CN114671866A - Aryl or heteroaryl pyridone or pyrimidone derivative and preparation method and application thereof - Google Patents

Aryl or heteroaryl pyridone or pyrimidone derivative and preparation method and application thereof Download PDF

Info

Publication number
CN114671866A
CN114671866A CN202011563650.9A CN202011563650A CN114671866A CN 114671866 A CN114671866 A CN 114671866A CN 202011563650 A CN202011563650 A CN 202011563650A CN 114671866 A CN114671866 A CN 114671866A
Authority
CN
China
Prior art keywords
group
formula
alkyl
compound
cycloalkyl
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
CN202011563650.9A
Other languages
Chinese (zh)
Inventor
吕彬华
崔大为
刘连军
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.)
Suzhou Zelgen Biopharmaceutical Co Ltd
Shanghai Zelgen Pharmatech Co Ltd
Original Assignee
Suzhou Zelgen Biopharmaceutical Co Ltd
Shanghai Zelgen Pharmatech 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 Suzhou Zelgen Biopharmaceutical Co Ltd, Shanghai Zelgen Pharmatech Co Ltd filed Critical Suzhou Zelgen Biopharmaceutical Co Ltd
Priority to CN202011563650.9A priority Critical patent/CN114671866A/en
Priority to US17/758,733 priority patent/US20230118795A1/en
Priority to PCT/CN2021/071331 priority patent/WO2021143693A1/en
Priority to EP21741983.7A priority patent/EP4092026A4/en
Priority to CN202180008854.2A priority patent/CN115175908A/en
Priority to PCT/CN2021/099875 priority patent/WO2021249563A1/en
Priority to PCT/CN2021/141360 priority patent/WO2022135591A1/en
Publication of CN114671866A publication Critical patent/CN114671866A/en
Pending legal-status Critical Current

Links

Classifications

    • 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
    • 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
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings

Abstract

The invention relates to aryl or heteroaryl pyridone or pyrimidone derivatives, and a preparation method and application thereof. Specifically, the compound has a structure shown in a formula (I), and the invention also discloses a preparation method of the compound and application of the compound as KRASG12CUse of inhibitors of KRASG12CHas good selective inhibition effect, better pharmacodynamics and pharmacokinetic performance and lower toxic and side effect.

Description

Aryl or heteroaryl pyridone or pyrimidone derivative and preparation method and application thereof
Technical Field
The invention belongs to the field of medicines, and particularly relates to an aryl or heteroaryl pyridone or pyrimidone derivative, and a preparation method and application thereof.
Background
Lung cancer is one of the leading causes of death in human cancers. Lung cancer can be divided into Small Cell Lung Cancer (SCLC) and non-small cell lung cancer (NSCLC) according to cell type, with NSCLC accounting for 85% of all lung cancer patients. The global NSCLC market was statistically estimated to be $ 209 billion in 2016, with the U.S. market taking up half, followed by Japan, Germany, and China. From the current trend, the market for non-small cell lung cancer is continuing to grow, and the global market is expected to reach $ 540 billion in 2023 (Nature, 2018; 553(7689):446- > 454).
At present, the main therapeutic drugs of NSCLC include chemotherapeutic drugs, molecular targeted drugs, tumor immunotherapy and the like. The chemotherapy drugs mainly comprise gemcitabine, paclitaxel, platinum drugs and the like, but the drugs generally have poor selectivity and high toxicity, so that relatively strong toxic and side effects are caused. In recent years, molecular targeted drugs gradually become research hotspots due to the obvious advantages of high selectivity, relatively small toxic and side effects, accurate treatment and the like. Existing NSCLC molecular targeted drugs include EGFR inhibitors (e.g., afatinib, gefitinib, erlotinib, lapatinib, dacomitinib, icotinib, pyrroltinib, norcetitinib (Rociletinib), ocitinib, etc.), ALK inhibitors (e.g., ceritinib, idetinib, bocatinib, loratinib, ocatinib, etc.), and VEGFR inhibitors (sorafenib, regorafenib, cabozantinib, sunitinib, doranitinib, etc.).
Within lung cancer patients, KRAS mutations are frequently detected, accounting for approximately 32% of all oncogene mutations. Wherein KRASG12CMutations account for 44% of all oncogene mutations within NSCLC. So far, the market has not been directed to KRAS G12CMutated drugs are approved for marketing.
Due to KRASG12CThe target proteins are pathologically associated with a variety of diseases, and there is therefore a need for novel KRASG12CThe inhibitor is used for clinical treatment. KRAS with high selectivity and high activityG12CThe inhibitor may be on KRASG12CCancer caused by mutationThese diseases are more effective treatments, and reduce the potential for off-target effects, thus having more urgent clinical needs.
Disclosure of Invention
The invention aims to provide a novel pair of KRASG12CCompounds with selective inhibition and/or better pharmacodynamic properties and uses thereof.
In a first aspect of the invention, there is provided a compound of formula (I), a stereoisomer, a tautomer, a crystalline form, a pharmaceutically acceptable salt, a hydrate, a solvate, or a prodrug thereof:
Figure BDA0002861043520000021
in the formula:
A. b are the same or different and are each independently selected from the group consisting of: CH. CR5Or N;
x is selected from the group consisting of: 4-14 membered saturated or unsaturated heterocyclic group, C4-C14Cycloalkyl radical, C6-C14Aryl or 5-14 membered heteroaryl, wherein said heterocyclyl, cycloalkyl, aryl or heteroaryl may optionally be substituted with one or more (e.g. 2, 3 or 4) R8Substituted;
u, V, W and Q are the same or different and are each independently selected from the group consisting of: CH. CR3Or N;
R1Selected from the group consisting of:
Figure BDA0002861043520000022
Figure BDA0002861043520000023
wherein, the first and the second end of the pipe are connected with each other,
Figure BDA0002861043520000024
represents a double bond "═ or a triple bond" ≡ ";
RAis absent or is independently selected from the group consisting of: hydrogen, deuterium, fluorine, cyano, or C1-C3An alkyl group; each RBIndependently selected from the group consisting of: hydrogen and deuteriumCyano or C1-C3An alkyl group; wherein the alkyl group may be substituted with one or more (e.g. 2, 3 or 4) substituents selected from the group consisting of: deuterium, halogen, cyano, amino, C3-C7Cycloalkyl, 4-7 membered heterocyclyl, NHR9Or NR9R10;R9And R10Each independently is C1-C3An alkyl group; or R9,R10Together with the N atom to which they are attached form a substituted or unsubstituted 4-8 membered heterocyclyl;
p is an integer of 1 or 2;
R2selected from the group consisting of substituted: c6-C14Aryl, 5-14 membered heteroaryl, wherein said substitution means substitution by one or more groups selected from the group consisting of: r ', -SR ', -SOR ', -SO2R'、-SO2NR'R”、-NR'SO2R ", -P (═ O) R' R"; with the proviso that C is6-C14Aryl, 5-14 membered heteroaryl, having at least one substituent selected from: -SR ', -SOR', -SO2R'、-SO2NR'R”、-NR'SO2R ", or-P (═ O) R' R"; r 'and R' are the same or different and are each independently selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C1-C6Alkyl radical, C3-C8Cycloalkyl radical, C4-C10Cycloalkenyl, 4-8 membered heterocyclyl, C 6-C14Aryl, 5-14 membered heteroaryl; or when R ' and R ' are attached to the same N atom, R ' together with the N atom to which they are attached form a substituted or unsubstituted 4-8 membered heterocyclyl;
R3selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C18Alkyl, deuterated C1-C18Alkyl, halo C1-C18Alkyl radical, C3-C20Cycloalkyl, C1-C18Alkoxy, deuterated C1-C18Alkoxy, halo C1-C18Alkoxy, halogen, nitro, hydroxyl, cyano, ester, amino, amido, sulfamide, carbamido, 4-20-membered heterocyclic radical, C6-C14Aryl, 5-14 membered heteroaryl;
l is selected from the group consisting of: a bond, -C (O) -, C1-C3An alkylene group;
R4selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C18Alkyl, deuterated C1-C18Alkyl, halo C1-C18Alkyl radical, C3-C20Cycloalkyl radical, C1-C18Alkoxy, deuterated C1-C18Alkoxy, halo C1-C18Alkoxy, amino, hydroxy, 4-20 membered heterocyclic group, C6-C14Aryl, 5-14 membered heteroaryl;
R5selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C18Alkyl, deuterated C1-C18Alkyl, halo C1-C18Alkyl radical, C3-C20Cycloalkyl radical, C1-C18Alkoxy, deuterated C1-C18Alkoxy, halo C1-C18Alkoxy, halogen, nitro, hydroxyl, cyano, ester, amino, amido, sulfamide, carbamido, 4-20-membered heterocyclic radical, C6-C14Aryl, 5-14 membered heteroaryl;
R6Selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C18Alkyl, deuterated C1-C18Alkyl, halo C1-C18Alkyl radical, C3-C20Cycloalkyl radical, C1-C18Alkoxy, deuterated C1-C18Alkoxy, halo C1-C18Alkoxy, 4-to 20-membered heterocyclic group, C6-C14Aryl, 5-14 membered heteroaryl;
R8independently selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C18Alkyl, deuterated C1-C18Alkyl, halo C1-C18Alkyl radical, C3-C20Cycloalkyl radical, C1-C18Alkoxy, deuterated C1-C18Alkoxy radical,Halogen substituted C1-C18Alkoxy, amino, hydroxy, 4-20 membered heterocyclic group, C6-C14Aryl, 5-14 membered heteroaryl;
wherein, the term "substituted" when not specifically stated, means substituted by one or more groups selected from the group consisting of: hydrogen, deuterium, C1-C18Alkyl, deuterated C1-C18Alkyl, halo C1-C18Alkyl radical, C3-C20Cycloalkyl radical, C1-C18Alkoxy, deuterated C1-C18Alkoxy, halo C1-C18Alkoxy radical, C6-C14Aryl, 5-14 membered heteroaryl, 4-20 membered heterocyclyl, halogen, nitro, hydroxy, cyano, ester, amine, NRbC(=O)ORe、OC(=O)Re、OC(=O)NRbRcAmide, sulfonamide, or ureido groups; rb、RcMay independently represent hydrogen, deuterium, C1-C6 alkyl, C3-C8 cycloalkyl, 4-8 membered heterocyclyl, 5-14 membered heteroaryl or C6-C14 aromatic ring, or RbAnd RcTogether with the N atom, may form a 4-8 membered heterocyclic group; r eMay independently represent hydrogen, C1-C6 alkyl, C3-C8 cycloalkyl, C2-C6 alkenyl, C3-C6 cycloalkenyl, C2-C6 alkynyl, 4-8 membered heterocyclyl, 5-14 membered heteroaryl, or C6-C14 aryl ring;
the limiting conditions are as follows:
when B is N, R1Selected from the group consisting of:
Figure BDA0002861043520000031
wherein p is 2;
when B is CH or CR5When R is1Selected from the group consisting of:
Figure BDA0002861043520000032
Figure BDA0002861043520000033
wherein p is 2;
when V is C (Cl), R2Is not selected from:
Figure BDA0002861043520000034
and L is not selected from a bond; and R4Is not selected from:
Figure BDA0002861043520000035
in another preferred embodiment, R8Independently selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C6Alkyl, deuterated C1-C6Alkyl, halo C1-C6Alkyl radical, C3-C8Cycloalkyl radical, C1-C6Alkoxy, deuterated C1-C6Alkoxy, halo C1-C6Alkoxy, amino, hydroxy, 4-8 membered heterocyclyl; the substitution means substitution with one or more groups selected from the group consisting of: hydrogen, deuterium, halogen, nitro, hydroxyl, cyano, ester, amine, amide, sulfonamide or urea groups.
In another preferred embodiment, R8Independently selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C18Alkyl, deuterated C1-C18Alkyl, halo C1-C18An alkyl group; wherein said substitution is by cyano.
In another preferred embodiment, R1Selected from the group consisting of: -C (O) C (R)A)=C(RB)2、-S(O)2C(RA)=C(RB)2、-NR6C(O)C(RA)=C(RB)2or-NR6S(O)2C(RA)=C(RB)2
Wherein R isAIndependently selected from the group consisting of: hydrogen, deuterium, fluoro, cyano, or C 1-C3An alkyl group; each RBThe same or different, and independently selected from the group consisting of: hydrogen, deuterium, cyano, or C1-C3An alkyl group; wherein the alkyl group may be substituted with one or more substituents selected from the group consisting of: deuterium, halogen, cyano, amino, C3-C7Cycloalkyl, 4-7 membered heterocyclyl, NHR9Or NR9R10;R9And R10Each independently is C1-C3An alkyl group; or R9,R10Together with the N atom to which they are attached form a substituted or unsubstituted 4-8 membered heterocyclyl;
R6selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C6Alkyl, deuterated C1-C6Alkyl, halo C1-C6Alkyl radical, C3-C8Cycloalkyl radical, C1-C6Alkoxy, deuterated C1-C6Alkoxy, halo C1-C6Alkoxy, 4-to 8-membered heterocyclic group, C6-C14Aryl, 5-14 membered heteroaryl;
wherein, the term "substituted" when not specifically stated, means substituted by one or more groups selected from the group consisting of: hydrogen, deuterium, C1-C6Alkyl, deuterated C1-C6Alkyl, halo C1-C6Alkyl radical, C3-C8Cycloalkyl radical, C1-C6Alkoxy, deuterated C1-C6Alkoxy, halo C1-C6Alkoxy radical, C6-C10Aryl, 5-10 membered heteroaryl, 4-8 membered heterocyclyl, halogen, nitro, hydroxy, cyano, ester, amine, amide, sulfonamide, or ureido;
the limiting conditions are as follows:
when B is N, R1Selected from the group consisting of: -C (O) C (R)A)=C(RB)2or-S (O)2C(RA)=C(RB)2
When B is CH or CR 5When R is1Selected from the group consisting of: -NR6C(O)C(RA)=C(RB)2or-NR6S(O)2C(RA)=C(RB)2
In another preferred embodiment, R1is-C (O) C (R)A)=C(RB)2Wherein RA is independently selected from the group consisting of: hydrogen, fluorine; each RBThe same or different, and independently selected from the group consisting of: hydrogen or C1-C3An alkyl group, wherein the alkyl group may be substituted with one or more substituents selected from the group consisting of: deuterium, halogen, cyano, amino, C3-C7Cycloalkyl, 4-7 membered heterocyclyl, NHR9Or NR9R10;R9And R10Each independently is C1-C3An alkyl group; or R9,R10Together with the N atom to which they are attached form a 4-8 membered heterocyclic group.
In another preferred embodiment, R2Selected from the group consisting of substituted: phenyl, 5-6 membered heteroaryl, wherein said substitution means substitution by one or more groups selected from the group consisting of: r', -SO2R'、-SO2NR'R”、-NR'SO2R ", -P (═ O) R' R"; r 'and R' are the same or different and are each independently selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C1-C6Alkyl radical, C3-C6Cycloalkyl radical, C4-C6Cycloalkenyl, 4-8 membered heterocyclyl, C6-C10Aryl, 5-10 membered heteroaryl; or when R ' and R ' are attached to the same N atom, R ' together with the N atom to which they are attached form a substituted or unsubstituted 4-6 membered heterocyclyl; wherein said substitution is by one or more groups selected from the group consisting of: hydrogen, deuterium, C 1-C6Alkyl, deuterated C1-C6Alkyl, halo C1-C6Alkyl radical, C3-C8Cycloalkyl radical, C1-C6Alkoxy, deuterated C1-C6Alkoxy, halo C1-C6Alkoxy radical, C6-C10Aryl, 5-10 membered heteroaryl, 4-8 membered heterocyclyl, halogen, nitro, hydroxy, cyano, ester, amine, amide, sulfonamide, or ureido.
In another preferred embodiment, R3Is halogen.
In another preferred embodiment, R4Selected from the group consisting of substituted or unsubstituted: phenyl or 5-6 membered heteroaryl, wherein said substitution is selected fromSubstituted with one or more groups of the following group: hydrogen, deuterium, halogen, ester group, NRbC(=O)ORe、OC(=O)Re、OC(=O)NRbRcAmino, halo C1-C18Alkyl (preferably halogenated C)1-C6Alkyl, more preferably halo C1-C3Alkyl), hydroxy; rb、RcMay independently represent hydrogen, deuterium, C1-C6 alkyl, C3-C8 cycloalkyl, 4-8 membered heterocyclyl, 5-14 membered heteroaryl or C6-C14 aromatic ring, or RbAnd RcTogether with the N atom, may form a 4-8 membered heterocyclic group; reCan independently represent hydrogen, C1-C6 alkyl, C3-C8 cycloalkyl, C2-C6 alkenyl, C3-C6 cycloalkenyl, C2-C6 alkynyl, 4-8 membered heterocyclyl, 5-14 membered heteroaryl, or C6-C14 aryl ring.
In another preferred embodiment, A, B are the same or different and are each independently CH or N.
In another preferred embodiment, Q is N.
In another preferred embodiment, U is N.
In another preferred embodiment, V, W are each independently CR3,R3Is H or halogen.
In another preferred embodiment, the compound of formula (I), its stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug has a structure represented by formula (II-a) or (II-B):
Figure BDA0002861043520000051
in the formula:
R1、R2、R4a, B, L, X, U, V, W, Q are as defined above.
In another preferred embodiment, the compound of formula (I), its stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug has the structure shown in formula (III):
Figure BDA0002861043520000052
R1、R2、R4x, L, U, V, W, Q are as defined above.
In another preferred embodiment, the compound of formula (I), its stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug has the structure shown in formula (IV):
Figure BDA0002861043520000053
in the formula:
R1、R2、R4、R8l, U, V, W, Q are as defined above.
In another preferred embodiment, the compound of formula (I), its stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug has the structure shown in formula (V):
Figure BDA0002861043520000061
In the formula:
R1、R2、R4、R8u, V, W, Q are as defined above.
In another preferred embodiment, the compound of formula (I), its stereoisomer, tautomer, crystalline form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug has the structure shown in formula (VI):
Figure BDA0002861043520000062
in the formula:
R1、R2、R4、R8u, V, Q are as defined above.
In another preferred embodiment, the compound of formula (I), its stereoisomer, tautomer, crystalline form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof is characterized by having the structure shown in formula (VII):
Figure BDA0002861043520000063
in the formula:
R1、R2、R4、R8v, Q are as defined above.
In another preferred embodiment, the compound of formula (I), its stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug has the structure shown in formula (VIII):
Figure BDA0002861043520000071
in the formula (I), the compound is shown in the specification,
r' "is selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C1-C6Alkyl radical, C3-C8Cycloalkyl radical, C4-C10Cycloalkenyl, 4-8 membered heterocyclyl, C6-C14Aryl, 5-14 membered heteroaryl, wherein said substitution means substitution by one or more groups selected from the group consisting of: deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C 1-C6Alkyl radical, C3-C8Cycloalkyl, C4-C10Cycloalkenyl, 4-8 membered heterocyclyl, C6-C14Aryl, 5-14 membered heteroaryl;
q is selected from: 1. 2, 3 or 4;
R1、R4、R8r', V, Q are as defined above.
In a further preferred embodiment of the method,
Figure BDA0002861043520000072
in the moiety, R8May be 1, 2, 3 or 4, or two adjacent R8May form C together with the C atom to which they are attached3-C6A cycloalkyl group.
In a further preferred embodiment of the method,
Figure BDA0002861043520000073
selected from:
Figure BDA0002861043520000074
Figure BDA0002861043520000075
in a further preferred embodiment of the method,
Figure BDA0002861043520000076
moieties are selected from:
Figure BDA0002861043520000077
in another preferred embodiment, R1Selected from:
Figure BDA0002861043520000078
Figure BDA0002861043520000079
in another preferred embodiment, the compound of formula (I), its stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug has the structure shown in formula (IX):
Figure BDA0002861043520000081
in the formula (I), the compound is shown in the specification,
R1selected from:
Figure BDA0002861043520000082
wherein R isASelected from: H. d, halogen or cyano; rB、RB’The same or different, each independently selected from: H. d, halogen, cyano, substituted or unsubstituted C1-C3An alkyl group; wherein said substitution is by one or more groups selected from the group consisting of: D. halogen, cyano, C1-C3Alkyl radical, C3-C6Cycloalkyl, 4-6 membered heterocyclyl or NRIVRV;RIV、RVThe same or different, each independently selected from: H. c1-C3Alkyl radical, C3-C6Cycloalkyl or 4-6 membered heterocyclyl; or RIV、RVAnd the adjacent N together form a 4-6 membered heterocyclyl;
R4R ', V, Q, R' "and q are as defined above.
In another preferred embodiment, R' "is selected from the group consisting of substituted or unsubstituted: c1-C6Alkyl radical, C3-C8Cycloalkyl radical, C4-C10Cycloalkenyl, 4-8 membered heterocyclyl, C6-C10Aryl, 5-10 membered heteroaryl, wherein said substitution means substitution by one or more groups selected from the group consisting of: deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C1-C6Alkyl radical, C3-C8Cycloalkyl, 4-8 membered heterocyclyl.
In another preferred embodiment, R' "is selected from the group consisting of substituted or unsubstituted: c1-C6Alkyl radical, C3-C6Cycloalkyl radical, C4-C6Cycloalkenyl, 4-6 membered heterocyclyl, wherein said substitution means substitution by one or more groups selected from the group consisting of: deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C1-C3An alkyl group.
In another preferred embodiment, R' "is selected from the group consisting of substituted or unsubstituted: c3-C8Cycloalkyl, 4-8 membered heterocyclyl, wherein said substitution means substitution by one or more groups selected from the group consisting of: deuterium, halogen, nitro, hydroxy, cyano, ester group,Amino, amido, C1-C3An alkyl group.
In another preferred embodiment, R2Selected from:
Figure BDA0002861043520000091
K=O、S、CH2or NH; f is 0, 1 or 2.
In another preferred embodiment, R 2Selected from the group consisting of:
Figure BDA0002861043520000092
k is independently O, S, CH2 or NH; e and f are each independently 0, 1 or 2, preferably H in the above groups may optionally be deuterium, halogen, nitro, hydroxy, cyano, ester, amine, amide, C1-C3Alkyl substitution.
In a further preferred embodiment of the method,
Figure BDA0002861043520000093
moieties are selected from the group consisting of:
Figure BDA0002861043520000101
k is independently O, S, CH2Or NH; e and f are each independently 0, 1 or 2, preferably H in the above groups may optionally be deuterium, halogen, nitro, hydroxy, cyano, ester, amine, amide, C1-C3Alkyl substitution.
In another preferred embodiment, the compound of formula (I), its stereoisomer, tautomer, crystalline form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug has the structure shown in formula (X) or (XI):
Figure BDA0002861043520000102
in the formula (I), the compound is shown in the specification,
R4selected from substituted or unsubstituted C6-C14Aryl or 5-10 membered heteroaryl, wherein said substitution means substitution by one or more (e.g. 2, 3, 4 or 5) groups selected from the group consisting of: deuterium, halogen, ester group, cyano group, NRbC(=O)ORe、OC(=O)Re、OC(=O)NRbRcAmino group, C1-C6Alkyl, halo C1-C6Alkyl, hydroxy; rb、RcCan independently represent hydrogen, deuterium, C1-C6Alkyl radical, C3-C8 cycloalkyl, 4-8 membered heterocyclyl, 5-14 membered heteroaryl or C6-C14Aromatic rings, or R bAnd RcTogether with the N atom may form a 4-8 membered heterocyclyl; r iseCan independently represent hydrogen, C1-C6Alkyl radical, C3-C8Cycloalkyl radical, C2-C6Alkenyl radical, C3-C6Cycloalkenyl radical, C2-C6Alkynyl, 4-8 membered heterocyclyl, 5-14 membered heteroaryl or C6-C14An aromatic ring;
rm is selected from the group consisting of substituted or unsubstituted: amino group, C1-C6Alkyl radical, C3-C6Cycloalkyl, 4-6 membered heterocyclyl, wherein said substitution is by one or more (e.g. 2, 3, 4) groups selected from the group consisting of: deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C1-C3Alkyl, C3-C6 cycloalkyl, 4-6 membered heterocyclyl;
rn is selected from the group consisting of substituted or unsubstituted: amino group, C1-C6Alkyl radical, C1-C6Alkoxy radical, C3-C6Cycloalkyl, -O-C3-C6Cycloalkyl radical, C1-C6Alkyl radical C3-C6Cycloalkyl, -O-C1-C6Alkyl radical C3-C6Cycloalkyl, 4-6 membered heterocyclyl, wherein said substitution is by one or more (e.g. 2, 3, 4) groups selected from the group consisting of: deuterium, halogen, nitro, hydroxy, cyano, ester groupAmino, amido, C1-C3Alkyl radical, C1-C3Haloalkyl, C3-C6Cycloalkyl, 4-6 membered heterocyclyl;
rx is selected from: f or Cl;
RAselected from: H. d, halogen, preferably RASelected from: h or F.
R' "is as defined above;
q' is selected from 0, 1, 2, or 3.
In another preferred embodiment, Rn is selected from the group consisting of substituted or unsubstituted: ethyl, n-propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclopropylamino, azetidinyl, azacyclohexyl, oxiranyl, oxetanyl, oxolanyl, wherein said substitution is by one or more (e.g. 2, 3, 4) groups selected from the group consisting of: deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C 1-C3An alkyl group.
In another preferred embodiment, Rm is selected from the group consisting of substituted or unsubstituted: methyl, ethyl, n-propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclopropylamino, azetidinyl, azacyclohexyl, oxirane, oxetanyl, oxolanyl, oxocyclohexyl, wherein the substitution is by one or more (e.g. 2, 3, 4) groups selected from: deuterium, halogen, nitro, hydroxy, cyano, ester, amine, amide, C1-C3An alkyl group.
In another preferred embodiment, R1、R2、R4、L、U、V、W、Q、p、A、B、X、Rn、Rm、Rx、RAR '", q and q' are the specific groups corresponding to the specific compounds in the examples.
In another preferred embodiment, the compound of formula (I), its stereoisomers, tautomers, crystalline forms, pharmaceutically acceptable salts, hydrates, solvates or prodrugs, is selected from the group consisting of:
Figure BDA0002861043520000111
Figure BDA0002861043520000121
Figure BDA0002861043520000131
Figure BDA0002861043520000141
Figure BDA0002861043520000151
Figure BDA0002861043520000161
Figure BDA0002861043520000171
Figure BDA0002861043520000181
Figure BDA0002861043520000191
Figure BDA0002861043520000201
Figure BDA0002861043520000211
Figure BDA0002861043520000221
Figure BDA0002861043520000231
Figure BDA0002861043520000241
Figure BDA0002861043520000251
Figure BDA0002861043520000261
Figure BDA0002861043520000271
Figure BDA0002861043520000281
Figure BDA0002861043520000291
Figure BDA0002861043520000301
Figure BDA0002861043520000311
Figure BDA0002861043520000321
Figure BDA0002861043520000331
Figure BDA0002861043520000341
Figure BDA0002861043520000351
Figure BDA0002861043520000361
Figure BDA0002861043520000371
Figure BDA0002861043520000381
Figure BDA0002861043520000391
Figure BDA0002861043520000401
Figure BDA0002861043520000411
Figure BDA0002861043520000421
Figure BDA0002861043520000431
Figure BDA0002861043520000441
Figure BDA0002861043520000451
Figure BDA0002861043520000461
Figure BDA0002861043520000471
in another preferred embodiment, the compound of formula (I), its stereoisomers, tautomers, crystalline forms, pharmaceutically acceptable salts, hydrates, solvates or prodrugs are not comprised
Figure BDA0002861043520000481
In another preferred embodiment, the compound of formula (I), its stereoisomers, tautomers, crystalline forms, pharmaceutically acceptable salts, hydrates, solvates or prodrugs are selected from the compounds shown in the examples.
In a second aspect of the present invention, there is provided a process for the preparation of a compound of formula (I), a stereoisomer, a tautomer, a crystalline form, a pharmaceutically acceptable salt, a hydrate, a solvate, or a prodrug thereof, comprising the steps of:
Figure BDA0002861043520000482
(i) the compound of formula P-1 is first reacted with oxalyl chloride in an inert solvent, such as tetrahydrofuran, and then with the amine-based compound R2-NH2Reacting to obtain a compound shown as a formula P-2;
(ii) in an inert solvent (e.g., tetrahydrofuran), in the presence of a first base, ring closure of the compound of formula P-2 to provide a compound of formula P-3;
(iii) reacting a compound of formula P-3 with phosphorus oxychloride in an inert solvent (e.g., acetonitrile) in the presence of a second base to provide a compound of formula P-4;
(iv) a compound of formula P-4 is reacted with a base such as N, N-diisopropylethylamine in an inert solvent such as acetonitrile
Figure BDA0002861043520000483
Obtaining a compound of formula P-5 through coupling or substitution reaction;
(v) deprotecting a compound of formula P-5 in an inert solvent (e.g., dichloromethane) in the presence of an acid (e.g., trifluoroacetic acid) to provide a compound of formula P-6;
(vi) a compound of formula P-6 is reacted with R in an inert solvent such as dichloromethane in the presence of a base such as N, N-diisopropylethylamine1E, obtaining a compound shown in a formula P-7 through coupling, substitution or acylation reaction;
(vii) in an inert solvent (e.g. dioxane/water), a base (e.g. potassium acetate) and a catalyst (e.g. [1,1' -bis (diphenylphosphino) ferrocene) ]Palladium dichloride) in the presence of a compound of the formula P-7 and R4-L-E1Obtaining the compound of formula (I) through coupling, substitution or acylation reaction;
in the formula (I), the compound is shown in the specification,
e is halogen, OH, OCOR1、OCO(iBu), and the like;
E1is-BH2、-B(OH)2
Figure BDA0002861043520000491
-Sn(Bu)3-ZnBr, etc.;
PG is an amino protecting group selected from the group consisting of: boc, Bn, Cbz or Fmoc;
y and Z are leaving groups selected from the group consisting of: halogen or OTf;
the first base is selected from the group consisting of: KHMDS, NaHMDS, LiHMDS, NaH,NaOMe, NaOEt ortBuONa;
The second base is selected from the group consisting of: TEA, DIPEA, DMAP or N, N-dimethylaniline;
R1、R2、R4l, A, B, X, U, V, W and Q are as defined in the first aspect.
In a third aspect of the invention, there is provided a pharmaceutical composition comprising one or more compounds of formula (I) according to the first aspect, stereoisomers, tautomers, crystal forms, pharmaceutically acceptable salts, hydrates, solvates or prodrugs thereof; and a pharmaceutically acceptable carrier.
In another preferred embodiment, the pharmaceutical composition further comprises a drug selected from the group consisting of: PD-1 inhibitors (e.g., nivolumab, pembrolizumab, pidilizumab, cemipimab, JS-001, SHR-120, BGB-A317, IBI-308, GLS-010, GB-226, STW204, HX008, HLX10, BAT1306, AK105, LZM 009, or biologically similar drugs thereof), PD-L1 inhibitors (e.g., Duvalimab, alemtuzumab, Avelumab (avelumab), CS1001, KN035, HLX20, SHR-1316, BGB-A333, JS003, CS1003, KL-A167, F520, GR, MSB2311, or biologically similar drugs thereof), CD20 antibodies (e.g., rituximab, obituzumab, orituzumab, veltutumumab, tositumomab, 131I-tuximab, ibritumomab, itumomab, 90Y-itumomab, In-90, ritumumab, In-5932, and so on-47), antibody (e.g., Hulomab, Hututumumab), Hututumumab, Tumtuzumab, Writumumab, Wolb-L-D-E-D-E-D-E-D-E-D-E-D-E-, CC-90002, TTI-621, TTI-622, OSE-172, SRF-231, ALX-148, NI-1701, SHR-1603, IBI188, IMM01, ALK inhibitors (e.g., Ceritinib, Aleptinib, Bugatinib, Laratinib, Ocatinib), PI3K inhibitors (e.g., Idiranib, Duvelisib, Dactlisib, Taselisib, Bimiralisib, Omipalisib, Bupalisib, etc.), BTK inhibitors (e.g., Ibrutinib, Tirabutinib, Acatinib, Zabritinib, Vebrutinib, etc.), EGFR inhibitors (e.g., Afatinib, Gefitinib, erlotinib, Lapatinib, Darkatinib, Icotinib, Netinib, Sapatinib, Napatinib, pyrroltinib, Hirtitinib, HDAC, erlotinib, Galatinib, e, Galatinib, e, vorinostat, fimepinastat, drosrinostat, entinostat, daciclast, Quisinostat, tacrine, etc.), CDK inhibitors (e.g., palbociclib, ribbociclib, Abemaciclib, micciib, Trilaciclib, Lerociclib, etc.), MEK inhibitors (e.g., semetinib (AZD6244), trametinib (GSK1120212), PD0325901, seru 0126, pimatiib (AS-703026), PD184352(CI-1040), etc.), mTOR inhibitors (e.g., vistuertib, etc.), SHP2 inhibitors (e.g., RMC-4630, JAB-3068, TNO155, etc.), or combinations thereof.
In another preferred embodiment, there is provided a method for preparing a pharmaceutical composition comprising the steps of: mixing a pharmaceutically acceptable carrier with a compound of formula (I), a stereoisomer, a tautomer, a crystalline form, a pharmaceutically acceptable salt, a hydrate, a solvate, or a prodrug of the first aspect of the invention, thereby forming a pharmaceutical composition.
In a fourth aspect of the present invention, there is provided a use of a compound of formula (I), a stereoisomer, a tautomer, a crystalline form, a pharmaceutically acceptable salt, a hydrate, a solvate, or a prodrug thereof, according to the first aspect, or a pharmaceutical composition according to the third aspect, for the preparation of a medicament for the prophylaxis and/or treatment of KRASG12COr an expression level of the polypeptide.
In a fifth aspect of the invention, there is provided a method of preventing and/or treating KRASG12CA disease associated with the activity or expression level of (a), comprising the steps of: administering to a patient in need thereof an effective amount of a compound of formula (I), a stereoisomer, a tautomer, a crystalline form, a pharmaceutically acceptable salt, a hydrate, a solvate, or a prodrug thereof, described in the first aspect, or administering a pharmaceutical composition described in the third aspect.
In another preferred embodiment, the disease is a tumor or a dysregulated disease.
In another preferred embodiment, the disease is selected from the group consisting of: lung cancer, breast cancer, prostate cancer, esophageal cancer, colorectal cancer, bone cancer, kidney cancer, stomach cancer, liver cancer, colorectal cancer, melanoma, lymphoma, leukemia, brain tumor, myeloma, soft tissue sarcoma, pancreatic cancer, skin cancer.
In a sixth aspect, the invention provides a non-diagnostic, non-therapeutic inhibition of KRASG12CThe method of (1), comprising the steps of: administering to a patient in need thereof an effective amount of a compound of formula (I), a stereoisomer, a tautomer, a crystalline form, a pharmaceutically acceptable salt, a hydrate, a solvate, or a prodrug thereof, described in the first aspect, or administering a pharmaceutical composition described in the third aspect.
In a seventh aspect of the invention, there is provided an in vitro inhibitor for inhibiting KRASG12CThe method comprises the following steps: contacting a compound of the first aspect, a stereoisomer, a tautomer, a crystalline form, a pharmaceutically acceptable salt, a hydrate, a solvate, or a prodrug thereof, or a composition of the third aspect, with a somatic cell.
In another preferred embodiment, the somatic cells are from a primate (e.g., human).
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. Not to be reiterated herein, but to the extent of space.
Detailed Description
The present inventors have conducted extensive and intensive studies for a long time to unexpectedly prepare a novel class of KRASG12CCompounds with selective inhibition and/or better pharmacodynamic properties. On this basis, the inventors have completed the present invention.
Term(s) for
In the present invention, unless otherwise specified, the terms used have the ordinary meanings well known to those skilled in the art.
When a substituent is described by a general formula written from left to right, the substituent also includes chemically equivalent substituents obtained when the formula is written from right to left. For example, -CH2O-is equivalent to-OCH2-。
The term "alkyl" refers to a straight or branched chain alkyl group, which may includeAnd the number of carbon atoms, wherein "C" is1-C18Alkyl is meant to include 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17 or 18 carbon atoms, preferably for example C1-C2、C1-C3、C1-C4、C1-C5、C1-C6、C1-C7、C1-C8、C1-C9、C1-C10、C2-C3、C2-C4、C2-C5、C2-C6、C3-C4、C3-C5、C3-C6、C4-C5、C4-C6Or C5-6. Typical "alkyl" groups include, but are not limited to, methyl, ethyl, propyl, isopropyl, n-butyl, t-butyl, isobutyl, tert-butyl, and,
Figure BDA0002861043520000511
Pentyl, isopentyl, heptyl, 4-dimethylpentyl, octyl, 2, 4-trimethylpentyl, nonyl, decyl, undecyl, dodecyl and the like. In the present invention, the alkyl group also includes substituted alkyl groups. "substituted alkyl" means an alkyl group which is substituted at one or more positions, especially 1 to 4 substituents, and may be substituted at any position.
The term "cycloalkyl" refers to a fully saturated cyclic hydrocarbon group, wherein "C" is3-C20Cycloalkyl "means a fully saturated cyclic hydrocarbon compound group containing 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 carbon atoms, including 1 to 4 rings, each ring containing 3 to 8 carbon atoms. Preferably is C3-C4、C3-C5、C3-C6、C3-C7、C3-C8、C3-C9、C3-C10. "substituted cycloalkyl" means that one or more positions in the cycloalkyl group are substituted, especially 1 to 4 substituents, which may be substituted at any position. In the invention, a "ringAlkyl "is intended to include" substituted cycloalkyl ".
The term "heterocyclyl" refers to a fully saturated or partially unsaturated cyclic group, wherein "3-20 membered heterocyclyl" refers to a fully saturated or partially unsaturated cyclic group (including but not limited to, e.g., 3-7 membered monocyclic, 6-11 membered bicyclic, or 8-16 membered tricyclic ring system) containing 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 ring atoms, wherein at least one heteroatom is present in the ring having at least one carbon atom. Each heteroatom-containing heterocycle may carry 1, 2, 3 or 4 heteroatoms selected from nitrogen, oxygen or sulfur, where nitrogen or sulfur may be oxidized and nitrogen may also be quaternized. The heterocyclic group may be attached to any heteroatom or residue of a carbon atom of the ring or ring system molecule. Typical monocyclic heterocyclic groups include, but are not limited to, azetidinyl, pyrrolidinyl, oxetanyl, pyrazolinyl, imidazolinyl, imidazolidinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, tetrahydrofuranyl, piperidinyl, piperazinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, hexahydroazepinyl, 4-piperidyl, tetrahydropyranyl, morphinyl, thiomorpholinyl sulfoxide, thiomorpholinyl sulfone, 1, 3-dioxanyl, and tetrahydro-1, 1-dioxythiophene, and the like. Polycyclic heterocyclic groups include spiro, fused and bridged heterocyclic groups; wherein the heterocyclic groups of the spiro, fused and bridged rings are optionally linked to other groups by single bonds, or are further linked to other cycloalkyl, heterocyclic, aryl and heteroaryl groups by any two or more atoms in the ring; heterocyclic groups may be substituted or unsubstituted.
The term "aryl" refers to an aromatic cyclic hydrocarbon compound group, wherein "C6-C14 aryl" refers to an aromatic cyclic hydrocarbon compound group containing 6, 7, 8, 9, 10, 11, 12, 13 or 14 ring carbon atoms, having 1-5 rings, especially monocyclic and bicyclic groups such as phenyl, biphenyl or naphthyl. Where the aromatic ring contains two or more aromatic rings (bicyclic, etc.), the aromatic rings of the aryl group may be linked by a single bond (e.g., biphenyl), or fused (e.g., naphthalene, anthracene, etc.). "substituted aryl" means an aryl group which is substituted at one or more positions, especially 1 to 3 substituents, and can be substituted at any position.
The term "heteroaryl" refers to an aromatic cyclic hydrocarbon compound group containing 1 to 4 heteroatoms, wherein the heteroatoms are selected from oxygen, nitrogen and sulfur. Wherein "5-14 membered heteroaryl" refers to an aromatic cyclic hydrocarbon compound group containing 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 ring atoms containing 1-4 heteroatoms selected from N, O, S. Heteroaryl is preferably a 5 to 10 membered ring, more preferably 5 or 6 membered, and includes, but is not limited to, pyrrolyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, thiadiazolyl, isothiazolyl, furanyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, triazolyl, tetrazolyl, and the like.
The term "alkoxy" refers to an alkoxy group having a straight or branched chain or cyclic structure, wherein "C1-C18 alkoxy" refers to a straight or branched chain or cyclic alkoxy group having 1 to 18 carbon atoms, including C1-C18 alkyl-O-, -C1-C6 alkyl-O-C1-C6 alkyl, preferably C1-C8 alkoxy, more preferably C1-C6 alkoxy, including but not limited to methoxy, ethoxy, propoxy, isopropoxy, butoxy, and the like.
"cycloalkenyl" refers to a cyclic hydrocarbon group having one or more double bonds, where "C" is4-C10Cycloalkenyl "means a cyclic hydrocarbon group containing 4, 5, 6, 7, 8, 9 or 10 carbon atoms with one or more double bonds, preferably C4-C6Cycloalkenyl groups, including but not limited to: cyclobutenyl, cyclopentenyl, cyclopentadienyl, cyclohexenyl, cyclohexadienyl, and the like.
The term "ester group" refers to a group with the structure-COOR, wherein R represents hydrogen, alkyl or substituted alkyl, cycloalkyl or substituted cycloalkyl, cycloalkenyl or substituted cycloalkenyl, aryl or substituted aryl, heteroaryl or substituted heteroaryl, heterocyclyl or substituted heterocyclyl. Wherein alkyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl have the definitions as described above.
The term "amineGroup "means a group with the structure-NRR ', where R and R' may independently represent hydrogen, alkyl or substituted alkyl, cycloalkyl or substituted cycloalkyl, cycloalkenyl or substituted cycloalkenyl, aryl or substituted aryl, heteroaryl or substituted heteroaryl, heterocyclyl or substituted heterocyclyl. Wherein alkyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl have the definitions as described above. R and R 'may be the same or different, and when R and R' are both H, the amino group is-NH2. Examples of amine groups include, but are not limited to, methylamino, dimethylamino, ethylamino, diethylamino, propylamino, isopropylamino, butylamino, and the like.
The term "amido" refers to a group with the structure-CONRR ', where R and R' may independently represent hydrogen, alkyl or substituted alkyl, cycloalkyl or substituted cycloalkyl, cycloalkenyl or substituted cycloalkenyl, aryl or substituted aryl, heteroaryl or substituted heteroaryl, heterocyclyl or substituted heterocyclyl. Wherein alkyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl have the definitions as described above. R and R' may be the same or different.
The term "sulfonamide" refers to a sulfonamide group having the structure-SO 2NRR ', wherein R and R' may independently represent hydrogen, alkyl or substituted alkyl, cycloalkyl or substituted cycloalkyl, cycloalkenyl or substituted cycloalkenyl, aryl or substituted aryl, heteroaryl or substituted heteroaryl, heterocyclyl or substituted heterocyclyl. Wherein alkyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl are as defined above. R and R' may be the same or different.
The term "aminosulfonyl" refers to compounds having the structure-NRSO2R 'wherein R and R' may independently represent hydrogen, alkyl or substituted alkyl, cycloalkyl or substituted cycloalkyl, cycloalkenyl or substituted cycloalkenyl, aryl or substituted aryl, heteroaryl or substituted heteroaryl, heterocyclyl or substituted heterocyclyl. Wherein alkyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl have the definitions as described above. R and R' may be the same or different.
The term "ureido" refers to a group having the structure-NRCONR 'R ", where R, R' and R" may independently represent hydrogen, alkyl or substituted alkyl, cycloalkyl or substituted cycloalkyl, cycloalkenyl or substituted cycloalkenyl, aryl or substituted aryl, heteroaryl or substituted heteroaryl, heterocyclyl or substituted heterocyclyl. Wherein alkyl, cycloalkyl, cycloalkenyl, aryl, heteroaryl, heterocyclyl have the definitions as described above. R, R' and R "may be the same or different.
When the substituent is a non-terminal substituent or the relevant group is deprived of an H atom, it is a subunit of the corresponding group, typically a divalent group, e.g., an alkyl group deprived of an H atom is an alkylene group (e.g., methylene, ethylene, propylene, isopropylene)
Figure BDA0002861043520000531
) Butylene (e.g. butyl ene)
Figure BDA0002861043520000532
) Pentylene (e.g. ethylene)
Figure BDA0002861043520000533
) Hexamethylene (e.g. hexamethylene)
Figure BDA0002861043520000534
) Heptylene (e.g. ethylene)
Figure BDA0002861043520000535
) Etc.), cycloalkyl corresponding cycloalkylene (e.g.:
Figure BDA0002861043520000536
Figure BDA0002861043520000537
etc.), heterocyclo-corresponding heterocyclylene (e.g.:
Figure BDA0002861043520000538
) Cycloalkyl corresponding heterocyclylene (e.g.:
Figure BDA0002861043520000539
etc.), alkoxy groupsRadical-corresponding alkyleneoxy (-CH)2O-、-CH2CH2-O-CH2-、-CH2OCH2CH2CH2-) and the like.
The term "halogen" or "halo" refers to chlorine, bromine, fluorine, iodine.
The term "halo" means that H in a group is substituted with halogen.
The term "deuterated" means that the H in the group is replaced with deuterium.
The term "hydroxy" refers to a group with the structure OH.
The term "nitro" refers to a group with the structure NO2A group of (1).
The term "cyano" refers to a group with the structure CN.
The term "selected from the group consisting of substituted and unsubstituted" means that the H atom of the selected group is substituted or unsubstituted, whereas the selected group, absent the H atom, is not substituted.
Unless otherwise stated, it is assumed that any heteroatom that is not in a valence state has sufficient hydrogen to replenish its valence state.
As described herein, the compounds of the present invention can be taken with any number of substituents or functional groups to expand their inclusion range. In general, the term "substituted", whether appearing before or after the term "optional", in the formula of the present invention including substituents, means that the hydrogen radical is replaced with a substituent of the indicated structure. When multiple positions in a particular structure are substituted with multiple particular substituents, each position of the substituent may be the same or different. The term "substituted" as used herein includes all permissible substitutions of organic compounds. In a broad sense, permissible substituents include acyclic, cyclic, branched unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic organic compounds. In the present invention, the heteroatom nitrogen may have a hydrogen substituent or any permissible organic compound described hereinabove to supplement its valence state. Furthermore, the present invention is not intended to limit in any way the permissible substitution of organic compounds. The present invention recognizes that the combination of substituents and variable groups is excellent in the treatment of diseases, such as infectious diseases or proliferative diseases, in the form of stable compounds. The term "stable" as used herein refers to compounds that are stable enough to maintain the structural integrity of the compound when tested for a sufficient period of time, and preferably are effective for a sufficient period of time, and are used herein for the purposes described above.
In the present invention, the term "substituted" means that one or more hydrogen atoms on a specified group are replaced with a specified substituent. Particular substituents are those described correspondingly in the preceding text or as present in the examples. Unless otherwise specified, a substituted group may have a substituent selected from a specific group at any substitutable position of the group, and the substituent may be the same or different at each position. In the present invention, unless otherwise specified, the groups include corresponding substituent groups and subunits, such as: alkyl includes substituted alkyl, cycloalkyl includes substituted cycloalkyl, aryl includes substituted aryl, heteroaryl includes substituted heteroaryl, heterocyclyl includes substituted heterocyclyl, and the like. It will be understood by those skilled in the art that the combinations of substituents contemplated by the present invention are those that are stable or chemically achievable. Typical substitutions include, but are not limited to, one or more of the following groups: such as hydrogen, deuterium, halogen (e.g. mono-or polyhalo-substituents, the latter being e.g. trifluoromethyl or containing Cl3Alkyl of (i), nitrile, nitro, oxygen (e.g., ═ O), trifluoromethyl, trifluoromethoxy, cycloalkyl, C2-C6 alkenyl, C4-C10 cycloalkenyl, C2-C6 alkynyl, heterocyclyl, aryl, heteroaryl, OR a、SRa、S(=O)Re、S(=O)2Re、P(=O)2Re、S(=O)2ORe、P(=O)2ORe、NRbRc、NRbS(=O)2Re、NRbP(=O)2Re、S(=O)2NRbRc、P(=O)2NRbRc、C(=O)ORd、C(=O)Ra、C(=O)NRbRc、OC(=O)Ra、OC(=O)NRbRc、NRbC(=O)ORe、NRdC(=O)NRbRc、NRdS(=O)2NRbRc、NRdP(=O)2NRbRc、NRbC(=O)RaOr NRbP(=O)2ReWherein R is present thereinaMay independently represent hydrogen, deuterium, C1-C6 alkyl, C3-C8 cycloalkyl, C2-C6 alkenyl, C3-C10 cycloalkenyl, C2-C6 alkynyl, 4-8 membered heterocyclyl, 5-14 membered heteroaryl, or C6-C14 aryl, Rb、RcAnd RdMay independently represent hydrogen, deuterium, C1-C6 alkyl, C3-C8 cycloalkyl, 4-8 membered heterocyclyl, 5-14 membered heteroaryl, or C6-C14 aromatic ring, or RbAnd RcTogether with the N atom may form a heterocyclic ring; r iseCan independently represent hydrogen, C1-C6 alkyl, C3-C8 cycloalkyl, C2-C6 alkenyl, C3-C6 cycloalkenyl, C2-C6 alkynyl, 4-8 membered heterocyclyl, 5-14 membered heteroaryl, or C6-C14 aryl ring. The above-mentioned typical substituents such as alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, heterocyclyl, heteroaryl or aryl and their corresponding substituents and subunits may be optionally substituted, wherein said alkyl, cycloalkyl, cycloalkenyl, heterocyclyl, heteroaryl or aryl has the definition as described above.
Active ingredient
As used herein, the terms "compound of the invention" or "active ingredient of the invention" are used interchangeably to refer to a compound of formula (I), or a pharmaceutically acceptable salt, hydrate, solvate, isotopic compound (e.g., deuterated compound), or prodrug thereof. The term also includes racemates, optical isomers.
The compound of formula (I) has the following structure:
Figure BDA0002861043520000541
wherein R is1、R2、R4L, U, V, W, Q, A, B, X are as defined above.
Preferably, the compound of formula (I) has a structure represented by formula (II-A) or (II-B):
Figure BDA0002861043520000551
in the formula:
R1、R2、R4a, B, L, X, U, V, W, Q are as defined above.
Preferably, the compound of formula (I) has the structure shown in formula (III):
Figure BDA0002861043520000552
R1、R2、R4x, L, U, V, W, Q are as defined above.
Preferably, the compound of formula (I) has the structure shown in formula (IV):
Figure BDA0002861043520000553
in the formula:
R1、R2、R4、R8l, U, V, W, Q are as defined above.
Preferably, the compound of formula (I) has the structure shown in formula (V):
Figure BDA0002861043520000554
in the formula:
R1、R2、R4、R8u, V, W, Q are as defined above.
Preferably, the compound of formula (I) has the structure of formula (VI):
Figure BDA0002861043520000561
in the formula:
R1、R2、R4、R8u, V, Q are as defined above.
Preferably, the compound of formula (I) has the structure shown in formula (VII):
Figure BDA0002861043520000562
in the formula:
R1、R2、R4、R8v, Q are as defined above.
Preferably, in each of the above formulas (i.e., I-VII), R1Selected from the group consisting of: -C (O) C (R)A)=C(RB)2、-S(O)2C(RA)=C(RB)2、-NR6C(O)C(RA)=C(RB)2or-NR6S(O)2C(RA)=C(RB)2
Wherein R isAIndependently selected from the group consisting of: hydrogen, deuterium, fluoro, cyano, or C1-C3An alkyl group; each RBThe same or different, and independently selected from the group consisting of: hydrogen, deuterium, cyano, or C 1-C3An alkyl group; wherein the alkyl group may be substituted with one or more (e.g., 2, 3, 4 or 5) substituents selected from the group consisting of: deuterium, halogen, cyano, amino, C3-C7Cycloalkyl, 4-7 membered heterocyclyl, NHR9Or NR9R10;R9And R10Each independently is C1-C3An alkyl group; or R9,R10Together with the N atom to which they are attached form a substituted or unsubstituted 4-8 membered heterocyclyl;
R6selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C6Alkyl, deuterated C1-C6Alkyl, halo C1-C6Alkyl radical, C3-C8Cycloalkyl radical, C1-C6Alkoxy, deuterated C1-C6Alkoxy, halo C1-C6Alkoxy radical4-8 membered heterocyclic group, C6-C14Aryl, 5-14 membered heteroaryl;
wherein, the term "substituted" when not specifically stated, means substituted by one or more groups selected from the group consisting of: hydrogen, deuterium, C1-C6Alkyl, deuterated C1-C6Alkyl, halo C1-C6Alkyl radical, C3-C8Cycloalkyl radical, C1-C6Alkoxy, deuterated C1-C6Alkoxy, halo C1-C6Alkoxy radical, C6-C10Aryl, 5-10 membered heteroaryl, 4-8 membered heterocyclyl, halogen, nitro, hydroxy, cyano, ester, amine, NRbC(=O)ORe、OC(=O)NRbRcAmide, sulfonamide, or ureido groups; rb、RcMay independently represent hydrogen, deuterium, C1-C6 alkyl, C3-C8 cycloalkyl, 4-8 membered heterocyclyl, 5-14 membered heteroaryl or C6-C14 aromatic ring, or RbAnd R cTogether with the N atom may form a 4-8 membered heterocyclyl; r iseMay independently represent hydrogen, C1-C6 alkyl, C3-C8 cycloalkyl, C2-C6 alkenyl, C3-C6 cycloalkenyl, C2-C6 alkynyl, 4-8 membered heterocyclyl, 5-14 membered heteroaryl, or C6-C14 aryl ring;
the limiting conditions are as follows:
when B is N, R1Selected from the group consisting of: -C (O) C (R)A)=C(RB)2or-S (O)2C(RA)=C(RB)2
When B is CH or CR5When R is1Selected from the group consisting of: -NR6C(O)C(RA)=C(RB)2or-NR6S(O)2C(RA)=C(RB)2More preferably, R1is-C (O) C (R)A)=C(RB)2Wherein R isAIndependently selected from the group consisting of: hydrogen, fluorine; each RBThe same or different, and independently selected from the group consisting of: hydrogen or C1-C3An alkyl group, wherein the alkyl group may be substituted with one or more substituents selected from the group consisting of: deuterium, halogen, cyano, amino, C3-C7CycloalkanesRadical, 4-7 membered heterocyclyl radical, NHR9Or NR9R10;R9And R10Each independently is C1-C3An alkyl group; or R9,R10Together with the N atom to which they are attached form a 4-8 membered heterocyclic group.
Preferably, in each of the above formulae, R2Selected from the group consisting of substituted: phenyl, 5-6 membered heteroaryl, wherein said substitution means substitution by one or more (e.g. 2, 3, 4 or 5) groups selected from the group consisting of: r', -SO2R'、-SO2NR'R”、-NR'SO2R ", -P (═ O) R' R"; r 'and R' are the same or different and are each independently selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C 1-C6Alkyl radical, C3-C6Cycloalkyl, C4-C6Cycloalkenyl, 4-8 membered heterocyclyl, C6-C10Aryl, 5-10 membered heteroaryl; or when R ' and R ' are attached to the same N atom, R ' together with the N atom to which they are attached form a substituted or unsubstituted 4-6 membered heterocyclyl; wherein said substitution means substitution by one or more (e.g. 2, 3, 4 or 5) groups selected from: hydrogen, deuterium, C1-C6Alkyl, deuterated C1-C6Alkyl, halo C1-C6Alkyl radical, C3-C8Cycloalkyl radical, C1-C6Alkoxy, deuterated C1-C6Alkoxy, halo C1-C6Alkoxy radical, C6-C10Aryl, 5-10 membered heteroaryl, 4-8 membered heterocyclyl, halogen, nitro, hydroxy, cyano, ester, amine, amide, sulfonamide, or ureido.
Preferably, in each of the above formulae, R4Selected from the group consisting of substituted or unsubstituted: phenyl or 5-6 membered heteroaryl, wherein said substitution means substitution by one or more (such as 2, 3, 4 or 5) groups selected from the group consisting of: hydrogen, deuterium, halogen, ester group, cyano group, NRbC(=O)ORe、OC(=O)Re、OC(=O)NRbRcAmino group, C1-C18Alkyl (preferably C)1-C6Alkyl, more preferably C1-C3Alkyl), halo C1-C18Alkyl (preferably halogenated C)1-C6Alkyl, more preferably halogenated C1-C3Alkyl), hydroxy; rb、RcMay independently represent hydrogen, deuterium, C1-C6 alkyl, C3-C8 cycloalkyl, 4-8 membered heterocyclyl, 5-14 membered heteroaryl or C6-C14 aromatic ring, or R bAnd RcTogether with the N atom, may form a 4-8 membered heterocyclic group; reCan independently represent hydrogen, C1-C6 alkyl, C3-C8 cycloalkyl, C2-C6 alkenyl, C3-C6 cycloalkenyl, C2-C6 alkynyl, 4-8 membered heterocyclyl, 5-14 membered heteroaryl, or C6-C14 aryl ring.
Preferably, in each of the above formulae, Q is N.
Preferably, in each of the above formulae, V, W is each independently CR3,R3Is H or halogen; preferably, R3Is halogen.
Preferably, in the formulae (IV), (V), (VI), (VII), R8Independently selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C6Alkyl, deuterated C1-C6Alkyl, halo C1-C6Alkyl radical, C3-C8Cycloalkyl radical, C1-C6Alkoxy, deuterated C1-C6Alkoxy, halo C1-C6Alkoxy, amino, hydroxy, 4-8 membered heterocyclyl; said substitution means substitution by one or more (e.g. 2, 3, 4 or 5) groups selected from the group consisting of: hydrogen, deuterium, halogen, nitro, hydroxyl, cyano, ester, amine, amide, sulfonamide, or urea; more preferably, R8Independently selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C6Alkyl, deuterated C1-C6Alkyl, halo C1-C6Alkyl, wherein said substitution is by cyano.
Preferably, R8Is methyl.
Preferably, the compound of formula (I), its stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate or prodrug has the structure shown in formula (VIII):
Figure BDA0002861043520000581
In the formula (I), the compound is shown in the specification,
r' "is selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C1-C6Alkyl radical, C3-C8Cycloalkyl radical, C4-C10Cycloalkenyl, 4-8 membered heterocyclyl, C6-C14Aryl, 5-14 membered heteroaryl, wherein said substitution means substitution by one or more groups selected from the group consisting of: deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C1-C6Alkyl radical, C3-C8Cycloalkyl radical, C4-C10Cycloalkenyl, 4-8 membered heterocyclyl, C6-C14Aryl, 5-14 membered heteroaryl;
q is selected from: 1. 2, 3, or 4;
R1、R4、R8r', V, Q are as defined above.
Preferably, the compound of formula (I), its stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate or prodrug has the structure shown in formula (IX):
Figure BDA0002861043520000582
in the formula (I), the compound is shown in the specification,
R1、R4r ', V, R' ", Q and Q are as defined above.
Preferably, in the formulae II to VII,
Figure BDA0002861043520000583
moieties are selected from:
Figure BDA0002861043520000584
preferably, in the formula I,
Figure BDA0002861043520000585
moieties are selected from:
Figure BDA0002861043520000586
preferably, in the formula I-II,
Figure BDA0002861043520000587
is partially made of
Figure BDA0002861043520000588
Wherein Rx is selected from: hydrogen, 20 deuterium, C1-C3Alkyl, deuterated C1-C3Alkyl, halo C1-C3Alkyl radical, C3-C6Cycloalkyl radical, C1-C3Alkoxy, halogen, nitro, hydroxy, cyano, ester, 4-6 membered heterocyclyl, preferably,
Figure BDA0002861043520000591
Is partially as
Figure BDA0002861043520000592
Preferably, in the formulae III to IX,
Figure BDA0002861043520000593
is partially as
Figure BDA0002861043520000594
Wherein Rx is selected from: hydrogen, deuterium, C1-C3Alkyl, deuterated C1-C3Alkyl, halo C1-C3Alkyl radical, C3-C6Cycloalkyl, C1-C3Alkoxy, halogen, nitro, hydroxy, cyano, ester, 4-6 membered heterocyclyl, preferably,
Figure BDA0002861043520000595
is partially made of
Figure BDA0002861043520000596
Preferably, in the formulae I-VII, R2Selected from the group consisting of:
Figure BDA0002861043520000597
k is independently O, S, CH2 or NH; e and f are each independently 0, 1 or 2, preferably H in the above groups may optionally be deuterium, halogen, nitro, hydroxy, cyano, ester, amine, amide, C1-C3Alkyl substitution.
Preferably, the compound of formula (I), its stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate or prodrug has the structure shown in formula (VIII):
Figure BDA0002861043520000598
in the formula (I), the compound is shown in the specification,
r' "is selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C1-C6Alkyl radical, C3-C8Cycloalkyl radical, C4-C10Cycloalkenyl, 4-8 membered heterocyclyl, C6-C14Aryl, 5-14 membered heteroaryl, wherein said substitution means substitution by one or more groups selected from the group consisting of: deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C 1-C6Alkyl radical, C3-C8Cycloalkyl, C4-C10Cycloalkenyl, 4-8 membered heterocyclyl, C6-C14Aryl, 5-14 membered heteroaryl;
R1、R4、R8r', V, Q, and q are as defined above.
Preferably, in formula VIII, R' "is selected from the group consisting of substituted or unsubstituted:C1-C6alkyl radical, C3-C8Cycloalkyl radical, C4-C10Cycloalkenyl, 4-8 membered heterocyclyl, C6-C10Aryl, 5-10 membered heteroaryl, wherein said substitution means substitution by one or more groups selected from the group consisting of: deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C1-C6Alkyl radical, C3-C8Cycloalkyl, 4-8 membered heterocyclyl.
Preferably, in formula VIII, R' "is selected from the group consisting of substituted or unsubstituted: c1-C6Alkyl radical, C3-C6Cycloalkyl radical, C4-C6Cycloalkenyl, 4-6 membered heterocyclyl, wherein said substitution means substitution by one or more groups selected from the group consisting of: deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C1-C3An alkyl group.
Preferably, in the formulae VIII to IX,
Figure BDA0002861043520000601
moieties are selected from the group consisting of:
Figure BDA0002861043520000602
k is independently O, S, CH2 or NH; e and f are each independently 0, 1 or 2, preferably H in the above groups may optionally be deuterium, halogen, nitro, hydroxy, cyano, ester, amine, amide, C1-C3Alkyl substitution.
Preferably, the compound of formula (I), its stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate or prodrug has the structure shown in formula (X) or (XI):
Figure BDA0002861043520000603
In the formula (I), the compound is shown in the specification,
R4selected from substituted or unsubstitutedSubstituted C6-C14Aryl or 5-10 membered heteroaryl, wherein said substitution means substitution by one or more (such as 2, 3, 4 or 5) groups selected from: deuterium, halogen, ester group, cyano group, NRbC(=O)ORe、OC(=O)Re、OC(=O)NRbRcAmino group, C1-C6Alkyl, halo C1-C6Alkyl, hydroxy; r isb、RcMay independently represent hydrogen, deuterium, C1-C6 alkyl, C3-C8 cycloalkyl, 4-8 membered heterocyclyl, 5-14 membered heteroaryl or C6-C14 aromatic ring, or RbAnd RcTogether with the N atom, may form a 4-8 membered heterocyclic group; reMay independently represent hydrogen, C1-C6 alkyl, C3-C8 cycloalkyl, C2-C6 alkenyl, C3-C6 cycloalkenyl, C2-C6 alkynyl, 4-8 membered heterocyclyl, 5-14 membered heteroaryl, or C6-C14 aryl ring;
rm is selected from the group consisting of substituted or unsubstituted: amino group, C1-C6Alkyl radical, C3-C6Cycloalkyl, 4-6 membered heterocyclyl, wherein said substitution means substitution by one or more groups selected from the group consisting of: deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C1-C3Alkyl, C3-C6 cycloalkyl, 4-6 membered heterocyclyl;
rn is selected from the group consisting of substituted or unsubstituted: amino group, C1-C6Alkyl radical, C1-C6Alkoxy radical, C3-C6Cycloalkyl, -O-C3-C6Cycloalkyl radical, C1-C6Alkyl radical C3-C6Cycloalkyl, -O-C1-C6Alkyl radical C3-C6Cycloalkyl, 4-6 membered heterocyclyl, wherein said substitution means substitution by one or more groups selected from the group consisting of: deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C 1-C3Alkyl radical, C1-C3Haloalkyl, C3-C6 cycloalkyl, 4-6 membered heterocyclyl;
rx is selected from: f or Cl;
RAselected from: H. d, halogen, or cyano, preferably RASelected from: h or F.
R' "is as defined above;
q' is selected from 0, 1, 2, or 3.
Preferably of the formula X-XI, R4Selected from substituted or unsubstituted phenyl or 5-6 membered heteroaryl, wherein said substitution is by one or more (e.g. 2, 3, 4 or 5) groups selected from: deuterium, halogen, ester group, cyano group, NRbC(=O)ORe、OC(=O)Re、OC(=O)NRbRcAmino group, C1-C3Alkyl, halo C1-C3Alkyl, hydroxy; rb、RcMay independently represent hydrogen, deuterium, C1-C3 alkyl, C3-C6 cycloalkyl, 4-6 membered heterocyclyl, 5-6 membered heteroaryl or phenyl, or RbAnd RcTogether with the N atom, may form a 4-6 membered heterocyclic group; reCan independently represent hydrogen, C1-C3 alkyl, C3-C6 cycloalkyl.
Preferably, in the formula X-XI, Rn is selected from the following substituted or unsubstituted groups: ethyl, n-propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclopropylamino, azetidinyl, azacyclohexyl, oxiranyl, oxetanyl, oxolanyl, wherein said substitution is by one or more groups selected from the group consisting of: deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C 1-C3An alkyl group.
Preferably, in formula X-XI, Rm is selected from the group consisting of substituted or unsubstituted: methyl, ethyl, n-propyl, isopropyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclopropylamino, azetidinyl, azacyclohexyl, oxiranyl, oxetanyl, oxolanyl, wherein said substitution is by one or more groups selected from the group consisting of: deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C1-C3An alkyl group.
The salts which the compounds of the invention may form are also within the scope of the invention. Unless otherwise indicated, the compounds of the present invention are understood to include salts thereof. The term "salt" as used herein refers to a salt formed from an inorganic or organic acid and a base in either an acidic or basic form. Furthermore, when a compound of the present invention contains a basic moiety, including but not limited to pyridine or imidazole, and an acidic moiety, including but not limited to carboxylic acid, zwitterions ("inner salts") that may form are included within the scope of the term "salt(s)". Pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts are preferred, although other salts are also useful, e.g., in isolation or purification steps during manufacture. The compounds of the invention may form salts, for example, by reacting compound I with an amount of acid or base, e.g. an equivalent amount, and salting out in a medium, or lyophilizing in an aqueous solution.
The compounds of the invention may contain basic moieties, including but not limited to amine or pyridine or imidazole rings, which may form salts with organic or inorganic acids. Typical acids which may form salts include acetates (e.g. with acetic acid or trihaloacetic acid such as trifluoroacetic acid), adipates, alginates, ascorbates, aspartates, benzoates, benzenesulfonates, bisulfates, borates, butyrates, citrates, camphorates, camphorsulfonates, cyclopentanepropionates, diglycolates, dodecylsulfates, ethanesulfonates, fumarates, glucoheptonates, glycerophosphates, hemisulfates, heptanoates, hexanoates, hydrochlorides, hydrobromides, hydroiodides, hydroxyethanesulfonates (e.g. 2-hydroxyethanesulfonates), lactates, maleates, methanesulfonates, naphthalenesulfonates (e.g. 2-naphthalenesulfonates), nicotinates, nitrates, oxalates, pectinates, persulfates, phenylpropionates (e.g. 3-phenylpropionates), phosphates, propionates, citrates, and the like, Picrates, pivalates, propionates, salicylates, succinates, sulfates (e.g., with sulfuric acid), sulfonates, tartrates, thiocyanates, tosylates, e.g., p-toluenesulfonate, dodecanoate, and the like
Certain compounds of the present invention may contain acidic moieties, including but not limited to carboxylic acids, which may form salts with various organic or inorganic bases. Typical salts with bases include ammonium salts, alkali metal salts such as sodium, lithium, potassium salts, alkaline earth metal salts such as calcium, magnesium salts, and salts with organic bases (e.g., organic amines) such as benzathine, dicyclohexylamine, hydrabamine (salt with N, N-bis (dehydroabietyl) ethylenediamine), N-methyl-D-glucamine, N-methyl-D-glucamide, t-butylamine, and salts with amino acids such as arginine, lysine, and the like. The basic nitrogen-containing groups may be combined with halide quaternary ammonium salts, such as small molecule alkyl halides (e.g., methyl, ethyl, propyl, and butyl chlorides, bromides, and iodides), dialkyl sulfates (e.g., dimethyl, diethyl, dibutyl, and diamyl sulfates), long chain halides (e.g., decyl, dodecyl, tetradecyl, and tetradecyl chlorides, bromides, and iodides), aralkyl halides (e.g., benzyl and phenyl bromides), and the like.
Prodrugs and solvates of the compounds of the invention are also contemplated. The term "prodrug" as used herein refers to a compound that undergoes chemical conversion by metabolic or chemical processes to yield a compound, salt, or solvate of the present invention when used in the treatment of a related disease. The compounds of the present invention include solvates, such as hydrates.
The compounds, salts or solvates of the invention, in tautomeric forms (for example amides and imine ethers) may be present. All of these tautomers are part of the present invention.
All stereoisomers of the compounds (e.g., those asymmetric carbon atoms that may exist due to various substitutions), including enantiomeric and diastereomeric forms thereof, are contemplated within the invention. The individual stereoisomers of the compounds of the invention may not be present in combination with the other isomers (e.g. as a pure or substantially pure optical isomer having a particular activity), or may be present as a mixture, e.g. as a racemate, or as a mixture with all or a portion of the other stereoisomers. The chiral center of the invention has two S or R configurations, and is defined by the International Union of theory and applied chemistry (IUPAC) proposed in 1974. The racemic forms can be resolved by physical methods such as fractional crystallization, or by separation of the crystals by derivatization into diastereomers, or by chiral column chromatography. The individual optical isomers can be obtained from the racemates by any suitable method, including, but not limited to, conventional methods such as salt formation with an optically active acid followed by crystallization.
The compounds of the present invention, obtained by preparing, isolating and purifying the compound in sequence, have a weight content of 90% or more, for example, 95% or more, 99% or more ("very pure" compounds), as set forth in the text. Such "very pure" compounds of the invention are also part of the invention herein.
All configurational isomers of the compounds of the invention are within the scope of the invention, whether in mixture, pure or very pure form. The definition of compounds in the present invention encompasses both cis (Z) and trans (E) olefin isomers, as well as cis and trans isomers of carbocyclic and heterocyclic rings.
Throughout the specification, groups and substituents may be selected to provide stable fragments and compounds.
Specific functional groups and definitions of chemical terms are detailed below. For purposes of the present invention, the chemical Elements are compatible with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics,75thD. as defined in. The definition of a particular functional group is also described herein. In addition, the basic principles of Organic Chemistry, as well as specific functional groups and reactivities are also described in "Organic Chemistry", Thomas Sorrell, University Science Books, Sausaltito: 1999, which is incorporated by reference in its entirety.
Certain compounds of the present invention may exist in specific geometric or stereoisomeric forms. The present invention encompasses all compounds, including cis and trans isomers, R and S enantiomers, diastereomers, (D) isomer, (L) isomer, racemic mixtures and other mixtures thereof. Further, the asymmetric carbon atom may represent a substituent such as an alkyl group. All isomers, as well as mixtures thereof, are encompassed by the present invention.
According to the present invention, the mixture of isomers may contain a variety of isomer ratios. For example, in a mixture of only two isomers, the following combinations are possible: 50:50, 60:40, 70:30, 80:20, 90:10, 95:5, 96:4, 97:3, 98:2, 99:1, or 100:0, all ratios of isomers are within the scope of the invention. Similar ratios, as well as ratios that are mixtures of more complex isomers, are also within the scope of the invention, as would be readily understood by one of ordinary skill in the art.
The invention also includes isotopically-labeled compounds, equivalent to those disclosed herein as the original compound. In practice, however, it will often occur that one or more atoms are replaced by an atom having a different atomic weight or mass number. Examples of isotopes that can be listed as compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, respectively 2H、3H、13C、11C、14C、15N、18O、17O、31P、32P、35S、18F and36and (4) Cl. The compounds of the present invention, or enantiomers, diastereomers, isomers, or pharmaceutically acceptable salts or solvates thereof, wherein isotopes or other isotopic atoms containing those compounds are within the scope of the present invention. Certain isotopically-labelled compounds of the invention, e.g.3H and14among these, the radioactive isotope of C is useful in tissue distribution experiments of drugs and substrates. Tritium, i.e.3H and carbon-14, i.e.14C, their preparation and detection are relatively easy. Is the first choice among isotopes. In addition, heavier isotopes such as deuterium, i.e.2H, due to its good metabolic stability, may be advantageous in certain therapies, such as increased half-life in vivo or reduced dose, and therefore, may be preferred in certain circumstances. Isotopically labeled compounds can be prepared by conventional methods by substituting a readily available isotopically labeled reagent for a non-isotopically labeled reagent using the protocols disclosed in the examples.
If it is desired to design the synthesis of a particular enantiomer of a compound of the invention, it may be prepared by asymmetric synthesis or by derivatization with chiral auxiliary agents, separation of the resulting diastereomeric mixture and removal of the chiral auxiliary agent to give the pure enantiomer. Alternatively, if the molecule contains a basic functional group, such as an amino acid, or an acidic functional group, such as a carboxyl group, diastereomeric salts can be formed therewith with an appropriate optically active acid or base, and the isolated enantiomers can be obtained in pure form by conventional means such as fractional crystallization or chromatography.
Metabolites of the compounds referred to herein and pharmaceutically acceptable salts thereof, as well as prodrugs that are convertible in vivo into the structures of the compounds referred to herein and pharmaceutically acceptable salts thereof, are also encompassed by the claims herein.
Preparation method
The process for preparing the compounds of formula (I) according to the invention is described in more detail below, but these particular processes do not limit the invention in any way. The compounds of the present invention may also be conveniently prepared by optionally combining various synthetic methods described in the present specification or known in the art, and such combinations may be readily carried out by those skilled in the art to which the present invention pertains.
Typically, the compounds of the present invention are prepared by the following procedures, wherein the starting materials and reagents used, unless otherwise specified, are commercially available or synthesized according to the reported literature.
Figure BDA0002861043520000641
(i) The compound of formula P-1 is first reacted with oxalyl chloride in an inert solvent, such as tetrahydrofuran, and then with the amine-based compound R2-NH2Reacting to obtain a compound shown as a formula P-2;
(ii) in an inert solvent (such as tetrahydrofuran), under the action of a first base, the compound of the formula P-2 is subjected to ring closure to obtain a compound of the formula P-3;
(iii) reacting the compound of formula P-3 with phosphorus oxychloride in an inert solvent (e.g., acetonitrile) in the presence of a second base to obtain a compound of formula P-4;
(iv) A compound of formula P-4 is reacted with a base such as N, N-diisopropylethylamine in an inert solvent such as acetonitrile
Figure BDA0002861043520000642
Obtaining a compound of formula P-5 through coupling or substitution reaction;
(v) deprotecting a compound of formula P-5 in an inert solvent (e.g., dichloromethane) in the presence of an acid (e.g., trifluoroacetic acid) to provide a compound of formula P-6;
(vi) a compound of formula P-6 is reacted with R in an inert solvent such as dichloromethane in the presence of a base such as N, N-diisopropylethylamine1E, obtaining a compound shown in a formula P-7 through coupling, substitution or acylation reaction;
(vii) in an inert solvent (e.g. dioxane/water), a base (e.g. potassium acetate) and a catalyst (e.g. [1,1' -bis (diphenylphosphino) ferrocene)]Palladium dichloride) in the presence of a compound of the formula P-7 and R4-L-E1Obtaining the compound of formula (I) through coupling, substitution or acylation reaction;
in the formula (I), the compound is shown in the specification,
e is halogen, OH, OCOR1、OCO(iBu), and the like;
E1is-BH2、-B(OH)2
Figure BDA0002861043520000651
-Sn(Bu)3-ZnBr, etc.;
PG is an amino protecting group selected from the group consisting of: boc, Bn, Cbz or Fmoc;
y and Z are leaving groups selected from the group consisting of: halogen or OTf;
the first base is selected from the group consisting of: KHMDS, NaHMDS, LiHMDS, NaH, NaOMe, NaOEt ortBuONa;
The second base is selected from the group consisting of: TEA, DIPEA, DMAP or N, N-dimethylaniline;
R1、R2、R4L, A, B, X, U, V, W and Q are as defined above.
In the above reaction steps, a reaction solvent, a reaction catalyst, a base used for the reaction, a reaction temperature, a reaction time, and the like may be selected by those skilled in the art according to a specific reactant.
Pharmaceutical compositions and methods of administration
The pharmaceutical composition is used for preventing and/or treating the following diseases: inflammation, cancer, cardiovascular disease, infection, immunological disease, metabolic disease.
The compounds of formula (I) may be combined with other drugs known to treat or ameliorate similar conditions. When administered in combination, the mode of administration and dosage of the original drug may be maintained unchanged while the compound of formula (I) is administered simultaneously or subsequently. When the compound of the formula (I) is administered simultaneously with one or more other drugs, a pharmaceutical composition containing one or more known drugs together with the compound of the formula (I) can be preferably used. The pharmaceutical combination may also comprise administering the compound of formula (I) in combination with one or more other known drugs, over an overlapping period of time. When the compounds of formula (I) are administered in pharmaceutical combination with one or more other drugs, the dosage of the compounds of formula (I) or known drugs may be lower than the dosage for their administration alone.
Drugs or active ingredients that may be used in combination with the compounds of formula (I) include, but are not limited to: PD-1 inhibitors (e.g., nivolumab, pembrolizumab, pidilizumab, cemipimab, JS-001, SHR-120, BGB-A317, IBI-308, GLS-010, GB-226, STW204, HX008, HLX10, BAT1306, AK105, LZM 009, or biologically similar drugs thereof), PD-L1 inhibitors (e.g., Duvalimab, alemtuzumab, Avelumab (avelumab), CS1001, KN035, HLX20, SHR-1316, BGB-A333, JS003, CS1003, KL-A167, F520, GR, MSB2311, or biologically similar drugs thereof), CD20 antibodies (e.g., rituximab, obituzumab, orituzumab, veltutumumab, tositumomab, 131I-tuximab, ibritumomab, itumomab, 90Y-itumomab, In-90, ritumumab, In-5932, and so on-47), antibody (e.g., Hulomab, Hututumumab), Hututumumab, Tumtuzumab, Writumumab, Wolb-L-D-E-D-E-D-E-D-E-D-E-D-E-, CC-90002, TTI-621, TTI-622, OSE-172, SRF-231, ALX-148, NI-1701, SHR-1603, IBI188, IMM01, ALK inhibitors (e.g., Ceritinib, Aletinib, bugatinib, Laratinib, Ocatinib), PI3K inhibitors (e.g., Ivariansib, Duvelisib, Dactlisib, Taselisib, Bimiralisib, Omipalisib, Buparlisib, etc.), BTK inhibitors (e.g., ibrutinib, Tiradinutinib, Acatinib, Tuseltinib, Vebrutinib, etc.), EGFR inhibitors (e.g., Afatinib, gefitinib, erlotinib, lapatinib, dacatinib, erlotinib, Canotinib, canertinib, sunitinib, VEGFR, erlotinib, etc.), HDAC-NI inhibitors (e, Nicotinib, Ivortinib, VEGFR, etc.), non-Sorafitinib, non-A inhibitors (e, Sorafitinib, etc.), BTX-I-II, Ivoritinib, etc.), and other inhibitors such as, non-I, Ivoritinib, etc.), and other inhibitors (e, etc.), inhibitors such as inhibitors (e, etc.), non-I, non-HDAC, non-I, etc.), and others, etc.), and non-I, etc., inhibitors such as inhibitors (e, non-HDAC, non-I, HDAC, non-I, HDAC, non-I, HDAC, Ivoritinib, HDAC, Ivor, Vorinostat, fimepinastat, Droxinostat, entinostat, daciclast, Quisinostat, tacrine, etc.), CDK inhibitors (e.g., palbociclib, ribociclib, Abemaciclib, micciclib, Lerociclib, etc.), MEK inhibitors (e.g., simetinib (AZD6244), tremetinib (GSK1120212), PD0325901, seru 0126, pimatiib (AS-703026), PD184352(CI-1040), etc.), mTOR inhibitors (e.g., visturtib, etc.), SHP2 inhibitors (e.g., RMC-4630, JAB-3068, TNO155, etc.), or combinations thereof.
Dosage forms of the pharmaceutical composition of the present invention include (but are not limited to): injection, tablet, capsule, aerosol, suppository, pellicle, dripping pill, topical liniment, controlled release type or delayed release type or nanometer preparation.
The pharmaceutical composition of the present invention comprises the compound of the present invention or a pharmacologically acceptable salt thereof in a safe and effective amount range and a pharmacologically acceptable excipient or carrier. Wherein "safe and effective amount" means: the amount of the compound is sufficient to significantly improve the condition without causing serious side effects. Typically, the pharmaceutical composition contains 1-2000mg of a compound of the invention per dose, more preferably, 10-1000mg of a compound of the invention per dose. Preferably, said "dose" is a capsule or tablet.
"pharmaceutically acceptable carrier" refers to: one or more compatible solid or liquid fillers or gel substances which are suitable for human use and must be of sufficient purity and sufficiently low toxicity. By "compatible" is meant herein that the components of the composition are capable of intermixing with and with the compounds of the present invention without significantly diminishing the efficacy of the compounds. Examples of pharmaceutically acceptable carrier moieties are cellulose and its derivatives (e.g. sodium carboxymethylcellulose) Sodium ethylcellulose, cellulose acetate, and the like), gelatin, talc, solid lubricants (e.g., stearic acid, magnesium stearate), calcium sulfate, vegetable oils (e.g., soybean oil, sesame oil, peanut oil, olive oil, and the like), polyols (e.g., propylene glycol, glycerin, mannitol, sorbitol, and the like), emulsifiers
Figure BDA0002861043520000661
Wetting agents (e.g., sodium lauryl sulfate), coloring agents, flavoring agents, stabilizers, antioxidants, preservatives, pyrogen-free water, and the like.
The mode of administration of the compounds or pharmaceutical compositions of the present invention is not particularly limited, and representative modes of administration include (but are not limited to): oral, intratumoral, rectal, parenteral (intravenous, intramuscular or subcutaneous), and topical administration.
Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules. In these solid dosage forms, the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with the following ingredients: (a) fillers or extenders, for example, starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) binders, for example, hydroxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (c) humectants, for example, glycerol; (d) disintegrating agents, for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) slow solvents, such as paraffin; (f) absorption accelerators, e.g., quaternary ammonium compounds; (g) wetting agents, such as cetyl alcohol and glycerol monostearate; (h) adsorbents, for example, kaolin; and (i) lubricants, for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In capsules, tablets and pills, the dosage forms may also comprise buffering agents.
Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared using coatings and shells such as enteric coatings and other materials well known in the art. They may contain opacifying agents and the release of the active compound or compounds in such a composition may be delayed in release in a certain part of the digestive tract. Examples of embedding components which can be used are polymeric substances and wax-like substances. If desired, the active compound may also be in microencapsulated form with one or more of the above-mentioned excipients.
Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly employed in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1, 3-butylene glycol, dimethylformamide and oils, in particular, cottonseed, groundnut, corn germ, olive, castor and sesame oils or mixtures of such materials and the like.
In addition to these inert diluents, the compositions can also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.
Compositions for parenteral injection may comprise physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols and suitable mixtures thereof.
Dosage forms of the compounds of the present invention for topical administration include ointments, powders, patches, sprays, and inhalants. The active ingredient is mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants which may be required if desired.
The therapeutic methods of the invention can be administered alone or in combination with other therapeutic means or agents.
When the pharmaceutical composition is used, a safe and effective amount of the compound of the present invention is suitable for mammals (such as human beings) to be treated, wherein the administration dosage is a pharmaceutically-considered effective administration dosage, and for a human body with a weight of 60kg, the daily administration dosage is usually 1-2000 mg, preferably 50-1000 mg. Of course, the particular dosage will also take into account such factors as the route of administration, the health of the patient, and the like, which are within the skill of the skilled practitioner.
The invention also provides a preparation method of the pharmaceutical composition, which comprises the following steps: mixing a pharmaceutically acceptable carrier with a compound of formula (I) as described herein, or a crystalline form, a pharmaceutically acceptable salt, hydrate, or solvate thereof, to form a pharmaceutical composition.
The present invention also provides a method of treatment comprising the steps of: administering a compound of formula (I), or a crystalline form, a pharmaceutically acceptable salt, hydrate, or solvate thereof, as described herein, or a pharmaceutical composition as described herein, to a subject in need thereof for selectively inhibiting KRASG12C
Compared with the prior art, the invention has the following main advantages:
(1) the compound is right to KRASG12CHas good selective inhibition effect;
(2) the compound has better pharmacodynamics and pharmacokinetic performance and lower toxic and side effects.
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.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. In addition, any methods and materials similar or equivalent to those described herein can be used in the methods of the present invention. The preferred embodiments and materials described herein are exemplary only.
The structure of the compounds of the invention is determined by Nuclear Magnetic Resonance (NMR) and liquid mass spectrometry (LC-MS).
NMR was detected using a Bruker AVANCE-400 nuclear magnetic instrument, and the assay solvent contained deuterated dimethylsulfoxide (DMSO-d)6) Deuterated acetone (CD)3COCD3) Deuterated chloroform (CDCl)3) And deuterated methanol (CD)3OD), and internal standards are Tetramethylsilane (TMS), chemical shifts are measured in parts per million (ppm).
Liquid chromatography-mass spectrometry (LC-MS) was detected using a Waters SQD2 mass spectrometer. HPLC measurements were performed using an Agilent1100 high pressure chromatograph (Microsorb 5micron C18100 x 3.0.0 mm column).
Thin layer chromatography silica gel plate is blue island GF254 silica gel plate, TLC is 0.15-0.20mm, and preparative thin layer chromatography is 0.4-0.5 mm. Column chromatography generally uses Qingdao silica gel 200-300 mesh silica gel as a carrier.
The starting materials in the examples of the present invention are known and commercially available or may be used or synthesized according to literature reports in the art.
All reactions of the present invention are carried out under continuous magnetic stirring in a dry inert gas (e.g., nitrogen or argon) at temperatures in degrees celsius, unless otherwise specified.
Examples
Example 14 preparation of- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -1- (4-methyl-2- (methylsulfonyl) pyridin-3-yl) pyrido [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000691
Step 1 preparation of 4-methyl-2- (methylsulfonyl) -3-nitropyridine
2-chloro-4-methyl-3-nitropyridine (3g, 17.4mmol) was dissolved in 25 ml of dimethyl sulfoxide, and sodium methylsulfinate (2.7g, 26.2mmol) was added to the solution. The mixture is stirred at 120 ℃ for 1 hour, quenched with 100 ml of water and then extracted 3 times with 100 ml of ethyl acetate. The combined organic phases were washed 3 times with 50 ml brine, then the organic phases were dried and concentrated. The residual solid was slurried with ethanol/methanol/ethyl acetate (40 ml/5 ml), filtered off with suction and dried to give the title compound (2.3g, 61%).
Step 2 preparation of 4-methyl-2- (methylsulfonyl) pyridin-3-amine
4-methyl-2- (methylsulfonyl) -3-nitropyridine (2.2g, 10.2mmol) was dissolved in 110 mL of methanol and 10% palladium on carbon (50% w/w, 660mg) was added to the solution. The mixture was stirred at room temperature for 5 hours under a hydrogen atmosphere. The catalyst was removed by suction filtration, the filtrate was concentrated, and the residue was separated with a silica gel column (petroleum ether: ethyl acetate: 5: 1 to 2: 1) to obtain the objective compound (820mg, yield: 41%).
LC-MS:m/z 187(M+H)+
Step 3 preparation of 2, 6-dichloro-5-fluoro-N- (((4-methyl-2- (methylsulfonyl) pyridin-3-yl) carbamoyl) nicotinamide
2, 6-dichloro-5-fluoronicotinamide (873mg, 4.2mmol) was dissolved in 15 mL of anhydrous tetrahydrofuran, to which was slowly added dropwise a solution of oxalyl chloride (3.6mL, 42.0mmol) in dichloromethane (4.5 mL). After the addition was complete, the mixture was stirred at 75 ℃ under reflux for 2 hours and then concentrated to dryness under reduced pressure. The residue was diluted with 15 ml of anhydrous tetrahydrofuran and cooled to zero degrees. 4-methyl-2- (methylsulfonyl) pyridin-3-amine (820mg, 4.4mmol) was dissolved in 6mL of anhydrous tetrahydrofuran and added dropwise to the solution. The reaction mixture was stirred at zero degrees for 2 hours, quenched with saturated ammonium chloride/saturated brine (V: V ═ 1: 1, 30mL), and then extracted 3 times with dichloromethane/methanol (V: V ═ 10: 1, 30 mL). The combined organic phases were dried, concentrated, and the residual solid was slurried with petroleum ether/ethyl acetate (2: 1, 25mL), suction filtered, and dried to give the title compound (1.36g, yield: 77%).
LC-MS:m/z 421(M+H)+
Step 4 preparation of 7-chloro-6-fluoro-1- (4-methyl-2- (methylsulfonyl) pyridin-3-yl) pyridin [2,3-d ] pyrimidine-2, 4(1H, 3H) -dione
Suspending 2, 6-dichloro-5-fluoro-N- (((4-methyl-2- (methylsulfonyl) pyridin-3-yl) carbamoyl) nicotinamide (1.13g, 2.7mmol) in 34 mL of tetrahydrofuran, dropwise adding bis (trimethylsilyl) amino potassium (1 mol of tetrahydrofuran solution, 6.2mL, 6.2mmol) under ice bath, clarifying the reaction solution after completion of dropwise addition, stirring the reaction solution at 60 ℃ for 2 hours, quenching 40 mL of saturated ammonium chloride, extracting with 40 mL of ethyl acetate for 3 times, combining ethyl acetate layers, drying, concentrating, slurrying the residual solid with petroleum ether/ethyl acetate (V: V ═ 1: 1, 25mL), suction-filtering, and drying to obtain the objective compound (870mg, yield: 84%).
LC-MS:m/z 385(M+H)+
Preparation of (S) -tert-butyl 4- (7-chloro-6-fluoro-1- (4-methyl-2- (methylsulfonyl) pyridin-3-yl) -2-oxo-1, 2-dihydropyridine [2,3-d ] pyrimidin-4-yl) -3-methylpiperazine-1-carboxylate in step 5
7-chloro-6-fluoro-1- (4-methyl-2- (methylsulfonyl) pyridin-3-yl) pyridine [2,3-d ] pyrimidine-2, 4(1H, 3H) -dione (300mg, 0.8mmol) was suspended in 9 mL acetonitrile, N-diisopropylethylamine (0.77mL, 4.7mmol) and phosphorus oxychloride (0.36mL, 3.9mmol) were added dropwise, and the reaction solution became clear. The reaction mixture was stirred at 80 ℃ for 1 hour, and concentrated to dryness under reduced pressure. The residue was dissolved in 12 mL acetonitrile, cooled to zero, and N, N-diisopropylethylamine (0.4mL, 2.3mmol) and tert-butyl (S) -3-methylpiperazine-1-carboxylate (188mg, 0.9mmol) were added. The reaction was stirred at room temperature for 1 hour, quenched with saturated sodium bicarbonate solution (20mL), and extracted 3 times with 20mL of ethyl acetate. The combined ethyl acetate layers were dried, concentrated, and subjected to silica gel column separation (petroleum ether: ethyl acetate: 3: 1 to pure ethyl acetate) to obtain the objective compound (328mg, yield: 74%).
LC-MS:m/z 567(M+H)+
Preparation of (S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -7-chloro-6-fluoro-1- (4-methyl-2- (methylsulfonyl) pyridin-3-yl) pyrido [2,3-d ] pyrimidin-2 (1H) -one in step 6
(S) -tert-butyl 4- (7-chloro-6-fluoro-1- (4-methyl-2- (methylsulfonyl) pyridin-3-yl) -2-oxo-1, 2-dihydropyridine [2,3-d ] pyrimidin-4-yl) -3-methylpiperazine-1-carboxylate (328mg, 0.6mmol) was dissolved in 4 ml of dichloromethane, 1ml of trifluoroacetic acid was added, the reaction solution was stirred at room temperature for 2 hours, concentrated to dryness, and the residue was co-evaporated with 15 ml of dichloromethane 3 times to give a crude product. The crude product was dissolved in 6 mL of dichloromethane, cooled to zero, and a solution of N, N-diisopropylethylamine (0.38mL, 2.3mmol) and acryloyl chloride (63mg, 0.7mmol) in dichloromethane (1mL) was added dropwise. The reaction was stirred at zero degrees for 30 minutes, quenched with 20 ml of saturated sodium bicarbonate and extracted 3 times with 20 ml of dichloromethane. The dichloromethane layers were combined, dried, concentrated, and the residue was separated with a silica gel column (dichloromethane: methanol 60: 1) to obtain the objective compound (200mg, yield: 67%).
LC-MS:m/z 521(M+H)+
Step 7 preparation of 4- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -1- (4-methyl-2- (methylsulfonyl) pyridin-3-yl) pyrido [2,3-d ] pyrimidin-2 (1H) -one
(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -7-chloro-6-fluoro-1- (4-methyl-2- (methylsulfonyl) pyridin-3-yl) pyrido [2,3-d ] pyrimidin-2 (1H) -one (126mg, 0.24mmol), (2-fluoro-6-hydroxyphenyl) boronic acid (49mg, 0.31mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane complex (20mg, 0.024mmol), and potassium acetate (95mg, 0.97mmol) were suspended in a mixed solvent of dioxane/water (7mL/0.7mL), replaced with nitrogen 3 times, and stirred at 90 degrees with heating for 2.5 hours. After the reaction solution was cooled to room temperature, 20 ml of a half-saturated sodium bicarbonate solution was added, and extracted 3 times with 20 ml of ethyl acetate. The ethyl acetate layers were combined, dried, concentrated, and the residue was separated with a silica gel column (dichloromethane: methanol 100: 1 to 60: 1) to obtain the objective compound (45mg, yield: 31%).
LC-MS:m/z 597(M+H)+1H NMR(400MHz,CDCl3)δ8.65-8.63(m,1H),8.57(brs,1H),7.89(t,J=8.8Hz,1H),7.60(d,J=4.4Hz,1H),7.26(m,1H),6.71-6.56(m,3H),6.40(d,J=16.4Hz,1H),5.81(d,J=10.4Hz,1H),5.10-4.50(m,3H),4.10-3.50(m,3H),3.27(d,J=2.4Hz,3H),3.26-3.00(m,1H),2.28(d,J=6.8Hz,3H),1.53(m,3H).
The following compounds were synthesized according to the procedure of example 1, starting from different starting materials:
example 24- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -1- (2-methyl-4- (methylsulfonyl) pyridin-3-yl) pyrido [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000711
LC-MS:m/z 597(M+H)+1H NMR(400MHz,CDCl3)δ8.93(d,J=5.2Hz,1H),8.65(brs,1H),7.95-7.89(m,2H),7.26(m,1H),6.72-6.75(m,3H),6.45-6.39(m,1H),5.85-5.81(m,1H),5.25-4.30(m,3H),4.20-3.50(m,3H),3.30-3.00(m,4H),2.46(d,J=10.0Hz,3H),1.51-1.48(m,3H).
Example 32- ((2S) -4-acryloyl-1- (6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -1- (2-methyl-6- (methylsulfonyl) phenyl) -2-oxo-1, 2-dihydropyridin [2,3-d ] pyrimidin-4-yl) piperazin-2-yl) acetonitrile
Figure BDA0002861043520000712
LC-MS:m/z 621(M+H)+1H NMR(400MHz,CDCl3)δ9.01-8.91(m,1H),8.11(t,J=7.2Hz,1H),7.95(brs,1H),7.75(t,J=8.4Hz,1H),7.70-7.64(m,1H),7.26(m,1H),6.71-6.59(m,3H),6.46(d,J=16.4Hz,1H),5.88(d,J=10.4Hz,1H),5.50-5.30(m,1H),4.75-3.40(m,6H),3.10(d,J=4.8Hz,3H),3.05-2.80(m,2H),2.18(d,J=10.8Hz,3H).
Example 44- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -1- (2- (isopropylsulfo) -6-methylphenyl) pyridine [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000721
LC-MS:m/z 624(M+H)+1H NMR(400MHz,CDCl3)δ8.98-8.94(m,1H),8.04(d,J=8.0Hz,1H),7.87(t,J=8.0Hz,1H),7.72(d,J=7.6Hz,1H),7.62(t,J=8.0Hz,1H),7.26(m,1H),6.71-6.54(m,3H),6.44-6.39(m,1H),5.83-5.80(m,1H),5.10-4.30(m,3H),4.10-3.00(m,5H),2.16(d,J=8.4Hz,3H),1.52(brs,3H),1.31-1.28(m,3H),1.16-1.14(m,3H).
Example 54- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -1- (2-ethyl-6- (methylsulfonyl) phenyl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) pyridine [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000722
LC-MS:m/z 610(M+H)+1H NMR(400MHz,CDCl3)δ8.99(s,1H),8.12(d,J=7.6Hz,1H),7.87(dd,J=9.2Hz,3.2Hz,1H),7.78(d,J=7.6Hz,1H),7.71(t,J=7.6Hz,1H),7.26(m,1H),6.69-6.54(m,3H),6.44-6.38(m,1H),5.83-5.80(m,1H),5.10-4.38(m,3H),4.10-3.50(m,3H),3.30-3.00(m,4H),2.59-2.41(m,2H),1.58-1.49(m,3H),1.21-1.16(m,3H).
Example 5-1 two isomers, example 5A and example 5B, were obtained by chiral separation:
Figure BDA0002861043520000723
example 5A: LC-MS: m/z 610(M + H)+1H NMR(400MHz,CDCl3)δ9.02(s,1H),8.14(d,J=7.3Hz,1H),7.90(d,J=8.5Hz,1H),7.81(d,J=7.1Hz,1H),7.73(t,J=7.8Hz,1H),7.33–7.23(m,1H),6.76–6.53(m,3H),6.44(d,J=16.3Hz,1H),5.84(d,J=10.0Hz,1H),5.03(m,1H),4.57(m,2H),4.22–3.41(m,3H),3.35–3.00(m,4H),2.48(m,2H),1.51(s,3H),1.21(t,J=6.7Hz,3H).
Example 5B: LC-MS: m/z 610(M + H)+1H NMR(400MHz,CDCl3)δ9.01(s,1H),8.21–8.07(m,1H),7.90(d,J=9.3Hz,1H),7.81(d,J=7.0Hz,1H),7.73(t,J=7.8Hz,1H),7.35–7.23(m,1H),6.68(m,3H),6.44(d,J=16.5Hz,1H),5.84(d,J=10.2Hz,1H),5.12–4.29(m,3H),3.99(m,1H),3.69(m,2H),3.22(m,4H),2.69–2.37(m,2H),1.66–1.43(m,3H),1.25–1.12(m,3H).
Example 64- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -1- (2-cyclopropyl-6- (methylsulfonyl) phenyl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) pyrido [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000731
LC-MS:m/z 622(M+H)+1H NMR(400MHz,CDCl3)δ9.00(s,1H),8.08(d,J=7.6Hz,1H),7.88(d,J=9.2Hz,1H),7.67-7.63(m,1H),7.46(d,J=7.6Hz,1H),7.29-7.23(m,1H),6.75-6.50(m,3H),6.44-6.39(m,1H),5.83-5.80(m,1H),5.04-4.38(m,3H),4.06-3.55(m,3H),3.24-3.02(m,4H),1.55-1.49(m,4H),0.85-0.56(m,4H).
Example 6-1 two isomers, example 5A and example 5B, were obtained by chiral separation:
Figure BDA0002861043520000732
Example 6A: LC-MS: m/z 622(M + H)+1H NMR(400MHz,CDCl3)δ9.03(s,1H),8.10(d,J=7.2Hz,1H),7.90(d,J=8.9Hz,1H),7.67(t,J=7.9Hz,1H),7.48(dd,J=7.9,1.0Hz,1H),7.40–7.17(m,1H),6.81–6.54(m,3H),6.43(dd,J=16.7,1.3Hz,1H),5.84(d,J=10.3Hz,1H),5.08(m,1H),4.59(m,2H),4.25–3.44(m,3H),3.31–2.99(m,4H),1.70(m,1H),1.51(s,3H),0.96–0.78(m,2H),0.65(m,2H).
Example 6B: LC-MS: m/z 622(M + H)+1H NMR(400MHz,CDCl3)δ9.02(s,1H),8.10(d,J=7.5Hz,1H),7.91(d,J=9.2Hz,1H),7.67(t,J=7.9Hz,1H),7.47(d,J=7.7Hz,1H),7.36–7.19(m,1H),6.68(m,3H),6.43(d,J=16.6Hz,1H),5.84(d,J=10.3Hz,1H),4.69(m,3H),3.98(m,1H),3.70(m,2H),3.35–3.02(m,4H),1.63(m,4H),0.87(m,2H),0.67(m,2H).
Example 74- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -1- (2- (cyclopropylsulfonyl) -6-methylphenyl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) pyridinyl [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000741
LC-MS:m/z 622(M+H)+1H NMR(400MHz,CDCl3)δ9.00(s,1H),7.97-7.85(m,2H),7.71(d,J=7.6Hz,1H),7.61(t,J=8.0Hz,1H),7.29-7.23(m,1H),6.69-6.63(m,3H),6.44-6.39(m,1H),5.83-5.80(m,1H),5.05-4.37(m,3H),4.02-3.63(m,3H),3.23-2.78(m,2H),2.19(d,J=10.8Hz,3H),1.41-1.37(m,4H).1.03-0.96(m,3H).
Example 84- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -1- (2-chloro-6- (methylsulfonyl) phenyl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) pyrido [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000742
LC-MS:m/z 616(M+H)+1H NMR(400MHz,CDCl3)δ8.81(s,1H),8.19(d,J=8.0Hz,1H),7.95-7.85(m,2H),7.70(t,J=8.4Hz,1H),7.33-7.23(m,1H),6.74-6.50(m,3H),6.10(d,J=16.8Hz,1H),6.10(dd,J=10.4Hz,8Hz;1H),5.15-4.30(m,3H),4.15-3.39(m,3H),3.31-2.95(m,4H),1.51(s,3H).
Example 94- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -7- (2- (difluoromethyl) -6-fluorophenyl) -6-fluoro-1- (2-methyl-6- (methylsulfonyl) phenyl) pyrido [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000743
LC-MS:m/z 630(M+H)+1H NMR(400MHz,CDCl3)δ8.01(d,J=7.6Hz,1H),7.87-7.83(m,1H),7.64-7.62(m,1H),7.56-7.46(m,3H),7.26-7.22(m,1H),6.69-6.30(m,3H),5.82(dd,J=10.4Hz,1.6Hz,1H),5.04-4.36(m,3H),4.05-3.62(m,3H),3.27-3.08(m,4H),2.16-2.13(m,3H),1.60-1.49(m,3H).
Example 104- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -1- (2-isopropyl-6- (methylsulfonyl) phenyl) pyridine [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000751
LC-MS:m/z 624(M+H)+1H NMR(400MHz,CDCl3)δ8.98(s,1H),8.12(dd,J=8.0Hz,1.6Hz,1H),7.89-7.82(m,2H),7.74(t,J=8.0Hz,1H),7.26(m,1H),6.69-6.62(m,3H),6.42(dd,J=16.8Hz,1.6Hz,1H),5.82(dd,J=10.4Hz,1.6Hz,1H),5.15-4.65(m,3H),4.15-3.50(m,3H),3.30-3.00(m,4H),2.85-2.70(m,1H),1.56-1.48(m,3H),1.25(dd,J=6.8Hz,2.4Hz,3H),1.05(t,J=6.4Hz,3H).
Example 10-1 two isomers, example 10A and example 10B, were obtained by chiral separation:
Figure BDA0002861043520000752
example 10A: LC-MS: m/z 624(M + H)+1H NMR(400MHz,DMSO)δ10.23(s,1H),8.31(t,J=10.5Hz,1H),7.95–7.75(m,2H),7.66(t,J=7.9Hz,1H),7.26(m,1H),6.98–6.78(m,1H),6.77–6.59(m,2H),6.22(d,J=16.6Hz,1H),5.78(dd,J=10.5,2.1Hz,1H),4.92(s,1H),4.37(m,2H),4.10(m,1H),3.88–3.46(m,2H),3.18(m,1H),2.65(d,J=6.0Hz,1H),1.34(d,J=6.6Hz,3H),1.10(d,J=6.8Hz,3H),1.00(d,J=6.8Hz,3H).
Example 10B: LC-MS: m/z 624(M + H)+1H NMR(400MHz,DMSO)δ10.28(s,1H),8.33(t,J=9.2Hz,1H),7.98–7.77(m,2H),7.66(t,J=7.8Hz,1H),7.26m,1H),6.88(m,1H),6.79–6.52(m,2H),6.22(dd,J=16.6,6.0Hz,1H),5.78(dd,J=10.4,2.2Hz,1H),4.89(s,1H),4.55–3.91(m,3H),3.65(m,1H),3.47(m,1H),3.08(m,1H),2.67(m,1H),1.30(s,3H),1.11(d,J=6.8Hz,3H),1.02(m,3H).
Example 11N- (2- (4- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -2-oxopyrano [2,3-d ] pyrimidin-1 (2H) -yl) -3-methylphenyl) -N-methylmethanesulfonamide
Figure BDA0002861043520000761
LC-MS:m/z 625(M+H)+1H NMR(400MHz,CDCl3)δ8.64(brs,1H),7.86-7.78(m,1H),7.47(td,J=7.6Hz,1.6Hz;1H),7.40(d,J=7.2Hz,1H),7.32-7.21(m,2H),6.70-6.50(m,3H),6.40(dd,J=16.8Hz,1.6Hz;1H),6.10(dd,J=10.4Hz,1.6Hz;1H),5.05-4.25(m,3H),4.10-3.46(m,3H),3.31-2.99(m,4H),2.85(d,J=2.0Hz,3H),2.17(s,3H),1.54-1.42(m,3H).
Example 124- ((2S, 5R) -4-acryloyl-2, 5-dimethylpiperazin-1-yl) -1- (2-ethyl-6- (methylsulfonyl) phenyl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) pyrido [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000762
LC-MS:m/z 624(M+H)+1H NMR(400MHz,CDCl3)δ9.03(t,J=18.4Hz,1H),8.14-8.10(m,1H),7.89-7.68(m,3H),7.28-7.23(m,1H),6.71-6.36(m,4H),,5.83-5.79(m,1H),5.12-3.47(m,6H),3.14-3.10(m,3H),2.55-2.37(m,2H),1.51-1.16(m,9H).
Example 13(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -1- (2-cyclopropyl-6- (methylsulfonyl) phenyl) -6-fluoro-7- (2-fluorophenyl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000763
LC-MS:m/z 606(M+H)+1H NMR(400MHz,CDCl3)δ7.99(d,J=7.6Hz,1H),7.80-7.77(m,1H),7.55(t,J=8.0Hz,1H),7.42-7.38(m,2H),7.31-7.26(m,1H),7.16-7.08(m,2H),6.70-6.52(m,1H),6.41(dd,J=16.8Hz,1.6Hz,1H),5.81(dd,J=10.4Hz,1.6Hz,1H),5.04-4.36(m,3H),4.05-3.58(m,3H),3.24-3.04(m,4H),1.67-1.60(m,1H),1.54-1.47(m,3H),0.82-0.76(m,2H),0.66-0.53(m,2H).
Example 142- (4- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -2-oxopyrano [2,3-d ] pyrimidin-1 (2H) -yl) -N, N, 3-trimethylbenzenesulfonamide
Figure BDA0002861043520000771
LC-MS:m/z 625(M+H)+1H NMR(400MHz,CDCl3)δ9.04(brs,1H),7.96(d,J=8.0Hz,1H),7.89-7.81(m,1H),7.80(d,J=7.6Hz,1H),7.57(t,J=7.6Hz,1H),7.30-7.22(m,1H),6.71-6.51(m,3H),6.41(dd,J=16.4Hz,1.6Hz;1H),5.82(dd,J=10.4Hz,1.6Hz;1H),5.16-2.92(m,7H),2.73(s,6H),2.50(d,J=7.2Hz,3H),1.50-1.41(m,3H).
Example 15(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -1- (2-cyclopropyl-6- (methylsulfonyl) phenyl) -7- (2, 6-difluorophenyl) -6-fluoropyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000772
LC-MS:m/z 624(M+H)+1H NMR(400MHz,CDCl3)δ7.95(d,J=8.0Hz,1H),7.83-7.78(m,1H),7.50(t,J=8.0Hz,1H),7.42-7.33(m,2H),6.92(t,J=8.4Hz,2H),6.70-6.53(m,1H),6.41(dd,J=16.4Hz,1.6Hz,1H),5.81(dd,J=10.4Hz,2.0Hz,1H),5.08-4.28(m,3H),4.09-3.58(m,3H),3.31-3.07(m,4H),1.65-1.57(m,1H),1.57-1.45(m,3H),0.78-0.74(m,2H),0.66-0.55(m,2H).
Example 164- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -1- (4-isopropyl-2- (methylsulfonyl) pyridin-3-yl) pyridinyl [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000773
LC-MS:m/z 625(M+H)+1H NMR(400MHz,CDCl3)δ8.73-8.70(m,1H),8.50-8.41(m,1H),7.89(dd,J=9.2Hz,2.4Hz,1H),7.67(d,J=5.2Hz,1H),7.26(m,1H),6.70-6.50(m,3H),6.40(d,J=16.8Hz,1H),5.81(d,J=10.4Hz,1H),5.10-4.34(m,3H),4.04-3.60(m,3H),3.28-3.09(m,4H),3.00-2.80(m,1H),1.57-1.49(m,3H),1.27(dd,J=6.8Hz,1.6Hz,3H),1.07(t,J=6.4Hz,3H).
Example 16-1 two isomers were obtained by chiral separation, example 16A and example 16B:
Figure BDA0002861043520000781
example 16A: LC-MS: m/z625(M + H)+1H NMR(400MHz,DMSO)δ10.22(s,1H),8.70(d,J=4.7Hz,1H),8.34(d,J=9.3Hz,1H),7.87(d,J=4.8Hz,1H),7.26(dd,J=15.2,7.8Hz,1H),6.85(dd,J=15.8,10.0Hz,1H),6.79–6.56(m,2H),6.21(d,J=16.2Hz,1H),5.77(d,J=10.5Hz,1H),4.97(s,1H),4.22(m,3H),3.61(m,2H),3.30–2.90(m,4H),2.81(m,1H),1.31(d,J=6.3Hz,3H),1.12(d,J=6.6Hz,3H),1.03(d,J=6.5Hz,3H).
Example 16B: LC-MS: m/z625(M + H)+1H NMR(400MHz,DMSO)δ10.22(s,1H),8.70(d,J=5.0Hz,1H),8.31(t,J=10.7Hz,1H),7.87(d,J=5.0Hz,1H),7.26(dd,J=15.4,8.1Hz,1H),7.01–6.77(m,1H),6.76–6.57(m,2H),6.29–6.11(m,1H),5.77(dd,J=10.5,2.0Hz,1H),4.86(s,1H),4.50–3.93(m,3H),3.78–3.40(m,2H),3.30–2.94(m,4H),2.80(m,1H),1.31(d,J=6.5Hz,3H),1.13(d,J=6.8Hz,3H),1.03(d,J=6.7Hz,3H).
Example 17(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -7- (2-chlorophenyl) -1- (2-cyclopropyl-6- (methylsulfonyl) phenyl) -6-fluoropyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000782
LC-MS:m/z 622(M+H)+1H NMR(400MHz,CDCl3)δ7.96(d,J=7.6Hz,1H),7.80-7.77(m,1H),7.51(t,J=8.0Hz,1H),7.41-7.28(m,3H),7.26(m,2H),6.68-6.54(m,1H),6.42(dd,J=16.8Hz,2.0Hz,1H),5.81(dd,J=10.4Hz,1.6Hz,1H),5.08-4.33(m,3H),4.07-3.59(m,3H),3.28-3.09(m,4H),1.69-1.65(m,1H),1.57-1.47(m,3H),0.82-0.76(m,2H),0.67-0.51(m,2H).
Example 184- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -1- (2-isopropyl-4- (methylsulfonyl) pyridin-3-yl) pyridinyl [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000791
LC-MS:m/z 625(M+H)+1H NMR(400MHz,CDCl3)δ9.02(t,J=8.8Hz,1H),8.67(t,J=4.8Hz,1H),7.93-7.89(m,2H),7.28-7.26(m,1H),6.71-6.40(m,4H),5.84-5.81(m,1H),4.48-3.68(m,7H),3.15(s,3H),2.97-2.93(m,1H),1.59-1.07(m,9H).
Example 18-1 two isomers, example 18A and example 18B, were obtained by chiral separation:
Figure BDA0002861043520000792
example 18A: LC-MS: m/z 625(M + H)+1H NMR(400MHz,DMSO)δ10.31(s,1H),8.96(d,J=5.0Hz,1H),8.40(t,J=10.2Hz,1H),7.89(d,J=5.0Hz,1H),7.32(dd,J=15.3,8.3Hz,1H),7.02–6.84(m,1H),6.83–6.64(m,2H),6.26(dd,J=16.6,5.1Hz,1H),5.82(dd,J=10.4,2.3Hz,1H),4.96(s,1H),4.29(m,3H),3.58(m,2H),3.36–2.87(m,5H),1.43–1.33(m,3H),1.17(d,J=6.7Hz,3H),1.10–1.00(m,3H).
Example 18B: LC-MS: m/z 625(M + H)+1H NMR(400MHz,DMSO)δ10.24(s,1H),8.90(d,J=5.0Hz,1H),8.35(t,J=10.4Hz,1H),7.83(d,J=5.0Hz,1H),7.26(dd,J=15.3,8.2Hz,1H),6.95–6.79(m,1H),6.78–6.57(m,2H),6.21(d,J=16.6Hz,1H),5.77(dd,J=10.5,2.1Hz,1H),4.97(s,1H),4.22(m,3H),3.86–3.43(m,2H),3.29–2.81(m,5H),1.32(t,J=7.0Hz,3H),1.12(t,J=8.2Hz,3H),1.02(d,J=6.6Hz,3H).
Example 194- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-7- (2-fluoro-6-methoxyphenyl) -1- (4-isopropyl-2- (methylsulfonyl) pyridin-3-yl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000793
LC-MS:m/z 639(M+H)+1H NMR(400MHz,CDCl3)δ8.66(d,J=5.2Hz,1H),7.79-7.75(m,1H),7.58(d,J=4.4Hz,1H),7.33-7.29(m,1H),6.71-6.60(m,3H),6.41-6.38(m,1H),5.80(d,J=10.4Hz,1H),5.04-4.35(m,3H),4.00-3.60(m,6H),3.18(m,4H),2.92(m,1H),1.48(m,3H),1.23(d,J=6.8Hz,3H),1.09(m,3H).
Example 20(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -7- (2-chlorophenyl) -6-fluoro-1- (4-isopropyl-2- (methylsulfonyl) pyridin-3-yl) pyridine [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000801
LC-MS:m/z 625(M+H)+。δ8.67-8.65(m,1H),7.81-7.77(m,1H),7.57(d,J=4.8Hz,1H),7.40-7.27(m,4H),6.61(m,1H),6.42-6.38(m,1H),5.80(d,J=10.4Hz,1H),5.16-4.36(m,3H),4.02-3.59(m,3H),3.17(m,4H),2.95(m,1H),1.49(m,3H),1.25(d,J=6.8Hz,3H),1.06(d,J=6.8Hz,3H).
Example 20-1 two isomers, example 20A and example 20B, were obtained by chiral separation:
Figure BDA0002861043520000802
example 20A: LC-MS: m/z 625(M + H)+1H NMR(400MHz,DMSO)δ8.71(d,J=5.0Hz,1H),8.41(t,J=9.3Hz,1H),7.88(d,J=5.0Hz,1H),7.56(d,J=7.1Hz,1H),7.50(dd,J=7.4,1.6Hz,1H),7.42(t,J=7.5Hz,1H),7.38–7.25(m,1H),6.86(dd,J=16.6,9.3Hz,1H),6.22(d,J=16.4Hz,1H),5.89–5.69(m,1H),5.02(s,1H),4.23(m,3H),3.93–3.57(m,2H),3.30–3.01(m,4H),2.88(m,1H),1.31(d,J=6.5Hz,3H),1.15(d,J=6.8Hz,3H),1.02(d,J=6.8Hz,3H).
Example 20B: LC-MS: m/z 625(M + H)+1H NMR(400MHz,DMSO)δ8.71(d,J=5.0Hz,1H),8.37(t,J=10.5Hz,1H),7.88(d,J=5.0Hz,1H),7.48(m,3H),7.30(d,J=7.3Hz,1H),6.87(d,J=10.4Hz,1H),6.22(d,J=16.4Hz,1H),5.78(d,J=12.5Hz,1H),4.86(s,1H),4.62–3.99(m,3H),3.58(m,2H),3.11(m,4H),2.94–2.81(m,1H),1.33(d,J=6.6Hz,3H),1.15(d,J=6.8Hz,3H),1.02(d,J=6.8Hz,3H).
Example 216-fluoro-7- (2-fluoro-6-hydroxyphenyl) -4- ((S) -4- (2-fluoroacryloyl) -2-methylpiperazin-1-yl) -1- (2-isopropyl-6- (methylsulfonyl) phenyl) pyridine [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000811
LC-MS:m/z 642(M+H)+1H NMR(400MHz,CDCl3)δ8.97(s,1H),8.13-8.11(m,1H),7.88-7.82(m,2H),7.76-7.72(m,1H),7.28-7.22(m,1H),6.66-6.62(m,2H),5.48-5.35(m,1H),5.26-5.22(m,1H),5.02-4.90(m,1H),4.53-3.65(m,6H),3.11(s,3H),2.82-2.73(m,1H),1.60-1.03(m,9H).
Example 22(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -7- (2-chloro-4-fluorophenyl) -6-fluoro-1- (4-isopropyl-2- (methylsulfonyl) pyridin-3-yl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000812
LC-MS:m/z 643(M+H)+1H NMR(400MHz,CDCl3)δ8.68-8.67(m,1H),7.81-7.78(m,1H),7.59(d,J=5.2Hz,1H),7.34(m,1H),7.14(dd,J=4.4,2.4Hz,1H),7.02-6.99(m,1H),6.61-6.58(m,1H),6.4-6.38(m,1H),5.82-5.80(m,1H),5.01-3.62(m,6H),3.18(m,4H),2.96(m,1H),1.48(m,3H),1.26(d,J=6.8Hz,3H),1.07-1.05(m,3H).
Example 23(S) -7- (2-chlorophenyl) -6-fluoro-4- (4- (2-fluoroacryloyl) -2-methylpiperazin-1-yl) -1- (2-isopropyl-6- (methylsulfonyl) phenyl) pyridine [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000813
LC-MS:m/z 642(M+H)+1H NMR(400MHz,CDCl3)δ7.98(d,J=6.8Hz,1H),7.79-7.75(m,1H),7.70-7.69(m,1H),7.60-7.56(m,1H),7.39-7.19(m,4H),5.46-5.33(m,1H),5.25-5.20(m,1H),4.93(m,1H),4.65-3.20(m,6H),3.07(s,3H),2.81-2.75(m,1H),1.52(d,J=6.8Hz,3H),1.23(d,J=6.8Hz,3H),1.02-1.00(m,3H).
Example 242- (4- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-1- (4-isopropyl-2- (methylsulfonyl) pyridin-3-yl) -2-oxo-1, 2-dihydropyridine [2,3-d ] pyrimidin-7-yl) -3-fluorophenyl acetate
Figure BDA0002861043520000821
LC-MS:m/z 667(M+H)+1H NMR(400MHz,CDCl3)δ8.68-8.67(m,1H),7.85-7.79(m,1H),7.56(d,J=4.8Hz,1H),7.43-7.37(m,1H),7.02-6.95(m,2H),6.70-6.53(m,1H),6.42-6.38(m,1H),5.81-5.79(m,1H),5.08-3.63(m,6H),3.17(m,4H),2.76(m,1H),2.02(s,3H),1.46(m,3H),1.20(d,J=6.8Hz,3H),0.99(m,3H).
Example 254- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -1- (2-cyclobutyl-6- (methylsulfonyl) phenyl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) pyridine [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000822
LC-MS:m/z 636(M+H)+1H NMR(400MHz,CDCl3)δ8.97(s,1H),8.11(d,J=6.8Hz,1H),8.10-7.70(m,3H),7.28-7.22(m,1H),6.69-6.60(m,3H),6.42-6.39(m,1H),5.83-5.80(m,1H),5.05-3.64(m,6H),3.12(m,5H),2.30-2.25(m,1H),2.11-2.08(m,2H),1.85-1.71(m,3H),1.47(d,J=6.4Hz,3H).
Example 25-1 two isomers, example 25A and example 25B, were obtained by chiral separation:
Figure BDA0002861043520000823
example 25A: LC-MS: m/z 636(M + H)+1H NMR(400MHz,DMSO)δ10.24(s,1H),8.32(t,J=9.3Hz,1H),7.89(dd,J=7.8,1.1Hz,1H),7.76(d,J=7.3Hz,1H),7.65(m,1H),7.27(m,1H),6.87(m,1H),6.79–6.58(m,2H),6.22(m,1H),5.78(m,1H),4.87(m,1H),4.21(m,3H),3.78–3.42(m,2H),3.31–2.91(m,5H),2.19–1.87(m,3H),1.87–1.57(m,3H),1.33(m,3H).
Example 25B: LC-MS: m/z 636(M + H)+1H NMR(400MHz,DMSO)δ10.24(s,1H),8.33(t,J=10.1Hz,1H),7.89(dd,J=7.8,1.1Hz,1H),7.76(d,J=7.3Hz,1H),7.66(m,1H),7.27(m,1H),6.99–6.79(m,1H),6.79–6.58(m,2H),6.21(m,1H),5.77(m,1H),4.88(s,1H),4.55–3.97(m,3H),3.77–3.42(m,2H),3.30–2.89(m,5H),2.13–1.88(m,3H),1.89–1.57(m,3H),1.29(m,3H).
Example 26(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -7- (2-chloro-3-fluorophenyl) -6-fluoro-1- (4-isopropyl-2- (methylsulfonyl) pyridin-3-yl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000831
LC-MS:m/z 643(M+H)+1H NMR(400MHz,CDCl3)δ8.67-8.66(m,1H),7.83-7.80(m,1H),7.58(d,J=4.2Hz,1H),7.28-7.13(m,3H),6.61-6.58(m,1H),6.42-6.39(m,1H),5.83-5.80(m,1H),5.34-3.62(m,6H),3.18(m,4H),2.97(m,1H),1.49(m,3H),1.27-1.25(m,3H),1.07-1.06(m,3H).
Example 274- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -7- (2-chloro-6-hydroxyphenyl) -6-fluoro-1- (4-isopropyl-2- (methylsulfonyl) pyridin-3-yl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000832
LC-MS:m/z 641(M+H)+1H NMR(400MHz,CDCl3)δ8.61(m,1H),7.90-7.86(m,1H),7.61-7.60(m,1H),7.26-7.18(m,1H),6.96(d,J=8.0Hz,1H),6.84(d,J=8.4Hz,1H),6.60-6.57(m,1H),6.42-6.38(m,1H),5.82-5.79(m,1H),5.06-3.61(m,6H),3.30(s,3H),2.95-2.88(m,2H),1.54-1.50(m,3H),1.28-1.26(m,3H),1.02(m,3H).
Example 284- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -7- (2-chloro-6-fluorophenyl) -6-fluoro-1- (4-isopropyl-2- (methylsulfonyl) pyridin-3-yl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000841
LC-MS:m/z 643(M+H)+1H NMR(400MHz,CDCl3)δ8.66(m,1H),7.87-7.83(m,1H),7.58(d,J=4.8Hz,1H),7.36-7.31(m,1H),7.23-7.21(m,1H),7.03-7.02(m,1H),6.61-6.58(m,1H),6.42-6.38(m,1H),5.82-5.79(m,1H),5.06-3.61(m,6H),3.27-3.12(m,4H),2.92(m,1H),1.53-1.50(m,3H),1.25-1.23(m,3H),1.08(m,3H).
Example 294- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -1- (3-isopropyl-5- (methylsulfonyl) pyrimidin-4-yl) pyridine [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000842
LC-MS:m/z 625(M+H)+1H NMR(400MHz,CDCl3)δ9.25(s,1H),9.06(s,1H),8.58(s,1H),7.90(d,J=9.3Hz,1H),7.32–7.23(m,1H),6.74–6.51(m,3H),6.42(dd,J=16.7,1.6Hz,1H),5.89–5.77(m,1H),5.27–3.45(m,6H),3.17(s,4H),2.83(m,1H),1.56–1.41(m,3H),1.30(m,3H),1.14(t,J=6.5Hz,3H).
Example 29-1 two isomers, example 29A and example 29B, were obtained by chiral separation:
Figure BDA0002861043520000843
example 29A: LC-MS: m/z625(M + H)+
Example 29B: LC-MS: m/z625(M + H)+
Example 30(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -7- (2-chlorophenyl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000851
LC-MS:m/z 624(M+H)+1H NMR(400MHz,CDCl3)δ7.98(dd,J=7.8,1.3Hz,1H),7.84–7.73(m,1H),7.70(d,J=6.8Hz,1H),7.58(t,J=7.9Hz,1H),7.44–7.30(m,2H),7.28–7.15(m,2H),6.61(m,1H),6.41(dd,J=16.8,1.7Hz,1H),5.81(d,J=10.5Hz,1H),5.13–3.47(m,6H),3.08(m,4H),2.80(m,1H),1.50(m,3H),1.24(t,J=6.4Hz,3H),1.01(d,J=6.8Hz,3H).
Example 30-1 two isomers, example 30A and example 30B, were obtained by chiral separation:
Figure BDA0002861043520000852
example 30A: LC-MS: m/z 624(M + H)+1H NMR(400MHz,DMSO)δ8.38(t,J=9.3Hz,1H),7.86(m,2H),7.66(t,J=7.8Hz,1H),7.47(m,3H),7.22(d,J=7.2Hz,1H),6.92–6.74(m,1H),6.22(d,J=16.6Hz,1H),5.77(d,J=10.7Hz,1H),4.96(brs,1H),4.23(m,3H),3.87–3.44(m,2H),3.28–3.07(m,1H),3.01(s,3H),2.71(d,J=6.1Hz,1H),1.30(t,J=14.4Hz,3H),1.11(d,J=6.7Hz,3H),0.94(t,J=28.8Hz,3H).
Example 30B: LC-MS: m/z 624(M + H)+1H NMR(400MHz,DMSO)δ8.37(m,1H),7.86(m,2H),7.66(t,J=7.5Hz,1H),7.47(m,3H),7.22(d,J=7.0Hz,1H),6.96–6.68(m,1H),6.22(d,J=15.4Hz,1H),5.77(d,J=10.3Hz,1H),4.88(brs,1H),4.53–3.94(m,3H),3.76–3.45(m,2H),3.33–2.60(m,5H),1.30(t,J=15.2Hz,3H),1.09(t,J=15.7Hz,3H),0.95(t,J=27.8Hz,3H).
Example 312- (4- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) -2-oxo-1, 2-dihydropyridine [2,3-d ] pyrimidin-7-yl) -3-fluorophenylacetate
Figure BDA0002861043520000853
LC-MS:m/z 666(M+H)+1H NMR(400MHz,CDCl3)δ7.91(d,J=7.3Hz,1H),7.76(s,1H),7.60(d,J=7.4Hz,1H),7.52(t,J=7.6Hz,1H),7.32(m,1H),6.99–6.76(m,2H),6.55(m,1H),6.34m,1H),5.75(m,1H),5.23–3.42(m,6H),3.15(m,4H),2.54(s,1H),1.94(s,3H),1.57–1.30(m,3H),1.07(m,3H),0.85(m,3H).
Example 31-1 two isomers, example 31A and example 31B, were obtained by chiral separation:
Figure BDA0002861043520000861
example 31A: LC-MS: m/z 666(M + H)+1H NMR(400MHz,DMSO)δ8.38(t,J=9.3Hz,1H),7.89(dd,J=7.8,1.3Hz,1H),7.80(dt,J=14.3,7.2Hz,1H),7.66(t,J=7.8Hz,1H),7.56(td,J=8.4,6.6Hz,1H),7.25(t,J=8.7Hz,1H),7.12(d,J=7.8Hz,1H),6.87(m,1H),6.22(d,J=16.7Hz,1H),5.78(d,J=10.6Hz,1H),4.93(brs,1H),4.35(m,2H),4.10(m,1H),3.86–3.46(m,2H),3.24(m,1H),2.99(m,3H),2.60(m,1H),2.13–1.86(m,3H),1.35(d,J=6.5Hz,3H),1.09(d,J=6.6Hz,3H),0.90(t,J=12.5Hz,3H).
Example 31B: LC-MS: m/z 666(M + H)+1H NMR(400MHz,DMSO)δ8.41(t,J=8.8Hz,1H),7.89(dd,J=7.8,1.4Hz,1H),7.82(dt,J=7.9,3.9Hz,1H),7.66(t,J=7.8Hz,1H),7.56(td,J=8.4,6.6Hz,1H),7.25(t,J=8.8Hz,1H),7.12(d,J=8.0Hz,1H),7.00–6.78(m,1H),6.34–6.09(m,1H),5.78(dd,J=10.4,2.3Hz,1H),4.90(brs,1H),4.56–3.94(m,3H),3.63(m,2H),3.31–2.88(m,4H),2.66(m,1H),2.09–1.87(m,3H),1.31(m,3H),1.09(d,J=6.6Hz,3H),0.93(m,3H).
Example 32(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -7- (2-chloro-3-fluorophenyl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000862
LC-MS:m/z 642(M+H)+1H NMR(400MHz,CDCl3)δ8.03–7.91(m,1H),7.80(dd,J=8.6,5.5Hz,1H),7.70(d,J=7.0Hz,1H),7.59(t,J=7.8Hz,1H),7.21(m,2H),7.02(d,J=7.4Hz,1H),6.61(s,1H),6.41(dd,J=16.8,1.6Hz,1H),5.81(d,J=10.5Hz,1H),5.24–3.42(m,6H),3.08(s,4H),2.80(m,1H),1.47(m,3H),1.25(m,3H),1.06–0.93(m,3H).
Example 32-1 two isomers, example 32A and example 32B, were obtained by chiral separation:
Figure BDA0002861043520000871
example 32A: LC-MS: m/z 642(M + H) +1H NMR(400MHz,DMSO)δ8.42(t,J=9.4Hz,1H),7.86(m,2H),7.67(t,J=7.8Hz,1H),7.58–7.23(m,2H),7.12(m,1H),6.91(m,1H),6.22(d,J=16.9Hz,1H),5.78(d,J=11.3Hz,1H),4.97(brs,1H),4.23(m,3H),3.90–3.56(m,2H),3.20(m,1H),3.01(s,3H),2.73(m,1H),1.32(d,J=6.4Hz,3H),1.11(d,J=6.7Hz,3H),0.98(d,J=6.7Hz,3H).
Example 32B: LC-MS: m/z 642(M + H)+1H NMR(400MHz,DMSO)δ8.40(t,J=9.9Hz,1H),7.87(m,2H),7.67(t,J=7.8Hz,1H),7.59–7.38(m,2H),7.10(d,J=7.5Hz,1H),6.87(dd,J=26.4,15.8Hz,1H),6.22(d,J=16.5Hz,1H),5.90–5.66(m,1H),4.87(brs,1H),4.24(m,3H),3.82–3.45(m,2H),3.18(m,1H),3.01(s,3H),2.74(m,1H),1.31(t,J=6.4Hz,3H),1.11(d,J=6.8Hz,3H),0.98(d,J=6.8Hz,3H).
Example 33(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -7- (2-chlorophenyl) -6-fluoro-1- (2-isopropyl-4- (methylsulfonyl) pyridin-3-yl) pyridine [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000872
LC-MS:m/z 625(M+H)+
Example 33-1 two isomers were obtained by chiral separation, example 33A and example 33B:
Figure BDA0002861043520000873
example 33A: LC-MS: m/z 625(M + H)+
Example 33B: LC-MS: m/z 625(M + H)+
Example 34(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -7- (2-chloro-3-fluorophenyl) -6-fluoro-1- (2-isopropyl-4- (methylsulfonyl) pyridin-3-yl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000881
LC-MS:m/z 643(M+H)+
Example 34-1 two isomers, example 34A and example 34B, were obtained by chiral separation:
Figure BDA0002861043520000882
example 34A: LC-MS: m/z 643(M + H)+
Example 34B: LC-MS: m/z 643(M + H)+
Example 35(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -7- (2-chlorophenyl) -1- (2-cyclobutyl-6- (methylsulfonyl) phenyl) -6-fluoropyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000883
LC-MS:m/z 636(M+H)+
Example 35-1 two isomers, example 35A and example 35B, were obtained by chiral separation:
Figure BDA0002861043520000884
example 35A: LC-MS: m/z 636(M + H)+
Example 35B: LC-MS: m/z 636(M + H)+
Example 36(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -7- (2-chloro-3-fluorophenyl) -1- (2-cyclobutyl-6- (methylsulfonyl) phenyl) -6-fluoropyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000891
LC-MS:m/z 654(M+H)+
Example 36-1 two isomers, example 36A and example 36B, were obtained by chiral separation:
Figure BDA0002861043520000892
example 36A: LC-MS: m/z 654(M + H)+
Example 36B: LC-MS: m/z 654(M + H)+
Example 37(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -7- (2-chloropyridin-3-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000893
LC-MS:m/z 625(M+H)+
Example 37-1 two isomers, example 37A and example 37B, were obtained by chiral separation:
Figure BDA0002861043520000901
example 37A: LC-MS: m/z 625(M+H)+
Example 37B: LC-MS: m/z 625(M + H)+
Example 38(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -7- (3-chloropyridin-4-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000902
LC-MS:m/z 625(M+H)+
Example 38-1 two isomers, example 38A and example 38B, were obtained by chiral separation:
Figure BDA0002861043520000903
example 38A: LC-MS: m/z 625(M + H)+
Example 38B: LC-MS: m/z 625(M + H)+
Example 39(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -7- (4-chloropyridin-3-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000904
LC-MS:m/z 625(M+H)+
Example 39-1 two isomers, example 39A and example 39B, were obtained by chiral separation:
Figure BDA0002861043520000911
example 39A: LC-MS: m/z 625(M + H)+
Example 39B: LC-MS: m/z 625(M + H) +
Example 40(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -7- (3-chloropyridin-2-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000912
LC-MS:m/z 625(M+H)+
Example 40-1 two isomers, example 39A and example 39B, were obtained by chiral separation:
Figure BDA0002861043520000913
example 40A: LC-MS: m/z 625(M + H)+
Example 40B: LC-MS: m/z 625(M + H)+
Example 41(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -7- (2-aminopyridin-3-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000914
LC-MS:m/z 606(M+H)+
Example 41-1 two isomers, example 41A and example 41B, were obtained by chiral separation:
Figure BDA0002861043520000921
example 41A: LC-MS: m/z 606(M + H)+
Example 41B: LC-MS: m/z 606(M + H)+
Example 42(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -7- (3-aminopyridin-4-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000922
LC-MS:m/z 606(M+H)+
Example 42-1 two isomers, example 42A and example 42B, were obtained by chiral separation:
Figure BDA0002861043520000923
example 42A: LC-MS: m/z 606(M + H)+
Example 42B: LC-MS: m/z 606(M + H)+
Example 43(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -7- (4-aminopyridin-3-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000931
LC-MS:m/z 606(M+H)+
Example 43-1 two isomers, example 43A and example 43B, were obtained by chiral separation:
Figure BDA0002861043520000932
example 43A: LC-MS: m/z 606(M + H)+
Example 43B: LC-MS: m/z 606(M + H)+
Example 44(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -7- (3-aminopyridin-2-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000933
LC-MS:m/z 606(M+H)+
Example 44-1 two isomers, example 44A and example 44B, were obtained by chiral separation:
Figure BDA0002861043520000934
example 44A: LC-MS: m/z 606(M + H)+
Example 44B: LC-MS: m/z 606(M + H)+
Example 452- (4- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) -2-oxo-1, 2-dihydropyridin [2,3-d ] pyrimidin-7-yl) -3-fluorophenylpropionate
Figure BDA0002861043520000941
LC-MS:m/z 680(M+H)+
Example 45-1 two isomers, example 45A and example 45B, were obtained by chiral separation:
Figure BDA0002861043520000942
example 45A: LC-MS: m/z 680(M + H)+
Example 45B: LC-MS: m/z 680(M + H)+
Example 462- (4- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) -2-oxo-1, 2-dihydropyridin [2,3-d ] pyrimidin-7-yl) -3-fluorophenylisobutyrate
Figure BDA0002861043520000943
LC-MS:m/z 694(M+H)+
Example 46-1 two isomers, example 46A and example 46B, were obtained by chiral separation:
Figure BDA0002861043520000944
example 46A: LC-MS: m/z 694(M + H) +
Example 46B: LC-MS: m/z 694(M + H)+
Example 472- (4- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) -2-oxo-1, 2-dihydropyridine [2,3-d ] pyrimidin-7-yl) -3-fluorophenylmethylcarbamate
Figure BDA0002861043520000951
LC-MS:m/z 681(M+H)+
Example 47-1 two isomers, example 47A and example 47B, were obtained by chiral separation:
Figure BDA0002861043520000952
example 47A: LC-MS: m/z 681(M + H)+
Example 47B: LC-MS: m/z 681(M + H)+
Example 48 methyl (2- (4- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) -2-oxo-1, 2-dihydropyridine [2,3-d ] pyrimidin-7-yl) -3-fluorophenyl) carbamate
Figure BDA0002861043520000953
LC-MS:m/z 681(M+H)+
Example 48-1 two isomers, example 48A and example 48B, were obtained by chiral separation:
Figure BDA0002861043520000954
example 48A: LC-MS: m-z 681(M+H)+
Example 48B: LC-MS: m/z 681(M + H)+
Example 491- (2- (4- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) -2-oxo-1, 2-dihydropyridine [2,3-d ] pyrimidin-7-yl) -3-fluorophenyl) -3-methylurea
Figure BDA0002861043520000961
LC-MS:m/z 680(M+H)+
Example 49-1 gave two isomers by chiral separation, example 49A and example 49B:
Figure BDA0002861043520000962
example 49A: LC-MS: m/z 680(M + H)+
Example 49B: LC-MS: m/z 680(M + H)+
Example 50N- (2- (4- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) -2-oxo-1, 2-dihydropyridine [2,3-d ] pyrimidin-7-yl) -3-fluorophenyl) -methanesulfonamide
Figure BDA0002861043520000963
LC-MS:m/z 701(M+H)+
Example 50-1 two isomers, example 50A and example 50B, were obtained by chiral separation:
Figure BDA0002861043520000964
example 50A: LC-MS: m/z 701(M + H)+
Example 50B: LC-MS: m/z 701(M + H)+
Example 514 preparation of- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -1- (2- (methylsulfonyl) phenyl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000971
Step 1 preparation of 2, 6-dichloro-5-fluoro-N- (((2- (methylsulfonyl) phenyl) carbamoyl) nicotinamide
2, 6-dichloro-5-fluoronicotinamide (420mg, 2.0mmol) was dissolved in anhydrous tetrahydrofuran (7mL), and a solution of oxalyl chloride (1.7mL, 20.0mmol) in dichloromethane (2mL) was slowly added dropwise to this solution. After the addition was complete, the mixture was stirred under reflux at 75 ℃ for 2h and then concentrated to dryness under reduced pressure. The residue was diluted with anhydrous tetrahydrofuran (7mL) and cooled to 0 ℃. 2- (methylsulfonyl) aniline (360mg, 2.1mmol) was dissolved in anhydrous tetrahydrofuran (3mL) and added dropwise to the solution. The reaction mixture was stirred at 0 ℃ for 2h, quenched with saturated ammonium chloride/saturated brine (V/V-1/1, 20mL), and extracted 3 times with dichloromethane/methanol (V/V-10/1, 20 mL). The combined organic phases were dried, concentrated, and the residual solid was slurried with petroleum ether/ethyl acetate (V/V. 3/1, 15mL), suction-filtered, and dried to give the desired product (645mg, yield: 79%).
LC-MS:m/z 406(M+H)+
Step 2 preparation of 7-chloro-6-fluoro-1- (2- (methylsulfonyl) phenyl) pyrido [2,3-d ] pyrimidine-2, 4(1H, 3H) -dione
Suspending 2, 6-dichloro-5-fluoro-N- (((2- (methylsulfonyl) phenyl) carbamoyl) nicotinamide (645mg, 1.6mmol) in tetrahydrofuran (15mL), dropwise adding potassium bis (trimethylsilyl) amide (1 mol in tetrahydrofuran, 3.6mL, 3.6mmol) under ice bath, clarifying the reaction solution after completion of dropwise addition, stirring the reaction solution at room temperature for 16h, quenching with saturated ammonium chloride (20mL), extracting with ethyl acetate (20mL) 3 times, combining ethyl acetate layers, drying, concentrating, slurrying the residual solid with petroleum ether/ethyl acetate (V/V ═ 3/1, 10mL), suction-filtering, and drying to obtain the desired product (500mg, yield: 85%).
LC-MS:m/z 370(M+H)+1H NMR(400MHz,DMSO-d6)δ12.32(s,1H),8.50(d,J=7.2Hz,1H),8.15(dd,J=8.0Hz,1.2Hz,1H),7.97-7.92(m,1H),7.86-7.81(m,1H),7.66(dd,J=8.0Hz,1.2Hz,1H),3.09(s,3H).
Preparation of (S) -tert-butyl 4- (7-chloro-6-fluoro-1- (2- (methylsulfonyl) phenyl) -2-oxo-1, 2-dihydropyrido [2,3-d ] pyrimidin-4-yl) -3-methylpiperazine-1-carboxylic acid
7-chloro-6-fluoro-1- (2- (methylsulfonyl) phenyl) pyrido [2,3-d ] pyrimidine-2, 4(1H, 3H) -dione (450mg, 1.2mmol) was suspended in acetonitrile (10mL), N-diisopropylethylamine (1.2mL, 7.3mmol) and phosphorus oxychloride (0.6mL, 6.1mmol) were added dropwise, and the reaction solution was clarified. The reaction mixture was stirred at 80 ℃ for 4h and concentrated to dryness under reduced pressure. The residue was dissolved in acetonitrile (10mL), cooled to 0 ℃, N-diisopropylethylamine (0.6mL, 3.7mmol) and tert-butyl (S) -3-methylpiperazine-1-carboxylate (290mg, 1.5mmol) were added, the reaction was stirred at room temperature for 1h, quenched with half-saturated sodium bicarbonate solution (40mL) and extracted 3 times with ethyl acetate (30 mL). The combined ethyl acetate layers were dried, concentrated, and purified with a silica gel column (petroleum ether/ethyl acetate: 3/1 to 1/2.5) to obtain the objective product (460mg, yield: 68%).
LC-MS:m/z 552(M+H)+
Preparation of (S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -7-chloro-6-fluoro-1- (2- (methylsulfonyl) phenyl) pyridin [2,3-d ] pyrimidin-2 (1H) -one in step 4
(S) -tert-butyl 4- (7-chloro-6-fluoro-1- (2- (methylsulfonyl) phenyl) -2-oxo-1, 2-dihydropyrido [2,3-d ] pyrimidin-4-yl) -3-methylpiperazine-1-carboxylic acid (500mg, 0.9mmol) was dissolved in dichloromethane (10mL), trifluoroacetic acid (2mL) was added, the reaction solution was stirred for 2h at room temperature, concentrated to dryness, and the residue was co-evaporated with dichloromethane (15mL) 3 times to give the crude product. The crude product was dissolved in dichloromethane (8mL), cooled to 0 deg.C, and a solution of N, N-diisopropylethylamine (0.6mL, 3.6mmol) and acryloyl chloride (110mg, 1.2mmol) in dichloromethane (1mL) was added dropwise. The reaction mixture was stirred at 0 ℃ for 30min, quenched with saturated sodium bicarbonate (30mL), and extracted with dichloromethane (20mL) 3 times, the dichloromethane layers were combined, dried, concentrated, and the residue was purified with silica gel column (dichloromethane/methanol: 60/1) to give the objective product (380mg, yield: 83%).
LC-MS:m/z 506(M+H)+
Step 5 preparation of 4- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -1- (2- (methylsulfonyl) phenyl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -7-chloro-6-fluoro-1- (2- (methylsulfonyl) phenyl) pyridine [2,3-d ] pyrimidin-2 (1H) -one (150mg, 0.3mmol), (2-fluoro-6-hydroxyphenyl) boronic acid (60mg, 0.4mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane complex (24mg, 0.03mmol) and potassium acetate (120mg, 1.2mmol) were suspended in a mixed solvent of dioxane/water (7.5mL/0.75mL), displaced with nitrogen 3 times, and stirred at 90 ℃ for 2H. After the reaction solution was cooled to room temperature, a half-saturated sodium bicarbonate solution (20mL) was added, and extracted 3 times with ethyl acetate (20 mL). The ethyl acetate layers were combined, dried, concentrated, and the residue was purified with a silica gel column (dichloromethane/methanol: 100/1 to 60/1) to obtain the objective product (90mg, yield: 52%).
LC-MS:m/z 582(M+H)+1H NMR(400MHz,CDCl3)δ8.97-8.94(m,1H),8.27(d,J=8.0Hz,1H),7.90-7.84(m,2H),7.79-7.74(m,1H),7.46-7.42(m,1H),7.29-7.23(m,1H),6.71-6.57(m,3H),6.44-6.38(m,1H),5.82(d,J=11.2Hz,1H),5.12-4.32(m,3H),4.07-3.63(m,3H),3.24-3.01(m,4H),1.50(s,3H).
The following compounds were synthesized according to the procedure of example 51, starting from different starting materials:
example 524- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -1- (2- (isopropylsulfonyl) phenyl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000991
LC-MS:m/z 610(M+H)+1H NMR(400MHz,CDCl3)δ8.91-8.86(m,1H),8.22-8.19(m,1H),7.88-7.82(m,2H),7.75-7.72(m,1H),7.45-7.42(m,1H),7.28-7.23(m,1H),6.71-6.63(m,3H),6.44-6.39(m,1H),5.83-5.80(m,1H),5.04-4.79(m,3H),4.56-3.90(m,4H),3.71-3.48(m,1H),1.50(s,3H),1.33-1.30(m,3H),1.18-1.16(m,3H).
Example 534- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -1- (2-methyl-6- (methylsulfonyl) phenyl) pyridine [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520000992
LC-MS:m/z 596(M+H)+1H NMR(400MHz,CDCl3)δ8.12-8.11(m,1H),7.89-7.87(m,1H),7.75-7.63(m,2H),7.26(m,1H),6.69-6.64(m 3H),6.44-6.40(m,1H),5.84-5.81(m,1H),5.00-4.40(m,3H),4.07-3.65(m,3H),3.25-3.16(m,4H),2.20-2.18(m,3H),1.58-1.50(m,3H).
Example 53-1 two isomers, example 53A and example 53B, were obtained by chiral separation:
Figure BDA0002861043520000993
example 53A: LC-MS: m/z 596(M + H)+
Example 53B: LC-MS: m/z 596(M + H)+
Example 544- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -1- (2- (methylsulfonyl) pyridin-3-yl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520001001
LC-MS:m/z 583(M+H)+1H NMR(400MHz,CDCl3)δ8.80(dd,J=4.8,1.2Hz,1H),8.37-8.25(m,1H),8.05(dd,J=8.0,1.2Hz,1H),7.90-7.87(m,1H),7.26(dd,J=14.6,7.6Hz,1H),6.92(m,1H),6.82-6.65(m,2H),6.23-6.18(m,1H),5.76(dd,J=6.4,2.4Hz,1H),4.98-4.82(m,1H),4.44-4.00(m,3H),3.81-3.62(m,2H),3.24-3.00(m,4H),1.34-1.29(m,3H).
Example 554- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -1- (4- (methylsulfonyl) pyridin-3-yl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520001002
LC-MS:m/z 583(M+H)+1H NMR(400MHz,CDCl3)δ9.06(d,J=4.2Hz),8.75-8.74(m,1H),8.59-8.57(m,1H),8.10(d,J=4.2Hz),7.87(d,J=11.2Hz,1H),7.30-7.25(m,1H),6.72-6.57(m,3H),6.64-6.58(m,1H),5.84-5.82(m,1H),5.29-4.28(m,3H),4.10-3.61(m,3H),3.22-2.92(m,4H0,1.59-1.48(m,3H).
Example 562- (1-acryloyl-4- (6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -1- (2- (methylsulfonyl) phenyl) -2-oxo-1, 2-dihydropyridin [2,3-d ] pyrimidin-4-yl) piperazin-2-yl) acetonitrile
Figure BDA0002861043520001003
LC-MS:m/z 607(M+H)+1H NMR(400MHz,CDCl3)δ8.29-8.24(m,1H),7.96-7.76(m,3H),7.47-7.43(m,1H),7.29(m,1H),6.70-6.56(m,3H),6.45-6.41(m,1H),5.00(brs,1H),4.56-3.70(m,6H),3.16(m,3H),3.16-3.10(m,1H),2.98-2.79(m,1H).
Example 572- (1-acryloyl-4- (6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -1- (2-methyl-6- (methylsulfonyl) phenyl) -2-oxo-1, 2-dihydropyridin [2,3-d ] pyrimidin-4-yl) piperazin-2-yl) acetonitrile
Figure BDA0002861043520001011
LC-MS:m/z 621(M+H)+1H NMR(400MHz,CDCl3)δ8.12-7.97(m,2H),7.76-7.72(m,1H),7.67-7.64(m,1H),7.29(m,1H),6.69-6.46(m,3H),6.42-6.41(m,1H),5.88-5.86(m,1H),5.00(brs,1H),4.54-3.77(m,6H),3.12(m,4H),2.81-2.77(m,1H),2.20-2.17(m,3H).
Example 582- (1-acryloyl-4- (6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -1- (2- (methylsulfonyl) pyridin-3-yl) -2-oxo-1, 2-dihydropyridin [2,3-d ] pyrimidin-4-yl) piperazin-2-yl) acetonitrile
Figure BDA0002861043520001012
LC-MS:m/z 608(M+H)+1H NMR(400MHz,CDCl3)δ8.82-8.81(m,1H),7.99-7.76(m,3H),7.30(m,1H),6.70-6.56(m,3H),6.44-6.40(m,1H),5.87-5.84(m,1H),5.01(brs,1H),4.61-3.60(m,6H),3.29(m,3H),2.96-2.76(m,2H).
Example 594- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -7- (2-chloro-6-fluorophenyl) -6-fluoro-1- (2-methyl-6- (methylsulfonyl) phenyl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520001013
LC-MS:m/z 614(M+H)+1H NMR(400MHz,CDCl3)δ7.95(d,J=3.6Hz,1H),7.82(t,J=8.4Hz,1H),7.60(d,J=3.2Hz,1H),7.50(t,J=8.4Hz,1H),7.34-7.29(m,1H),7.20(d,J=8Hz,1H),7.02(m,1H),6.60(m,1H),6.39(dd,J=1.6Hz,17.2Hz,1H),5.80(dd,J=1.2Hz,10.4Hz,1H),5.20-4.20(m,3H),4.10-3.55(m,3H),3.40-3.05(m,4H),2.17-2.15(m,3H),1,.51(m,3H).
Example 602- (4-acryloyl-1- (6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -1- (2- (methylsulfonyl) phenyl) -2-oxo-1, 2-dihydropyrido [2,3-d ] pyrimidin-4-yl) piperazin-2-yl) acetonitrile
Figure BDA0002861043520001021
LC-MS:m/z 607(M+H)+1H NMR(400MHz,CDCl3)δ8.87-8.77(m,1H),8.22-8.19(m,1H),7.86-7.80(m,3H),7.69(m,1H),7.37-7.36(m,1H),7.25(m,1H),6.66-6.53(m,3H),6.39(d,J=17.2Hz,1H),5.81(d,J=10Hz,1H),5.30-5.20(m,1H),4.80-3.95(m,3H),3.90-3.30(m,3H),3.09-2.88(m,5H).
Example 612- (4-acryloyl-1- (6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -1- (2- (methylsulfonyl) phenyl) -2-oxo-1, 2-dihydropyrido [2,3-d ] pyrimidin-4-yl) piperazin-2-yl) acetamide
Figure BDA0002861043520001022
LC-MS:m/z 625(M+H)+1H NMR(400MHz,CDCl3)δ8.20-7.75(m,2H),7.60-7.40(m,2H),7.11-7.01(m,2H),6.84-7.53(m,5H),6.36-6.29(m,1H),5.79(m,1H),5.44-5.23(m,1H),5.50-3.98(m,4H),3.81-3.070(m,6H),2.85-2.63(m,2H).
Example 622- (4- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) -2-oxo-1, 2-dihydropyridin [2,3-d ] pyrimidin-7-yl) -3-fluorophenyl cyclopropane
Figure BDA0002861043520001023
LC-MS:m/z 692(M+H)+
Example 62-1 two isomers, example 62A and example 62B, were obtained by chiral separation:
Figure BDA0002861043520001031
example 62A: LC-MS: m/z 692(M + H)+
Example 62B: LC-MS: m/z 692(M + H)+
Example 634 preparation of- ((2S, 5R) -4-acryloyl-2, 5-dimethylpiperazin-1-yl) -1- (2-cyclobutyl-6- (methylsulfonyl) phenyl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) pyrido [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520001032
Step 1 preparation of 2-bromo-6- (methylsulfonyl) aniline
1-bromo-3- (methylsulfonyl) -2-nitrobenzene (44g, 158mmol), iron powder (44g, 788mmol) and ammonium chloride (83g, 1.6mol) were suspended in a mixed solvent of ethanol/water (600mL/120mL), and stirred at 75 ℃ for 2 hours. After the reaction solution was cooled to room temperature, the solid was removed by suction filtration, and 500 ml of water was added to the filtrate, followed by extraction with 500 ml of ethyl acetate 2 times. The ethyl acetate layers were combined, washed with 500 ml of brine, dried, concentrated, and the residue was separated on a silica gel column (petroleum ether: ethyl acetate: 10: 1 to 2: 1) to obtain the objective product (30g, yield: 77%).
Step 2 preparation of 2-cyclobutyl-6- (methylsulfonyl) aniline
Zinc powder (52g,800mmol, Acros) was suspended in tetrahydrofuran (100mL) solvent, followed by addition of trimethylchlorosilane (8.7g,80mmol) and stirring with heating at 75 ℃ for half an hour. Then, bromocyclobutane (54g,400mmol) was added, the reaction solution was cooled to room temperature, 2-bromo-6- (methylsulfonyl) aniline (10g,40mmol) and [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium dichloromethane complex (3.3g, 4mmol) were added, and the mixture was stirred at 75 ℃ for three hours. The reaction solution was quenched with 30 ml of 1M phosphoric acid and extracted 2 times with ethyl acetate. The ethyl acetate layers were combined, dried, concentrated, and the residue was separated with a silica gel column (petroleum ether: ethyl acetate: 10: 1 to 2: 1) to obtain the objective product (8.1g, yield: 90%).
LC-MS:m/z 226(M+H)+
Step 3 preparation of 2, 6-dichloro-N- (((2-cyclobutyl-6- (methylsulfonyl) phenyl) carbamoyl) -5-fluoronicotinamide
2, 6-dichloro-5-fluoronicotinamide (7.7g, 37mmol) was dissolved in 100ml of anhydrous tetrahydrofuran, to which oxalyl chloride (47g, 370mmol) was slowly added dropwise. After the addition was complete, the mixture was stirred at 75 ℃ under reflux for 2 hours and then concentrated to dryness under reduced pressure. The residue was diluted with 100ml of anhydrous tetrahydrofuran and cooled to zero degrees. 2-cyclobutyl-6- (methylsulfonyl) aniline (8.8g, 39mmol) was dissolved in 50ml of anhydrous tetrahydrofuran and added dropwise to the solution. The reaction was stirred at zero degrees for 2 hours, quenched with saturated ammonium chloride/saturated brine (1: 1, 100mL), and extracted 2 times with ethyl acetate (50 mL). The combined organic phases were dried, concentrated, and the residual solid was slurried with petroleum ether/ethyl acetate (5: 1, 200mL), suction filtered, and dried to give the desired product (12.7g, yield: 75%).
LC-MS:m/z 460(M+H)+
Step 4 preparation of 7-chloro-1- (2-cyclobutyl-6- (methylsulfonyl) phenyl) -6-fluoropyrido [2,3-d ] pyrimidine-2, 4(1H, 3H) -dione
Dissolving 2, 6-dichloro-N- (((2-cyclobutyl-6- (methylsulfonyl) phenyl) carbamoyl) -5-fluoronicotinamide 5(37.3g, 81.1mmol) in 550 mL tetrahydrofuran, dropwise adding bis (trimethylsilyl) amino potassium (1mol/L, 186.5mL, 186.5mmol) in ice bath, stirring the reaction solution at 25 ℃ for 16 hours after the dropwise addition, quenching with 200mL saturated ammonium chloride, extracting with 1000 mL ethyl acetate for 2 times, combining ethyl acetate layers, drying, concentrating, pulping the residual solid with petroleum ether/ethyl acetate (1: 1, 300mL), filtering with suction, and drying to obtain the target product (23.2g, yield: 68%).
LC-MS:m/z 424(M+H)+
Preparation of (2R, 5S) -tert-butyl 4- (7-chloro-1- (2-cyclobutyl-6- (methylsulfonyl) phenyl) -6-fluoro-2-oxo-1, 2-dihydropyrido [2,3-d ] pyrimidin-4-yl) -2, 5-dimethylpiperazine-1-carboxylate in step 5
7-chloro-1- (2-cyclobutyl-6- (methylsulfonyl) phenyl) -6-fluoropyrido [2,3-d ] pyrimidine-2, 4(1H, 3H) -dione (23.2g, 54.7mmol) was suspended in 350 ml acetonitrile, N-diisopropylethylamine (42.3g, 328mmol) and phosphorus oxychloride (33.5g, 219mmol) were added dropwise and the reaction solution was clarified. The reaction mixture was stirred at 80 ℃ for 1 hour, and concentrated to dryness under reduced pressure. The residue was dissolved in 350 ml acetonitrile, cooled to zero and N, N-diisopropylethylamine (42.3g, 328mmol) and 2, 5-dimethylpiperazine-1-carboxylic acid (2R, 5S) -tert-butyl ester (14g, 65.6mmol) were added. The reaction was stirred at room temperature for 1 hour, quenched with half-saturated sodium bicarbonate solution (400mL), and extracted 2 times with 1000 mL of ethyl acetate. The combined ethyl acetate layers were dried, concentrated, and subjected to silica gel column separation (petroleum ether: ethyl acetate: 5: 1 to 1: 1) to obtain the objective product (21.2g, yield: 63%).
LC-MS:m/z 620(M+H)+
Step 6 preparation of 4- ((2S, 5R) -4-acryloyl-2, 5-dimethylpiperazin-1-yl) -7-chloro-1- (2-cyclobutyl-6- (methylsulfonyl) phenyl) -6-fluoropyridin [2,3-d ] pyrimidin-2 (1H) -one
(2R, 5S) -tert-butyl 4- (7-chloro-1- (2-cyclobutyl-6- (methylsulfonyl) phenyl) -6-fluoro-2-oxo-1, 2-dihydropyrido [2,3-d ] pyrimidin-4-yl) -2, 5-dimethylpiperazine-1-carboxylate (21.2g, 34.2mmol) was dissolved in 200ml of dichloromethane, 65 ml of trifluoroacetic acid was added, the reaction solution was stirred for 2 hours at room temperature, concentrated to dryness, and the residue was co-evaporated with 100 ml of dichloromethane 2 times to give the crude product. The crude product was dissolved in 200mL of dichloromethane, cooled to zero degrees, and a solution of triethylamine (17.3g, 171mmol) and acryloyl chloride (4g, 44.5mmol) in dichloromethane (20mL) was added dropwise. The reaction mixture was stirred at zero degrees for 30 minutes and at room temperature for 30 minutes. 200ml of saturated sodium bicarbonate are quenched and extracted 2 times with 800 ml of dichloromethane. The organic phase was dried and concentrated, and the residue was separated with a silica gel column (dichloromethane: methanol: 60: 1) to obtain the objective product (13.6g, yield: 69%).
LC-MS:m/z 574(M+H)+
Step 7 preparation of 4- ((2S, 5R) -4-acryloyl-2, 5-dimethylpiperazin-1-yl) -1- (2-cyclobutyl-6- (methylsulfonyl) phenyl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) pyrido [2,3-d ] pyrimidin-2 (1H) -one
4- ((2S, 5R) -4-acryloyl-2, 5-dimethylpiperazin-1-yl) -7-chloro-1- (2-cyclobutyl-6- (methylsulfonyl) phenyl) -6-fluoropyridin [2,3-d ] pyrimidin-2 (1H) -one (13.6g, 23.7mmol), (2-fluoro-6-hydroxyphenyl) boronic acid (5.2g, 33.1mmol), [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride dichloromethane complex (1.94g, 2.4mmol) and potassium acetate (9.3g, 95mmol) were suspended in a mixed solvent of dioxane/water (200mL/20mL), displaced with nitrogen 3 times, and stirred at 90 ℃ for 4 hours. After the reaction solution was cooled to room temperature, 200ml of a half-saturated sodium bicarbonate solution was added, and extracted 2 times with 500 ml of ethyl acetate. The ethyl acetate layers were combined, dried, concentrated, and the residue was separated with a silica gel column (dichloromethane: methanol 100: 1 to 50: 1) to obtain the objective product (8.6g, yield: 56%).
LC-MS:m/z 650(M+H)+1H NMR(400MHz,CDCl3)δ9.07-8.93(m,1H),8.14-8.09(m,1H),7.90-7.67(m,3H),7.29-7.23(m,1H),6.71-6.52(m,3H),6.40(m,1H),5.80(m,1H),5.23-3.30(m,6H),3.15-3.08(m,3H),2.33-2.20(m,1H),2.17-2.05(m,1H),1.96-1.65(m,3H),1.60-1.32(m,8H).
Example 63-1 two isomers, examples 63A and 63B, were obtained by chiral separation:
Figure BDA0002861043520001051
compound 63A
LC-MS:m/z 650(M+H)+1H NMR(400MHz,DMSO)δ10.21(s,1H),8.32(d,J=9.1Hz,1H),7.88(d,J=7.7Hz,1H),7.75–7.59(m,2H),7.27(m,1H),6.93–6.60(m,3H),6.19(m,1H),5.74(m,1H),4.93–4.38(m,2H),4.23–3.40(m,3H),3.30–3.23(m,1H),2.99(s,3H),2.18–1.87(m,3H),1.85–1.52(m,3H),1.38–1.10(m,7H).
Compound 63B
LC-MS:m/z 650(M+H)+1H NMR(400MHz,DMSO)δ10.23(s,1H),8.34(m,1H),7.89(d,J=7.7Hz,1H),7.77(m,1H),7.66(t,J=7.8Hz,1H),7.27(m,1H),6.95–6.60(m,3H),6.19(m,1H),5.75(m,1H),4.93–4.38(m,2H),4.18–3.45(m,3H),3.28(m,1H),2.96(s,3H),2.15–1.86(m,3H),1.86–1.56(m,3H),1.35–0.98(m,7H).
The following compounds were synthesized according to the procedure of example 63, starting from different starting materials:
example 64(S) -3- (4- (4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) -2-oxo-1, 2-dihydropyridine [2,3-d ] pyrimidin-7-yl) -4-chlorobenzonitrile
Figure BDA0002861043520001061
LC-MS:m/z 649(M+H)+1H NMR(400MHz,CDCl3)δ7.99(d,J=7.6Hz,1H),7.84(dd,J=8.8Hz,6.4Hz,1H),7.72(d,J=7.6Hz,1H),7.64-7.60(m,2H),7.53-7.48(m,2H),6.61(s,1H),6.40(d,J=16.8Hz,1H),5.80(d,J=12.0Hz,1H),5.06-3.62(m,6H),3.33-3.01(m,4H),2.80(s,1H),1.51(s,3H),1.24(d,J=6.8Hz,3H),1.03(d,J=6.8Hz,1H).
Example 65(S) -3- (4- (4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) -2-oxo-1, 2-dihydropyrido [2,3-d ] pyrimidin-7-yl) -2-chlorobenzonitrile
Figure BDA0002861043520001062
LC-MS:m/z 649(M+H)+1H NMR(400MHz,CDCl3)δ8.34(d,J=2.4Hz,1H),8.07-7.98(m,2H),7.91-7.63(m,5H),7.43(t,J=7.6Hz,1H),7.03-6.81(m,1H),5.10-3.51(m,6H),3.44-3.14(m,1H),3.06(s,3H),2.78-2.58(m,1H),1.56-1.45(m,3H),1.26-1.18(m,3H),1.13-1.05(m,3H).
Example 66(S) -4- (4- (4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) -2-oxo-1, 2-dihydropyrido [2,3-d ] pyrimidin-7-yl) -3-chlorobenzonitrile
Figure BDA0002861043520001071
LC-MS:m/z 649(M+H)+1H NMR(400MHz,CDCl3)δ8.34(d,J=2.0Hz,1H),8.07-7.99(m,2H),7.82-7.77(m,1H),7.76-7.64(m,4H),7.59(d,J=8.4Hz,1H),7.05-6.84(m,1H),5.10-3.12(m,7H),3.06(s,3H),2.79-2.58(m,1H),1.56-1.41(m,3H),1.26-1.18(m,3H),1.13-1.05(m,3H).
Example 672- (4- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) -2-oxo-1, 2-dihydropyrido [2,3-d ] pyrimidin-7-yl) -3-chlorobenzonitrile
Figure BDA0002861043520001072
LC-MS:m/z 649(M+H)+1H NMR(400MHz,CDCl3)δ8.34(d,J=2.4Hz,1H),8.03(d,J=8.0Hz,1H),7.94(d,J=15.6Hz,1H),7.79(d,J=7.2Hz,1H),7.76-7.63(m,4H),7.41(t,J=8.0Hz,1H),7.25-7.16(m,1H),5.01-3.60(m,6H),3.38-3.10(m,1H),3.07(s,3H),2.78-2.62(m,1H),1.56-1.46(m,3H),1.26-1.18(m,3H),1.13-1.05(m,3H).
Example 68(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) -7- (1-methyl-1H-pyrazol-5-yl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520001073
LC-MS:m/z 594(M+H)+1H NMR(400MHz,CDCl3)δ8.05(d,J=7.8Hz,1H),7.82-7.78(m,2H),7.73(t,J=7.8Hz,1H),7.47(d,J=1.8Hz,1H),6.94(s,1H),6.59(d,J=17.9Hz,1H),6.42(dd,J=16.7,1.6Hz,1H),5.81(d,J=10.4Hz,1H),5.01-3.67(m,7H),3.61(s,3H),3.19(s,3H),2.75(s,1H),1.52(s,3H),1.28-1.23(m,3H),1.04(d,J=5.8Hz,3H).
Example 69(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) -7- (1H-pyrazol-5-yl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520001081
LC-MS:m/z 580(M+H)+。H NMR(400MHz,CDCl3)δ8.07(d,J=7.8Hz,1H),7.86-7.77(m,2H),7.73(t,J=7.8Hz,1H),7.61(d,J=1.8Hz,1H),6.83(s,1H),6.58(d,J=17.9Hz,1H),6.40(dd,J=16.7Hz,1.6Hz,1H),5.81(d,J=10.4Hz,1H),5.01-3.67(m,6H),3.09(s,4H),2.75(s,1H),1.52(s,3H),1.28-1.23(m,3H),1.04(d,J=5.8Hz,3H).
Example 70(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) -7- (1-methyl-1H-imidazol-5-yl) pyrido [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520001082
LC-MS:m/z 594(M+H)+1H NMR(400MHz,CDCl3)δ8.34(d,J=2.4Hz,1H),8.03(d,J=8.0Hz,1H),7.79(d,J=7.6Hz,1H),7.76-7.60(m,3H),7.57-7.45(m,2H),6.86-6.63(m,1H),5.06-3.12(m,10H),3.07(s,3H),2.78-2.59(m,1H),1.56-1.41(m,3H),1.26-1.18(m,3H),1.13-1.05(m,3H).
Example 71(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) -7- (2-oxo-1, 2-dihydropyridin-3-yl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520001083
LC-MS:m/z 607(M+H)+1H NMR(400MHz,CDCl3)δ11.34(s,1H),8.33(d,J=2.8Hz,1H),8.06-7.88(m,2H),7.79(d,J=7.6Hz,1H),7.76-7.60(m,3H),7.56(d,J=15.2Hz,1H),7.37(d,J=6.0Hz,1H),6.39(t,J=6.4Hz,1H),5.01-3.52(m,7H),3.07(s,3H),2.78-2.62(m,1H),1.56-1.46(m,3H),1.26-1.18(m,3H),1.13-1.05(m,3H).
Example 72(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) -7- (6-oxo-1, 6-dihydropyridin-2-yl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520001091
LC-MS:m/z 607(M+H)+1H NMR(400MHz,CDCl3)δ13.26(brs,1H),8.35(d,J=2.4Hz,1H),8.04(d,J=8.0Hz,1H),7.87-7.66(m,4H),7.56-7.52(m,1H),7.42-7.37(m,1H),6.52(d,J=9.2Hz,1H),6.46(d,J=6.8Hz,1H),5.14-4.24(m,4H),3.80-3.66(m,2H),3.33-3.08(m,4H),2.73-2.68(m,1H),1.56-1.50(m,3H),1.26-1.18(m,3H),1.13-1.05(m,3H).
Example 73(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) -7- (1-methyl-2-oxo-1, 2-dihydropyridin-3-yl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520001092
LC-MS:m/z 621(M+H)+.1H NMR(400MHz,CDCl3)δ8.33(d,J=2.4Hz,1H),8.11-7.94(m,2H),7.90(d,J=8.0Hz,1H),7.75-7.63(m,2H),7.59-7.51(m,2H),7.38(d,J=6.4Hz,1H),6.30(t,J=6.8Hz,1H),4.99-4.02(m,4H),3.77-3.48(m,5H),3.31-3.10(m,1H),3.07(s,3H),2.78-2.62(m,1H),1.55-1.43(m,3H),1.26-1.18(m,3H),1.13-1.05(m,3H).
Example 74(S) -4- (4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) -7- (1-methyl-6-oxo-1, 6-dihydropyridin-2-yl) pyridin [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520001093
LC-MS:m/z 621(M+H)+.1H NMR(400MHz,CDCl3)δ8.35(d,J=2.4Hz,1H),8.03(d,J=7.6Hz,1H),7.80(d,J=7.2Hz,1H),7.76-7.65(m,3H),7.31(t,J=8.0Hz,1H),6.94-6.74(m,1H),6.63(d,J=9.2Hz,1H),6.46-6.35(m,1H),5.10-3.64(m,6H),3.60(s,3H),3.45-3.09(m,1H),3.06(s,3H),2.78-2.62(m,1H),1.59-1.43(m,3H),1.23(d,J=6.8Hz,3H),1.10(d,J=5.6Hz,3H).
Example 754- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -7- (6-chloro-1H-indazol-7-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) pyrido [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520001101
LC-MS:m/z 664(M+H)+.1H NMR(400MHz,CDCl3)δ12.99(brs,1H),8.57-8.20(m,3H),8.02(d,J=7.2Hz,1H),7.86-7.72(m,2H),7.67(t,J=8.0Hz,1H),7.59(d,J=7.6Hz,1H),7.23-7.03(m,2H),5.19-3.24(m,7H),3.06(s,3H),2.78-2.58(m,1H),1.60-1.42(m,3H),1.30-1.15(m,3H),1.14-0.99(m,3H).
Example 764- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -7- (1, 4-dimethyl-1H-imidazol-5-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) pyrido [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520001102
LC-MS:m/z 608(M+H)+.1H NMR(400MHz,CDCl3)δ8.34(d,J=2.4Hz,1H),8.03(d,J=8.0Hz,1H),7.79(d,J=6.8Hz,1H),7.76-7.64(m,3H),7.43(s,1H),6.61-6.44(m,1H),5.10-3.37(m,10H),3.07(s,3H),2.78-2.59(m,1H),2.42(s,3H),1.56-1.41(m,3H),1.28-1.18(m,3H),1.16-1.02(m,3H).
Example 772- (4- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-1- (2-isopropyl-6- (methylsulfonyl) phenyl) -2-oxo-1, 2-dihydropyridine [2,3-d ] pyrimidin-7-yl) -3-fluorobenzoic acid methyl ester
Figure BDA0002861043520001103
LC-MS:m/z 666(M+H)+.1H NMR(400MHz,CDCl3)δ8.34(d,J=2.4Hz,1H),8.04-7.96(m,2H),7.80-7.65(m,4H),7.41-7.36(m,1H),7.33-7.26(m,1H),7.00-6.87(m,1H),4.97-4.14(m,4H),3.94(s,3H),3.76-3.65(m,2H),3.30-3.07(m,4H),2.71(s,1H),1.55-1.49(m,3H),1.29-1.23(m,3H),1.11-1.08(m,3H).
Example 784- ((2S, 5R) -4-acryloyl-2, 5-dimethylpiperazin-1-yl) -1- (2-isopropyl-6- (methylsulfonyl) phenyl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) pyrido [2,3-d ] pyrimidin-2 (1H) -one preparation
Figure BDA0002861043520001111
LC-MS:m/z 638(M+H)+.
Example 78-1 two isomeric examples 78A and 78B were obtained by chiral separation:
Figure BDA0002861043520001112
example 78A
LC-MS:m/z 638(M+H)+.1H NMR(400MHz,DMSO)δ10.21(brs,1H),8.28(m,1H),7.85(m,2H),7.65(t,J=7.8Hz,1H),7.26(m,1H),6.95–6.58(m,3H),6.19(m,1H),5.82–5.69(m,1H),4.96–4.41(m,2H),4.30–3.38(m,4H),2.95(s,3H),2.72–2.55(m,1H),1.40–0.93(m,12H).
Example 78B
LC-MS:m/z 638(M+H)+.1H NMR(400MHz,DMSO)δ10.28(brs,1H),8.51–8.33(m,1H),8.00–7.81(m,2H),7.70(t,J=7.8Hz,1H),7.31(m,1H),7.00–6.65(m,3H),6.24(m,1H),5.80(m,1H),5.02–4.46(m,2H),4.27–3.57(m,4H),2.97(s,3H),2.70–2.58(m,1H),1.43–0.97(m,12H).
Example 794- ((2S, 5R) -4-acryloyl-2, 5-dimethylpiperazin-1-yl) -6-chloro-7- (2-fluoro-6-hydroxyphenyl) -1- (2-isopropyl-6- (methylsulfonyl) phenyl) pyrido [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520001113
LC-MS:m/z 654(M+H)+.
Example 79-1 two isomers, examples 79A and 79B, were obtained by chiral separation:
Figure BDA0002861043520001121
example 79A
LC-MS:m/z 638(M+H)+.1H NMR(400MHz,CDCl3)δ8.11(t,J=7.0Hz,2H),7.78(d,J=6.8Hz,1H),7.68(t,J=7.8Hz,1H),7.26–7.20(m,1H),6.78–6.49(m,3H),6.40(m,1H),5.81(m,1H),5.11(m,2H),4.53–4.19(m,1H),4.17–3.80(m,2H),3.61(m,1H),3.13(d,J=5.6Hz,3H),2.81–2.61(m,1H),1.52–0.94(m,12H).
Example 79B
LC-MS:m/z 638(M+H)+.1H NMR(400MHz,DMSO)δ10.12(brs,1H),8.48(s,1H),7.92–7.75(m,2H),7.65(t,J=7.8Hz,1H),7.22(m,1H),6.92–6.53(m,3H),6.19(m,1H),5.84–5.69(m,1H),4.70(m,2H),4.19–3.47(m,4H),2.90(s,3H),2.57(m,1H),1.37–0.90(m,12H).
Example 804- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) -1- (2-isopropyl-6- (isopropylsulfonyl) phenyl) pyrido [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520001122
LC-MS:m/z 652(M+H)+.1H NMR(400MHz,CDCl3)δ8.91(brs,1H),8.04(d,J=7.2Hz,1H),7.87(dd,J=9.2Hz,3.2Hz,1H),7.81(dd,J=8.0Hz,1.2Hz,1H),7.72-7.67(m,1H),7.26(overlap,1H),6.69-6.54(m,3H),6.42(dd,J=16.8Hz,1.6Hz,1H),5.81(dd,J=10.8Hz,1.6Hz,1H),5.10-3.40(m,7H),3.30-3.00(m,1H),2.75-2.60(m,1H),1.50-1.48(m,3H),1.31-1.24(m,6H),1.17-1.15(m,3H),1.02-0.99(m,3H).
Example 80-1 two isomers, examples 80A and 80B, were obtained by chiral separation:
Figure BDA0002861043520001131
example 80A
LC-MS:m/z 652(M+H)+.1H NMR(400MHz,DMSO)δ10.19(brs,1H),8.28(t,J=9.7Hz,1H),7.90–7.76(m,2H),7.64(t,J=7.8Hz,1H),7.26(m,1H),6.97–6.58(m,3H),6.21(m,1H),5.77(m,1H),4.86(m,1H),4.49–3.96(m,3H),3.83–3.48(m,2H),3.28–3.02(m,2H),2.59(m,1H),1.34(m,3H),1.18–0.89(m,12H).
Example 80B
LC-MS:m/z 652(M+H)+.1H NMR(400MHz,DMSO)δ10.21(brs,1H),8.38(s,1H),7.82(m,2H),7.74–7.58(m,1H),7.26(m,1H),7.05–6.58(m,3H),6.22(m,1H),5.88–5.67(m,1H),4.92(m,1H),4.63–3.95(m,3H),3.74(m,2H),3.29(m,1H),3.05(m,1H),2.62(m,1H),1.35–1.19(m,3H),1.18–0.82(m,12H).
Example 814- ((2S, 5R) -4-acryloyl-2, 5-dimethylpiperazin-1-yl) -1- (2-cyclopropyl-6- (methylsulfonyl) phenyl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) pyrido [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520001132
LC-MS:m/z 636(M+H)+.1H NMR(400MHz,CDCl3)δ9.16-8.90(m,1H),8.14-8.02(m,1H),7.92-7.82(m,1H),7.65(t,J=8.0Hz,1H),7.54-7.41(m,1H),7.30-7.21(m,1H),6.73-6.50(m,3H),6.40(t,J=15.2Hz,1H),5.81(t,J=8.8Hz,1H),5.18-3.67(m,6H),3.18-3.07(m,3H),1.53-1.19(m,7H),0.93-0.53(m,4H).
Example 81-1 two isomeric examples 81A and 81B were obtained by chiral separation:
Figure BDA0002861043520001133
example 81A
LC-MS:m/z 636(M+H)+.1H NMR(400MHz,DMSO)δ10.22(brs,1H),8.31(d,J=9.0Hz,1H),7.86(d,J=7.6Hz,1H),7.58(t,J=7.8Hz,1H),7.54–7.44(m,1H),7.26(dd,J=15.3,8.0Hz,1H),6.96–6.52(m,3H),6.19(m,1H),5.79–5.65(m,1H),4.97–4.36(m,2H),4.16(m,1H),3.95–3.41(m,3H),3.00(s,3H),1.67–1.46(m,1H),1.24(m,6H),0.75–0.43(m,4H).
Example 81B
LC-MS:m/z 636(M+H)+.1H NMR(400MHz,DMSO)δ10.29(brs,1H),8.34(dd,J=8.9,4.0Hz,1H),7.85(d,J=7.5Hz,1H),7.58(t,J=7.8Hz,1H),7.43(d,J=7.7Hz,1H),7.26(dd,J=15.4,8.0Hz,1H),6.95–6.55(m,3H),6.19(m,1H),5.75(m,1H),4.74(m,2H),4.22–3.51(m,4H),2.96(s,3H),1.64–1.48(m,1H),1.25(m,6H),0.63(m,4H).
Example 824- ((2S, 5R) -4-acryloyl-2, 5-dimethylpiperazin-1-yl) -1- (2-ethyl-6- (isopropylsulfonyl) phenyl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) pyrido [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520001141
LC-MS:m/z 652(M+H)+.1H NMR(400MHz,CDCl3)δ9.10-8.09(m,1H),8.06-8.03(m,1H),7.88-7.85(m,1H),7.80-7.76(m,1H),7.70-7.65(m,1H),7.28-7.22(m,1H),6.70-6.55(m,3H),6.39(t,J=13.6Hz,1H),5.80-5.78(m,1H),5.07-3.40(m,7H),2.52-2.49(m,1H),2.36-2.33(m,1H),1.50-1.41(m,3H),1.40-1.29(m,6H),1.27-1.17(m,6H).
Example 82-1 two isomeric examples 82A and 82B were obtained by chiral separation:
Figure BDA0002861043520001142
example 82A
LC-MS:m/z 652(M+H)+.1H NMR(400MHz,DMSO)δ10.16(brs,1H),8.29–8.11(m,1H),7.78(t,J=7.0Hz,2H),7.63(t,J=7.8Hz,1H),7.27(m,1H),6.96–6.58(m,3H),6.19(m,1H),5.75(m,1H),4.66(m,2H),4.46–4.13(m,2H),3.94–3.57(m,2H),3.23(m,1H),2.31(dd,J=14.7,7.2Hz,2H),1.38(d,J=6.5Hz,3H),1.22(m,3H),1.10–0.86(m,9H).
Example 82B
LC-MS:m/z 652(M+H)+.1H NMR(400MHz,DMSO)δ10.13(brs,1H),8.37(t,J=8.2Hz,1H),7.79(t,J=6.4Hz,2H),7.64(t,J=7.7Hz,1H),7.26(m,1H),6.93–6.57(m,3H),6.19(m,1H),5.75(m,1H),5.05–4.31(m,2H),4.21–3.52(m,4H),3.17–2.99(m,1H),2.30(d,J=6.9Hz,2H),1.40–0.84(m,15H).
Example 834- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -1- (2-ethyl-6- (isopropylsulfonyl) phenyl) -6-fluoro-7- (2-fluoro-6-hydroxyphenyl) pyrido [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520001151
LC-MS:m/z 638(M+H)+.1H NMR(400MHz,CDCl3)δ8.96(brs,1H),8.05(d,J=7.6Hz,1H),7.89-7.85(m,1H),7.78(d,J=8Hz,1H),7.70-7.68(m,1H),7.28-7.22(m,1H),6.70-6.61(m,3H),6.43-6.39(m,1H),5.83-5.80(m,1H),5.00-3.06(m,8H),2.58-2.50(m,1H),2.39-2.35(m,1H),1.33-1.28(m,6H),1.26-1.15(m,6H).
Example 83-1 two isomers, examples 83A and 83B, were obtained by chiral separation:
Figure BDA0002861043520001152
example 83A
LC-MS:m/z 638(M+H)+.1H NMR(400MHz,DMSO)δ10.19(brs,1H),8.28(d,J=8.8Hz,1H),7.78(t,J=6.9Hz,2H),7.63(t,J=7.7Hz,1H),7.26(m,1H),6.75(m,3H),6.21(m,1H),5.77(m,1H),4.84(s,1H),4.57–3.99(m,3H),3.78–3.49(m,2H),3.19(m,2H),2.41–2.18(m,2H),1.36(d,J=6.4Hz,3H),1.12–0.81(m,9H).
Example 83B
LC-MS:m/z 638(M+H)+.1H NMR(400MHz,DMSO)δ9.31(br,1H),8.35(s,1H),7.84–7.70(m,2H),7.63(t,J=7.8Hz,1H),7.25(m,1H),6.96–6.55(m,3H),6.21(m,1H),5.77(m,1H),4.90(m,1H),4.59–3.94(m,3H),3.66(m,2H),3.07(m,2H),2.39–2.23(m,2H),1.31–1.15(m,3H),1.15–0.89(m,9H).
Example 844- ((2S, 5R) -4-acryloyl-2, 5-dimethylpiperazin-1-yl) -6-chloro-1- (2-ethyl-6- (methylsulfonyl) phenyl) -7- (2-fluoro-6-hydroxyphenyl) pyrido [2,3-d ] pyrimidin-2 (1H) -one
Figure BDA0002861043520001161
LC-MS:m/z 640(M+H)+.1H NMR(400MHz,CDCl3)δ8.19-8.06(m,2H),8.06-7.83(m,1H),7.75(t,J=8.0Hz,1H),7.66(dt,J=8.0Hz,2.0Hz,1H),7.26-7.20(m,1H),6.77-6.50(m,3H),6.40(t,J=15.2Hz,1H),5.81(t,J=8.8Hz,1H),5.21-3.64(m,6H),3.17-3.05(m,3H),2.60-2.31(m,2H),1.55-1.34(m,6H),1.22-1.11(m,3H).
Example 84-1 two isomeric examples 84A and 84B were obtained by chiral separation:
Figure BDA0002861043520001162
example 84A
LC-MS:m/z 640(M+H)+.1H NMR(400MHz,DMSO)δ10.20(brs,1H),8.42(d,J=10.9Hz,1H),7.88(d,J=7.7Hz,1H),7.73(s,1H),7.60m,1H),7.22(m,1H),6.94–6.52(m,3H),6.19(m,1H),5.85–5.69(m,1H),4.65(m,2H),4.37–4.05(m,2H),3.74(m,2H),2.97(s,3H),2.33(m,2H),1.45–0.86(m,9H).
Example 84B
LC-MS:m/z 640(M+H)+.1H NMR(400MHz,DMSO)δ10.21(brs,1H),8.48(m,1H),7.88(d,J=7.7Hz,1H),7.74(s,1H),7.63(t,J=7.8Hz,1H),7.23(dd,J=15.4,8.0Hz,1H),6.82(dt,J=16.8,10.0Hz,1H),6.74–6.54(m,2H),6.19(m,1H),5.85–5.64(m,1H),4.71(m,2H),3.95(m,4H),2.95(s,3H),2.31(m,2H),1.39–0.97(m,9H).
Biological test evaluation
The following biological test examples further illustrate the present invention, but these examples are not meant to limit the scope of the present invention.
Compound Pair NCI-H358 (KRAS)G12CMutation) cell experiment of antiproliferative activity of cells.
Experimental procedure
To the peripheral wells of 384 microwell plates 40. mu.L of phosphate buffer was added, followed by 40. mu.L of the test cell suspension to the other wells, and the microwell plates were then placed in a carbon dioxide incubator overnight.
The test compounds were diluted in a gradient of 10 concentrations (from 50. mu.M to 0.003. mu.M) and 100nL of each was added to the corresponding well of the microplate. After dosing, 40. mu.L of phosphate buffer was added to each well at line A, P and columns 1 and 24, and the plate was incubated for 5 days in a carbon dioxide incubator.
mu.L of Promega CellTiter-Glo reagent was added to each well of the plate, followed by shaking at room temperature for 10min to stabilize the luminescence signal, which was then read using a Pekinelmer Envision multi-label analyzer.
Finally, IC of the compound was calculated using GraphPad Prism software50And (5) obtaining values, and drawing a fitting curve.
Compound couple NCI-H358 (KRAS) as an example of the present inventionG12CMutant) cell antiproliferative activity is shown in table 1.
TABLE 1 antiproliferative activity of the compounds of the examples of the invention
Figure BDA0002861043520001171
Figure BDA0002861043520001181
Figure BDA0002861043520001191
As can be seen from table 1:
example Compounds of the invention for KRASG12CThe mutant NCI-H358 cells showed very good cell antiproliferative activity, especially compounds containing a phenylsulfone structure.
Pharmacokinetic test evaluation
Evaluation of mouse pharmacokinetic test
Male ICR mice, weighing about 20-30g, were fasted overnight and then gavaged with 30mg/kg of the compound of the present invention, a solution of example 10B, 16B, 18A, 63B, 78B [ CMC/TW80 as vehicle ]. Blood was collected at 0.5, 1.0, 2.0, 4.0, 6.0, 8.0, 12 and 24h after administration of the compound of the present invention, and the concentration of the compound of the present invention in plasma was determined by LC/MS/MS, respectively.
Figure BDA0002861043520001201
The test results are shown in Table 2
Table 2 summary of pharmacokinetic parameters (n ═ 4, mean)
Figure BDA0002861043520001202
According to the detection result, the compounds with similar structures, wherein the compounds containing the phenylsulfone structure have better pharmacokinetic properties compared with the compounds with the pyridine sulfone structure.
Evaluation of rat pharmacokinetic testing
Male SD rats weighing about 220g were fasted overnight and then were gavaged with 15mg/kg of a solution of the compound of the present invention or the control compound AMG510 [ DMSO/PEG400 as vehicle ]. Blood was collected at 0.5, 1.0, 2.0, 4.0, 6.0, 8.0, 12 and 24 after administration of the compound of the present invention, or at 0.5, 1.0, 2.0, 4.0, 6.0, 8.0, 12 and 24 after 7 days of continuous administration, respectively, and the concentration of the compound of the present invention or the control compound AMG510 in plasma was determined by LC/MS.
The detection result shows that the compound of the invention has good pharmacokinetic property relative to AMG 510.
Pharmacodynamic test evaluation of antitumor activity
1. H358 CDX tumor model
100uL of the solution containing 5x106NCI-H358 tumor cell suspension was inoculated subcutaneously into the right back of nude mice. Mice were monitored daily for health and measurements were started when tumors grew to reach. The tumor volume calculation formula is 0.5xLxW2, wherein L, W represents the tumor length and width, respectively. Tumor growth to-200 mm3Mice were randomly grouped. Mice were gavaged daily with a corresponding dose (20mg/Kg) of compound in solution while monitoring their general state. Tumors were measured 2 times per week and body weights were measured twice per week. Test resultsAs shown in table 3.
TABLE 3 evaluation of pharmacodynamic test for antitumor Activity
Figure BDA0002861043520001203
Figure BDA0002861043520001211
The results show that the compounds containing the phenylsulfone structure (such as the compound 63B and the example 78B) have better drug effects than the control compound AMG 510.
2. MIA PaCa-2 CDX tumor model
100uL of 5x106The MIA PaCa-2 tumor cell suspension was subcutaneously inoculated into the right posterior abdomen of nude mice. Mice were monitored daily for health and measurements were started when tumors grew to reach. The formula for calculating the tumor volume is 0.5xLxW 2Wherein L, W represents tumor length and width, respectively. Tumor growth to 150mm3Mice were randomly grouped. Mice were gavaged daily with the corresponding dose (3, 10mg/Kg) of CMC-Na suspension of compound while monitoring their general state. Tumors were measured 3 times per week and body weights were measured twice per week.
The detection result shows that the compound has good anti-tumor effect.
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 (13)

1. A compound of formula (I), a stereoisomer, a tautomer, a crystalline form, a pharmaceutically acceptable salt, a hydrate, a solvate, or a prodrug thereof:
Figure FDA0002861043510000011
in the formula:
A. b are the same or different and are each independently selected from the group consisting of: CH. CR5Or N;
x is selected from the group consisting of: 4-14 membered saturated or unsaturated heterocyclic group, C4-C14Cycloalkyl radical, C6-C14Aryl or 5-14 membered heteroaryl, wherein said heterocyclyl, cycloalkyl, aryl or heteroaryl may be optionally substituted with one or more R 8Substituted;
u, V, W and Q are the same or different and are each independently selected from the group consisting of: CH. CR3Or N;
R1selected from the group consisting of:
Figure FDA0002861043510000012
Figure FDA0002861043510000013
wherein the content of the first and second substances,
Figure FDA0002861043510000014
represents a double bond "═ or a triple bond" ≡ ";
RAis absent or is independently selected from the group consisting of: hydrogen, deuterium, fluoro, cyano, or C1-C3An alkyl group; each RBIndependently selected from the group consisting of: hydrogen, deuterium, cyano, or C1-C3An alkyl group; wherein the alkyl group may be substituted with one or more substituents selected from the group consisting of: deuterium, halogen, cyano, amino, C3-C7Cycloalkyl, 4-7 membered heterocyclyl, NHR9Or NR9R10;R9And R10Each independently is C1-C3An alkyl group; or R9,R10Together with the N atom to which they are attached form a substituted or unsubstituted 4-8 membered heterocyclyl;
p is an integer of 1 or 2;
R2selected from the group consisting of substituted:C6-C14Aryl, 5-14 membered heteroaryl, wherein said substitution means substitution by one or more groups selected from the group consisting of: r ', -SR ', -SOR ', -SO2R'、-SO2NR'R”、-NR'SO2R ", -P (═ O) R' R"; with the proviso that C is6-C14Aryl, 5-14 membered heteroaryl, having at least one substituent selected from: -SR ', -SOR', -SO2R'、-SO2NR'R”、-NR'SO2R ", or-P (═ O) R' R"; r 'and R' are the same or different and are each independently selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C 1-C6Alkyl radical, C3-C8Cycloalkyl, C4-C10Cycloalkenyl, 4-8 membered heterocyclyl, C6-C14Aryl, 5-14 membered heteroaryl; or when R ' and R ' are attached to the same N atom, R ' together with the N atom to which they are attached form a substituted or unsubstituted 4-8 membered heterocyclyl;
R3selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C18Alkyl, deuterated C1-C18Alkyl, halo C1-C18Alkyl radical, C3-C20Cycloalkyl radical, C1-C18Alkoxy, deuterated C1-C18Alkoxy, halo C1-C18Alkoxy, halogen, nitro, hydroxyl, cyano, ester, amino, amido, sulfamide, carbamido, 4-20-membered heterocyclic radical, C6-C14Aryl, 5-14 membered heteroaryl;
l is selected from the group consisting of: a bond, -C (O) -, C1-C3An alkylene group;
R4selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C18Alkyl, deuterated C1-C18Alkyl, halo C1-C18Alkyl radical, C3-C20Cycloalkyl radical, C1-C18Alkoxy, deuterated C1-C18Alkoxy, halo C1-C18Alkoxy, amino, hydroxy, 4-20 membered heterocyclic group, C6-C14Aryl, 5-14 membered heteroaryl;
R5selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C18Alkyl, deuterated C1-C18Alkyl, halo C1-C18Alkyl radical, C3-C20Cycloalkyl radical, C1-C18Alkoxy, deuterated C1-C18Alkoxy, halo C1-C18Alkoxy, halogen, nitro, hydroxyl, cyano, ester, amino, amido, sulfamide, carbamido, 4-20-membered heterocyclic radical, C 6-C14Aryl, 5-14 membered heteroaryl;
R6selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C18Alkyl, deuterated C1-C18Alkyl, halo C1-C18Alkyl radical, C3-C20Cycloalkyl radical, C1-C18Alkoxy, deuterated C1-C18Alkoxy, halo C1-C18Alkoxy, 4-to 20-membered heterocyclic group, C6-C14Aryl, 5-14 membered heteroaryl;
R8independently selected from the group consisting of substituted or unsubstituted: hydrogen, deuterium, C1-C18Alkyl, deuterated C1-C18Alkyl, halo C1-C18Alkyl radical, C3-C20Cycloalkyl radical, C1-C18Alkoxy, deuterated C1-C18Alkoxy, halo C1-C18Alkoxy, amino, hydroxy, 4-20 membered heterocyclic group, C6-C14Aryl, 5-14 membered heteroaryl;
wherein, the term "substituted" when not specifically stated, means substituted by one or more groups selected from the group consisting of: hydrogen, deuterium, C1-C18Alkyl, deuterated C1-C18Alkyl, halo C1-C18Alkyl radical, C3-C20Cycloalkyl radical, C1-C18Alkoxy, deuterated C1-C18Alkoxy, halo C1-C18Alkoxy radical, C6-C14Aryl, 5-14 membered heteroaryl, 4-20 membered heterocyclyl, halogen, nitro, hydroxy, cyano, ester, amine, NRbC(=O)ORe、OC(=O)Re
OC(=O)NRbRcAmide, sulfonamide, or ureido groups; rb、RcMay independently represent hydrogen, deuterium, C1-C6 alkyl, C3-C8 cycloalkyl, 4-8 membered heterocyclyl, 5-14 membered heteroaryl or C6-C14 aromatic ring, or RbAnd RcTogether with the N atom, may form a 4-8 membered heterocyclic group; r eMay independently represent hydrogen, C1-C6 alkyl, C3-C8 cycloalkyl, C2-C6 alkenyl, C3-C6 cycloalkenyl, C2-C6 alkynyl, 4-8 membered heterocyclyl, 5-14 membered heteroaryl, or C6-C14 aryl ring;
the limiting conditions are as follows:
when B is N, R1Selected from the group consisting of:
Figure FDA0002861043510000021
wherein p is 2;
when B is CH or CR5When R is1Selected from the group consisting of:
Figure FDA0002861043510000022
Figure FDA0002861043510000023
wherein p is 2;
when V is C (Cl), R2Is not selected from:
Figure FDA0002861043510000024
and L is not selected from a bond; and
R4is not selected from:
Figure FDA0002861043510000025
2. a compound of formula (I), its stereoisomers, tautomers, crystalline forms, pharmaceutically acceptable salts, hydrates, solvates or prodrugs according to claim 1, having the structure shown in formula (III):
Figure FDA0002861043510000031
R1、R2、R4x, L, U, V, W, Q are as defined in claim 1.
3. A compound of formula (I), its stereoisomers, tautomers, crystalline forms, pharmaceutically acceptable salts, hydrates, solvates or prodrugs according to claim 1, having the structure shown in formula (IV):
Figure FDA0002861043510000032
in the formula:
R1、R2、R4、R8l, U, V, W, Q are as defined in claim 1.
4. A compound of formula (I), its stereoisomers, tautomers, crystalline forms, pharmaceutically acceptable salts, hydrates, solvates or prodrugs according to claim 1, having the structure shown in formula (V):
Figure FDA0002861043510000033
In the formula:
R1、R2、R4、R8u, V, W, Q are as defined in claim 1.
5. A compound of formula (I), its stereoisomers, tautomers, crystalline forms, pharmaceutically acceptable salts, hydrates, solvates or prodrugs according to claim 1, having the structure shown in formula (VI):
Figure FDA0002861043510000034
in the formula:
R1、R2、R4、R8u, V, Q are as defined in claim 1.
6. A compound of formula (I), its stereoisomers, tautomers, crystalline forms, pharmaceutically acceptable salts, hydrates, solvates or prodrugs according to claim 1, having the structure shown in formula (VII):
Figure FDA0002861043510000041
in the formula:
R1、R2、R4、R8v, Q are as defined in claim 1.
7. A compound of formula (I), its stereoisomers, tautomers, crystalline forms, pharmaceutically acceptable salts, hydrates, solvates or prodrugs thereof according to claims 1-6, characterized in that it has the structure shown in formula (VIII):
Figure FDA0002861043510000042
in the formula (I), the compound is shown in the specification,
r' "is selected from the group consisting of substituted or unsubstituted: deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C1-C6Alkyl radical, C3-C8Cycloalkyl radical, C4-C10Cycloalkenyl, 4-8 membered heterocyclyl, C6-C14Aryl, 5-14 membered heteroaryl, wherein said substitution means substitution by one or more groups selected from the group consisting of: deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C 1-C6Alkyl radical, C3-C8Cycloalkyl, C4-C10Cycloalkenyl, 4-8 membered heterocyclyl, C6-C14Aryl, 5-14 membered heteroaryl;
q is selected from: 1. 2, 3 or 4;
R1、R4、R8r', V, Q are as defined in claim 1.
8. A compound of formula (I), its stereoisomers, tautomers, crystalline forms, pharmaceutically acceptable salts, hydrates, solvates or prodrugs according to claim 7, having the structure shown in formula (IX):
Figure FDA0002861043510000043
in the formula (I), the compound is shown in the specification,
R1selected from:
Figure FDA0002861043510000051
wherein R isASelected from: H. d, halogen or cyano; rB、RB’The same or different, each independently selected from: H. d, halogen, cyano, substituted or unsubstituted C1-C3An alkyl group; wherein said substitution is by one or more groups selected from the group consisting of: D. halogen, cyano, C1-C3Alkyl radical, C3-C6Cycloalkyl, 4-6 membered heterocyclyl or NRIVRV;RIV、RVThe same or different, each independently selected from: H. c1-C3Alkyl radical, C3-C6Cycloalkyl or 4-6 membered heterocyclyl; or RIV、RVAnd the adjacent N together form a 4-6 membered heterocyclyl;
R4、R'、V、q, R' "and q are as defined in claim 7.
9. A compound of formula (I), a stereoisomer, a tautomer, a crystalline form, a pharmaceutically acceptable salt, a hydrate, a solvate, or a prodrug thereof, according to any one of claims 1 to 8, characterized in that it has the structure shown in formula (X) or (XI):
Figure FDA0002861043510000052
In the formula (I), the compound is shown in the specification,
R4selected from substituted or unsubstituted C6-C14Aryl or 5-10 membered heteroaryl, wherein said substitution means substitution by one or more (e.g. 2, 3, 4 or 5) groups selected from the group consisting of: deuterium, halogen, ester group, cyano group, NRbC(=O)ORe、OC(=O)Re、OC(=O)NRbRcAmino group, C1-C6Alkyl, halo C1-C6Alkyl, hydroxy; rb、RcCan independently represent hydrogen, deuterium, C1-C6Alkyl radical, C3-C8Cycloalkyl, 4-8 membered heterocyclyl, 5-14 membered heteroaryl or C6-C14Aromatic rings, or RbAnd RcTogether with the N atom, may form a 4-8 membered heterocyclic group; reCan independently represent hydrogen, C1-C6Alkyl radical, C3-C8Cycloalkyl radical, C2-C6Alkenyl radical, C3-C6Cycloalkenyl radical, C2-C6Alkynyl, 4-8 membered heterocyclyl, 5-14 membered heteroaryl, or C6-C14 aromatic ring;
rm is selected from the group consisting of substituted or unsubstituted: amino group, C1-C6Alkyl radical, C3-C6Cycloalkyl, 4-6 membered heterocyclyl, wherein said substitution means substitution by one or more groups selected from the group consisting of: deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C1-C3Alkyl radical, C3-C6Cycloalkyl, 4-6 membered heterocyclyl;
rn is selected from the group consisting of substituted or unsubstituted: amino group, C1-C6Alkyl radical, C1-C6Alkoxy radical, C3-C6Cycloalkyl, -O-C3-C6Cycloalkyl radical, C1-C6Alkyl radical C3-C6Cycloalkyl, -O-C1-C6Alkyl radical C3-C6Cycloalkyl, 4-6 membered heterocyclyl, wherein said substitution means substitution by one or more groups selected from the group consisting of: deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C 1-C3Alkyl radical, C1-C3Haloalkyl, C3-C6Cycloalkyl, 4-6 membered heterocyclyl;
rx is selected from: f or Cl;
RAselected from: H. d, halogen;
q' is selected from 0, 1, 2 or 3;
r' "is selected from the group consisting of substituted or unsubstituted: deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C1-C6Alkyl radical, C3-C8Cycloalkyl radical, C4-C10Cycloalkenyl, 4-8 membered heterocyclyl, C6-C14Aryl, 5-14 membered heteroaryl, wherein said substitution means substitution by one or more groups selected from the group consisting of: deuterium, halogen, nitro, hydroxy, cyano, ester, amino, amide, C1-C6Alkyl radical, C3-C8Cycloalkyl radical, C4-C10Cycloalkenyl, 4-8 membered heterocyclyl, C6-C14Aryl, 5-14 membered heteroaryl.
10. A compound of formula (I), its stereoisomers, tautomers, crystalline forms, pharmaceutically acceptable salts, hydrates, solvates or prodrugs thereof according to claim 1, wherein said compound is selected from the group consisting of:
Figure FDA0002861043510000061
Figure FDA0002861043510000071
Figure FDA0002861043510000081
Figure FDA0002861043510000091
Figure FDA0002861043510000101
Figure FDA0002861043510000111
Figure FDA0002861043510000121
Figure FDA0002861043510000131
Figure FDA0002861043510000141
Figure FDA0002861043510000151
Figure FDA0002861043510000161
Figure FDA0002861043510000171
Figure FDA0002861043510000181
Figure FDA0002861043510000191
Figure FDA0002861043510000201
Figure FDA0002861043510000211
Figure FDA0002861043510000221
Figure FDA0002861043510000231
Figure FDA0002861043510000241
Figure FDA0002861043510000251
Figure FDA0002861043510000261
Figure FDA0002861043510000271
Figure FDA0002861043510000281
Figure FDA0002861043510000291
Figure FDA0002861043510000301
Figure FDA0002861043510000311
Figure FDA0002861043510000321
Figure FDA0002861043510000331
Figure FDA0002861043510000341
Figure FDA0002861043510000351
Figure FDA0002861043510000361
Figure FDA0002861043510000371
Figure FDA0002861043510000381
Figure FDA0002861043510000391
Figure FDA0002861043510000401
Figure FDA0002861043510000411
Figure FDA0002861043510000421
11. a process for preparing a compound of formula (I), a stereoisomer, a tautomer, a crystalline form, a pharmaceutically acceptable salt, a hydrate, a solvate, or a prodrug thereof, comprising the steps of:
Figure FDA0002861043510000431
(i) in an inert solvent, the compound of formula P-1 is reacted first with oxalyl chloride and then with the amine-based compound R 2-NH2Reacting to obtain a compound shown as a formula P-2;
(ii) in an inert solvent, under the action of a first base, a compound of a formula P-2 is subjected to ring closure to obtain a compound of a formula P-3;
(iii) in an inert solvent, the compound of the formula P-3 and phosphorus oxychloride react under the action of a second alkali to obtain a compound of the formula P-4;
(iv) reacting a compound of formula P-4 with a base in an inert solvent in the presence of a base
Figure FDA0002861043510000432
Obtaining a compound of formula P-5 through coupling or substitution reaction;
(v) deprotecting the compound of formula P-5 in an inert solvent in the presence of an acid to give a compound of formula P-6;
(vi) a compound of formula P-6 with R in an inert solvent in the presence of a base1E, obtaining a compound shown in a formula P-7 through coupling, substitution or acylation reaction;
(vii) in an inert solvent in the presence of a base and a catalyst, the compounds of the formulae P-7 and R4-L-E1Obtaining the compound of formula (I) through coupling, substitution or acylation reaction;
in the formula (I), the compound is shown in the specification,
e is selected from: halogen, OH, OCOR1、OCO(iBu);
E1Selected from: -BH2、-B(OH)2
Figure FDA0002861043510000433
-Sn(Bu)3、-ZnBr;
PG is an amino protecting group selected from the group consisting of: boc, Bn, Cbz or Fmoc;
y and Z are leaving groups selected from the group consisting of: halogen or OTf;
the first base is selected from the group consisting of: KHMDS, NaHMDS, LiHMDS, NaH, NaOMe, NaOEt ortBuONa;
The second base is selected from the group consisting of: TEA, DIPEA, DMAP or N, N-dimethylaniline;
R1、R2、R4L, A, B, X, U, V, W and Q are defined as in claim 1.
12. A pharmaceutical composition comprising one or more compounds of formula (I) according to claim 1, stereoisomers, tautomers, crystalline forms, pharmaceutically acceptable salts, hydrates, solvates or prodrugs thereof; and a pharmaceutically acceptable carrier.
13. Use of a compound of formula (I), a stereoisomer, a tautomer, a crystalline form, a pharmaceutically acceptable salt, a hydrate, a solvate, or a prodrug thereof, according to claim 1, or of a pharmaceutical composition according to claim 12, for the preparation of a medicament for the prophylaxis and/or treatment of KRASG12COr an expression level of the compound (b).
CN202011563650.9A 2020-01-13 2020-12-25 Aryl or heteroaryl pyridone or pyrimidone derivative and preparation method and application thereof Pending CN114671866A (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
CN202011563650.9A CN114671866A (en) 2020-12-25 2020-12-25 Aryl or heteroaryl pyridone or pyrimidone derivative and preparation method and application thereof
US17/758,733 US20230118795A1 (en) 2020-01-13 2021-01-12 Aryl or heteroaryl pyridone or pyrimidine derivative, preparation method and use thereof
PCT/CN2021/071331 WO2021143693A1 (en) 2020-01-13 2021-01-12 Aryl or heteroaryl pyridone or pyrimidine derivative, preparation method and use thereof
EP21741983.7A EP4092026A4 (en) 2020-01-13 2021-01-12 Aryl or heteroaryl pyridone or pyrimidine derivative, preparation method and use thereof
CN202180008854.2A CN115175908A (en) 2020-01-13 2021-01-12 Aryl or heteroaryl pyridone or pyrimidone derivative and preparation method and application thereof
PCT/CN2021/099875 WO2021249563A1 (en) 2020-06-12 2021-06-11 Aryl or heteroaryl pyridone or pyrimidone derivative, preparation method therefor and application thereof
PCT/CN2021/141360 WO2022135591A1 (en) 2020-12-25 2021-12-24 Aryl or heteroaryl pyridone or pyrimidone derivative and preparation method therefor and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202011563650.9A CN114671866A (en) 2020-12-25 2020-12-25 Aryl or heteroaryl pyridone or pyrimidone derivative and preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN114671866A true CN114671866A (en) 2022-06-28

Family

ID=82071144

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202011563650.9A Pending CN114671866A (en) 2020-01-13 2020-12-25 Aryl or heteroaryl pyridone or pyrimidone derivative and preparation method and application thereof

Country Status (2)

Country Link
CN (1) CN114671866A (en)
WO (1) WO2022135591A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115175908A (en) * 2020-01-13 2022-10-11 苏州泽璟生物制药股份有限公司 Aryl or heteroaryl pyridone or pyrimidone derivative and preparation method and application thereof

Family Cites Families (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG11201906223TA (en) * 2016-12-22 2019-08-27 Amgen Inc Benzisothiazole, isothiazolo[3,4-b]pyridine, quinazoline, phthalazine, pyrido[2,3-d]pyridazine and pyrido[2,3-d]pyrimidine derivatives as kras g12c inhibitors for treating lung, pancreatic or colorectal cancer
JOP20190272A1 (en) * 2017-05-22 2019-11-21 Amgen Inc Kras g12c inhibitors and methods of using the same
EP3679040B1 (en) * 2017-09-08 2022-08-03 Amgen Inc. Inhibitors of kras g12c and methods of using the same
MA52501A (en) * 2018-05-04 2021-03-10 Amgen Inc KRAS G12C INHIBITORS AND THEIR PROCEDURES FOR USE
US20190375749A1 (en) * 2018-06-11 2019-12-12 Amgen Inc. Kras g12c inhibitors and methods of using the same
MX2020012261A (en) * 2018-06-12 2021-03-31 Amgen Inc Kras g12c inhibitors encompassing a piperazine ring and use thereof in the treatment of cancer.
JP7377679B2 (en) * 2018-11-19 2023-11-10 アムジエン・インコーポレーテツド Combination therapy comprising a KRASG12C inhibitor and one or more additional pharmaceutically active agents for the treatment of cancer
CA3128062A1 (en) * 2019-01-29 2020-08-06 Brightgene Bio-Medical Technology Co., Ltd. Benzopyridone heterocyclic compound and use thereof
EP3972967A4 (en) * 2019-05-21 2023-07-12 InventisBio Co., Ltd. Heterocyclic compounds, preparation methods and uses thereof
JP2022534765A (en) * 2019-05-29 2022-08-03 上▲海▼翰森生物医▲薬▼科技有限公司 Nitrogen-containing heterocyclic derivative regulator, method of preparation and use thereof
CN112552294B (en) * 2019-09-10 2023-12-19 上海翰森生物医药科技有限公司 Piperazine heterocyclic derivative-containing inhibitor, preparation method and application thereof
WO2021011417A1 (en) * 2019-07-12 2021-01-21 The Regents Of The University Of California Chemically controlled monoclonal antibody target engagement
CN112390818B (en) * 2019-08-12 2023-08-22 劲方医药科技(上海)有限公司 Substituted heteroaromatic dihydro pyrimidinone derivatives, their preparation and pharmaceutical use
CN112390796B (en) * 2019-08-19 2023-06-27 贝达药业股份有限公司 KRAS G12C inhibitor and application thereof in medicine
EP4045047A1 (en) * 2019-10-15 2022-08-24 Amgen Inc. Combination therapy of kras inhibitor and shp2 inhibitor for treatment of cancers
WO2021081212A1 (en) * 2019-10-24 2021-04-29 Amgen Inc. Pyridopyrimidine derivatives useful as kras g12c and kras g12d inhibitors in the treatment of cancer
CN112225734B (en) * 2019-10-25 2021-12-07 南京瑞捷医药科技有限公司 KRAS G12C inhibitors and uses thereof
CN113286794B (en) * 2019-11-04 2024-03-12 北京加科思新药研发有限公司 KRAS mutein inhibitors
CN111377918B (en) * 2019-11-29 2021-03-02 苏州信诺维医药科技有限公司 KRAS inhibitor compound
WO2021113595A1 (en) * 2019-12-06 2021-06-10 Beta Pharma, Inc. Phosphorus derivatives as kras inhibitors
CN112920183A (en) * 2019-12-06 2021-06-08 南京圣和药业股份有限公司 Compounds as KRAS-G12C inhibitors and uses thereof
US20230028414A1 (en) * 2019-12-16 2023-01-26 Amgen Inc. Dosing regimen of kras g12c inhibitor
WO2021120045A1 (en) * 2019-12-18 2021-06-24 InventisBio Co., Ltd. Heterocyclic compounds, preparation methods and uses thereof
CN113651814B (en) * 2019-12-19 2022-06-17 北京加科思新药研发有限公司 KRAS mutein inhibitors
CN113061132B (en) * 2020-01-01 2023-11-14 上海凌达生物医药有限公司 Condensed ring lactam compound, preparation method and application
CN111205286B (en) * 2020-01-13 2022-12-13 中科苏州药物研究院 Nitrile methyl piperazine derivative as KRAS G12C mutant protein inhibitor and application thereof
CN113105448A (en) * 2020-01-13 2021-07-13 苏州泽璟生物制药股份有限公司 Aryl or heteroaryl pyridone or pyrimidone derivative and preparation method and application thereof
CN112159405B (en) * 2020-02-04 2021-09-14 广州必贝特医药技术有限公司 Pyridopyrimidinone compounds and application thereof
CN112574199B (en) * 2020-05-20 2021-05-18 首药控股(北京)股份有限公司 Heterocyclic compounds as Kras-G12C inhibitors

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115175908A (en) * 2020-01-13 2022-10-11 苏州泽璟生物制药股份有限公司 Aryl or heteroaryl pyridone or pyrimidone derivative and preparation method and application thereof

Also Published As

Publication number Publication date
WO2022135591A1 (en) 2022-06-30

Similar Documents

Publication Publication Date Title
CN115175908A (en) Aryl or heteroaryl pyridone or pyrimidone derivative and preparation method and application thereof
KR20220119088A (en) KRAS mutant protein inhibitor
CN112694475A (en) Cycloalkyl and heterocycloalkyl inhibitors, and preparation method and application thereof
CN113666923A (en) Alkoxy alkyl substituted heterocyclic inhibitor and preparation method and application thereof
WO2021249563A1 (en) Aryl or heteroaryl pyridone or pyrimidone derivative, preparation method therefor and application thereof
JP2022549171A (en) Fused pyridone compounds, and methods of preparation and uses thereof
CN112778301A (en) Tetrahydropyridopyrimidine inhibitor and preparation method and application thereof
CN115109078A (en) Pyrimidopyridine inhibitor and preparation method and application thereof
CN113105448A (en) Aryl or heteroaryl pyridone or pyrimidone derivative and preparation method and application thereof
CN114423751B (en) Novel heterocyclic compounds useful as selective AURORA a inhibitors
CN115043842A (en) Amino-substituted bicyclic inhibitor and preparation method and application thereof
WO2021003310A1 (en) Heterocyclic compounds as bet inhibitors
CN114835719A (en) Substituted bicyclic aromatic heterocyclic amine inhibitor and preparation method and application thereof
CN114835703A (en) Substituted pyrimidopyridone inhibitor and preparation method and application thereof
CN112824410A (en) Aza-heptacyclic inhibitor and preparation method and application thereof
CN114981273B (en) Heterocyclic amide compound, pharmaceutically acceptable salt thereof, preparation method and application thereof
CN112867717B (en) Compounds useful as kinase inhibitors and uses thereof
EP4234548A1 (en) Substituted benzo or pyridopyrimidine amine inhibitor, preparation method therefor, and application thereof
CN116217591A (en) Pyridopyrimidine derivatives serving as KRAS G12D mutation inhibitors
WO2017098440A1 (en) Substituted benzazinones as antibacterial compounds
CN113801113A (en) Aryl or heteroaryl pyridone or pyrimidone derivative and preparation method and application thereof
CN113045569B (en) Compounds useful as RET kinase inhibitors and uses thereof
CN112707892A (en) Pyridazinone or pyridazine compound and derivative and pharmaceutical composition thereof
CN112824420B (en) Compounds useful as EGFR kinase inhibitors and uses thereof
CN114671866A (en) Aryl or heteroaryl pyridone or pyrimidone derivative and preparation method and application thereof

Legal Events

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