CN114195805A - DNA-PK selective inhibitor and preparation method and application thereof - Google Patents

DNA-PK selective inhibitor and preparation method and application thereof Download PDF

Info

Publication number
CN114195805A
CN114195805A CN202010998179.XA CN202010998179A CN114195805A CN 114195805 A CN114195805 A CN 114195805A CN 202010998179 A CN202010998179 A CN 202010998179A CN 114195805 A CN114195805 A CN 114195805A
Authority
CN
China
Prior art keywords
alkyl
methyl
dihydro
purin
pyran
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
CN202010998179.XA
Other languages
Chinese (zh)
Inventor
陈曦
侯登
陈坤成
雷永珂
任仁
闵汪洋
刘志华
刘希杰
张凯
孙永亮
路畅
张苗
孙颖慧
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.)
Capital Pharmaceutical Holdings Beijing Co ltd
Original Assignee
Capital Pharmaceutical Holdings Beijing 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 Capital Pharmaceutical Holdings Beijing Co ltd filed Critical Capital Pharmaceutical Holdings Beijing Co ltd
Priority to CN202010998179.XA priority Critical patent/CN114195805A/en
Priority to CN202180061261.2A priority patent/CN116406272A/en
Priority to PCT/CN2021/107304 priority patent/WO2022017368A1/en
Publication of CN114195805A publication Critical patent/CN114195805A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • 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
    • C07D473/00Heterocyclic compounds containing purine ring systems
    • C07D473/26Heterocyclic compounds containing purine ring systems with an oxygen, sulphur, or nitrogen atom directly attached in position 2 or 6, but not in both
    • C07D473/32Nitrogen atom
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D513/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00
    • C07D513/22Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for in groups C07D463/00, C07D477/00 or C07D499/00 - C07D507/00 in which the condensed system contains four or more hetero rings

Landscapes

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

Abstract

The application relates to a DNA-PK selective inhibitor shown as a formula (I) and a preparation method and application thereof. The application comprises the application of the compound shown in the formula (I) in the preparation of medicaments for treating diseases related to DNA-PKA series of reactions, such as cyclization and alkylation, to give the compounds of the invention.

Description

DNA-PK selective inhibitor and preparation method and application thereof
Technical Field
The present invention relates to compounds that selectively inhibit the activity of DNA-PK proteins, to methods of making these compounds and salts thereof, and to methods of using such compounds and salts for the treatment of DNA-PK mediated diseases, including cancer.
Background
DNA-PK is a trimer consisting of a catalytic subunit DNA-PKcs (size 470kD) and 2 heterodimers composed of regulatory subunits Ku70 and Ku80, which polymerize upon contact with fragmented DNA. Wherein the DNA-PKcs is one of phosphatidylinositol-3 kinase family members, and participates in various biochemical reaction processes: repair of double-stranded DNA breaks (DSBs), signaling of programmed death, gene surveillance, maintenance of telomere structure, etc. (FASEB J, 2005, 19 (7): 704-715.). Radiation and a great deal of anticancer drugs can directly or indirectly act on DNA or DNA metabolic processes, so that DNA damage is caused, and a series of cellular reactions such as damaged DNA repair and the like are initiated, and the repair results in the improvement of cell survival, which is also one of the mechanisms of tumor cells for resisting radiotherapy and chemotherapy. Cell Res, 2008, 18 (1): 114-:
1) v (D) J chain rearrangements of immunoglobulin and T cell receptors, such as deletion of DNA-PKcs or Ku protein, Severe Combined Immunodeficiency (SCID) occurs in mammalian cells
2) The telomere structure is maintained to be stable, and the lack of Ku or DNA-PKcs can cause unstable genome, cell growth retardation and premature senility;
3) DNA-PKcs is a serine/threonine kinase, a member of PI-3-K (phosphine tip in to 1-3-kinase) kinase family (including A TM, A TR, etc.), and plays a role in the functions of cell signal transduction and cell cycle after DNA damage. (Int J radial Oncol Biol Phys, 2005, 61 (3): 915-.
A range of factors can induce DNA double strand breaks, including chemotherapy, radiation therapy, and PARP inhibitors such as olaparib. DNA-PK inhibitors have the potential to sensitize these therapies. A plurality of DNA-PK selective inhibitors enter a clinical stage at present all over the world, wherein two medicaments enter a clinical second stage, but no related medicament is on the market so far, and the requirements of the related medicament are not met. The DNA-PK selective inhibitor provided by the invention has high activity, strong drug resistance and small clinical side effect, can effectively enhance the sensitivity of radiotherapy and chemotherapy in tumor treatment, and has better economic value and application prospect.
Disclosure of Invention
The invention provides a DNA-PK selective inhibitor which is a compound shown as a general formula (I) or pharmaceutically acceptable salt, solvate, polymorph or isomer thereof, and also provides a series of compounds shown as the general formula (I) and pharmaceutically acceptable salt, solvate, polymorph or isomer thereof, a pharmaceutical composition containing the compounds, and a method for treating diseases by using the compounds.
In one aspect, the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt, solvate, polymorph or isomer thereof
Figure BSA0000220128980000021
Wherein the content of the first and second substances,
ring A is 6-10 membered aryl or 5-12 membered heteroaryl,
ring B is a 4-12 membered heterocyclic ring, S on ring B may be optionally oxidized,
z is O or S, and the compound is,
X2is CR2Or the number of N is greater than the number of N,
X1is CRR4O, S, or NR6
R1Is H, C1-6Alkyl, or C3-8Cycloalkyl, said alkyl and cycloalkyl being optionally substituted by halogen, -CN, -OH, -NH2、-O-C1-6Alkyl, or-NR-C1-6Alkyl substitution;
R7and R8Each independently selected from halogen, CN, C1-6Alkyl, -O-C1-6Alkyl and-NR-C1-6Alkyl, which may optionally be substituted by halogen, -CN, -OH, -NH2、-O-C1-6Alkyl, or-NR-C1-6The substitution of the alkyl group is carried out,
m and n are each independently 0, 1, 2, or 3,
R3is R5or-X3-R5
R4Is R6or-X3-R6
X3Each independently is-O-, -S-, or-NR-,
R5and R6Each independently selected from H and C1-6Alkyl, or R5And R6Taken together to form- (CH)2)p-X-(CH2)q-, wherein X is a bond, -O-, -S, -N (R) -, -CO-, -C (O) NR-, -C (O) O-, 6-10 membered arylene, 5-12 membered heteroarylene, or 3-12 membered heterocycle, and- (CH)2)p-X-(CH2)qCH in (E)2Optionally substituted by a halogen, and optionally substituted by a halogen,
p and q are each independently 0, 1, 2, 3, or 4, and p + q is 1, 2, 3, 4, 5, or 6,
R2selected from H, halogen, CHF2、CF3、-OH、-NH2、CN、C1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, -O-C1-6Alkyl, - (CH)2)1-6-CN、-(CH2)1-6-O-C1-6Alkyl, - (CH)2)1-3-OH、-CHO、-(CO)NH2- (CO) NHR, - (CO) OR and-NR-C1-6An alkyl group.
Each R is independently H or C1-6An alkyl group;
in certain embodiments, R5And R6Each independently selected from H and C1-6Alkyl, or R5And R6Taken together to form- (CH)2)p-X-(CH2)q-, wherein X is a bond, -O-, -S, -N (R) -, -CO-, -C (O) NR-, -C (O) O-, a 6-to 10-membered arylene, a 5-to 12-membered heteroarylene, or a 3-to 12-membered heterocycle;
in certain embodiments, when R5And R6Each independently selected from H or C1-6When alkyl, R2Selected from halogen, CH2F、CHF2、CF3、-OH、-NH2、CN、C1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, -O-C1-6Alkyl, - (CH)2)1-6-CN、-(CH2)1-6-O-C1-6Alkyl, or-NR-C1-6An alkyl group, a carboxyl group,
when R is5And R6Taken together to form- (CH)2)p-X-(CH2)qWhen is,R2Selected from H, halogen, CHF2、CF3、-OH、-NH2、CN、C1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, -O-C1-6Alkyl, - (CH)2)1-6-CN、-(CH2)1-6-O-C1-6Alkyl and-NR-C1-6An alkyl group, a carboxyl group,
r is H or C1-6An alkyl group;
in certain embodiments, R2Selected from H, halogen, CHF2、CF3、-OH、-NH2、CN、C1-6Alkyl, -O-C1-6Alkyl, - (CH)2)1-3-CN、-(CH2)1-3-O-C1-6Alkyl, - (CH)2)1-3-OH、-CHO、-(CO)NH2- (CO) OR and-NR-C1-6Alkyl, R is H or C1-6An alkyl group; preferably, R2Selected from H, halogen, CHF2、CF3、CN、C1-6Alkyl and- (CO) NH2(ii) a More preferably, R2Selected from halogen, CHF2、CF3、CN、C1-6Alkyl and- (CO) NH2
In certain embodiments, when
Figure BSA0000220128980000031
Is composed of
Figure BSA0000220128980000032
Figure BSA0000220128980000033
Or
Figure BSA0000220128980000034
When R is2Selected from halogen, CH2F、CHF2、CF3、-OH、-NH2、CN、C1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, -O-C1-6Alkyl, - (CH)2)1-6-CN、-(CH2)1-6-O-C1-6Alkyl and-NR-C1-6An alkyl group, a carboxyl group,
r is H or C1-6An alkyl group;
in certain embodiments, R1Is C1-6Alkyl or C3-8Cycloalkyl, said alkyl and cycloalkyl being optionally substituted by halogen, -CN, -OH, -NH2、-O-C1-6Alkyl, or-NR-C1-6Alkyl substituted, R is H or C1-6Alkyl, preferably said alkyl and cycloalkyl groups may be optionally substituted by halogen or-CN;
in certain embodiments, R1Is C1-6An alkyl group;
in some embodiments of the invention, the compounds of the invention are selected from:
Figure BSA0000220128980000035
Figure BSA0000220128980000041
or a pharmaceutically acceptable salt, solvate, polymorph or isomer thereof.
It is noted that the compounds and salts described in this specification can exist in solvated as well as unsolvated forms; the atoms of these compounds and salts described in this specification may exist as their isotopes; furthermore, the compounds and salts described in this specification may exist in optically active or racemic forms via one or more asymmetric carbon atoms.
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of formula (I) or a pharmaceutically acceptable salt, solvate, polymorph, or tautomer thereof. In some embodiments, the pharmaceutical compositions of the present invention further comprise a pharmaceutically acceptable excipient.
In another aspect, the present invention provides a method of treating a DNA-PK related disease, said method comprising administering to a subject an effective amount of a compound of formula (I) or a pharmaceutically acceptable salt, solvate, polymorph or isomer thereof, or a pharmaceutical composition thereof;
in another aspect, the present invention provides the use of a compound of the present invention, or a pharmaceutically acceptable salt, solvate, polymorph or tautomer thereof, or a pharmaceutical composition thereof, for the manufacture of a medicament for the treatment of a DNA-PK related disease.
In some embodiments of the invention, the DNA-PK related disease is cancer; preferably, the cancer is colorectal cancer, glioblastoma, gastric cancer, ovarian cancer, diffuse large B-cell lymphoma, chronic lymphocytic leukemia, acute myelogenous leukemia, head and neck squamous cell carcinoma, breast cancer, prostate cancer, bladder cancer, hepatocellular carcinoma, small cell lung cancer, or non-small cell lung cancer.
Detailed Description
Exemplary embodiments utilizing the principles of the present invention are set forth in the following detailed description of the invention. The features and advantages of the present invention may be better understood by reference to the following summary.
It should be understood that the scope of the various aspects of the invention is defined by the claims and that methods and structures within the scope of these claims and their equivalents are intended to be covered thereby.
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, explanatory and are not restrictive of any inventive subject matter. Unless otherwise indicated, the use of "or", "or" means "and/or" and, in addition, the use of the term "including" as well as other forms, such as "includes", "including", and "including", is not limiting.
Certain chemical terms
The terms "optionally," "optionally," or "optionally" mean that the subsequently described event or circumstance may or may not occur, and that the description includes instances where said event or circumstance occurs and instances where it does not. And, optionally substituted groups may be unsubstituted (e.g.: CH)2CH3) Fully substituted (e.g.: -CF2CF3) Monosubstituted (e.g.: -CH2CH2F) Or any level between mono-and fully substituted (e.g.: -CH2CHF2、-CF2CH3、-CFHCHF2Etc.). It will be appreciated by those skilled in the art that any group containing one or more substituents will not incorporate any substitution or substitution pattern which is sterically impossible and/or cannot be synthesized.
Unless otherwise indicated, conventional methods within the skill of the art are employed, such as mass spectrometry, nuclear magnetism, high performance liquid chromatography, infrared and ultraviolet/visible spectroscopy, and pharmacological methods unless a specific definition is set forth, the nomenclature and experimental procedures and techniques described herein relating to analytical chemistry, organic synthetic chemistry, and pharmaceutical and medicinal chemistry are known in the art. 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. In this specification, groups and substituents thereof may be selected by one of ordinary skill 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, -CH2O-is equivalent to-OCH2-.
As used herein, the terms "group", "chemical group" or "chemical group" refer to a particular portion or functional group of a molecule. Chemical groups are often considered as chemical entities embedded in or attached to a molecule.
Some of the chemical groups named herein may be referred to by a shorthand notation for the total number of carbon atoms. E.g. C1-6Alkyl describes an alkyl group, as defined below, having a total of 1 to 6 carbon atoms. The total number of carbon atoms indicated by shorthand notation does not include carbon atoms on possible substituents.
The terms "halogen", "halo" or "halide" refer to bromine, chlorine, fluorine or iodine.
The compounds of the invention may contain one or more (e.g. one, two, three or four) isotopic substitutions. For example, in the compounds, H may be in any isotopic form, including1H、2H (D or deuterium) and3h (T or tritium); c may be in any isotopic form, including12C、13C and14c; o may be in any isotopic form, including16O and18o, and the like.
As used herein, the terms "aromatic", "aromatic ring", "aromatic" and "aromatic-cyclic" refer to a planar ring portion of one or more rings having a delocalized electron-conjugated system of 4n +2 electrons, where n is an integer. The aromatic compound may be optionally substituted and may be monocyclic or fused-ring polycyclic. The term aromatic compound includes all carbocyclic rings (e.g., benzene rings) and rings containing one or more heteroatoms (e.g., pyridine).
The terms "heteroatom" or "hetero", as used herein alone or as part of another ingredient, refer to atoms other than carbon and hydrogen, heteroatoms are independently selected from, but not limited to, oxygen, nitrogen, sulfur, phosphorus, silicon, selenium, and tin.
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.
The term "alkyl" as used herein, alone or in combination, refers to an optionally substituted straight or optionally substituted branched chain monovalent saturated hydrocarbon having 1 to 12 carbon atoms, preferably 1 to 8 carbon atoms, more preferably 1 to 6 carbon atoms, attached to the rest of the molecule by a single bond, such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, n-hexyl, n-heptyl, 2-methylhexyl, 3 methylhexyl, n-octyl, n-nonyl, n-decyl, and the like.
The term "alkenyl" as used herein, alone or in combination, refers to an optionally substituted straight or optionally substituted branched chain monovalent hydrocarbon radical having one or more C ═ C double bonds and having from 2 to about 10 carbon atoms, more preferably from 2 to about 6 carbon atoms2) 1-propenyl (CH)2CH=CH2) Isopropenyl (C (CH)3)=CH2) Butenyl, 1, 3-butadienyl and the like. When a numerical range is present for alkenyl as defined herein, e.g. "C2-C6Alkenyl "or" C2-6The "alkenyl group" means an alkenyl group which may be composed of 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms or 6 carbon atoms, and the alkenyl group herein also covers the case where no numerical range is specified.
The term "alkynyl", as used herein, alone or in combination, refers to an optionally substituted straight or branched chain monovalent hydrocarbon radical having one or more C ≡ C triple bonds and having 2 to about 10 carbon atoms, more preferably 2 to about 6 carbon atoms. Examples include, but are not limited to, ethynyl, 2-propynyl, 2-butynyl, 1, 3-butadiynyl, and the like2-C6Alkynyl "or" C2-6Alkynyl "meansAlkynyl groups that can be composed of 2 carbon atoms, 3 carbon atoms, 4 carbon atoms, 5 carbon atoms, or 6 carbon atoms, and alkynyl groups herein also encompass instances where no numerical range is specified.
The term "aryl" refers to an all-carbon monocyclic or fused ring having a fully conjugated pi-electron system, having 6 to 14 carbon atoms, preferably 6 to 12 carbon atoms, most preferably 6 carbon atoms. Aryl groups may be unsubstituted or substituted with one or more substituents, examples of which include, but are not limited to, alkyl, alkyloxy, aryl, aralkyl, amino, halo, hydroxy, sulfonyl, sulfinyl, phosphoryl, and heteroalicyclic. Non-limiting examples of unsubstituted aryl groups include, but are not limited to, phenyl, naphthyl, and anthracenyl.
The term "arylene" as used herein, alone or in combination, refers to a divalent group derived from a monovalent aryl group as defined above.
The term "heteroaryl" refers to a monocyclic or fused ring of 5 to 12 ring atoms, having 5, 6, 7, 8, 9, 10, 11 or 12 ring atoms, containing 1, 2, 3 or 4 ring atoms selected from N, O, S, the remaining ring atoms being C, and having a fully conjugated pi-electron system. Heteroaryl groups may be unsubstituted or substituted, and the substituents include, but are not limited to, alkyl, alkyloxy, aryl, aralkyl, amino, halo, hydroxy, cyano, nitro, carbonyl, and heteroalicyclic. Non-limiting examples of unsubstituted heteroaryl groups include, but are not limited to, pyrrolyl, furanyl, thienyl, imidazolyl, oxazolyl, pyrazolyl, pyridyl, pyrimidinyl, pyrazinyl, quinolinyl, isoquinolinyl, tetrazolyl, triazinyl.
The term "heteroarylene" as used herein, alone or in combination, refers to a divalent radical derived from a monovalent heteroaryl group as defined above.
The term "heterocycle" refers to an aliphatic heterocycle. When the number of carbon atoms of the heterocyclic ring is indicated herein (e.g. C)3-6Heterocyclic ring), at least one non-carbon atom (heteroatom) necessarily being present in the ring. E.g. "C3-6The designation "heterocyclic" relates only to the number of carbon atoms in the ring and not to the total number of atoms in the ring. For example, the designation "4-to 6-membered heterocycle" refers to the ringThe total number of atoms contained in (i.e., a four, five or six membered ring in which at least one atom is a carbon atom, at least one atom is a heteroatom, and the remaining 2 to 4 atoms are carbon atoms or heteroatoms.) for a heterocyclic ring having two or more heteroatoms, the two or more heteroatoms may be the same as or different from each other. The heterocyclic ring may be optionally substituted. As used herein, a "heterocycle" preferably contains from about 5 to about 20 or 5 to 10 or 5-8 or 5-6 backbone ring-forming atoms.
The term "polymorph" or "polymorph" as used herein means that the compounds of the present invention have multiple lattice morphologies. 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.
Unless otherwise specified, the compounds of the present invention contain olefinic double bonds including E and Z isomers.
It is understood that the compounds of the present invention may contain asymmetric centers which may independently be in the R or S configuration. It will be apparent to those skilled in the art that some of the compounds of the present invention may also exhibit cis-trans isomerism. The present invention also relates to pharmaceutical compositions comprising a compound of the invention and a pharmaceutical composition comprising a compound of the invention, as well as to pharmaceutical compositions comprising a compound of the invention and a pharmaceutically acceptable carrier.
The term "pharmaceutically acceptable salts" as used herein includes both acid and base salts.
"pharmaceutically acceptable acid addition salts" refers to those salts formed with inorganic acids such as, but not limited to, acetic acid, 2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, capric acid, caproic acid, carbonic acid, cinnamic acid, citric acid, and the like, that retain the biological potency and properties of the free acid of the compound, are not biologically or otherwise undesirable. Salts formed by reaction with an inorganic base include, but are not limited to, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like.
Salt-forming organic bases include, but are not limited to, primary, secondary, tertiary, cyclic amines, and the like, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, diethanolamine, ethanolamine, dicyclohexylamine, ethylenediamine, purine, piperazine, piperidine, choline, caffeine, and the like. Particularly preferred organic bases are isopropylamine, diethylamine, ethanolamine, trimethylamine, dicyclohexylamine, choline, and caffeine.
Crystallization often produces solvates of the compounds of the present invention. The term "solvate" as used herein refers to a combination of one or more molecules of the 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. 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 present invention may be true solvates, but in other cases, the compounds of the present invention may also retain water only by chance or a mixture of water and 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.
The term "pharmaceutical composition" as used herein refers to a formulation mixed with a compound of the present invention and a vehicle generally accepted in the art for delivering biologically active compounds to a mammal, such as a human. Such media comprise all pharmaceutically acceptable carriers.
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, isotonic agents, solvents, or emulsifiers that have been approved by the relevant governmental authorities for use in humans and domestic animals.
The term "subject," "patient," "subject" or "individual" as used herein refers to an individual, including mammals and non-mammals, suffering from a disease, disorder or condition, among others. 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; examples of non-human mammals include, but are not limited to, birds and fish, among others. 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 or 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 previously been 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) alleviating the disease or condition, i.e., causing 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 term "effective amount," "therapeutically effective amount," or "pharmaceutically effective amount" as used herein refers to an amount of at least one agent or compound that is sufficient to alleviate, to some extent, one or more of the symptoms of the disease or disorder being treated, after administration. For example, an "effective amount" for treatment is the amount of a composition comprising a compound disclosed herein that is required to provide clinically significant relief from a condition.
The terms "administration," "administering," "administration," and the like, as used herein, refer to methods capable of delivering a compound or composition to a desired site for biological action. In preferred embodiments, the compounds and compositions discussed herein are administered orally.
The anti-cancer treatments described herein may be useful as monotherapy or may include conventional surgery, radiotherapy or chemotherapy in addition to the administration of a compound having formula (I); such conventional surgery, radiation therapy or chemotherapy may be administered simultaneously, sequentially or separately with a compound of formula (I) for treatment.
Preparation of the Compounds of the invention
The following reaction scheme illustrates the process for preparing the compounds of the present invention.
It will be appreciated that in the following description, combinations of substituents and/or variables of the formula are permitted only in the context of forming stable compounds.
It will also be appreciated by those skilled in the art that in the schemes described below, the functional groups of the intermediate compounds may need to be protected by suitable protecting groups. Suitable hydroxyl protecting groups include trialkylsilyl or diarylalkylsilyl groups (e.g., tert-butylmethylsilyl, tert-butyldiphenylsilyl, or trimethylsilyl), tetrahydropyranyl, benzyl, and the like. Suitable amino, amidino and guanidine protecting groups include t-butyloxycarbonyl, benzyloxycarbonyl and the like suitable protecting groups for mercapto include-C (O) -R '(R' represents alkyl, aryl or arylalkyl), p-methoxybenzyl, trityl and the like. Suitable carboxyl protecting groups include alkyl, aryl or arylalkyl esters. Protecting groups may be added or removed by standard techniques known to those skilled in the art.
Examples
The following non-limiting examples are illustrative only and do not limit the invention in any way.
Reagents are commercially available from national pharmaceutical group chemical reagents, Beijing, Inc., Afa Aesar, or Beijing Bailingwei technologies Inc., and these reagents can be used directly without further purification unless otherwise indicated.
Unless otherwise stated, the following reactions are carried out at room temperature, in anhydrous solvents, under positive pressure of nitrogen or argon, or using a drying tube; the reaction bottle is provided with a rubber diaphragm so as to add the substrate and the reagent through an injector; glassware was dried and/or heat dried.
Unless otherwise stated, column chromatography purification was performed using 200-300 mesh silica gel from Qingdao oceanic plants; preparation of thin-layer chromatography silica gel precast slab (HSGF254) produced by Nicoti chemical industry research institute was used; MS is measured by a Thermo LCQ fly model (ESI) liquid chromatography-mass spectrometer; the optical rotation was measured by using an SGW-3 automatic polarimeter, Shanghai Spanish Meter, Ltd.
Nuclear magnetic data (1H NMR) was run at 400MHz using a Varian instrument. The solvent used for nuclear magnetic data is CDCl3、CD3OD、D2O, DMSO-d6, based on tetramethylsilane (0.00ppm) or based on residual solvent (CDCl)3:7.26ppm;CD3OD:3.31ppm;D2O: 4.79 ppm; d 6-DMSO: 2.50 ppm). When indicating the diversity of the peak shapes, the following abbreviations represent the different peak shapes: s (singlet), d (doublet), t (triplet), q (quartet), m (multiplet), br (broad), dd (doublet of doublets), dt (doublet of triplets). If the coupling constant is given, it is given in Hertz (Hz).
Abbreviations:
Figure BSA0000220128980000091
intermediate 1: 4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000101
Step 1: 4- ((2-chloro-5-nitropyrimidin-4-yl) amino) tetrahydro-2H-pyran-4-carbonitrile
2, 4-dichloro-5-nitropyrimidine (1.94g) and triethylamine (1.01g) were dissolved in tetrahydrofuran (50mL), and 4-aminotetrahydro-2H-pyran-4-carbonitrile (1.26g) was slowly added under ice bath. The reaction was warmed to room temperature and stirred overnight. The reaction solution was poured into saturated aqueous ammonium chloride (200mL), extracted with ethyl acetate (100mL), the extracts were concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether: ethyl acetate 3: 1 (V: V)) to give a white solid (1.9 g).
Step 2: 4- ((5-amino-2-chloropyrimidin-4-yl) amino) tetrahydro-2H-pyran-4-carbonitrile
Reduced iron powder (1.5g) was added to a solution of 4- ((2-chloro-5-nitropyrimidin-4-yl) amino) tetrahydro-2H-pyran-4-carbonitrile (1.9g) in acetic acid (30mL) at room temperature, then the reaction was raised to 45 ℃ and stirred for 2 hours, cooled to room temperature, poured into ice water, and extracted with ethyl acetate (100 mL); the extract was washed with saturated brine, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol ═ 20: 1 (V: V)) to give a white solid (1.3 g).
And step 3: 4- (2-chloro-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
The 4- ((5-amino-2-chloropyrimidin-4-yl) amino) tetrahydro-2H-pyran-4-carbonitrile (1.3g) and CDI (1.3g) in step 2 were dissolved in tetrahydrofuran (50mL), heated to 65 ℃ under nitrogen protection and stirred for 2 hours, cooled to room temperature, the reaction mixture was poured into water (150mL), the reaction mixture was extracted with dichloromethane, the extract was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: petroleum ether: ethyl acetate ═ 3: 1 (V: V)) to give a pale yellow solid (1g).
And 4, step 4: 4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
60% (mineral oil) of sodium hydride (200mg) was slowly added to a DMF (20mL) solution of 4- (2-chloro-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (1g) obtained in step 3 at 0 ℃ and after the addition was completed, stirring was continued at 0 ℃ for 0.5 hour, iodomethane (1.41g) was then slowly added to the reaction solution, and after the reaction was completed, the reaction solution was poured into a saturated aqueous ammonium chloride solution and stirred continuously, a yellow precipitate was generated, and was filtered and dried to obtain a yellow solid (900 mg).
Intermediate 2: 2-chloro-7-methyl-9- (tetrahydro-2H-pyran-4-yl) -7, 9-dihydro-8H-purin-8-one (synthesized according to the method described in patent CN110177791A 1)
Figure BSA0000220128980000102
Intermediate 4: 7-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-6-amine (synthesized according to the procedure described in patent CN110177791A 1)
Figure BSA0000220128980000111
Intermediate 6: 2-chloro-7-methyl-9- (piperidin-4-yl) -7, 9-dihydro-8H-purin-8-one (synthesized according to the method described in patent WO2019/238929A 1)
Figure BSA0000220128980000112
Example 29: 2- (2- ((7-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-6-yl) amino) -8-oxo-9- (tetrahydro-2H-pyran-4-yl) -8, 9-dihydro-7H-purin-7-yl) acetonitrile
Figure BSA0000220128980000113
Step 1: 2- (2-chloro-8-oxo-9- (tetrahydro-2H-pyran-4-yl) -8, 9-dihydro-7H-purin-7-yl) acetonitrile
To a solution of 2-chloro-9- (tetrahydro-2H-pyran-4-yl) -7, 9-dihydro-8H-purin-8-one (254mg) in DMF (10mL) at 0 deg.C was slowly added 60% (mineral oil) sodium hydride (80mg) and stirring continued at 0 deg.C for 0.5H. Then, bromoacetonitrile (150mg) is slowly added into the reaction solution, and after the reaction is finished, the reaction solution is poured into saturated ammonium chloride aqueous solution and extracted by ethyl acetate; the extract was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the title compound (77 mg).
Step 2: 2- (2- ((7-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-6-yl) amino) -8-oxo-9- (tetrahydro-2H-pyran-4-yl) -8, 9-dihydro-7H-purin-7-yl) acetonitrile
The above-described 2- (2-chloro-8-oxo-9- (tetrahydro-2H-pyran-4-yl) -8, 9-dihydro-7H-purin-7-yl) acetonitrile (58mg), intermediate 47-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-6-amine (30mg), RuPhos Pd G3(20mg) and cesium carbonate (160mg) were dissolved in dioxane (10mL) under nitrogen, and the reaction flask was replaced with nitrogen three times and heated to 100 ℃ for 3 hours with stirring. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (45 mg).
1H NMR(400MHz,DMSO-d6)δ9.05(s,1H),8.82(s,1H),8.36(s,1H),8.20(s,1H),7.69(s,1H),5.06(s,2H),4.34-4.43(m,1H),3.92(dd,J=11.6Hz,4.0Hz,2H),3.37(t,J=11.6Hz,2H),2.40-2.50(m,2H),2.35(s,3H),1.64-1.72(m,2H)。
Example 30: 7-methyl-2- ((7-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-6-yl) amino) -9- (1-oxidotrih-2H-thiophen-4-yl) -7, 9-dihydro-8H-purin-8-one
Figure BSA0000220128980000121
2-chloro-7-methyl-9- (1-oxotetrahydro-2H-thiopyran-4-yl) -7, 9-dihydro-8H-purin-8-one (obtained following the synthesis of intermediate 1 using 4-aminotetrahydro-2H-thiopyran 1-oxide as the starting material) (60mg), intermediate 47-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-6-amine (30mg), RuPhos Pd G3(9mg) and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 15: 1 (V: V)) to give the objective compound (43mg).
1H NMR(400MHz,DMSO-d6)δ9.03(s,0.5H),9.01(s,0.5H),8.65(s,0.5H),8.60(s,0.5H),8.35(s,0.5H),8.33(s,0.5H),8.05(s,0.5H),8.03(s,0.5H),7.68(s,0.5H),7.65(s,0.5H),4.32-4.47(m,1H),3.30-3.36(m,1H),3.26(s,1.5H),3.25(s,1.5H),2.94-3.14(m,2H),2.76-2.86(m,2H),2.48-2.60(m,1H),2.34(s,3H),1.95-2.04(m,1H),1.69-1.77(m,1H)。
Example 31: 4- (7-methyl-2- ((7-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) piperidine-1-carbonitrile
Figure BSA0000220128980000122
Intermediate 62-chloro-7-methyl-9- (piperidin-4-yl) -7, 9-dihydro-8H-purin-8-one (268mg), cyanogen bromide (110mg) were dissolved in THF (20mL) at room temperature, triethylamine (200mg) was slowly added and stirring continued for 2 hours. The reaction solution was poured into water and extracted with ethyl acetate, and the organic phase was washed with a saturated aqueous sodium chloride solution, dried over anhydrous sodium sulfate, and concentrated. The residue was purified by column chromatography on silica gel (eluent: petroleum ether: ethyl acetate 1: 1 (V: V)) to give 4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) piperidine-1-carbonitrile (120 mg). The above 4- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) piperidine-1-carbonitrile (59mg), intermediate 47-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-6-amine (30mg), RuPhos Pd G3(9mg) and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (36mg).
1H NMR(400MHz,DMSO-d6)δ9.04(s,1H),8.67(s,1H),8.33(s,1H),8.05(s,1H),7.67(s,1H),4.22-4.35(m,1H),3.42-3.50(m,2H),3.26(s,3H),3.10-3.22(m,2H),2.44-2.56(m,2H),2.35(s,3H),1.69-1.79(m,2H)。
Example 32: 27,42-dimethyl-28,29-dihydro-27H-5-oxa-3-aza-2 (9, 2) -purine-1 (4, 1) -piperidine-4 (1, 4) -phencynonane-28-ketones
Figure BSA0000220128980000131
This compound was synthesized according to the synthesis method of example 7 using alternative phenols as starting materials.
1H NMR(400MHz,DMSO-d6)δ8.46(s,1H),7.98(s,1H),6.96(d,J=8.4Hz,1H),6.85(s,1H),6.78(d,J=7.6Hz,1H),4.04-4.20(m,2H),3.77-4.03(m,1H),3.22(s,3H),2.52-2.79(m,2H),2.04-2.30(m,4H),2.01(s,3H),1.58-1.78(m,4H),1.28-1.56(m,4H)。
Example 33: 9- (1, 3-Dimethoxypropan-2-yl) -7-methyl-2- ((7-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-6-yl) amino) -7, 9-dihydro-8H-purin-8-one
Figure BSA0000220128980000132
2-chloro-9- (1, 3-dimethoxypropan-2-yl) -7-methyl-7, 9-dihydro-8H-purin-8-one (obtained following the synthesis of intermediate 1) (58mg), intermediate 47-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-6-amine (30mg), RuPhos Pd G3(9mg) and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 15: 1 (V: V)) to give the objective compound (43mg).
1H NMR(400MHz,DMSO-d6)δ9.08(s,1H),8.63(s,1H),8.34(s,1H),8.08(s,1H),7.68(s,1H),4.63-4.70(m,1H),3.87(t,J=9.6Hz,2H),3.56(dd,J=10.4Hz,5.6Hz,2H),3.28(s,3H),3.16(s,6H),2.35(s,3H)。
Example 34: 7-methyl-2- ((3-methyl-3H-imidazo [4, 5-c ] pyridin-2-yl) amino) -9- (tetrahydro-2H-pyran-4-yl) -7, 9-dihydro-8H-purin-8-one
Figure BSA0000220128980000133
Under the protection of nitrogen, the intermediate 2: 2-chloro-7-methyl-9- (tetrahydro-2H-pyran-4-yl) -7, 9-dihydro-8H-purin-8-one (54mg), 3-methyl-3H-imidazo [4, 5-c ] pyridin-2-amine (30mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL), heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 15: 1 (V: V)) to give the objective compound (13mg).
1H NMR(400MHz,DMSO-d6)δ8.50-8.66(m,1H),8.23(d,J=4.8Hz,1H),8.13(s,1H),7.37(d,J=4.8Hz,1H),4.37-4.52(m,1H),3.89-4.02(m,2H),3.60(s,3H),3.37-3.52(m,5H),2.47-2.59(m,2H),1.57-1.70(m,2H)。
Example 35: 7-cyclopropyl-2- ((7-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-6-yl) amino) -9- (tetrahydro-2H-pyran-4-yl) -7, 9-dihydro-8H-purin-8-one
Figure BSA0000220128980000141
Step 1: 2-chloro-7-cyclopropyl-9- (tetrahydro-2H-pyran-4-yl) -7, 9-dihydro-8H-purin-8-one
To a solution of 2-chloro-9- (tetrahydro-2H-pyran-4-yl) -7, 9-dihydro-8H-purin-8-one (254mg) in 1, 2-dichloroethane (10mL) was added cyclopropylboronic acid (100mg), copper acetate (20mg) and pyridine (160mg) in that order at room temperature, and after the addition was complete, stirring was continued for 12 hours with open mouth. After the reaction is finished, pouring the reaction solution into a saturated ammonium chloride aqueous solution, and extracting with ethyl acetate; the extract was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (eluent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the title compound (150 mg).
Step 2: 7-cyclopropyl-2- ((7-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-6-yl) amino) -9- (tetrahydro-2H-pyran-4-yl) -7, 9-dihydro-8H-purin-8-one
The above-mentioned 2-chloro-7-cyclopropyl-9- (tetrahydro-2H-pyran-4-yl) -7, 9-dihydro-8H-purin-8-one (60mg), intermediate 47-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-6-amine (30mg), RuPhos Pd G3(9mg) and cesium carbonate (160mg) were dissolved in dioxane (10mL) under nitrogen, the reaction flask was replaced with nitrogen three times and heated to 100 ℃ with stirring for 2 hours, cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the resultant residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol 20: 1 (V: V)) to obtain the objective compound (45 mg).
1H NMR(400MHz,DMSO-d6)δ9.05(s,1H),8.66(s,1H),8.33(s,1H),8.02(s,1H),7.67(s,1H),4.28-4.40(m,1H),3.86-3.97(m,2H),3.34-3.42(m,2H),2.83-2.91(m,1H),2.39-2.52(m,2H),2.35(s,3H),1.56-1.68(m,2H),0.80-0.99(m,4H)。
Example 36: 2- ((1, 4-dimethyl-6-oxo-1, 6-dihydropyridin-3-yl) amino) -7-methyl-9- (tetrahydro-2H-pyran-4-yl) -7, 9-dihydro-8H-purin-8-one
Figure BSA0000220128980000142
Under the protection of nitrogen, the intermediate 2: 2-chloro-7-methyl-9- (tetrahydro-2H-pyran-4-yl) -7, 9-dihydro-8H-purin-8-one (54mg), 5-amino-1, 4-dimethylpyridin-2 (1H) -one (28mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL), heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol 15: 1 (V: V)) to give the objective compound (15 mg).
1H NMR(400MHz,DMSO-d6)δ8.21(s,1H),7.94(s,1H),7.62(s,1H),6.23(s,1H),4.29-4.38(m,1H),3.92(dd,J=11.6Hz,4.4Hz,2H),3.37(t,J=11.6Hz,2H),3.35(s,3H),3.23(s,3H),2.41-2.51(m,2H),1.98(s,3H),1.60(dd,J=12.4Hz,2.4Hz,2H)。
Example 37: 13, 20-dimethyl-1, 5, 10, 15, 17, 20, 23-heptaazapentacyclic [14.5.2.2 ]2,5.110, 14.019,22]Six carbon-12, 14(24), 16, 18, 22-pentene-11, 21-dione
Figure BSA0000220128980000151
This compound was synthesized according to the synthesis procedure of example 7 using the alternative nitropyridinones as starting materials.
1H NMR(400MHz,DMSO-d6)δ8.49(s,1H),8.16(s,1H),7.86(s,1H),6.24(s,1H),4.21-4.30(m,1H),3.80-3.86(m,2H),3.26-3.32(m,5H),2.82-2.88(m,2H),2.62-2.73(m,2H),2.17-2.28(m,5H),1.49-1.69(m,6H)。
Example 38: 4- (7-methyl-2- ((6-methylimidazo [1, 2-a ] pyridin-7-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000152
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 6-methylimidazo [1, 2-a ] pyridin-7-amine (30mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (41 mg).
1H NMR(400MHz,DMSO)δ8.59-8.71(m,2H),8.14(s,1H),7.76-7.92(m,1H),7.36-7.55(m,1H),6.68-6.82(m,1H),3.81-3.92(m,2H),3.46-3.60(m,2H),3.27(s,3H),2.65-2.78(m,2H),2.52-2.61(m,2H),2.24(s,3H)。
Example 39: 4- (2- ((4-chloro-7-methylquinolin-6-yl) amino) -7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000153
The intermediates 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 4-chloro-7-methylquinolin-6-amine (synthesized according to the method described in patent WO2019/238929a 1) (38mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (31mg).
1H NMR(400MHz,DMSO)δ8.80(s,1H),8.63(d,J=4.4Hz,1H),8.43(s,1H),8.27(s,1H),7.91(s,1H),7.60(d,J=4.4Hz,1H),3.84-3.92(m,2H),3.51-3.60(m,2H[),3.31(s,3H),2.62-2.80(m,4H),2.50(s,3H)。
Example 40: 4- (2- ((4-methoxy-7-methylquinolin-6-yl) amino) -7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000161
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 4-methoxy-7-methylquinolin-6-amine (synthesized according to the method described in patent WO2019/238929a 1) (38mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL), heated to 100 ℃ and stirred for 2 hours, cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the title compound (45 mg).
1H NMR(400MHz,DMSO)δ8.67(s,1H),8.55(d,J=4.8Hz,1H),8.34(s,1H),8.23(s,1H),7.74(s,1H),6.88(d,J=5.2Hz,1H),3.97(s,3H),3.81-3.88(m,2H),3.52-3.60(m,2H),3.30(s,3H),2.62-2.77(m,4H),2.44(s,3H).
Example 41: 4- (7-methyl-2- ((7-methyl- [1, 2, 4] triazolo [4, 3-a ] pyridin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000162
Step 1: 7-methyl- [1, 2, 4] triazolo [4, 3-a ] pyridin-6-amine
6-bromo-7-methyl- [1, 2, 4] triazolo [4, 3-a ] pyridine (212mg), RuPhos Pd G3(84mg), benzophenone imine (182mg) and cesium carbonate (650mg) were dissolved in toluene (15mL) under nitrogen, heated to 100 ℃ and stirred for 12 hours, cooled to room temperature, filtered, the solid was washed with dichloromethane, the filtrate was concentrated under reduced pressure, the residue was dissolved in tetrahydrofuran (30mL) and 1M dilute hydrochloric acid (10mL) was slowly added, after stirring for 1 hour, a saturated aqueous sodium bicarbonate solution was added to the system until the pH was neutral, then ethyl acetate was added for extraction, the residue was purified by column chromatography (eluent: dichloromethane: methanol ═ 20: 1 (V: V)) after the organic phase was evaporated to dryness to obtain 7-methyl- [1, 2, 4] triazolo [ 4], 3-a ] pyridin-6-amine (103 mg).
Step 2: 4- (7-methyl-2- ((7-methyl- [1, 2, 4] triazolo [4, 3-a ] pyridin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 7-methyl- [1, 2, 4] triazolo [4, 3-a ] pyridin-6-amine (30mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 10: 1 (V: V)) to give the objective compound (44mg).
1H NMR(400MHz,DMSO)δ9.07(s,1H),8.77(s,1H),8.35(s,1H),8.18(s,1H),7.68(s,1H),3.87-3.94(m,2H),3.52-3.60(m,2H),3.28(s,3H),2.68-2.76(m,2H),2.58-2.64(m,2H),2.35(s,3H)。
Example 42: 44-chloro-6, 6-difluoro-27,46-dimethyl-28,29-dihydro-27H-5-oxa-3-aza-2 (9, 2) -purine-1 (4, 1) -piperidine-4 (1, 3) -benzocycloheptane-287-diketones
Figure BSA0000220128980000171
This compound was synthesized according to the synthesis of example 7 using the alternative phenols and ethyl difluorobromoacetate as starting materials.
1H NMR(400MHz,DMSO-d6)δ8.34(s,1H),8.20(s,1H),7.96-7.98(m,1H),7.33(s,1H),4.68-4.77(m,1H),4.11-4.22(m,2H),3.41-3.48(m,1H),3.30(s,3H),2.96-3.04(m,1H),2.50-2.68(m,1H),2.46-2.53(m,1H),2.26(s,3H),1.94-2.02(m,1H),1.68-1.75(m,1H)。
Example 43: 4- (7-methyl-2- ((2-methyl-1, 8-naphthyridin-3-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000172
Step 1: 2-methyl-1, 8-naphthyridin-3-amine
2-methyl-1, 8-naphthyridine-3-carboxylic acid (188mg) and DPPA (300mg) were dissolved in toluene (20mL), and triethylamine (0.5mL) was added slowly at room temperature and stirred for 1 hour. To the system was added 0.5mL of water, followed by heating to 80 ℃ under nitrogen atmosphere and stirring for 2 hours, cooling to room temperature, pouring the reaction solution into water (150mL), extracting the reaction solution with ethyl acetate, washing the extract with saturated brine, drying over anhydrous sodium sulfate, filtering, concentrating the filtrate under reduced pressure, and purifying the residue by silica gel column chromatography (eluent: petroleum ether: ethyl acetate ═ 2: 1 (V: V)) to obtain a pale yellow solid (60 mg).
Step 2: 4- (7-methyl-2- ((2-methyl-1, 8-naphthyridin-3-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 2-methyl-1, 8-naphthyridin-3-amine (32mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (41 mg).
1H NMR(400MHz,DMSO)δ8.99(s,1H),8.86(dd,J=4.4Hz,2.0Hz,1H),8.64(s,1H),8.25-8.27(m,2H),7.50(dd,J=8-7.6Hz,4.4Hz,1H),3.86-3.93(m,2H),3.53-3.61(m,2H),3.31(s,3H),2.68-2.78(m,5H),2.58-2.65(m,2H).
Example 44: 4- (7-methyl-2- ((7-methylquinazolin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000181
The intermediates 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 7-methyl quinazolin-6-amine (synthesized according to the method described in patent WO2019/238929a 1) (32mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (39 mg).
1H NMR(400MHz,DMSO)δ9.36(s,1H),9.09(s,1H),8.84(s,1H),8.45(s,1H),8.28(s,1H),7.83(s,1H),3.88-3.94(m,2H),3.54-3.62(m,2H),3.31(s,3H),2.74-2.82(m,2H),2.58-2.64(m,2H),2.53(s,3H)。
Example 45: 2- ((7-chloro- [1, 2, 4] triazolo [1, 5-a ] pyridin-6-yl) amino) -7-methyl-9- (tetrahydro-2H-pyran-4-yl) -7, 9-dihydro-8H-purin-8-one
Figure BSA0000220128980000182
The intermediates 22-chloro-7-methyl-9- (tetrahydro-2H-pyran-4-yl) -7, 9-dihydro-8H-purin-8-one (54mg), 7-chloro- [1, 2, 4] triazolo [1, 5-a ] pyridin-6-amine (synthesized following the procedure described for intermediate 4 in patent WO2019/238929a 1) (34mg), RuPhos Pd G3(9mg) and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (18 mg).
1H NMR(400MHz,DMSO)δ10.19(s,1H),8.29(s,1H),7.95(s,1H),7.85(s,1H),7.37(s,1H),4.52-4.61(m,1H),4.15(dd,J=11.6Hz,4.4Hz,2H),3.51-3.59(m,2H),3.43(s,3H),2.72-2.82(m,2H),1.71-1.77(m,2H)。
Example 46: 4- (2- ((7-chloro- [1, 2, 4] triazolo [1, 5-a ] pyridin-6-yl) amino) -7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000183
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 7-chloro- [1, 2, 4] triazolo [1, 5-a ] pyridin-6-amine (34mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours, cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (39 mg).
1H NMR(400MHz,DMSO)δ10.04(s,1H),8.30(s,1H),8.03(s,1H),7.86(s,1H),7.37(s,1H),4.10-4.17(m,2H),3.89-3.95(m,2H),3.43(s,3H),2.77-2.87(m,4H)。
Example 47: 4- (7-methyl-2- ((7-methylbenzothiazol-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000191
Step 1: 7-methylbenzothiazol-6-amine
6-bromo-7-methylbenzo [ d ] thiazole (228mg), RuPhos Pd G3(80mg), benzophenone imine (182mg) and cesium carbonate (650mg) were dissolved in toluene (10mL) under nitrogen, heated to 100 ℃ and stirred for 5 hours, cooled to room temperature, filtered, the solid washed with dichloromethane then the filtrate was concentrated under reduced pressure, the residue was dissolved in tetrahydrofuran (30mL) and 1M dilute hydrochloric acid (10mL) was slowly added, stirring was continued for 1 hour and then saturated aqueous sodium bicarbonate was added to the system until the pH was neutral. Extraction was performed with ethyl acetate and the organic phase was evaporated to dryness and the residue was purified by column chromatography (developing solvent: dichloromethane: methanol 20: 1 (V: V)) to give 7-methylbenzothiazol-6-amine (73 mg).
Step 2: 4- (7-methyl-2- ((7-methylbenzothiazol-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 7-methylbenzothiazol-6-amine (33mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (37mg).
1H NMR(400MHz,DMSO)δ9.25(s,1H),8.93(s,1H),8.12(s,1H),7.84(d,J=8.4Hz,1H),7.61(d,J=8.4Hz,1H),3.82-3.92(m,2H),3.46-3.60(m,2H),3.33(s,3H),2.65-2.76(m,2H),2.50-2.59(m,2H),2.41(s,3H)。
Example 48: 4- (7-methyl-2- ((6-methylbenzo [ d ] [1, 3] dioxin-5-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000192
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 6-methylbenzo [ d ] [1, 3] dioxan-5-amine (30mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (45 mg).
1H NMR(400MHz,DMSO)δ8.40(s,1H),8.07(s,1H),6.95(s,1H),6.74(s,1H),5.92(s,2H),3.86-3.92(m,2H),3.53-3.61(m,2H),3.25(s,3H),2.68-2.77(m,2H),2.54-2.61(m,2H),2.07(s,3H)。
Example 49: 4- (7-methyl-2- ((7-methylbenzothiazol-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000201
Step 1: 7-methylbenzothiazol-6-amine
6-bromo-7-methylbenzo [ d ] thiazole (228mg), RuPhos Pd G3(80mg), benzophenone imine (182mg) and cesium carbonate (650mg) were dissolved in toluene (10mL) under nitrogen, heated to 100 ℃ and stirred for 5 hours, cooled to room temperature, filtered, the solid washed with dichloromethane then the filtrate was concentrated under reduced pressure, the residue was dissolved in tetrahydrofuran (30mL) and 1M dilute hydrochloric acid (10mL) was slowly added, stirring was continued for 1 hour and then saturated aqueous sodium bicarbonate was added to the system until the pH was neutral. Extraction was performed with ethyl acetate, and the organic phase was evaporated to dryness and the residue was purified by column chromatography (developing solvent: dichloromethane: methanol 20: 1 (V: V)) to give 5-methylbenzothiazol-6-amine (40 mg).
Step 2: 4- (7-methyl-2- ((7-methylbenzothiazol-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 7-methylbenzothiazol-6-amine (33mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (31mg).
1H NMR(400MHz,DMSO)δ9.25(s,1H),8.93(s,1H),8.12(s,1H),7.84(d,J=8.4Hz,1H),7.61(d,J=8.4Hz,1H),3.82-3.92(m,2H),3.46-3.60(m,2H),3.33(s,3H),2.65-2.76(m,2H),2.50-2.59(m,2H),2.41(s,3H)。
Example 50: 4- (7-methyl-2- ((6-methylpyrazolo [1, 5-a ] pyrimidin-5-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000202
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 6-methylpyrazol [1, 5-a ] pyrimidin-5-amine (32mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (22 mg).
1H NMR(400MHz,DMSO)δ9.39(s,1H),8.75(s,1H),8.33(s,1H),7.90(d,J=2.0Hz,1H),6.15(d,J=2.0Hz,1H),3.89-3.95(m,2H),3.54-3.62(m,2H),3.33(s,3H),2.82-2.91(m,2H),2.61-2.67(m,2H),2.34(s,3H)。
Example 51: 4- (7-methyl-2- ((3-methyl-1, 5-naphthyridin-2-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000211
Step 1: 3-methyl-1, 5-naphthyridin-2-amine
Under the protection of nitrogen, 3-bromo-1, 5-naphthyridin-2-amine (224mg) and PdCl are added2(dppf) (90mg), trimethylcyclotriboroxane (250mg) and potassium carbonate (278mg) were dissolved in dioxane (20mL) and water (5mL), heated to 100 ℃ and stirred for 2 hours. After cooling to room temperature, the reaction mixture was poured into water (100mL) and extracted with ethyl acetate, and the organic phase was evaporated to dryness, and the residue was purified by column chromatography (eluent: dichloromethane: methanol ═ 20: 1 (V: V)) to give 3-methyl-1, 5-naphthyridin-2-amine (90 mg).
Step 2: 4- (7-methyl-2- ((3-methyl-1, 5-naphthyridin-2-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 3-methyl-1, 5-naphthyridin-2-amine (32mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (10mL) under nitrogen, heated to 100 ℃ and stirred for 3 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (49 mg).
1H NMR(400MHz,DMSO)δ9.42(s,1H),8.73(d,J=3.2Hz,1H),8.30(s,1H),8.11(s,1H),8.01(d,J=8.8Hz,1H),7.58(dd,J=8.8Hz,4.0Hz,1H),3.82-3.89(m,2H),349-3.57(m,2H),3.32(s,3H),2.79-2.88(m,2H),2.58-2.65(m,2H),2.45(s,3H)。
Example 52: 4- (7-methyl-2- ((7-methylpyridine [2, 3-b ] pyrazin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000212
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 7-methylpyridine [2, 3-b ] pyrazin-6-amine (32mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (14 mg).
1H NMR(400MHz,DMSO)δ9.64(s,1H),8.85(s,1H),8.72(s,1H),8.36(s,1H),8.18(s,1H),3.86-3.96(m,2H),3.50-3.61(m,2H),3.34(s,3H),2.70-2.90(m,4H),2.47(s,3H)。
Example 53: 4- (2- ((4, 7-dimethylquinolin-6-yl) amino) -7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000221
Step 1: 4, 7-dimethylquinolin-6-amines
Under the protection of nitrogen, 4-chloro-7-methylquinolin-6-amine (192mg), PdCl2(dppf) (90mg), trimethylcyclotriboroxane (250mg) and potassium carbonate (278mg) were dissolved in dioxane (20mL) and water (5mL), heated to 100 ℃ and stirred for 4 hours, cooled to room temperature, the reaction solution was poured into water (100mL), ethyl acetate was added and extracted, the organic phase was dried and evaporated to dryness, and the residue was purified by column chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give 3-methyl-1, 5-naphthyridin-2-amine (100mg).
Step 2: 4- (2- ((4, 7-dimethylquinolin-6-yl) amino) -7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 4, 7-dimethylquinolin-6-amine (34mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (10mL) under nitrogen, heated to 100 ℃ and stirred for 3 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (37mg).
1H NMR(400MHz,DMSO)δ8.74(s,1H),8.57(d,J=4.4Hz,1H),8.20(s,1H),8.16(s,1H),7.81(s,1H),7.23(d,J=4.4Hz,1H),3.81-3.88(m,2H),3.50-3.57(m,2H),3.29(s,3H),2.70-2.79(m,2H),2.54-2.61(m,5H),2.43(s,3H)。
Example 54: 27,47-dimethyl-28,29-dihydro-27H-5-oxa-3-aza-4 (6, 4) -quinoline-2 (9, 2) -purine-1 (4, 1) -piperidinepepane-28-ketones
Figure BSA0000220128980000222
This compound was synthesized according to the synthesis method of example 7 using 7-methyl-6-nitroquinolin-4-ol as the starting material.
1H NMR(400MHz,DMSO)δ8.96(s,1H),8.49(d,J=4.8Hz,1H),8.20(s,1H),8.05(s,1H),7.71(s,1H),6.93(d,J=5.2Hz,1H),4.31-4.40(m,2H),4.18-4.30(m,1H),3.32(s,3H),2.90-3.03(m,4H),2.76-2.89(m,2H),2.52(s,3H),2.38-2.50(m,2H),1.56-1.66(m,2H)。
Example 55: 44-fluoro-27,46-dimethyl-28,29-dihydro-27H-5-oxa-3-aza-2 (9, 2) -purine-1 (4, 1) -piperidine-4 (1, 3) -benzocycloheptane-28-ketones
Figure BSA0000220128980000231
This compound was synthesized according to the synthesis method of example 7 using alternative phenols as starting materials.
1H NMR(400MHz,DMSO)δ8.25(s,1H),8.17(s,1H),8.03(d,J=8.0Hz,1H),7.02(d,J=12.4Hz,1H),4.26-4.51(m,3H),3.31(s,3H),3.12-3.30(m,2H),2.81-3.11(m,4H),2.67-2.81(m,2H),2.21(s,3H),1.49-1.69(m,2H)。
Example 56: 4- (2- ((4-methoxy-7-methylquinazolin-6-yl) amino) -7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000232
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 4-methoxy-7-methyl-quinazolin-6-amine (38mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours, cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (48 mg).
1H NMR(400MHz,DMSO)δ8.76(s,1H),8.63(s,1H),8.42(s,1H),8.27(s,1H),7.74(s,1H),4.07(s,3H),3.84-3.92(m,2H),3.54-3.61(m,2H),3.31(s,3H),2.62-2.79(m,4H),2.48(s,3H).
Example 57: 4- (7-methyl-2- ((6-methyl- [1, 2, 5] thiadiazole [3, 4-b ] pyridin-5-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000233
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 6-methyl- [1, 2, 5] thiadiazolo [3, 4-b ] pyridin-5-amine (33mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (17 mg).
1H NMR(400MHz,DMSO)δ9.62(s,1H),8.41(s,1H),8.16(s,1H),3.90-3.98(m,2H),3.56-3.63(m,2H),3.35(s,3H),2.85-2.94(m,2H),2.68-2.76(m,2H),2.47(s,3H)。
Example 58: 44-fluoro-27,46-dimethyl-28,29-dihydro-27H-3, 5-diaza-2 (9, 2) -purine-1 (4, 1) -piperidine-4 (1, 3) -benzocycloheptane-28-ketones
Figure BSA0000220128980000241
This compound was synthesized by following the synthesis procedure of example 7 using 2-fluoro-4-methyl-5-nitroaniline as the starting material.
1H NMR(400MHz,DMSO)δ8.19(s,1H),8.02(s,1H),7.56(d,J=8.4Hz,1H),6.86(d,J=12.0Hz,1H),5.63-5.70(br,1H),4.52-4.65(m,1H),3.20-3.53(m,11H),2.72-2.86(m,2H),2.16(s,3H),1.74-1.86(m,2H)。
Example 59: 27,45-dimethyl-28,29-dihydro-27H-5-oxa-3-aza-2 (9, 2) -purine-4 (4, 2) -pyrimidine-1 (4, 1) -piperidinepepane-28-ketones
Figure BSA0000220128980000242
This compound was synthesized following the synthesis procedure of example 7 using 5-methylcytosine as starting material.
1H NMR(400MHz,DMSO)δ8.73(s,1H),8.34(s,1H),8.09(s,1H),4.51-4.63(m,1H),4.40-4.47(m,2H),3.74-3.88(m,2H),3.35(s,3H),3.13-3.22(m,4H),2.78-2.95(m,2H),2.18(s,3H),1.67-1.82(m,2H)。
Example 60: 4- (7-methyl-2- ((5-methylbenzothiazol-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000243
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 5-methylbenzothiazol-6-amine (33mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (34 mg).
1H NMR(400MHz,DMSO)δ9.19(s,1H),8.70(s,1H),8.36(s,1H),8.19(s,1H),7.89(s,1H),3.86-3.92(m,2H),3.52-3.59(m,2H),3.28(s,3H),2.67-2.75(m,2H),2.56-2.63(m,2H),2.37(s,3H)。
Example 61: 4- (2- ((5, 7-Dimethylquinolin-6-yl) amino) -7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000244
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 5, 7-dimethylquinolin-6-amine (34mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (25mg).
1H NMR(400MHz,DMSO)δ8.80(s,1H),8.76(s,1H),8.40(d,J=7.6Hz,1H),8.03(s,1H),7.74(s,1H),7.41-7.48(m,1H),3.56-3.90(m,2H),3.32-3.52(m,2H),3.23(s,3H),2.50-2.74(m,2H),2.44(s,3H),2.31(s,3H),1.35-1.64(m,2H)。
Example 62: 4- (7-methyl-2- ((8-methylquinolin-7-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000251
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 8-methylquinolin-7-amine (32mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (26 mg).
1H NMR(400MHz,DMSO)δ8.97(s,1H),8.84(dd,J=4.0Hz,1.6Hz,1H),8.24(dd,J=8.4Hz,1.6Hz,1H),8.17(s,1H),7.81(d,J=8.8Hz,1H),7.71(d,J=9.2Hz,1H),7.40(dd,J=8.4Hz,4.0Hz,1H),3.83-3.91(m,2H),3.50-3.58(m,2H),3.28(s,3H),2.68-2.76(m,2H),2.62(s,3H),2.54-2.61(m,2H)。
Example 63: 4- (2- ((7-fluoro-5-methylquinolin-6-yl) amino) -7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000252
Step 1: 6-bromo-7-fluoro-5-methylquinoline
Adding concentrated H2SO4(13.7g) was slowly added to glycerol (8.63g) maintaining the temperature below 70 ℃ followed by the addition of 4-bromo-3-fluoro-5-methylaniline (6.1g) which was then warmed to 85 ℃ and stirred for 40 minutes. Potassium iodide (0.30g), iodine (0.34g) and water (1.50mL) were added, and the mixture was stirred at 135 ℃ for 4 hours. Cooling to room temperature, pouring into ice to quench the reaction, and filtering with diatomaceous earth. The filtrate was adjusted to pH 7 with ammonia and extracted thoroughly with ethyl acetate. The organic phase was dried over anhydrous sodium sulfate and concentrated, and the residue was subjected to column chromatography to give 6-bromo-7-fluoro-5-methylquinoline (1.5g) and the isomer 6-bromo-5-fluoro-7-methylquinoline (1.1g), respectively.
Step 2: 7-fluoro-5-methylquinolin-6-amine
6-bromo-7-fluoro-5-methylquinoline (240mg), RuPhos Pd G3(80mg), benzophenone imine (182mg) and cesium carbonate (650mg) were dissolved in toluene (10mL) under nitrogen, heated to 100 ℃ and stirred for 5 hours, cooled to room temperature, filtered, the solid was washed with dichloromethane, the filtrate was concentrated under reduced pressure, the residue was dissolved in tetrahydrofuran (30mL), 1M dilute hydrochloric acid (10mL) was slowly added, stirring was continued for 1 hour, a saturated aqueous sodium bicarbonate solution was added to the system until the pH was neutral, ethyl acetate was added for extraction, and the organic phase was evaporated to dryness and the residue was purified by column chromatography (eluent: dichloromethane: methanol ═ 20: 1 (V: V)) to give 7-fluoro-5-methylquinolin-6-amine (120 mg).
And step 3: 4- (2- ((7-fluoro-5-methylquinolin-6-yl) amino) -7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 7-fluoro-5-methylquinolin-6-amine (35mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (42 mg).
1H NMR(400MHz,DMSO)δ9.01(s,1H),8.85(d,J=2.8Hz,1H),8.47(d,J=8.4Hz,1H),8.07(s,1H),7.63(d,J=10.8Hz,1H),7.51(dd,J=8.8Hz,4.4Hz,1H),3.74-3.83(m,2H),3.43-3.53(m,2H),3.25(s,3H),2.61-2.71(m,2H),2.53(s,3H),2.46-2.52(m,2H)。
Example 64: 4- (2- ((5-fluoro-7-methylquinolin-6-yl) amino) -7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000261
The target compound (30mg) was synthesized in two steps using 6-bromo-5-fluoro-7-methylquinoline obtained in step one of example 63 as a starting material.
1H NMR(400MHz,DMSO)δ8.92(s,1H),8.86(dd,J=4.0Hz,1.2Hz,1H),8.36(d,J=8.4Hz,1H),8.10(s,1H),7.76(s,1H),7.51(dd,J=8.4Hz,4.0Hz,1H),3.70-3.78(m,2H),3.41-3.48(m,2H),3.26(s,3H),2.59-2.68(m,2H),2.44-2.50(m,2H),2.41(s,3H)。
Example 65: 4- (2- ((2, 2-difluoro-6-methylbenzo [ d ] [1, 3] dioxol-5-yl) amino) -7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000262
Step 1: 2, 2-difluoro-6-methylbenzo [ d ] [1, 3] dioxin-5-amine
5-bromo-2, 2-difluoro-6-methylbenzo [ d ] [1, 3] dioxine (250mg), RuPhos Pd G3(80mg), benzophenone imine (182mg) and cesium carbonate (650mg) were dissolved in toluene (10mL) under nitrogen, heated to 100 ℃ and stirred for 5 hours. Cooled to room temperature, filtered, the solid washed with dichloromethane then the filtrate was concentrated under reduced pressure, the residue was dissolved in tetrahydrofuran (30mL) and 1M dilute hydrochloric acid (10mL) was added slowly, stirring was continued for 1 hour, and saturated aqueous sodium bicarbonate was added to the system until the pH was neutral. Extraction was performed with ethyl acetate, and the organic phase was evaporated to dryness and the residue was purified by column chromatography (eluent: dichloromethane: methanol ═ 20: 1 (V: V)) to give 2, 2-difluoro-6-methylbenzo [ d ] [1, 3] dioxin-5-amine (95 mg).
Step 2: 4- (2- ((2, 2-difluoro-6-methylbenzo [ d ] [1, 3] dioxol-5-yl) amino) -7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 2-difluoro-6-methylbenzo [ d ] [1, 3] dioxin-5-amine (38mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours, cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (36mg).
1H NMR(400MHz,DMSO)δ7.90(s,1H),7.69(s,1H),6.90(s,1H),6.58(s,1H),4.03-4.10(m,2H),3.82-3.90(m,2H),3.37(s,3H),2.72-2.82(m,4H),2.29(s,3H).
Example 66: 4- (7-methyl-2- ((7-methyl-2, 3-dihydrobenzo [ b ] [1, 4] dioxin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000271
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 7-methyl-2, 3-dihydrobenzo [ b ] [1, 4] dioxin-6-amine (31mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours, cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (43mg).
1H NMR(400MHz,DMSO)δ8.34(s,1H),8.07(s,1H),6.91(s,1H),6.64(s,1H),4.16(s,4H),3.87-3.93(m,2H),3.54-3.62(m,2H),3.25(s,3H),2.69-2.77(m,2H),2.56-2.63(m,2H),2.05(s,3H)。
Example 67: 4- (7-methyl-2- ((7-methylbenzo [ b ] [1, 4] dioxin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000272
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 7-methylbenzo [ b ] [1, 4] dioxin-6-amine (33mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours, cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (27 mg).
1H NMR(400MHz,DMSO)δ8.41(s,1H),8.11(s,1H),6.87(s,1H),6.53(s,1H),6.13(s,2H),3.89-3.96(m,2H),3.55-3.63(m,2H),3.26(s,3H),2.69-2.77(m,2H),2.57-2.63(m,2H),2.03(s,3H)。
Example 68: 4- (2- ((8-fluoro-7-methylquinolin-6-yl) amino) -7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000281
The title compound (20mg) was synthesized in 3 steps using 4-bromo-2-fluoro-3-methylaniline as the starting material in analogy to example 63.
1H NMR(400MHz,DMSO)δ8.93(s,1H),8.76(dd,J=4.4Hz,1.6Hz,1H),8.25(s,1H),8.22(d,J=8.4Hz,1H),8.08(s,1H),7.49(dd,J=8.4Hz,4.4Hz,1H),3.85-3.92(m,2H),3.52-3.60(m,2H),3.30(s,3H),2.70-2.79(m,2H),2.58-2.64(m,2H),2.36(d,J=2.4Hz,3H)。
Example 69: 4- (2- ((7- (difluoromethyl) quinolin-6-yl) amino) -7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000282
The title compound (22mg) was synthesized in 3 steps using 4-bromo-3- (difluoromethyl) aniline as the starting material in analogy to example 63.
1H NMR(400MHz,DMSO)δ8.95(s,1H),8.86(d,J=2.8Hz,1H),8.34(s,1H),8.29(d,J=8.4Hz,1H),8.24(s,1H),8.18(s,1H),7.57(dd,J=8.4Hz,4.4Hz,1H),7.45(t,J=54.2Hz,1H),3.84-3.91(m,2H),3.52-3.59(m,2H),3.31(s,3H),2.68-2.77(m,2H),2.56-2.62(m,2H)。
Example 70: 27,45-dimethyl-28,29-dihydro-27H-5-oxa-3-aza-2 (9, 2) -purine-4 (4, 2) -pyridine-1 (4, 1) -piperidinepepane-28-ketones
Figure BSA0000220128980000283
This compound was synthesized by following the synthesis procedure of example 7 using 2-bromo-5-methyl-4-nitropyridine as starting material.
1H NMR(400MHz,DMSO)δ8.45(s,1H),8.26(s,1H),8.02(s,1H),7.80(s,1H),4.35-4.45(m,1H),4.26-4.32(m,2H),3.32(s,3H),3.07-3.15(m,2H),2.62-2.88(m,6H),2.18(s,3H),1.37-1.45(m,2H)。
Example 71: 4- (7- (methyl-d 3) -2- ((7-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000284
Figure BSA0000220128980000291
Analogously to example 1, CD was used in the synthesis of intermediate 13I (deuterated iodomethane) instead of CH3The target compound (33mg) is synthesized by 2 steps by taking the I as a substitute raw material.
1H NMR(400MHz,DMSO)δ9.06(s,1H),8.76(s,1H),8.35(s,1H),8.18(s,1H),7.67(s,1H),3.87-3.93(m,2H),3.52-3.60(m,2H),2.68-2.76(m,2H),2.58-2.64(m,2H),2.35(s,3H)。
Example 72: 4- (7- (methyl-d 3) -2- ((7-methylquinolin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000292
Using the same procedure as in example 71, 7-methylquinolin-6-amine as a surrogate amine was subjected to a catalytic coupling reaction to give the title compound (34 mg).
1H NMR(400MHz,DMSO)δ8.69-8.71(m,2H),8.26(s,1H),8.25(s,1H),8.14-8.17(m,1H),7.81(s,1H),7.39(dd,J=8.4Hz,4.0Hz,1H),3.85-3.92(m,2H),3.53-3.60(m,2H),2.72-2.79(m,2H),2.59-2.65(m,2H),2.47(s,3H)。
Example 73: 44-chloro-27,46-dimethyl-28,29-dihydro-27H-5-thia-3-aza-2 (9, 2) -purine-1 (4, 1) -piperidine-4 (1, 3) -benzocycloheptane-28-ketones
Figure BSA0000220128980000293
This compound was synthesized by following the synthesis procedure of example 7 using 2-chloro-4-methyl-5-nitrobenzenethiol as starting material.
1H NMR(400MHz,DMSO)δ8.23(s,1H),7.88(s,1H),7.20(s,1H),6.86(s,1H),4.47-4.58(m,1H),3.41(s,3H),3.21(t,J=7.2Hz,2H),3.12-3.19(m,2H),3.09(t,J=7.2Hz,2H),2.82-2.97(m,4H),2.29(s,3H),1.47-1.55(m,2H)。
Example 74: 54-chloro-37,56-dimethyl-38,39-dihydro-37H-6-oxo-4-aza-3 (9, 2) -purine-2 (1, 4) -piperidine-1, 5(1, 3) -dibenzosuberane-38-ketones
Figure BSA0000220128980000294
This compound was synthesized by following the synthesis procedure of example 7 using 2-chloro-4-methyl-5-nitrophenol and 3-bromobenzyl bromide as starting materials.
1H NMR(400MHz,DMSO)δ7.79(s,1H),7.43(s,1H),7.31(s,1H),7.12(s,1H),7.09(t,J=8.0Hz,1H),6.86-6.90(m,1H),6.50(d,J=7.6Hz,1H),5.27(s,1H),5.24(s,2H),4.64-4.74(m,1H),4.05-4.13(m,2H),3.39(s,3H),3.27(t,J=13.6Hz,2H),2.59-2.72(m,2H),2.22(s,3H),1.61-1.69(m,2H)。
Example 75: 4- (7-methyl-2- ((6-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-7-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000301
Step 1: (E) -N' - (4-iodo-5-methylpyridin-2-yl) -N, N-dimethylformamide
4-iodo-5-methylpyridin-2-amine (2.34g) was dissolved in toluene (50mL) at room temperature, 1-dimethoxy-N, N-dimethylmethylamine (4.0mL) was added thereto, and the reaction mixture was stirred at reflux for 3 hours. Cooled to room temperature and concentrated under reduced pressure to give the title compound (2.5g) as a yellow solid.
Step 2: (E) -N-hydroxy-N' - (4-iodo-5-methylpyridin-2-yl) carboxamide
Hydroxylamine hydrochloride (1.4g) was added to (E) -N' - (4-iodo-5-methylpyridin-2-yl) -N, N-dimethylformamide (2.5g) in MeOH (50mL) at room temperature. The reaction mixture was stirred at reflux for 2h, cooled to room temperature, poured into water and extracted with EtOAc (100 mL). The extract was washed with saturated brine (50mL), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the title compound (2g) as a yellow solid.
And step 3: 7-iodo-6-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridines
2, 2, 2-trifluoroacetic anhydride (2.1g) was added to (E) -N-hydroxy-N' - (4-iodo-5-methylpyridin-2-yl) formamide (2g) in THF (50mL) at 0 deg.C. The reaction mixture was stirred at room temperature for 18 hours and then concentrated. The reaction solution was poured into water, extracted with ethyl acetate, and the organic phase was evaporated to dryness and the residue was purified by column chromatography (eluent: dichloromethane: methanol 50: 1 (V: V)) to give the title compound (1.2g).
And 4, step 4: 6-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-7-amine
7-iodo-6-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridine (259mg), RuPhos Pd G3(80mg), benzophenone imine (182mg), and cesium carbonate (650mg) were dissolved in toluene (10mL) under nitrogen, heated to 100 ℃ and stirred for 7 hours. Cooled to room temperature, filtered, the solid was washed with dichloromethane, the filtrate was concentrated under reduced pressure, the residue was dissolved in tetrahydrofuran (30mL), 1M dilute hydrochloric acid (10mL) was slowly added, stirring was continued for 1 hour, and a saturated aqueous sodium bicarbonate solution was added to the system until the pH was neutral. Extraction was performed with ethyl acetate, and the organic phase was evaporated to dryness and the residue was purified by column chromatography (eluent: dichloromethane: methanol ═ 10: 1 (V: V)) to give 6-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-7-amine (90 mg).
And 5: 4- (7-methyl-2- ((6-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-7-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 6-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-7-amine (30mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 10: 1 (V: V)) to give the objective compound (56 mg).
1H NMR(400MHz,DMSO)δ8.71(s,1H),8.68(s,1H),8.33(s,1H),8.27(s,1H),8.24(s,1H),3.95-4.01(m,2H),3.58-3.67(m,2H),3.32(s,3H),2.75-2.84(m,2H),2.67-2.73(m,2H),2.34(s,3H).
Example 76: 7-methyl-2- ((6-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-7-yl) amino) -9- (tetrahydro-2H-pyran-4-yl) -7, 9-dihydro-8H-purin-8-one
Figure BSA0000220128980000311
Under the protection of nitrogen, the intermediate 2: 2-chloro-7-methyl-9- (tetrahydro-2H-pyran-4-yl) -7, 9-dihydro-8H-purin-8-one (54mg), 6-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-7-amine (30mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL), heated to 100 ℃ and stirred for 2 hours, cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 15: 1 (V: V)) to give the objective compound (43mg).
1H NMR(400MHz,DMSO)δ8.70(s,1H),8.53(s,1H),8.39(s,1H),8.23(s,2H),4.38-4.50(m,1H),3.90-4.01(m,2H),3.40(t,J=11.2Hz,2H),3.32(s,3H),2.44-2.58(m,2H),2.36(s,3H),1.62-1.72(m,2H).
Example 77: 4- (7- (methyl-d 3) -2- ((6-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-7-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000312
Following example 71, 6-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-7-amine was used as a surrogate amine to undergo a catalytic coupling reaction to give the title compound (44mg).
1H NMR(400MHz,DMSO)δ8.71(s,1H),8.68(s,1H),8.33(s,1H),8.27(s,1H),8.24(s,1H),3.94-4.02(m,2H),3.58-3.67(m,2H),2.74-2.84(m,2H),2.66-2.74(m,2H),2.34(s,3H)。
Example 78: 4- (7-methyl-2- ((7-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carboxamide
Figure BSA0000220128980000313
At room temperature, 4- (7-methyl-2- ((7-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (30mg) was dissolved in THF (10mL), 1M NaOH (1mL) was added and heated to 50 ℃ and stirred for 2 hours, the filtrate was cooled to room temperature, the filtrate was concentrated under reduced pressure, and the residue was purified by column chromatography (eluent: dichloromethane: methanol ═ 10: 1 (V: V)) to give the objective compound (17 mg).
1H NMR(400MHz,DMSO)δ8.93(s,1H),8.55(s,1H),8.33(s,1H),8.10(s,1H),7.66(s,1H),7.33(s,1H),7.13(s,1H),3.62-3.70(m,2H),3.38-3.48(m,2H),3.25(s,3H),2.97-3.06(m,2H),2.32(s,3H),2.01-2.11(m,2H).
Example 79: 4- (7-methyl-2- ((5-methylbenzo [ d ] oxazol-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000321
Step 1: 5-methylbenzo [ d ] oxazol-6-amines
6-bromo-5-methylbenzo [ d ] oxazole (212mg), RuPhos Pd G3(80mg), benzophenone imine (182mg) and cesium carbonate (650mg) were dissolved in toluene (10mL) under nitrogen, heated to 100 ℃ and stirred for 5 hours, cooled to room temperature, filtered, the solid washed with dichloromethane then the filtrate was concentrated under reduced pressure, the residue was dissolved in tetrahydrofuran (30mL) and 1M dilute hydrochloric acid (10mL) was slowly added, stirring was continued for 1 hour and then saturated aqueous sodium bicarbonate was added to the system until the pH was neutral. Ethyl acetate was added for extraction, and the organic phase was evaporated to dryness and the residue was purified by column chromatography (developing solvent: dichloromethane: methanol 20: 1 (V: V)) to give 5-methylbenzo [ d ] oxazol-6-amine (50 mg).
Step 2: 4- (7-methyl-2- ((5-methylbenzo [ d ] oxazol-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 5-methylbenzo [ d ] oxazol-6-amine (30mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 3 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol 15: 1 (V: V)) to give the objective compound (33 mg).
1H NMR(400MHz,DMSO)δ8.62(s,1H),8.57(s,1H),8.17(s,1H),7.95(s,1H),7.58(s,1H),3.86-3.92(m,2H),3.52-3.60(m,2H),3.28(s,3H),2.69-2.79(m,2H),2.56-2.63(m,2H),2.32(s,3H).
Example 80: (E) -27,47-dimethyl-28,29-dihydro-27H-5-thia-3-aza-2 (9, 2) -purine-4 (6, 3) - [1, 2, 4]]Triazole [4, 3-a ]]Pyridine-1 (4, 1) -piperidine cycloheptane-28-ketones
Figure BSA0000220128980000322
This compound was synthesized by following the synthesis procedure of example 7 using 7-methyl-6-nitro- [1, 2, 4] triazolo [4, 3-a ] pyridine-3-thiol as the starting material.
1H NMR(400MHz,DMSO)δ8.83(s,1H),8.70(s,1H),8.01(s,1H),7.49(s,1H),3.72-3.83(m,1H),3.21(s,3H),3.08-3.14(m,2H),2.59-2.72(m,4H),2.25(s,3H),1.72-1.87(m,2H),1.63-1.71(m,2H),1.24-1.33(m,2H)。
Example 81: 4- (7-methyl-2- ((6-methylbenzothiazol-5-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000331
Intermediate 14- (2-chloro-7-methyl-8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile (59mg), 6-methylbenzothiazol-5-amine (33mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL) under nitrogen, heated to 100 ℃ and stirred for 2 hours, cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 20: 1 (V: V)) to give the objective compound (47 mg).
1H NMR(400MHz,DMSO)δ9.24(s,1H),8.67(s,1H),8.25(s,1H),8.17(s,1H),7.92(s,1H),3.85-3.92(m,2H),3.52-3.60(m,2H),3.28(s,3H),2.70-2.79(m,2H),2.58-2.65(m,2H),2.35(s,3H)。
Example 82: 4- (7- (fluoromethyl) -2- ((7-methyl- [1, 2, 4] triazolo [1, 5-a ] pyridin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000332
Analogously to example 1, the intermediate 1 was synthesized by fluorinationIodomethane instead of CH3I. Cesium carbonate is used as a substitute for sodium hydride as an alkali to obtain a target compound (13mg) through 2 steps of synthesis.
1H NMR(400MHz,DMSO)δ9.50(s,1H),8.26(s,1H),8.14(s,1H),7.58(s,1H),6.82(s,1H),5.90(d,J=53.6Hz,2H),4.07-4.14(m,2H),3.84-3.92(m,2H),2.73-2.83(m,4H),2.49(s,3H)。
Example 83: 4- (7- (fluoromethyl) -2- ((7-methylquinolin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000333
The reaction was performed in analogy to example 75, using 7-methylquinolin-6-amine as the substituting amine for the catalytic coupling reaction to give the title compound (16mg).
1H NMR(400MHz,DMSO)δ8.75(dd,J=4.0Hz,1.6Hz,1H),8.63(s,1H),8.14-8.19(m,2H),7.93(s,1H),7.34(dd,J=8.0Hz,4.0Hz,1H),7.14(s,1H),5.89(d,J=53.6Hz,2H),4.04-4.10(m,2H),3.84-3.91(m,2H),2.76-2.86(m,4H),2.57(s,3H)。
Example 84: 7-methyl-2- ((7-methyl- [1, 2, 4] triazolo [4, 3-a ] pyridin-6-yl) amino) -9- (tetrahydro-2H-pyran-4-yl) -7, 9-dihydro-8H-purin-8-one
Figure BSA0000220128980000341
Under the protection of nitrogen, the intermediate 2: 2-chloro-7-methyl-9- (tetrahydro-2H-pyran-4-yl) -7, 9-dihydro-8H-purin-8-one (54mg), 7-methyl- [1, 2, 4] triazolo [4, 3-a ] pyridin-6-amine (30mg), RuPhos Pd G3(9mg), and cesium carbonate (130mg) were dissolved in dioxane (20mL), heated to 100 ℃ and stirred for 2 hours. Cooled to room temperature, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by thin layer chromatography (developing solvent: dichloromethane: methanol ═ 15: 1 (V: V)) to give the objective compound (34 mg).
1H NMR(400MHz,DMSO)δ9.08(s,1H),8.65(s,1H),8.34(s,1H),8.05(s,1H),7.67(s,1H),4.33-4.43(m,1H),3.92(dd,J=11.2Hz,4.0Hz,2H),3.33-3.42(m,2H),3.27(s,3H),2.41-2.54(m,2H),2.36(s,3H),1.60-1.68(m,2H)。
Example 85: 4- (7- (methyl-d 3) -2- ((7-methyl- [1, 2, 4] triazolo [4, 3-a ] pyridin-6-yl) amino) -8-oxo-7, 8-dihydro-9H-purin-9-yl) tetrahydro-2H-pyran-4-carbonitrile
Figure BSA0000220128980000342
The procedure was followed as in example 71, and 7-methyl- [1, 2, 4] triazolo [4, 3-a ] pyridin-6-amine was used as a substitute amine to conduct a catalytic coupling reaction to give the title compound (24 mg).
1H NMR(400MHz,DMSO)δ9.58(s,1H),8.26(s,1H),7.97(s,1H),7.57(s,1H),6.68(s,1H),4.08-4.13(m,2H),3.86-3.92(m,2H),2.75-2.85(m,4H),2.49(s,3H).
Biological Activity assay
DNA-PK compound bioactivity test method
1. Determination of proliferative Activity of Compounds in MDA-MB-468 cells
The patent relates to a detection method for cell proliferation inhibition, which is established under the condition of combination of a DNA-PK inhibitor established in human breast cancer cells MDA-MB-468 and a chemotherapeutic drug Doxorubicin. The specific method comprises the following steps: human breast cancer cells MDA-MB-468 cells were cultured in RPMI-1640 medium (purchased from Biological Industries, BI) supplemented with 10% fetal bovine serum (FBS, purchased from Hyclone) and 1% penicillin/streptomycin double antibody (P/S, purchased from Life Techology) under the conditions of (37 ℃, 5% CO 2). The day before compound detection, MDA-MB-468 cells were plated in 96-well plates (#3917, purchased from Corning) at a concentration of 1000 cells/190. mu.L/well. After 24 hours Doxorubicin was added to a final concentration of 10nM (DMSO final concentration 0.1%), the test compound was diluted 3-fold in 100% DMSO starting at 10mM (10 concentrations in total), then 2. mu.L of each concentration was added to 48. mu.L of RPMI-1640 medium, and 5. mu.L of each diluted test compound was added to the plated cell suspension. After incubation of the compounds with cells in a Cell incubator for 120h (5 days), the medium was aspirated and incubated again for 5-10 min with 25. mu.L of Cell-Titer Glo (G7570, purchased from Promega) reagent. Fluorescence values were then read on a CLARIO starPlus (purchased from BMG) plate reader and the data were calculated using GraphPad Prism software to obtain IC50 values for inhibition of cell proliferation by this compound.
Partial examples cellular Activity data
Compound numbering Active IC in MDA-MB-468 cells50(nM)
Example 39 116
Example 40 29
EXAMPLE 41 74
Example 42 80
Example 43 161
Example 44 28
Example 48 23
Example 53 59
Example 54 58
Example 56 27
Example 60 23
Example 67 98
Example 71 114
Example 72 34
2. Pharmacokinetic data for compounds:
male SD rats are from Beijing Wittingle laboratory animal technology, Inc., the rats are divided into groups of 3 rats, and suspension of samples to be tested (5mg/kg, suspension is 0.5% HPMC, 0.1% Tween 80in H) is orally administrated2O). Animals were fasted overnight prior to the experiment, with the fasting time ranging from 10 hours prior to dosing to 4 hours post-dosing. Blood was collected at 0.25, 0.5, 1, 2, 4, 6, 8, and 24 hours post-dose, respectively. After isoflurane anesthesia by using a small animal anesthesia machine, 0.3mL of whole blood is collected through an eyeground venous plexus, the whole blood is placed in a heparin anticoagulation tube, a sample is centrifuged at 4000rpm and 4 ℃ for 5 minutes, and the plasma is transferred to a centrifuge tube and stored at-80 ℃ until analysis. Sample use in plasmaProtein precipitation extraction and the extract was analyzed by LC/MS.
Figure BSA0000220128980000351

Claims (9)

1. A compound of formula (I) or a pharmaceutically acceptable salt, solvate, polymorph or isomer thereof,
Figure FSA0000220128970000011
wherein the content of the first and second substances,
ring A is 6-10 membered aryl or 5-12 membered heteroaryl,
ring B is a 4-12 membered heterocyclic ring, S on ring B may be optionally oxidized,
z is O or S, and the compound is,
X2is CR2Or the number of N is greater than the number of N,
X1is CRR4O, S, or NR6
R1Is H, C1-6Alkyl, or C3-8Cycloalkyl, said alkyl and cycloalkyl being optionally substituted by halogen, -CN, -OH, -NH2、-O-C1-6Alkyl, or-NR-C1-6The substitution of the alkyl group is carried out,
R7and R8Each independently selected from halogen, CN, C1-6Alkyl, -O-C1-6Alkyl and-NR-C1-6Alkyl, which may optionally be substituted by halogen, -CN, -OH, -NH2、-O-C1-6Alkyl, or-NR-C1-6The substitution of the alkyl group is carried out,
m and n are each independently 0, 1, 2, or 3,
R3is R5or-X3-R5
R4Is R6or-X3-R6
X3Each independently is-O-, -S-, or-NR-,
R5and R6Each independently selected from H and C1-6Alkyl, or R5And R6Taken together to form- (CH)2)p-X-(CH2)q-, wherein X is a bond, -O-, -S, -N (R) -, -CO-, -C (O) NR-, -C (O) O-, 6-10 membered arylene, 5-12 membered heteroarylene, or 3-12 membered heterocycle, and- (CH)2)p-X-(CH2)qCH in (E)2Optionally substituted by a halogen, and optionally substituted by a halogen,
p and q are each independently 0, 1, 2, 3, or 4, and p + q is 1, 2, 3, 4, 5, or 6,
R2selected from H, halogen, CHF2、CF3、-OH、-NH2、CN、C1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, -O-C1-6Alkyl, - (CH)2)1-6-CN、-(CH2)1-6-O-C1-6Alkyl, - (CH)2)1-3-OH、-CHO、-(CO)NH2- (CO) NHR, - (CO) OR and-NR-C1-6An alkyl group.
R is H or C1-6An alkyl group.
2. A compound according to claim 1 or a pharmaceutically acceptable salt, solvate, polymorph or isomer thereof, wherein
When R is5And R6Each independently selected from H or C1-6When alkyl, R2Selected from halogen, CH2F、CHF2、CF3、-OH、-NH2、CN、C1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, -O-C1-6Alkyl, - (CH)2)1-6-CN、-(CH2)1-6-O-C1-6Alkyl, or-NR-C1-6An alkyl group, a carboxyl group,
when R is5And R6Taken together to form- (CH)2)p-X-(CH2)qWhen is, R2Selected from H, halogen, CHF2、CF3、-OH、-NH2、CN、C1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, -O-C1-6Alkyl, - (CH)2)1-6-CN、-(CH2)1-6-O-C1-6Alkyl and-NR-C1-6An alkyl group, a carboxyl group,
r is H or C1-6An alkyl group.
3. A compound according to claim 1, or a pharmaceutically acceptable salt, solvate, polymorph or isomer thereof, wherein
Figure FSA0000220128970000021
Is composed of
Figure FSA0000220128970000022
Figure FSA0000220128970000023
Or
Figure FSA0000220128970000024
When R is2Selected from halogen, CH2F、CHF2、CF3、-OH、-NH2、CN、C1-6Alkyl radical, C2-6Alkenyl radical, C2-6Alkynyl, -O-C1-6Alkyl, - (CH)2)1-6-CN、-(CH2)1-6-O-C1-6Alkyl and-NR-C1-6An alkyl group, a carboxyl group,
r is H or C1-6An alkyl group.
4. The compound according to claim 1, or a pharmaceutically acceptable salt, solvate, polymorph or isomer thereof, wherein R1Is C1-6Alkyl, which may optionally be substituted by halogen, -CN, -OH, -NH2、-O-C1-6Alkyl, or-NR-C1-6Alkyl substituted, R is H or C1-6An alkyl group.
5. The following compounds:
Figure FSA0000220128970000025
Figure FSA0000220128970000031
or a pharmaceutically acceptable salt, solvate, polymorph or isomer thereof.
6. A pharmaceutical composition comprising a compound according to any one of claims 1-5, or a pharmaceutically acceptable salt, solvate, polymorph, or isomer thereof.
7. Use of a compound according to any one of claims 1 to 5 or a pharmaceutically acceptable salt, solvate, polymorph or isomer thereof or a pharmaceutical composition according to claim 6 for the manufacture of a medicament for the treatment of DNA-PK related diseases.
8. The use of claim 7, wherein the DNA-PK related disease is cancer.
9. The use of claim 7, wherein the DNA-PK associated disease is colorectal cancer, glioblastoma, gastric cancer, ovarian cancer, diffuse large B-cell lymphoma, chronic lymphocytic leukemia, acute myelogenous leukemia, squamous cell carcinoma of the head and neck, breast cancer, prostate cancer, bladder cancer, hepatocellular carcinoma, small cell lung cancer, or non-small cell lung cancer.
CN202010998179.XA 2020-07-20 2020-09-18 DNA-PK selective inhibitor and preparation method and application thereof Pending CN114195805A (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CN202010998179.XA CN114195805A (en) 2020-09-18 2020-09-18 DNA-PK selective inhibitor and preparation method and application thereof
CN202180061261.2A CN116406272A (en) 2020-07-20 2021-07-20 DNA-PK selective inhibitor and preparation method and application thereof
PCT/CN2021/107304 WO2022017368A1 (en) 2020-07-20 2021-07-20 Dna-pk selective inhibitor, preparation method therefor and use thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202010998179.XA CN114195805A (en) 2020-09-18 2020-09-18 DNA-PK selective inhibitor and preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN114195805A true CN114195805A (en) 2022-03-18

Family

ID=80645289

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010998179.XA Pending CN114195805A (en) 2020-07-20 2020-09-18 DNA-PK selective inhibitor and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN114195805A (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114634522A (en) * 2020-12-15 2022-06-17 江苏恒瑞医药股份有限公司 Purine ketone derivative, preparation method and medical application thereof
WO2023036156A1 (en) * 2021-09-07 2023-03-16 首药控股(北京)股份有限公司 Dna-pk selective inhibitor, and preparation method therefor and use thereof

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN114634522A (en) * 2020-12-15 2022-06-17 江苏恒瑞医药股份有限公司 Purine ketone derivative, preparation method and medical application thereof
WO2023036156A1 (en) * 2021-09-07 2023-03-16 首药控股(北京)股份有限公司 Dna-pk selective inhibitor, and preparation method therefor and use thereof

Similar Documents

Publication Publication Date Title
CN113646314B (en) FGFR4 kinase inhibitor and preparation method and application thereof
JP6888006B2 (en) Pyrazolo, pyrazolo, imidazole-pyrimidine and pyridine compounds that inhibit MNK1 and MNK2
CN113956272A (en) DNA-PK selective inhibitor and preparation method and application thereof
CN116406272A (en) DNA-PK selective inhibitor and preparation method and application thereof
JP2022538917A (en) Heterocyclic compounds as BET inhibitors
CN114302886B (en) Triazolopyridazine derivative, preparation method, pharmaceutical composition and application thereof
CN114195805A (en) DNA-PK selective inhibitor and preparation method and application thereof
JP2021534250A (en) Highly active STING protein agonist
JP7357146B2 (en) Azaheteroaryl compounds and their uses
CN111718348A (en) WEE1 inhibitor, and preparation and application thereof
JP2022528047A (en) Wee1 inhibitor, its manufacture and use
CN115028644A (en) SOS1 inhibitor heterocyclic compounds
JP6816287B2 (en) Pyridine and 5-membered aromatic ring compounds, their production methods and uses
CN108699065A (en) Tricyclic compounds and composition as kinase inhibitor
CN114072404B (en) RET selective inhibitor and preparation method and application thereof
CN114605414A (en) DNA-PK selective inhibitor and preparation method and application thereof
CN114573584A (en) DNA-PK selective inhibitor and preparation method and application thereof
CN114634521A (en) DNA-PK selective inhibitor and preparation method and application thereof
CN114907384A (en) DNA-PK selective inhibitor and preparation method and application thereof
CN114075218A (en) USP7 inhibitor
CN112724144A (en) WEE1 inhibitor, and preparation and application thereof
CN112209925A (en) RET selective inhibitor and preparation method and application thereof
CN111662292A (en) FGFR4 kinase inhibitor and preparation method and application thereof
CN115433206A (en) DNA-PK selective inhibitor and preparation method and application thereof
CN117940437A (en) DNA-PK selective inhibitor and preparation method and application thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
WD01 Invention patent application deemed withdrawn after publication
WD01 Invention patent application deemed withdrawn after publication

Application publication date: 20220318