WO2023116594A1 - 取代的稠杂环化合物及其制备方法与应用 - Google Patents

取代的稠杂环化合物及其制备方法与应用 Download PDF

Info

Publication number
WO2023116594A1
WO2023116594A1 PCT/CN2022/139864 CN2022139864W WO2023116594A1 WO 2023116594 A1 WO2023116594 A1 WO 2023116594A1 CN 2022139864 W CN2022139864 W CN 2022139864W WO 2023116594 A1 WO2023116594 A1 WO 2023116594A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
cancer
membered
pharmaceutically acceptable
acceptable salt
Prior art date
Application number
PCT/CN2022/139864
Other languages
English (en)
French (fr)
Inventor
龙凯
Original Assignee
上海安诺达生物科技有限公司
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 上海安诺达生物科技有限公司 filed Critical 上海安诺达生物科技有限公司
Priority to CN202280012432.7A priority Critical patent/CN116761804A/zh
Publication of WO2023116594A1 publication Critical patent/WO2023116594A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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
    • 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
    • C07D473/34Nitrogen atom attached in position 6, e.g. adenine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers

Definitions

  • the invention relates to a substituted condensed heterocyclic compound and its preparation method and application.
  • ATR Rad3-related
  • cancer cells Unlike normal cells, cancer cells exhibit genomic instability, a key hallmark of cancer, due to defective DNA damage responses and/or increased replication stress.
  • the characteristics of cancer cells that are different from normal cells make it easier to accumulate gene mutations, rearrangements, copy number changes and other events during the division of cancer cells, which may promote the clonal evolution of cancer cells; but at the same time, These features are also defects that create cancer cell-specific vulnerabilities. Taking full advantage of these vulnerabilities is expected to enhance the efficacy of anticancer treatments, thereby improving the prognosis of cancer patients (Pilié.P.G.et al, 2019, Nat.Rev.Clin.Oncol.16,81-104).
  • Cancer cells are more dependent on ATR than normal cells, which is one of the above-mentioned vulnerabilities.
  • oncogene-induced replication stress is a major source of genomic instability in cancer
  • cancer cell survival has become highly dependent on a proficient and well-established replication stress response and thus susceptible to ATR inhibitors.
  • Early work in cell lines and mouse models has provided preclinical support for this concept.
  • mice with reduced ATR levels are highly resistant to cancer development.
  • One object of the present invention is to provide a novel compound as an ATR inhibitor.
  • Another object of the present invention is to provide a preparation method of the compound.
  • Another object of the present invention is to provide the application of the compound.
  • Another object of the present invention is to provide a pharmaceutical composition comprising the compound and its application.
  • Another object of the present invention is to provide intermediates for the preparation of said compounds.
  • Another object of the present invention is to provide a preparation method of the intermediate.
  • the present invention provides a compound of formula I or a pharmaceutically acceptable salt thereof,
  • Y 1 , Y 2 , Y 3 , and Y 4 are each independently CH or N, and at least one of Y 1 and Y 2 is N, and at least one of Y 3 and Y 4 is CH;
  • R is a 5-membered and 6-membered heteroaryl ring group containing n heteroatoms or a 6-membered and 6-membered heteroaryl ring group, or a 5-membered or 6-membered aromatic ring group containing m heteroatoms, wherein n is selected from An integer of 1-4, m is selected from an integer of 0-3, and the heteroatom is selected from one or both of O, N, and S; the 5-membered and 6-membered heteroaromatic ring group containing n heteroatoms or Among the 6-membered and 6-membered heteroaryl ring groups, the heteroaryl ring group is optionally selected from halogen, -OH, -OMe, -NH 2 , -NHMe, -NMe 2 , -CN, -CONH 2 , -Me Substituted by one or more substituents; in the 5-membered or 6-membered aromatic ring group containing m heteroatoms, the aromatic ring
  • R 2 is -OH, C 1-6 alkoxy, -NR 3 R 4 , -CN, C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 cycloalkane A group or a 4-6 membered heterocycloalkyl group, the heteroatom is selected from one of O and N; the C 1-6 alkyl, C 2-6 alkenyl, C 2-6 alkynyl, C 3-6 Cycloalkyl or 4-6 membered heterocycloalkyl is optionally substituted by one or more substituents selected from halogen, -OH, C 1-6 alkoxy, -NR 5 R 6 , -CN;
  • R 3 and R 4 are each independently hydrogen or C 1-6 alkyl
  • R 5 and R 6 are each independently hydrogen or C 1-6 alkyl, or -NR 5 R 6 together represent azetidinyl, tetrahydropyrrolyl, piperidinyl, morpholinyl or 4-methyl Basepiperazinyl.
  • Y 3 and Y 4 are respectively CH; Y 1 and Y 2 are each independently CH or N, and At least one of Y1 and Y2 is N. That is, these specific embodiments of the present invention provide the compound of structure shown in following formula I-1:
  • Y 1 , Y 2 , R 1 , and R 2 are each independently as defined above.
  • Y 1 and Y 2 are N respectively.
  • Y 1 and Y 2 are respectively N, and Y 3 and Y 4 are respectively CH. That is, these specific embodiments of the present invention provide the compound of structure shown in following formula I-2:
  • R 1 and R 2 are each independently as defined above.
  • R 1 is a 5-membered and 6-membered heteroaromatic ring group containing n heteroatoms, and n is selected from 1 An integer of -4, the heteroatom is selected from one or both of O, N, and S; the heteroaryl ring group is optionally selected from halogen, -OH, -OMe, -NH 2 , -NHMe, -NMe 2 , -CN, -CONH 2 , -Me are substituted by one or more substituents.
  • R is a group selected from the following groups:
  • R is hydroxyl, methoxyl, ethoxyl, amino, methylamino, dimethylamino , cyano, methyl, ethyl, propyl, isopropyl, vinyl, propenyl, allyl, ethynyl, propynyl, propargyl, cyclopropyl, hydroxymethyl, methoxymethyl 2-hydroxyethyl, 2-methoxyethyl, 2-dimethylaminoethyl, N-morpholinyl or 4-methylpiperazinyl.
  • the compound of formula I of the present invention in the compound of formula I of the present invention or a pharmaceutically acceptable salt thereof, the compound may be selected from:
  • the present invention also provides an intermediate compound, which includes but is not limited to: any intermediate in the above synthetic route for preparing the compound or a pharmaceutically acceptable salt thereof.
  • the intermediate has the same basic structural unit (substantially the same basic core part or basic ring) as the compound of formula I or a pharmaceutically acceptable salt thereof, or the basic structural unit of the intermediate is contained in In the chemical structure of the compound of formula I, or a pharmaceutically acceptable salt thereof.
  • the intermediate compound has the structure shown in formula II:
  • Y 1 , Y 2 , Y 3 , Y 4 and R 2 are each independently as defined above;
  • R3 is halogen or Wherein said halogen is preferably chlorine.
  • the present invention also provides a method for preparing a compound of structure shown in formula I or a pharmaceutically acceptable salt thereof, the method comprising:
  • the intermediate compound represented by formula II is coupled with R 1 -M to prepare the compound of formula I.
  • R3 is When, M is halogen, preferably bromine
  • Y 1 , Y 2 , Y 3 , Y 4 , R 1 and R 2 are each independently as defined above.
  • the method for preparing the compound represented by formula I or a pharmaceutically acceptable salt thereof of the present invention further includes the process of preparing the intermediate compound.
  • the method for preparing the compound of the structure shown in Formula I or a pharmaceutically acceptable salt thereof of the present invention includes but is not limited to: Synthesizing according to any one of Preparation Example 1 to Preparation Example 27 Routes Prepare the compound of formula I or a pharmaceutically acceptable salt thereof.
  • each substituent of the prepared compound is not limited to the scope of the specific compounds in Preparation Example 1 to Preparation Example 27, and the definition of each substituent can refer to the definition range of each substituent of the aforementioned compound of formula I.
  • the present invention also provides a pharmaceutical composition, which comprises: the compound of the structure represented by formula I or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • the present invention also provides the use of the compound or its pharmaceutically acceptable salt or the pharmaceutical composition as an ATR kinase inhibitor.
  • the compound of the present invention or its pharmaceutically acceptable salt or the pharmaceutical composition is specifically used as an ATR inhibitor, it may be for non-therapeutic purposes (such as in vitro experimental research purposes, etc.), or for therapeutic purposes. That is, the present invention provides the use of the compound or a pharmaceutically acceptable salt thereof or the pharmaceutical composition in the preparation of a medicament for inhibiting ATR kinase.
  • the present invention also provides the application of the compound or its pharmaceutically acceptable salt or the pharmaceutical composition in the preparation of medicine for treating hyperproliferative diseases.
  • the compound of the present invention or a pharmaceutically acceptable salt thereof is used as a pharmaceutical active ingredient for the treatment of hyperproliferative diseases, it includes administering the compound or a pharmaceutically acceptable salt thereof to a subject in need or The steps of the pharmaceutical composition.
  • the hyperproliferative disease includes cancer.
  • cancers may include, for example, melanoma, brain tumor, esophageal cancer, gastric cancer, liver cancer, pancreatic cancer, colorectal cancer, lung cancer, kidney cancer, breast cancer, cervical cancer, ovarian cancer, prostate cancer, skin cancer, neuroblastoma , glioma, sarcoma, bone cancer, uterine cancer, endometrial cancer, head and neck cancer, multiple myeloma, B-cell lymphoma, polycythemia vera, leukemia, thyroid tumor, bladder cancer or gallbladder cancer, etc.
  • pharmaceutically acceptable refers to those compounds, materials, compositions and/or dosage forms which, within the scope of sound medical judgment, are suitable for use in contact with human and animal tissues without excessive Toxicity, irritation, allergic reaction or other problems or complications, commensurate with a reasonable benefit/risk ratio.
  • pharmaceutically acceptable salt refers to a salt of a compound of the present invention, which is prepared from a compound having a specific substituent found in the present invention and a relatively non-toxic acid or base.
  • base addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of base, either neat solution or in a suitable inert solvent.
  • Pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic ammonia or magnesium salts or similar salts.
  • acid addition salts can be obtained by contacting the neutral form of such compounds with a sufficient amount of the acid, either neat solution or in a suitable inert solvent.
  • Examples of pharmaceutically acceptable acid addition salts include salts of inorganic acids including, for example, hydrochloric acid, hydrobromic acid, nitric acid, carbonic acid, bicarbonate, phosphoric acid, dihydrogenphosphate, dihydrogenphosphate, sulfuric acid, Hydrogen sulfate, hydriodic acid, phosphorous acid, etc.; and organic acid salts, such as acetic acid, propionic acid, isobutyric acid, maleic acid, malonic acid, benzoic acid, succinic acid, suberic acid, Fumaric acid, lactic acid, mandelic acid, phthalic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, tartaric acid, methanesulfonic acid and similar acids; also salts of amino acids such as arginine and the like , and salts of organic acids such as glucuronic acid. Certain specific compounds of the present invention contain basic and acidic functional groups and can thus be converted into
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound containing acid groups or bases by conventional chemical methods.
  • such salts are prepared by reacting the free acid or base form of these compounds with a stoichiometric amount of the appropriate base or acid in water or an organic solvent or a mixture of both.
  • keys with wedge-shaped solid lines and dotted wedge keys Indicates the absolute configuration of a stereocenter, with a straight solid-line bond and straight dashed keys Indicates the relative configuration of the stereocenter, with a wavy line Indicates wedge-shaped solid-line bond or dotted wedge key or with tilde Indicates a straight solid line key and straight dashed keys
  • substituted means that any one or more hydrogen atoms on a specific atom are replaced by a substituent, which may include deuterium and hydrogen variants, as long as the valence of the specific atom is normal and the substituted compound is stable .
  • Oxygen substitution does not occur on aromatic groups.
  • optionally substituted means that it may or may not be substituted, unless otherwise specified, and the type and number of substituents can be arbitrary on the basis of chemically realizable.
  • any variable eg, R
  • its definition is independent at each occurrence.
  • said group may optionally be substituted with up to two R, with independent options for each occurrence of R.
  • substituents and/or variations thereof are permissible only if such combinations result in stable compounds.
  • one of the variables When one of the variables is selected from deletion, it means its absence, such as R3 in CR 3 is selected from deletion, which means that the structure is actually C.
  • substituent When the enumerated substituent does not indicate which atom it is connected to the substituted group, this substituent can be bonded through any atom, for example, phenyl can be used as a substituent through any atom on the benzene ring. The carbon atom is attached to the group being substituted.
  • alkyl is used to denote a linear or branched saturated hydrocarbon group, which may be monosubstituted (such as -CH 2 F) or polysubstituted (such as -CF 3 ), which may be monovalent (such as methyl), divalent (such as methylene) or multivalent (such as methine).
  • alkyl groups examples include methyl (Me), ethyl (Et), propyl (e.g., n-propyl and isopropyl), butyl (e.g., n-butyl, isobutyl, s-butyl , t-butyl), pentyl (eg, n-pentyl, isopentyl, neopentyl) and the like.
  • cycloalkyl includes any stable cyclic or polycyclic hydrocarbon group, any carbon atom is saturated, may be monosubstituted or polysubstituted, and may be monovalent, divalent or polyvalent.
  • examples of such cycloalkyl groups include, but are not limited to, cyclopropyl, norbornyl, [2.2.2]bicyclooctane, [4.4.0]bicyclodecane, and the like.
  • alkenyl means a straight or branched chain hydrocarbon group having at least one carbon-carbon double bond
  • C2-6 alkenyl means any of the above-mentioned Hydrocarbyl
  • C 2-4 alkenyl means a hydrocarbyl as described above having 2, 3 or 4 carbon atoms, it being understood that where said alkenyl contains more than one carbon-carbon double bond, said double bonds may be separated from each other separate or conjugated to each other.
  • C alkenyl include, but are not limited to, vinyl, allyl, butenyl, pentenyl, hexenyl, butadienyl, pentadienyl, hexadienyl, and the like.
  • alkynyl means a straight or branched chain hydrocarbon group having at least one carbon-carbon triple bond
  • C2-6 alkynyl means any of the above-mentioned groups having 2, 3, 4, 5 or 6 carbon atoms Hydrocarbyl
  • C 2-4 alkynyl means the above-mentioned hydrocarbon groups having 2, 3 or 4 carbon atoms
  • exemplary "C 2-6 alkynyl” include, but are not limited to, ethynyl, propynyl, butynyl, Pentynyl, hexynyl, methylpropynyl, 4-methyl-1-butynyl and the like.
  • halogen by itself or as part of another substituent means a fluorine (F), chlorine (Cl), bromine (Br) or iodine (I) atom.
  • alkoxy denotes an alkyl group attached to the rest of the molecule through an oxygen atom, wherein the alkyl group has a meaning as described herein.
  • C 1-5 alkoxy includes C 1 , C 2 , C 3 , C 4 and C 5 alkoxy.
  • alkoxy groups include, but are not limited to: methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, n-pentoxy, and S- Pentyloxy.
  • the alkoxy groups may be optionally substituted with one or more substituents described herein.
  • amino (amino) refers to -NH2 , -NH(alkyl) or -N(alkyl)(alkyl).
  • aromatic ring means a polyunsaturated aromatic alkane monocyclic ring, which may be mono- or polysubstituted.
  • 4-6 membered heterocycloalkyl means a saturated monovalent monocyclic hydrocarbon ring containing 3, 4 or 5 carbon atoms, and one or more selected from O, NR a A heteroatom group, wherein, R a represents a hydrogen atom or a C 1-6 alkyl group; the "4-6 membered heterocycloalkyl group” can pass through any carbon atom, or, if present, a nitrogen atom, and The rest of the molecule is connected.
  • heteromatic ring means an aromatic ring containing one to four heteroatoms selected from one or more of N, O and S.
  • fused heteroaromatic ring means a fused heterocyclic ring containing two or more aromatic properties.
  • 5-membered and 6-membered heteroaromatic ring means a condensed 5-membered and 6-membered heteroaryl ring, at least one of which contains more than one heteroatom (including but not limited to O, S or N), the entire group is aromatic, non-limiting examples include benzo 5-membered heteroaryl, 6-membered heteroaryl and 5-membered heteroaryl.
  • 6-membered and 6-membered heteroaryl ring means a 6-membered and 6-membered condensed heteroaryl ring, at least one of which contains more than one heteroatom (including but not (limited to O, S or N), the entire group is aromatic, non-limiting examples include benzo 6-membered heteroaryl, 6-membered heteroaryl and 6-membered heteroaryl.
  • the term “pharmaceutically acceptable salt” or “pharmaceutically acceptable salt thereof” means that the compound of the present invention retains the biological effectiveness and properties of the free acid or free base, and the free acid is obtained by combining with Non-toxic inorganic base or organic base, the salt obtained by reacting the free base with non-toxic inorganic acid or organic acid.
  • the term “pharmaceutical composition” refers to a mixture of one or more compounds or pharmaceutically acceptable salts of the present invention and other chemical components, wherein, “other chemical components” refers to pharmaceutical acceptable carrier, excipient and/or one or more other therapeutic agents.
  • Carrier refers to a material that does not produce significant irritation to an organism and that does not abrogate the biological activity and properties of the administered compound.
  • Excipient refers to an inert substance added to a pharmaceutical composition to facilitate administration of the compound.
  • Non-limiting examples include calcium carbonate, calcium phosphate, sugars, starches, cellulose derivatives (including microcrystalline cellulose), gelatin, vegetable oils, polyethylene glycols, diluents, granulating agents, lubricants, binders agents and disintegrants.
  • the compound of the invention has good ATR enzyme inhibitory activity and inhibitory activity on tumor cell proliferation, and has potential application value in treating diseases related to cell proliferation.
  • High-performance liquid chromatography was performed using a Thermo U3000 high-pressure liquid chromatograph.
  • High-performance liquid phase preparation uses Hanbang DAC-50 or Shimadzu LC-20AP preparative chromatograph.
  • Reaction monitoring uses thin-layer chromatography or liquid chromatography-mass spectrometry.
  • the developer systems used in thin-layer chromatography include: dichloromethane and methanol system, petroleum ether and ethyl acetate system, the volume ratio of the solvent is adjusted according to the polarity of the compound, or adjusted by adding a small amount of triethylamine.
  • Liquid mass spectrometry uses Waters ACQUITY Arc/ACQUITY QDa or Thermo U3000-ISQ EC liquid mass spectrometer.
  • the eluent system includes: dichloromethane and methanol system, petroleum ether and ethyl acetate system, and the volume ratio of the solvent is adjusted according to the polarity of the compound, or adjusted by adding a small amount of triethylamine.
  • reaction temperature is room temperature (20°C-30°C), and the solvents are all dried and purified according to standard methods.
  • the third step 4-bromo-6-chloro-1-(2-(trimethylsilyl)ethoxymethyl)-1H-pyrrolo[2,3-b]pyridine
  • Arylboronic acid pinacol esters are commercially available or prepared from aryl halides and diboronic acid pinacol esters using the Miyaura boronation reaction.
  • Aryl boronic acids are commercially available or prepared by reacting aryl halides with butyllithium and borate esters.
  • reaction solution was cooled to room temperature, ethyl acetate (20 mL) was added, the resulting mixture was filtered through celite, and the filtrate was concentrated under reduced pressure to obtain the crude product of the title compound (300 mg). It was used directly in the next step without further purification.
  • the third step (R)-2-((4-(3-methylmorpholinyl)-2-(1H-pyrrolo[2,3-b]pyridin-4-yl)-7H-pyrrolo[ 2,3-d]pyrimidin-7-yl)sulfonyl)ethanol
  • reaction solution was cooled to room temperature, ethyl acetate (50 mL) was added, and washed with water. The layers were separated, and the organic phase was dried over anhydrous sodium sulfate. After filtration, the filtrate was concentrated under reduced pressure and purified by preparative HPLC to obtain compound 25 (20 mg, yield 32%).
  • reaction solution was cooled to room temperature, ethyl acetate (80 mL) was added, the resulting mixture was filtered through celite, and the filtrate was concentrated under reduced pressure to obtain the crude product of the title compound (252 mg). It was used directly in the next step without further purification.
  • Compound 1' is a known compound and was prepared according to the synthetic route described in the literature (ACS Med. Chem. Lett. 2015, 6, 42–46).
  • Compound 7' is a known compound, which was prepared by referring to the synthetic route described in the literature (ACS Med. Chem. Lett. 2015, 6, 42-46). Compound 7' is a mixture of two diastereoisomers (the ratio is about 1:1), and the following two diastereoisomers were obtained by HPLC chiral resolution.
  • the NMR characterization is as follows:
  • ATR kinase (Eurofins, catalog number 14-953); 5-FAM-AK-17 (Gill Biochemical, catalog number 524315); 1x kinase buffer (50 mM HEPES pH 7.5, 0.0015% Brij- 35, 1 M MnCl 2 ); Stop Buffer (100 mM HEPES pH 7.5, 0.015% Brij-35, 0.2% Coating Reagent #3, 50 mM EDTA); Caliper EZ Reader (Caliper Life Sciences).
  • LoVo cells ATCC, catalog number CCL-29
  • F-12K medium Invitrogen, catalog number 21127-022
  • fetal bovine serum Biological Industries, catalog number 04-002-1A
  • GlutaMAX GlutaMAX
  • Trypsin-EDTA Invitrogen, Cat. No. 25200-072
  • CellTiter-Glo Kit Promega, Cat. No. G7558.
  • Table 4 shows the inhibitory effect of the compounds of the examples on the in vitro proliferation of LoVo cells.
  • the inhibitory activity of compound 1 of the present invention to ATR kinase and the inhibitory activity of LoVo cell proliferation were compared with the comparative compound 4' and compound 5', as shown in Table 5.

Abstract

一种取代的稠杂环化合物及其制备方法与应用。提供了一种具有结构式(I)的化合物或其药学上可接受的盐。所述化合物、或其药学上可接受的盐可作为ATR抑制剂,具有较高活性。

Description

取代的稠杂环化合物及其制备方法与应用 技术领域
本发明是关于一种取代的稠杂环化合物及其制备方法与应用。
背景技术
在每个细胞周期中,人类细胞必须准确而有效地复制整个基因组。这是一项极具挑战性的复杂工程,因为它意味着要复制超过60亿个DNA片断(碱基),而且对准确率和效率的要求极高。在这个过程中,不可避免地会有许多障碍阻止或影响DNA的正常复制。因此,正常的人类细胞具有强大的应答机制,以确保整个基因组在每个细胞周期中准确地复制一次(Saldivar J.C.et al,2017,Nat.Rev.Mol.Cell Biol.18,622-636)。
人类细胞中有诸多调控分子参与维持基因组的稳定性和完整性,其中的重要一员是共济失调毛细血管扩张突变基因和Rad3相关(ATR)蛋白激酶。当细胞内DNA损伤或复制压力产生时,ATR被募集至适当位置,继而被激活。激活后的ATR在诸多方面发挥重要的调控作用,包括阻滞细胞周期、抑制复制起点、促进脱氧核苷酸的合成、启动复制叉等等。
与正常细胞不同,癌细胞由于存在DNA损伤应答的缺陷和/或复制压力的增加,其基因组具有不稳定性,这也正是癌症的一个关键标志。癌细胞有别于正常细胞的这些特点,使得癌细胞在***过程中,基因的突变、重排、拷贝数变化等事件更容易积累,进而有可能促进癌细胞的克隆进化;但与此同时,这些特点也是缺陷,会造成癌细胞特异性的脆弱性。充分利用这些脆弱性,有望增强抗癌治疗的效果,从而改善癌症患者的预后(Pilié.P.G.et al,2019,Nat.Rev.Clin.Oncol.16,81-104)。
癌细胞相较于正常细胞更依赖于ATR,这就是上述脆弱性之一。鉴于癌基因诱导的复制压力是癌症基因组不稳定性的主要来源,癌细胞的生存变得高度依赖于熟练完善的复制压力应答,因此容易受到ATR抑制剂的影响。细胞系和小鼠模型的早期工作已经为这一概念提供了临床前支持。此外,还有证据表明,ATR水平降低的小鼠对癌症的发展具有高度抵抗力。这些结果突出了在癌症治疗中靶向ATR的潜力。因此,开发高活性且具有良好成药性的ATR抑制剂,有望为人类提供抗击癌症的新武器(Lecona,E.et al,2018,Nat.Rev.Cancer 18,586–595)。
现有技术WO2014089379A1、WO2011154737A1、WO2016020320A1等分别公开了具有ATR激酶抑制活性的化合物,有数个ATR激酶抑制剂已经进入临床试验阶段,其中包括:
Figure PCTCN2022139864-appb-000001
此外,现有技术(ACS Med.Chem.Lett.2015,6,42-46)中还公开了以下具有ATR激酶抑制活性的化合物:
Figure PCTCN2022139864-appb-000002
但到目前为止,尚无ATR激酶抑制剂上市,因此本领域仍然亟需新的ATR抑制剂,特别是具有高活性以及良好成药性的ATR抑制剂。
发明内容
本发明的一个目的在于提供一种新的化合物,以作为ATR抑制剂。
本发明的另一目的在于提供所述化合物的制备方法。
本发明的另一目的在于提供所述化合物的应用。
本发明的另一目的在于提供包含所述化合物的药物组合物及其应用。
本发明的另一目的在于提供制备所述化合物的中间体。
本发明的另一目的在于提供所述中间体的制备方法。
一方面,本发明提供了一种具有结构式I的化合物或其药学上可接受的盐,
Figure PCTCN2022139864-appb-000003
其中:
Y 1、Y 2、Y 3、Y 4各自独立地为CH或N,并且,Y 1和Y 2中至少之一为N,Y 3和Y 4中至少之一为CH;
R 1为含n个杂原子的5元并6元杂芳环基或6元并6元杂芳环基、或含m个杂原子的5元或6元芳环基,其中,n选自1-4的整数,m选自0-3的整数,杂原子选自O、N、S中的一种或两种;所述含n个杂原子的5元并6元杂芳环基或6元并6元杂芳环基中,杂芳环基任选被选自卤素、-OH、-OMe、-NH 2、-NHMe、-NMe 2、-CN、-CONH 2、-Me中的一个或多个取代基所取代;所述含m个杂原子的5元或6元芳环基中,芳环基任选被选自4-6元杂环烷基、卤素、-OH、-OMe、-NH 2、-NHMe、-NMe 2、-CN、-CONH 2、-Me中的一个或多个取代基所取代;上述芳环基在被4-6元杂环烷基取代时,可以以单键相连,也可以彼此形成并环;
R 2为-OH、C 1-6烷氧基、-NR 3R 4、-CN、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 3-6环烷基或4-6元杂环烷基,杂原子选自O、N中的一种;所述C 1-6烷基、C 2-6烯基、C 2-6炔基、C 3-6环烷基或4-6元杂环烷基任选被选自卤素、-OH、C 1-6烷氧基、-NR 5R 6、-CN中的一个或多个取代基所取代;
R 3、R 4各自独立地为氢或C 1-6烷基;
R 5、R 6各自独立地为氢或C 1-6烷基,或者,-NR 5R 6一起代表氮杂环丁烷基、四氢吡咯基、哌啶基、吗啉基或4-甲基哌嗪基。
根据本发明的一些具体实施方案,本发明的所述式I化合物或其药学上可接受的盐中,Y 3和Y 4分别为CH;Y 1、Y 2各自独立地为CH或N,且Y 1与Y 2中至少之一为N。即,本发明的这些具体实施方案提供了如下式I-1所示结构的化合物:
Figure PCTCN2022139864-appb-000004
式I-1中,Y 1、Y 2、R 1、R 2各自独立地如前述所定义。
根据本发明的一些具体实施方案,本发明的所述式I化合物或其药学上可接受的盐中,Y 1和Y 2分别为N。
根据本发明的一些具体实施方案,本发明的所述式I化合物或其药学上可接受的盐中,Y 1和Y 2分别为N,Y 3和Y 4分别为CH。即,本发明的这些具体实施方案提供了如下式I-2所示结构的化合物:
Figure PCTCN2022139864-appb-000005
式I-2中,R 1、R 2各自独立地如前述所定义。
根据本发明的一些具体实施方案,本发明的所述式I化合物或其药学上可接受的盐中,R 1为含n个杂原子的5元并6元杂芳环基,n选自1-4的整数,杂原子选自O、N、S中的一种或两种;所述杂芳环基任选被选自卤素、-OH、-OMe、-NH 2、-NHMe、-NMe 2、-CN、-CONH 2、-Me中的一个或多个取代基所取代。
根据本发明的一些具体实施方案,本发明的所述式I化合物或其药学上可接受的盐中,R 1为选自以下的基团:
Figure PCTCN2022139864-appb-000006
Figure PCTCN2022139864-appb-000007
根据本发明的一些具体实施方案,本发明的所述式I化合物或其药学上可接受的盐中,R 2为羟基、甲氧基、乙氧基、氨基、甲胺基、二甲胺基、氰基、甲基、乙基、丙基、异丙基、乙烯基、丙烯基、烯丙基、乙炔基、丙炔基、炔丙基、环丙基、羟甲基、甲氧基甲基、2-羟乙基、2-甲氧基乙基、2-二甲胺基乙基、N-吗啉基或4-甲基哌嗪基。
根据本发明的一些具体实施方案,本发明的所述式I化合物或其药学上可接受的盐中,所述化合物可选自:
Figure PCTCN2022139864-appb-000008
Figure PCTCN2022139864-appb-000009
Figure PCTCN2022139864-appb-000010
另一方面,本发明还提供了一种中间体化合物,所述的中间体包括但不限于:上述制备所述的化合物或其药学上可接受的盐的合成路线中的任一中间体。优选地,所述中间体具有与式I的化合物、或其药学上可接受的盐相同的基本结构单元(基本相同的基本核心部分或者基本的环),或者中间体的基本结构单元包含在了式I的化合物、或其药学上可接受的盐的化学结构中。
在本发明的一些具体实施方案中,所述中间体化合物具有式II所示结构:
Figure PCTCN2022139864-appb-000011
式II中,Y 1、Y 2、Y 3、Y 4与R 2各自独立地如前述所定义;
R 3为卤素或
Figure PCTCN2022139864-appb-000012
其中所述卤素优选为氯。
另一方面,本发明还提供了一种制备式I所示结构的化合物或其药学上可接受的盐的方法,该方法包括:
Figure PCTCN2022139864-appb-000013
以式II所示的中间体化合物,与R 1-M发生偶联反应,制备具有结构式I的化合物。
其中:
R 3为卤素时,M为
Figure PCTCN2022139864-appb-000014
R 3
Figure PCTCN2022139864-appb-000015
时,M为卤素优选为溴;
Y 1、Y 2、Y 3、Y 4、R 1和R 2各自独立地如前述所定义。
根据本发明的具体实施方案,本发明的制备式I所示结构的化合物或其药学上可接受的盐的方法,还包括制备所述的中间体化合物的过程。
根据本发明的一些具体实施方案,本发明的制备式I所示结构的化合物或其药学上可接受的盐的方法,包括但不限于:按照制备实施例1至制备实施例27中任一合成路线制备得到式I的化合物或其药学上可接受的盐。其中,所制备得到的化合物的各取代基并不限于制备实施例1至制备实施例27中具体化合物的范围,各取代基的定义可参照前述式I化合物的各取代基的定义范围。
另一方面,本发明还提供了一种药物组合物,其包括:本发明所述的式I所示结构的化合物或其药学上可接受的盐,以及药学上可接受的载体。
本发明还提供了所述化合物或其药学上可接受的盐或所述药物组合物作为ATR激酶抑制剂的应用。本发明的化合物或其药学上可接受的盐或所述的药物组合物作为ATR抑制剂具体应用时,可以是非治疗目的的(如体外实验研究目的等),也可以是治疗目的的。即,本发明提供了所述的化合物或其药学上可接受的盐或所述的药物组合物在制备用于抑制ATR激酶的药物中的应用。
本发明还提供了所述的化合物或其药学上可接受的盐或所述的药物组合物在制备用于治疗过度增殖性疾病的药物中的应用。本发明所述的化合物或其药学上可接受的盐在作为药物活性成分用于治疗过度增殖性疾病时,包括向有需要的受试者给予所述的化合物或其药学上可接受的盐或所述的药物组合物的步骤。
本发明提供的所述应用中,其中所述过度增殖性疾病包括癌症。所述癌症例如可包括黑色素瘤、脑瘤、食管癌、胃癌、肝癌、胰腺癌、结肠直肠癌、肺癌、肾癌、乳腺癌、***、卵巢癌、***癌、皮肤癌、神经母细胞瘤、神经胶质瘤、肉瘤、骨癌、子宫癌、子宫内膜癌、头颈肿瘤、多发性骨髓瘤、B-细胞淋巴瘤、真性红细胞增多症、白血病、甲状腺肿瘤、膀胱癌或胆囊癌等。
定义和说明
除非另有说明,本文所用的下列术语和短语旨在具有下列含义。一个特定的术语或短语在没有特别定义的情况下不应该被认为是不确定的或不清楚的,而应该按照普通的含义去理解。 当本文中出现商品名时,意在指代其对应的商品或其活性成分。
术语“药学上可接受的”是针对那些化合物、材料、组合物和/或剂型而言,它们在可靠的医学判断的范围之内,适用于与人类和动物的组织接触使用,而没有过多的毒性、刺激性、过敏性反应或其它问题或并发症,与合理的利益/风险比相称。
术语“药学上可接受的盐”是指本发明化合物的盐,由本发明发现的具有特定取代基的化合物与相对无毒的酸或碱制备。当本发明的化合物中含有相对酸性的功能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的碱与这类化合物的中性形式接触的方式获得碱加成盐。药学上可接受的碱加成盐包括钠、钾、钙、铵、有机氨或镁盐或类似的盐。当本发明的化合物中含有相对碱性的官能团时,可以通过在纯的溶液或合适的惰性溶剂中用足够量的酸与这类化合物的中性形式接触的方式获得酸加成盐。药学上可接受的酸加成盐的实例包括无机酸盐,所述无机酸包括例如盐酸、氢溴酸、硝酸、碳酸、碳酸氢根、磷酸、磷酸-氢根、磷酸二氢根、硫酸、硫酸氢根、氢碘酸、亚磷酸等;以及有机酸盐,所述有机酸包括如乙酸、丙酸、异丁酸、马来酸、丙二酸、苯甲酸、琥珀酸、辛二酸、反丁烯二酸、乳酸、扁桃酸、邻苯二甲酸、苯磺酸、对甲苯磺酸、柠檬酸、酒石酸和甲磺酸等类似的酸;还包括氨基酸(如精氨酸等)的盐,以及如葡萄糖醛酸等有机酸的盐。本发明的某些特定的化合物含有碱性和酸性的官能团,从而可以被转换成任一碱或酸加成盐。
本发明的药学上可接受的盐可由含有酸根或碱基的母体化合物通过常规化学方法合成。一般情况下,这样的盐的制备方法是:在水或有机溶剂或两者的混合物中,经由游离酸或碱形式的这些化合物与化学计量的适当的碱或酸反应来制备。
除非另有说明,用楔形实线键
Figure PCTCN2022139864-appb-000016
和楔形虚线键
Figure PCTCN2022139864-appb-000017
表示一个立体中心的绝对构型,用直形实线键
Figure PCTCN2022139864-appb-000018
和直形虚线键
Figure PCTCN2022139864-appb-000019
表示立体中心的相对构型,用波浪线
Figure PCTCN2022139864-appb-000020
表示楔形实线键
Figure PCTCN2022139864-appb-000021
或楔形虚线键
Figure PCTCN2022139864-appb-000022
或用波浪线
Figure PCTCN2022139864-appb-000023
表示直形实线键
Figure PCTCN2022139864-appb-000024
和直形虚线键
Figure PCTCN2022139864-appb-000025
“任选”或“任选地”指的是随后描述的事件或状况可能但不是必需出现的,并且该描述包括其中所述事件或状况发生的情况以及所述事件或状况不发生的情况。
术语“被取代”是指特定原子上的任意一个或多个氢原子被取代基取代,可以包括重氢和氢的变体,只要特定原子的价态是正常的并且取代后的化合物是稳定的。当取代基为氧(即=O)时,意味着两个氢原子被取代。氧取代不会发生在芳香基上。术语“任选被取代”是指可以被取代,也可以不被取代,除非另有规定,取代基的种类和数目在化学上可以实现的基础上可以是任意的。
当任何变量(例如R)在化合物的组成或结构中出现一次以上时,其在每一种情况下的定义都是独立的。因此,例如,如果一个基团被0-2个R所取代,则所述基团可以任选地至多被两个R所取代,并且每种情况下的R都有独立的选项。此外,取代基和/或其变体的组合只有在这样的组合会产生稳定的化合物的情况下才是被允许的。
当其中一个变量选自键时,表示其连接的两个基团直接相连,比如A-L-Z中L代表键时表示该结构实际上是A-Z。
当其中一个变量选自缺失时,表示其不存在,比如C-R 3中R 3选自缺失时表示该结构实际上是C。
当所列举的取代基中没有指明其通过哪一个原子连接到被取代的基团上时,这种取代基可以通过其任何原子相键合,例如,苯基作为取代基可以通过苯环上任意一个碳原子连接到被取代的基团上。
除非另有规定,术语“烷基”用于表示直链或支链的饱和烃基,可以是单取代(如-CH 2F)或多取代的(如-CF 3),可以是一价(如甲基)、二价(如亚甲基)或者多价(如次甲基)。烷基的例子包括甲基(Me),乙基(Et),丙基(如,n-丙基和异丙基),丁基(如,n-丁基,异丁基,s-丁基,t-丁基),戊基(如,n-戊基,异戊基,新戊基)等。
除非另有规定,“环烷基”包括任何稳定的环状或多环烃基,任何碳原子都是饱和的,可以是单取代或多取代的,可以是一价、二价或者多价。这些环烷基的实例包括,但不限于,环丙基、降冰片烷基、[2.2.2]二环辛烷、[4.4.0]二环癸烷等。
除非另有规定,术语“烯基”表示具有至少一个碳碳双键的直链或支链烃基,“C 2-6烯基”表示具有2、3、4、5或6个碳原子的上述烃基、“C 2-4烯基”表示具有2、3或4个碳原子的上述烃基,应当理解,在所述烯基含有多于一个碳碳双键的情况下,所述双键可以彼此分离或彼此共轭。示例性“C 2-6烯基”包括但不限于,乙烯基、烯丙基、丁烯基、戊烯基、己烯基、丁二烯基、戊二烯基、己二烯基等。
除非另有规定,术语“炔基”表示具有至少一个碳碳三键的直链或支链烃基,“C 2-6炔基”表示具有2、3、4、5或6个碳原子的上述烃基、“C 2-4炔基”表示具有2、3或4个碳原子的上述烃基,示例性“C 2-6炔基”包括但不限于,乙炔基、丙炔基、丁炔基、戊炔基、己炔基、甲基丙炔基、4-甲基-1-丁炔基等。
除非另有规定,术语“卤素”本身或作为另一取代基的一部分表示氟(F)、氯(Cl)、溴(Br)或碘(I)原子。
除非另有规定,术语“烷氧基”表示烷基基团通过氧原子与分子其余部分相连,其中烷基基团具有如本发明所述含义。除非另有规定,C 1-5烷氧基包括C1、C2、C3、C4和C5的烷氧基。烷氧基的例子包括但不限于:甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、仲丁氧基、叔丁氧基、正戊氧基和S-戊氧基。所述烷氧基基团可以任选地被一个或多个本发明描述的取代基所取代。
除非另有规定,术语“胺基(氨基)”是指是-NH 2、-NH(烷基)或者-N(烷基)(烷基)。
除非另有规定,术语“芳环”表示多不饱和的芳香族烷烃单环,可以是单取代或多取代的。
除非另有规定,术语“4-6元杂环烷基”表示饱和的单价的单环烃环,其环上含有3、4或5个 碳原子,以及一或多个选自O、NR a的杂原子基团,其中,R a代表氢原子或C 1-6烷基;所述“4-6元杂环烷基”可以通过任何一个碳原子,或,如果存在的话,氮原子,与分子的其余部分相连接。
除非另有规定,术语“杂芳环”表示含有一至四个杂原子的芳环,杂原子选自N、O和S中的一种或多种。
除非另有规定,术语“稠杂芳环”表示含有两个以上芳香性的稠杂环。
除非另有规定,术语“5元并6元杂芳环”表示5元并6元的稠合杂芳环,2个并环中至少有1个环含有1个以上的杂原子(包括但不限于O、S或N),整个基团具有芳香性,非限制实施例包括了苯并5元杂芳基、6元杂芳环并5元杂芳环。
除非另有规定,术语“6元并6元杂芳环”表示6元并6元的稠合杂芳环,2个并环中至少有1个环含有1个以上的杂原子(包括但不限于O、S或N),整个基团具有芳香性,非限制实施例包括了苯并6元杂芳基、6元杂芳环并6元杂芳环。
除非另有规定,术语“药学上可接受的盐”或者“其药学上可接受的盐”是指本发明化合物保持游离酸或者游离碱的生物有效性和特性,且所述的游离酸通过与无毒的无机碱或者有机碱,所述的游离碱通过与无毒的无机酸或者有机酸反应获得的盐。
除非另有规定,术语“药物组合物”是指一种或多种本发明所述化合物或药学上可接受的盐与其它化学组分形成的混合物,其中,“其它化学组分”是指药学上可接受的载体、赋形剂和/或一种或多种其它治疗剂。“载体”是指不会对生物体产生明显刺激且不会消除所给予化合物的生物活性和特性的材料。“赋形剂”是指加入到药物组合物中以促进化合物给药的惰性物质。非限制性实施例包括碳酸钙、磷酸钙、糖、淀粉、纤维素衍生物(包括微晶纤维素)、明胶、植物油、聚乙二醇类、稀释剂、成粒剂、润滑剂、粘合剂和崩解剂。
本发明化合物具有良好的ATR酶抑制活性,以及对肿瘤细胞增殖的抑制活性,在治疗与细胞增殖相关的疾病中具有潜在的应用价值。
具体实施方式
下面将结合实施例对本发明的实施方案进行详细描述,但是本领域技术人员会理解,下列实施例仅用于说明本发明,而不应视为限制本发明的范围。实施例中未注明具体条件者,按照常规条件或制造商建议的条件进行。所用试剂或仪器未注明生产厂商者,均为可以通过市购获得的常规产品。
化合物的结构通过核磁共振或质谱来确定。核磁共振使用BRUKER 400M核磁仪测定,测定溶剂为氘代二甲基亚砜(DMSO-d 6)或氘代氯仿(CDCl 3),内标为四甲基硅烷(TMS),化学位移(δ)以10 -6(ppm)作为单位给出。质谱使用Waters ACQUITY Arc/ACQUITY QDa或Thermo U3000-ISQ EC液质联用仪测定。
高效液相色谱法分析使用Thermo U3000高压液相色谱仪。高效液相制备使用汉邦DAC-50或岛津LC-20AP制备型色谱仪。
反应监测使用薄层色谱或液质联用。薄层色谱使用的展开剂体系有:二氯甲烷和甲醇体系,石油醚和乙酸乙酯体系,溶剂的体积比根据化合物的极性不同而进行调节,或者加入少量三乙胺等进行调节。液质联用使用Waters ACQUITY Arc/ACQUITY QDa或Thermo U3000-ISQ EC液质联用仪。
柱层析一般使用200~300目硅胶为载体。洗脱剂的体系包括:二氯甲烷和甲醇体系,石油醚和乙酸乙酯体系,溶剂的体积比根据化合物的极性不同而进行调节,或者加入少量三乙胺等进行调节。
以下实施例中无特殊说明,反应温度为室温(20℃~30℃),溶剂均按照标准方法干燥纯化。
在常规的合成法以及本发明的实施例中,下述缩写的含义如下所示:
缩写                   含义
Pd(dppf)Cl 2            [1,1’-双(二苯基膦)二茂铁]二氯化钯
Pd(XantPhos)Cl 2        4,5-双二苯基膦-9,9-二甲基氧杂蒽二氯化钯
实施例1:(R)-3-甲基-4-(1’-(甲基磺酰基)-1H,1’H-[4,6’-二吡咯并[2,3-b]吡啶]-4’-基)吗啉(化合物1)
Figure PCTCN2022139864-appb-000026
第一步:4-溴-1H-吡咯并[2,3-b]吡啶-7-氧化物
将4-溴-1H-吡咯并[2,3-b]吡啶(10.0g,50.8mmol)溶于四氢呋喃(200mL)中,冷却至0℃,加入间氯过氧苯甲酸(17.5g,101.5mmol),然后在室温下搅拌反应4小时,有固体析出。过滤收集固体,将其溶解后经硅胶柱层析纯化,得到标题化合物(7.8g,收率72%)。
LC-MS(ESI)m/z 213.0,215.0[M+H] +.
第二步:4-溴-6-氯-1H-吡咯并[2,3-b]吡啶
将4-溴-1H-吡咯并[2,3-b]吡啶-7-氧化物(5.0g,23.5mmol)溶于N,N-二甲基甲酰胺(250mL)中,加热至50℃,加入甲磺酰氯(13.5g,118.0mmol),再加热至75℃搅拌反应1小时。将反应液冷却至室温,减压浓缩,经制备HPLC纯化,得到标题化合物(2.2g,收率41%)。
LC-MS(ESI)m/z 230.9,232.9[M+H] +.
第三步:4-溴-6-氯-1-(2-(三甲基硅基)乙氧基甲基)-1H-吡咯并[2,3-b]吡啶
将4-溴-6-氯-1H-吡咯并[2,3-b]吡啶(1.1g,4.8mmol)溶于四氢呋喃(35mL)中,在室温下缓慢加入60%的氢化钠(400mg,10.0mmol),搅拌反应5分钟,再加入2-(三甲基硅基)乙氧基甲基氯(2.5g,15.0mmol),继续在室温下搅拌反应,直至原料完全转化。加少量水淬灭反应,再加入二氯甲烷(100mL),然后用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经硅胶柱层析纯化,得到标题化合物(1.5g,收率87%)。
LC-MS(ESI)m/z 361.0,363.0[M+H] +.
第四步:(R)-4-(6-氯-1-(2-(三甲基硅基)乙氧基甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-3-甲基吗啉
将4-溴-6-氯-1-(2-(三甲基硅基)乙氧基甲基)-1H-吡咯并[2,3-b]吡啶(1.0g,2.8mmol)、(R)-3-甲基吗啉(280mg,2.8mmol)溶于1,4-二氧六环(20mL)中。用氮气除气并在氮气保护下,加入Pd(XantPhos)Cl 2(210mg,0.28mmol)和叔丁醇钠(270mg,2.8mmol),然后加热至80℃,在氮气保护下搅拌反应6小时。将反应液冷却至室温,加入二氯甲烷(100mL),再用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经硅胶柱层析纯化,得到标题化合物(437mg,收率41%)。
LC-MS(ESI)m/z 382.2[M+H] +.
第五步:(R)-4-(6-氯-1H-吡咯并[2,3-b]吡啶-4-基)-3-甲基吗啉
向反应瓶中加入(R)-4-(6-氯-1-(2-(三甲基硅基)乙氧基甲基)-1H-吡咯并[2,3-b]吡啶-4-基)-3-甲基吗啉(110mg,0.29mmol)和三氟乙酸(1mL)。该混合物在室温下搅拌反应,直至原料完全转化。将反应液减压浓缩,加入四氢呋喃(5mL)和1M氢氧化钠水溶液(2mL),所得混合物在室温下搅拌30分钟,再加入乙酸乙酯(50mL),然后用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,得到标题化合物粗品。对其不作进一步纯化,直接用于下一步。
LC-MS(ESI)m/z 252.1[M+H] +.
第六步:(R)-4-(6-氯-1-(甲基磺酰基)-1H-吡咯并[2,3-b]吡啶-4-基)-3-甲基吗啉
将上一步所得的(R)-4-(6-氯-1H-吡咯并[2,3-b]吡啶-4-基)-3-甲基吗啉粗品全部溶于四氢呋喃(6mL)中,在室温下缓慢加入60%的氢化钠(48mg,1.2mmol),搅拌反应5分钟,再逐滴加入甲磺酰氯(172mg,1.5mmol),继续在室温下搅拌反应3小时。加少量水淬灭反应,再加入乙酸乙酯(40mL),然后用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经硅胶柱层析纯化,得到标题化合物(76mg,收率80%)。
LC-MS(ESI)m/z 330.1[M+H] +.
第七步:(R)-3-甲基-4-(1’-(甲基磺酰基)-1H,1’H-[4,6’-二吡咯并[2,3-b]吡啶]-4’-基)吗啉
向反应瓶中加入(R)-4-(6-氯-1-(甲基磺酰基)-1H-吡咯并[2,3-b]吡啶-4-基)-3-甲基吗啉(50mg, 0.15mmol),4-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)-1H-吡咯并[2,3-b]吡啶(73.5mg,0.3mmol),Pd(dppf)Cl 2(22mg,0.03mmol)和碳酸钠(47.5mg,0.45mmol),再加入1,4-二氧六环(2.5mL)和水(0.5mL)。用氮气除气并在氮气保护下,将上述混合物加热至100℃,搅拌反应3小时。将反应液冷却至室温,加入二氯甲烷(20mL),再用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经制备HPLC纯化,得到化合物1(51mg,收率82%)。
LC-MS(ESI)m/z 412.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.77(s,1H),8.32(d,J=5.0Hz,1H),7.67(d,J=5.1Hz,1H),7.61(d,J=4.1Hz,1H),7.57(dd,J=3.5,2.5Hz,1H),7.31(s,1H),7.27(dd,J=3.4,1.9Hz,1H),6.98(d,J=4.2Hz,1H),4.42-4.34(m,1H),4.03-3.95(m,1H),3.84(dd,J=11.3,2.7Hz,1H),3.79(s,3H),3.73(d,J=11.3Hz,1H),3.69-3.59(m,2H),3.51(td,J=12.3,3.5Hz,1H),1.18(d,J=6.7Hz,3H).
13C NMR(100MHz,DMSO-d 6)δ153.11,152.00,150.79,149.64,143.44,139.42,127.84,123.59,118.25,114.68,111.58,105.64,104.16,101.94,71.45,67.08,51.42,43.70,42.51,13.89.
采用与实施例1第七步相类似的偶联反应,将4-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)-1H-吡咯并[2,3-b]吡啶换作其他芳香基硼酸频哪醇酯或芳香基硼酸,可由中间体(R)-4-(6-氯-1-(甲基磺酰基)-1H-吡咯并[2,3-b]吡啶-4-基)-3-甲基吗啉合成化合物2至化合物11。芳香基硼酸频哪醇酯为市售或采用Miyaura硼化反应由芳基卤代物与联硼酸频哪醇酯制备。芳香基硼酸为市售或由芳基卤代物与丁基锂、硼酸酯反应制备。
实施例2:(R)-4-(6-(1H-吲哚-4-基)-1-(甲基磺酰基)-1H-吡咯并[2,3-b]吡啶-4-基)-3-甲基吗啉(化合物2)
Figure PCTCN2022139864-appb-000027
按如上所示反应,得到化合物2(54mg,收率87%)。
LC-MS(ESI)m/z 411.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.26(s,1H),7.57(dd,J=7.4,0.9Hz,1H),7.55(d,J=4.1Hz,1H),7.48(d,J=8.1Hz,1H),7.44(t,J=2.8Hz,1H),7.24–7.18(m,2H),7.16(s,1H),6.94(d,J=4.2Hz,1H),4.36–4.28(m,1H),4.02–3.94(m,1H),3.87–3.78(m,4H),3.72(d,J=11.4Hz,1H),3.65(td,J=11.2,3.3Hz,1H),3.55(d,J=12.1Hz,1H),3.48(td,J=11.8,3.4Hz,1H),1.17(d,J=6.6Hz,3H).
实施例3:(R)-3-甲基-4-(1-(甲基磺酰基)-6-(1H-吡咯并[2,3-c]吡啶-4-基)-1H-吡咯并[2,3-b]吡啶-4-基)吗啉(化合物3)
Figure PCTCN2022139864-appb-000028
按如上所示反应,得到化合物3(102mg,收率82%)。
LC-MS(ESI)m/z 412.3[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.76(s,1H),8.79(s,1H),8.71(s,1H),7.71(t,J=2.8Hz,1H),7.56(d,J=4.2Hz,1H),7.32-7.27(m,1H),7.25(s,1H),6.96(d,J=4.2Hz,1H),4.41-4.33(m,1H),3.94-4.02(m,1H),3.83(dd,J=11.6,2.8Hz,1H),3.79(s,3H),3.73(d,J=11.5Hz,1H),3.69–3.59(m,2H),3.50(td,J=12.4,3.4Hz,1H),1.19(d,J=6.6Hz,3H).
实施例4:(R)-3-甲基-4-(1’-(甲基磺酰基)-1H,1’H-[5,6’-二吡咯并[2,3-b]吡啶]-4’-基)吗啉(化合物4)
Figure PCTCN2022139864-appb-000029
按如上所示反应,得到化合物4(50mg,收率80%)。
LC-MS(ESI)m/z 412.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.79(s,1H),9.02(d,J=2.1Hz,1H),8.66(d,J=2.2Hz,1H),7.55–7.48(m,2H),7.25(s,1H),6.91(d,J=4.2Hz,1H),6.56(dd,J=3.4,1.8Hz,1H),4.43–4.35(m,1H),3.98(dd,J=10.7,3.0Hz,1H),3.87–3.79(m,4H),3.73(d,J=11.6Hz,1H),3.69–3.60(m,2H),3.51(td,J=12.8,3.6Hz,1H),1.18(d,J=6.6Hz,3H).
实施例5:(R)-3-甲基-4-(1’-(甲基磺酰基)-1H,1’H-[3,6’-二吡咯并[2,3-b]吡啶]-4’-基)吗啉(化合物5)
Figure PCTCN2022139864-appb-000030
按如上所示反应,得到化合物5(53mg,收率85%)。
LC-MS(ESI)m/z 412.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ12.01(d,J=2.1Hz,1H),8.99(dd,J=7.9,1.7Hz,1H),8.37(d,J=2.8Hz,1H),8.27(dd,J=4.7,1.7Hz,1H),7.45(d,J=4.1Hz,1H),7.21–7.14(m,2H),6.87(d,J=4.2Hz,1H),4.38–4.29(m,1H),3.98(d,J=11.3Hz,1H),3.83(dd,J=11.3,3.0Hz,1H),3.77–3.69(m,4H),3.65(td,J=11.2,3.2Hz,1H),3.57–3.42(m,2H),1.16(d,J=6.6Hz,3H).
实施例6:(R)-3-甲基-4-(3-甲基-1’-(甲基磺酰基)-1H,1’H-[4,6’-二吡咯并[2,3-b]吡啶]-4’-基)吗啉(化合物6)
Figure PCTCN2022139864-appb-000031
按如上所示反应,得到化合物6(33mg,收率34%)。
LC-MS(ESI)m/z 426.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.47(s,1H),8.24(d,J=4.8Hz,1H),7.58(d,J=4.2Hz,1H),7.30–7.23(m,1H),7.08(d,J=4.8Hz,1H),6.98(d,J=4.2Hz,1H),6.82(s,1H),4.37–4.29(m,1H),3.97–3.89(m,1H),3.80(dd,J=11.3,2.8Hz,1H),3.74–3.67(m,4H),3.61(td,J=11.3,3.0Hz,1H),3.55–3.41(m,2H),1.94(s,3H),1.16(d,J=6.6Hz,3H).
实施例7:(R)-4-(6-(6-氟-1H-吲哚-4-基)-1-(甲基磺酰基)-1H-吡咯并[2,3-b]吡啶-4-基)-3-甲基吗啉(化合物7)
Figure PCTCN2022139864-appb-000032
按如上所示反应,得到化合物7(56mg,收率86%)。
LC-MS(ESI)m/z 429.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.32(s,1H),7.57(d,J=4.1Hz,1H),7.47(dd,J=11.2,2.3Hz,1H),7.45–7.43(m,1H),7.26(dd,J=9.5,2.2Hz,1H),7.23–7.20(m,1H),7.18(s,1H),6.95(d,J=4.2Hz,1H),4.40–4.32(m,1H),4.02–3.94(m,1H),3.83(dd,J=11.4,2.7Hz,1H),3.79(s,3H),3.72(d,J=11.2Hz,1H),3.69–3.57(m,2H),3.49(td,J=12.4,3.5Hz,1H),1.18(d,J=6.7Hz,3H).
实施例8:(R)-4-(6-(7-氟-1H-吲哚-4-基)-1-(甲基磺酰基)-1H-吡咯并[2,3-b]吡啶-4-基)-3-甲基吗啉(化合物8)
Figure PCTCN2022139864-appb-000033
按如上所示反应,得到化合物8(35mg,收率54%)。
LC-MS(ESI)m/z 429.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.75(s,1H),7.59–7.53(m,2H),7.51(t,J=2.7Hz,1H),7.33–7.29(m,1H),7.13(s,1H),7.04(dd,J=11.0,8.2Hz,1H),6.93(d,J=4.2Hz,1H),4.37–4.29(m,1H),4.02–3.94(m,1H),3.83(dd,J=11.3,2.9Hz,1H),3.79(s,3H),3.72(d,J=10.6Hz,1H),3.65(td,J=11.3,2.9Hz,1H),3.56(d,J=11.6Hz,1H),3.48(td,J=12.2,3.5Hz,1H),1.17(d,J=6.6Hz,3H).
实施例9:(R)-3-甲基-4-(1-(甲基磺酰基)-6-(1H-吡唑-5-基)-1H-吡咯并[2,3-b]吡啶-4-基)吗啉(化合物9)
Figure PCTCN2022139864-appb-000034
按如上所示反应,得到化合物9(45mg,收率82%)。
LC-MS(ESI)m/z 362.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ12.98(s,1H),7.82(s,1H),7.51(d,J=4.1Hz,1H),7.31(s,1H),6.90(d,J=4.2Hz,1H),6.83(s,1H),4.34–4.22(m,1H),3.98(d,J=10.8Hz,1H),3.93–3.77(m,4H),3.71(d,J=11.2Hz,1H),3.67–3.57(m,1H),3.50–3.39(m,2H),1.14(d,J=6.5Hz,3H).
实施例10:(R)-3-甲基-4-(1-(甲基磺酰基)-6-(1H-吡唑-4-基)-1H-吡咯并[2,3-b]吡啶-4-基)吗啉(化合物10)
Figure PCTCN2022139864-appb-000035
按如上所示反应,得到化合物10(43mg,收率78%)。
LC-MS(ESI)m/z 362.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ13.03(s,1H),8.35(s,1H),8.08(s,1H),7.44(d,J=4.1Hz,1H),6.98(s,1H),6.84(d,J=4.1Hz,1H),4.35–4.27(m,1H),3.96(d,J=13.1Hz,1H),3.85–3.77(m,4H), 3.71(d,J=11.3Hz,1H),3.62(td,J=11.5,3.3Hz,1H),3.51(d,J=12.1Hz,1H),3.45(dd,J=11.4,3.4Hz,1H),1.13(d,J=6.6Hz,3H).
实施例11:(R)-4-(6-(1H-苯并[d]咪唑-2-基)-1-(甲基磺酰基)-1H-吡咯并[2,3-b]吡啶-4-基)-3-甲基吗啉(化合物11)
Figure PCTCN2022139864-appb-000036
按如上所示反应,得到化合物11(23mg,收率40%)。
LC-MS(ESI)m/z 412.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ12.68(s,1H),7.73–7.68(m,2H),7.66–7.61(m,2H),7.30–7.19(m,2H),6.99(d,J=4.2Hz,1H),4.39–4.31(m,1H),4.06–3.96(m,4H),3.84(dd,J=11.5,2.5Hz,1H),3.74(d,J=11.2Hz,1H),3.65(td,J=11.0,3.8Hz,1H),3.59–3.50(m,2H),1.18(d,J=6.6Hz,3H).
实施例12:(R)-3-甲基-4-(1-(甲基磺酰基)-6-(7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡咯并[2,3-b]吡啶-4-基)-吗啉(化合物12)
Figure PCTCN2022139864-appb-000037
第一步:(R)-3-甲基-4-(1-(甲基磺酰基)-6-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)-1H-吡咯并[2,3-b]吡啶-4-基)吗啉
向反应瓶中加入(R)-4-(6-氯-1-(甲基磺酰基)-1H-吡咯并[2,3-b]吡啶-4-基)-3-甲基吗啉(200mg,0.61mmol),联硼酸频哪醇酯(230mg,0.91mmol),Pd(dppf)Cl 2(88mg,0.12mmol)和醋酸钾(101mg,1.03mmol),再加入1,4-二氧六环(4mL)。用氮气除气并在氮气保护下,将上述混合物加热至110℃,搅拌反应1.5小时。将反应液冷却至室温,加入乙酸乙酯(20mL),所得混合物用硅藻土过滤,滤液减压浓缩,得到标题化合物粗品(300mg)。对其不作进一步纯化,直接用于下一步。
第二步:(R)-3-甲基-4-(1-(甲基磺酰基)-6-(7H-吡咯并[2,3-d]嘧啶-4-基)-1H-吡咯并[2,3-b]吡啶-4-基)-吗啉
向反应瓶中加入(R)-3-甲基-4-(1-(甲基磺酰基)-6-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)-1H-吡咯并[2,3-b]吡啶-4-基)吗啉(200mg粗品,按0.40mmol计),4-溴-7H-吡咯并[2,3-d]嘧啶 (48mg,0.24mmol),Pd(dppf)Cl 2(51mg,0.07mmol)和碳酸钠(153mg,1.44mmol),再加入1,4-二氧六环(5mL)和水(1mL)。用氮气除气并在氮气保护下,将上述混合物加热至85℃,搅拌反应1小时。将反应液冷却至室温,加入二氯甲烷(25mL),再用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经制备HPLC纯化,得到化合物12(40mg,收率40%)。
LC-MS(ESI)m/z 413.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ12.19(s,1H),8.85(s,1H),8.01(s,1H),7.72–7.63(m,3H),7.02(d,J=4.2Hz,1H),4.40–4.32(m,1H),4.02(d,J=11.0Hz,1H),3.85(dd,J=11.5,2.8Hz,1H),3.79–3.70(m,4H),3.65(td,J=10.7,4.0Hz,1H),3.59–3.50(m,2H),1.18(d,J=6.6Hz,3H).
实施例13:(R)-4-(6-(3H-咪唑并[4,5-b]吡啶-7-基)-1-(甲基磺酰基)-1H-吡咯并[2,3-b]吡啶-4-基)-3-甲基吗啉(化合物13)
Figure PCTCN2022139864-appb-000038
按如上所示反应(与实施例12第二步相类似),得到化合物13(20mg,收率35%)。
LC-MS(ESI)m/z 413.1[M+H] +.
1H NMR(400MHz,CDCl 3)δ12.00(s,1H),8.66(d,J=5.1Hz,1H),8.41(s,1H),7.65(d,J=5.2Hz,1H),7.51(d,J=4.1Hz,1H),7.29(s,1H),6.74(d,J=4.1Hz,1H),4.31–4.23(m,1H),4.15–4.08(m,1H),3.97(dd,J=11.4,3.0Hz,1H),3.85(d,J=11.0Hz,1H),3.80(td,J=11.4,2.9Hz,1H),3.68(td,J=11.7,3.5Hz,1H),3.53(d,J=12.1Hz,1H),3.40(s,3H),1.34(d,J=6.7Hz,3H).
实施例14:(R)-4-(1’-(异丙基磺酰基)-1H,1’H-[4,6’-二吡咯并[2,3-b]吡啶]-4’-基)-3-甲基吗啉(化合物14)
Figure PCTCN2022139864-appb-000039
采用与实施例1第六步、第七步相类似的反应,将实施例1第六步中的甲磺酰氯换作异丙基磺酰氯,由中间体(R)-4-(6-氯-1H-吡咯并[2,3-b]吡啶-4-基)-3-甲基吗啉经如上所示的两步反应,得到化合物14(111mg,两步收率分别为92%、45%)。
LC-MS(ESI)m/z 440.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.74(s,1H),8.31(d,J=5.0Hz,1H),7.68(d,J=5.1Hz,1H),7.61–7.56(m,2H),7.36(dd,J=3.5,2.0Hz,1H),7.33(s,1H),7.00(d,J=4.2Hz,1H),4.45–4.35(m,2H),3.99(dd,J=11.7,2.5Hz,1H),3.83(dd,J=11.4,2.8Hz,1H),3.73(d,J=11.3Hz,1H),3.69–3.60(m,2H),3.51(td,J=12.7,3.4Hz,1H),1.29(d,J=6.9Hz,3H),1.23(d,J=6.8Hz,3H),1.19(d,J=6.6Hz,3H).
实施例15:(R)-4-(1’-(环丙基磺酰基)-1H,1’H-[4,6’-二吡咯并[2,3-b]吡啶]-4’-基)-3-甲基吗啉(化合物15)
Figure PCTCN2022139864-appb-000040
采用与实施例1第六步、第七步相类似的反应,将实施例1第六步中的甲磺酰氯换作环丙基磺酰氯,由中间体(R)-4-(6-氯-1H-吡咯并[2,3-b]吡啶-4-基)-3-甲基吗啉经如上所示的两步反应,得到化合物15(109mg,两步收率分别为90%、50%)。
LC-MS(ESI)m/z 438.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.75(s,1H),8.32(d,J=5.0Hz,1H),7.68(d,J=5.1Hz,1H),7.60–7.55(m,2H),7.35(dd,J=3.5,1.9Hz,1H),7.32(s,1H),6.96(d,J=4.2Hz,1H),4.41–4.33(m,1H),4.02–3.96(m,1H),3.84(dd,J=11.4,2.8Hz,1H),3.73(d,J=11.1Hz,1H),3.70–3.59(m,2H),3.51(td,J=12.5,3.7Hz,1H),3.47–3.39(m,1H),1.43–1.29(m,2H),1.19(d,J=6.6Hz,3H),1.14–1.06(m,2H).
实施例16:(R)-N,N-二甲基-4’-(3-甲基吗啉基)-1H,1’H-[4,6’-二吡咯并[2,3-b]吡啶]-1’-磺酰胺(化合物16)
Figure PCTCN2022139864-appb-000041
采用与实施例1第六步、第七步相类似的反应,将实施例1第六步中的甲磺酰氯换作二甲胺基磺酰氯,由中间体(R)-4-(6-氯-1H-吡咯并[2,3-b]吡啶-4-基)-3-甲基吗啉经如上所示的两步反应,得到化合物16(148mg,两步收率分别为91%、60%)。
LC-MS(ESI)m/z 441.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.74(s,1H),8.31(d,J=5.0Hz,1H),7.61(d,J=5.1Hz,1H),7.59–7.55(m,2H),7.28(dd,J=3.4,1.9Hz,1H),7.25(s,1H),6.91(d,J=4.2Hz,1H),4.39–4.31(m,1H),4.02–3.95(m,1H),3.84(dd,J=11.4,2.7Hz,1H),3.72(d,J=11.2Hz,1H),3.69–3.56(m,2H),3.50(td,J=12.0,3.4Hz,1H),2.89(s,6H),1.18(d,J=6.6Hz,3H).
实施例17:(R)-3-甲基-4-(7-(甲基磺酰基)-2-(1H-吡咯并[2,3-b]吡啶-4-基)-7H-吡咯并[2,3-d]嘧啶-4-基)吗啉(化合物17)
Figure PCTCN2022139864-appb-000042
第一步:(R)-4-(2-氯-7H-吡咯并[2,3-d]嘧啶-4-基)-3-甲基吗啉
将2,4-二氯-7H-吡咯并[2,3-d]嘧啶(1.00g,5.3mmol)和(R)-3-甲基吗啉(2.70g,26.7mmol)溶于1,4-二氧六环(25mL)中。缓慢加入三乙胺(3.80g,37.6mmol),再加热至90℃搅拌反应8小时。将反应液冷却至室温,加入二氯甲烷(100mL),再用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经硅胶柱层析纯化,得到标题化合物(1.02g,收率76%)。
LC-MS(ESI)m/z 253.1[M+H] +.
第二步:(R)-4-(2-氯-7-(甲基磺酰基)-7H-吡咯并[2,3-d]嘧啶-4-基)-3-甲基吗啉
将(R)-4-(2-氯-7H-吡咯并[2,3-d]嘧啶-4-基)-3-甲基吗啉(152mg,0.6mmol)溶于四氢呋喃(7.5mL)中,在室温下缓慢加入60%的氢化钠(48mg,1.2mmol),搅拌反应5分钟,再逐滴加入甲磺酰氯(137mg,1.2mmol)。继续在室温下搅拌反应,直至原料完全转化。加少量水淬灭反应,再加入二氯甲烷(100mL),然后用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经硅胶柱层析纯化,得到标题化合物(149mg,收率75%)。
LC-MS(ESI)m/z 331.1[M+H] +.
第三步:(R)-3-甲基-4-(7-(甲基磺酰基)-2-(1H-吡咯并[2,3-b]吡啶-4-基)-7H-吡咯并[2,3-d]嘧啶-4-基)吗啉
向反应瓶中加入(R)-4-(2-氯-7-(甲基磺酰基)-7H-吡咯并[2,3-d]嘧啶-4-基)-3-甲基吗啉(180mg,0.54mmol),4-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)-1H-吡咯并[2,3-b]吡啶(264mg,1.08mmol),Pd(dppf)Cl 2(80mg,0.11mmol)和碳酸钾(299mg,2.16mmol),再加入1,4-二氧六环(17.5mL)和水(3.5mL)。用氮气除气并在氮气保护下,将上述混合物加热至90℃,搅拌反应1小时。将反应液冷却至室温,加入二氯甲烷(100mL),再用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经制备HPLC纯化,得到化合物17(130mg,收率58%)。
LC-MS(ESI)m/z 413.1[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.79(s,1H),8.35(d,J=5.0Hz,1H),8.04(d,J=5.0Hz,1H),7.63–7.57(m,2H),7.48(dd,J=3.2,1.9Hz,1H),7.02(d,J=4.1Hz,1H),4.92–4.80(m,1H),4.62–4.50(m,1H),4.04(dd,J=11.1,3.0Hz,1H),3.85–3.71(m,5H),3.60(td,J=12.2,3.2Hz,1H),3.55–3.45(m,1H),1.36(d,J=6.8Hz,3H).
13C NMR(100MHz,DMSO-d 6)δ158.60,157.15,153.14,151.17,143.19,137.50,128.22,123.07,118.76,115.52,105.90,104.00,102.74,71.17,67.13,48.81,48.79,42.64,15.25.
采用与实施例17第三步相类似的偶联反应,将4-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)-1H-吡咯并[2,3-b]吡啶换作其他芳香基硼酸频哪醇酯或芳香基硼酸,可由中间体(R)-4-(2-氯-7-(甲基磺酰基)-7H-吡咯并[2,3-d]嘧啶-4-基)-3-甲基吗啉合成化合物18至化合物20以及化合物27。
实施例18:(R)-4-(2-(1H-吲哚-4-基)-7-(甲基磺酰基)-7H-吡咯并[2,3-d]嘧啶-4-基)-3-甲基吗啉(化合物18)
Figure PCTCN2022139864-appb-000043
按如上所示反应,得到化合物18(52mg,收率42%)。
LC-MS(ESI)m/z 412.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.26(s,1H),8.15(d,J=7.5Hz,1H),7.62–7.42(m,4H),7.21(t,J=7.7Hz,1H),6.97(d,J=4.1Hz,1H),4.92–4.77(m,1H),4.61–4.46(m,1H),4.09–3.99(m,1H),3.85–3.70(m,5H),3.65–3.55(m,1H),3.53–3.42(m,1H),1.35(d,J=6.7Hz,3H).
实施例19:(R)-3-甲基-4-(7-(甲基磺酰基)-2-(1H-吡咯并[2,3-c]吡啶-4-基)-7H-吡咯并[2,3-d]嘧啶-4-基)吗啉(化合物19)
Figure PCTCN2022139864-appb-000044
按如上所示反应,得到化合物19(81mg,收率65%)。
LC-MS(ESI)m/z 413.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.79(s,1H),9.17(s,1H),8.83(s,1H),7.74(t,J=2.6Hz,1H),7.56–7.51(m,2H),6.99(d,J=4.1Hz,1H),4.91–4.80(m,1H),4.61–4.48(m,1H),4.04(dd,J=10.8,2.9Hz,1H),3.85–3.71(m,5H),3.60(td,J=11.8,2.8Hz,1H),3.55–3.45(m,1H),1.36(d,J=6.7Hz,3H).
实施例20:(R)-4-(2-(6-氟-1H-吲哚-4-基)-7-(甲基磺酰基)-7H-吡咯并[2,3-d]嘧啶-4-基)-3-甲基吗啉(化合物20)
Figure PCTCN2022139864-appb-000045
按如上所示反应,得到化合物20(35mg,收率34%)。
LC-MS(ESI)m/z 430.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.33(s,1H),7.92(dd,J=11.5,2.5Hz,1H),7.59(br s,1H),7.55(d,J=4.0Hz,1H),7.48(t,J=2.8Hz,1H),7.32(dd,J=9.2,2.5Hz,1H),6.99(d,J=4.2Hz,1H),4.90–4.76(m,1H),4.59–4.46(m,1H),4.08–4.00(m,1H),3.85–3.71(m,5H),3.64–3.55(m,1H),3.53–3.43(m,1H),1.35(d,J=6.8Hz,3H).
实施例21:(R)-2-((4-(3-甲基吗啉基)-2-(1H-吡咯并[2,3-b]吡啶-4-基)-7H-吡咯并[2,3-d]嘧啶-7-基)磺酰基)乙醇(化合物21)
Figure PCTCN2022139864-appb-000046
第一步:(R)-4-(2-氯-7-((2-甲氧基乙基)磺酰基)-7H-吡咯并[2,3-d]嘧啶-4-基)-3-甲基吗啉
将(R)-4-(2-氯-7H-吡咯并[2,3-d]嘧啶-4-基)-3-甲基吗啉(200mg,0.79mmol)溶于四氢呋喃(5mL)中,在室温下缓慢加入60%的氢化钠(63mg,1.58mmol),搅拌反应5分钟,再逐滴加入2-甲氧基乙基磺酰氯(251mg,1.58mmol)。继续在室温下搅拌反应,直至原料完全转化。加少量水淬灭反应,再加入乙酸乙酯(50mL),然后用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经硅胶柱层析纯化,得到标题化合物(273mg,收率92%)。
LC-MS(ESI)m/z 375.1[M+H] +.
第二步:(R)-2-((2-氯-4-(3-甲基吗啉基)-7H-吡咯并[2,3-d]嘧啶-7-基)磺酰基)乙醇
将(R)-4-(2-氯-7-((2-甲氧基乙基)磺酰基)-7H-吡咯并[2,3-d]嘧啶-4-基)-3-甲基吗啉(270mg,0.72mmol)溶于二氯甲烷(20mL)中,冷却至0℃后加入三溴化硼(1.21g,4.83mmol),继续在室温下搅拌反应1小时。加入水(20mL)淬灭反应,减压浓缩除去二氯甲烷,再用乙酸乙酯(50mL)萃取,分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经硅胶柱层析纯化, 得到标题化合物(223mg,收率86%)。
LC-MS(ESI)m/z 361.1[M+H] +.
第三步:(R)-2-((4-(3-甲基吗啉基)-2-(1H-吡咯并[2,3-b]吡啶-4-基)-7H-吡咯并[2,3-d]嘧啶-7-基)磺酰基)乙醇
向反应瓶中加入(R)-2-((2-氯-4-(3-甲基吗啉基)-7H-吡咯并[2,3-d]嘧啶-7-基)磺酰基)乙醇(100mg,0.28mmol),4-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)-1H-吡咯并[2,3-b]吡啶(135mg,0.55mmol),Pd(dppf)Cl 2(40mg,0.055mmol)和醋酸钾(27mg,0.28mmol),再加入1,4-二氧六环(4mL)。用氮气除气并在氮气保护下,将上述混合物加热至85℃,搅拌反应4小时。将反应液冷却至室温,加入乙酸乙酯(50mL),再用水(20mL)洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经制备HPLC纯化,得到化合物21(49mg,收率40%)。
LC-MS(ESI)m/z 443.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.77(s,1H),8.34(d,J=5.0Hz,1H),8.04(d,J=5.0Hz,1H),7.60(t,J=3.0Hz,1H),7.54(d,J=4.1Hz,2H),7.51(dd,J=3.4,2.0Hz,1H),6.99(d,J=4.1Hz,1H),4.94–4.80(m,2H),4.62–4.48(m,1H),4.13(t,J=5.9Hz,2H),4.04(dd,J=11.3,3.5Hz,1H),3.81(d,J=11.5Hz,1H),3.78–3.68(m,3H),3.60(td,J=11.6,2.7Hz,1H),3.55–3.45(m,1H),1.36(d,J=6.7Hz,3H).
实施例22:(R)-3-甲基-4-(9-(甲基磺酰基)-2-(1H-吡咯并[2,3-b]吡啶-4-基)-9H-嘌呤-6-基)吗啉(化合物22)
Figure PCTCN2022139864-appb-000047
第一步:(R)-4-(2-氯-9H-嘌呤-6-基)-3-甲基吗啉
将2,6-二氯-9H-嘌呤(1.0g,5.3mmol)溶于N,N-二甲基甲酰胺(15mL)中,缓慢加入(R)-3-甲基吗啉(2.1g,21mmol)和三乙胺(5.4g,53mmol),再加热至120℃搅拌反应1小时。将反应液冷却,减压浓缩,残余物经硅胶柱层析纯化,得到标题化合物(1.2g,收率89%)。
LC-MS(ESI)m/z 254.1[M+H] +.
第二步:(R)-4-(2-氯-9-(甲基磺酰基)-9H-嘌呤-6-基)-3-甲基吗啉
将(R)-4-(2-氯-9H-嘌呤-6-基)-3-甲基吗啉(800mg,3.15mmol)溶于四氢呋喃(15mL)中,在室温下缓慢加入60%的氢化钠(505mg,12.6mmol),搅拌反应5分钟,再逐滴加入甲磺酰氯(1.44g,12.6mmol)。继续在室温下搅拌反应3小时。加少量水淬灭反应,再加入乙酸乙酯(50mL),然后用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经硅胶柱 层析纯化,得到标题化合物(785mg,收率75%)。
LC-MS(ESI)m/z 332.1[M+H] +.
第三步:(R)-3-甲基-4-(9-(甲基磺酰基)-2-(1H-吡咯并[2,3-b]吡啶-4-基)-9H-嘌呤-6-基)吗啉
向反应瓶中加入(R)-4-(2-氯-9-(甲基磺酰基)-9H-嘌呤-6-基)-3-甲基吗啉(100mg,0.30mmol),4-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)-1H-吡咯并[2,3-b]吡啶(183mg,0.75mmol),Pd(dppf)Cl 2(44mg,0.060mmol)和醋酸钾(118mg,1.20mmol),再加入1,4-二氧六环(10mL)和水(0.25mL)。用氮气除气并在氮气保护下,将上述混合物加热至85℃,搅拌反应1小时。将反应液冷却至室温,加入乙酸乙酯(20mL),再用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经制备HPLC纯化,得到化合物22(31mg,收率25%)。
LC-MS(ESI)m/z 414.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.83(s,1H),8.55(s,1H),8.36(d,J=5.0Hz,1H),8.04(d,J=5.0Hz,1H),7.63(t,J=3.0Hz,1H),7.39(dd,J=3.4,2.0Hz,1H),5.56–5.38(br,1H),5.18–5.06(br,1H),4.05(d,J=8.6Hz,1H),3.88(s,3H),3.84(d,J=11.2Hz,1H),3.75(dd,J=11.5,3.0Hz,1H),3.65–3.47(m,2H),1.40(d,J=6.8Hz,3H).
实施例23:(R)-3-甲基-4-(1-(甲基磺酰基)-6-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑并[3,4-d]嘧啶-4-基)吗啉(化合物23)
Figure PCTCN2022139864-appb-000048
第一步:(R)-4-(6-氯-1H-吡唑并[3,4-d]嘧啶-4-基)-3-甲基吗啉
将4,6-二氯-1H-吡唑并[3,4-d]嘧啶(0.40g,2.1mmol)溶于1,4-二氧六环(2mL)中,缓慢加入(R)-3-甲基吗啉(0.86g,8.5mmol)和三乙胺(2.14g,21.1mmol),再加热至120℃搅拌反应2小时。将反应液冷却,减压浓缩,残余物经硅胶柱层析纯化,得到标题化合物(0.50g,收率93%)。
LC-MS(ESI)m/z 254.1[M+H] +.
第二步:(R)-4-(6-氯-1-(甲基磺酰基)-1H-吡唑并[3,4-d]嘧啶-4-基)-3-甲基吗啉
将(R)-4-(6-氯-1H-吡唑并[3,4-d]嘧啶-4-基)-3-甲基吗啉(650mg,2.56mmol)溶于四氢呋喃(4mL)中,在室温下缓慢加入60%的氢化钠(205mg,5.13mmol),搅拌反应5分钟,再逐滴加入甲磺酰氯(587mg,5.12mmol)。继续在室温下搅拌反应1小时。加少量水淬灭反应,再加入乙酸乙酯(30mL),然后用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经硅胶柱层析纯化,得到标题化合物(765mg,收率90%)。
LC-MS(ESI)m/z 332.1[M+H] +.
第三步:(R)-3-甲基-4-(1-(甲基磺酰基)-6-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡唑并[3,4-d]嘧啶-4-基)吗啉
向反应瓶中加入(R)-4-(6-氯-1-(甲基磺酰基)-1H-吡唑并[3,4-d]嘧啶-4-基)-3-甲基吗啉(100mg,0.30mmol),4-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)-1H-吡咯并[2,3-b]吡啶(147mg,0.60mmol),Pd(dppf)Cl 2(44mg,0.060mmol)和氟化钾(70mg,1.20mmol),再加入1,4-二氧六环(5mL)。用氮气除气并在氮气保护下,将上述混合物加热至90℃,搅拌反应1小时。将反应液冷却至室温,加入乙酸乙酯(20mL),再用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经制备HPLC纯化,得到化合物23(19mg,收率15%)。
LC-MS(ESI)m/z 414.1[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.84(s,1H),8.70(s,1H),8.37(d,J=5.0Hz,1H),8.10(d,J=5.0Hz,1H),7.66–7.62(m,1H),7.55–7.51(m,1H),4.72–7.42(br,2H),4.07(d,J=8.4Hz,1H),3.84(d,J=12.0Hz,1H),3.77(d,J=12.0Hz,1H),3.70(s,3H),3.67–3.48(m,2H),1.40(d,J=6.7Hz,3H).
实施例24:(R)-3-甲基-4-(3-(甲基磺酰基)-5-(1H-吡咯并[2,3-b]吡啶-4-基)-3H-咪唑并[4,5-b]吡啶-7-基)吗啉(化合物24)
Figure PCTCN2022139864-appb-000049
第一步:(R)-4-(5-氯-3H-咪唑并[4,5-b]吡啶-7-基)-3-甲基吗啉
将5,7-二氯-3H-咪唑并[4,5-b]吡啶(500mg,2.66mmol)溶于N,N-二异丙基乙胺(9mL,51.7mmol)中,室温下加入(R)-3-甲基吗啉(3mL,26.4mmol),再加热至145℃搅拌反应8天。将反应液冷却至室温,加入乙酸乙酯(50mL),再用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经硅胶柱层析纯化,得到标题化合物(605mg,收率90%)。
LC-MS(ESI)m/z 253.1[M+H] +.
第二步:(R)-4-(5-氯-3-(甲基磺酰基)-3H-咪唑并[4,5-b]吡啶-7-基)-3-甲基吗啉
将(R)-4-(5-氯-3H-咪唑并[4,5-b]吡啶-7-基)-3-甲基吗啉(350mg,1.39mmol)溶于四氢呋喃(20mL)中,在室温下缓慢加入60%的氢化钠(180mg,4.50mmol),搅拌反应5分钟,再逐滴加入甲磺酰氯(518mg,4.52mmol)。继续在室温下搅拌反应,直至原料完全转化。加少量水淬灭反应,再加入二氯甲烷(100mL),然后用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经硅胶柱层析纯化,得到标题化合物(357mg,收率78%)。
LC-MS(ESI)m/z 331.1[M+H] +.
第三步:(R)-3-甲基-4-(3-(甲基磺酰基)-5-(1H-吡咯并[2,3-b]吡啶-4-基)-3H-咪唑并[4,5-b]吡啶-7-基)吗啉
向反应瓶中加入(R)-4-(5-氯-3-(甲基磺酰基)-3H-咪唑并[4,5-b]吡啶-7-基)-3-甲基吗啉(20mg,0.06mmol),4-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)-1H-吡咯并[2,3-b]吡啶(30mg,0.12mmol),Pd(dppf)Cl 2(9mg,0.012mmol)和氟化钾(20mg,0.344mmol),再加入1,4-二氧六环(2mL)和水(0.1mL)。用氮气除气并在氮气保护下,将上述混合物加热至85℃,搅拌反应2小时。将反应液冷却至室温,加入二氯甲烷(50mL),再用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经制备HPLC纯化,得到化合物24(11mg,收率44%)。
LC-MS(ESI)m/z 413.1[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.79(s,1H),8.51(s,1H),8.32(d,J=5.0Hz,1H),7.66(d,J=5.0Hz,1H),7.58(dd,J=3.4,2.5Hz,1H),7.30(s,1H),7.17(dd,J=3.4,1.9Hz,1H),5.25–5.14(m,1H),4.48–4.33(m,1H),4.02(dd,J=11.5,3.5Hz,1H),3.87(s,3H),3.79(d,J=2.2Hz,2H),3.64(td,J=11.7,2.7Hz,1H),3.45(td,J=12.7,3.6Hz,1H),1.26(d,J=6.7Hz,3H).
实施例25:(R)-3-甲基-4-(1-(甲基磺酰基)-6-(1H-吡咯并[2,3-c]吡啶-4-基)-1H-咪唑并[4,5-c]吡啶-4-基)吗啉(化合物25)
Figure PCTCN2022139864-appb-000050
(R)-4-(6-氯-1H-咪唑并[4,5-c]吡啶-4-基)-3-甲基吗啉按照文献(ACS Med.Chem.Lett.2015,6,42–46)所述合成路线制得。
第一步:(R)-4-(6-氯-1-(甲基磺酰基)-1H-咪唑并[4,5-c]吡啶-4-基)-3-甲基吗啉
将(R)-4-(6-氯-1H-咪唑并[4,5-c]吡啶-4-基)-3-甲基吗啉(150mg,0.59mmol)溶于四氢呋喃(10mL)中,在室温下缓慢加入60%的氢化钠(48mg,1.20mmol),搅拌反应5分钟,再逐滴加入甲磺酰氯(136mg,1.19mmol),继续在室温下搅拌反应,直至原料完全转化。加少量水淬灭反应,再加入乙酸乙酯(200mL),然后用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经硅胶柱层析纯化,得到标题化合物(150mg,收率76%)。
LC-MS(ESI)m/z 331.1[M+H] +.
第二步:(R)-3-甲基-4-(1-(甲基磺酰基)-6-(1H-吡咯并[2,3-c]吡啶-4-基)-1H-咪唑并[4,5-c]吡啶-4-基)吗啉
向反应瓶中加入(R)-4-(6-氯-1-(甲基磺酰基)-1H-咪唑并[4,5-c]吡啶-4-基)-3-甲基吗啉(50mg, 0.15mmol),(1H-吡咯并[2,3-c]吡啶-4-基)硼酸(49mg,0.30mmol),Pd(dppf)Cl 2(11mg,0.015mmol)和碳酸钠(32mg,0.30mmol),再加入1,4-二氧六环(1.5mL)和水(0.5mL)。用氮气除气并在氮气保护下,将上述混合物加热至80℃,搅拌反应2小时。将反应液冷却至室温,加入乙酸乙酯(50mL),再用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经制备HPLC纯化,得到化合物25(20mg,收率32%)。
LC-MS(ESI)m/z 413.1[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.79(s,1H),8.78(s,1H),8.66(s,1H),8.49(s,1H),7.73(t,J=2.7Hz,1H),7.64(s,1H),6.94–6.89(m,1H),5.48–5.38(m,1H),4.89(d,J=13.4Hz,1H),4.01(dd,J=11.1,2.7Hz,1H),3.83–3.72(m,5H),3.60(td,J=11.7,2.6Hz,1H),3.47–3.38(m,1H),1.31(d,J=6.7Hz,3H).
实施例26:(R)-3-甲基-4-(1-(甲基磺酰基)-6-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡咯并[3,2-c]吡啶-4-基)吗啉(化合物26)
Figure PCTCN2022139864-appb-000051
第一步:(R)-4-(6-氯-1H-吡咯并[3,2-c]吡啶-4-基)-3-甲基吗啉
将4,6-二氯-1H-吡咯并[3,2-c]吡啶(500mg,2.67mmol)溶于N,N-二异丙基乙胺(9mL,51.7mmol)中,室温下加入(R)-3-甲基吗啉(3mL,26.4mmol),再加热至145℃搅拌反应8天。将反应液冷却至室温,加入乙酸乙酯(50mL),再用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经硅胶柱层析纯化,得到标题化合物(603mg,收率90%)。
LC-MS(ESI)m/z 252.1[M+H] +.
第二步:(R)-4-(6-氯-1-(甲基磺酰基)-1H-吡咯并[3,2-c]吡啶-4-基)-3-甲基吗啉
将(R)-4-(6-氯-1H-吡咯并[3,2-c]吡啶-4-基)-3-甲基吗啉(550mg,2.19mmol)溶于四氢呋喃(20mL)中,在室温下缓慢加入60%的氢化钠(367mg,9.18mmol),搅拌反应5分钟,再逐滴加入甲磺酰氯(740mg,6.46mmol)。继续在室温下搅拌反应,直至原料完全转化。加少量水淬灭反应,再加入二氯甲烷(100mL),然后用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经硅胶柱层析纯化,得到标题化合物(591mg,收率82%)。
LC-MS(ESI)m/z 330.1[M+H] +.
第三步:(R)-3-甲基-4-(1-(甲基磺酰基)-6-(1H-吡咯并[2,3-b]吡啶-4-基)-1H-吡咯并[3,2-c]吡啶-4-基)吗啉
向反应瓶中加入(R)-4-(6-氯-1-(甲基磺酰基)-1H-吡咯并[3,2-c]吡啶-4-基)-3-甲基吗啉(20mg,0.06mmol),4-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)-1H-吡咯并[2,3-b]吡啶(30mg,0.12mmol),Pd(dppf)Cl 2(9mg,0.012mmol)和碳酸钠(20mg,0.19mmol),再加入1,4-二氧六环(1mL)和水(0.2mL)。用氮气除气并在氮气保护下,将上述混合物加热至100℃,搅拌反应2小时。将反应液冷却至室温,加入二氯甲烷(20mL),再用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经制备HPLC纯化,得到化合物26(10mg,收率40%)。
LC-MS(ESI)m/z 412.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.79(s,1H),8.31(d,J=5.0Hz,1H),7.93(s,1H),7.64–7.61(m,2H),7.59(t,J=3.0Hz,1H),7.08(d,J=3.8Hz,1H),6.90(dd,J=3.5,1.9Hz,1H),4.66–4.58(m,1H),4.08(d,J=12.8Hz,1H),4.02–3.96(m,1H),3.80(dd,J=11.4,2.9Hz,1H),3.73(d,J=10.8Hz,1H),3.63(td,J=11.3,2.6Hz,1H),3.58(s,3H),3.49(td,J=12.5,3.4Hz,1H),1.26(d,J=6.7Hz,3H).
实施例27:(R)-4-(2-(1H-苯并[d]咪唑-2-基)-7-(甲基磺酰基)-7H-吡咯并[2,3-d]嘧啶-4-基)-3-甲基吗啉(化合物27)
Figure PCTCN2022139864-appb-000052
按如上所示反应(与实施例17第三步相类似),得到化合物27(30mg,收率35%)。
LC-MS(ESI)m/z 413.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ9.19(s,1H),8.65(d,J=8.0Hz,1H),7.78(d,J=7.9Hz,1H),7.52(d,J=4.1Hz,1H),7.45–7.39(m,1H),7.38–7.32(m,1H),7.03(d,J=4.1Hz,1H),4.91–4.75(br,1H),4.65–4.49(br,1H),4.02(dd,J=11.2,3.0Hz,1H),3.80(d,J=11.5Hz,1H),3.77–3.70(m,4H),3.59(td,J=11.6,2.5Hz,1H),3.54–3.45(m,1H),1.37(d,J=6.8Hz,3H).
实施例28:(R)-N-甲基-1-(4-(3-甲基吗啉基)-7-(甲基磺酰基)-7H-吡咯并[2,3-d]嘧啶-2-基)-1H-苯并[d]咪唑-2-胺(化合物28)
Figure PCTCN2022139864-appb-000053
向反应瓶中加入(R)-4-(2-氯-7-(甲基磺酰基)-7H-吡咯并[2,3-d]嘧啶-4-基)-3-甲基吗啉(100mg,0.30mmol),N-甲基-1H-苯并[d]咪唑-2-胺(89mg,0.60mmol),三(二亚苄基丙酮)二钯(28mg, 0.031mmol),2-二环己基膦-2’,4’,6’-三异丙基联苯(29mg,0.061mmol)和碳酸铯(296mg,0.91mmol),再加入1,4-二氧六环(2mL)。用氮气除气并在氮气保护下,将上述混合物加热至85℃,搅拌反应1小时。将反应液冷却至室温,加入二氯甲烷(80mL),再用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经制备HPLC纯化,得到化合物28(28mg,收率21%)。
LC-MS(ESI)m/z 442.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ8.56(q,J=4.5Hz,1H),8.24(d,J=7.9Hz,1H),7.51(d,J=4.1Hz,1H),7.28(d,J=7.7Hz,1H),7.14–7.08(m,1H),7.05(d,J=4.1Hz,1H),7.04–6.98(m,1H),4.78–4.68(m,1H),4.41–4.31(m,1H),4.10–4.01(m,1H),3.86–3.72(m,2H),3.65(s,3H),3.63–3.51(m,2H),3.08(d,J=4.8Hz,3H),1.39(d,J=6.8Hz,3H).
实施例29:(R)-3-甲基-4-(2-(6-甲基-1H-吲哚-4-基)-7-(甲基磺酰基)-7H-吡咯并[2,3-d]嘧啶-4-基)吗啉(化合物29)
Figure PCTCN2022139864-appb-000054
第一步:(R)-3-甲基-4-(7-(甲基磺酰基)-2-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)-7H-吡咯并[2,3-d]嘧啶-4-基)吗啉
向反应瓶中加入(R)-4-(2-氯-7-(甲基磺酰基)-7H-吡咯并[2,3-d]嘧啶-4-基)-3-甲基吗啉(200mg,0.60mmol),联硼酸频哪醇酯(384mg,1.51mmol),Pd(dppf)Cl 2(88mg,0.12mmol)和醋酸钾(245mg,2.50mmol),再加入1,4-二氧六环(2mL)。用氮气除气并在氮气保护下,将上述混合物加热至100℃,搅拌反应2小时。将反应液冷却至室温,加入乙酸乙酯(80mL),所得混合物用硅藻土过滤,滤液减压浓缩,得到标题化合物粗品(252mg)。对其不作进一步纯化,直接用于下一步。
第二步:(R)-3-甲基-4-(2-(6-甲基-1H-吲哚-4-基)-7-(甲基磺酰基)-7H-吡咯并[2,3-d]嘧啶-4-基)吗啉
向反应瓶中加入(R)-3-甲基-4-(7-(甲基磺酰基)-2-(4,4,5,5-四甲基-1,3,2-二氧杂环戊硼烷-2-基)-7H-吡咯并[2,3-d]嘧啶-4-基)吗啉(126mg粗品,按0.30mmol计),4-溴-6-甲基-1H-吲哚(25mg,0.12mmol),Pd(dppf)Cl 2(17.3mg,0.024mmol)和碳酸钠(50mg,0.47mmol),再加入1,4-二 氧六环(5mL)和水(1mL)。用氮气除气并在氮气保护下,将上述混合物加热至85℃,搅拌反应2小时。将反应液冷却至室温,加入二氯甲烷(40mL),再用水洗涤。分液,有机相用无水硫酸钠干燥。过滤,滤液减压浓缩,经制备HPLC纯化,得到化合物29(26mg,收率51%)。
LC-MS(ESI)m/z 426.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.08(s,1H),7.96(d,J=1.5Hz,1H),7.51(d,J=4.1Hz,1H),7.50–7.47(m,1H),7.36(t,J=2.7Hz,1H),7.32(d,J=1.5Hz,1H),6.96(d,J=4.1Hz,1H),4.88–4.78(m,1H),4.60–4.50(m,1H),4.05(dd,J=11.2,3.5Hz,1H),3.86–3.71(m,5H),3.60(td,J=11.7,2.9Hz,1H),3.52–3.41(m,1H),2.48(s,3H),1.35(d,J=6.8Hz,3H).
实施例30:(R)-4-(4-(3-甲基吗啉基)-7-(甲基磺酰基)-7H-吡咯并[2,3-d]嘧啶-2-基)-1H-吲哚-6-甲腈(化合物30)
Figure PCTCN2022139864-appb-000055
按如上所示反应(与实施例29第二步相类似),得到化合物30(30mg,收率46%)。
LC-MS(ESI)m/z 437.1[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.87(s,1H),8.36(d,J=1.5Hz,1H),8.02(t,J=1.2Hz,1H),7.80(t,J=2.8Hz,1H),7.75–7.71(m,1H),7.57(d,J=4.1Hz,1H),7.01(d,J=4.2Hz,1H),4.90–4.78(m,1H),4.62–4.48(m,1H),4.11–4.01(m,1H),3.86–3.70(m,5H),3.60(td,J=11.5,11.1,2.5Hz,1H),3.54–3.44(m,1H),1.36(d,J=6.7Hz,3H).
实施例31:(R)-4-(2-(6-氯-1H-吲哚-4-基)-7-(甲基磺酰基)-7H-吡咯并[2,3-d]嘧啶-4-基)-3-甲基吗啉(化合物31)
Figure PCTCN2022139864-appb-000056
按如上所示反应(与实施例29第二步相类似),得到化合物31(18mg,收率27%)。
LC-MS(ESI)m/z 446.1[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.41(s,1H),8.08(d,J=2.0Hz,1H),7.59(t,J=2.5Hz,1H),7.57(d,J=1.6Hz,1H),7.55(d,J=4.1Hz,1H),7.52(t,J=2.8Hz,1H),6.99(d,J=4.1Hz,1H),4.88–4.76(m,1H),4.58–4.46(m,1H),4.05(dd,J=11.0,2.8Hz,1H),3.86–3.70(m,5H),3.60(td,J=11.8,2.4Hz,1H),3.54–3.43(m,1H),1.36(d,J=6.8Hz,3H).
实施例32:(R)-4-(2-(6-甲氧基-1H-吲哚-4-基)-7-(甲基磺酰基)-7H-吡咯并[2,3-d]嘧啶-4-基)-3-甲基吗啉(化合物32)
Figure PCTCN2022139864-appb-000057
按如上所示反应(与实施例29第二步相类似),得到化合物32(30mg,收率25%)。
LC-MS(ESI)m/z 442.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.04(s,1H),7.78(d,J=2.4Hz,1H),7.52(d,J=4.1Hz,1H),7.49–7.45(m,1H),7.31(t,J=2.7Hz,1H),7.06(dd,J=2.3,0.9Hz,1H),6.97(d,J=4.2Hz,1H),4.89–4.77(m,1H),4.57–4.45(m,1H),4.04(dd,J=11.0,3.0Hz,1H),3.83(s,3H),3.81–3.71(m,5H),3.60(td,J=11.6,2.8Hz,1H),3.53–3.43(m,1H),1.35(d,J=6.7Hz,3H).
实施例33:(R)-4-(7-(乙基磺酰基)-2-(1H-吡咯并[2,3-b]吡啶-4-基)-7H-吡咯并[2,3-d]嘧啶-4-基)-3-甲基吗啉(化合物33)
Figure PCTCN2022139864-appb-000058
按如上所示反应(与实施例17第二步、第三步相类似),得到化合物33(195mg,两步收率分别为92%、79%)。
LC-MS(ESI)m/z 427.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.78(s,1H),8.34(d,J=5.0Hz,1H),8.04(d,J=5.0Hz,1H), 7.63–7.57(m,2H),7.50(dd,J=3.4,2.0Hz,1H),7.03(d,J=4.1Hz,1H),4.92–4.80(m,1H),4.62–4.50(m,1H),4.09–3.95(m,3H),3.85–3.71(m,2H),3.61(td,J=11.6,2.7Hz,1H),3.55–3.45(m,1H),1.37(d,J=6.8Hz,3H),1.12(t,J=7.3Hz,3H).
实施例34:(R)-N,N-二甲基-4-(3-甲基吗啉基)-2-(1H-吡咯并[2,3-b]吡啶-4-基)-7H-吡咯并[2,3-d]嘧啶-7-磺酰胺(化合物34)
Figure PCTCN2022139864-appb-000059
按如上所示反应(与实施例17第二步、第三步相类似),得到化合物34(40mg,两步收率分别为13%、32%)。
LC-MS(ESI)m/z 442.2[M+H] +.
1H NMR(400MHz,DMSO-d 6)δ11.74(s,1H),8.33(d,J=5.0Hz,1H),7.99(d,J=5.0Hz,1H),7.62–7.56(m,2H),7.53(dd,J=3.4,2.0Hz,1H),6.96(d,J=4.1Hz,1H),4.90–4.80(m,1H),4.60–4.50(m,1H),4.08–4.00(m,1H),3.84–3.71(m,2H),3.65–3.56(m,1H),3.55–3.45(m,1H),2.92(s,6H),1.37(d,J=6.7Hz,3H).
对比例
分别合成表1中所示结构的化合物1’至化合物7’作为对比例化合物。
表1 对比例化合物
Figure PCTCN2022139864-appb-000060
Figure PCTCN2022139864-appb-000061
化合物1’为已知化合物,按照文献(ACS Med.Chem.Lett.2015,6,42–46)所述合成路线制得。
化合物2’按照以下合成路线制得:
Figure PCTCN2022139864-appb-000062
化合物3’按照以下合成路线制得:
Figure PCTCN2022139864-appb-000063
化合物4’按照以下合成路线制得:
Figure PCTCN2022139864-appb-000064
化合物5’按照以下合成路线制得:
Figure PCTCN2022139864-appb-000065
化合物6’按照以下合成路线制得:
Figure PCTCN2022139864-appb-000066
化合物7’为已知化合物,参照文献(ACS Med.Chem.Lett.2015,6,42–46)所述合成路线制得。化合物7’为两个非对映异构体的混合物(比例约为1:1),经HPLC手性拆分得到如下两个非对映异构体,核磁表征如下:
Figure PCTCN2022139864-appb-000067
非对映异构体一: 1H NMR(400MHz,Chloroform-d)δ8.84(s,1H),8.80(br s,1H),8.72(s,1H),8.04(s,1H),7.50(d,J=3.1Hz,1H),7.23(s,1H),7.09(d,J=3.1Hz,1H),5.62–5.55(m,1H),5.51(q,J=7.1Hz,1H),5.05(d,J=13.4Hz,1H),4.07(dd,J=11.2,3.4Hz,1H),3.91(dd,J=11.4,3.1Hz,1H),3.84(d,J=11.3Hz,1H),3.74(td,J=11.7,2.8Hz,1H),3.59(td,J=12.9,3.5Hz,1H),2.72(s,3H),2.11(d,J=7.1Hz,3H),1.44(d,J=6.8Hz,3H).
非对映异构体二: 1H NMR(400MHz,Chloroform-d)δ8.97(br s,1H),8.84(s,1H),8.72(s,1H),8.05(s,1H),7.50(d,J=3.2Hz,1H),7.23(s,1H),7.09(d,J=3.1Hz,1H),5.64–5.56(m,1H),5.52(q,J=7.0Hz,1H),5.02(d,J=13.5Hz,1H),4.07(dd,J=11.2,3.5Hz,1H),3.91(dd,J=11.4,3.1Hz,1H),3.84(d,J=11.3Hz,1H),3.75(td,J=11.8,2.8Hz,1H),3.58(td,J=12.9,3.6Hz,1H),2.69(s,3H),2.11(d,J=7.1Hz,3H),1.44(d,J=6.8Hz,3H).
生物学测试例
在常规的生物学测试以及以下测试例中,下述缩写的含义如下所示:
缩写                         含义
ATP                          腺嘌呤核苷三磷酸
HEPES                        4-羟乙基哌嗪乙磺酸
Brij-35                      聚氧乙烯月桂醚
EDTA                         乙二胺四乙酸
DMSO                         二甲基亚砜
测试例1:体外ATR激酶抑制实验
实验方法:利用Caliper EZ Reader基于微流体芯片技术的迁移率检测技术,在ATP浓度为K m的情况下,用迁移率改变法(Mobility Shift Assay)测定化合物在体外对ATR激酶的抑制活性。
实验材料和仪器:ATR激酶(Eurofins,产品目录号14-953);5-FAM-AK-17(吉尔生化,产品目录号524315);1倍激酶缓冲液(50mM HEPES pH 7.5,0.0015%Brij-35,1M MnCl 2);终止缓冲液(100mM HEPES pH 7.5,0.015%Brij-35,0.2%Coating Reagent#3,50mM EDTA);Caliper EZ Reader(Caliper Life Sciences)。
实验步骤:(1)将待测化合物用DMSO溶解并连续进行4倍稀释,共10个浓度;(2)使用Echo移液***移取0.06μL不同浓度的化合物至384孔板中;(3)用ATR激酶与1倍激酶缓冲液配成ATR激酶浓度为10nM的2倍激酶溶液;用5-FAM-AK-17、ATP与1倍激酶缓冲液配成5-FAM-AK-17浓度为6μM、ATP浓度为4μM的2倍底物溶液;(4)在上述384孔板中加入2倍激酶溶液10μL,室温下孵育10分钟;再加入2倍底物溶液10μL,28℃下孵育4小时;(5)加入终止缓冲液30μL以终止反应,再将384孔板放入Caliper EZ Reader,读取转化率数据。
数据处理:(1)通过转化率计算抑制率:“max”为DMSO空白对照组(未加化合物)的转化率,“min”为无酶对照组(未加ATR激酶)的转化率,“conversion”为化合物测试组的转化率,抑制率=(max-conversion)/(max-min)×100%;(2)使用Excel插件XLFit(版本号5.4.0.8)拟合曲线并计算IC 50值。
实验结果:实施例化合物对ATR激酶的抑制作用,如表2所示。
表2
Figure PCTCN2022139864-appb-000068
由上述结果可知,实施例化合物对ATR激酶具有良好的抑制作用。
对比例化合物1’至化合物6’对ATR激酶的抑制作用,如表3所示。
表3
对比例化合物编号 ATR IC 50(nM)
化合物1’ 56
化合物2’ 63
化合物3’ 31
化合物4’ 20
化合物5’ 15
化合物6’ 268
化合物7’ 28
测试例2:细胞增殖抑制实验
实验方法:采用CellTiter-Glo细胞活力检测试剂盒,测试化合物对人结肠癌细胞LoVo体外增殖的抑制作用。
实验材料:LoVo细胞(ATCC,产品目录号CCL-29);F-12K培养基(Invitrogen,产品目录号21127-022);胎牛血清(Biological Industries,产品目录号04-002-1A);GlutaMAX(Gibco,产品目录号35050-061);0.25%胰酶-EDTA(Invitrogen,产品目录号25200-072);CellTiter-Glo试剂盒(Promega,产品目录号G7558)。
实验步骤:(1)将F-12K培养基、胎牛血清、GlutaMAX配制成完全培养基,用于培养LoVo细胞;(2)取生长状态良好的LoVo细胞,用胰酶消化后转至离心管中,离心,弃去上清,加入新鲜培养基重悬细胞,用Vi-CELL XR细胞计数仪计数,将细胞悬液的密度调至25000~30000细胞/mL,在96孔细胞培养板中按100μL/孔加入细胞悬液,同时设未加细胞的空白对照组,将培养板置于二氧化碳培养箱中孵育过夜;(3)使用HP D300超微量加样器将待测化合物的DMSO溶液分配到上述96孔细胞培养板中,每孔加入0.5μL,每个化合物10个浓度(3倍稀释),同时设未加化合物的DMSO对照组,将培养板继续在二氧化碳培养箱中孵育96小时;(4)将培养板取出,平衡至室温,每孔加入CellTiter-Glo试剂100μL,避光振荡10分钟,孵育10分钟;(5)将培养板放入EnSpire读板,记录发光值RLU。
数据处理:(1)通过发光值计算抑制率:“RLU DMSO”为未加化合物的DMSO对照组的发光值,“RLU blank”为未加细胞的空白对照组的发光值,“RLU compound”为化合物测试组的发光值,抑制率=[1-(RLU compound-RLU blank)/(RLU DMSO-RLU blank)]×100%;(2)使用Excel插件XLFit拟合曲线并计算IC 50值。
实验结果:实施例化合物对LoVo细胞体外增殖的抑制作用,如表4所示。
表4
Figure PCTCN2022139864-appb-000069
由上述结果可知,本发明实施例化合物对LoVo细胞的增殖具有良好的抑制作用。
将本发明实施例化合物1对ATR激酶的抑制活性及对LoVo细胞增殖的抑制活性与对比例化合物4’、化合物5’进行比较,如表5所示。
表5
Figure PCTCN2022139864-appb-000070
以上所述仅是本发明的具体实施方式,应当指出,对于本技术领域的普通技术人员来说,在不脱离本发明原理的前提下,还可以做出若干改进和润饰,这些改进和润饰也应视为本发明的保护范围。

Claims (15)

  1. 一种具有结构式I的化合物或其药学上可接受的盐,
    Figure PCTCN2022139864-appb-100001
    其中:
    Y 1、Y 2、Y 3、Y 4各自独立地为CH或N,并且,Y 1和Y 2中至少之一为N,Y 3和Y 4中至少之一为CH;
    R 1为含n个杂原子的5元并6元杂芳环基或6元并6元杂芳环基、或含m个杂原子的5元或6元芳环基,其中,n选自1-4的整数,m选自0-3的整数,杂原子选自O、N、S中的一种或两种;所述含n个杂原子的5元并6元杂芳环基或6元并6元杂芳环基中,杂芳环基任选被选自卤素、-OH、-OMe、-NH 2、-NHMe、-NMe 2、-CN、-CONH 2、-Me中的一个或多个取代基所取代;所述含m个杂原子的5元或6元芳环基中,芳环基任选被选自4-6元杂环烷基、卤素、-OH、-OMe、-NH 2、-NHMe、-NMe 2、-CN、-CONH 2、-Me中的一个或多个取代基所取代;上述芳环基在被4-6元杂环烷基取代时,以单键相连或彼此形成并环;
    R 2为-OH、C 1-6烷氧基、-NR 3R 4、-CN、C 1-6烷基、C 2-6烯基、C 2-6炔基、C 3-6环烷基或4-6元杂环烷基,杂原子选自O、N中的一种;所述C 1-6烷基、C 2-6烯基、C 2-6炔基、C 3-6环烷基或4-6元杂环烷基任选被选自卤素、-OH、C 1-6烷氧基、-NR 5R 6、-CN中的一个或多个取代基所取代;
    R 3、R 4各自独立地为氢或C 1-6烷基;
    R 5、R 6各自独立地为氢或C 1-6烷基,或者,-NR 5R 6一起代表氮杂环丁烷基、四氢吡咯基、哌啶基、吗啉基或4-甲基哌嗪基。
  2. 根据权利要求1所述的化合物或其药学上可接受的盐,其中:
    Y 3和Y 4分别为CH;Y 1、Y 2各自独立地为CH或N,且Y 1与Y 2中至少之一为N。
  3. 根据权利要求1或2所述的化合物或其药学上可接受的盐,其中:
    Y 1和Y 2分别为N。
  4. 根据权利要求1-3任一项所述的化合物或其药学上可接受的盐,其中:
    R 1为含n个杂原子的5元并6元杂芳环基,n选自1-4的整数,杂原子选自O、N、S中的一种或两种;所述杂芳环基任选被选自卤素、-OH、-OMe、-NH 2、-NHMe、-NMe 2、-CN、-CONH 2、-Me中的一个或多个取代基所取代。
  5. 根据权利要求1-3任一项所述的化合物或其药学上可接受的盐,其中:
    R 1为选自以下的基团:
    Figure PCTCN2022139864-appb-100002
  6. 根据权利要求1-5任一项所述的化合物或其药学上可接受的盐,其中,
    R 2为-OH、C 1-3烷氧基、-NR 3R 4、-CN、C 1-4烷基、C 2-4烯基、C 2-4炔基、C 3-4环烷基或4-6元杂环烷基,杂原子选自O、N中的一种;所述C 1-4烷基、C 2-4烯基、C 2-4炔基、C 3-4环烷基或4-6元杂环烷基任选被选自卤素、-OH、C 1-3烷氧基、-NR 5R 6、-CN中的一个或多个取代基所取代;
    R 3、R 4各自独立地为氢或C 1-3烷基;
    R 5、R 6各自独立地为氢或C 1-3烷基,或者,-NR 5R 6一起代表氮杂环丁烷基、四氢吡咯基、哌啶基、吗啉基或4-甲基哌嗪基。
  7. 根据权利要求1-6任一项所述的化合物或其药学上可接受的盐,其中:
    R 2为羟基、甲氧基、乙氧基、氨基、甲胺基、二甲胺基、氰基、甲基、乙基、丙基、异丙基、乙烯基、丙烯基、烯丙基、乙炔基、丙炔基、炔丙基、环丙基、羟甲基、甲氧基甲基、2-羟乙基、2-甲氧基乙基、2-二甲胺基乙基、N-吗啉基或4-甲基哌嗪基。
  8. 根据权利要求1所述的化合物或其药学上可接受的盐,其中,所述化合物选自:
    Figure PCTCN2022139864-appb-100003
    Figure PCTCN2022139864-appb-100004
    Figure PCTCN2022139864-appb-100005
  9. 一种中间体化合物,其具有式II所示结构:
    Figure PCTCN2022139864-appb-100006
    式II中,Y 1、Y 2、Y 3、Y 4与R 2各自独立地如权利要求1-8任一项所定义;
    R 3为卤素或
    Figure PCTCN2022139864-appb-100007
    其中所述卤素优选为氯。
  10. 一种制备权利要求1-8任一项所述的化合物或其药学上可接受的盐的方法,该方法包括:
    Figure PCTCN2022139864-appb-100008
    以权利要求9所述的中间体化合物,与R 1-M发生偶联反应,制备具有结构式I的化合物;
    其中:
    R 3为卤素时,M为
    Figure PCTCN2022139864-appb-100009
    R 3
    Figure PCTCN2022139864-appb-100010
    时,M为卤素优选为溴;
    Y 1、Y 2、Y 3、Y 4、R 1和R 2各自独立地如权利要求1-8任一项中所定义。
  11. 根据权利要求10所述的方法,该方法还包括制备权利要求9所述的中间体化合物的过程。
  12. 一种药物组合物,其包括:权利要求1-8任一项所述的化合物或其药学上可接受的盐,以及药学上可接受的载体。
  13. 权利要求1-8任一项所述的化合物或其药学上可接受的盐或权利要求12所述的药物组合物在制备用于抑制ATR激酶的药物中的应用。
  14. 权利要求1-8任一项所述的化合物或其药学上可接受的盐或权利要求12所述的药物组合物在制备用于治疗过度增殖性疾病的药物中的应用。
  15. 根据权利要求14所述的应用,其中所述过度增殖性疾病包括癌症;
    所述癌症例如可包括黑色素瘤、脑瘤、食管癌、胃癌、肝癌、胰腺癌、结肠直肠癌、肺癌、肾癌、乳腺癌、***、卵巢癌、***癌、皮肤癌、神经母细胞瘤、神经胶质瘤、肉瘤、骨癌、子宫癌、子宫内膜癌、头颈肿瘤、多发性骨髓瘤、B-细胞淋巴瘤、真性红细胞增多症、白血病、甲状腺肿瘤、膀胱癌或胆囊癌。
PCT/CN2022/139864 2021-12-21 2022-12-19 取代的稠杂环化合物及其制备方法与应用 WO2023116594A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202280012432.7A CN116761804A (zh) 2021-12-21 2022-12-19 取代的稠杂环化合物及其制备方法与应用

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202111571407.6A CN116283960A (zh) 2021-12-21 2021-12-21 取代的稠杂环化合物及其制备方法与应用
CN202111571407.6 2021-12-21

Publications (1)

Publication Number Publication Date
WO2023116594A1 true WO2023116594A1 (zh) 2023-06-29

Family

ID=86783781

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/139864 WO2023116594A1 (zh) 2021-12-21 2022-12-19 取代的稠杂环化合物及其制备方法与应用

Country Status (2)

Country Link
CN (2) CN116283960A (zh)
WO (1) WO2023116594A1 (zh)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110257170A1 (en) * 2008-10-03 2011-10-20 Merck Serono Sa 4-morpholino-pyrido[3,2-d]pyrimidines
CN102850355A (zh) * 2011-06-29 2013-01-02 四川大学 9-磺酰基-9h-嘌呤衍生物及其制备方法和用途
CN103068391A (zh) * 2010-06-11 2013-04-24 阿斯利康(瑞典)有限公司 吗啉代嘧啶及其治疗用途
CN111205310A (zh) * 2018-11-22 2020-05-29 上海迪诺医药科技有限公司 杂环稠合嘧啶衍生物、其药物组合物及应用

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20110257170A1 (en) * 2008-10-03 2011-10-20 Merck Serono Sa 4-morpholino-pyrido[3,2-d]pyrimidines
CN103068391A (zh) * 2010-06-11 2013-04-24 阿斯利康(瑞典)有限公司 吗啉代嘧啶及其治疗用途
CN102850355A (zh) * 2011-06-29 2013-01-02 四川大学 9-磺酰基-9h-嘌呤衍生物及其制备方法和用途
CN111205310A (zh) * 2018-11-22 2020-05-29 上海迪诺医药科技有限公司 杂环稠合嘧啶衍生物、其药物组合物及应用

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
BARSANTI PAUL A, PAN YUE, LU YIPIN, JAIN RAMA, COX MATTHEW, AVERSA ROBERT J., DILLON MICHAEL P., ELLING ROBERT, HU CHENG, JIN XIAN: "Structure-Based Drug Design of Novel, Potent, and Selective Azabenzimidazoles (ABI) as ATR Inhibitors", ACS MEDICINAL CHEMISTRY LETTERS, AMERICAN CHEMICAL SOCIETY, US, vol. 6, no. 1, 8 January 2015 (2015-01-08), US , pages 42 - 46, XP093073759, ISSN: 1948-5875, DOI: 10.1021/ml500352s *

Also Published As

Publication number Publication date
CN116761804A (zh) 2023-09-15
CN116283960A (zh) 2023-06-23

Similar Documents

Publication Publication Date Title
TWI725266B (zh) Fgfr4抑制劑、其製備方法與藥學上的應用
RU2745035C1 (ru) Ингибитор fgfr и его применение
WO2020073945A1 (zh) 双环类衍生物抑制剂、其制备方法和应用
AU2009258115B2 (en) Diazacarbazoles and methods of use
CA2808543C (en) Pyrrolopyrimidine compounds and uses thereof
TW202110843A (zh) 含氮雜環類衍生物調節劑、其製備方法和應用
JP7131837B2 (ja) Csf-1r阻害活性を有するアザアリール誘導体、その製造方法および応用
TWI822868B (zh) Fgfr4抑制劑、包含其的藥物組合物及其用途
WO2011073263A1 (en) 1, 7 - diazacarbazoles and their use in the treatment of cancer
WO2021244609A1 (zh) 具有大环结构的化合物及其用途
JP2021503479A (ja) 置換ヘテロアリール化合物及び使用方法
CN115260187A (zh) 吡啶酮化合物及其用途
WO2023036252A1 (zh) 吡咯并嘧啶类或吡咯并吡啶类衍生物及其医药用途
WO2020221209A1 (zh) 一种cd73抑制剂,其制备方法和应用
TW202028195A (zh) 作為TGF-βR1抑制劑的化合物及其應用
WO2023116594A1 (zh) 取代的稠杂环化合物及其制备方法与应用
WO2015014283A1 (zh) 蛋白酪氨酸激酶抑制剂及其应用
TWI732249B (zh) Fgfr抑制劑、其製備方法和應用
JP2023530768A (ja) Fgfrおよびその突然変異阻害剤、その製造方法と応用
WO2020207419A1 (zh) 哌嗪酰胺衍生物,其制备方法及其在医药上的用途
US20150368244A1 (en) Diazacarbazoles and methods of use
CN115141202A (zh) 嘧啶并吡嗪酮化合物及其用途
CN111039941A (zh) 一种含氮杂环化合物、其制备方法及应用

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 202280012432.7

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22909907

Country of ref document: EP

Kind code of ref document: A1