CN112300194B - Condensed ring pyridone compounds, preparation method and application - Google Patents

Condensed ring pyridone compounds, preparation method and application Download PDF

Info

Publication number
CN112300194B
CN112300194B CN202010747468.2A CN202010747468A CN112300194B CN 112300194 B CN112300194 B CN 112300194B CN 202010747468 A CN202010747468 A CN 202010747468A CN 112300194 B CN112300194 B CN 112300194B
Authority
CN
China
Prior art keywords
independently selected
group
substituted
cancer
compound
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.)
Active
Application number
CN202010747468.2A
Other languages
Chinese (zh)
Other versions
CN112300194A (en
Inventor
万惠新
查传涛
马金贵
潘建峰
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shanghai Lingda Biomedical Co Ltd
Original Assignee
Shanghai Lingda Biomedical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shanghai Lingda Biomedical Co Ltd filed Critical Shanghai Lingda Biomedical Co Ltd
Publication of CN112300194A publication Critical patent/CN112300194A/en
Application granted granted Critical
Publication of CN112300194B publication Critical patent/CN112300194B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/14Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • 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
    • C07D471/22Heterocyclic 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 systems contains four or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains three hetero rings
    • C07D487/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/22Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains four or more hetero rings

Abstract

The invention discloses a condensed ring pyridone compound, a preparation method and application thereof. In particular to a fused ring pyridone compound shown as a general formula I, or pharmaceutically acceptable salt thereof, or enantiomer, diastereoisomer, tautomer, solvate, polymorph or prodrug thereof, a preparation method and pharmaceutical application thereof, wherein the definition of each group is described in the specification.

Description

Condensed ring pyridone compounds, preparation method and application
Technical Field
The invention belongs to the field of pharmaceutical chemistry, and particularly relates to a fused ring pyridone compound, a compound with Ras mutein inhibition activity, a preparation method and application.
Background
RAS is the first oncogene identified in human tumors and was first found in two murine sarcoma viruses. There are three members of the RAS gene family, Hras, Kras, Nras. In human tumors, Kras mutations are most common, accounting for approximately 85%. Previous studies have shown that Kras mutations are carcinogenic because codon 12 is missense mutated, altering the structure of the Kras protein and keeping it activated at all times. Ras plays a major role in signal pathway transmission, mainly activating kinases controlling gene transcription, thereby regulating cell differentiation and proliferation, and is closely related to survival, proliferation, migration, metastasis and angiogenesis of tumor cells. According to statistics, Kras G12C mutation exists in 11% -16% of lung adenocarcinoma cases, and part of pancreatic cancer, colorectal cancer, ovarian cancer and bile duct cancer is caused by Kras mutation. However, over thirty years ago since the first discovery of Kras oncogene, the targeting drugs for EGFR, BCL and other common protooncogenes have been developed for several generations, and the targeting drugs for Kras have not been successfully developed. Historically, targeted drugs against KRas pathway mutant tumors have focused primarily on farnesyl transferase inhibitors and Raf-MEK pathway inhibitors, but with little success. In recent years, inhibitors aiming at KRas specific gene mutation are developed into hot spots, and part of inhibitors gradually go from preclinical hatching to clinical research, such as KRas G12C inhibitors AMG510, MRTX1257 and the like, and show certain curative effect in early clinical experiments. The first clinical data of the first global KRASG12C inhibitor AMG510 was finally promulgated by the american clinical oncology institute held in 6 months 2019, in which clinical studies the installed drug AMG510 was shown to prevent tumor growth in most non-small cell lung and colorectal cancer patients with KRas mutations. Therefore, finding and searching for a target drug against KRas specific mutant gene with high specificity and excellent drug availability is a major hotspot in the industry.
Disclosure of Invention
One of the technical problems to be solved by the invention is to provide a novel KRas G12C inhibitor for preparing a tumor treatment medicament.
The scheme for solving the technical problems is as follows:
a fused ring pyridone compound shown as a general formula I, or pharmaceutically acceptable salt thereof, or enantiomer, diastereoisomer, tautomer, solvate, polymorph or prodrug thereof,
Figure BDA0002608844410000021
in the formula:
r1 is independently selected from hydrogen, deuterium, halogen, cyano, nitro, C1-C6Alkyl radical, C1-C6alkyl-SO2-、C1-C6alkyl-SO-, or C1-C6A haloalkyl group;
r2 and R3 are independently selected from hydrogen, halogen, cyano, nitro and C1-C6Alkyl radical, C1-C6alkyl-SO2-、C1-C6alkyl-SO-, N (R)2a)(R2b)-(CH2) x-; or, R9And R10Together forming a 5-to 10-membered quilt C1-C6An alkyl-substituted nitrogen-containing heterocycloalkyl group; wherein R is2aAnd R2bEach independently selected from hydrogen or C1-C6Alkyl, x is selected from any integer of 0-5;
w, W1, W2 are independently selected from CR4 or N, R4 is independently selected from H, deuterium, halogen, cyano, hydroxy, amino, C1-C6Alkyl radical, C1-C6Alkoxy, haloalkyl, haloalkoxy, alkenyl, alkynyl, 3-to 8-membered cycloalkyl or heterocycloalkyl, and the like;
ra, Rb, Rc, Rd, Re, Rf and Rg are respectively and independently selected from hydrogen, deuterium, C1-C6 alkyl, alkoxy, haloalkyl and the like, or Ra, Rb, Rc, Rd, Re, Rf and Rg form a 3-8-membered saturated or partially unsaturated ring system between every two;
ar is independently selected from a 5-12 membered aromatic ring or aromatic condensed ring, a 5-12 membered aromatic heterocycle or aromatic condensed heterocycle; and the Ar ring may be substituted with one or more of the following groups: hydrogen, deuterium, halogen, C1-C6 alkyl, alkoxy, 3-8 membered cycloalkyl or heterocycloalkyl, C1-C6 haloalkoxy, C2-C6 alkenyl, C2-C6 alkynyl, substituted or unsubstituted amino, amide, sulfonamide, etc.;
z is independently selected from O or NR5, R5 is independently selected from H, deuterium, C1-C6Alkyl radical, C1-C6Alkoxy, haloalkyl, haloalkoxy, alkenyl, alkynyl, 3-to 8-membered cycloalkyl or heterocycloalkyl, and the like;
ri and Rj are independently selected from H, deuterium, halogen, C1-C6Alkyl radical, C1-C6Alkoxy, haloalkyl, haloalkoxy, alkenyl, alkynyl, 3-8 membered cycloalkyl or heterocycloalkyl, etc., or when Z is NR5, Ri and Rj may be oxo; or Ri and Rj may both form a 3-8 membered saturated or partially unsaturated or unsaturated ring system with each other; or Ri and Rj may form a 3-8 membered saturated or partially unsaturated or unsaturated ring system with Z, respectively;
r is independently selected from hydrogen, deuterium, C1-C20 alkyl, C2-12 alkenyl, C2-C12 alkynyl, 3-12 membered cycloalkyl or heterocycloalkyl, 5-10 membered aryl or heteroaryl, etc.;
wherein said heteroaryl group contains 1 to 3 heteroatoms selected from the group consisting of: n, O, P or S, the heterocycloalkyl group containing 1 to 3 heteroatoms selected from the group consisting of: n, O, P or S, said ring system including spiro, bridged, fused, etc. saturated or partially unsaturated ring systems.
In some embodiments, the compound having the general formula (I), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, solvate, polymorph, or prodrug thereof, preferably a compound represented by the general formulae (IIA), (IIB), (IIC), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, solvate, polymorph, or prodrug thereof:
Figure BDA0002608844410000031
wherein R6 is independently selected from hydrogen, deuterium, C1-C6 alkyl, alkoxy, haloalkyl, haloalkoxy, and the like; r, R1, R2, R3, Ra, Rb, Rc, Rd, Re, Rf, Rg, W1, W2, Ri, Rj are as defined above.
In other embodiments, a compound having the general formula (1), or a pharmaceutically acceptable salt thereof, or an enantiomer, diastereomer, tautomer, solvate, polymorph, or prodrug thereof, characterized by:
r1 is preferably selected from hydrogen, deuterium, fluoro, methyl, cyano, etc.;
ra, Rb, Rc, Rd, Re, Rf, Rg are each independently preferably selected from hydrogen, deuterium, fluorine, methyl;
w1 and W are each independently preferably selected from CR4, R4 is independently selected from hydrogen, deuterium, halogen, cyano, hydroxy, amino, C1-C4 alkyl, C1-C4 alkoxy, C2-C4 alkenyl, C2-C4 alkynyl, C1-C4 alkoxy, 3-6 membered cycloalkyl, C1-C4 haloalkyl, C1-C4 haloalkoxy, and the like;
w2 is independently preferably selected from N or CH, C-F, C-Cl, C-Me, C-OMe, etc.;
ar is independently preferably a monocyclic aromatic group such as a substituted or unsubstituted phenyl group or pyridyl group, or a substituted or unsubstituted bicyclic aromatic group such as a naphthyl group, a naphthyridinyl group, an indazolyl group, a benzimidazolyl group, or a benzothiazolyl group; said one or more substituents are preferably selected from the group consisting of: hydrogen, deuterium, halogen, alkyl of C1-C4, hydroxy, amino, cyano, C1-C4 alkoxy, C1-C4 haloalkyl, C1-C4 haloalkoxy, and the like;
r is independently preferably selected from substituted or unsubstituted C6-C20 alkyl, substituted or unsubstituted 3-8 membered cycloalkyl or heterocycloalkyl, substituted or unsubstituted 5-10 membered aryl or heteroaryl; further preferably a C6-C20 branched alkyl group, a 5-to 8-membered cycloalkyl or heterocycloalkyl group, a phenyl group, a pyridyl group or the like;
the ranges for the other groups are as defined above.
A process for preparing a compound of formula I, said process comprising steps a-c:
a) converting a compound of formula (a) to an intermediate (B) by a transition metal catalysed coupling reaction with an arylboronic acid or arylboronic ester or arylmetal reagent (Ar-M); and
b) removing the protective group PG from the compound of the general formula (B) through a conventional functional group to obtain a compound of a general formula (C);
c) the compound of the general formula (C) and acrylic acid or acryloyl chloride are subjected to condensation reaction under proper conditions to generate the general formula (I).
Figure BDA0002608844410000041
The definition of each group is as described above;
preferably, said steps a), b), c) are each carried out in a solvent, and said solvent is selected from the group consisting of: water, methanol, ethanol, isopropanol, butanol, ethylene glycol methyl ether, N-methyl pyrrolidone, dimethyl sulfoxide, tetrahydrofuran, toluene, dichloromethane, 1, 2-dichloroethane, acetonitrile, N-dimethylformamide, N-dimethylacetamide, dioxane, or a combination thereof.
Preferably, the transition metal catalyst is selected from the group consisting of: tris (dibenzylideneacetone) dipalladium (Pd)2(dba)3) Tetrakis (triphenylphosphine) palladium (Pd (PPh)3)4) Palladium acetate, palladium chloride, dichlorobis (triphenylphosphine) palladium, palladium trifluoroacetate, triphenylphosphine palladium acetate, [1,1' -bis (diphenylphosphino) ferrocene]Palladium dichloride, bis (tri-o-phenylphosphino) palladium dichloride, 1, 2-bis (diphenylphosphino) ethane palladium dichloride, or a combination thereof; the catalyst ligand is selected from the group consisting of: tri-tert-butylphosphine, tri-tert-butylphosphine tetrafluoroborate, tri-n-butylphosphine, triphenylphosphine, tri-p-benzylphosphine, tricyclohexylphosphine, tri-o-phenylphosphine, or a combination thereof.
Preferably, the condensing agent is selected from the group consisting of: DCC, DIC, CDI, EDCI, HOAt, HOBt, BOP, PyBOP, HATU, TBTU, and the like, or combinations thereof.
Preferably, the inorganic base is selected from the group consisting of: sodium hydride, potassium hydroxide, sodium acetate, potassium tert-butoxide, sodium tert-butoxide, potassium fluoride, cesium fluoride, potassium phosphate, potassium carbonate, potassium bicarbonate, sodium carbonate, sodium bicarbonate, or combinations thereof; the organic base is selected from the group consisting of: pyridine, triethylamine, N, N-diisopropylethylamine, 1, 8-diazabicyclo [5.4.0] undec-7-ene (DBU), lithium hexamethyldisilazide, sodium hexamethyldisilazide, lutidine, or a combination thereof.
Preferably, the acid is selected from the group consisting of: hydrochloric acid, sulfuric acid, phosphoric acid, methanesulfonic acid, toluenesulfonic acid, trifluoroacetic acid, formic acid, acetic acid, trifluoromethanesulfonic acid, or combinations thereof.
Preferably, the reducing agent is selected from the group consisting of: iron powder, zinc powder, stannous chloride, sodium thiosulfate, sodium sulfite, hydrogen and the like.
The invention provides a class of preferred compounds of formula (I) including, but not limited to, the following structures:
Figure BDA0002608844410000051
Figure BDA0002608844410000061
the invention also aims to provide a medicament for treating or preventing tumors and a composition thereof. The technical scheme for realizing the purpose is as follows:
a pharmaceutical composition for treating tumor comprises the fused ring pyridone compound shown in the general formula (I), or pharmaceutically acceptable salt thereof, or enantiomer, diastereomer, tautomer, solvate, polymorph or prodrug thereof and pharmaceutically acceptable carrier.
Another object of the present invention is to provide a use of the above compound. The technical scheme for realizing the purpose is as follows:
the condensed ring pyridone compound shown in the general formula (I) or pharmaceutically acceptable salt thereof, or enantiomer, diastereoisomer, tautomer, solvate, polymorph or prodrug thereof are used for preparing medicaments for treating diseases related to the activity or expression amount of Ras mutein, particularly medicaments for treating tumors. The tumor is independently selected from non-small cell lung cancer, lung adenocarcinoma, lung squamous carcinoma, breast cancer, prostatic cancer, liver cancer, skin cancer, gastric cancer, intestinal cancer, cholangiocarcinoma, brain cancer, leukemia, lymph cancer, fibroma, sarcoma, basal cell carcinoma, glioma, renal cancer, melanoma, bone cancer, thyroid cancer, nasopharyngeal cancer, pancreatic cancer, etc.
The invention relates to a compound with the structural characteristics of a general formula (I), which can inhibit various tumor cells, particularly can efficiently kill tumors related to abnormal KRas G12C mutant protein signal pathways, and is a treatment medicament with a brand-new action mechanism.
It is to be understood that within the scope of the present invention, the above-described features of the present invention and those specifically described below (e.g., in the examples) may be combined with each other to form new or preferred embodiments. The space is not described herein in a repeated fashion.
Detailed Description
The inventor has made a long-term and intensive study to prepare a compound with a novel structure shown in formula I, and finds that the compound has a better inhibitory activity for inhibiting the KRas G12C protein, the compound has a specific inhibitory effect on the KRas G12C protein at a very low concentration (which can be as low as less than 100nM), the inhibitory activity on the KRas G12C-related cell proliferation is quite excellent, and the compound has a stronger killing effect on KRas G12C-positive tumor cells at a very low concentration (which can be as low as less than 10nM), so that the compound can be used for treating related diseases such as tumors caused by KRas G12C mutation or abnormal expression. Based on the above findings, the inventors have completed the present invention.
Term(s) for
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the claimed subject matter belongs. All patents, patent applications, and publications cited herein are incorporated by reference in their entirety unless otherwise indicated.
It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the subject matter claimed. In this application, the use of the singular also includes the plural unless specifically stated otherwise. It must be noted that, as used in this specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. It should also be noted that the use of "or", "or" means "and/or" unless stated otherwise. Furthermore, the term "comprising" as well as other forms, such as "includes," "including," and "containing," are not limiting.
Definitions for the terms of the standardization sector can be found in the literature references including Carey and Sundberg "ADVANCED ORGANIC CHEMISTRY 4TH ED." Vols.A (2000) and B (2001), Plenum Press, New York. Unless otherwise indicated, conventional methods within the skill of the art are employed, such as mass spectrometry, NMR, IR and UV/VIS spectroscopy, and pharmacological methods. Unless a specific definition is set forth, the terms used herein in the pertinent description of analytical chemistry, organic synthetic chemistry, and pharmaceutical chemistry are known in the art. Standard techniques can be used in chemical synthesis, chemical analysis, pharmaceutical preparation, formulation and delivery, and treatment of patients. For example, the reaction and purification can be carried out using the instructions of the kit from the manufacturer, or according to the methods known in the art or the instructions of the present invention. The techniques and methods described above can generally be practiced according to conventional methods well known in the art, as described in various general and more specific documents referred to and discussed in this specification. In the present specification, groups and substituents thereof may be selected by one skilled in the art to provide stable moieties and compounds.
When a substituent is described by a general formula written from left to right, the substituent also includes chemically equivalent substituents obtained when the formula is written from right to left. For example, -CH 2O-is equivalent to-OCH 2-.
The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. All documents, or portions of documents, cited in this application, including but not limited to patents, patent applications, articles, books, operating manuals, and treatises, are hereby incorporated by reference in their entirety.
Certain chemical groups defined herein are preceded by a shorthand notation to indicate the total number of carbon atoms present in the group. For example, C1-6 alkyl refers to an alkyl group as defined below having a total of 1 to 6 carbon atoms. The total number of carbon atoms in the shorthand notation excludes carbons that may be present in a substituent of the group.
In addition to the foregoing, the following terms, when used in the specification and claims of this application, have the meanings indicated below, unless otherwise specifically indicated.
In the present application, the term "halogen" means fluorine, chlorine, bromine or iodine; "hydroxy" means an-OH group; "hydroxyalkyl" refers to an alkyl group as defined below substituted with a hydroxyl (-OH) group; "carbonyl" refers to a-C (═ O) -group; "nitro" means-NO2(ii) a "cyano" means-CN; "amino" means-NH2(ii) a "substituted amino" refers to an amino group substituted with one or two alkyl, alkylcarbonyl, aralkyl, heteroaralkyl groups as defined below, e.g., monoalkylamino, dialkylamino, alkylamido, aralkylamino, heteroaralkylamino; "carboxyl" means-COOH.
In the present application, the term "alkyl", as a group or as part of another group (e.g. as used in groups such as halogen-substituted alkyl), means a straight or branched hydrocarbon chain group consisting only of carbon and hydrogen atoms, containing no unsaturated bonds, having, for example, from 1 to 12 (preferably from 1 to 8, more preferably from 1 to 6) carbon atoms and being attached to the rest of the molecule by single bonds. Examples of alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 2-methylbutyl, 2-dimethylpropyl, n-hexyl, heptyl, 2-methylhexyl, 3-methylhexyl, octyl, nonyl, decyl, and the like.
In the present application, the term "alkenyl" as a group or part of another group means a straight or branched hydrocarbon chain group consisting of only carbon atoms and hydrogen atoms, containing at least one double bond, having, for example, 2 to 14 (preferably 2 to 10, more preferably 2 to 6) carbon atoms, and being connected to the rest of the molecule by a single bond, such as, but not limited to, vinyl, propenyl, allyl, but-1-enyl, but-2-enyl, pent-1, 4-dienyl, and the like.
In the present application, the term "alkynyl" as a group or part of another group means a straight or branched hydrocarbon chain group consisting solely of carbon and hydrogen atoms, containing at least one triple bond and optionally one or more double bonds, having for example 2 to 14 (preferably 2 to 10, more preferably 2 to 6) carbon atoms and being connected to the rest of the molecule by single bonds, such as but not limited to ethynyl, prop-1-ynyl, but-1-ynyl, pent-1-en-4-ynyl and the like.
In the present application, the term "cycloalkyl" as a group or part of another group means a stable non-aromatic monocyclic or polycyclic hydrocarbon group consisting of only carbon atoms and hydrogen atoms, which may include fused, bridged or spiro ring systems, having 3 to 15 carbon atoms, preferably having 3 to 10 carbon atoms, more preferably having 3 to 8 carbon atoms, and which is saturated or unsaturated and may be attached to the rest of the molecule by a single bond via any suitable carbon atom. Unless otherwise specifically indicated in the specification, carbon atoms in cycloalkyl groups may be optionally oxidized. Examples of cycloalkyl include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl, cyclooctyl, 1H-indenyl, 2, 3-indanyl, 1,2,3, 4-tetrahydro-naphthyl, 5,6,7, 8-tetrahydro-naphthyl, 8, 9-dihydro-7H-benzocyclohepten-6-yl, 6,7,8, 9-tetrahydro-5H-benzocycloheptenyl, 5,6,7,8,9, 10-hexahydro-benzocyclooctenyl, fluorenyl, bicyclo [2.2.1] heptyl, 7-dimethyl-bicyclo [2.2.1] heptyl, bicyclo [2.2.1] heptenyl, bicyclo [2.2.2] octyl, bicyclo [3.1.1] heptyl, bicyclo [3.2.1] octyl, bicyclo [2.2.2] octenyl, Bicyclo [3.2.1] octenyl, adamantyl, octahydro-4, 7-methylene-1H-indenyl, octahydro-2, 5-methylene-pentalenyl and the like.
In this application, the term "heterocyclyl" as a group or part of another group means a stable 3-to 20-membered non-aromatic cyclic group consisting of 2 to 14 carbon atoms and 1 to 6 heteroatoms selected from nitrogen, phosphorus, oxygen, and sulfur. Unless otherwise specifically indicated in the specification, a heterocyclic group may be a monocyclic, bicyclic, tricyclic or higher ring system, which may include fused ring systems, bridged ring systems or spiro ring systems; wherein the nitrogen, carbon or sulfur atom in the heterocyclic group may be optionally oxidized; the nitrogen atoms may optionally be quaternized; and the heterocyclic group may be partially or fully saturated. The heterocyclic group may be attached to the rest of the molecule via a carbon atom or a heteroatom and by a single bond. In heterocyclic groups containing fused rings, one or more of the rings may be aryl or heteroaryl as defined below, provided that the point of attachment to the rest of the molecule is a non-aromatic ring atom. For the purposes of the present invention, heterocyclyl is preferably a stable 4-to 11-membered non-aromatic monocyclic, bicyclic, bridged or spiro group containing 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur, more preferably a stable 4-to 8-membered non-aromatic monocyclic, bicyclic, bridged or spiro group containing 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur. Examples of heterocyclyl groups include, but are not limited to: pyrrolidinyl, morpholinyl, piperazinyl, homopiperazinyl, piperidinyl, thiomorpholinyl, 2, 7-diaza-spiro [3.5] nonan-7-yl, 2-oxa-6-aza-spiro [3.3] heptan-6-yl, 2, 5-diaza-bicyclo [2.2.1] heptan-2-yl, azetidinyl, pyranyl, tetrahydropyranyl, thiopyranyl, tetrahydrofuranyl, oxazinyl, dioxolanyl, tetrahydroisoquinolinyl, decahydroisoquinolinyl, imidazolinyl, imidazolidinyl, quinolizinyl, thiazolidinyl, isothiazolidinyl, isoxazolidinyl, indolinyl, octahydroindolyl, octahydroisoindolyl, pyrrolidinyl, pyrazolidinyl, phthalimidyl, and the like.
In this application, the term "aryl" as a group or as part of another group means a conjugated hydrocarbon ring system group having 6 to 18 carbon atoms, preferably having 6 to 10 carbon atoms. For the purposes of the present invention, an aryl group may be a monocyclic, bicyclic, tricyclic or higher polycyclic ring system and may also be fused to a cycloalkyl or heterocyclic group as defined above, provided that the aryl group is attached to the remainder of the molecule by a single bond via an atom on the aromatic ring. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, anthracenyl, phenanthrenyl, fluorenyl, 2, 3-dihydro-1H-isoindolyl, 2-benzoxazolinone, 2H-1, 4-benzoxazin-3 (4H) -one-7-yl, and the like.
In the present application, the term "arylalkyl" refers to an alkyl group as defined above substituted with an aryl group as defined above.
In this application, the term "heteroaryl" as a group or part of another group means a 5-to 16-membered conjugated ring system group having 1 to 15 carbon atoms (preferably having 1 to 10 carbon atoms) and 1 to 6 heteroatoms selected from nitrogen, oxygen and sulfur in the ring. Unless otherwise specifically indicated in the specification, a heteroaryl group may be a monocyclic, bicyclic, tricyclic or higher ring system, and may also be fused to a cycloalkyl or heterocyclic group as defined above, provided that the heteroaryl group is attached to the rest of the molecule by a single bond via an atom on the aromatic ring. The nitrogen, carbon or sulfur atoms in the heteroaryl group may be optionally oxidized; the nitrogen atoms may optionally be quaternized. For the purposes of the present invention, heteroaryl is preferably a stable 5-to 12-membered aromatic group containing 1 to 5 heteroatoms selected from nitrogen, oxygen and sulfur, more preferably a stable 5-to 10-membered aromatic group containing 1 to 4 heteroatoms selected from nitrogen, oxygen and sulfur or a 5-to 6-membered aromatic group containing 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur. Examples of heteroaryl groups include, but are not limited to, thienyl, imidazolyl, pyrazolyl, thiazolyl, oxazolyl, oxadiazolyl, isoxazolyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl, benzimidazolyl, benzopyrazolyl, indolyl, furyl, pyrrolyl, triazolyl, tetrazolyl, triazinyl, indolizinyl, isoindolyl, indazolyl, isoindolyl, purinyl, quinolyl, isoquinolyl, diazonaphthyl, naphthyridinyl, quinoxalinyl, pteridinyl, carbazolyl, carbolinyl, phenanthridinyl, phenanthrolinyl, acridinyl, phenazinyl, isothiazolyl, benzothiazolyl, benzothienyl, oxazolyl, cinnolinyl, quinazolinyl, thiophenyl, indolizinyl, orthophenanthrolidinyl, isoxazolyl, phenoxazinyl, phenothiazinyl, 4,5,6, 7-tetrahydrobenzo [ b ] thienyl, naphthopyridyl, pyridinyl, and the like, [1,2,4] triazolo [4,3-b ] pyridazine, [1,2,4] triazolo [4,3-a ] pyrazine, [1,2,4] triazolo [4,3-c ] pyrimidine, [1,2,4] triazolo [4,3-a ] pyridine, imidazo [1,2-b ] pyridazine, imidazo [1,2-a ] pyrazine and the like.
In the present application, the term "heteroarylalkyl" refers to an alkyl group as defined above substituted with a heteroaryl group as defined above.
In this application, "optional" or "optionally" means that the subsequently described event or circumstance may or may not occur, and that the description includes instances where the event or circumstance occurs and instances where it does not. For example, "optionally substituted aryl" means that the aryl group is substituted or unsubstituted, and the description includes both substituted and unsubstituted aryl groups.
The terms "moiety," "structural moiety," "chemical moiety," "group," "chemical group" as used herein refer to a specific fragment or functional group in a molecule. Chemical moieties are generally considered to be chemical entities that are embedded in or attached to a molecule.
"stereoisomers" refers to compounds that consist of the same atoms, are bonded by the same bonds, but have different three-dimensional structures. The present invention is intended to cover various stereoisomers and mixtures thereof.
When the compounds of the present invention contain olefinic double bonds, the compounds of the present invention are intended to include both E-and Z-geometric isomers unless otherwise specified.
"tautomer" refers to an isomer formed by the transfer of a proton from one atom of a molecule to another atom of the same molecule. All tautomeric forms of the compounds of the invention are also intended to be included within the scope of the invention.
The compounds of the present invention or pharmaceutically acceptable salts thereof may contain one or more chiral carbon atoms and may therefore give rise to enantiomers, diastereomers and other stereoisomeric forms. Each chiral carbon atom may be defined as (R) -or (S) -, based on stereochemistry. The present invention is intended to include all possible isomers, as well as racemates and optically pure forms thereof. The compounds of the invention may be prepared by selecting as starting materials or intermediates racemates, diastereomers or enantiomers. Optically active isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, e.g., crystallization and chiral chromatography.
Conventional techniques for preparing/separating individual isomers include Chiral synthesis from suitable optically pure precursors, or resolution of racemates (or racemates of salts or derivatives) using, for example, Chiral high performance liquid chromatography, as described, for example, in Gerald Gubitz and Martin G.Schmid (Eds.), Chiral separation, Methods and Protocols, Methods in Molecular Biology, Vol.243, 2004; m. Stalcup, Chiral Separations, Annu. Rev. anal. chem.3:341-63, 2010; fumiss et al (eds.), VOGEL' S ENCYCOPEDIA OF PRACTICAL ORGANIC CHEMISTRY 5. TH ED., Longman Scientific and Technical Ltd., Essex,1991, 809-816; heller, acc, chem, res, 1990,23,128.
In the present application, the term "pharmaceutically acceptable salts" includes pharmaceutically acceptable acid addition salts and pharmaceutically acceptable base addition salts.
"pharmaceutically acceptable acid addition salts" refers to salts with inorganic or organic acids which retain the biological effectiveness of the free base without other side effects. Inorganic acid salts include, but are not limited to, hydrochloride, hydrobromide, sulfate, nitrate, phosphate, and the like; organic acid salts include, but are not limited to, formates, acetates, 2-dichloroacetates, trifluoroacetates, propionates, caproates, caprylates, caprates, undecylenates, glycolates, gluconates, lactates, sebacates, adipates, glutarates, malonates, oxalates, maleates, succinates, fumarates, tartrates, citrates, palmitates, stearates, oleates, cinnamates, laurates, malates, glutamates, pyroglutamates, aspartates, benzoates, methanesulfonates, benzenesulfonates, p-toluenesulfonates, alginates, ascorbates, salicylates, 4-aminosalicylates, napadisylates, and the like. These salts can be prepared by methods known in the art.
"pharmaceutically acceptable base addition salts" refers to salts with inorganic or organic bases which maintain the biological effectiveness of the free acid without other side effects. Salts derived from inorganic bases include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like. Preferred inorganic salts are ammonium, sodium, potassium, calcium and magnesium salts. Salts derived from organic bases include, but are not limited to, the following: primary, secondary and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, diethanolamine, triethanolamine, dimethylethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purine, piperazine, piperidine, N-ethylpiperidine, polyamine resins, and the like. Preferred organic bases include isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine. These salts can be prepared by methods known in the art.
"polymorph" refers to different solid crystalline phases of certain compounds of the present invention in the solid state due to the presence of two or more different molecular arrangements. Certain compounds of the present invention may exist in more than one crystalline form and the present invention is intended to include the various crystalline forms and mixtures thereof.
Typically, crystallization will result in solvates of the compounds of the invention. The term "solvate" as used herein refers to an aggregate comprising one or more molecules of the compound of the present invention and one or more solvent molecules. The solvent may be water, in which case the solvate is a hydrate. Alternatively, the solvent may be an organic solvent. Thus, the compounds of the present invention may exist as hydrates, including monohydrates, dihydrate, hemihydrate, sesquihydrates, trihydrate, tetrahydrate, and the like, as well as the corresponding solvated forms. The compounds of the invention may form true solvates, but in some cases it is also possible to retain only adventitious water or a mixture of water plus a portion of adventitious solvent. The compounds of the invention may be reacted in a solvent or precipitated or crystallized from a solvent. Solvates of the compounds of the invention are also included within the scope of the invention.
The invention also includes prodrugs of the above compounds. In the present application, the term "prodrug" denotes a compound that can be converted under physiological conditions or by solvolysis to the biologically active compound of the invention. Thus, the term "prodrug" refers to a pharmaceutically acceptable metabolic precursor of a compound of the invention. Prodrugs may not be active when administered to a subject in need thereof, but are converted in vivo to the active compounds of the invention. Prodrugs are generally rapidly converted in vivo to yield the parent compound of the invention, for example, by hydrolysis in blood. Prodrug compounds generally provide solubility, histocompatibility, or sustained release advantages in mammalian organisms. Prodrugs include known amino protecting groups and carboxyl protecting groups. Specific methods for preparing prodrugs can be found in Saulnier, M.G., et al, bioorg.Med.chem.Lett.1994,4, 1985-1990; greenwald, r.b., et al, j.med.chem.2000,43,475.
In the present application, a "pharmaceutical composition" refers to a formulation of a compound of the present invention with a vehicle generally accepted in the art for delivery of biologically active compounds to a mammal (e.g., a human). The medium includes a pharmaceutically acceptable carrier. The purpose of the pharmaceutical composition is to facilitate administration to an organism, facilitate absorption of active ingredients and exert biological activity.
The term "pharmaceutically acceptable" as used herein refers to a substance (e.g., carrier or diluent) that does not affect the biological activity or properties of the compounds of the present invention and is relatively non-toxic, i.e., the substance can be administered to an individual without causing an adverse biological response or interacting in an adverse manner with any of the components contained in the composition.
As used herein, a "pharmaceutically acceptable carrier" includes, but is not limited to, any adjuvant, carrier, excipient, glidant, sweetener, diluent, preservative, dye/colorant, flavoring agent, surfactant, wetting agent, dispersing agent, suspending agent, stabilizing agent, isotonic agent, solvent, or emulsifying agent that is approved by the relevant governmental regulatory agency for human or livestock use.
The "tumor" and "diseases related to abnormal cell proliferation" include, but are not limited to, leukemia, gastrointestinal stromal tumor, histiocytic lymphoma, non-small cell lung cancer, pancreatic cancer, squamous cell lung cancer, lung adenocarcinoma, breast cancer, prostate cancer, liver cancer, skin cancer, epithelial cell cancer, cervical cancer, ovarian cancer, intestinal cancer, nasopharyngeal cancer, brain cancer, bone cancer, esophageal cancer, melanoma, renal cancer, oral cancer, and the like.
The terms "preventing," "prevention," and "prevention" as used herein include reducing the likelihood of occurrence or worsening of a disease or disorder in a patient.
As used herein, the term "treatment" and other similar synonyms include the following meanings:
(i) preventing the occurrence of a disease or condition in a mammal, particularly when such mammal is susceptible to the disease or condition, but has not been diagnosed as having the disease or condition;
(ii) inhibiting the disease or disorder, i.e., arresting its development;
(iii) alleviating the disease or condition, i.e., causing regression of the state of the disease or condition; or
(iv) Alleviating the symptoms caused by the disease or disorder.
The terms "effective amount," "therapeutically effective amount," or "pharmaceutically effective amount" as used herein, refer to an amount of at least one agent or compound that is sufficient to alleviate one or more symptoms of the disease or disorder being treated to some extent after administration. The result may be a reduction and/or alleviation of signs, symptoms, or causes, or any other desired change in a biological system. For example, an "effective amount" for treatment is the amount of a composition comprising a compound disclosed herein that is clinically necessary to provide a significant remission effect of the condition. An effective amount suitable in any individual case can be determined using techniques such as a dose escalation assay.
The terms "administering," "administration," "administering," and the like as used herein refer to a method capable of delivering a compound or composition to a desired site for biological action. These methods include, but are not limited to, oral routes, via the duodenal route, parenteral injection (including intravenous, subcutaneous, intraperitoneal, intramuscular, intraarterial injection or infusion), topical administration, and rectal administration. Administration techniques useful for The compounds and methods described herein are well known to those skilled in The art, for example, in Goodman and Gilman, The pharmaceutical Basis of Therapeutics, current ed.; pergamon; and Remington's, Pharmaceutical Sciences (current edition), Mack Publishing Co., Easton, Pa. In preferred embodiments, the compounds and compositions discussed herein are administered orally.
The terms "drug combination", "administering other treatment", "administering other therapeutic agent" and the like as used herein refer to a drug treatment obtained by mixing or combining more than one active ingredient, including fixed and unfixed combinations of active ingredients. The term "fixed combination" refers to the simultaneous administration of at least one compound described herein and at least one co-agent to a patient in the form of a single entity or a single dosage form. The term "non-fixed combination" refers to the simultaneous administration, concomitant administration, or sequential administration at variable intervals of at least one compound described herein and at least one synergistic formulation to a patient as separate entities. These also apply to cocktail therapy, for example the administration of three or more active ingredients.
It will also be appreciated by those skilled in the art that in the processes described below, the functional groups of the intermediate compounds may need to be protected by suitable protecting groups. Such functional groups include hydroxyl, amino, mercapto and carboxylic acid. Suitable hydroxy protecting groups include trialkylsilyl or diarylalkylsilyl groups (e.g.tert-butyldimethylsilyl, tert-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like. Suitable protecting groups for amino, amidino and guanidino include t-butyloxycarbonyl, benzyloxycarbonyl and the like. Suitable thiol protecting groups include-C (O) -R "(where R" is alkyl, aryl or aralkyl), p-methoxybenzyl, trityl and the like. Suitable carboxyl protecting groups include alkyl, aryl or aralkyl esters.
Protecting groups may be introduced and removed according to standard techniques known to those skilled in the art and as described herein. The use of protecting Groups is described in detail in Greene, T.W. and P.G.M.Wuts, Protective Groups in Organic Synthesis, (1999),4th Ed., Wiley. The protecting group may also be a polymeric resin.
The invention will be further illustrated with reference to specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. Experimental procedures without specific conditions noted in the following examples, generally according to conventional conditions, or according to conditions recommended by the manufacturer. Percentages and parts are by weight unless otherwise indicated.
The first preparation method of the intermediate comprises the following steps: synthesis of fused ring pyridone compounds
Referring to synthetic routes and methods of patents WO2019110751A1 and WO2018217651A1, fused ring pyridone intermediate compounds 1A-1H are prepared.
Figure BDA0002608844410000141
Figure BDA0002608844410000151
And a second intermediate preparation method comprises the following steps: synthesis of piperazine compound
Referring to the synthetic routes and methods of WO2019110751A1 and US20190062330A1, piperazine intermediate compounds 2A-2C are prepared
Figure BDA0002608844410000152
Examples general preparative method one
Figure BDA0002608844410000153
The first step is as follows: dissolving the fused ring pyridone intermediate (1eq.) in anhydrous tetrahydrofuran, sequentially adding N, N-Diisopropylethylamine (DIPEA) (1.6eq.) and piperazine intermediate (1.5eq.) and heating and refluxing for 18 hours under the protection of nitrogen. Monitoring the reaction completion by Thin Layer Chromatography (TLC), cooling to room temperature, concentrating under reduced pressure, adding water and dichloromethane into the residue, separating the phase, extracting the water phase with dichloromethane three times, drying the extract with anhydrous sodium sulfate, concentrating under reduced pressure, separating and purifying the residue with silica gel column chromatography to obtain the target product, and confirming the structure by nuclear magnetic and mass spectrometry.
The second step is that: the product of the first step (1eq.) was dissolved in N, N-Dimethylformamide (DMF), bis (trimethylsilyl) aminolithium (LiHMDS) (3eq.) was added, and the mixture was heated to 100 ℃ under nitrogen and stirred for 8 hours. And monitoring the reaction by TLC (thin layer chromatography), cooling to room temperature, pouring into saturated ammonium chloride aqueous solution, separating out solids, filtering, drying a filter cake in vacuum to obtain a target product, and confirming the structure by adopting nuclear magnetism and mass spectrum.
The third step: the second-step product (1eq.) was dissolved in a mixed solvent of anhydrous dioxane/water (4/1), and boric acid or potassium trifluoroborate salt (2eq.), anhydrous potassium carbonate powder (2.5eq.) and [1,1' -bis (diphenylphosphino) ferrocene ] palladium dichloride (pd (dppf) Cl2) (0.1eq.) were sequentially added thereto, and the mixture was heated under reflux for 2 hours under nitrogen protection. TLC monitoring reaction is complete, cooling to room temperature, decompression concentrating, diluting the remainder with dichloromethane, washing with saturated ammonium chloride solution and saturated salt solution in turn, drying with anhydrous sodium sulfate, filtering, decompression concentrating, separating and purifying the remainder with silica gel column chromatography to obtain the target product, and confirming the structure by nuclear magnetism and mass spectrum.
The fourth step: the product of the third step (1eq.) was dissolved in methanol, and 4M HCl/methanol solution (20eq.) was added and stirred at room temperature for 3 hours. TLC monitors the reaction to be complete, and the reaction is concentrated under reduced pressure, the residue is dissolved in dichloromethane, DIPEA (3eq.) and acryloyl chloride (1eq.) are added in turn at 0 ℃, the mixture is stirred for 0.5 hour, the reaction solution is washed by saturated ammonium chloride solution and saturated common salt solution, anhydrous sodium sulfate is dried, the filtration and the concentration under reduced pressure are carried out, the residue is separated and purified by silica gel column chromatography to obtain the target compound, and the structure is confirmed by nuclear magnetism and mass spectrometry.
EXAMPLES general preparation method II
Figure BDA0002608844410000161
The first step is as follows: dissolving the fused ring pyridone intermediate (1eq.) in anhydrous tetrahydrofuran, sequentially adding DIPEA (1.6eq.) and piperazine intermediate (1.5eq.), and heating and refluxing for 18 hours under the protection of nitrogen. TLC monitoring reaction is completed, cooling to room temperature, decompression concentrating, adding water and dichloromethane into residues for phase separation, extracting water phase with dichloromethane for three times, drying extraction liquid anhydrous sodium sulfate, decompression concentrating, separating and purifying the residues by silica gel column chromatography to obtain target products, and confirming the structure by adopting nuclear magnetism and mass spectrum.
The second step is that: dissolving the product of the first step (1eq.) in anhydrous glacial acetic acid, slowly adding reduced iron powder (3eq.), heating to 80 ℃ under the protection of nitrogen, and stirring for 0.5 hour. After the reaction was completed, the mixture was filtered through celite, washed with ethyl acetate, and the filtrate was concentrated. Diluting the residue with dichloromethane, washing with saturated sodium bicarbonate solution and saturated saline solution in sequence, drying with anhydrous sodium sulfate, filtering, concentrating under reduced pressure, separating and purifying the residue with silica gel column chromatography to obtain the target product, and confirming the structure by nuclear magnetism and mass spectrometry.
The third step: the second-step product (1eq.) was dissolved in DMF, sodium hydrogen (1.5eq.) was added at 0 ℃, stirred for 0.5 h, methyl iodide (1.1eq.) was added, and the reaction was allowed to proceed overnight at room temperature. After the reaction is finished, decompressing and concentrating the reaction liquid, diluting the remainder with dichloromethane, washing the remainder with saturated sodium bicarbonate solution and saturated saline solution in sequence, drying the remainder with anhydrous sodium sulfate, filtering the mixture, decompressing and concentrating the mixture, separating and purifying the remainder by silica gel column chromatography to obtain a target product, and confirming the structure by adopting nuclear magnetism and mass spectrometry.
The fourth step: dissolving the product of the third step (1eq.) in a mixed solvent of anhydrous dioxane and water (4/1), and sequentially adding boric acid or potassium trifluoroborate salt (2eq.), anhydrous potassium carbonate powder (2.5eq.) and Pd (dppf) Cl2(0.1eq.) and reflux with heating under nitrogen for 2 hours. TLC monitoring reaction is complete, cooling to room temperature, decompression concentrating, diluting the remainder with dichloromethane, washing with saturated ammonium chloride solution and saturated salt solution in turn, drying with anhydrous sodium sulfate, filtering, decompression concentrating, separating and purifying the remainder with silica gel column chromatography to obtain the target product, and confirming the structure by nuclear magnetism and mass spectrum.
The fifth step: the product of the fourth step (1eq.) was dissolved in methanol, and 4M hydrogen chloride/methanol solution (20eq.) was added and stirred at room temperature for 3 hours. TLC monitors the reaction to be complete, and the reaction is concentrated under reduced pressure, the residue is dissolved in dichloromethane, DIPEA (3eq.) and acryloyl chloride (1eq.) are added in turn at 0 ℃, the mixture is stirred for 0.5 hour, the reaction solution is washed by saturated ammonium chloride solution and saturated common salt solution, anhydrous sodium sulfate is dried, the filtration and the concentration under reduced pressure are carried out, the residue is separated and purified by silica gel column chromatography to obtain the target compound, and the structure is confirmed by nuclear magnetism and mass spectrometry.
Examples general preparative method three
Figure BDA0002608844410000171
The first step is as follows: the condensed ring pyridone amide intermediate (1eq.) is dissolved in a mixed solvent of acetonitrile/triethylamine (3/1), phosphorus pentasulfide (1eq.) is added in batches, and the mixture is heated and refluxed for 3 hours under the protection of nitrogen. After the reaction is finished, cooling to room temperature, separating out solids, filtering, drying a filter cake in vacuum to obtain a target product, and confirming the structure by adopting nuclear magnetism and mass spectrometry.
The second step is that: dissolving the product of the first step (1eq.) in a mixed solvent of ethanol and DIPEA (5/1), adding 2, 2-dimethoxyethylamine (1eq.), and heating and refluxing for 6 hours under the protection of nitrogen. After the reaction is finished, cooling to room temperature, decompressing and concentrating, dissolving the residue in glacial acetic acid, heating to 100 ℃, and stirring for 2 hours. Cooling to room temperature, concentrating under reduced pressure, diluting the residue with dichloromethane, washing with saturated sodium bicarbonate solution and saturated sodium chloride solution sequentially, drying with anhydrous sodium sulfate, filtering, concentrating under reduced pressure, separating and purifying the residue with silica gel column chromatography to obtain the target product, and confirming the structure by nuclear magnetism and mass spectrometry.
The third step: dissolving the second-step product (1eq.) in a mixed solvent of anhydrous dioxane and water (4/1), and sequentially adding boric acid or potassium trifluoroborate salt (2eq.), anhydrous potassium carbonate powder (2.5eq.) and Pd (dppf) Cl2(0.1eq.) and reflux with heating under nitrogen for 2 hours. TLC monitoring reaction is complete, cooling to room temperature, decompression concentrating, diluting the remainder with dichloromethane, washing with saturated ammonium chloride solution and saturated salt solution in turn, drying with anhydrous sodium sulfate, filtering, decompression concentrating, separating and purifying the remainder with silica gel column chromatography to obtain the target product, and confirming the structure by nuclear magnetism and mass spectrum.
The fourth step: the product of the third step (1eq.) was dissolved in methanol, and 4M HCl in methanol (20eq.) was added and stirred at room temperature for 3 hours. TLC monitors the reaction to be complete, and the reaction is concentrated under reduced pressure, the residue is dissolved in dichloromethane, DIPEA (3eq.) and acryloyl chloride (1eq.) are added in turn at 0 ℃, the mixture is stirred for 0.5 hour, the reaction solution is washed by saturated ammonium chloride solution and saturated common salt solution, anhydrous sodium sulfate is dried, the filtration and the concentration under reduced pressure are carried out, the residue is separated and purified by silica gel column chromatography to obtain the target compound, and the structure is confirmed by nuclear magnetism and mass spectrometry.
Examples general preparative method four
Figure BDA0002608844410000181
The first step is as follows: the fused ring pyridone sulfamide intermediate (1eq.) is dissolved in tetrahydrofuran, and hydrazine hydrate (1.5eq.) is added and heated under reflux for 6 hours under the protection of nitrogen. After the reaction is finished, cooling to room temperature, carrying out reduced pressure concentration, separating out solids, filtering, carrying out vacuum drying on a filter cake to obtain a target product, and confirming the structure by adopting nuclear magnetism and mass spectrometry.
The second step is that: the product of the first step (1eq.) was dissolved in dichloromethane, trimethyl orthoformate (4eq.) was added, and after stirring for ten minutes trifluoroacetic acid (1eq.) was added. And continuously stirring for one hour at room temperature, concentrating under reduced pressure, separating and purifying the residue by using a silica gel column chromatography to obtain a target product, and confirming the structure by adopting nuclear magnetism and mass spectrometry.
The third step: dissolving the second-step product (1eq.) in a mixed solvent of anhydrous dioxane and water (4/1), and sequentially adding boric acid or potassium trifluoroborate salt (2eq.), anhydrous potassium carbonate powder (2.5eq.) and Pd (dppf) Cl2(0.1eq.) and reflux with heating under nitrogen for 2 hours. TLC monitoring reaction is complete, cooling to room temperature, decompression concentrating, diluting the remainder with dichloromethane, washing with saturated ammonium chloride solution and saturated salt solution in turn, drying with anhydrous sodium sulfate, filtering, decompression concentrating, separating and purifying the remainder with silica gel column chromatography to obtain the target product, and confirming the structure by nuclear magnetism and mass spectrum.
The fourth step: the product of the third step (1eq.) was dissolved in methanol, and 4M HCl in methanol (20eq.) was added and stirred at room temperature for 3 hours. TLC monitors the reaction to be complete, and the reaction is concentrated under reduced pressure, the residue is dissolved in dichloromethane, DIPEA (3eq.) and acryloyl chloride (1eq.) are added in turn at 0 ℃, the mixture is stirred for 0.5 hour, the reaction solution is washed by saturated ammonium chloride solution and saturated common salt solution, anhydrous sodium sulfate is dried, the filtration and the concentration under reduced pressure are carried out, the residue is separated and purified by silica gel column chromatography to obtain the target compound, and the structure is confirmed by nuclear magnetism and mass spectrometry.
Examples general preparative method five
Figure BDA0002608844410000192
The first step is as follows: dissolving the fused ring pyridone sulfamide intermediate (1eq.) in a mixed solvent of methanol/concentrated ammonia water (1/1), placing in a sealed tank, adding a catalytic amount of p-toluenesulfonic acid, heating to 100 ℃, and stirring for 6 hours. After the reaction, the reaction solution was concentrated under reduced pressure. The residue was diluted with dichloromethane, washed successively with a saturated sodium bicarbonate solution and a saturated sodium chloride solution, dried over anhydrous sodium sulfate, filtered, and concentrated under reduced pressure, and the residue was used directly in the next reaction.
The second step is that: the crude product of the first step (1eq.) was suspended in isopropanol and dimethylformamide-dimethylacetal (1.2eq.) was added dropwise at room temperature. After the dropwise addition, the reaction solution was refluxed for three hours under heating, cooled to room temperature, added with hydroxylamine hydrochloride (1.2eq.) and stirred at 50 ℃ overnight. The reaction solution was cooled to room temperature, concentrated under reduced pressure, dried, and the residue was suspended in anhydrous tetrahydrofuran, cooled in an ice bath, and then trifluoroacetic anhydride (1.5eq.) was slowly added dropwise. After the addition was complete, the ice bath was removed and stirred at room temperature overnight. Slowly dripping saturated sodium bicarbonate solution into the reaction solution, extracting with dichloromethane, washing with water and saturated saline solution in sequence, drying with anhydrous sodium sulfate, filtering, concentrating under reduced pressure, separating and purifying the residue with silica gel column chromatography to obtain the target product, and confirming the structure by adopting nuclear magnetism and mass spectrometry.
The third step: dissolving the second-step product (1eq.) in a mixed solvent of anhydrous dioxane and water (4/1), and sequentially adding boric acid or potassium trifluoroborate salt (2eq.), anhydrous potassium carbonate powder (2.5eq.) and Pd (dppf) Cl2(0.1eq.) under nitrogen protectionHot reflux for 2 hours. TLC monitoring reaction is complete, cooling to room temperature, decompression concentrating, diluting the remainder with dichloromethane, washing with saturated ammonium chloride solution and saturated salt solution in turn, drying with anhydrous sodium sulfate, filtering, decompression concentrating, separating and purifying the remainder with silica gel column chromatography to obtain the target product, and confirming the structure by nuclear magnetism and mass spectrum.
The fourth step: the product of the third step (1eq.) was dissolved in methanol, and 4M HCl in methanol (20eq.) was added and stirred at room temperature for 3 hours. TLC monitors the reaction to be complete, and the reaction is concentrated under reduced pressure, the residue is dissolved in dichloromethane, DIPEA (3eq.) and acryloyl chloride (1eq.) are added in turn at 0 ℃, the mixture is stirred for 0.5 hour, the reaction solution is washed by saturated ammonium chloride solution and saturated common salt solution, anhydrous sodium sulfate is dried, the filtration and the concentration under reduced pressure are carried out, the residue is separated and purified by silica gel column chromatography to obtain the target compound, and the structure is confirmed by nuclear magnetism and mass spectrometry.
Examples
The following compounds of examples were prepared using intermediates 1-2 and other commercial reagents as starting materials, by synthetic methods of examples general preparative methods one to five, respectively.
Figure BDA0002608844410000191
Figure BDA0002608844410000201
Figure BDA0002608844410000211
Figure BDA0002608844410000221
Test example 1: test of Effect of Compounds of the present invention on the cell proliferation of NCI-H358, MiaPaca-2 and phosphorylation of downstream Signal ERK
Test method one (2D) NCI-H358 (Lung)Carcinoma) and MiaPaca-2 cells (pancreatic cancer) cells (100 μ L/well, 20000 cells/mL) were seeded in 96-well culture plates and supplemented with 10% fetal bovine serum and 1% penicillin/streptomycin sulfate. Cells were treated with a 0.5% dimethyl sulfoxide blank, diluted with an initial 10 μ M solution of test compound diluted three times with an eight gradient, and incubated in a 5% CO2 incubator for a period of time (3-7 days). At the end of the incubation, 10. mu.L of MTT stock solution (5mg/mL) was added to each well. The plates were incubated at 37 ℃ for 4 hours and then the medium was removed. Dimethylsulfoxide (100 μ L) was added to each well, followed by sufficient shaking. The absorbance of the formazan product was measured at 570nm on a Thermo Scientific Varioskan Flash multimodal reader. IC was obtained by fitting dose-response data to a three-parameter nonlinear regression model using GraphPad Prism 6.0 software50The value is obtained.
As a result, the compounds of the examples provided in the present invention have proliferation inhibitory activity, IC, on NCI-H358 and MiaPaca-2 cells50All less than 5000nM, some examples being ICs such as 1, 3, 6,7,8, 12, 13, 14, 15, etc50Values were less than 500 nM.
Test method two (3D) tumor cells in logarithmic growth phase were diluted to a certain concentration with culture medium and seeded in 96-well plates with ultra-low attachment surface at 80. mu.L/well. Cells were incubated overnight at 37 ℃ in a humidity chamber. The next day the plate was added serial dilutions of test compound (10 concentrations, 3-fold dilution), 20 μ L/well and incubated in incubator for 96 h. Taking out the plate, standing at room temperature, and adding the same volume
Figure BDA0002608844410000231
Incubation with 3D reagent for 1h, En VisionTMThe plate reader detects the signal. The signal was converted to percent inhibition using the following equation: % inhibition 100- [ (test compound signal-median minimum signal)/(median maximum signal-median minimum signal) x 100]. Maximum signal is the signal value from wells without inhibitor and minimum signal is the signal value from wells containing a reference inhibitor sufficient to completely inhibit cell proliferation, a four-parameter non-linear regression fit curve is performed on the percent inhibition for each concentration of compound and the IC is calculated50
As a result: the example compounds provided herein have proliferation-inhibiting activity, IC, on NCI-H358 and MiaPaca-2 cells50Less than 1000nM each, some examples, e.g., examples 1, 3, 8, 12, 13, 15, etc., IC, of cell proliferation inhibitory activity on NCI-H358 and MiaPaca-250Less than 200 nM.
Test method three (ERK phosphorylation): miapaca-2 or H358 cells were seeded at a certain concentration in 96-well plates and placed at 37 ℃ in 5% CO2The next day the plate was incubated overnight with serial dilutions of test compounds (5 concentrations, 3 fold dilutions) for 24H (Miapaca-2) or 3H (H358), followed by lysis of the cells with lysis solutions containing protease and phosphatase inhibitors to extract the protein, and western blot to detect the level of p-ERK.
As a result: some of the compounds of the examples provided by the invention have obvious inhibition effect on the level of phosphorylated ERK of NCI-H358 and MiaPaca-2 and IC (integrated circuit) as examples 1, 3, 8, 12, 13, 15 and the like50Less than 500 nM.
All documents referred to herein are incorporated by reference into this application as if each were individually incorporated by reference. Furthermore, it should be understood that various changes and modifications of the present invention can be made by those skilled in the art after reading the above teachings of the present invention, and these equivalents also fall within the scope of the present invention as defined by the appended claims.

Claims (7)

1. A fused ring pyridone compound shown in formula I or pharmaceutically acceptable salt thereof,
Figure FDA0003378875300000011
in the formula:
r1 is independently selected from hydrogen, halogen, cyano, C1-C6An alkyl group;
r2 and R3 are independently selected from hydrogen, halogen and C1-C6An alkyl group;
w2 is CH or N;
w, W1 are independently selected from CR4 or N, R4 are independently selected from halogen, alkenyl or 3-5 membered cycloalkyl;
ra, Rb, Rc, Rd, Re, Rf, Rg are each independently selected from hydrogen, C1-C6 alkyl; and when Ra is different from Rb, the carbon atom to which Ra and Rb are attached is in the R configuration;
ar is independently selected from a 5-12 membered aromatic ring or aromatic fused ring group, a 5-12 membered aromatic heterocycle or aromatic fused heterocyclic group; the 5-12 membered aromatic heterocycle or aromatic fused heterocycle group contains 1-3 heteroatoms independently selected from N, O, P, S; further, one or more hydrogen atoms on Ar may be substituted with: halogen, hydroxy, C1-C6 alkyl or amino;
z is independently selected from NR5, R5 is independently selected from H, C1-C6An alkyl group;
when Z is NR5, Ri and Rj are oxo;
r is independently selected from unsubstituted 5-10 membered aryl, unsubstituted 5-10 membered heteroaryl, substituted 5-10 membered aryl or substituted 5-10 membered heteroaryl; each of said 5-10 membered heteroaryl groups contains 1-3 heteroatoms independently selected from N, O, P, S; when R is substituted, the substituent is independently selected from C1-C6 alkyl, and the number of the substituent is one or more.
2. The compound of claim 1, or a pharmaceutically acceptable salt thereof, wherein:
r1 is selected from hydrogen;
ra, Rb, Rc, Rd, Re, Rf and Rg are respectively and independently selected from hydrogen and methyl;
w2 is CH;
w1 is independently selected from CR4, R4 is independently selected from halogen, C2-C4 alkenyl, 3-5 membered cycloalkyl;
w is independently selected from N or CR4, R4 is independently selected from halogen;
ar is independently selected from substituted phenyl, substituted pyridyl, substituted naphthyl, substituted indazolyl, substituted benzimidazolyl, substituted benzothiazolyl; the one or more substituents are independently selected from the group consisting of: halogen, hydroxyl, alkyl of C1-C4, amino;
r is independently selected from substituted 5-10 membered aryl or heteroaryl; the substituents are independently selected from methyl, isopropyl.
3. The compound of claim 2, or a pharmaceutically acceptable salt thereof, wherein:
w1 is CR4, R4 is independently selected from halogen, vinyl or cyclopropyl;
w is N or C-F;
r is substituted pyridyl, and the substituents are independently selected from methyl and isopropyl.
4. A fused ring pyridone compound or a pharmaceutically acceptable salt thereof, which is characterized in that the compound is any one of the following compounds:
Figure FDA0003378875300000021
5. the use of a compound of any one of claims 1-4, or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of a disease associated with a Ras protein mutation.
6. The use according to claim 5, wherein the disease is a tumor selected from the group consisting of lung cancer, pancreatic cancer, liver cancer, colorectal cancer, bile duct cancer, brain cancer, leukemia, lymphoma, melanoma, thyroid cancer and nasopharyngeal cancer.
7. A pharmaceutical composition, comprising:
(i) an effective amount of a compound of any one of claims 1-4, or a pharmaceutically acceptable salt thereof; and
(ii) a pharmaceutically acceptable carrier.
CN202010747468.2A 2019-07-30 2020-07-29 Condensed ring pyridone compounds, preparation method and application Active CN112300194B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2019106946147 2019-07-30
CN201910694614 2019-07-30

Publications (2)

Publication Number Publication Date
CN112300194A CN112300194A (en) 2021-02-02
CN112300194B true CN112300194B (en) 2022-01-14

Family

ID=74483343

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010747468.2A Active CN112300194B (en) 2019-07-30 2020-07-29 Condensed ring pyridone compounds, preparation method and application

Country Status (1)

Country Link
CN (1) CN112300194B (en)

Families Citing this family (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20230227472A1 (en) * 2019-09-20 2023-07-20 Shanghai Jemincare Pharmaceuticals Co., Ltd Fused pyridone compound, and preparation method therefor and use thereof
CN116947885A (en) * 2019-09-20 2023-10-27 上海济煜医药科技有限公司 Condensed pyridone compound, preparation method and application thereof
AU2020374844B2 (en) * 2019-10-30 2024-01-25 Genfleet Therapeutics (Shanghai) Inc. Substituted heterocyclic fused cyclic compound, preparation method therefor and pharmaceutical use thereof
CN116406363A (en) * 2020-08-21 2023-07-07 浙江海正药业股份有限公司 Tetracyclic derivative, preparation method and medical application thereof
CN116670136A (en) * 2020-12-31 2023-08-29 正大天晴药业集团股份有限公司 Tetracyclic compound and medical application thereof
TW202246267A (en) * 2021-03-25 2022-12-01 大陸商上海濟煜醫藥科技有限公司 Fused pyridone compound salt form and crystal form, and application thereof
CN116848111A (en) * 2021-03-26 2023-10-03 浙江海正药业股份有限公司 KRAS inhibitor key intermediate and preparation method thereof
CN115124533A (en) * 2021-03-26 2022-09-30 浙江海正药业股份有限公司 Tetracyclic derivative, preparation method and medical application thereof
WO2022223037A1 (en) * 2021-04-22 2022-10-27 劲方医药科技(上海)有限公司 Salt or polymorph of kras inhibitor
US20230106174A1 (en) 2021-05-05 2023-04-06 Revolution Medicines, Inc. Ras inhibitors
KR20240017811A (en) 2021-05-05 2024-02-08 레볼루션 메디슨즈, 인크. RAS inhibitors for the treatment of cancer
CA3226720A1 (en) * 2021-07-23 2023-01-26 Suzhou Zanrong Pharma Limited Kras g12d inhibitors and uses thereof
WO2023030517A1 (en) * 2021-09-06 2023-03-09 Suzhou Zanrong Pharma Limited Kras g12c inhibitors and uses thereof
TW202325298A (en) * 2021-11-01 2023-07-01 大陸商江蘇恆瑞醫藥股份有限公司 A nitrogen-containing tetracyclic compound, a preparation method and medical use thereof
TW202327593A (en) * 2021-12-24 2023-07-16 大陸商勁方醫藥科技(上海)有限公司 Polymorph of kras inhibitor and preparation method for polymorph and use thereof
CN116327956A (en) * 2022-04-01 2023-06-27 劲方医药科技(上海)有限公司 Pharmaceutical composition, use thereof and method for treating cancer
WO2024041621A1 (en) * 2022-08-25 2024-02-29 Jacobio Pharmaceuticals Co., Ltd. K-ras mutant protein inhibitors

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104844573A (en) * 2015-04-17 2015-08-19 中国药科大学 Miazines BTK inhibitor, preparation method and medical application thereof
WO2016044772A1 (en) * 2014-09-18 2016-03-24 Araxes Pharma Llc Combination therapies for treatment of cancer
WO2018217651A1 (en) * 2017-05-22 2018-11-29 Amgen Inc. Kras g12c inhibitors and methods of using the same
WO2020239123A1 (en) * 2019-05-31 2020-12-03 上海翰森生物医药科技有限公司 Aromatic heterocyclic derivative modulator and preparation method therefor and use thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016044772A1 (en) * 2014-09-18 2016-03-24 Araxes Pharma Llc Combination therapies for treatment of cancer
TW201625305A (en) * 2014-09-18 2016-07-16 亞瑞克西斯製藥公司 Combination therapies for treatment of cancer
CN104844573A (en) * 2015-04-17 2015-08-19 中国药科大学 Miazines BTK inhibitor, preparation method and medical application thereof
WO2018217651A1 (en) * 2017-05-22 2018-11-29 Amgen Inc. Kras g12c inhibitors and methods of using the same
WO2020239123A1 (en) * 2019-05-31 2020-12-03 上海翰森生物医药科技有限公司 Aromatic heterocyclic derivative modulator and preparation method therefor and use thereof

Also Published As

Publication number Publication date
CN112300194A (en) 2021-02-02

Similar Documents

Publication Publication Date Title
CN112300194B (en) Condensed ring pyridone compounds, preparation method and application
CN111704611B (en) Aryl spiro SHP2 inhibitor compound, preparation method and application
CN111153901B (en) Nitrogen-containing fused heterocyclic SHP2 inhibitor compound, preparation method and application
CN112142735B (en) Condensed cyanopyridine compound, preparation method and application
CA3177261A1 (en) Benzothiazolyl biaryl compound, and preparation method and use
AU2019374194A1 (en) Spiro aromatic ring compound and application thereof
CN109721599B (en) Amino-substituted nitrogen-containing fused ring compound and preparation method and application thereof
CN109721600B (en) Nitrogen-containing fused ring compounds and preparation method and application thereof
CN112300196A (en) Piperidine condensed ring compound, preparation method and application
CN113527299B (en) Nitrogen-containing condensed ring compound, preparation method and application
CN110964012B (en) Fused heterocyclic biaryl benzyl alcohol compound, preparation method and application
CN112457326B (en) Aromatic heterocyclic lactam compound, preparation method and application
CN110857300B (en) Condensed ring triazole compounds, preparation method and application
CN111518100A (en) Cyclopropenoarylbenzofuran substituted nitrogen heteroaryl compound and application thereof
CN112094269B (en) Saturated six-membered ring heterocyclic compound, preparation method and application
CN112300173B (en) Nitrogen-containing polycyclic compounds, preparation method and application
CN114524810A (en) Pyrimidine heterocyclic compound, preparation method and application
CN112778336A (en) Nitrogen-containing fused ring STING regulator compounds, preparation method and application
WO2020187123A1 (en) Pyrrole amidopyridone compound, preparation method therefor and use thereof
WO2023143147A1 (en) Pyridazopyridone compounds, pharmaceutical composition thereof and use thereof
CN114907350B (en) Nitrogen-containing condensed ring compound, preparation method and application
CN111057048B (en) Aminopyrazine/pyridine compound, preparation method and application
CN115960098A (en) Preparation method and application of nitrogen-containing fused ring compounds
CN111763217B (en) Thieno-nitrogen heterocyclic compounds, preparation method and application
CN110229151B (en) Indolizine compound, preparation method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant
EE01 Entry into force of recordation of patent licensing contract
EE01 Entry into force of recordation of patent licensing contract

Application publication date: 20210202

Assignee: GENFLEET THERAPEUTICS (SHANGHAI) Inc.

Assignor: Shanghai Lingda biomedical Co.,Ltd.

Contract record no.: X2024980002019

Denomination of invention: A class of fused pyridine ketone compounds, preparation methods, and applications

Granted publication date: 20220114

License type: Common License

Record date: 20240207