0, 1,2,3,4 and 5, and n is selected from 0, 1,2,3,4 and 5.
13. The compound of claim 12, or a pharmaceutically acceptable form thereof, wherein the compound of formula I is selected from the group consisting of compounds of formulae (II-1) to (IV-3) below:
Figure FDA0002678062010000042
Figure FDA0002678062010000051
wherein R is1、R2、R3、R4、X4、X5Ring a and ring B are as defined in claim 1, m is selected from 0, 1,2,3,4 and 5, and n is selected from 0, 1,2,3,4 and 5.
14. The compound, or pharmaceutically acceptable form thereof, according to claim 13, wherein the compound of formula I is any one of the following:
Figure FDA0002678062010000061
15. a pharmaceutical composition comprising an effective amount of a compound according to any one of claims 1 to 14, or a pharmaceutically acceptable form thereof, which is a pharmaceutically acceptable salt, or a stereoisomer, or a tautomer, or a hydrate, or a solvate, or a metabolite, or a prodrug, and at least one pharmaceutical excipient.
16. Use of a compound of any one of claims 1 to 14, or a pharmaceutically acceptable form thereof, or a pharmaceutical composition of claim 15, in the manufacture of a medicament of an SHP-2 inhibitor, said pharmaceutically acceptable form being a pharmaceutically acceptable salt, or a stereoisomer, or a tautomer, or a hydrate, or a solvate, or a metabolite, or a prodrug.
17. Use of a compound according to any one of claims 1 to 14, or a pharmaceutically acceptable form thereof, or a pharmaceutical composition according to claim 15, for the manufacture of a medicament for the treatment and/or prevention of cancer, including noonan syndrome, leopard syndrome, juvenile myelomonocytic leukemia, neuroblastoma, melanoma, acute myelogenous leukemia, breast cancer, esophageal cancer, lung cancer, colon cancer, stomach cancer, anaplastic large-cell lymphoma, glioblastoma, non-small cell lung cancer or head and neck tumors, preferably non-small cell lung cancer, esophageal cancer and head and neck tumors.
CN202010954235.XA 2020-09-11 2020-09-11 Pyrimido five-membered nitrogen heterocyclic compound and use thereof Pending CN114163457A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202010954235.XA CN114163457A (en) 2020-09-11 2020-09-11 Pyrimido five-membered nitrogen heterocyclic compound and use thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010954235.XA CN114163457A (en) 2020-09-11 2020-09-11 Pyrimido five-membered nitrogen heterocyclic compound and use thereof

Publications (1)

Publication Number Publication Date
CN114163457A true CN114163457A (en) 2022-03-11

Family

ID=80476085

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010954235.XA Pending CN114163457A (en) 2020-09-11 2020-09-11 Pyrimido five-membered nitrogen heterocyclic compound and use thereof

Country Status (1)

Country Link
CN (1) CN114163457A (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022235870A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Ras inhibitors for the treatment of cancer
WO2022235866A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Covalent ras inhibitors and uses thereof
WO2022235864A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Ras inhibitors
WO2023172940A1 (en) 2022-03-08 2023-09-14 Revolution Medicines, Inc. Methods for treating immune refractory lung cancer
WO2023240263A1 (en) 2022-06-10 2023-12-14 Revolution Medicines, Inc. Macrocyclic ras inhibitors

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022235870A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Ras inhibitors for the treatment of cancer
WO2022235866A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Covalent ras inhibitors and uses thereof
WO2022235864A1 (en) 2021-05-05 2022-11-10 Revolution Medicines, Inc. Ras inhibitors
WO2023172940A1 (en) 2022-03-08 2023-09-14 Revolution Medicines, Inc. Methods for treating immune refractory lung cancer
WO2023240263A1 (en) 2022-06-10 2023-12-14 Revolution Medicines, Inc. Macrocyclic ras inhibitors

Similar Documents

Publication Publication Date Title
CN111484477B (en) Benzopyridone heterocyclic compound and application thereof
CN114163457A (en) Pyrimido five-membered nitrogen heterocyclic compound and use thereof
WO2021043077A1 (en) Substituted pyrazine compound and preparation method therefor and use thereof
KR20230019101A (en) Benzothiazolyl biaryl compounds, methods of preparation and uses
KR101783642B1 (en) Heterocyclic Compounds and Pharmaceutical Compositions Comprising the Same
TW201734018A (en) Substituted pyrrolopyrimidine CDK inhibitor, pharmaceutical composition containing same and use thereof
CN111153899A (en) Substituted pyridine compound, preparation method and application thereof
JP2017508779A (en) Substituted 4,5,6,7-tetrahydro-pyrazolo [1,5-α] pyrazine derivatives and 5,6,7,8-tetrahydro-4H-pyrazolo [1,5-α] [1, ROS1 inhibitors 4] Diazepine derivatives
WO2017128917A1 (en) Parazole condensed-ring derivatives and preparation method thereof and application thereof in treatment of cancers, inflammation and immune diseases
WO2022193871A1 (en) Preparation and use of krasg12d mutant protein inhibitor
CN112745335A (en) Tri-heterocyclic compound and application thereof
JP2021501215A (en) Amino-substituted nitrogen-containing condensed ring compound, its preparation method and use
CN113024544A (en) Cyano-containing heterocyclic compound and application thereof
CN112851663A (en) Fused heterocyclic compound and application thereof
CN116568681A (en) SOS1 inhibitor, pharmaceutical composition containing same and application thereof
WO2022063297A1 (en) Quinazoline derivative, preparation method therefor and use thereof
CN116194446A (en) SOS1 inhibitor, pharmaceutical composition containing same and application thereof
CN112867717A (en) Compounds useful as kinase inhibitors and uses thereof
CN112707905A (en) Tri-heterocyclic compound and preparation method and application thereof
CN115073469B (en) Preparation and application of pyrrolopyrimidine compound as kinase inhibitor
KR102485731B1 (en) JAK Enzyme Inhibitors and Manufacturing Methods and Uses Thereof
KR20230058466A (en) Methods for preparing novel RHO-associated protein kinase inhibitors and intermediates in manufacturing methods
CN115073451A (en) KRAS G12D Preparation and application of mutant protein inhibitor
CN114605390A (en) Compound with CDK kinase inhibitory activity, pharmaceutical composition and use thereof
CN115028633A (en) Preparation and application of pyrrolopyrimidine compounds

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