CN114621244A - Pyridine derivative and application thereof in medicine - Google Patents

Pyridine derivative and application thereof in medicine Download PDF

Info

Publication number
CN114621244A
CN114621244A CN202111526178.6A CN202111526178A CN114621244A CN 114621244 A CN114621244 A CN 114621244A CN 202111526178 A CN202111526178 A CN 202111526178A CN 114621244 A CN114621244 A CN 114621244A
Authority
CN
China
Prior art keywords
compound
methylpyridin
isopropyl
pyrimidin
pyrido
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN202111526178.6A
Other languages
Chinese (zh)
Other versions
CN114621244B (en
Inventor
张靖
魏用刚
尹杰
孙毅
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.)
Chengdu Baiyu Pharmaceutical Co Ltd
Original Assignee
Chengdu Baiyu Pharmaceutical 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 Chengdu Baiyu Pharmaceutical Co Ltd filed Critical Chengdu Baiyu Pharmaceutical Co Ltd
Publication of CN114621244A publication Critical patent/CN114621244A/en
Application granted granted Critical
Publication of CN114621244B publication Critical patent/CN114621244B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/12Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains three hetero rings
    • C07D491/14Ortho-condensed systems
    • C07D491/147Ortho-condensed systems the condensed system containing one ring with oxygen as ring hetero atom and two rings with nitrogen as ring hetero atom
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The application discloses a compound shown as a general formula (I) or pharmaceutically acceptable salt thereof and application thereof in medicines

Description

Pyridine derivative and application thereof in medicine
Technical Field
The invention relates to pyridine derivatives and their use in medicine.
Background
RAS gene is one of the human important protooncogenes, and plays a key role in regulating and controlling cell signal transduction and the generation and development of tumors; and the most frequently mutated oncogene in human cancers, RAS gene mutations are present in about 30% of cancers. The human RAS super-protein family contains three genes (KRAS, NRAS and HRAS, respectively) encoding four proteins (KRAS-4A, KRAS-4B, NRAS and HRAS). Wherein KRAS is the most common subtype in the RAS family and is also the most mutable gene; KRAS mutations were detected in 86% of RAS mutations, in 86-96% of pancreatic, 40-54% of colorectal and 20-37% of lung cancers, and also occur in cancers such as biliary duct, cervical, bladder, liver and breast (Kessler et al, 2019).
The KARS proteins belong to the small GTPase (small GTPase) family of proteins. In normal physiological states, KRAS protein effects modulation of the signaling pathway by transitioning between inactive (GDP-bound) and active (GTP-bound) states (Downward, 2003): (ii) is in an inactivated state when KRAS binds Guanosine Diphosphate (GDP); activation when bound to Guanosine Triphosphate (GTP) can activate downstream signaling pathways. The transition and balance between these two states is regulated by two classes of proteins: (1) guanine nucleotide exchange factors (GEF) can promote the combination of KRAS and GTP, improve the dissociation efficiency of GDP from KRAS and promote the activation of KRAS; (2) GTPase activating/activating proteins (GAPs) can improve the relatively weak GTPase activity of KRAS protein, promote GTP hydrolysis to GDP and reduce the influence of KRAS on downstream signal paths. Under physiological conditions, KRAS has stronger binding capacity with GDP, so KRAS in cells is normally in an inactivated state; KRAS is activated by binding to GTP, and can promote cell survival, proliferation, cytokine release, etc. through multiple downstream signaling pathways such as MAPK, PI3K, and Ral-GEFs (Liu et al, 2019).
When KRAS is mutated or changed in conformation, the KRAS has reduced GTP hydrolysis activity, increased GDP binding stability and hindered interaction with GAP, so that KRAS protein is activated for a long time, and a large amount of downstream cell signals are stimulated to continuously promote cells to continue to grow and divide, thereby finally causing cancer. Current research indicates that the most common mutation mode of KRAS is single-point mutations (single-point mutations) occurring at codon 12, 13 and 61, wherein the mutation at codon 12 accounts for approximately 82% of the KRAS mutation. Missense mutations at these sites in somatic cells interfere with the GTPase activity of KRAS itself: e.g., mutation at position 12 (G12) persistently activates KRAS by interfering with GAP protein binding and GTP hydrolysis; the mutation at position 13 (G13) results in a decrease in GAP binding capacity; the mutation at position 61 (Q61) affects the stability of the GTP hydrolyzed intermediate (Ostrem & shocat, 2016). Besides the occurrence of tumors caused by the change of KRAS gene, KRAS mutant cells have wide influence on the microenvironment causing tumor deterioration through paracrine action, can promote the secretion of various cytokines, chemokines and growth factors to influence the tumor microenvironment on fibroblasts, immune cells and the like, and can also modify and reprogram stromal cells. These studies fully confirm that the KRAS gene can be an important anti-tumor drug target.
The KRAS inhibitor has been studied for decades, and the target is generally considered to be difficult to prepare before, and the following two main reasons exist: (1) KRAS protein has low affinity for guanosine phosphate (GDP or GTP) on the picomolar (pM) scale, and guanosine phosphate concentrations in cells can reach the millimolar (mM) level, which makes it difficult for general drugs (guanosine phosphate analogs) to competitively bind to KRAS protein (Waters & Der, 2018); (2) the guanosine phosphate binding site of the KRAS protein also lacks regions suitable for binding of other small molecule compounds. Recent studies found that small molecule compounds can inhibit tumors with KRAS-G12C mutation compared to wild-type KRAS protein. Because codon 12 of KRAS protein is close to the nucleoside binding domain and catalytic center of the protein, after cysteine mutation at this site, small molecule compounds can influence the function of KRAS protein by irreversible covalent binding with cysteine, while small molecule compounds are more prone to bind KRAS-GDP protein, reducing GTP affinity for protein (lita, Solomon, Li, Hansen, & Rosen, 2016).
The early developed small molecule compound ARS-853 against KRAS-G12C tumor showed good activity in vitro, effectively inhibited the conversion of GDP and GTP, but lacked in vivo activity (Patricelli et al, 2016). Subsequent further optimization of the structure resulted in ARS-1620, which showed good pharmacological activity against KRAS-G12C tumor cells in vitro and in vivo, but had no significant effect on other KRAS gene mutant cells (Janes et al, 2018). Many KRAS inhibitor drugs are currently in clinical research, but according to the published data, the clinical efficacy of KRAS inhibitors in tumor patients still needs to be further improved. Therefore, a new generation of KRAS-G12C mutation inhibitors with high efficacy and safety needs to be developed to better meet clinical needs.
Disclosure of Invention
One or more embodiments of the present application provide a compound represented by general formula (I), or a pharmaceutically acceptable salt or stereoisomer thereof:
Figure BDA0003409054530000021
wherein
X, Y, Z are each independently CH2Or O;
ring B is
Figure BDA0003409054530000022
Said B ring being optionally substituted by 1,2 or 3R2Substitution;
R1is H or halogen;
R2is H or C1-6An alkyl group;
C1is composed of
Figure BDA0003409054530000023
C2Is composed of
Figure BDA0003409054530000024
In one or more embodiments, X, Y, Z are each independently CH2Or O;
ring B is
Figure BDA0003409054530000031
Said B ring being optionally substituted by 1,2 or 3R2Substitution;
R1is H or halogen;
R2is H or C1-6An alkyl group;
C1is composed of
Figure BDA0003409054530000032
C2Is composed of
Figure BDA0003409054530000033
In one or more embodiments, one of X and Z is O and the other is CH2(ii) a Y is CH2
R1Is H;
ring B is
Figure BDA0003409054530000034
Said B ring being optionally substituted by 1 or 2R2Substitution;
R2is C1-6An alkyl group;
C1is composed of
Figure BDA0003409054530000035
C2Is composed of
Figure BDA0003409054530000036
One or more embodiments of the present application provide a compound represented by general formula (II), or a pharmaceutically acceptable salt or stereoisomer thereof:
Figure BDA0003409054530000037
wherein
X, Y, Z are each independently CH2Or O;
n is 0, 1,2, 3;
ring B is 4-10 membered heterocycloalkyl containing 1,2 or 3N atoms, optionally substituted with 1,2 or 3R4Substitution;
R3is H or halogen;
R4is H or C1-6An alkyl group;
C1is composed of
Figure BDA0003409054530000041
C2Is composed of
Figure BDA0003409054530000042
In one or more embodiments, one of X and Z is O and the other is CH2(ii) a Y is CH2
n is 0 or 1;
ring B is
Figure BDA0003409054530000043
Said B ring being optionally substituted by 1 or 2R4Substitution;
R3is H;
R4is C1-6An alkyl group;
C1is composed of
Figure BDA0003409054530000044
C2Is composed of
Figure BDA0003409054530000045
One or more embodiments of the present application provide a compound represented by general formula (III), or a pharmaceutically acceptable salt or stereoisomer thereof:
Figure BDA0003409054530000046
wherein
X, Y, Z are each independently CH2Or O;
R1、R2each independently is H or C1-6An alkyl group;
R3is H or halogen;
C1is composed of
Figure BDA0003409054530000051
C2Is composed of
Figure BDA0003409054530000052
In one or more embodiments, one of X and Z is O and the other is CH2(ii) a Y is CH2
R1And R2Is C1-6An alkyl group;
C1is composed of
Figure BDA0003409054530000053
C2Is composed of
Figure BDA0003409054530000054
One or more embodiments of the present application provide a compound, or a pharmaceutically acceptable salt or stereoisomer thereof, having one of the following structures:
Figure BDA0003409054530000055
Figure BDA0003409054530000061
Figure BDA0003409054530000071
one or more embodiments of the present application provide a pharmaceutical composition comprising:
(1) a compound of the present application or a stereoisomer, solvate, metabolite, pharmaceutically acceptable salt, co-crystal, or prodrug thereof;
(2) optionally one or more other active ingredients; and
(3) a pharmaceutically acceptable carrier and/or excipient.
One or more embodiments of the present application provide a use of a compound or pharmaceutical composition of the present application in the manufacture of a medicament for preventing or treating cancer, an anti-tumor medicament, a medicament for treating a KRAS mutation-mediated disease, or a KRAS inhibitor.
In one or more embodiments, the cancer is cholangiocarcinoma, cervical cancer, bladder cancer, liver cancer, or breast cancer.
One or more embodiments of the present application provide a compound or pharmaceutical composition of the present application for use as a medicament.
One or more embodiments of the present application provide a compound or pharmaceutical composition of the present application for preventing or treating cancer, anti-tumor, or treating KRAS mutation-mediated diseases.
One or more embodiments of the present application provide a compound or pharmaceutical composition of the present application for use as a KRAS inhibitor.
One or more embodiments of the present application provide a method for preventing or treating cancer, an anti-tumor, or treating a KRAS mutation-mediated disease, comprising administering to a subject in need thereof a compound or pharmaceutical composition of the present application.
Unless stated to the contrary, the terms used in the specification and claims have the following meanings.
Carbon, hydrogen, oxygen, sulfur, nitrogen or F, Cl, Br, I referred to in the groups and compounds of the invention include isotopes thereof, and carbon, hydrogen, oxygen, sulfur or nitrogen referred to in the groups and compounds of the invention are optionally further replaced by one or more of their corresponding isotopes, wherein isotopes of carbon include12C、13C and14c, isotopes of hydrogen including protium (H), deuterium (D, also called deuterium), tritium (T, also called deuterium), isotopes of oxygen including16O、17O and18isotopes of O, sulfur including32S、33S、34S and36isotopes of S, nitrogen include14N and15isotopes of N, F include17F and19isotopes of F, chlorine including35Cl and37isotopes of Cl, bromine including79Br and81Br。
"alkyl" refers to a straight or branched chain saturated aliphatic hydrocarbon group of 1 to 20 carbon atoms, preferably an alkyl group of 1 to 8 (e.g., 1,2,3,4, 5, 6,7, 8) carbon atoms, more preferably an alkyl group of 1 to 6 carbon atoms, and further preferably an alkyl group of 1 to 4 carbon atoms. Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, neo-butyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, and various branched isomers thereof; when the alkyl group is substituted, it may be optionally further substituted with 1 or more substituents. "alkoxy" refers to a group formed by replacement of at least 1 carbon atom in an alkyl group with an oxygen atom. Non-limiting examples include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentoxy, n-hexoxy, cyclopropoxy, and cyclobutoxy. The alkyl group is defined as the "alkyl" group as defined above.
"alkenyl" means a straight or branched chain unsaturated aliphatic hydrocarbon group consisting of 2 to 20 carbon atoms, preferably 2 to 12 (e.g., 2,3,4, 5, 6,7, 8,9, 10) carbon atoms, containing 1 to 10 (e.g., 1,2,3,4, 5, 6,7, 8,9, 10, 11, 12) carbon atoms, more preferably 2 to 8 carbon atoms, and still more preferably 2 to 6 carbon atoms. Non-limiting examples include vinyl, propen-2-yl, buten-2-yl, penten-4-yl, hexen-2-yl, hexen-3-yl, hepten-2-yl, hepten-3-yl, hepten-4-yl, octen-3-yl, nonen-3-yl, decen-4-yl and undecen-3-yl. The alkenyl group may be optionally further substituted with 1 or more substituents.
"alkynyl" means a straight or branched chain unsaturated aliphatic hydrocarbon group containing 1 to 10 (e.g., 1,2,3,4, 5, 6,7, 8,9, or 10) carbon-carbon triple bonds, consisting of 2 to 20 carbon atoms, preferably 2 to 12 (e.g., 2,3,4, 5, 6,7, 8,9, 10, 11, or 12) carbon atoms, more preferably 2 to 8 carbon atoms, and even more preferably 2 to 6 carbon atoms. Non-limiting examples include ethynyl, propyn-1-yl, propyn-2-yl, butyn-1-yl, butyn-2-yl, butyn-3-yl, 3-dimethylbutyn-2-yl, pentyn-1-yl, pentyn-2-yl, hexyn-1-yl, 1-heptyn-1-yl, heptyn-3-yl, heptyn-4-yl, octyn-3-yl, nonyn-3-yl, decyn-4-yl, undec-3-yl, dodecyn-4-yl. The alkynyl group may be optionally further substituted with one or more substituents.
"aryl" means a substituted or unsubstituted aromatic ring which may be a 5 to 8 membered (e.g., 5, 6,7, 8 membered) monocyclic, 5 to 12 membered (e.g., 5, 6,7, 8,9, 10, 11, 12 membered) bicyclic, or 10 to 15 membered (e.g., 10, 11, 12, 13, 14, 15 membered) tricyclic ring system which may be bridged or spiro, non-limiting examples of which include phenyl, naphthyl. The aryl group may be optionally further substituted with 1 or more substituents.
"heteroaryl" refers to a substituted or unsubstituted aromatic ring, which can be a 3 to 8 membered (e.g., 3,4, 5, 6,7, 8 membered) monocyclic, 5 to 12 membered (e.g., 5, 6,7, 8,9, 10, 11, 12 membered) bicyclic, or 10 to 15 membered (e.g., 10, 11, 12, 13, 14, 15 membered) tricyclic ring system, and contains 1 to 6 (e.g., 1,2,3,4, 5, 6) heteroatoms selected from N, O or S, preferably 5 to 8 membered heteroaryl, wherein the optionally substituted 1 to 4 (e.g., 1,2,3, 4) N, S in the ring of the heteroaryl can be oxidized to various oxidation states. Heteroaryl groups may be attached at a heteroatom or carbon atom, heteroaryl groups may be bridged or spiro, non-limiting examples include cyclic pyridyl, furyl, thienyl, pyranyl, pyrrolyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, piperidinylbenzimidazolyl, benzopyridyl, pyrrolopyridyl. Heteroaryl is optionally further substituted with 1 or more substituents.
"carbocyclyl" or "carbocycle" refers to a saturated or unsaturated aromatic or non-aromatic ring. When aromatic, it is as defined above for "aryl"; when non-aromatic, it may be a 3 to 10 membered (e.g., 3,4, 5, 6,7, 8,9, 10 membered) monocyclic, 4 to 12 membered (e.g., 4,5, 6,7, 8,9, 10, 11, 12 membered) bicyclic, or 10 to 15 membered (e.g., 10, 11, 12, 13, 14, 15 membered) tricyclic ring system, which may be bridged or spiro, non-limiting examples of which include cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopentyl-1-alkenyl, 1-cyclopentyl-2-alkenyl, 1-cyclopentyl-3-alkenyl, cyclohexyl, 1-cyclohexyl-2-alkenyl, 1-cyclohexyl-3-alkenyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl, cyclododecyl, or a pharmaceutically acceptable salt thereof,
Figure BDA0003409054530000081
Said "carbocyclyl" or "carbocycle" is optionally further substituted with 1 or more substituents.
"Heterocyclyl" or "heterocycle" refers to a saturated or unsaturated aromatic or non-aromatic heterocycle, which when aromatic is defined as "heteroaryl" as defined above; when a non-aromatic heterocycle, it may be a 3 to 10 membered (e.g. 3,4, 5, 6,7, 8,9, 10 membered) monocyclic, 4 to 12 membered (e.g. 4,5, 6,7, 8,9, 10, 11, 12 membered) bicyclic or 10 to 15 membered (e.g. 10, 11, 12, 13, 14, 15 membered) tricyclic ring system and contains 1 to 4 (e.g. 1,2,3, 4) heteroatoms selected from N, O or S, preferably a 3 to 8 membered heterocyclyl. From 1 to 4 (e.g., 1,2,3, 4) N, S optionally substituted in the ring of the "heterocyclyl" or "heterocycle" can be oxidized to various oxidation states; "heterocyclyl" or "heterocycle" may be attached at a heteroatom or carbon atom; "Heterocyclyl" or "heterocycle" may be a bridged or spiro ring. Non-limiting examples of "heterocyclyl" or "heterocycle" include epoxyethyl, epoxypropyl, aziridinyl, oxetanyl, azetidinyl, thietanyl, 1, 3-dioxolanyl, 1, 4-dioxolanyl, 1, 3-dioxanyl, azepinyl, oxepinyl, thiepinyl, oxazepinyl, diazepinyl, thiazepinyl, pyridyl, piperidyl, homopiperidinyl, furyl, thienyl, pyranyl, N-alkylpyrrolyl, pyrimidinyl, pyrazinyl, pyridazinyl, piperazinyl, homopiperazinyl, imidazolyl, piperidyl, morpholinyl, thiomorpholinyl, thialkyl, 1, 3-dithianyl, dihydrofuranyl, dithianyl, tetrahydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, tetrahydrothiopyranyl, tetrahydropyrrolyl, Tetrahydroimidazolyl, tetrahydrothiazolyl, tetrahydropyranyl, benzimidazolyl, benzopyridyl, pyrrolopyridyl, chromanyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl, 4H-pyranyl, dioxacyclohexyl, 1, 3-dioxolanyl, pyrazolinyl, dithianyl, dithienoalkyl, dihydrothienyl, pyrazolidinyl, imidazolinyl, imidazolidinyl, 1,2,3, 4-tetrahydroisoquinolinyl, 3-azabicyclo [3.1.0] hexyl, 3-azabicyclo [4.1.0] heptyl, azabicyclo [2.2.2] hexyl, 3H-indolinylquinolizinyl, N-pyridylurea, 1-dioxothiomorpholinyl, azabicyclo [3.2.1] octyl, azabicyclo [5.2.0] nonyl, oxatricyclo [5.3.1.1] dodecyl, Azaadamantyl and oxaspiro [3.3] heptanyl. Said "heterocyclyl" or "heterocycle" may optionally be further substituted with 1 or more substituents.
"cycloalkyl" refers to a saturated cyclic hydrocarbon group, the ring of which may be a 3 to 10-membered (e.g., 3,4, 5, 6,7, 8,9, 10-membered) monocyclic ring, a 4 to 12-membered (e.g., 4,5, 6,7, 8,9, 10, 11, 12-membered) bicyclic ring, or a 10 to 20-membered (e.g., 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20-membered) polycyclic ring system, the ring carbon atoms preferably being 3 to 10 carbon atoms, and more preferably 3 to 8 carbon atoms. Non-limiting examples of "cycloalkyl" include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclopropenyl, cyclobutenyl, cyclopentenyl, cyclohexenyl, cycloheptenyl, 1, 5-cyclooctadienyl, 1, 4-cyclohexadienyl, cycloheptatrienyl, and the like. When the cycloalkyl group is substituted, it may be optionally further substituted with 1 or more substituents.
"heterocycloalkyl" refers to a substituted or unsubstituted saturated nonaromatic cyclic group which may be a 3 to 8 membered (e.g., 3,4, 5, 6,7, 8 membered) monocyclic, 4 to 12 membered (e.g., 4,5, 6,7, 8,9, 10, 11, 12 membered) bicyclic, or 10 to 15 membered (e.g., 10, 11, 12, 13, 14, 15 membered) tricyclic ring system and contains 1,2 or 3 heteroatoms selected from N, O or S, preferably 3 to 8 membered heterocyclyl. Optionally substituted 1,2 or 3 of N, S in the ring of the "heterocycloalkyl" can be oxidized to various oxidation states; "heterocycloalkyl" can be attached to a heteroatom or a carbon atom; "heterocycloalkyl" can be a bridged or spiro ring. Non-limiting examples of "heterocycloalkyl" include oxiranyl, aziridinyl, oxetanyl, azetidinyl, 1, 3-dioxolanyl, 1, 4-dioxolanyl, 1, 3-dioxanyl, azepinyl, piperidinyl, perinyl, morpholinyl, thiomorpholinyl, 1, 3-dithianyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydropyrrolyl, tetrahydroimidazolyl, thiazolidinyl, tetrahydropyranyl, azabicyclo [3.2.1] octanyl, azabicyclo [5.2.0] nonanyl, oxatricyclo [5.3.1.1] dodecyl, azaadamantyl, and oxaspiro [3.3] heptanyl.
When the above-mentioned "alkyl", "alkoxy", "alkenyl", "alkynyl", "aryl", "heteroaryl", "carbocyclyl", "carbocycle", "heterocyclyl", "heterocycle", "cycloalkyl", "heterocycloalkyl" or "heterocyclyl" is substituted, it may be further substituted by 0, 1,2,3,4, 5, 6,7, 8,9 or 10 groups selected from F, Cl, Br, I, hydroxy, mercapto, nitro, cyano, amino, C1-6Alkylamino group, ═ O, C1-6Alkyl radical, C1-6Alkoxy radical、C2-6Alkenyl radical, C2-6Alkynyl, -NRq4Rq5、=NRq6、-C(=O)OC1-6Alkyl, -OC (═ O) C1-6Alkyl, -C (═ O) NRq4Rq5、C3-8Cycloalkyl radical, C3-8Heterocycloalkyl radical, C6-10Aryl radical, C5-10Heteroaryl, -C (═ O) OC6-10Aryl, -OC (═ O) C6-10Aryl, -OC (═ O) C5-10Heteroaryl, -C (═ O) OC5-10Heteroaryl, -OC (═ O) C3-8Heterocycloalkyl, -C (═ O) OC3-8Heterocycloalkyl, -OC (═ O) C3-8Cycloalkyl, -C (═ O) OC3-8Cycloalkyl, -NHC (═ O) C3-8Heterocycloalkyl, -NHC (═ O) C6-10Aryl, -NHC (═ O) C5-10Heteroaryl, -NHC (═ O) C3-8Cycloalkyl, -NHC (═ O) C3-8Heterocycloalkyl, -NHC (═ O) C2-6Alkenyl or-NHC (═ O) C2-6Alkynyl and wherein said substituent C1-6Alkyl radical, C1-6Alkoxy radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-8Cycloalkyl radical, C3-8Heterocycloalkyl radical, C6-10Aryl radical, C5-10Heteroaryl, -NHC (═ O) C6-10Aryl, -NHC (═ O) C5-10Heteroaryl, -NHC (═ O) C3-8Heterocycloalkyl or-NHC (═ O) C3-8The cycloalkyl group is optionally further substituted by 1 to 3 substituents selected from OH, F, Cl, Br, I, C1-6Alkyl radical, C1-6Alkoxy, -NRq4Rq5Or substituted with a substituent of ═ O; rq1Is selected from C1-6Alkyl radical, C1-6Alkoxy or C6-10An aryl group; rq2、Rq3Selected from H or C1-6An alkyl group; wherein R isq4、Rq5Selected from H, C1-6Alkyl, -NH (C ═ NR)q1)NRq2Rq3、-S(=O)2NRq2Rq3、-C(=O)Rq1or-C (═ O) NRq2Rq3Wherein said C1-6The alkyl is optionally further substituted by 1 or more groups selected from OH, F, Cl, Br, I, C1-6Alkyl radical, C1-6Alkoxy radical, C6-10Aryl radical, C5-10Heteroaryl group, C3-8Cycloalkyl or C3-8Substituted with a substituent of heterocycloalkyl; or Rq4And Rq5And the N atom forms a 3 to 8 membered heterocyclic ring which may contain 1 or more heteroatoms selected from N, O or S.
By "pharmaceutically acceptable salt" or "pharmaceutically acceptable salt thereof" is meant a salt of a compound of the invention that retains the biological effectiveness and properties of the free acid or free base obtained by reaction with a non-toxic inorganic or organic base, and the free base obtained by reaction with a non-toxic inorganic or organic acid.
"pharmaceutical composition" refers to a mixture of one or more compounds described herein, pharmaceutically acceptable salts or prodrugs thereof, and other chemical components, wherein "other chemical components" refers to pharmaceutically acceptable carriers, excipients, and/or one or more other therapeutic agents.
By "carrier" is meant a material that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound.
"excipient" refers to an inert substance added to a pharmaceutical composition to facilitate administration of a compound. Non-limiting examples include calcium carbonate, calcium phosphate, sugars, starches, cellulose derivatives (including microcrystalline cellulose), gelatin, vegetable oils, polyethylene glycols, diluents, granulating agents, lubricants, binders, and disintegrating agents.
By "prodrug" is meant a compound of the invention that is metabolically convertible in vivo to a biologically active compound. Prodrugs of the invention are prepared by modifying an amino or carboxyl group in a compound of the invention, which modification may be removed by routine manipulation or in vivo, to yield the parent compound. When a prodrug of the present invention is administered to a mammalian subject, the prodrug is cleaved to form a free amino or carboxyl group.
"cocrystal" refers to a crystal of an Active Pharmaceutical Ingredient (API) and a cocrystal former (CCF) bound by hydrogen bonding or other non-covalent bonds, wherein the API and CCF are both solid in their pure state at room temperature and a fixed stoichiometric ratio exists between the components. A co-crystal is a multi-component crystal that contains both a binary co-crystal formed between two neutral solids and a multicomponent co-crystal formed between a neutral solid and a salt or solvate.
"stereoisomers" refers to isomers resulting from the different arrangement of atoms in a molecule, including cis, trans isomers, enantiomers and conformational isomers.
"optional" or "optionally" or "selective" or "selectively" means that the subsequently described event or circumstance may, but need not, occur, and that the description includes instances where the event or circumstance occurs and instances where it does not. For example, "a heterocyclic group optionally substituted with an alkyl group" means that the alkyl group may, but need not, be present, and the description includes the case where the heterocyclic group is substituted with an alkyl group, and the case where the heterocyclic group is not substituted with an alkyl group.
Detailed Description
The following examples illustrate the technical solutions of the present invention in detail, but the scope of the present invention includes but is not limited thereto.
The structure of the compounds is determined by Nuclear Magnetic Resonance (NMR) or (and) Mass Spectrometry (MS). NMR shift (. delta.) of 10-6The units in (ppm) are given. NMR was measured using (Bruker Avance III 400 and Bruker Avance 300) nuclear magnetic spectrometers in deuterated dimethyl sulfoxide (DMSO-d)6) Deuterated chloroform (CDCl)3) Deuterated methanol (CD)3OD), internal standard Tetramethylsilane (TMS).
MS was measured by Agilent 6120B (ESI) and Agilent 6120B (APCI).
HPLC was carried out using an Agilent 1260DAD high pressure liquid chromatograph (Zorbax SB-C18100X 4.6mm, 3.5. mu.M).
The thin layer chromatography silica gel plate is HSGF254 of tobacco yellow sea or GF254 of Qingdao, the specification of silica gel plate used by Thin Layer Chromatography (TLC) is 0.15mm-0.20mm, and the specification of thin layer chromatography separation and purification product is 0.4mm-0.5 mm.
The column chromatography generally uses 200-mesh and 300-mesh silica gel of the Tibet yellow sea silica gel as a carrier.
Intermediate 1:
4-chloro-6- (2-isopropyl-4-methylpyridin-3-yl) -3, 6-dihydrofuran [2',3':4,5] pyridine [2,3-d ] pyrimidine-7, 9(2H,8H) -dione (intermediate 1)
4-chloro-6-(2-isopropyl-4-methylpyridin-3-yl)-3,6-dihydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidine-7,9(2H,8H)-dione
Figure BDA0003409054530000111
The first step is as follows:
3, 5-dihydrofuro [3,2-c ] pyridin-4 (2H) -one (3b)
3,5-dihydrofuro[3,2-c]pyridin-4(2H)-one
3a (5g, 37mmol) was dissolved in a mixed solvent of 50mL of methanol and 50mL of acetic acid, and palladium on carbon (3.5g) was added thereto, followed by reaction at 70 ℃ for 4 hours under normal pressure of hydrogen. The reaction was filtered, the filter cake was washed with 100mL of ethyl acetate, and the mother liquor was concentrated under reduced pressure to give compound 3b (white solid, 5g, 98% yield).
1H NMR(400MHz,DMSO-d6)δ11.56(s,1H),7.27(d,1H),5.98(d,1H),4.58(t,2H),2.88(t,2H)。
LC-MS m/z(ESI)=138.10[M+1]。
The second step is that:
4-chloro-2, 3-dihydrofuran [3,2-c ] pyridine (3c)
4-chloro-2,3-dihydrofuro[3,2-c]pyridine
3b (5g, 36.2mmol) was added slowly to phosphorus oxychloride (50mL) in an ice water bath and refluxed under nitrogen for 2 hours. The reaction mixture was directly concentrated under reduced pressure, diluted with ethyl acetate (20mL), slowly added dropwise to a saturated aqueous sodium carbonate solution (200mL), extracted with ethyl acetate, the combined organic phases were dried over anhydrous sodium sulfate, concentrated, and purified by silica gel column chromatography (petroleum ether: ethyl acetate ═ 20:1-5:1) to give compound 3c (white solid, 2.2g, yield 40%).
1H NMR(400MHz,DMSO-d6)δ8.09(d,1H),6.89(d,1H),4.71(t,2H),3.23(t,2H)。
LC-MS m/z(ESI)=156.10[M+1]。
The third step:
4-chloro-2, 3-dihydrofuran [3,2-c ] pyridine 5-oxide (3d)
4-chloro-2,3-dihydrofuro[3,2-c]pyridine 5-oxide
3c (2.2g, 14.1mmol) was dissolved in dichloromethane (20mL), m-chloroperoxybenzoic acid (7.3g, 42.3mmol) was added and the mixture was refluxed for 4 hours under nitrogen. The reaction was filtered and concentrated under reduced pressure to give compound 3d (white solid, 2g, 83% yield).
LC-MS m/z(ESI)=172.10[M+1]。
The fourth step:
4, 6-dichloro-2, 3-dihydrofuro [3,2-c ] pyridine (3e)
4,6-dichloro-2,3-dihydrofuro[3,2-c]pyridine
Phosphorus oxychloride (20mL) was added slowly to 3d (2g, 11.6mmol) in an ice-water bath and refluxed for 4 hours under nitrogen. The reaction mixture was directly concentrated under reduced pressure, diluted with ethyl acetate (10mL), slowly added dropwise to a saturated aqueous sodium carbonate solution (100mL), extracted with ethyl acetate, the combined organic phases were dried over anhydrous sodium sulfate and concentrated, and purified by silica gel column chromatography (petroleum ether: ethyl acetate ═ 20:1-4:1) to give compound 3e (white solid, 1.4g, yield 64%).
1H NMR(400MHz,DMSO-d6)δ7.07(s,1H),4.79(t,2H),3.22(t,2H)。
LC-MS m/z(ESI)=190.00[M+1]。
The fifth step:
4, 6-dichloro-2, 3-dihydrofuran [3,2-c ] pyridine-7-carboxylic acid (intermediate 1)
4,6-dichloro-2,3-dihydrofuro[3,2-c]pyridine-7-carboxylic acid
3e (1.4g, 7.4mmol) was dissolved in tetrahydrofuran (14mL), cooled in a dry ice acetone bath to-78 deg.C, n-butyllithium (5mL,1.6mol/L) was slowly added dropwise, the temperature was controlled below-70 deg.C for 1 hour, then dry ice was added thereto, and the reaction was slowly returned to room temperature for 1 hour. The reaction solution was slowly added dropwise to a saturated aqueous ammonium chloride solution (100mL), extracted with ethyl acetate, combined organic phases were dried and concentrated, and then petroleum ether: slurried ethyl acetate 5:1(10mL) and filtered to give intermediate 1 (white solid, 1.2g, 71% yield).
1H NMR(400MHz,DMSO-d6)δ13.98(s,1H),4.86(t,2H),3.26(t,2H)。
LC-MS m/z(ESI)=233.90[M+1]。
Intermediate 2:
Figure BDA0003409054530000121
the first step is as follows:
4, 6-dichloro-2, 3-dihydrofuran [3,2-c ] pyridine-7-carboxamide (3f)
4,6-dichloro-2,3-dihydrofuro[3,2-c]pyridine-7-carboxamide
Intermediate 1(1.8g, 7.8mmol) was added slowly dropwise thionyl chloride (20mL) in an ice-water bath, followed by reflux for 2 h. The reaction solution was concentrated and dissolved in tetrahydrofuran (2mL), and slowly added dropwise to ammonia water in an ice water bath: dichloromethane ═ 1:1(20mL), and the reaction was carried out at room temperature for 30 minutes. Concentration by extraction with dichloromethane afforded 4, 6-dichloro-2, 3-dihydrofuran [3,2-c ] pyridine-7-carboxamide 3f (1.7g, 96% yield).
1H NMR(400MHz,DMSO-d6)δ7.98(s,1H),7.82(s,1H),4.82(t,2H),3.26(t,2H)。
LC-MS m/z(ESI)=233.00[M+1]。
The second step is that:
4, 6-dichloro-N- (((2-isopropyl-4-methylpyridin-3-yl) carbamoyl) -2, 3-dihydrofuran [3,2-c ] pyridine-7-carboxamide (3g)
4,6-dichloro-N-((2-isopropyl-4-methylpyridin-3-yl)carbamoyl)-2,3-dihydrofuro[3,2-c]pyridine-7-carboxamide
2-isopropyl-4-methylpyridin-3-amine (1.2g, 8.0mmol) was dissolved in tetrahydrofuran, N-diisopropylethylamine (2.1g, 16mmol) was added thereto at room temperature, followed by stirring for 10 minutes, then chloroformic acid-2, 2, 2-trichloroethyl ester (2.5g, 12mmol) was slowly added dropwise, reacted for 10 minutes, and concentrated under reduced pressure. Dissolved in tetrahydrofuran (10mL) and added slowly dropwise to a solution of 3f (1.7g, 7.3mmol) and sodium hydride (60%) (584mg, 14.5mmol) in tetrahydrofuran (15mL) and allowed to react for 10 min. The reaction mixture was slowly added dropwise to saturated ammonium chloride (100mL), extracted with ethyl acetate, and concentrated. Purifying petroleum ether by column chromatography: gradient elution with ethyl acetate 6:1-1:1 afforded 3g (2.8g, 94% yield) of compound.
1H NMR(400MHz,DMSO-d6)δ11.31(s,1H),9.70(s,1H),8.33(d,1H),7.16(d,1H),4.89(m,2H),3.28(m,3H),2.19(d,3H),1.16(d,6H)。
LC-MS m/z(ESI)=409.10[M+1]。
The third step:
4-chloro-6- (2-isopropyl-4-methylpyridin-3-yl) -3, 6-dihydrofuran [2',3':4,5] pyridine [2,3-d ] pyrimidine-7, 9(2H,8H) -dione (intermediate 2)
4-chloro-6-(2-isopropyl-4-methylpyridin-3-yl)-3,6-dihydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidine-7,9(2H,8H)-dione
3g (2.8g, 7.5mmol) was dissolved in tetrahydrofuran (20mL) and potassium bis (trimethylsilyl) amide (15mL,15mmol,1mol/L) was slowly added dropwise under ice-water bath and refluxed for 2 hours. Slowly dripping the reaction solution into 200mL of saturated ammonium chloride aqueous solution, extracting and concentrating by ethyl acetate, and purifying petroleum ether by column chromatography: gradient elution with ethyl acetate 4:1-1:1 afforded intermediate 2 (white solid, 1.5g, 60% yield).
1H NMR(400MHz,DMSO-d6)δ11.90(s,1H),8.53(d,1H),7.28(d,1H),4.93(t,2H),3.18(t,2H),2.88(m,1H),2.10(s,3H),1.09(d,6H)。
LC-MS m/z(ESI)=391.10[M+1]。
Example 1
1- ((2S,5R) -4-acryloyl-2, 5-dimethylpiperazin-1-yl) -6- (2-fluoro-6-hydroxyphenyl) -4- (2-isopropyl-4-methylpyridin-3-yl) -4,7,8,9 tetrahydro-3H-cyclopenta [4,5] pyrido [2,3-d ] pyrimidin-3-one (Compound 1)
1-((2S,5R)-4-acryloyl-2,5-dimethylpiperazin-1-yl)-6-(2-fluoro-6-hydroxyphenyl)-4-(2-isopropyl-4-methylpyridin-3-yl)-4,7,8,9-tetrahydro-3H-cyclopenta[4,5]pyrido[2,3-d]pyrimidin-3-one
Figure BDA0003409054530000131
Figure BDA0003409054530000141
First step of
1, 3-dichloro-6, 7-dihydro-5H-cyclopenta [ c ] pyridine-4-carboxamide (1b)
1,3-dichloro-6,7-dihydro-5H-cyclopenta[c]pyridine-4-carboxamide
Compound 1a (2g, 9.4mmol) was dissolved in 30ml of aqueous sulfuric acid (v: v ═ 5:1) and stirred at 85 ℃ under reflux for 3 hours. The reaction was cooled to 0 ℃, quenched by dropwise addition to aqueous potassium carbonate, the pH of the solution was adjusted to neutral, and extracted with ethyl acetate (20mL × 4). The combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated to give 2.23g of crude product. Column chromatography gave compound 1b (white solid, 1.8g, 82% yield).
1H NMR(400MHz,DMSO-d6)δ8.03(s,1H),7.86(s,1H),2.97(t,2H),2.93(t,2H),2.12(p,2H);
LCMS m/z(ESI)=231.0[M+1]。
Second step of
1, 3-dichloro-N- (((2-isopropyl-4-methylpyridin-3-yl) carbamoyl) -6, 7-dihydro-5H-cyclopenta [ c ] pyridine-4-carboxamide (1c)
1,3-dichloro-N-((2-isopropyl-4-methylpyridin-3-yl)carbamoyl)-6,7-dihydro-5H-cyclopenta[c]pyridine-4-carboxamide
2-isopropyl-4-methylpyridin-3-amine (1g, 4.3mmol) was dissolved in tetrahydrofuran and N, N-diisopropylethylamine (1.11g, 8.6mmol) and 2,2, 2-trichloroethyl chloroformate (1.36g, 6.45mmol) were added. Stirring for 30 minutes at normal temperature. The reaction was quenched with 20ml of water, extracted with ethyl acetate, dried over anhydrous sodium sulfate, filtered, concentrated, and the solid was dissolved in 10ml of anhydrous tetrahydrofuran. Add tetrahydrofuran dissolved in compound 1b and cool to 0 ℃ and add sodium hydride (258mg, 6.45 mmol). Stir for 1.5 hours. The reaction solution was quenched by adding 50mL of a saturated aqueous ammonium chloride solution. Extraction with ethyl acetate (20mL 4), filtration and concentration gave 1.2g of crude product. Column chromatography gave compound 1c (yellow solid, 900mg, yield: 51%)
1H NMR(400MHz,DMSO-d6)δ11.38(s,1H),9.73(s,1H),8.34(d,1H),7.16(d,,1H),3.27(h,1H),3.09-2.83(m,4H),2.24-2.06(m,5H),1.17(d,6H)。
LCMS m/z(ESI)=407.1[M+1]
The third step
6-chloro-4- (2-isopropyl-4-methylpyridin-3-yl) -4,7,8, 9-tetrahydro-1H-cyclopenta [4,5] pyrido [2,3-d ] pyrimidine-1, 3(2H) -dione (1d)
6-chloro-4-(2-isopropyl-4-methylpyridin-3-yl)-4,7,8,9-tetrahydro-1H-cyclopenta[4,5]pyrido[2,3-d]pyrimidine-1,3(2H)-dione
Compound 1c (900mg, 2.2mmol) was dissolved in tetrahydrofuran, cooled to 0 ℃ and potassium bis (trimethylsilyl) amide (4.4mL,4.4mmol) was added. And heated to 75 deg.c and stirred for 3 hr. The reaction was cooled to room temperature, quenched with water, adjusted to neutral pH, extracted with ethyl acetate (10mL x 4), dried over anhydrous sodium sulfate, filtered, and concentrated to give 1g of crude product. Column chromatography gave compound 1d (white solid, 720mg, 88%)
1H NMR(400MHz,DMSO-d6)δ9.43(s,1H),8.46(d,1H),7.30(d,1H),3.40(t,2H),2.62(t,2H),2.52(p,1H),2.38(p,2H),2.21(s,3H),1.23(d,6H)。
LCMS m/z(ESI)=371.2[M+1]。
The fourth step
Tert-butyl (2R,5S) -4- (6-chloro-4- (2-isopropyl-4-methylpyridin-3-yl) -3-oxo-4, 7,8, 9-tetrahydro-3H-cyclopenta [4,5] pyrido [2,3-d ] pyrimidin-1-yl) -2, 5-dimethylpiperazine-1-carboxylic acid (1e)
tert-butyl(2R,5S)-4-(6-chloro-4-(2-isopropyl-4-methylpyridin-3-yl)-3-oxo-4,7,8,9-tetrahydro-3H-cyclopenta[4,5]pyrido[2,3-d]pyrimidin-1-yl)-2,5-dimethylpiperazine-1-carboxylate
Adding a compound 1d (300mg,0.81mmol) into a flask, adding N, N-diisopropylethylamine (523mg,4.05mmol) and phosphorus oxychloride (0.75mL,8.1mmol) under the protection of nitrogen, dropwise adding at 0 ℃, stirring at 85 ℃ to react, concentrating the reaction solution after 2h, adding 5mL of tetrahydrofuran solution to dissolve, adding N, N-diisopropylethylamine (523mg,4.05mmol) and (2R,5S) -2, 5-dimethylpiperazine-1-carboxylic acid tert-butyl ester (173.3mg, 0.81mmol) at low temperature, stirring at room temperature to react, adding ethyl acetate and water to extract after 0.5h, drying the organic phase with anhydrous sodium sulfate, concentrating to obtain a crude product of 320mg, and performing column chromatography to obtain a compound 1e (yellow solid, 290mg, 63% yield)
1H NMR(400MHz,DMSO-d6)δ8.62(d,1H),7.27(d,1H),4.05(h,1H),3.94(dd,1H),3.70(dd,1H),3.66-3.57(m,1H),3.52(dd,1H),3.50-3.35(m,2H),3.32-3.16(m,2H),3.02-2.91(m,1H),2.90-2.78(m,1H),2.33-2.23(m,1H),2.28-2.15(m,1H),2.15(s,3H),1.47(s,9H),1.14-1.01(m,9H),0.99(d,3H)。
LCMS m/z(ESI)=567.3[M+1]。
The fifth step
Tert-butyl (2R,5S) -4- (6- (2-fluoro-6-hydroxyphenyl) -4- (2-isopropyl-4-methylpyridin-3-yl) -3-oxo-4, 7,8, 9-tetrahydro-3H-cyclopenta [4,5] pyridin [2,3-d ] pyrimidin-1-yl) -2, 5-dimethylpiperazine-1-carboxylic acid (1f)
tert-butyl(2R,5S)-4-(6-(2-fluoro-6-hydroxyphenyl)-4-(2-isopropyl-4-methylpyridin-3-yl)-3-oxo-4,7,8,9-tetrahydro-3H-cyclopenta[4,5]pyrido[2,3-d]pyrimidin-1-yl)-2,5-dimethylpiperazine-1-carboxylate
Compound 1e (230mg, 0.41mmol), (2-fluoro-6-hydroxyphenyl) boronic acid (75.9mg, 0.48mmol), tetrakis (triphenylphosphine) palladium (93.83mg, 0.10mmol) and sodium acetate (159.96mg, 1.63mmol) were dissolved in 6mL of 1, 4-dioxane and 2mL of water and stirred at 100 ℃ for 2 hours under nitrogen. The reaction was quenched with 20mL of water, extracted with ethyl acetate (30mL x 3), dried over anhydrous sodium sulfate, filtered and concentrated. Column chromatography analysis gave compound 1f (yellow solid, 210mg, 81% yield).
1H NMR(400MHz,DMSO-d6)δ8.62(d,1H),8.19(s,1H),7.37-7.28(m,2H),7.30-6.90(m,1H),6.70(dd,1H),4.31(p,1H),4.11(dd,1H),3.74-3.60(m,2H),3.52-3.25(m,4H),3.18(dd,1H),3.08(dd,1H),3.01-2.86(m,1H),2.35-2.3(m,1H),2.24-2.12(m,4H),1.47(s,9H),1.13(dd,6H),0.99(dd,6H)。
LCMS m/z(ESI)=643.3[M+1]。
The sixth step
1- ((2S,5R) -4-acryloyl-2, 5-dimethylpiperazin-1-yl) -6- (2-fluoro-6-hydroxyphenyl) -4- (2-isopropyl-4-methylpyridin-3-yl) -4,7,8,9 tetrahydro-3H-cyclopenta [4,5] pyrido [2,3-d ] pyrimidin-3-one (Compound 1)
1-((2S,5R)-4-acryloyl-2,5-dimethylpiperazin-1-yl)-6-(2-fluoro-6-hydroxyphenyl)-4-(2-isopr opyl-4-methylpyridin-3-yl)-4,7,8,9-tetrahydro-3H-cyclopenta[4,5]pyrido[2,3-d]pyrimidin-3-one
Tert-butyl (2R,5S) -4- (6- (2-fluoro-6-hydroxyphenyl) -4- (2-isopropyl-4-methylpyridin-3-yl) -3-oxo-4, 7,8, 9-tetrahydro-3H-cyclopenta [4,5] pyridin [2,3-d ] pyrimidin-1-yl) -2, 5-dimethylpiperazine-1-carboxylic acid 1i (210mg, 0.33mmol) was dissolved in 4mL of dichloromethane, 1mL of trifluoroacetic acid was added dropwise at 0 ℃ after 1H, after which 2mL of methanol and sodium bicarbonate solid powder were added to adjust the pH to neutral, 5mL of dichloromethane was added to the reaction system, and the mixture was filtered and concentrated to dryness. 5mL of N, N-dimethylformamide was added to dissolve the mixture, acrylic acid (28.5mg, 0.4mmol) N, N-diisopropylethylamine (127.7mg,0.99mmol) and 1-propylphosphoric anhydride (50% N, N-dimethylformamide) (315mg, 0.495mmol) were added at 0 ℃ under nitrogen protection, the dropwise addition was completed, the mixture was stirred at room temperature, the reaction was completed after 2 hours, and the reaction solution was dried by spinning to obtain the objective compound 1 in a yield of 20% as a white solid (39 mg).
LCMS m/z(ESI)=597.3[M+1]
Example 2
1- (4-acryloyl-2-methylpiperazin-1-yl) -6- (2-fluoro-6-hydroxyphenyl) -4- (2-isopropyl-4-methylpyridin-3-yl) -4,7,8, 9-tetrahydro-3H-cyclopentyl [4,5] pyridinyl [2,3-d ] pyrimidin-3-one (compound 2)
1-(4-acryloyl-2-methylpiperazin-1-yl)-6-(2-fluoro-6-hydroxyphenyl)-4-(2-isopropyl-4-methylpyridin-3-yl)-4,7,8,9-tetrahydro-3H-cyclopenta[4,5]pyrido[2,3-d]pyrimidin-3-one
Figure BDA0003409054530000161
The first step is as follows:
4- (6-chloro-4- (2-isopropyl-4-methylpyridin-3-yl) -3-oxo-4, 7,8, 9-tetrahydro-3H-cyclopentyl [4,5] pyridin [2,3-d ] pyrimidin-1-yl) -3-methylpiperazine 1-carboxylic acid tert-butyl ester (2b)
tert-butyl-4-(6-chloro-4-(2-isopropyl-4-methylpyridin-3-yl)-3-oxo-4,7,8,9-tetrahydro-3H-cyclopenta[4,5]pyrido[2,3-d]pyrimidin-1-yl)-3-methylpiperazine-1-carboxylate
Compound 2a (300mg,0.81mmol) was added to a flask, N-diisopropylethylamine (523mg,4.05mmol) and phosphorus oxychloride (0.75mL,8.1mmol) were added dropwise at 0 ℃ and stirred at 85 ℃ to react, after 2h the reaction was concentrated, then 5mL tetrahydrofuran solution was added to dissolve, N-diisopropylethylamine (523mg,4.05mmol) and tert-butyl 3-methylpiperazine-1-carboxylate (162mg,0.81mmol) were added at low temperature and stirred to react after 0.5h, ethyl acetate and water were added to extract, the organic phase was dried over anhydrous sodium sulfate, the organic phase was spin-dried, and column chromatography was performed (petroleum ether: ethyl acetate ═ 1: 1) to give compound 2b (brown product, 280mg, yield 62.6%).
1H NMR(400MHz,DMSO-d6)δ8.62(d,1H),7.27(d,1H),4.05(h,1H),3.94(dd,1H),3.70(dd,1H),3.66-3.57(m,1H),3.52(dd,1H),3.50-3.35(m,2H),3.32-3.16(m,2H),3.02-2.91(m,1H),2.90-2.78(m,1H),2.33-2.23(m,1H),2.28-2.15(m,1H),2.15(s,3H),1.47(s,9H),1.14-1.01(m,6H),0.99(d,3H)。
LCMS m/z(ESI)=553.30[M+1]
The second step is that:
4- (6- (2-fluoro-6-hydroxyphenyl) -4- (2-isopropyl-4-methylpyridin-3-yl) -3-oxo-4, 7,8, 9-tetrahydro-3H-cyclopentyl [4,5] pyridin [2,3-d ] pyrimidin-1-yl) -3-methylpiperazine-1-carboxylic acid tert-butyl ester (2c)
tert-butyl4-(6-(2-fluoro-6-hydroxyphenyl)-4-(2-isopropyl-4-methylpyridin-3-yl)-3-oxo-4,7,8,9-tetrahydro-3H-cyclopenta[4,5]pyrido[2,3-d]pyrimidin-1-yl)-3-methylpiperazine-1-carboxylate
Compound 2b (280mg, 0.51mmol), (2-fluoro-6-hydroxyphenyl) boronic acid (95mg, 0.61mmol), [1,1' -bis (diphenylphosphino) ferrocene ] dichloropalladium (74.6mg, 0.10mmol) and potassium acetate (250mg, 2.55mmol) were dissolved in 4mL of 1, 4-dioxane and 1mL of water, the reaction was stirred at 100 ℃ under nitrogen, after 2h, 20mL of water and ethyl acetate (3 × 30mL) were added and extracted three times, the organic phase was collected, dried and analyzed by column chromatography (petroleum ether: ethyl acetate ═ 1: 1) to give the desired product 2c (pale yellow oil, 260mg, 82% yield).
1H NMR(400MHz,DMSO-d6)δ8.62(d,1H),8.19(s,1H),7.37-7.28(m,2H),7.30-6.90(m,1H),6.70(dd,1H),4.31(p,1H),4.11(dd,1H),3.74-3.60(m,2H),3.52-3.25(m,4H),3.18(dd,1H),3.08(dd,1H),3.01-2.86(m,1H),2.35-2.3(m,1H),2.24-2.12(m,4H),1.47(s,9H),1.13-1.11(m,3H),0.99(dd,6H)。
LCMS m/z(ESI)=629.30[M+1]
The third step:
1- (4-acryloyl-2-methylpiperazin-1-yl) -6- (2-fluoro-6-hydroxyphenyl) -4- (2-isopropyl-4-methylpyridin-3-yl) -4,7,8,9 tetrahydro-3H-cyclopenta [4,5] pyrido [2,3-d ] pyrimidin-3-one (Compound 2)
1-(4-acryloyl-2-methylpiperazin-1-yl)-6-(2-fluoro-6-hydroxyphenyl)-4-(2-isopropyl-4-methylpyridin-3-yl)-4,7,8,9-tetrahydro-3H-cyclopenta[4,5]pyrido[2,3-d]pyrimidin-3-one
Dissolving a compound 2c (260mg, 0.64mmol) in 4mL of dichloromethane, dropwise adding 1mL of trifluoroacetic acid at 0 ℃, after 1h of reaction, adding 2mL of methanol and sodium bicarbonate solid powder to adjust the pH to be neutral, adding 5mL of dichloromethane in the reaction system, filtering, spin-drying the filtrate, adding 3mL of N, N-dimethylformamide to dissolve, adding acrylic acid (46mg, 0.64mmol) under nitrogen protection, adding N, N-diisopropylethylamine (413mg,3.2mmol) and 1-propylphosphoric anhydride (244mg, 0.768mmol) at 0 ℃, after dropwise addition, stirring at room temperature, after 2h of reaction, spin-drying the reaction solution to prepare and isolate a target compound 2 (20 mg of white solid, 5% yield).
1H NMR(400MHz,DMSO-d6)δ8.62(d,1H),8.19(s,1H),7.37-7.28(m,2H),7.30-6.90(m,1H),6.70(dd,1H),6.62-6.64(m,1H),6.04-6.01(m,1H),5.58-5.42(m,1H),4.31(p,1H),4.11(dd,1H),3.74-3.60(m,2H),3.52-3.25(m,4H),3.18(dd,1H),3.08(dd,1H),3.01-2.86(m,1H),2.35-2.3(m,1H),2.24-2.12(m,4H),1.21-1.01(m,3H),0.99(m,6H)。
LCMS m/z(ESI)=583.30[M+1]
Example 3
9- ((2S,5R) -4-acryloyl-2, 5-dimethylpiperazin-1-yl) -6- (2-isopropyl-4-methylpyridin-3-yl) -4- (5-methyl-1H-indazol-4-yl) -3, 6-dihydrofuro [2',3':4,5] pyrido [2,3-d ] pyrimidin-7 (2H) -one (Compound 3)
9-((2S,5R)-4-acryloyl-2,5-dimethylpiperazin-1-yl)-6-(2-isopropyl-4-methylpyridin-3-yl)-4-(5-methyl-1H-indazol-4-yl)-3,6-dihydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-7(2H)-one
Figure BDA0003409054530000181
The first step is as follows:
tert-butyl (2R,5S) -4- (4-chloro-6- (2-isopropyl-4-methylpyridin-3-yl) -7-oxo-2, 3,6, 7-tetrahydrofuran [2',3':4,5] pyrido [2,3-d ] pyrimidin-9-yl) -2, 5-dimethylpiperazine-1-carbox-ylic acid (3h)
tert-butyl(2R,5S)-4-(4-chloro-6-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-2,3,6,7-tetrahydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-9-yl)-2,5-dimethylpiperazine-1-carboxylate
Intermediate 2(1g, 2.6mmol) was dissolved in acetonitrile (10mL), N-diisopropylethylamine (1.66g, 12.8mmol) was added under ice-water bath, and phosphorus oxychloride (10mL) was slowly added dropwise and refluxed for 2 hours. The reaction mixture was concentrated under reduced pressure, dissolved in tetrahydrofuran, and slowly added dropwise to tetrahydrofuran of (2R,5S) -tert-butyl 2, 5-dimethyl-1-piperazinecarboxylate (0.6g, 2.8mmol) and N, N-diisopropylethylamine (1.66g, 12.8mmol) in an ice-water bath to react at room temperature for 2 hours. The reaction was added dropwise to a saturated aqueous sodium bicarbonate solution (50mL), extracted with ethyl acetate, concentrated, and purified by column chromatography to dichloromethane: gradient elution with ethyl acetate 4:1-1:2 afforded compound 3h (1.2g, 83%).
LC-MS m/z(ESI)=569.30[M+1]。
The second step is that:
tert-butyl (2R,5S) -4- (6- (2-isopropyl-4-methylpyridin-3-yl) -4- (5-methyl-1H-indazol-4-yl) -7-oxo-2, 3,6, 7-tetrahydrofuran [2',3':4,5] pyridine [2,3-d ] pyrimidin-9-yl) -2, 5-dimethylpiperazine-1-carboxylic acid (3i)
tert-butyl(2R,5S)-4-(6-(2-isopropyl-4-methylpyridin-3-yl)-4-(5-methyl-1H-indazol-4-yl)-7-oxo-2,3,6,7-tetrahydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-9-yl)-2,5-dimethylpiperazine-1-carboxylate
Dissolve 3H (0.3g, 0.51mmol), 5-methyl-1H-indazole-4-boronic acid (112mg, 0.63mmol), sodium carbonate (224mg, 2.1mmol), palladium tetrakistriphenylphosphine (61mg, 0.05mmol) in 1, 4-dioxane: water 4: 1(5mL) at 100 ℃ for 4 h. Quenching with 50mL of saturated aqueous sodium bicarbonate solution, extracting with ethyl acetate, concentrating, purifying by column chromatography with dichloromethane: methanol 80: 1-10: gradient elution 1 afforded compound 3i (white solid, 300mg, 86% yield).
LC-MS m/z(ESI)=665.40[M+1]。
The third step:
9- ((2S,5R) -4-acryloyl-2, 5-dimethylpiperazin-1-yl) -6- (2-isopropyl-4-methylpyridin-3-yl) -4- (5-methyl-1H-indazol-4-yl) -3, 6-dihydrofuro [2',3':4,5] pyrido [2,3-d ] pyrimidin-7 (2H) -one (Compound 3)
9-((2S,5R)-4-acryloyl-2,5-dimethylpiperazin-1-yl)-6-(2-isopropyl-4-methylpyridin-3-yl)-4-(5-methyl-1H-indazol-4-yl)-3,6-dihydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-7(2H)-one
3i (300mg, 0.45mmol) was dissolved in dichloromethane (4mL), and trifluoroacetic acid (1mL) was slowly added dropwise under an ice-water bath, followed by reaction at room temperature for 30 minutes. After concentration under reduced pressure, the resulting mixture was dissolved in N, N-dimethylformamide (3mL), N-diisopropylethylamine (466mg, 3.6mmol) was added dropwise, and 1-propylphosphoric anhydride (50%) (431, 0.68mmol) was slowly added dropwise under an ice-water bath, and the reaction was carried out at room temperature for 2 hours. The reaction mixture was concentrated under reduced pressure to give compound 3 (white solid, 60mg, yield 21%).
LC-MS m/z(ESI)=619.30[M+1]。
Example 4
9- ((2S,5R) -4-acryloyl-2, 5-dimethylpiperazin-1-yl) -4- (2-fluoro-6-hydroxyphenyl) -6- (2-isopropyl-4-methylpyridin-3-yl) -3, 6-dihydrofuro [2',3':4,5] pyrido [2,3-d ] pyrimidin-7 (2H) -one (Compound 4)
9-((2S,5R)-4-acryloyl-2,5-dimethylpiperazin-1-yl)-4-(2-fluoro-6-hydroxyphenyl)-6-(2-isopropyl-4-methylpyridin-3-yl)-3,6-dihydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-7(2H)-one
Figure BDA0003409054530000191
The first step is as follows:
9- ((2S,5R) -4-acryloyl-2, 5-dimethylpiperazin-1-yl) -4- (2-fluoro-6-hydroxyphenyl) -6- (2-isopropyl-4-methylpyridin-3-yl) -3, 6-dihydrofuro [2',3':4,5] pyrido [2,3-d ] pyrimidin-7 (2H) -one (4a)
9-((2S,5R)-4-acryloyl-2,5-dimethylpiperazin-1-yl)-4-(2-fluoro-6-hydroxyphenyl)-6-(2-isopropyl-4-methylpyridin-3-yl)-3,6-dihydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-7(2H)-one
Dissolve 3h (0.3g, 0.51mmol), 2-fluoro-6-hydroxyphenylboronic acid (98.6mg, 0.63mmol), sodium carbonate (224mg, 2.1mmol), palladium tetrakistriphenylphosphine (61mg, 0.05mmol) in 1, 4-dioxane: water-4: 1(5mL) at 100 ℃ for 4 h. Quenching with 50mL of saturated aqueous sodium bicarbonate solution, extracting with ethyl acetate, concentrating, purifying by column chromatography with dichloromethane: methanol 80: 1-10: gradient elution of 1 afforded compound 4a (white solid, 290mg, 85% yield).
LC-MS m/z(ESI)=645.30[M+1]。
The second step is that:
9- ((2S,5R) -4-acryloyl-2, 5-dimethylpiperazin-1-yl) -4- (2-fluoro-6-hydroxyphenyl) -6- (2-isopropyl-4-methylpyridin-3-yl) -3, 6-dihydrofuro [2',3':4,5] pyrido [2,3-d ] pyrimidin-7 (2H) -one (Compound 4)
9-((2S,5R)-4-acryloyl-2,5-dimethylpiperazin-1-yl)-4-(2-fluoro-6-hydroxyphenyl)-6-(2-isopropyl-4-methylpyridin-3-yl)-3,6-dihydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-7(2H)-one
4a (290mg, 0.45mmol) was dissolved in dichloromethane (4mL), and trifluoroacetic acid (1mL) was slowly added dropwise under an ice-water bath, followed by reaction at room temperature for 30 minutes. After concentration under reduced pressure, the resulting solution was dissolved in N, N-dimethylformamide (3mL), N-diisopropylethylamine (466mg, 3.6mmol) was added dropwise, and 1-propylphosphoric anhydride (50%) (431, 0.68mmol) was slowly added dropwise under an ice-water bath, and the reaction was carried out at room temperature for 2 hours. After the reaction solution was concentrated under reduced pressure, compound 4 (white solid, 50mg, yield 19%) was obtained.
LC-MS m/z(ESI)=599.30[M+1]。
Example 5
9- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6- (2-isopropyl-4-methylpyridin-3-yl) -4- (5-methyl-1H-indazol-4-yl) -3, 6-dihydrofuro [2',3':4,5] pyrido [2,3-d ] pyrimidin-7 (2H) -one (Compound 5)
9-((S)-4-acryloyl-2-methylpiperazin-1-yl)-6-(2-isopropyl-4-methylpyridin-3-yl)-4-(5-methyl-1H-indazol-4-yl)-3,6-dihydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-7(2H)-one
Figure BDA0003409054530000201
The first step is as follows:
tert-butyl (S) -4- (4-chloro-6- (2-isopropyl-4-methylpyridin-3-yl) -7-oxo-2, 3,6, 7-tetrahydrofuran [2',3':4,5] pyrido [2,3-d ] pyrimidin-9-yl) -3-methylpiperazine-1-carboxylic acid (5a)
tert-butyl(S)-4-(4-chloro-6-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-2,3,6,7-tetrahydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-9-yl)-3-methylpiperazine-1-carboxylate
Intermediate 2(500mg, 1.3mmol) was dissolved in acetonitrile (5mL), N-diisopropylethylamine (865mg, 6.7mmol) was added under an ice-water bath, and phosphorus oxychloride (5mL) was slowly added dropwise under reflux for 2 hours. The reaction mixture was concentrated under reduced pressure, dissolved in tetrahydrofuran, and slowly added dropwise to tetrahydrofuran containing (S) -4-N-tert-butoxycarbonyl-2-methylpiperazine (322mg, 1.6mmol) and N, N-diisopropylethylamine (865g, 6.7mmol) in an ice-water bath to react at room temperature for 2 hours. The reaction was added dropwise to a saturated aqueous sodium bicarbonate solution (50mL), extracted with ethyl acetate, concentrated, and purified by column chromatography to dichloromethane: gradient elution with ethyl acetate 4:1-1:2 gave compound 5a (600mg, 81%).
LC-MS m/z(ESI)=555.20[M+1]。
The second step is that:
tert-butyl (3S) -4- (6- (2-isopropyl-4-methylpyridin-3-yl) -4- (5-methyl-1H-indazol-4-yl) -7-oxo-2, 3,6, 7-tetrahydrofuran [2',3':4,5] pyrido [2,3-d ] pyrimidin-9-yl) -3-methylpiperazine-1-carboxylic acid (5b)
tert-butyl(3S)-4-(6-(2-isopropyl-4-methylpyridin-3-yl)-4-(5-methyl-1H-indazol-4-yl)-7-oxo-2,3,6,7-tetrahydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-9-yl)-3-methylpiperazine-1-carboxylate
Dissolve 5a (0.3g, 0.54mmol), 5-methyl-1H-indazole-4-boronic acid (112mg, 0.63mmol), sodium carbonate (224mg, 2.1mmol), palladium tetrakistriphenylphosphine (61mg, 0.05mmol) in 1, 4-dioxane: water-4: 1(5mL) at 100 ℃ for 4 h. Quenching with 50mL of saturated aqueous sodium bicarbonate solution, extracting with ethyl acetate, concentrating, purifying by column chromatography with dichloromethane: methanol 80: 1-10: gradient elution 1 afforded compound 5b (white solid, 280mg, 80% yield).
LC-MS m/z(ESI)=651.30[M+1]。
The third step:
9- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -6- (2-isopropyl-4-methylpyridin-3-yl) -4- (5-methyl-1H-indazol-4-yl) -3, 6-dihydrofuro [2',3':4,5] pyrido [2,3-d ] pyrimidin-7 (2H) -one (Compound 5)
9-((S)-4-acryloyl-2-methylpiperazin-1-yl)-6-(2-isopropyl-4-methylpyridin-3-yl)-4-(5-methyl-1H-indazol-4-yl)-3,6-dihydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-7(2H)-one
5b (280mg, 0.43mmol) was dissolved in dichloromethane (4mL) and trifluoroacetic acid (1mL) was slowly added dropwise under an ice-water bath and reacted at room temperature for 30 minutes. After concentration under reduced pressure, the resulting mixture was dissolved in N, N-dimethylformamide (3mL), N-diisopropylethylamine (444mg, 3.4mmol) was added dropwise, and 1-propylphosphoric anhydride (50%) (410, 0.65mmol) was slowly added dropwise in an ice-water bath, followed by reaction at room temperature for 2 hours. The reaction mixture was concentrated under reduced pressure to give compound 5 (white solid, 53mg, yield 20%).
LC-MS m/z(ESI)=605.30[M+1]。
Example 6
9- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -4- (2-fluoro-6-hydroxyphenyl) -6- (2-isopropyl-4-methylpyridin-3-yl) -3, 6-dihydrofuro [2',3':4,5] pyrido [2,3-d ] pyrimidin-7 (2H) -one (Compound 6)
9-((S)-4-acryloyl-2-methylpiperazin-1-yl)-4-(2-fluoro-6-hydroxyphenyl)-6-(2-isopropyl-4-methylpyridin-3-yl)-3,6-dihydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-7(2H)-one
Figure BDA0003409054530000211
The first step is as follows:
tert-butyl (3S) -4- (4- (2-fluoro-6-hydroxyphenyl) -6- (2-isopropyl-4-methylpyridin-3-yl) -7-oxo-2, 3,6, 7-tetrahydrofuro [2',3':4,5] pyrido [2,3-d ] pyrimidin-9-yl) -3-methylpiperazine-1-carboxylic acid (6a)
tert-butyl(3S)-4-(4-(2-fluoro-6-hydroxyphenyl)-6-(2-isopropyl-4-methylpyridin-3-yl)-7-oxo-2,3,6,7-tetrahydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-9-yl)-3-methylpiperazine-1-carboxylate
Dissolve 5a (0.3g, 0.54mmol), 2-fluoro-6-hydroxyphenylboronic acid (98.6mg, 0.63mmol), sodium carbonate (224mg, 2.1mmol), palladium tetrakistriphenylphosphine (61mg, 0.05mmol) in 1, 4-dioxane: water 4: 1(5mL) at 100 ℃ for 4 h. Quenching with 50mL of saturated aqueous sodium bicarbonate solution, extracting with ethyl acetate, concentrating, purifying by column chromatography with dichloromethane: methanol 80: 1-10: gradient elution of 1 afforded Compound 6a (white solid, 282mg, 82% yield)
LC-MS m/z(ESI)=631.30[M+1]。
The second step is that:
9- ((S) -4-acryloyl-2-methylpiperazin-1-yl) -4- (2-fluoro-6-hydroxyphenyl) -6- (2-isopropyl-4-methylpyridin-3-yl) -3, 6-dihydrofuro [2',3':4,5] pyrido [2,3-d ] pyrimidin-7 (2H) -one (Compound 6)
9-((S)-4-acryloyl-2-methylpiperazin-1-yl)-4-(2-fluoro-6-hydroxyphenyl)-6-(2-isopropyl-4-methylpyridin-3-yl)-3,6-dihydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-7(2H)-one
6a (282mg, 0.44mmol) was dissolved in dichloromethane (4mL), and trifluoroacetic acid (1mL) was slowly added dropwise under an ice-water bath, followed by reaction at room temperature for 30 minutes. After concentration under reduced pressure, the resulting mixture was dissolved in N, N-dimethylformamide (3mL), N-diisopropylethylamine (461mg, 3.6mmol) was added dropwise, and 1-propylphosphoric anhydride (50%) (427mg, 0.67mmol) was slowly added dropwise in an ice-water bath, followed by reaction at room temperature for 2 hours. After the reaction solution was concentrated under reduced pressure, compound 6 (white solid, 40mg, yield 15%) was obtained.
LC-MS m/z(ESI)=585.30[M+1]。
Example 7
9- (2-acryloyl-2, 7-diazaspiro [3.5] nonan-7-yl) -4- (2-fluorophenyl) -6- (2-isopropyl-4-methylpyridin-3-yl) -3, 6-dihydrofuran [2',3':4,5] pyrido [2,3-d ] pyrimidin-7 (2h) -one (compound 7)
9-(2-acryloyl-2,7-diazaspiro[3.5]nonan-7-yl)-4-(2-fluorophenyl)-6-(2-isopropyl-4-methylpyridin-3-yl)-3,6-dihydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-7(2H)-one
Figure BDA0003409054530000221
Synthesis according to the synthesis of compound 3, preparative synthesis gave compound 7 (white solid, 43mg, 25% yield).
1H NMR(400MHz,DMSO-d6)δ8.36(d,1H),7.26-7.17(m,1H),7.16(d,1H),6.70(d,1H),6.63(t,1H),6.34(dd,1H),6.12(dd,1H),5.68(dd,1H),4.92-4.87(m,1H),4.05-4.01(m,2H),3.77-3.72(m,2H),3.60-3.57(m,4H),3.14-2.93(m,2H),2.77-2.68(m,1H),2.67-2.59(m,1H),2.34-2.30(m,1H),1.95-1.83(m,7H),1.06(d,3H),0.94(d,3H)。
LCMS m/z(ESI)=595.2[M+1]。
Example 8
9- (6-acryloyl-2, 6-diazaspiro [3.3] heptan-2-yl) -4- (2-fluoro-6-hydroxyphenyl) -6- (2-isopropyl-4-methylpyridin-3-yl) -3, 6-dihydrofuran [2',3':4,5] pyrido [2,3-d ] pyrimidin-7 (2H) -one (Compound 8)
9-(6-acryloyl-2,6-diazaspiro[3.3]heptan-2-yl)-4-(2-fluoro-6-hydroxyphenyl)-6-(2-isopropyl-4-methylpyridin-3-yl)-3,6-dihydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-7(2H)-one
Figure BDA0003409054530000222
Following the procedure for the synthesis of compound 3, preparative synthesis gave compound 8 (slightly yellowish solid, 36mg, 31% yield).
1H NMR(600MHz,DMSO-d6)δ10.09(s,1H),8.34(d,1H),7.24-7.16(m,1H),7.14(d,1H),6.70(d,1H),6.63(t,1H),6.33-6.28(m,1H),6.11(dd,1H),5.69-5.67(m,1H),4.93-4.85(m,2H),4.82-4.76(m,2H),4.52-4.34(m,4H),4.22-4.11(m,2H),3.09-2.95(m,2H),2.71-2.66(m,1H),1.88(s,3H),1.05(d,3H),0.93(d,3H)。
LCMS m/z(ESI)=583.2[M+1]。
Example 9
9- (2-acryloyl-2, 7-diazaspiro [3.5] nonan-7-yl) -4- (2-fluoro-6-hydroxyphenyl) -6- (2-isopropyl-4-methylpyridin-3-yl) -3, 6-dihydrofuran [2',3':4,5] pyrido [2,3-d ] pyrimidin-7 (2h) -one (Compound 9)
9-(2-acryloyl-2,7-diazaspiro[3.5]nonan-7-yl)-4-(2-fluoro-6-hydroxyphenyl)-6-(2-isopropyl-4-methylpyridin-3-yl)-3,6-dihydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-7(2H)-one
Figure BDA0003409054530000231
Synthesis according to the synthesis of compound 3, preparative synthesis gave compound 9 (white solid, 27mg, 42% yield).
1H NMR(400MHz,DMSO-d6)δ10.05(s,1H),8.35(d,1H),7.23-7.13(m,1H),7.15(d,1H),6.71(d,1H),6.65(t,1H),6.34-6.27(m,1H),6.15(dd,1H),5.70-5.62(m,1H),4.91-4.88(m,1H),4.07-4.02(m,2H),3.79-3.73(m,2H),3.61-3.58(m,4H),3.11-2.90(m,2H),2.78-2.67(m,1H),2.68-2.61(m,1H),2.36-2.31(m,1H),1.99-1.85(m,6H),1.09(d,3H),0.91(d,3H)。
LCMS m/z(ESI)=611.3[M+1]。
Example 10
9- (4-Acryloylpiperazin-1-yl) -4- (2-fluoro-6-hydroxyphenyl) -6- (2-isopropyl-4-methylpyridin-3-yl) -3, 6-dihydrofuran [2',3':4,5] pyrido [2,3-d ] pyrimidin-7 (2H) -one (Compound 10)
9-(4-acryloylpiperazin-1-yl)-4-(2-fluoro-6-hydroxyphenyl)-6-(2-isopropyl-4-methylpyridin-3-yl)-3,6-dihydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-7(2H)-one
Figure BDA0003409054530000232
Synthesis according to the synthesis of compound 3, preparative synthesis gave compound 10 (white solid, 18mg, 45% yield).
1H NMR(400MHz,DMSO-d6)δ10.01(s,1H),8.33(d,1H),7.23-7.15(m,1H),7.16(d,1H),6.73(d,1H),6.67(t,1H),6.35-6.26(m,1H),6.18(dd,1H),5.71-5.64(m,1H),4.17-4.05(m,2H),3.76-3.70(m,4H),3.63-3.59(m,4H),3.15-2.93(m,2H),3.14-2.99(m,1H),2.35(m,3H),1.14(d,3H),0.98(d,3H)。
LCMS m/z(ESI)=671.2[M+1]。
Example 11
9- (6-acryloyl-2, 6-diazaspiro [3.3] heptan-2-yl) -4- (2-fluorophenyl) -6- (2-isopropyl-4-methylpyridin-3-yl) -3, 6-dihydrofuran [2',3':4,5] pyrido [2,3-d ] pyrimidin-7 (2h) -one (compound 11)
9-(6-acryloyl-2,6-diazaspiro[3.3]heptan-2-yl)-4-(2-fluorophenyl)-6-(2-isopropyl-4-methylpyridin-3-yl)-3,6-dihydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-7(2H)-one
Figure BDA0003409054530000241
Synthesis according to the synthesis of compound 3, preparative synthesis gave compound 11 (white solid, 28mg, 35% yield).
1H NMR(600MHz,DMSO-d6)δ8.45(dd,1H),7.85-6.95(m,4H),6.31(dd,1H),6.21-6.00(m,2H),5.66(ddd,1H),4.90(t,1H),4.78(s,1H),4.52-4.37(m,1H),4.17(d,1H),3.72(s,4H),3.4(d,4H),3.14(t,1H),2.71(p,1H),1.94(d,2H),1.21-0.90(m,6H)。
LCMS m/z(ESI)=567.2[M+1]。
Example 12
9- (6-acryloyl-2, 6-diazaspiro [3.3] heptan-2-yl) -4- (2-hydroxyphenyl) -6- (2-isopropyl-4-methylpyridin-3-yl) -3, 6-dihydrofuran [2',3':4,5] pyrido [2,3-d ] pyrimidin-7 (2h) -one (Compound 12)
9-(6-acryloyl-2,6-diazaspiro[3.3]heptan-2-yl)-4-(2-hydroxyphenyl)-6-(2-isopropyl-4-methylpyridin-3-yl)-3,6-dihydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-7(2H)-one
Figure BDA0003409054530000242
Synthesis according to the method for synthesizing compound 3, Synthesis was conducted to give compound 12 (white solid, 18mg, yield 39%).
1H NMR(600MHz,DMSO-d6)δ11.69(s,1H),8.51(d,1H),7.60(dd,1H),7.35-7.21(m,2H),6.87(t,1H),6.74(d,1H),6.31(dd,1H),6.11(dd,1H),5.69(dd,1H),5.04-4.87(m,2H),4.75(s,2H),4.56-4.36(m,4H),4.25-3.97(m,2H),3.67-3.44(m,2H),2.72(p,1H),1.93(s,3H),1.08(d,3H),0.94(d,3H)。
LCMS m/z(ESI)=565.25[M+1]。
Example 13
9- (((1-Acryloylazetidin-3-yl) methyl) amino) -4- (2-hydroxyphenyl) -6- (2-isopropyl-4-methylpyridin-3-yl) -3, 6-dihydrofuro [2',3':4,5] pyrido [2,3-d ] pyrimidin-7 (2h) -one (Compound 13)
9-(((1-acryloylazetidin-3-yl)methyl)amino)-4-(2-hydroxyphenyl)-6-(2-isopropyl-4-methylpyridin-3-yl)-3,6-dihydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-7(2H)-one
Figure BDA0003409054530000251
Synthesis according to the method for synthesizing compound 3, Synthesis was conducted to give compound 13 (white solid, 33mg, yield 37%).
1H NMR(400MHz,DMSO-d6)δ11.42(s,1H),8.51(t,1H),8.30(d,1H),7.78-7.50(m,1H),7.39-7.10(m,2H),6.84(t,1H),6.78-6.60(m,1H),6.28(ddd,1H),6.09(ddd,1H),5.67-5.52(m,1H),4.92-4.76(m,2H),3.91-3.65(m,2H),3.34(d,2H),2.99(d,2H),2.88(p,1H),2.79-2.64(m,1H),1.97(d,2H),1.23(s,3H),1.08(dt,3H),1.03-0.88(m,3H)。
LCMS m/z(ESI)=553.1[M+1]。
Example 14
9- (((1-Acryloylazetidin-3-yl) methyl) amino) -4- (3-fluorophenyl) -6- (2-isopropyl-4-methylpyridin-3-yl) -3, 6-dihydrofuran [2',3':4,5] pyrido [2,3-d ] pyrimidin-7 (2h) -one (Compound 14)
9-(((1-acryloylazetidin-3-yl)methyl)amino)-4-(3-fluorophenyl)-6-(2-isopropyl-4-methylpyridin-3-yl)-3,6-dihydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-7(2H)-one
Figure BDA0003409054530000252
Synthesis according to the method for synthesizing compound 3, Synthesis was conducted to give compound 14 (white solid, 35mg, yield 41%).
1H NMR(400MHz,DMSO-d6)δ8.48(dd,1H),7.98(s,1H),7.44(d,2H),7.34-7.17(m,3H),6.43-6.16(m,1H),6.07(s,1H),5.75-5.55(m,1H),5.00(d,,1H),4.32(t,1H),4.03(dd,1H),3.81(d,2H),2.77-2.61(m,2H),2.33(q,1H),2.01(s,1H),1.93(s,2H),1.23(s,4H),1.08(t,3H),0.97-0.76(m,3H)。
LCMS m/z(ESI)=555.24[M+1]。
Example 15
9- ((1-Acryloylazetidin-3-yl) amino) -4- (2-fluorophenyl) -6- (2-isopropyl-4-methylpyridin-3-yl) -3, 6-dihydrofuran [2',3':4,5] pyrido [2,3-d ] pyrimidin-7 (2H) -one (Compound 15)
9-((1-acryloylazetidin-3-yl)amino)-4-(2-fluorophenyl)-6-(2-isopropyl-4-methylpyridin-3-yl)-3,6-dihydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-7(2H)-one
Figure BDA0003409054530000261
Synthesis according to the synthesis of compound 3, preparative synthesis gave compound 15 (white solid, 25mg, 21% yield).
1H NMR(400MHz,DMSO-d6)δ8.72(dd,1H),7.87(s,1H),7.34(d,2H),7.29-7.21(m,3H),6.21-6.06(m,1H),5.92(s,1H),5.71-5.59(m,1H),5.03(d,1H),4.66(t,1H),4.21(dd,1H),2.61-2.42(m,2H),2.31(q,1H),2.13(s,1H),1.80(s,2H),1.37(s,4H),1.08(t,3H),0.96-0.72(m,3H)。
LCMS m/z(ESI)=541.23[M+1]。
Example 16
9- ((1-Acryloylazetidin-3-yl) amino) -4- (2-fluoro-6-hydroxy-phenyl) -6- (2-isopropyl-4-methylpyridin-3-yl) -3, 6-dihydrofuran [2',3':4,5] pyrido [2,3-d ] pyrimidin-7 (2h) -one (compound 16)
9-((1-acryloylazetidin-3-yl)amino)-4-(2-fluoro-6-hydroxyphenyl)-6-(2-isopropyl-4-methylpyridin-3-yl)-3,6-dihydrofuro[2',3':4,5]pyrido[2,3-d]pyrimidin-7(2H)-one
Figure BDA0003409054530000262
Following the procedure for the synthesis of compound 3, preparative synthesis gave compound 16 (slightly yellowish solid, 35mg, 26% yield).
1H NMR(400MHz,DMSO-d6)δ9.87(s,1H),8.63(dd,1H),7.77(s,1H),7.52(d,2H),7.33-7.22(m,2H),6.29-6.02(m,1H),5.98(s,1H),5.80-5.62(m,1H),5.23(d,,1H),4.56(t,1H),4.22(dd,1H),2.51-2.32(m,2H),2.21(q,1H),2.13(s,1H),1.88(s,2H),1.42(s,4H),1.12(t,3H),0.90-0.75(m,3H)。
LCMS m/z(ESI)=557.3[M+1]。
IC50: refers to the concentration of the compound at which the activity of KRAS is 50% inhibited.
DMEM: dulbecco's modified eagle Medium;
FBS: fetal bovine serum;
NBT: nitro blue tetrazolium.
In the examples, the reaction temperature is room temperature, and the optimum reaction temperature is 20 ℃ to 30 ℃;
biological assay
Soft agar gel clone formation assay
(1) Bottom high concentration agarose gel layer: high concentration agarose gels were plated in 96-well plates (50. mu.L/well).
(2) Upper low concentration agarose gel layer: low concentration agarose and cell-containing medium were mixed at 1:1, mixing and spreading on a bottom agarose gel layer (100 mu L/hole; the used cells are NCI-H358 and MiaPaca cells, the cell amount is 10000 cells/hole); after cooling and solidification, the cells were incubated at 37 ℃ overnight.
(3) Test compounds were prepared as 10mM stock, diluted in growth medium (1:5) in a 8-10 concentration gradient (starting concentration 10. mu.M), added to 96 wells containing supernatant agar gel-cells (50. mu.L/well); setting a solvent control hole; each concentration is 2 times repeated, and the culture medium is placed in a carbon dioxide incubator for 10 to 14 days.
(4) On day 7, the medium containing the candidate compound was changed once and the growth of cell clones was observed.
(5) After the culture was completed, the cells were stained with NBT, the number of clones formed was counted, and GraphPad Prism 8 software was fitted with IC50The value is obtained.
The results show that all compounds had IC formed on cell clones50Less than 10 mu M, has good inhibition effect on KRAS mutant tumor cells, wherein the effects of the compound 6 and the compound 10 are obvious.
Compound (I) IC50nM(NCI-H358 cell)
Compound 6 293.7
Compound 10 256.9
While the present invention has been described in detail with respect to the specific embodiments thereof, it will be understood by those skilled in the art that the above embodiments are illustrative and not to be construed as limiting the present invention, and that various changes and modifications can be made therein by those skilled in the art without departing from the spirit of the invention and within the scope of the appended claims.

Claims (10)

1. A compound of formula (I), or a pharmaceutically acceptable salt or stereoisomer thereof:
Figure FDA0003409054520000011
wherein
X, Y, Z are each independently CH2Or O;
ring B is
Figure FDA0003409054520000012
Said B ring being optionally substituted by 1,2 or 3R2Substitution;
R1is H or halogen;
R2is H or C1-6An alkyl group;
C1is composed of
Figure FDA0003409054520000013
C2Is composed of
Figure FDA0003409054520000014
2. The compound of claim 1, or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
X, Y, Z are each independently CH2Or O;
ring B is
Figure FDA0003409054520000015
Said B ring being optionally substituted by 1,2 or 3R2Substitution;
R1is H or halogen;
R2is H or C1-6An alkyl group;
C1is composed of
Figure FDA0003409054520000016
C2Is composed of
Figure FDA0003409054520000021
3. The compound of claim 1, or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
One of X and Z is O and the other is CH2(ii) a Y is CH2
R1Is H;
ring B is
Figure FDA0003409054520000022
Said B ring being optionally substituted by 1 or 2R2Substitution;
R2is C1-6An alkyl group;
C1is composed of
Figure FDA0003409054520000023
C2Is composed of
Figure FDA0003409054520000024
4. A compound of formula (II), or a pharmaceutically acceptable salt or stereoisomer thereof:
Figure FDA0003409054520000025
wherein
X, Y, Z are each independently CH2Or O;
n is 0, 1,2, 3;
ring B is 4-10 membered heterocycloalkyl containing 1,2 or 3N atoms, optionally substituted with 1,2 or 3R4Substitution;
R3is H or halogen;
R4is H or C1-6An alkyl group;
C1is composed of
Figure FDA0003409054520000026
C2Is composed of
Figure FDA0003409054520000031
5. The compound of claim 4, or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
One of X and Z is O and the other is CH2(ii) a Y is CH2
n is 0 or 1;
ring B is
Figure FDA0003409054520000032
Said B ring being optionally substituted by 1 or 2R4Substitution;
R3is H;
R4is C1-6An alkyl group;
C1is composed of
Figure FDA0003409054520000033
C2Is composed of
Figure FDA0003409054520000034
6. A compound of formula (III), or a pharmaceutically acceptable salt or stereoisomer thereof:
Figure FDA0003409054520000035
wherein
X, Y, Z are each independently CH2Or O;
R1、R2each independently is H or C1-6An alkyl group;
R3is H or halogen;
C1is composed of
Figure FDA0003409054520000036
C2Is composed of
Figure FDA0003409054520000037
7. The compound of claim 6, or a pharmaceutically acceptable salt or stereoisomer thereof, wherein
One of X and Z is O and the other is CH2(ii) a Y is CH2
R1And R2Is C1-6An alkyl group;
C1is composed of
Figure FDA0003409054520000041
C2Is composed of
Figure FDA0003409054520000042
8. A compound, or a pharmaceutically acceptable salt or stereoisomer thereof, having one of the following structures:
Figure FDA0003409054520000043
Figure FDA0003409054520000051
Figure FDA0003409054520000061
9. a pharmaceutical composition comprising:
(1) a compound of any one of claims 1-8 or a stereoisomer, solvate, metabolite, pharmaceutically acceptable salt, co-crystal or prodrug thereof;
(2) optionally one or more other active ingredients; and
(3) a pharmaceutically acceptable carrier and/or excipient.
10. Use of a compound of any one of claims 1-8 or a pharmaceutical composition of claim 9 in the manufacture of a medicament for the prevention or treatment of cancer, an antineoplastic agent, a medicament for the treatment of a KRAS mutation-mediated disease, or a KRAS inhibitor; preferably, the cancer is cholangiocarcinoma, cervical cancer, bladder cancer, liver cancer or breast cancer.
CN202111526178.6A 2020-12-14 2021-12-14 Pyridine derivative and application thereof in medicine Active CN114621244B (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN202011461273 2020-12-14
CN2020114612738 2020-12-14
CN202110159721 2021-02-05
CN2021101597217 2021-02-05

Publications (2)

Publication Number Publication Date
CN114621244A true CN114621244A (en) 2022-06-14
CN114621244B CN114621244B (en) 2023-08-18

Family

ID=81898155

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202111526178.6A Active CN114621244B (en) 2020-12-14 2021-12-14 Pyridine derivative and application thereof in medicine

Country Status (1)

Country Link
CN (1) CN114621244B (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992008719A1 (en) * 1990-11-08 1992-05-29 Basf Aktiengesellschaft SUBSTITUTED PYRIDO[2,3-d]PYRIMIDIN-2,4(1H,3H)-DIONES
CN112574224A (en) * 2019-09-30 2021-03-30 上海迪诺医药科技有限公司 KRAS G12C inhibitor and application thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992008719A1 (en) * 1990-11-08 1992-05-29 Basf Aktiengesellschaft SUBSTITUTED PYRIDO[2,3-d]PYRIMIDIN-2,4(1H,3H)-DIONES
CN112574224A (en) * 2019-09-30 2021-03-30 上海迪诺医药科技有限公司 KRAS G12C inhibitor and application thereof

Also Published As

Publication number Publication date
CN114621244B (en) 2023-08-18

Similar Documents

Publication Publication Date Title
ES2928169T3 (en) 4-(3H-indol-5-yl)-N-(pyridin-2-yl)pyrimidin-2-amine derivatives as protein kinase inhibitors, and method of preparation and medical use thereof
CN104125959B (en) Heteroaryl pyridone and aza-pyridone compounds as inhibitors of BTK activity
EP3935049A1 (en) Carboxamide-pyrimidine derivatives as shp2 antagonists
CN104024255B (en) Alkylated piperazine compounds as inhibitors of BTK activity
RU2468027C2 (en) Phosphoinositide-3-kinase inhibitors and methods for use thereof
EP3952998A1 (en) Pyrimidinone derivatives as shp2 antagonists
CN115232121B (en) Pyridine derivative and application thereof in medicine
EP4043465A1 (en) Substituted aromatic fused ring derivative and composition comprising same, and use thereof
EP3444250A1 (en) Fgfr4 inhibitor, preparation method therefor, and applications thereof
CN113121574B (en) Purine derivatives and their use in medicine
JP2019500413A5 (en)
CN112552294A (en) Piperazine heterocyclic derivative inhibitor, preparation method and application thereof
KR20130110149A (en) Phthalazinone ketone derivative, preparation method thereof, and pharmaceutical use thereof
CN110914277B (en) Imidazo [1,2-b ] pyrimido [4,5-d ] pyridazin-5 (6H) -one compounds and application thereof
JP2021515767A (en) Identification and use of ERK5 inhibitors
EP3498707A1 (en) Fgfr4 inhibitor and preparation method and use thereof
CN118055933A (en) Selective PARP1 inhibitors and uses thereof
KR20210088675A (en) Macrocyclic tyrosine kinase inhibitors and uses thereof
CN112867717A (en) Compounds useful as kinase inhibitors and uses thereof
CN115466273A (en) Substituted alkynyl heterocyclic compounds
CN115803328A (en) Piperazine-2, 3-dione derivative and application thereof in medicine
CN113968856B (en) Compounds with kinase inhibitory activity
CN114621244B (en) Pyridine derivative and application thereof in medicine
AU2020281411A1 (en) Tetracyclic compounds as Cdc7 inhibitors
CN114805359B (en) Preparation method and application of acetylenic heterocyclic compound FGFR inhibitor

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
GR01 Patent grant
GR01 Patent grant