CN112851587A - Alkyne heterocyclic compound for treating cancer and preparation method and application thereof - Google Patents

Alkyne heterocyclic compound for treating cancer and preparation method and application thereof Download PDF

Info

Publication number
CN112851587A
CN112851587A CN202110080572.5A CN202110080572A CN112851587A CN 112851587 A CN112851587 A CN 112851587A CN 202110080572 A CN202110080572 A CN 202110080572A CN 112851587 A CN112851587 A CN 112851587A
Authority
CN
China
Prior art keywords
membered
cycloalkyl
cancer
compound
alkyl
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
CN202110080572.5A
Other languages
Chinese (zh)
Inventor
梁永宏
严文广
曾兆森
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.)
Yaoya Technology Shanghai Co ltd
Original Assignee
Yaoya Technology Shanghai 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 Yaoya Technology Shanghai Co ltd filed Critical Yaoya Technology Shanghai Co ltd
Priority to CN202110080572.5A priority Critical patent/CN112851587A/en
Publication of CN112851587A publication Critical patent/CN112851587A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/34One oxygen 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
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/47One nitrogen atom and one oxygen or sulfur atom, e.g. cytosine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, directly attached to ring carbon atoms
    • C07D239/46Two or more oxygen, sulphur or nitrogen atoms
    • C07D239/48Two nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Abstract

Hair brushThe invention discloses a selective inhibitor of a clinical mutant of FGFR protein tyrosine kinase, a compound shown in a general formula (I) and a medicinal salt thereof, a pharmaceutical composition containing the compound and/or the medicinal salt thereof, and application of the compound or the medicinal salt thereof in medicaments for treating or preventing FGFR kinase related diseases, particularly tumors.

Description

Alkyne heterocyclic compound for treating cancer and preparation method and application thereof
The technical field is as follows:
the invention relates to an acetylenic heterocyclic compound or a pharmaceutically acceptable salt thereof as an FGFR inhibitor; a pharmaceutical composition containing the alkynylated heterocyclic compound or a pharmaceutically acceptable salt thereof; a process for producing the alkynylated heterocyclic compound or a pharmaceutically acceptable salt thereof; the application of the alkynylated heterocyclic compound or pharmaceutically acceptable salt thereof or a pharmaceutical composition containing the alkynylated heterocyclic compound or pharmaceutically acceptable salt thereof in preparing medicaments for treating and/or preventing FGFR related diseases, particularly tumors.
Background art:
fibroblast Growth Factor Receptors (FGFR) are Receptor Tyrosine Kinases (RTK), and the FGFR family mainly comprises four subtypes of FGFR1-, FGFR2, FGFR3 and FGFR 4. FGFR1 is a transmembrane protein belonging to the receptor tyrosine kinase, consisting of three major components: namely an extracellular domain, a transmembrane domain and a cell domain, the extracellular domain is a binding domain of the ligand Fibroblast Growth Factors (FGFs). FGFs are also a polygenic family, and there are 19 members, namely FGF1 also known as acidic fibroblast growth factor (aFGF), FGF2 also known as basic fibroblast growth-related document factor (basic FGF, bFGF), which have biological activities of stimulating the growth of fibroblasts, vascular endothelial cells, smooth muscle cells and nerve cells, and FGFR1, which is a high affinity receptor. After FGF is combined with an extracellular segment of FGFR1, a tyrosine kinase active region of an inner segment of a receptor cell firstly generates autophosphorylation, then receptor target protein generates transphosphorylation, and a signal of a ligand is transmitted to a cell nucleus through a family structure and family relation of a protein cascade reaction, so that the effects of promoting damage repair, embryonic development and skeleton formation are achieved.
However, when FGFR is mutated or overexpressed, it causes excessive activation of the FGFR signaling pathway and further induces normal cell carcinogenesis. Wherein, over-activation of RAS-RAF-MAPK stimulates cell proliferation and differentiation; over-activation of PI3K-AKT results in inhibition of apoptosis; SATA is closely related to promoting tumor invasion and metastasis and enhancing tumor immune escape capacity; the PLC gamma signal channel is an important way for regulating and controlling the metastasis of tumor cells. Next Generation Sequencing (NGS) on 4853 solid tumor types showed, according to a study published in Clinical Cancer Research in 2015, FGFR aberrations (abortions) and abnormal activation were found in approximately 7.1% of cancers, mostly gene amplification (66%), followed by mutations (26%) and rearrangements (8%). FGFR distortion exists in almost all detected malignant tumors, and the cancers with high incidence rate include urothelial carcinoma, cholangiocarcinoma, breast cancer, endometrial carcinoma, squamous epithelial carcinoma and the like; meanwhile, the abnormal activation of FGFR is also found in tumors such as lung cancer, liver cancer, breast cancer and the like.
There are currently some non-FGFR specific drugs on the market, such as Sunitinib from pfizer, lentitini from Eisai, and nintedanib from Boehringer ingelheimer. Whereas the only FGFR inhibitors approved by the FDA to be marketed are balversa (erdafitinib) and pemazyre (pemigatinib). Small molecule inhibitors of FGFR1/2/3 entering the clinic are: infigrtinib (BGJ398) and AZD4547, fisogatinib (BLU-554), Roblitiniib (FGF401), H3B6527, lucitanib (E-3810), Futibatinib (TAS-120), RPN1371, ICP-192, derazatinib, 3D185, BPI-17509, HMPL-453.
Although the development of FGFR inhibitors has attracted the deployment of numerous companies both at home and abroad, and although 2 FGFR inhibitors are already on the market, there is still a need to develop new compounds due to the prospects they show in the treatment of various malignancies. Through continuous efforts, the invention designs an irreversible inhibitor which has proprietary intellectual property rights and shows excellent activity on FGFR-1-4 protein kinase.
Disclosure of Invention
In order to solve the above problems, the present invention provides a novel FGFR tyrosine kinase inhibitor compound represented by formula (I) or a stereoisomer, hydrate, solvate, or pharmaceutically acceptable salt thereof:
Figure BDA0002909122720000021
wherein:
x1, X2, X3, X4 and X5 are each independently CR1Or N, and at least one of X1, X2, X3, X4 and X5 is N; ring B is a phenyl ring or a 5-6 membered heteroaromatic ring wherein the phenyl and heteroaromatic rings are optionally substituted by one or more G1;
R1independently selected from H, cyano, halogen, C1-6Alkyl radical, C3-6Cycloalkyl, 3-6 membered heterocycloalkyl, -OR2、-NR2R3、-C(O)NR2R3Wherein said alkyl, cycloalkyl OR heterocycloalkyl is optionally substituted by cyano, halogen, -OR4、-NR4R5、C1-6Alkyl radical, C3-6Cycloalkyl or 3-6 membered heterocycloalkyl;
a is independently selected from-C0-4Alkyl-, -CR6R7-、-C1-2Alkyl (R)6)(OH)-、-C(O)-、-CR6R7O-、-OCR6R7-、-SCR6R7-、-CR6R7S-、-NR6-、-NR6C(O)-、-C(O)NR6-、-NR6C(O)NR7-、-CF2-、-O-、-S-、-S(O)m-、-NR6S(O)2-、-S(O)2NR6-;
Y is absent or C is selected3-8Cycloalkyl, 3-8 membered heterocycloalkyl, 5-12 membered fused alkyl, 5-12 membered fused heterocyclyl, 5-12 membered spiro cyclic group, 5-12 membered spiro heterocyclic group, aromatic group or heteroaromatic group, wherein said cycloalkyl, heterocycloalkyl, spiro cyclic group, fused heterocyclic group, spiro heterocyclic group, aromatic group or heteroaromatic group is optionally substituted with one or more G2Substituted;
z is independently selected from cyano, -NR8CN、
Figure BDA0002909122720000031
Bond a is a double or triple bond;
when a is a double bond, Ra、RbAnd RcEach independently selected from H, cyano, halogen, C1-6Alkyl radical, C3-6Cycloalkyl or 3-6 membered heterocycleAnd (4) a base. Wherein said alkyl, cycloalkyl and heterocyclyl are optionally substituted by 1 or more G3Substituted;
Raand RbOr RbAnd RcOptionally taken together with the carbon atom to which they are attached to form a 3-6 membered ring optionally containing heteroatoms;
when bond a is a triple bond, RaAnd RcIs absent, RbIndependently selected from H, cyano, halogen, C1-6Alkyl radical, C3-6Cycloalkyl or 3-6 membered heterocyclyl by one or more G4Substituted;
R8independently selected from H, C1-6Alkyl radical, C3-6Cycloalkyl or 3-6 membered heterocyclyl, wherein said alkyl, cycloalkyl and heterocyclyl are optionally substituted by 1 or more G5Substituted;
G1、G2、G3、G4and G5Each independently selected from cyano, halogen, C1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-8Cycloalkyl or 3-8 membered heterocyclyl, C6-10Aryl, 5-10 membered heteroaryl, -OR9、-OC(O)NR9R10、-C(O)OR9、-C(O)NR9R10、-C(O)R9、-NR9R10、-NR9C(O)R10、-NR9C(O)NR10R11、-S(O)mR9or-NR9S(O)mR10Wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl are optionally substituted by 1 or more cyano, halogen, C1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-8Cycloalkyl or 3-8 membered heterocyclyl, C6-10Aryl, 5-10 membered heteroaryl, -OR12、-OC(O)NR12R13、-C(O)OR12、-C(O)NR12R13、-C(O)R12、-NR12R13、-NR12C(O)R13、-NR12C(O)NR13R14、-S(O)mR12or-NR12S(O)mR13By substitution ofSubstituted by a group;
R3、R4、R5、R6、R7、R8、R9、R11、R12、R13and R14Each independently selected from cyano, halogen, C1-6Alkyl radical, C3-8Cycloalkyl or 3-8 membered monocyclic heterocyclyl, monocyclic heteroaryl or phenyl; and is
m is 1 or 2.
The compounds according to the general structural formula (I) of the present invention include one of the compounds of numbers 101-130, but are not limited to the following compounds:
Figure BDA0002909122720000032
Figure BDA0002909122720000041
Figure BDA0002909122720000051
Figure BDA0002909122720000061
the present invention provides methods for the above novel FGFR inhibitors or isomers, hydrates, solvates, polymorphs, pharmaceutically acceptable salts thereof. The method is characterized by comprising the following steps:
firstly, an intermediate SW-2a or SW-2b is obtained by an initial raw material SW-1a through an aromatic nucleophilic substitution reaction or a Suzuki coupling reaction, then an intermediate SW-3a or SW-3b is obtained by a Sonagashira coupling reaction, then an intermediate SW-3b is obtained by removing a Boc protecting group under an acidic condition or one step is omitted, and finally a compound with a structural general formula (I) is obtained by a nucleophilic addition reaction;
Figure BDA0002909122720000071
and secondly, obtaining an intermediate SW-2 a' from an initial raw material SW-1a through Sonagashira coupling reaction, then carrying out aromatic nucleophilic substitution reaction or Suzuki coupling reaction to obtain an intermediate SW-3a or SW-3b, then removing a Boc protecting group under an acidic condition to obtain an intermediate SW-3b or omitting the step, and finally carrying out nucleophilic addition reaction to obtain the compound with the structural general formula (I).
Figure BDA0002909122720000072
The compounds of the invention are capable of effectively inhibiting the activity of FGFR1, FGFR2, FGFR3 or FGFR4, which inhibit the IC of FGFR1, FGFR2, FGFR3 or FGFR450Is 100 to 1000nM, more preferably IC50Less than 100nM, optimal IC50Less than 10 nM.
The compounds of the invention are useful for the treatment or prevention of FGFR-associated tumors, such as non-small cell lung cancer, esophageal cancer, melanoma rhabdomyosarcoma, cellular carcinoma, multiple myeloma, breast cancer, ovarian cancer, endometrial cancer, cervical cancer, gastric cancer, colon cancer, bladder cancer, pancreatic cancer, lung cancer, prostate cancer, and liver cancer (e.g., hepatocellular carcinoma), more particularly liver cancer, gastric cancer, and bladder cancer. Thus, in a further aspect, the present invention provides a method of treating or preventing FGFR-mediated diseases (e.g. of a neoplasm), which comprises administering to a patient in need thereof a therapeutically effective amount of a compound of the present invention or a prodrug, stable isotope derivative, polymorph, solvate, pharmaceutically acceptable salt, isomer and mixtures thereof, or a pharmaceutical composition comprising the compound.
Another aspect of the present invention relates to a compound of formula (I) or a prodrug, stable isotope derivative, polymorph, solvate, pharmaceutically acceptable salt, isomer, and mixture thereof for pharmaceutical or medicinal use for treating or preventing FGFR mediated diseases, such as tumors or inflammatory diseases, including but not limited to non-small cell lung cancer, esophageal cancer, melanin, rhabdomyosarcoma, wild cell carcinoma, multiple myeloma, breast cancer, ovarian cancer, endometrial cancer, uterine cancer, gastric cancer, diaphragm cancer, bladder cancer, pancreatic cancer, lung cancer, prostate cancer.
The invention further relates to a pharmaceutical composition which comprises the compound or the prodrug, the stable isotope derivative, the pharmaceutically acceptable salt isomer and the mixture thereof, and pharmaceutically acceptable carriers, diluents and excipients.
Another aspect of the invention relates to the use of a compound of formula (I) or a prodrug stable isotope derivative thereof, a pharmaceutically acceptable salt, isomer and mixture thereof, or a pharmaceutical composition thereof for the manufacture of a medicament for the treatment or prevention of FGFR mediated diseases such as tumors and inflammatory diseases.
According to the present invention, the drug may be in any pharmaceutical dosage form including, but not limited to, tablets, sachets, solutions, lyophilized formulations, injections.
Certain chemical terms
Unless stated to the contrary, the following terms are used in the specification and claims.
Has the following meanings and is used herein in the manner ofx-y"denotes the range of the number of carbon atoms, wherein x and y are each an integer, e.g. C3-8Cycloalkyl denotes cycloalkyl having 3 to 8 carbon atoms, i.e. cycloalkyl having 3,4, 5,6, 7 or 8 carbon atoms. It is also understood that "C" is3-8"also includes any subrange therein, e.g. C3-7、C3-6、C4-7、C4-6、C5-6And the like.
"alkyl" refers to a straight or branched chain hydrocarbyl group containing 1 to 20 carbon atoms, for example 1 to 18 carbon atoms, 1 to 12 carbon atoms, 1 to 8 carbon atoms, 1 to 6 carbon atoms, or 1 to 4 carbon atoms. Non-limiting examples of alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1-dimethylpropyl, 1, 2-dimethylpropyl, 2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1, 2-trimethylpropyl, 1-dimethylbutyl, 1, 2-dimethylbutyl, 2-dimethylbutyl, 1, 3-dimethylbutyl, and 2-ethylbutyl. The alkyl group may be substituted or unsubstituted.
"alkenyl" refers to a straight or branched chain hydrocarbyl group containing at least one carbon-carbon double bond and typically 2 to 20 carbon atoms, e.g., 2 to 8 carbon atoms, 2 to 6 carbon atoms, or 2 to 4 carbon atoms. Non-limiting examples of alkenyl groups include ethenyl, 1-propenyl, 2-propenyl, 1-butenyl, 2-butenyl, 3-butenyl, 2-methyl-2-propenyl, 1, 4-pentadienyl, and 1, 4-butadienyl. The alkenyl group may be substituted or unsubstituted.
"alkynyl" refers to a straight or branched chain hydrocarbyl group containing at least one carbon-carbon triple bond and typically 2 to 20 carbon atoms, for example 2 to 8 carbon atoms, 2 to 6 carbon atoms, or 2 to 4 carbon atoms. Non-limiting examples of alkynyl groups include ethynyl, 1-propynyl, 2-propynyl, 1-butynyl, 2-butynyl, and 3-butynyl. The alkynyl group may be substituted or unsubstituted.
"cycloalkyl" refers to a saturated cyclic hydrocarbyl substituent containing from 3 to 14 carbon ring atoms. Cycloalkyl groups may be monocyclic, typically containing from 3 to 7 carbon ring atoms. Non-limiting examples of monocyclic cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl. Cycloalkyl groups may alternatively be bi-or tricyclic fused together, such as decahydronaphthyl, which may be substituted or unsubstituted.
"Heterocyclyl", "heterocycloalkyl", "heterocycle" means a stable 3-to 18-membered monovalent non-aromatic ring comprising 2 to 12 carbon atoms, 1 to 6 heteroatoms selected from nitrogen, oxygen and sulfur. Unless otherwise specified, a heterocyclyl group may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may contain fused, spiro or bridged ring systems, the nitrogen, carbon or sulfur of the heterocyclyl group may optionally be oxidized, the nitrogen atom may optionally be quaternized, and the heterocyclyl group may be partially or fully saturated. The heterocyclic group may be attached to the rest of the molecule through a single bond via a carbon or heteroatom in the ring. The heterocyclic group containing fused rings may contain one or more aromatic or heteroaromatic rings, provided that the atoms on the non-aromatic ring are attached to the rest of the molecule. For purposes of this application, a heterocyclyl group is preferably a stable 4-11 membered monovalent non-aromatic monocyclic or bicyclic ring containing 1-3 heteroatoms selected from nitrogen, oxygen and sulfur, and more preferably a stable 4-8 membered monovalent non-aromatic monocyclic ring containing 1-3 heteroatoms selected from nitrogen, oxygen and sulfur. Non-limiting examples of heterocyclyl groups include azepanyl, azetidinyl, decahydroisoquinolinyl, dihydrofuranyl, indolinyl, dioxolanyl, 1-dioxo-thiomorpholinyl, imidazolidinyl, imidazolinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, oxazinyl, piperazinyl, piperidinyl, 4-piperidinonyl, pyranyl, pyrazolidinyl, pyrrolidinyl, quinolizinyl, quinuclidinyl, tetrahydrofuranyl, tetrahydropyranyl and the like.
"spiroheterocyclyl" refers to a 5 to 20 membered polycyclic heterocyclic group which shares one atom (called the spiro atom) between single rings, wherein one or more ring atoms are selected from nitrogen, oxygen or a heteroatom of S (0) whose m is an integer of 0 to 2, and the remaining ring atoms are carbon. These may contain one or more double bonds, but none of the rings have a fully conjugated electronic system, preferably 6 to 14, more preferably 7 to 10. The spirocycloalkyl group is classified into a single spiroheterocyclyl group, a double spiroheterocyclyl group or a multiple spiroheterocyclyl group, preferably a single spirocycloalkyl group and a double spirocycloalkyl group, according to the number of spiro atoms shared between rings. More preferably a 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered or 5-membered/6-membered monocyclic group. Non-limiting examples of spirocycloalkyl groups include:
Figure BDA0002909122720000091
"fused heterocyclyl" means a 5 to 20 membered polycyclic heterocyclic group in which each ring in the system shares an adjacent pair of atoms with other rings in the system, one or more rings may contain one or more double bonds, but none of the rings has a fully conjugated pi-electron system in which one or more ring atoms are selected from nitrogen, oxygen or S (O)m(wherein m is an integer of 0 to 2) and the remaining ring atoms are carbon. Preferably 6 to 14 membered, more preferably7 to 10 yuan. They may be classified into bicyclic, tricyclic, tetracyclic or polycyclic fused heterocycloalkyl groups according to the number of constituent rings, preferably bicyclic or tricyclic, more preferably 5-or 6-membered bicyclic fused heterocyclic groups. Non-limiting examples of fused heterocyclic groups include:
Figure BDA0002909122720000101
"aryl" or "aryl" refers to an aromatic ring or fused polycyclic group containing 6 to 14 carbon atoms, preferably 6 to 10 members, such as phenyl and naphthyl, most preferably the aryl ring of the phenyl group may be fused to a heteroaryl, heterocyclyl or cycloalkyl ring, wherein the rings attached to the parent structure are aryl rings, non-limiting examples of which include:
"heteroaryl" or "heteroaryl" refers to a 5-16 membered ring system containing 1-15 carbon atoms, preferably 1-10 carbon atoms, 1-4 heteroatoms selected from nitrogen, oxygen and sulfur, at least one aromatic ring. Unless otherwise specified, heteroaryl groups may be monocyclic, bicyclic, tricyclic or tetracyclic ring systems, which may contain fused or bridged ring systems, provided that the point of attachment to the rest of the molecule is an aromatic ring atom, which may be selectively oxidized at nitrogen, carbon and sulfur atoms, and which may optionally be quaternized. For the purposes of the present invention, heteroaryl groups are preferably stable 4-11 membered monocyclic aromatic rings containing 1-3 heteroatoms selected from nitrogen, oxygen and sulfur, more preferably stable 5-8 membered monocyclic aromatic rings containing 1-3 heteroatoms selected from nitrogen, oxygen and sulfur. Non-limiting examples of heteroaryl groups include acridinyl, azepinyl, benzimidazolyl, benzindolyl, benzodioxinyl, benzodioxolyl, benzofuranonyl, benzofuranyl, benzonaphthofuranyl, benzopyranonyl, benzopyranyl, benzopyrazolyl, benzothiadiazolyl, benzothiazolyl, benzotriazolyl, furanyl, imidazolyl, indazolyl, indolyl, oxazolyl, purinyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, quinazolinyl, quinolinyl, quininyl, tetrazolyl, thiadiazolyl, thiazolyl, thienyl, triazinyl, triazolyl, and the like. In the present application, heteroaryl is preferably 5-8 membered heteroaryl comprising 1-3 heteroatoms selected from nitrogen, oxygen and sulfur, more preferably pyridyl, pyrimidinyl, thiazolyl. The heteroaryl group may be substituted or unsubstituted.
"halogen" means fluorine, chlorine, bromine or iodine.
"hydroxy" means-OH, and "amino" means-NH2"amido" means-NHCO-, "cyano" means-CN, and "nitro" means-NO2"isocyano" means-NC and "trifluoromethyl" means-CF3
The term "heteroatom" or "hetero", as used herein alone or as part of another ingredient, refers to atoms other than carbon and hydrogen, and is independently selected from, but not limited to, oxygen, nitrogen, sulfur, phosphorus, silicon, selenium, and tin, and in embodiments where two or more heteroatoms are present, the two or more heteroatoms may be the same as each other, or some or all of the two or more heteroatoms may be different.
The terms "fused" or "fused ring" as used herein, alone or in combination, refer to a cyclic structure in which two or more rings share one or more bonds.
The term "spiro" or "spirocyclic" as used herein, alone or in combination, refers to a cyclic structure in which two or more rings share one or more atoms.
"optionally" or "optionally" means that the subsequently described event or circumstance may, but need not, occur, and that the description includes where the event or circumstance occurs or does not occur-for example, "heterocyclic group optionally substituted with alkyl" means that alkyl may, but need not, be present, and that the description includes instances where the heterocyclic group is substituted with alkyl and instances where the heterocyclic group is not substituted with alkyl.
"substituted" means that one or more atoms, preferably 5, more preferably 1 to 3 atoms, in the group are independently substituted with a corresponding number of substituents. It goes without saying that the skilled person in the art is able to determine (experimentally or theoretically) possible or impossible substitutions without undue effort, when the substituents are in their possible chemical positions. Example (b)For example, having a free amine or hydroxyl group may be unstable in combination with a carbon atom having an unsaturated (e.g., olefinic) bond. Such substituents include, but are not limited to, hydroxy, amine, halogen, cyano, C1-6Alkyl radical, C1-6Alkoxy radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-8Cycloalkyl groups, and the like.
"pharmaceutical composition" refers to a composition containing one or more compounds described herein, or a pharmaceutically acceptable salt or prodrug thereof, and other ingredients such as pharmaceutically acceptable carriers and excipients. The purpose of the pharmaceutical composition is to promote administration to the organism, facilitate absorption of the active ingredient and further exert biological activity.
"isomers" refer to compounds having the same molecular formula but differing in the nature or order of their bonding of atoms or the spatial arrangement of their atoms, referred to as "isomers", and isomers differing in the spatial arrangement of their atoms, referred to as "stereoisomers". Stereoisomers include optical isomers, geometric isomers and conformational isomers. The compounds of the present invention may exist in the form of optical isomers. Depending on the configuration of the substituents around the chiral carbon atom, these optical isomers are either in the "R" or "S" configuration. Optical isomers, including enantiomers and diastereomers, and methods of preparing and separating optical isomers are known in the art.
Geometric isomers may also exist for the compounds of the present invention. The present invention contemplates various geometric isomers and mixtures thereof resulting from the distribution of substituents around carbon-carbon double bonds, carbon-nitrogen double bonds, cycloalkyl or heterocyclic groups. Substituents around carbon-carbon double bonds or carbon-nitrogen bonds are designated as either the Z or E configuration, substituents around cycloalkyl or heterocyclic rings are designated as either the cis or trans configuration.
The compounds of the invention may also exhibit tautomerism, such as keto-enol tautomerism.
It is to be understood that the present invention includes any tautomeric or stereoisomeric form and mixtures thereof, and is not to be limited solely to any one tautomeric or stereoisomeric form employed in the nomenclature or chemical structure of the compounds.
"isotopic" are those present in the compounds of the present inventionAll isotopes of atoms of (a). Isotopes include those atoms having the same atomic number but different mass numbers. Examples of isotopes suitable for incorporation into compounds of the invention are hydrogen, carbon, nitrogen, oxygen, phosphorus, fluorine and chlorine, such as but not limited to2H、3H、13C、14C、15N、18O、31P、32P、35S、18F and36and (4) Cl. Isotopically-labeled compounds of the present invention can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the accompanying examples using appropriate isotopically-labeled reagents in place of non-isotopically-labeled reagents. Such compounds have a variety of potential uses, for example, as standards and reagents in the determination of biological activity. In the case of stable isotopes, such compounds have the potential to favorably alter biological, pharmacological or pharmacokinetic properties.
By "prodrug" is meant that the compounds of the present invention can be administered in the form of a prodrug. Prodrugs refer to derivatives that are converted to the biologically active compounds of the present invention under physiological conditions in vivo, e.g., by oxidation, reduction, hydrolysis, and the like, each of which utilizes or proceeds without the participation of an enzyme. Examples of prodrugs are the following compounds: compounds in which the amine group in the compounds of the invention is acylated, alkylated or phosphorylated, for example eicosanoylamino, propylaminoylamino, pivaloyloxymethylamino, or in which the hydroxyl group is acylated, alkylated, phosphorylated or converted to a borate, for example acetoxy, palmitoyloxy, pivaloyloxy, succinyloxy, fumaroyloxy, propylaminoyloxy, or in which the carboxyl group is esterified or amidated, or in which the sulfhydryl group forms a disulfide bridge with a carrier molecule, for example a peptide, which selectively delivers a drug to the target and/or to the cytosol of the cell, can be prepared from the compounds of the invention according to well-known methods.
"pharmaceutically acceptable salt" or "pharmaceutically acceptable" refers to those made from pharmaceutically acceptable bases or acids, including inorganic bases or acids and organic bases or acids. Where the compounds of the invention contain one or more acidic or basic groups, the invention also includes their corresponding pharmaceutically acceptable salts. Thus, the compounds of the invention containing acidic groups can be present in the form of salts and can be used according to the invention, for example as alkali metal salts, alkaline earth metal salts or as ammonium salts. More specific examples of such salts include sodium, potassium, calcium, magnesium or salts with amines or organic amines, such as primary, secondary, tertiary, cyclic amines, and the like, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, diethanolamine, ethanolamine, dicyclohexylamine, ethylenediamine, purines, piperazine, piperidine, choline, caffeine, and the like, with particularly preferred organic bases being isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine. The compounds of the invention containing basic groups can be present in the form of salts and can be used according to the invention in the form of their addition to inorganic or organic acids. Examples of suitable acids include hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, phosphoric acid, methanesulfonic acid, p-toluenesulfonic acid, naphthalenedisulfonic acid, oxalic acid, acetic acid, tartaric acid, lactic acid, salicylic acid, benzoic acid, formic acid, propionic acid, pivalic acid, malonic acid, succinic acid, pimelic acid, fumaric acid, maleic acid, malic acid, sulfamic acid, phenylpropionic acid, gluconic acid, ascorbic acid, isonicotinic acid, citric acid, adipic acid, and other acids known to those skilled in the art. If the compounds of the invention contain both acidic and basic groups in the molecule, the invention also includes inner salts or betaine salts in addition to the salt forms mentioned. The salts are obtained by conventional methods known to the person skilled in the art, for example by contacting these with organic or inorganic acids or bases in solvents or dispersants or by anion exchange or cation exchange with other salts.
Thus, when reference is made in this application to "a compound", "a compound of the invention" or "a compound of the invention", all said compound forms are included, such as prodrugs, stable isotopic derivatives, pharmaceutically acceptable salts, isomers, meso-forms, racemates, enantiomers, diastereomers and mixtures thereof.
In this context, the term "tumor" includes both benign tumors and malignant tumors (e.g., cancers).
As used herein, the term "cancer" includes various malignancies in which FGFR kinase is involved in its development, including, but not limited to, non-small cell lung cancer, esophageal cancer, melanoma, striated muscle garnet, cellular carcinoma, multiple myeloma, breast cancer ovarian cancer, endometrial cancer, cervical cancer, gastric cancer, colon cancer, bladder cancer, pancreatic cancer, lung cancer, breast cancer, prostate cancer, and liver cancer (e.g., hepatocellular carcinoma), more specifically liver cancer, gastric cancer, and bladder cancer.
The terms "effective amount," "therapeutically effective amount," or "pharmaceutically effective amount" as used herein, refer to an amount of at least one agent or compound that is sufficient to alleviate one or more symptoms of the disease or disorder being treated to some extent after administration. The result may be a reduction and/or alleviation of signs, symptoms, or causes or any other desired change in a biological system. For example, an "effective amount" for treatment is the amount of a composition comprising a compound disclosed herein that is clinically necessary to provide a significant remission effect of the condition. An effective amount suitable in any individual case can be determined using techniques such as a dose escalation assay.
The term "polymorph" or "polymorph" as used herein means that the compounds of the present invention have multiple crystal lattice forms, some of the compounds of the present invention may have more than one crystal form, and the present invention encompasses all polymorphic forms or mixtures thereof.
Intermediate compounds of the present invention and polymorphs thereof are also within the scope of the present invention.
Crystallization often results in a solvate of a compound of the present invention, and the term "solvate" as used herein refers to an association of one or more molecules of a compound of the present invention and one or more molecules of a solvent.
The solvent may be water, in which case the solvate is a hydrate. In addition, an organic solvent may be used. Thus, the compounds of the present invention may exist as hydrates, including monohydrate, dihydrate, hemihydrate, trihydrate, tetrahydrate and the like, as well as the corresponding solvated forms. The compounds of the invention may be true solvates, but in other cases the compounds of the invention may also be present only occasionally as water or as a mixture of water with some other solvent the compounds of the invention may be reacted in a solvent or precipitated or crystallized in a solvent. Solvates of the compounds of the invention are also included within the scope of the invention.
As used herein, the term "acceptable" in reference to a formulation, composition or ingredient means that there is no lasting deleterious effect on the overall health of the subject being treated.
The term "pharmaceutically acceptable" as used herein refers to a substance (e.g., carrier or diluent) that does not affect the biological activity or properties of the compounds of the present invention and is relatively non-toxic, i.e., the substance can be administered to an individual without causing an adverse biological response or interacting in an adverse manner with any of the components contained in the composition.
"pharmaceutically acceptable carriers" include, but are not limited to, adjuvants, carriers, excipients, adjuvants, deodorants, diluents, preservatives, dyes/colorants, flavor enhancers, surfactants and wetting agents, dispersants, suspending agents, stabilizers, isotonizing agents, solvents, or emulsifiers that have been approved by the relevant governmental authorities for use in humans and domestic animals.
As used herein, the term "subject", "patient", "subject" or "individual" refers to an individual suffering from a disease, disorder or condition, and the like, including mammals and non-mammals, examples of which include, but are not limited to, any member of the class mammalia: humans, non-human primates (e.g., chimpanzees and other apes and monkeys); livestock, such as cattle, horses, sheep, goats, pigs; domestic animals such as rabbits, dogs, and cats; laboratory animals, including rodents, such as rats, mice, and guinea pigs, and the like. Examples of non-human mammals include, but are not limited to, birds, fish, and the like. In one embodiment related to the methods and compositions provided herein, the mammal is a human.
The term "treatment" as used herein refers to the treatment of a disease condition associated with a mammal, particularly a human, and includes
(i) Preventing the development of a disease or condition in a mammal, particularly a mammal that has been previously exposed to the disease or condition but has not been diagnosed as having the disease or condition;
(ii) inhibiting the disease or disorder, i.e., controlling its development;
(iii) relieving the disease or condition, i.e., slowing the regression of the disease or condition;
(iv) relieving symptoms caused by the disease or disorder.
The terms "disease" and "condition" as used herein may be used interchangeably and may have different meanings, as certain specific diseases or conditions have no known causative agent (and therefore the cause of the disease is not yet clear) and therefore are not considered as a disease but can be considered as an unwanted condition or syndrome, with more or less specific symptoms being confirmed by clinical researchers.
The terms "administering," "administration," "administering," and the like as used herein refer to methods that are capable of delivering a compound or composition to a desired site for biological action. Including, but not limited to, oral, via the duodenal route, parenteral injection (including intravenous, subcutaneous, intraperitoneal, intramuscular, intraarterial injection or infusion), topical administration, and rectal administration. In preferred embodiments, the compounds and compositions discussed herein are administered orally.
Synthesis method
The invention also provides a method for preparing the compound. The preparation of the compounds of the general formula (I) according to the invention can be carried out by the following exemplary methods and examples, which should not be construed as limiting the scope of the invention in any way. The compounds of the invention may also be synthesized by synthetic techniques known to those skilled in the art, or a combination of methods known in the art and those described herein may be used. The product of each step is obtained by separation techniques known in the art, including but not limited to extraction, filtration, distillation, crystallization, chromatography, and the like. The starting materials and chemical reagents required for the synthesis can be routinely synthesized or purchased according to the literature (reaxys).
Detailed Description
The starting materials and reagents used in the present invention are commercially available.
The invention is further illustrated by the following specific examples.
Example 1: preparation of N- (3- ((5- (phenylethynyl) pyrimidin-4-yl) oxy) phenyl) acrylamide (compound 101)
Figure BDA0002909122720000151
Step 1: synthesis of Compound 2
To a reaction flask were added compound 1(1.49g,10.0mmol), N-tert-butoxycarbonyl-3-aminophenol (2.09g,10.0mmol), potassium carbonate (2.07g,15.0mmol) and 30ml of N, N-dimethylformamide. The reaction was carried out at 60 ℃ for 2 hours with stirring. Cooled to room temperature, the reaction solution was diluted with ethyl acetate and water, and extracted with ethyl acetate. The organic phase was washed with water and saturated brine, dried over anhydrous sodium sulfate, and evaporated to dryness under reduced pressure. The residue was purified by column chromatography to give compound 2(2.73g, 85% yield).
Step 2: synthesis of Compound 3
To a reaction flask were added compound 2(1.61g,5.0mmol), phenylacetylene (2.09g,10.0mmol), bis (triphenylphosphine) palladium dichloride (351mg,0.5mmol), cuprous iodide (95mg,0.5mmol), triethylamine (2.53g,25.0mmol) and 40ml of N, N-dimethylformamide. The mixture was purged with nitrogen 3 times, and reacted at 80 ℃ overnight with stirring. Cooled to room temperature, the reaction solution was diluted with ethyl acetate and water, and extracted with ethyl acetate. The organic phase was washed with water and saturated brine, dried over anhydrous sodium sulfate, and evaporated to dryness under reduced pressure. The residue was purified by column chromatography to give compound 3(1.51g, yield 78%).
And step 3: synthesis of Compound 4
Intermediate 3(0.77g,2.0mmol), 4ml ethyl acetate, 4ml of 4N HCl in 1, 4-dioxane were added to the reaction flask. After stirring at room temperature for 2 hours, the reaction solution was neutralized with 1N sodium hydroxide solution and extracted with ethyl acetate. The organic phase was washed with saturated sodium bicarbonate and saturated brine, dried over anhydrous sodium sulfate, and the organic phase was evaporated to dryness under reduced pressure. Compound 4(0.54g, 95% yield) was obtained and used directly in the next step.
And 4, step 4: synthesis of Compound 101
Compound 4(287mg,1.0mmol), triethylamine (152mg,1.5mmol), and 4ml of tetrahydrofuran were added to a reaction flask, and after cooling in an ice-water bath, a solution of acryloyl chloride (136mg,1.5mmol) in 0.5ml of tetrahydrofuran was slowly added dropwise. Stirring was continued for 2 hours after the addition was complete. The reaction solution was quenched with methanol and evaporated to dryness under reduced pressure. The residue was purified by column chromatography to give compound 101(123mg, yield 35%) as a pale yellow solid.1H NMR(400MHz,DMSO-d6)δ:10.13(s,1H),8.97(s,1H),8.32(s,1H),7.52-7.41(m,5H),7.30-7.19(m,3H),6.73-6.70(m,1H),6.43(dd,1H),6.26(dd,1H),5.61(d,1H);LC/MS(ESI):m/z=342[M+H]+.
Example 2: preparation of N- (3- ((5- ((4-methoxybenzene) ethynyl) pyrimidin-4-yl) oxy) phenyl) acrylamide (compound 102)
Figure BDA0002909122720000161
The same procedure as in example 1 (intermediate was changed to p-methoxyphenylacetylene) was carried out to give compound 102(139mg, yield 42%, which is the final yield, the same applies hereinafter) as a pale yellow solid.1H NMR(400MHz,DMSO-d6)δ:10.14(s,1H),8.95(s,1H),8.31(s,1H),7.40-7.21(m,5H),6.98-6.70(m,3H),6.41(dd,1H),6.24(dd,1H),5.69(dd,1H),3.77(s,3H);LC/MS(ESI):m/z=372.1[M+H]+.
Example 3: preparation of N- (3- ((5- ((3, 5-difluorophenyl) ethynyl) pyrimidin-4-yl) oxy) phenyl) acrylamide (compound 103)
Figure BDA0002909122720000162
The same procedure as in example 1 (intermediate was changed to 3, 5-difluorophenylacetylene) was used to obtain compound 103(114mg, yield 32%) as a pale yellow solid.1HNMR(400MHz,DMSO-d6)δ:10.12(s,1H),8.96(s,1H),8.32(s,1H),7.30-7.18(m,3H),6.93-6.70(m,4H),6.42(dd,1H),6.19(dd,1H),5.63(dd,1H);
LC/MS(ESI):m/z=378.0[M+H]+.
Example 4: preparation of N- (3- ((5- ((3, 5-dimethoxybenzene) ethynyl) pyrimidin-4-yl) oxy) phenyl) acrylamide (compound 104)
Figure BDA0002909122720000171
The same procedure as in example 1 (intermediate was changed to 3, 5-dimethoxyphenylacetylene) was used to obtain compound 104(152mg, yield 46%) as a pale yellow solid.1H NMR(400MHz,DMSO-d6)δ:10.14(s,1H),8.96(s,1H),8.33(s,1H),7.30-7.19(m,3H),7.08(s,2H),6.72-6.69(m,1H),6.41-6.36(m,2H),6.21(dd,1H),5.65(dd,1H),3.77(s,6H);LC/MS(ESI):m/z=402.1[M+H]+.
Example 5: preparation of N- (3- ((6-amino-5- ((3, 5-dimethoxybenzene) ethynyl) pyrimidin-4-yl) oxy) phenyl) acrylamide (compound 105)
Figure BDA0002909122720000172
In a similar manner to example 4 (starting material was changed to 4-amino-5, 6-dichloropyrimidine), compound 105(99mg, yield 20%) was obtained as a yellow solid.1HNMR(400MHz,DMSO-d6)δ:10.14(s,1H),8.13(s,1H),7.31-7.17(m,3H),7.09(s,2H),6.72-6.69(m,1H),6.53(br s,2H),6.41-6.32(m,2H),6.23(dd,1H),5.64(dd,1H),3.77(s,6H);LC/MS(ESI):m/z=417.1[M+H]+.
Example 6: preparation of N- (3- ((3- ((3, 5-dimethoxybenzene) ethynyl) pyridin-4-yl) oxy) phenyl) acrylamide (Compound 106)
Figure BDA0002909122720000173
In a similar manner to example 4 (starting material was changed to 3, 4-dichloropyridine), compound 106(135mg, yield 41%) was obtained as a pale yellow solid.1HNMR(400MHz,DMSO-d6)δ:10.13(s,1H),8.76(s,1H),8.09(d,1H),7.32-7.18(m,3H),7.05(s,2H),6.72(d,1H),6.44-6.33(m,3H),6.23(dd,1H),5.61(dd,1H),3.77(s,6H);LC/MS(ESI):m/z=401.1[M+H]+.
Example 7: preparation of N- (3- ((4- ((3, 5-dimethoxybenzene) ethynyl) pyrimidin-5-yl) oxy) phenyl) acrylamide (compound 107)
Figure BDA0002909122720000181
In a similar manner to example 4 (starting material was changed to 4, 5-dichloropyrimidine), compound 107(119mg, yield 32%) was obtained as a yellow solid.1H NMR(400MHz,DMSO-d6)δ:10.13(s,1H),8.70(s,1H),8.33(s,1H),7.31-7.17(m,3H),7.08(s,2H),6.44-6.36(m,3H),6.21(dd,1H),5.64(dd,1H),3.77(s,6H);LC/MS(ESI):m/z=402.2[M+H]+.
Example 8: preparation of N- (3- ((3- ((3, 5-dimethoxybenzene) ethynyl) pyrazin-2-yl) oxy) phenyl) acrylamide (compound 108)
Figure BDA0002909122720000182
In a similar manner to example 4 (starting material was changed to 2, 3-dichloropyrazine) gave compound 108(105mg, yield 25%) as a yellow solid.1H NMR(400MHz,DMSO-d6)δ:10.13(s,1H),7.55(d,1H),7.45(d,1H),7.32-7.17(m,3H),7.08(s,2H),6.71-6.69(m,1H),6.45(dd,1H),6.35(s,1H),6.21(dd,1H),5.64(dd,1H),3.77(s,6H);LC/MS(ESI):m/z=402[M+H]+.
Example 9: preparation of N- (3- ((4-amino-6- ((3, 5-dimethoxybenzene) ethynyl) pyrimidin-2-yl) oxy) phenyl) acrylamide (compound 109)
Figure BDA0002909122720000183
In a similar manner to example 4 (starting material was changed to 4-amino-2, 6-diChloropyrimidine) gave compound 109(123mg, 26% yield) as a yellow solid.1HNMR(400MHz,DMSO-d6)δ:10.12(s,1H),7.32-7.17(m,3H),7.09(s,2H),6.70-6.68(m,1H),6.58(br s,2H),6.47(dd,1H),6.35(s,1H),6.23(dd,1H),5.82(s,1H),5.60(dd,1H),3.77(s,6H);LC/MS(ESI):m/z=417.1[M+H]+.
Example 10: preparation of N- (3- ((4-amino-6- ((3, 5-dimethoxybenzene) ethynyl) pyrimidin-2-yl) amino) phenyl) acrylamide (compound 110)
Figure BDA0002909122720000191
Step 1: synthesis of Compound 2a
Figure BDA0002909122720000192
Into a reaction flask were charged compound 1a (1.49g,10.0mmol), 1, 3-diphenylamine (2.09g,10.0mmol), premixed trifluoromethanesulfonic acid (1.50g,10.0mmol) and triethylamine (1.01g,10.0mmol), and 30ml of dimethyl sulfoxide. The reaction was allowed to proceed overnight at 90 ℃ with stirring. Cooled to room temperature, the reaction solution was diluted with ethyl acetate and water, and extracted with ethyl acetate. The organic phase was washed with water and saturated brine, dried over anhydrous sodium sulfate, and evaporated to dryness under reduced pressure. The residue was purified by column chromatography to give compound 2(1.53g, yield 65%).
The subsequent two steps were carried out in a similar manner to example 4 (arylyne and acryloyl, respectively) to give compound 110(89mg, 23% yield) as a yellow solid.1HNMR(400MHz,DMSO-d6)δ:10.13(s,1H),7.73(s,1H),7.35-7.29(m,1H),7.20-7.18(m,1H),7.08(s,2H),6.98-6.95(m,2H),6.61(br s,2H),6.47(dd,1H),6.35(s,1H),6.21(dd,1H),5.89(s,1H),5.60(dd,1H),3.77(s,6H);LC/MS(ESI):m/z=416.2[M+H]+.
Example 11: preparation of N- (3- (4-amino-6- ((3, 5-dimethoxybenzene) ethynyl) pyrimidin-5-yl)) phenyl) acrylamide (compound 111)
Figure BDA0002909122720000193
Step 1: synthesis of Compound 2b
Figure BDA0002909122720000201
Compound 1b (0.82g,5.0mmol), 3-aminophenylboronic acid (0.82g,6.0mmol), cesium carbonate (3.26g,10.0mmol), 40ml of 1, 4-dioxane, and 4ml of water were charged into a microwave reaction tube. After 3 times replacement with nitrogen, palladium acetate (56mg,0.25mmol) and 2-dicyclohexylphosphine-2 ',6' -dimethoxybiphenyl (205mg,0.5mmol) were added and reacted at 150 ℃ for 2 hours under microwave. Cooling to room temperature, passing the reaction solution through a silica gel short column, leaching with ethyl acetate, and evaporating the obtained solution under reduced pressure. The residue was purified by column chromatography to give compound 2b (0.64g, 58% yield).
The subsequent two steps were carried out in a similar manner to example 4 (arylyne and acryloyl, respectively) to give compound 110(115mg, 21% yield) as a yellow solid.1HNMR(400MHz,DMSO-d6)δ:10.12(s,1H),8.45(s,1H),7.35-7.22(m,3H),7.09(s,2H),6.65(br s,2H),6.47(dd,1H),6.34(s,1H),6.15(dd,1H),5.89(s,2H),5.62(dd,1H),3.77(s,6H);LC/MS(ESI):m/z=401.2[M+H]+.
Example 12: preparation of N- (3- (4-amino-6- ((3, 5-dimethoxyphenyl) ethynyl) pyrimidin-5-yl)) phenyl) acrylamide (compound 112)
Figure BDA0002909122720000202
In a similar manner to example 10 (starting material was changed to 4-amino-5, 6-dichloropyrimidine), compound 112(107mg, yield 28%) was obtained as a yellow solid.1HNMR(400MHz,DMSO-d6)δ:10.13(s,1H),8.24(s,1H),7.37-7.30(m,1H),7.20-7.18(m,1H),7.08(s,2H),6.98-6.95(m,2H),6.68(br s,2H),6.47(dd,1H),6.36(s,1H),6.21(dd,1H),5.89(s,1H),5.59(dd,1H),3.77(s,6H);LC/MS(ESI):m/z=416.1[M+H]+.
Example 13: preparation of 1- (3- (4-amino-6- ((3, 5-dimethoxybenzene) ethynyl) pyrimidin-2-yl) amino) pyrrolidin-1-yl) -2-propenyl-1-one (compound 113)
Figure BDA0002909122720000203
Step 1: synthesis of Compound 2c
Figure BDA0002909122720000211
To a reaction flask were added compound 1c (1.64g,10.0mmol), tert-butyl 3-aminopyrrolidine-1-carboxylate (2.24g,12.0mmol), 1, 8-diazabicycloundec-7-ene (2.28g,15.0mmol) and 30ml of N, N-dimethylformamide. The reaction was allowed to proceed overnight at 90 ℃ with stirring. Cooled to room temperature, the reaction solution was diluted with ethyl acetate and water, and extracted with ethyl acetate. The organic phase was washed with water and saturated brine, dried over anhydrous sodium sulfate, and evaporated to dryness under reduced pressure. The residue was purified by column chromatography to give compound 2(2.54g, 81% yield).
The subsequent three steps in a similar manner to example 4 (arylyne, deprotection group and acryloyl group, respectively) gave compound 113(115mg, yield 18%) as a pale yellow solid.1H NMR(400MHz,DMSO-d6)δ:7.08(s,2H),6.67(br s,2H),6.47(dd,1H),6.35(s,1H),6.21(dd,1H),5.89(s,1H),5.45(dd,1H),4.24(s,1H),3.82-3.31(m,10H),2.78-2.55(m,1H),1.98-1.75(m,2H);LC/MS(ESI):m/z=394.1[M+H]+.
Example 14: preparation of 1- (3- (4-amino-6- ((3, 5-dimethoxybenzene) ethynyl) pyrimidin-2-yl) amino) piperidin-1-yl) -2-propenyl-1-one (compound 114)
Figure BDA0002909122720000212
Using a method similar to example 13 (intermediate was changed to 1-tert-butoxycarbonyl-3-aminopiperidine), compound 114(103mg, yield 12%)) As a pale yellow solid.1H NMR(400MHz,DMSO-d6)δ:7.08(s,2H),6.67(br s,2H),6.50(dd,1H),6.35(s,1H),6.19(dd,1H),5.86(s,1H),5.42(dd,1H),4.34(s,1H),3.86-3.31(m,10H),2.68-2.61(m,1H),1.88-1.45(m,4H);LC/MS(ESI):m/z=408.2[M+H]+.
Example 15: preparation of 1- (4- (4-amino-6- ((3, 5-dimethoxybenzene) ethynyl) pyrimidin-2-yl) amino) piperidin-1-yl) -2-propenyl-1-one (compound 115)
Figure BDA0002909122720000221
Using a method similar to example 13 (intermediate was changed to 1-tert-butoxycarbonyl-4-aminopiperidine), compound 115(101mg, yield 11%) was obtained as a pale yellow solid.1H NMR(400MHz,DMSO-d6)δ:7.08(s,2H),6.68(br s,2H),6.55(dd,1H),6.35(s,1H),6.19(dd,1H),5.86(s,1H),5.32(dd,1H),4.24(s,1H),3.82(s,6H),3.05-2.91(m,4H),2.65-2.61(m,1H),1.82-1.55(m,4H);LC/MS(ESI):m/z=408.1[M+H]+.
Example 16: preparation of 1- (2- (4-amino-6- ((3, 5-dimethoxybenzene) ethynyl) pyrimidin-2-yl) amino) -7-azaspiro [3.5] nonan-7-yl) -2-propenyl-1-one (compound 116)
Figure BDA0002909122720000222
By a method similar to example 13 (intermediate exchanged for 2-amino-7-azaspiro [3.5]]Nonane-7-carboxylic acid tert-butyl ester) gave compound 116(108mg, 12% yield) as a pale yellow solid.1H NMR(400MHz,DMSO-d6)δ:7.08(s,2H),6.68(br s,2H),6.58(dd,1H),6.35(s,1H),6.12(dd,1H),5.87(s,1H),5.32(dd,1H),4.20(s,1H),3.82(s,6H),3.55-3.41(m,4H),3.15-3.04(m,1H),2.15-1.95(m,4H),1.55-1.43(m,4H);LC/MS(ESI):m/z=448.2[M+H]+.
Example 17: preparation of 1- (6- (4-amino-6- ((3, 5-dimethoxybenzene) ethynyl) pyrimidin-2-yl) amino) -2-azaspiro [3.3] heptan-2-yl) -2-propenyl-1-one (compound 117)
Figure BDA0002909122720000231
Using a method similar to example 13 (intermediate exchanged for 6-amino-2-azaspiro [3.3]]Heptane-2-carboxylic acid tert-butyl ester) gave compound 117(115mg, yield 15%) as a pale yellow solid.1H NMR(400MHz,DMSO-d6)δ:7.08(s,2H),6.68(br s,2H),6.59(dd,1H),6.35(s,1H),6.02(dd,1H),5.87(s,1H),5.34(dd,1H),4.18(s,1H),3.82(s,6H),3.65-3.61(m,4H),3.12-3.05(m,1H),2.18-1.95(m,4H);LC/MS(ESI):m/z=420.2[M+H]+.
Example 18: preparation of 1- (2- (4-amino-6- ((3, 5-dimethoxybenzene) ethynyl) pyrimidin-2-yl) amino) -6-azaspiro [3.4] octan-6-yl) -2-propenyl-1-one (compound 118)
Figure BDA0002909122720000232
By a method similar to example 13 (intermediate exchanged for 2-amino-6-azaspiro [3.4]]Octane-6-carboxylic acid tert-butyl ester) gave compound 118(115mg, yield 21%) as a pale yellow solid.1H NMR(400MHz,DMSO-d6)δ:7.08(s,2H),6.68(br s,2H),6.59(dd,1H),6.35(s,1H),6.12(dd,1H),5.86(s,1H),5.34(dd,1H),4.23(s,1H),3.82(s,6H),3.25-3.21(m,4H),3.11-3.05(m,1H),2.19-1.95(m,4H),1.61-1.53(m,2H);LC/MS(ESI):m/z=434.1[M+H]+.
Example 19: preparation of 1- (4- (6-amino-5- ((3, 5-dimethoxybenzene) ethynyl) pyrimidin-4-yl) amino) piperidin-1-yl) -2-propenyl-1-one (compound 119)
Figure BDA0002909122720000241
Using a method similar to example 12 (starting material was changed to 4-amino-5, 6-dichloropyrimidine and intermediate was changed to 1-tert-butoxycarbonyl-4-aminomethylpiperidine), compound 119(95mg, yield 10%) was obtained as a pale yellow solidA colored solid.1H NMR(400MHz,DMSO-d6)δ:8.23(s,1H),7.08(s,2H),6.70(br s,2H),6.56(dd,1H),6.35(s,1H),6.12(dd,1H),5.32(dd,1H),4.20(s,1H),3.82(s,6H),3.35-3.28(m,4H),3.05-2.98(m,2H),0.92-1.75(m,5H);LC/MS(ESI):m/z=422.2[M+H]+.
Example 20: preparation of 1- (3- (6-amino-5- ((3, 5-dimethoxybenzene) ethynyl) pyrimidin-4-yl) amino) pyrrolidin-1-yl) -2-propenyl-1-one (compound 120)
Figure BDA0002909122720000242
In a similar manner to example 12 (starting material was changed to 4-amino-5, 6-dichloropyrimidine and intermediate was changed to tert-butyl 3-aminopyrrolidine-1-carboxylate) was used to give compound 120(142mg, yield 21%) as a pale yellow solid.1H NMR(400MHz,DMSO-d6)δ:8.23(s,1H),7.08(s,2H),6.70(br s,2H),6.56(dd,1H),6.35(s,1H),6.12(dd,1H),5.32(dd,1H),4.34(s,1H),3.82(s,6H),3.55-3.31(m,4H),2.75-2.71(m,1H),1.92-1.65(m,2H);LC/MS(ESI):m/z=394.1[M+H]+.
Example 21: preparation of 1- (3- (6-amino-5- ((3, 5-dimethoxybenzene) ethynyl) pyrimidin-4-yl) amino) piperidin-1-yl) -2-propenyl-1-one (compound 121)
Figure BDA0002909122720000251
In a similar manner to example 12 (starting material was changed to 4-amino-5, 6-dichloropyrimidine and intermediate was changed to tert-butyl 3-aminopiperidine-1-carboxylate) was used to obtain compound 121(118mg, yield 13%) as a pale yellow solid.1H NMR(400MHz,DMSO-d6)δ:8.23(s,1H),7.08(s,2H),6.69(br s,2H),6.58(dd,1H),6.35(s,1H),6.09(dd,1H),5.34(dd,1H),4.31(s,1H),3.82(s,6H),3.65-3.30(m,4H),2.65-2.61(m,1H),1.82-1.45(m,4H);LC/MS(ESI):m/z=408.2[M+H]+.
Example 22: preparation of 1- (6- (6-amino-5- ((3, 5-dimethoxybenzene) ethynyl) pyrimidin-4-yl) amino) -1-azaspiro [3.3] heptan-1-yl) -2-propenyl-1-one (compound 122)
Figure BDA0002909122720000252
In a manner similar to that of example 12 (starting material was changed to 4-amino-5, 6-dichloropyrimidine and intermediate was changed to 6-amino-1-azaspiro [3.3]]Heptane-1-carboxylic acid tert-butyl ester) gave compound 122(126mg, yield 20%) as a light yellow solid.1H NMR(400MHz,DMSO-d6)δ:8.22(s,1H),7.08(s,2H),6.68(br s,2H),6.55(dd,1H),6.35(s,1H),6.12(dd,1H),5.35(dd,1H),4.22(s,1H),3.82(s,6H),3.55-3.51(m,2H),3.12-3.07(m,1H),2.49-2.15(m,6H);LC/MS(ESI):m/z=420.1[M+H]+.
Example 23: preparation of 1- (2- (6-amino-5- ((3, 5-dimethoxybenzene) ethynyl) pyrimidin-4-yl) amino) -6-azaspiro [3.4] octan-6-yl) -2-propenyl-1-one (compound 123)
Figure BDA0002909122720000261
In a manner similar to that of example 12 (starting material was changed to 4-amino-5, 6-dichloropyrimidine and intermediate was changed to 2-amino-6-azaspiro [3.4]]Octane-6-carboxylic acid tert-butyl ester) gave compound 123(132mg, yield 23%) as a pale yellow solid.1H NMR(400MHz,DMSO-d6)δ:8.23(s,1H),7.08(s,2H),6.70(br s,2H),6.57(dd,1H),6.35(s,1H),6.10(dd,1H),5.34(dd,1H),4.09(s,1H),3.82(s,6H),3.27-3.21(m,4H),3.11-3.05(m,1H),2.19-1.94(m,4H),1.59-1.50(m,2H);LC/MS(ESI):m/z=434.2[M+H]+.
Example 24: preparation of 1- (3- (6-amino-5- ((3, 5-dimethoxybenzene) ethynyl) pyrimidin-4-yl) amino) pyrrolidin-1-yl) -2-propenyl-1-one (compound 124)
Figure BDA0002909122720000262
In a manner similar to that of example 12 (seeThe starting material was changed to 4-amino-5, 6-dichloropyrimidine and the intermediate was changed to 1-tert-butoxycarbonyl-3- (aminomethyl) pyrrolidine) to give compound 124(125mg, yield 20%) as a pale yellow solid.1H NMR(400MHz,DMSO-d6)δ:8.23(s,1H),7.08(s,2H),6.69(br s,2H),6.58(dd,1H),6.35(s,1H),6.09(dd,1H),5.34(dd,1H),4.25(s,1H),3.82(s,6H),3.45-3.18(m,4H),3.15-2.98(m,2H),1.82-1.41(m,3H);LC/MS(ESI):m/z=408.2[M+H]+.
Example 25: preparation of 1- (4- (((6-amino-5- ((3, 5-dimethoxyphenyl) ethynyl) pyrimidin-4-yl) amino) methyl) piperidin-1-yl) -2-butyn-1-one (compound 125)
Figure BDA0002909122720000271
Intermediate 5- ((3, 5-Dimethoxybenzene) ethynyl) -N4- (piperidin-4-ylmethyl) pyrimidine-4, 6-diamino and 2-butynoyl chloride) was used in example 19 to give compound 125(108mg, 19% yield) as a pale yellow solid.1H NMR(400MHz,DMSO-d6)δ:8.23(s,1H),7.08(s,2H),6.70(br s,2H),6.35(s,1H),4.25(s,1H),3.82(s,6H),3.41-3.28(m,4H),3.05-2.99(m,2H),1.82(s,3H),1.78-1.34(m,5H);LC/MS(ESI):m/z=434.2[M+H]+.
Example 26: (E) preparation of (E) -1- (4- (((6-amino-5- ((3, 5-dimethoxyphenyl) ethynyl) pyrimidin-4-yl) amino) methyl) piperidin-1-yl) -3-methoxy-2-propenyl-1-one (compound 126)
Figure BDA0002909122720000272
The intermediate 5- ((3, 5-dimethoxybenzene) ethynyl) -N4- (piperidin-4-ylmethyl) pyrimidine-4, 6-diamino and (E) -3-methoxyacryloyl chloride) was used in example 19 to give compound 126(108mg, 12% yield) as a pale yellow solid.1H NMR(400MHz,DMSO-d6)δ:8.23(s,1H),7.28(d,1H),7.08(s,2H),6.69(br s,2H),6.35(s,1H),5.54(d,1H),4.21(s,1H),3.82(s,6H),3.77(s,3H)3.45-3.30(m,4H),3.09-2.99(m,2H),1.78-1.35(m,5H);LC/MS(ESI):m/z=452.1.[M+H]+.
Example 27: preparation of 1- (4- (((6-amino-5- ((3, 5-dimethoxyphenyl) ethynyl) pyrimidin-4-yl) amino) methyl) piperidin-1-yl) -2-propynyl-1-one (compound 127)
Figure BDA0002909122720000281
The intermediate 5- ((3, 5-dimethoxybenzene) ethynyl) -N4- (piperidin-4-ylmethyl) pyrimidine-4, 6-diamino and propiolic chloride) of example 19 was used to give compound 127(124mg, yield 23%) as a pale yellow solid.1H NMR(400MHz,DMSO-d6)δ:8.23(s,1H),7.08(s,2H),6.70(br s,2H),6.35(s,1H),4.25(s,1H),3.77(s,6H),3.39-3.28(m,4H),3.05-2.98(m,2H),2.72(s,1H),1.78-1.34(m,5H);LC/MS(ESI):m/z=420.1[M+H]+.
Example 28: ((3, 5-Dimethoxybenzene) ethynyl) -N4Preparation of (- (1-vinylsulfonyl) piperidin-4-yl) methyl) pyrimidine-4, 6-diamine) (Compound 128)
Figure BDA0002909122720000282
Intermediate 5- ((3, 5-dimethoxybenzene) ethynyl) -N4- (piperidin-4-ylmethyl) pyrimidine-4, 6-diamino and vinylsulfonyl chloride) was used in example 19 to give compound 128(146mg, 22% yield) as a pale yellow solid.1H NMR(400MHz,DMSO-d6)δ:8.23(s,1H),7.08(s,2H),6.94(dd,1H),6.68(br s,2H),6.35(s,1H),6.18(dd,1H),5.84(dd,1H),4.22(s,1H),3.79(s,6H),3.09-2.90(m,6H),1.76-1.32(m,5H);LC/MS(ESI):m/z=458.2[M+H]+.
Example 29: preparation of N- (1- (6-amino-5- ((3, 5-dimethoxyphenyl) ethynyl) pyrimidin-4-yl) pyrrolidin-3-yl) allylamide (compound 129)
Figure BDA0002909122720000291
In and out ofThe procedure similar to example 12 (starting material was changed to 4-amino-5, 6-dichloropyrimidine and intermediate was changed to 3-tert-butoxycarbonylaminopyrrolidine) gave compound 129(145mg, yield 25%) as a pale yellow solid. H NMR (400MHz, DMSO-d)6)δ:8.16(s,1H),7.71-7.10(m,4H),6.74(dd,1H),6.58-6.46(m,2H),5.60-5.50(m,1H),4.22(s,1H),3.79(s,6H),3.78-2.78(m,5H),1.94-1.62(m,2H);LC/MS(ESI):m/z=494.2[M+H]+.
Example 30: preparation of N- (1- (6-amino-5- ((3, 5-dimethoxyphenyl) ethynyl) pyrimidin-4-yl) piperidin-3-yl) allylamide (compound 130)
Figure BDA0002909122720000292
In a similar manner to example 12 (starting material was changed to 4-amino-5, 6-dichloropyrimidine and intermediate was changed to 3-tert-butoxycarbonylaminopiperidine), compound 130(129mg, yield 21%) was obtained as a pale yellow solid. H NMR (400MHz, DMSO-d)6)δ:8.16(s,1H),7.71-7.10(m,4H),6.74(dd,1H),6.58-6.46(m,2H),5.60-5.50(m,1H),4.22(s,1H),3.79(s,6H),3.29-2.90(m,5H),1.90-1.28(m,4H);LC/MS(ESI):m/z=408.2[M+H]+.
Example 31: in vitro activity inhibition assay for kinases FGFR1, FGFR2, FGFR3 and FGFR4
FGFR1, FGFR2, FGFR3 and FGFR4 protein kinase activities were determined by using a Caliper mobility shift assay (Caliper mobility shift assay). Compounds were dissolved in DMSO and diluted with kinase buffer, and 5 μ L of compound (10% DMS0) at 5-fold final reaction concentration was added to the 384-well plate. After adding 10. mu.L of a 2.5-fold enzyme solution (FGFR 1, FGFR2, FGFR3 and FGFR4, respectively), the mixture was incubated at room temperature for 10 minutes, and then 10. mu.L of a 2.5-fold substrate (FAM-labeled peptide and dATP) solution was added. After incubation at 28 ℃ for 30-60 minutes, 25. mu.L of stop buffer (pH 7.5100mM HEPES, 0.015% Brij-35, 0.2% Coating Reagent #3,50mM EDTA) was added to stop the reaction. Conversion data were read on a Caliper EZ Reader II (Caliper Life Sciences). The conversion was converted to inhibition data (% inhibition ═ max-sample conversion)/(max-min) × 100). Where max refers to DMSO control transferConversion rate, min, refers to the conversion rate of the enzyme-inactive control. The concentration and the inhibition rate of the compound are used as horizontal and vertical coordinates to draw a curve, XLFit excel add-in version4.3.1 software is used for fitting the curve and calculating IC50. The results of the assay are shown in the following table showing activity data of compounds 1-28 for the kinases FGFR1, FGFR2, FGFR3 and FGFR 4. Active utilization of IC50Characterization, wherein "A" represents IC50Less than or equal to 10 nM; "B" means 10<IC50Less than or equal to 100 nM; "C" means 100<IC50Less than or equal to 500 nM; "D" means 500<IC50≤2000nM。
Table 1 inhibitory activity against FGFR1, FGFR2, FGFR3 and FGFR4
Figure BDA0002909122720000301
Figure BDA0002909122720000311
Example 32: human hepatoma cell Hep3B survival assay
The human liver cancer Hep3B cell line is derived from ATCC. The cells were cultured in DMEM liquid medium, and fetal bovine serum (10% FBS) and penicillin-streptomycin (100,000U/L) were additionally added. The cells were maintained in culture at 37 ℃, 95% humidity and 5% carbon dioxide. For the experiments Hep3B cells were plated at a density of 3000 cells per well in 96-well plates at a cell suspension volume of 100PL per well and cells were cultured overnight to allow cell attachment. The following day, each compound was diluted in DMSO in a three-fold gradient, and a 1PL compound DMSO solution was added to the cell culture medium, with IM DMSO as a control, with three parallel side wells for each compound concentration. Cells were then placed in a 37 ℃ incubator and after 72 consecutive hours of compound treatment, 50 μ L of CellTiter-Glo (Promega, Madison Wis.) was added to the cell culture medium and the Relative Luminescence Units (RLU) of each well were determined and cell viability and compound activity (IC) were calculated50) Wherein "A" represents IC50Less than or equal to 10 nM; "B" means 10<IC50Less than or equal to 100 nM; "C" means 100<IC50Less than or equal to 500 nM; 'D' tableDisplay 500<IC50Less than or equal to 2000 nM. The results of the inhibitory activity of the example compounds on Hep3B cells are shown in table 2 below:
TABLE 2 inhibitory Activity on Hep3B cells
Sample numbering IC50(nM)
120 A
121 A
129 B
130 A

Claims (7)

1. A novel FGFR inhibitor with general structural formula (I) or its isomer, stable isotope derivative, hydrate, solvate, polymorph, and pharmaceutically acceptable salt
Figure FDA0002909122710000011
Wherein:
x1, X2, X3, X4 and X5 are each independently CR1Or N, and at least one of X1, X2, X3, X4 and X5 is N; ring B is a phenyl ring or a 5-6 membered heteroaromatic ring wherein the above phenyl and heteroaromatic rings are optionally substituted by one or more G1Substituted;
R1independently selected from H, cyano, halogen, C1-6Alkyl radical, C3-6Cycloalkyl, 3-6 membered heterocycloalkyl, -OR2、-NR2R3、-C(O)NR2R3Wherein said alkyl, cycloalkyl OR heterocycloalkyl is optionally substituted by cyano, halogen, -OR4、-NR4R5、C1-6Alkyl radical, C3-6Cycloalkyl or 3-6 membered heterocycloalkyl;
a is independently selected from-C0-4Alkyl-, -CR6R7-、-C1-2Alkyl (R)6)(OH)-、-C(O)-、-CR6R7O-、-OCR6R7-、-SCR6R7-、-CR6R7S-、-NR6-、-NR6C(O)-、-C(O)NR6-、-NR6C(O)NR7-、-CF2-、-O-、-S-、-S(O)m-、-NR6S(O)2-、-S(O)2NR6-;
Y is absent or C is selected3-8Cycloalkyl, 3-8 membered heterocycloalkyl, 5-12 membered fused alkyl, 5-12 membered fused heterocyclyl, 5-12 membered spiro cyclic group, 5-12 membered spiro heterocyclic group, aromatic group or heteroaromatic group, wherein said cycloalkyl, heterocycloalkyl, spiro cyclic group, fused heterocyclic group, spiro heterocyclic group, aromatic group or heteroaromatic group is optionally substituted with one or more G2Substituted;
z is independently selected from cyano, -NR8CN、
Figure FDA0002909122710000012
Bond a is a double or triple bond;
when a is a double bond, Ra、RbAnd RcEach independently selected from H, cyano, halogen, C1-6Alkyl radical, C3-6Cycloalkyl or 3-6 membered heterocyclyl. Wherein said alkyl, cycloalkyl and heterocyclyl are optionally substituted by 1 or more G3Substituted;
Raand RbOr RbAnd RcOptionally together with the carbon atom to which they are attachedTogether forming a 3-6 membered ring optionally containing heteroatoms;
when bond a is a triple bond, RaAnd RcIs absent, RbIndependently selected from H, cyano, halogen, C1-6Alkyl radical, C3-6Cycloalkyl or 3-6 membered heterocyclyl by one or more G4Substituted;
R8independently selected from H, C1-6Alkyl radical, C3-6Cycloalkyl or 3-6 membered heterocyclyl, wherein said alkyl, cycloalkyl and heterocyclyl are optionally substituted by 1 or more G5Substituted;
G1、G2、G3、G4and G5Each independently selected from cyano, halogen, C1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-8Cycloalkyl or 3-8 membered heterocyclyl, C6-10Aryl, 5-10 membered heteroaryl, -OR9、-OC(O)NR9R10、-C(O)OR9、-C(O)NR9R10、-C(O)R9、-NR9R10、-NR9C(O)R10、-NR9C(O)NR10R11、-S(O)mR9or-NR9S(O)mR10Wherein the alkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, heteroaryl are optionally substituted by 1 or more cyano, halogen, C1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, C3-8Cycloalkyl or 3-8 membered heterocyclyl, C6-10Aryl, 5-10 membered heteroaryl, -OR12、-OC(O)NR12R13、-C(O)OR12、-C(O)NR12R13、-C(O)R12、-NR12R13、-NR12C(O)R13、-NR12C(O)NR13R14、-S(O)mR12or-NR12S(O)mR13Substituted with the substituent(s);
R3、R4、R5、R6、R7、R8、R9、R11、R12、R13and R14Each is independentSelected from cyano, halogen, C1-6Alkyl radical, C3-8Cycloalkyl or 3-8 membered monocyclic heterocyclyl, monocyclic heteroaryl or phenyl; and is
m is 1 or 2.
2. A compound according to claim 1 or a prodrug, stable isotope derivative, pharmaceutically acceptable salt, solvate, polymorph or isomer thereof, and a mixture form thereof.
3. It is selected from the following compounds:
Figure FDA0002909122710000021
Figure FDA0002909122710000031
Figure FDA0002909122710000041
Figure FDA0002909122710000051
or a prodrug, stable isotope derivative, pharmaceutically acceptable salt, solvate, isomer, and mixtures and forms thereof.
4. A pharmaceutical composition comprising a compound of claims 1-3 or a prodrug, stable isotope derivative, pharmaceutically acceptable salt, solvate, polymorph or isomer thereof, and a pharmaceutically acceptable carrier.
5. Use of a compound according to any one of the claims or a prodrug, stable isotope derivative, pharmaceutically acceptable salt, solvate, polymorph or isomer thereof in the manufacture of a medicament for the treatment of an FGFR-mediated disease.
6. The use of claim 5, wherein the FGFR-mediated disease is one or more of non-small cell lung cancer, esophageal cancer, melanoma, gastric cancer, multiple myeloma, liver cancer, cholangiocarcinoma, prostate cancer, skin cancer, ovarian cancer, endometrial cancer, cervical cancer, bladder cancer, breast cancer, colon cancer, glioma, and rhabdomyosarcoma.
7. A compound according to any one of claims 5 or a prodrug, stable isotope derivative, pharmaceutically acceptable salt, solvate, polymorph or isomer thereof, and a pharmaceutically acceptable carrier for use as a medicament.
CN202110080572.5A 2021-01-21 2021-01-21 Alkyne heterocyclic compound for treating cancer and preparation method and application thereof Pending CN112851587A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110080572.5A CN112851587A (en) 2021-01-21 2021-01-21 Alkyne heterocyclic compound for treating cancer and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110080572.5A CN112851587A (en) 2021-01-21 2021-01-21 Alkyne heterocyclic compound for treating cancer and preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN112851587A true CN112851587A (en) 2021-05-28

Family

ID=76008567

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110080572.5A Pending CN112851587A (en) 2021-01-21 2021-01-21 Alkyne heterocyclic compound for treating cancer and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN112851587A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114057749A (en) * 2021-11-19 2022-02-18 药雅科技(上海)有限公司 Preparation method and application of irreversible alkyne heterocyclic compound FGFR inhibitor

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014011900A2 (en) * 2012-07-11 2014-01-16 Blueprint Medicines Inhibitors of the fibroblast growth factor receptor
CN104144915A (en) * 2012-02-28 2014-11-12 安斯泰来制药有限公司 Nitrogen-containing aromatic heterocyclic compound

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104144915A (en) * 2012-02-28 2014-11-12 安斯泰来制药有限公司 Nitrogen-containing aromatic heterocyclic compound
WO2014011900A2 (en) * 2012-07-11 2014-01-16 Blueprint Medicines Inhibitors of the fibroblast growth factor receptor
CN104540809A (en) * 2012-07-11 2015-04-22 蓝印药品公司 Inhibitors of the fibroblast growth factor receptor

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114057749A (en) * 2021-11-19 2022-02-18 药雅科技(上海)有限公司 Preparation method and application of irreversible alkyne heterocyclic compound FGFR inhibitor
CN114057749B (en) * 2021-11-19 2023-12-22 药雅科技(上海)有限公司 Preparation method and application of irreversible alkyne heterocyclic compound FGFR inhibitor

Similar Documents

Publication Publication Date Title
CN115073469B (en) Preparation and application of pyrrolopyrimidine compound as kinase inhibitor
JP2019522055A (en) Heterocyclic compounds used as FGFR inhibitors
JP2019520367A (en) Novel Heterocyclic Derivative Compound and Use Thereof
CN115073450A (en) KRAS G12C Preparation and application of mutant protein inhibitor
CN114436976A (en) Novel quinazoline derivative and preparation and application thereof
CN112851587A (en) Alkyne heterocyclic compound for treating cancer and preparation method and application thereof
CN115028633B (en) Preparation and application of pyrrolopyrimidine compound
CN114591334B (en) Dihydropyrazolopyrimidinone derivatives
CN114853723B (en) Preparation and application of indole compound BTK inhibitor
CN115181106B (en) Quinazoline KRAS G12D Preparation and application of mutant protein inhibitor
CN112939982A (en) Alkyne heterocyclic BTK inhibitor and preparation method and application thereof
CN114853739B (en) Acetylenic pyrazine FGFR inhibitor and preparation method and application thereof
CN114057749B (en) Preparation method and application of irreversible alkyne heterocyclic compound FGFR inhibitor
CN114805359B (en) Preparation method and application of acetylenic heterocyclic compound FGFR inhibitor
CN114853740B (en) Preparation method and application of acetylenic pyrimidine compound as FGFR inhibitor
CN115028634B (en) Acetylenic pyrazino heterocycle FGFR inhibitor and preparation method and application thereof
CN114853752B (en) Preparation and application of BTK inhibitor pyrido heterocyclic compound
CN115043832B (en) FGFR inhibitor acetylenic heterocyclic compound and preparation method and application thereof
CN114957241B (en) Preparation and Application of Heterocyclic Compounds as Kinase Inhibitors
CN115433190A (en) Preparation method and application of irreversible heterocyclic compound FGFR inhibitor
CN115043841B (en) Preparation and application of heterocyclic compound serving as BTK inhibitor
CN114957242B (en) Preparation and application of pyrido heterocyclic compounds as kinase inhibitors
CN115215868A (en) Preparation method and application of heterocyclic compound FGFR inhibitor
CN115141176B (en) Acetylenic indole FGFR inhibitor and preparation method and application thereof
CN115368381B (en) Preparation and application of heterocyclic 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
RJ01 Rejection of invention patent application after publication

Application publication date: 20210528

RJ01 Rejection of invention patent application after publication