WO2022121788A1 - 一种吡唑并氮杂卓类akt抑制剂 - Google Patents

一种吡唑并氮杂卓类akt抑制剂 Download PDF

Info

Publication number
WO2022121788A1
WO2022121788A1 PCT/CN2021/135266 CN2021135266W WO2022121788A1 WO 2022121788 A1 WO2022121788 A1 WO 2022121788A1 CN 2021135266 W CN2021135266 W CN 2021135266W WO 2022121788 A1 WO2022121788 A1 WO 2022121788A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
added
dihydro
solution
difluorophenyl
Prior art date
Application number
PCT/CN2021/135266
Other languages
English (en)
French (fr)
Inventor
马昌友
陈东晖
田禾
余方丹
周秋华
裴俊杰
苗雷
吴舰
Original Assignee
南京正大天晴制药有限公司
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 南京正大天晴制药有限公司 filed Critical 南京正大天晴制药有限公司
Priority to CN202180079206.6A priority Critical patent/CN116583525A/zh
Publication of WO2022121788A1 publication Critical patent/WO2022121788A1/zh

Links

Classifications

    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • 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
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/553Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one oxygen as ring hetero atoms, e.g. loxapine, staurosporine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains three hetero rings
    • C07D487/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/12Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains three hetero rings
    • C07D491/14Ortho-condensed systems
    • C07D491/147Ortho-condensed systems the condensed system containing one ring with oxygen as ring hetero atom and two rings with nitrogen as ring hetero atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/12Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D495/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/12Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
    • C07D498/14Ortho-condensed systems

Definitions

  • the invention belongs to the field of medicinal chemistry, and in particular relates to an AKT inhibitor, a preparation method thereof and medical use.
  • PI3K/AKT/mTOR pathway composed of phosphatidylinositol 3-kinase (PI3K) and its downstream protein AKT (also known as protein kinase B, PKB) and mammalian target of rapamycin (mTOR) is very important in cells.
  • Signal transduction pathway plays an extremely important biological function in the process of cell growth, survival, proliferation, apoptosis, angiogenesis, and autophagy. Aberrant activation of this pathway causes a range of diseases, including cancer, neuropathy, autoimmune diseases, and hemolymphatic disorders.
  • AKT a class of serine/threonine kinases, affects cell survival, growth, metabolism, proliferation, migration and differentiation through numerous downstream effectors.
  • AKT overactivation especially prostate cancer, pancreatic cancer, bladder cancer, ovarian cancer, and breast cancer.
  • AKT overactivation can lead to tumorigenesis, metastasis and drug resistance.
  • AKT has three subtypes: AKT1, AKT2, and AKT3.
  • each isoform consists of an amino-terminal PH domain (Pleckstrin homology domain), a central ATP-binding kinase domain, and a carboxy-terminal regulatory domain.
  • the targeted drugs for the PI3K/AKT/mTOR signaling pathway are mainly PI3K inhibitors and mTOR inhibitors, and AKT is at the core of this signal transduction pathway. Inhibition of AKT activity can not only avoid the serious side effects caused by the inhibition of upstream PI3K, but also avoid the negative feedback mechanism caused by the inhibition of downstream mTOR. Therefore, the search for effective and selective AKT inhibitors is an important direction for the development of current tumor-targeted drugs.
  • CN101631778A discloses a class of cyclopentadieno[D]pyrimidine derivatives
  • CN101578273A discloses a class of hydroxylated and methoxylated cyclopentadieno[D]pyrimidine derivatives
  • CN101511842A discloses a class of dihydrofuran Pyrimidine Derivatives
  • CN101970415A discloses a class of 5H-cyclopentadieno[d]pyrimidine derivatives, these compounds have an IC50 for inhibiting AKT1 of less than 10 ⁇ M.
  • the present invention provides the compound shown in formula I or a pharmaceutically acceptable salt thereof,
  • R 1 is selected from H or 2-(C 1 -C 4 alkyl)amino-2-oxoethyl
  • R 2 is selected from H, halogen or C 1 -C 4 alkyl
  • R is selected from halogen
  • X is selected from S, O or NH.
  • R 1 is selected from H or 2-(methylamino)-2-oxoethyl; in some typical embodiments, R 1 is selected from H.
  • R 2 is selected from chlorine, bromine, or methyl; in some typical embodiments, R 2 is selected from chlorine or bromine; in some more typical embodiments, R 2 is selected from bromine.
  • R 3 is selected from fluoro, chloro or bromo; in some typical embodiments, R 3 is selected from fluoro.
  • X is selected from S, O; in some typical embodiments, X is selected from S.
  • the aforementioned compound of formula I has the structure shown in formula II,
  • R 1 , R 2 , R 3 and X are as defined for compounds of formula I.
  • the present invention provides the following compounds or pharmaceutically acceptable salts thereof,
  • the present invention provides that a pharmaceutically acceptable salt is selected from the group consisting of hydrochloride or formate.
  • the present invention provides the following compounds,
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula I or a pharmaceutically acceptable salt thereof.
  • the present invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of a compound of formula I, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers.
  • compositions of the present invention may be administered by any suitable route or method, eg, orally or parenterally (eg, intravenously).
  • a therapeutically effective amount of a compound of formula I is from about 0.001 mg to 20 mg/Kg body weight/day, preferably from 0.01 mg to 10 mg/Kg body weight/day.
  • the pharmaceutical compositions of the present invention are typically provided in the form of tablets, capsules or solutions. Tablets may contain a compound of the present invention, or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier.
  • Such carriers include, but are not limited to, diluents, disintegrants, binders, lubricants, colorants or preservatives.
  • Capsules include hard capsules and soft capsules.
  • compositions of the present invention may be administered by intravenous injection, intramuscular injection or subcutaneous injection. It is usually provided as a sterile aqueous solution or suspension or as a lyophilized powder, adjusted for suitable pH and isotonicity.
  • the present invention also provides the use of a compound of formula I in the manufacture of a medicament for the prevention and/or treatment of AKT protein kinase mediated diseases or disease states.
  • the present invention also provides a method for preventing and/or treating an AKT protein kinase-mediated disease or disease state, comprising administering to an individual in need thereof a compound of formula I of the present invention or a pharmaceutical composition of the present invention .
  • the present invention also provides a compound of formula I of the present invention or a pharmaceutical composition of the present invention for use in the prevention and/or treatment of AKT protein kinase mediated diseases or disease states.
  • AKT protein kinase-mediated diseases or disease states include, but are not limited to, breast cancer, prostate cancer, or ovarian cancer.
  • the present invention provides a method for preparing a compound of formula I, including but not limited to the following synthetic schemes:
  • R 2 and X are as described above, wherein TBSO is tert-butyldimethylsiloxy.
  • the compound of formula I-1 and tert-butyldimethyl(2-propynyloxy)silane are prepared under the condition of catalyst (such as bis(triphenylphosphine)palladium dichloride and cuprous iodide) to prepare compound I-2;
  • catalyst such as bis(triphenylphosphine)palladium dichloride and cuprous iodide
  • compound I-2 is deprotected to obtain compound I-3
  • compound I-3 is prepared in the presence of a reducing agent (such as 10% wet palladium on carbon) to prepare compound I-4;
  • compound I-4 is subjected to bromination reaction to obtain compound I-5;
  • Compound I-5 reacts with 1H-pyrazole-3-boronic acid pinacol to obtain compound I-6;
  • compound I-6 undergoes hydroxy bromination to obtain compound I-7;
  • compound I-7 undergoes cyclization to obtain compound I-8 ;
  • I-8 compound is subjected to substitution reaction to prepare I-9 compound;
  • R 3 is as defined above, and R 4 is selected from C 1 -C 4 alkyl.
  • Compound II-1 reacts with (formylmethylene) triphenylphosphine to obtain compound II-2; compound II-2 reacts with tert-butyl 2-nitroethylcarbamate to obtain compound II-3; II -3 compound under the action of catalyst (such as triethylsilane, trifluoroacetic acid) to obtain II-4 compound; II-4 compound and di-tert-butyl dicarbonate, react to obtain II-5 compound; II-5 compound in reducing agent (such as 10% palladium hydroxide) to obtain compound II-6; compound II-3 reacts with allyltrimethylsilane to obtain compound II-7; compound II-7 undergoes oxidation reaction to obtain compound II-8; II The amide-forming reaction of the -8 compound gives the II-9 compound; the II-9 compound reduces the nitro group to obtain the II-10 compound.
  • catalyst such as triethylsilane, trifluoroacetic acid
  • R 1 , R 2 and X are as described in formula I above.
  • R 2 , R 3 and X are as described in the preceding formula I, and R 4 is selected from C 1 -C 4 alkyl groups.
  • the "compounds" of the present invention may be asymmetric, eg, having one or more chiral centers. Unless otherwise specified, a “compound” of the present invention refers to any one stereoisomer or a mixture of two or more stereoisomers. Stereoisomers include, but are not limited to, enantiomers and diastereomers.
  • the compounds of the present invention containing asymmetric carbon atoms can be isolated in optically pure form or as a mixture of two or more stereoisomers. Optically pure forms can be resolved from mixtures of two or more stereoisomers, or synthesized by using chiral starting materials or chiral reagents.
  • alkyl refers to saturated aliphatic hydrocarbon groups, including straight or branched chain saturated hydrocarbon groups, having the indicated number of carbon atoms.
  • C1-C4 alkyl includes C1 alkyl, C2 alkyl, C3 alkyl and C4 alkyl, examples include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl , isobutyl, tert-butyl, etc.
  • halogen refers to fluorine, chlorine, bromine and iodine.
  • hydroxy refers to -OH.
  • pharmaceutically acceptable salts refers to salts that retain the biological efficacy of the free acids and bases of a specified compound without biologically adverse effects. Examples are acid (including organic and inorganic acid) addition salts or base addition salts (including organic and inorganic bases).
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the acid or base containing parent compound 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 the two.
  • ⁇ ективное amount refers to a nontoxic but sufficient amount of a drug or agent to achieve the desired effect.
  • pharmaceutically acceptable carrier refers to those carriers which are not significantly irritating to the body and which do not impair the biological activity and properties of the active compound. Including but not limited to any diluents, disintegrants, binders, glidants, wetting agents that can be used in humans or animals and approved by the State Drug Administration.
  • TK tyrosine kinase
  • the compounds of the present invention can also be conveniently prepared by optionally combining various synthetic methods described in this specification or known in the art, and such combinations can be easily performed by those skilled in the art.
  • Reaction conditions a) ethyl 5-bromothiophene-2-carboxylate, tert-butyldimethyl(2-propynyloxy)silane, bis(triphenylphosphine)palladium dichloride, cuprous iodide, Triethylamine; b) tetrabutylammonium fluoride, tetrahydrofuran; c) hydrogen, 10% wet palladium on carbon, methanol; d) aluminum trichloride, liquid bromine, dichloromethane; e) 1H-pyrazole-3- pinacol borate, [1,1'-bis(diphenylphosphino)ferrocene]palladium(II) chloride, cesium carbonate, N,N-dimethylformamide, water; f) triphenylphosphine , carbon tetrabromide, dichloromethane; g) cesium carbonate, potassium iodide,
  • reaction solution was poured into saturated sodium thiosulfate solution (50 mL) and stirred for 10 min, then dichloromethane (2 ⁇ 50 mL) was added, the organic phase was washed with saturated sodium chloride solution (2 ⁇ 50 mL), dried, spin-dried, and the crude product was layered
  • the organic phase was washed with saturated sodium chloride solution (2 ⁇ 50 mL), dried over anhydrous sodium sulfate, and spin-dried.
  • Reaction conditions a) 3,4-difluorobenzaldehyde, (formylmethylene) triphenylphosphine, N,N dimethylformamide; b) (2S)-2-[diphenyl[(triphenylphosphine) Methylsilyl ester)oxy]methyl]-pyrrolidine, tert-butyl 2-nitroethylcarbamate, benzoic acid, dichloromethane; c)(4S,5S)4-(3,4-difluoro Phenyl)-2-hydroxy-5-nitropiperidine-1-carboxylic acid tert-butyl ester, triethylsilane, trifluoroacetic acid, dichloromethane; d) di-tert-butyl dicarbonate, triethylamine, dichloromethane Methane; e) hydrogen, 10% palladium hydroxide, ethanol.
  • the organic phase was washed with saturated sodium chloride solution (2 ⁇ 100 mL), dried over anhydrous sodium sulfate, and spin-dried.
  • Reaction conditions a) N-chlorosuccinimide, tetrahydrofuran; b) sodium hydroxide, methanol, water; c) 1-chloro-6,7-dihydro-5hydro-pyrazole[1,5- a] Thiophene[3,2-c]azepine-9-carboxylic acid, N,N-diisopropylethylamine, 2-(7-benzotriazole oxide)-N,N,N',N '-tetramethylurea hexafluorophosphate, dichloromethane; d) hydrogen chloride solution in dioxane, dichloromethane.
  • reaction solution was added to water (100 mL), and then ethyl acetate (2 ⁇ 100 mL) was added for extraction.
  • organic phase was washed with saturated sodium chloride solution (2 ⁇ 100 mL), dried over anhydrous sodium sulfate, and spin-dried.
  • Reaction conditions a) 6,7-dihydro-5H-pyrazolo[1,5-a]thieno[3,2-c]azepine-9-carboxylic acid ethyl ester, N-bromosuccinyl imine, tetrahydrofuran; b) sodium hydroxide, methanol, water; c) tert-butyl (3S,4S)3-amino-4-(3,4-difluorophenyl)piperidine-1-carboxylate, N, N-diisopropylethylamine, 2-(7-benzotriazole oxide)-N,N,N',N'-tetramethylurea hexafluorophosphate, dichloromethane; d) hydrogen chloride dioxygen Hexacyclic solution, dichloromethane;
  • reaction solution was added to water (100 mL), and then ethyl acetate (2 ⁇ 100 mL) was added for extraction.
  • organic phase was washed with saturated sodium chloride solution (2 ⁇ 100 mL), dried over anhydrous sodium sulfate, and spin-dried.
  • the product was isolated by preparation to obtain 50 mg of the product as a white solid.
  • reaction solution was added to water (100 mL), and then ethyl acetate (2 ⁇ 100 mL) was added for extraction.
  • organic phase was washed with saturated sodium chloride solution (2 ⁇ 100 mL), dried over anhydrous sodium sulfate, and spin-dried.
  • the product was isolated by preparation to obtain 80 mg of the product as a white solid.
  • Reaction conditions a) allyl trimethylsilane, boron trifluoride ether, dichloromethane; b) ruthenium trichloride, sodium periodate, dichloromethane, acetonitrile, water; c) 2M methylamine tetrahydrofuran solution ; N,N-diisopropylethylamine, 2-(7-benzotriazole oxide)-N,N,N',N'-tetramethylurea hexafluorophosphate, dichloromethane; d) Hydrogen, 10% palladium hydroxide, ethanol.
  • the organic phase was washed with saturated sodium chloride solution (2 ⁇ 100 mL), dried over anhydrous sodium sulfate, and spin-dried.
  • reaction solution was added to water (100mL), and then ethyl acetate (2 ⁇ 100mL) was added for extraction.
  • the organic phase was saturated with Washed with sodium chloride solution (2 ⁇ 100 mL), dried over anhydrous sodium sulfate, and spin-dried.
  • reaction solution was added to water (100 mL), and then ethyl acetate (2 ⁇ 100 mL) was added for extraction.
  • organic phase was washed with saturated sodium chloride solution (2 ⁇ 100 mL), dried over anhydrous sodium sulfate, and spin-dried.
  • Reaction conditions a) iodomethane, potassium carbonate, N,N-dimethylformamide; b) tert-butyldimethyl(2-propynyloxy)silane, [1,1'-bis(diphenyl) phosphine) ferrocene] palladium dichloride, cuprous iodide, triethylamine; c) tetrabutylammonium fluoride/tetrahydrofuran solution (1.0 M); d) hydrogen, 10% wet palladium on carbon, anhydrous ethanol; e) liquid bromine, aluminum trichloride, dichloromethane; f) 1H-pyrazole-3-boronic acid pinacol, bis(tri-tert-butylphosphine)palladium, sodium bicarbonate, dioxane, water; g ) carbon tetrabromide, triphenylphosphine, dichloromethane; h) cesium carbonate
  • Reaction conditions a) N-chlorosuccinimide, tetrahydrofuran; b) sodium hydroxide, methanol, water; c) (3S,4S)-3-amino-4-(3,4-difluorophenyl) ) tert-butyl piperidine-1-carboxylate, 2-(7-benzotriazole oxide)-N,N,N',N'-tetramethylurea hexafluorophosphate, diisopropylethylamine , dichloromethane; d) hydrogen chloride/1,4-dioxane solution (4.0 M).
  • Reaction conditions a) N-bromosuccinimide, tetrahydrofuran; b) sodium hydroxide, methanol, water; c) (3S,4S)-3-amino-4-(3,4-difluorophenyl) ) Piperidine-1-carboxylate tert-butyl ester, 2-(7-benzotriazole oxide)-N,N,N',N'-tetramethylurea hexafluorophosphate, diisopropylethylamine , dichloromethane; d) hydrogen chloride/1,4-dioxane solution (4.0 M).
  • Reaction conditions a) methylboronic acid, [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride, potassium carbonate, N,N-dimethylformamide; b) sodium hydroxide , methanol, water; c) (3S,4S)-3-amino-4-(3,4-difluorophenyl)piperidine-1-carboxylate tert-butyl ester, 2-(7-benzotriazepine oxide azole)-N,N,N',N'-tetramethylurea hexafluorophosphate, diisopropylethylamine, dichloromethane; d) hydrogen chloride/1,4-dioxane solution (4.0M) .
  • the reaction solution was poured into water (100 mL), and then ethyl acetate (100 mL x 2) was added for extraction.
  • the organic phase was washed with saturated sodium chloride solution (100 mL x 2), dried over anhydrous sodium sulfate, and spin-dried.
  • AKT1 (Item #01-101, Carna)
  • AKT3 (Item #PV3185, Invitrogen)
  • the final concentration is 1x Kinase Reaction Buffer: 14 mL of Kinase AKT1, 2, 3 in 1x Kinase Reaction Buffer is prepared by containing 2800 ⁇ L 5x Kinase Reaction Buffer, 140 ⁇ L 100 mM MgCl 2 , 140 ⁇ L 100 mM DTT, 10920 L ultrapure water.
  • Compound stock solutions (10 mM in DMSO) were first diluted to 100 [mu]M compound solutions with DMSO and then to 2.5 [mu]M compound working solutions (2.5% in DMSO) with 1x Kinase Reaction Buffer. Prepare a 2.5% DMSO solution using 1x Kinase Reaction Buffer, then dilute 2.5 ⁇ M compound working solution with a buffered reaction solution containing 2.5% DMSO at a dilution ratio of 3.16 7 times for a total of 8 concentrations (as shown in Table 2). ).
  • SA-XL665 and STK Antibody-Cryptate were formulated in assay buffer to the above table concentrations.
  • Enzyme preparation see Table 3, respectively 1.67 ⁇ final concentration of enzyme solution, add 6 ⁇ L to each well using an automatic liquid dispenser, and then spray the compound using a compound titrator; premix for 15 minutes;
  • ATP and substrate preparation see Table 3, prepare 5 ⁇ ATP and 5 ⁇ substrate respectively, after mixing the two, add 4 ⁇ L to each well using an automatic dispenser;
  • Example 1 Compound number AKT1/nM AKT2/nM AKT3/nM Example 1 ⁇ 0.01 0.71 1.12 Example 2 ⁇ 0.01 1.06 0.66 Example 3 0.24 1.12 4.29 Example 4 0.24 1.18 1.60 Example 5 1.00 15.35 10.45 Example 6 1.20 11.04 5.09 Example 7 2.40 26.35 23.96 Example 8 2.11 20.22 12.63 GDC-0068 5.96 43.04 51.23
  • Example 10 Inhibition test of the compound of the present invention on LNCaP cell proliferation
  • LNCaP human prostate cancer cells, purchased from Nanjing Kebai Biotechnology Co., Ltd.
  • medium: 1640+15% FBS (purchased from Gibco) and cultured in a 37° C., 5% CO 2 incubator.
  • the above cells in the logarithmic growth phase were plated in a 96-well plate at a cell density of 6000 cells/well, and a blank control group was set at the same time.
  • the signal value of the test substance the mean value of the fluorescence signal of the cell + medium + compound group;
  • Signal value of blank group mean value of fluorescence signal of medium group (containing 0.5% DMSO);
  • Signal value of negative control group mean value of fluorescence signal of cell + medium group (containing 0.5% DMSO).
  • Example 11 Inhibition test of the compounds of the present invention on the proliferation of TOV21G cells
  • TOV21G human ovarian cancer cells, purchased from Nanjing Kebai Biotechnology Co., Ltd.
  • the above cells in the logarithmic growth phase were plated in 96-well plates at a cell density of 2000 cells/well, and a blank control group was set at the same time.
  • DMSO dimethyl sulfoxide
  • Signal value of blank group mean value of fluorescence signal of medium group (containing 0.5% DMSO);
  • Signal value of negative control group mean value of fluorescence signal of cell + medium group (containing 0.5% DMSO).
  • Table 6 shows the IC50s of compounds for inhibition of TOV21G cell proliferation.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)

Abstract

本发明公开了一种抑制AKT的吡唑并氮杂卓类化合物,该吡唑并氮杂卓类化合物的结构如通式I所示,各取代基的定义如说明书所述,本发明还提供了其制备方法。本发明的吡唑并氮杂卓类化合物具有显著的AKT抑制活性,能够用作AKT抑制剂。

Description

一种吡唑并氮杂卓类AKT抑制剂 技术领域
本发明属于药物化学领域,具体涉及AKT抑制剂、其制备方法以及医药用途。
背景技术
磷脂酰肌醇3-激酶(PI3K)及其下游蛋白AKT(又称蛋白激酶B,PKB)和哺乳动物雷帕霉素靶蛋白(mTOR)组成的PI3K/AKT/mTOR通路作为细胞内非常重要的信号转导途径,在细胞的生长、存活、增殖、凋亡、血管生成、自吞噬等过程中发挥着极其重要的生物学功能。该通路的异常激活会引起一系列的疾病,包括癌症、神经病变、自身免疫性疾病和血液淋巴系统疾病。
AKT,是一类丝氨酸/苏氨酸激酶,通过下游众多效应器影响细胞存活、生长、代谢、增殖、迁移和分化。超过50%的人类肿瘤存在AKT过度活化的现象,尤以前列腺癌、胰腺癌、膀胱癌、卵巢癌、乳腺癌为主。AKT过活化可导致肿瘤发生、转移以及耐药性产生。AKT具有三种亚型:AKT1、AKT2和AKT3。作为典型的蛋白激酶,每种亚型均由氨基末端的PH结构域(Pleckstrin homology domain)、中部结合ATP的激酶结构域以及羧基末端的调节结构域组成。3种亚型约80%的氨基酸序列同源,仅在PH结构域和激酶结构域连接区变化较大。目前,针对PI3K/AKT/mTOR信号通路的靶向药物主要是PI3K抑制剂和mTOR抑制剂,而AKT处于该信号转导通路的核心部位。抑制AKT活性既可避免抑制上游PI3K造成的严重副作用,也可避免抑制下游mTOR引起的负反馈机制影响药效。因此,寻找有效和选择性的AKT抑制剂是当前肿瘤靶向药物研发的重要方向。CN101631778A公开了一类环戊二烯并[D]嘧啶衍生物,CN101578273A公开了一类羟基化和甲氧基化的环戊二烯并[D]嘧啶衍生物,CN101511842A公开了一类二氢呋喃并嘧啶衍生物,CN101970415A公开了一类5H-环戊二烯并[d]嘧啶衍生物,这些化合物具有小于10μM的抑制AKT1的IC 50
发明内容
一方面,本发明提供了式I所示的化合物或其药学上可接受的盐,
Figure PCTCN2021135266-appb-000001
其中:
R 1选自H或2-(C 1-C 4烷基)氨基-2-氧代乙基;
R 2选自H、卤素或C 1-C 4烷基;
R 3选自卤素;
X选自S、O或NH。
在一些实施方案中,R 1选自H或2-(甲基氨基)-2-氧代乙基;在一些典型的实施方案中,R 1选自H。
在一些实施方案中,R 2选自氯、溴或甲基;在一些典型的实施方案中,R 2选自氯或溴;在一些更为典型的实施方案中,R 2选自溴。
在一些实施方案中,R 3选自氟、氯或溴;在一些典型的实施方案中,R 3选自氟。
在一些实施方案中,X选自S、O;在一些典型的实施方案中,X选自S。
在一些实施方案中,前述式I化合物具有如式II所示的结构,
Figure PCTCN2021135266-appb-000002
其中,R 1、R 2、R 3和X定义如式I化合物中所定义的。
在一些特定的实施方案中,本发明提供了以下化合物或其药学上可接受的盐,
Figure PCTCN2021135266-appb-000003
在一些特定的实施方案中,本发明提供药学上可接受的盐选自盐酸盐或甲酸盐。
在一些特定的实施方案中,本发明提供了以下化合物,
Figure PCTCN2021135266-appb-000004
Figure PCTCN2021135266-appb-000005
另一方面,本发明还提供了一种药物组合物,其包含治疗有效量的式I化合物或其药学上可接受的盐。
在一些实施方案中,本发明还提供了一种药物组合物,其包含治疗有效量的式I化合物或其药学上可接受的盐,以及一种或多种药学上可接受的载体。
本发明所述的药物组合物可以通过任何适用的途径或方法给药,例如通过口服或肠胃外(例如,静脉内)给药。式I化合物的治疗有效量为从约0.001mg到20mg/Kg体重/天,优选从0.01mg到10mg/Kg体重/天。对于口服途径给药,本发明的药物组合物通常以片剂、胶囊剂或溶液的形式提供。片剂可以包含本发明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。所述载体包括但不限于稀释剂、崩解剂、粘合剂、润滑剂、着色剂或防腐剂。胶囊剂包括硬胶囊剂和软胶囊剂。
对于胃肠道外途径给药,本发明的药物组合物可以通过静脉内注射、肌内注射或皮下注射给药。其通常以无菌水溶液或混悬液或冻干粉末提供,并调节合适的pH和等渗性。
另一方面,本发明还提供了式I化合物在制备用于预防和/或治疗AKT蛋白激酶介导的疾病或疾病状态的药物中的用途。
另一方面,本发明还提供了用于预防和/或治疗AKT蛋白激酶介导的疾病或疾病状态的方法,其包括向有需要的个体给予本发明的式I化合物或本发明的药物组合物。
另一方面,本发明还提供了用于预防和/或治疗AKT蛋白激酶介导的疾病或疾病状态的本发明的式I化合物或本发明的药物组合物。
所述AKT蛋白激酶介导的疾病或疾病状态的实例包括但不限于乳腺癌、前列腺癌或卵巢癌。
还一方面,本发明提供一种制备式I化合物的方法,包括但不限于以下合成方案:
中间体I-8及I-10的合成方案:
Figure PCTCN2021135266-appb-000006
其中,R 2、X的定义如前所述,其中TBSO为叔丁基二甲基硅氧基。
式I-1化合物与叔丁基二甲基(2-丙炔氧基)硅烷在催化剂(如二(三苯基膦)二氯化钯和碘化亚铜)条件下制备I-2化合物;I-2化合物脱保护基得I-3化合物;I-3化合物在还原剂(如10%湿钯碳)存在下制备I-4化合物;I-4化合物进行溴代反应得I-5化合物;I-5化合物与1H-吡唑-3-硼酸频哪酯反应得I-6化合物;I-6化合物进行羟基溴代得I-7化合物;I-7化合物经成环反应得I-8化合物;I-8化合物进行取代反应制备I-9化合物;I-9化合物进行水解反应制备I-10化合物。
中间体II-6及II-10合成方案:
Figure PCTCN2021135266-appb-000007
其中,R 3的定义如上文所述,R 4选自C 1-C 4烷基。
II-1化合物与(甲酰基亚甲基)三苯基膦反应制得II-2化合物;II-2化合物与2-硝基乙基氨基甲酸叔丁酯成环反应得II-3化合物;II-3化合物在催化剂(如三乙基硅烷,三氟乙酸)作用下得到II-4化合物;II-4化合物与二碳酸二叔丁酯,反应得到II-5化合物;II-5化合物在还原剂(如10%氢氧化钯)条件下还原得II-6化合物;II-3化合物与烯丙基三甲基硅烷反应得II-7化合物;II-7化合物进行氧化反应得II-8化合物;II-8化合物成酰胺反应得到II-9化合物;II-9化合物还原硝基得到II-10化合物。
合成方案III:
Figure PCTCN2021135266-appb-000008
其中,R 1、R 2、X的定义如前式I所述。
化合物II-6和化合物I-10进行成酰胺反应得到式III-2;式III-2脱保护得式III-3化合物。合成方案IV:
Figure PCTCN2021135266-appb-000009
其中,R 2、R 3、X的定义如前式I所述,R 4选自C 1-C 4烷基。
化合物II-10和化合物I-10进行成酰胺反应得到式III-2;式III-2脱保护得式III-3化合物。
具体实施方式
相关定义
除非有特定说明,下列用在说明书和权利要求书中的术语具有下述含义:
本发明“化合物”可以是不对称的,例如,具有一个或多个手性中心。除非另有说明,本发明的“化合物”指的是任意一种立体异构体或两种以上的立体异构体的混合物。立体异构体包括但不限于对映异构体和非对映异构体。本发明的含有不对称碳原子的化合物可以以光学活性纯的形式或两种以上的立体异体的混合物的形式被分离得到。光学活性纯的形式可以从两种以上的立体异构体的混合物中进行拆分,或通过使用手性原料或手性试剂合成。
术语“任选”或“任选地”是指随后描述的事件或情况可能发生或可能不发生,该描述包括发生所述事件或情况和不发生所述事件或情况。本文中的数字范围,是指给定范围中的各个整数。例如,“C1-C4”是指该基团可具有1个碳原子、2个碳原子3个碳原子、4个碳原子。
术语“烷基”指饱和的脂族烃基团,包括直链的或支链的饱和烃基,所述烃基具有所示出的碳原子数。如术语“C1-C4烷基”包括C1烷基、C2烷基、C3烷基和C4烷基,实例包括,但不限于,甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基等。
术语“卤素”指氟、氯、溴和碘。
术语“羟基”指-OH。
术语“药学上可接受的盐”是指保留了特定化合物的游离酸和碱的生物学效力而没有生物学不良作用的盐。例如酸(包括有机酸和无机酸)加成盐或碱加成盐(包括有机碱和无机碱)。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。
术语“有效量”或“治疗有效量”是指无毒的但能达到预期效果的药物或药剂的足够用量。
术语“药学上可接受的载体”是指对机体无明显刺激作用,而且不会损害该活性化合物的 生物活性及性能的那些载体。包括但不限于国家药品监督管理局许可的可用于人或动物的任何稀释剂、崩解剂、粘合剂、助流剂、润湿剂。
权利要求书和说明书中所使用的简称其含义如下:
M:mol/L
mM:mmol/L
nM:nmol/L
Boc:叔丁氧羰基
DMB:2,4-二甲氧基苄基
NMP:N-甲基吡咯烷酮
DMAP:对二甲氨基吡啶
DMF:N,N-二甲基甲酰胺
h:小时
min:分
TK:酪氨酸激酶
制备方法:
下面更具体地描述本发明的化合物的制备方法,但这些具体的制备方法不对本发明的范围构成任何限制。此外,反应条件如反应物、溶剂、碱、所用化合物的量、反应温度、反应时间等不限于下面的实例。
本发明的化合物还可以任选地将在本说明书中描述的或本领域已知的各种合成方法组合起来而方便制得,这样的组合可由本领域的技术人员容易地进行。
流程A:
Figure PCTCN2021135266-appb-000010
反应条件:a)5-溴噻吩-2-羧酸乙酯,叔丁基二甲基(2-丙炔氧基)硅烷,二(三苯基膦)二氯化钯,碘化亚铜,三乙胺;b)四丁基氟化铵,四氢呋喃;c)氢气,10%湿钯碳,甲醇;d)三氯化铝,液溴,二氯甲烷;e)1H-吡唑-3-硼酸频哪酯,[1,1'-双(二苯基膦)二茂铁]氯化钯(II),碳酸铯,N,N-二甲基甲酰胺,水;f)三苯基膦,四溴化碳,二氯甲烷;g)碳酸铯,碘化钾,N,N-二甲基甲酰胺。
中间体1 6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酸乙酯
Figure PCTCN2021135266-appb-000011
a)5-(3-((叔丁基二甲基甲硅烷基)氧基)丙-1-基-1-基)噻吩-2-羧酸乙酯
20℃氮气保护下,将5-溴噻吩-2-羧酸乙酯(6g),叔丁基二甲基(2-丙炔氧基)硅烷(5.22g),二(三苯基膦)二氯化钯(1.79g),碘化亚铜(0.97g)溶于三乙胺(30mL)中,反应液在90℃下搅拌8h。反应液旋干,直接用于下一步。
b)5-(3-羟基丙-1-炔-1-基)噻吩-2-羧酸乙酯
将5-(3-((叔丁基二甲基甲硅烷基)氧基)丙-1-基-1-基)噻吩-2-羧酸乙酯(6g)溶于四氢呋喃(60mL)中,滴加四丁基氟化铵的四氢呋喃溶液(20mL)20℃下搅拌16h。加入的混合物用水(100mL)稀释。混合物用乙酸乙酯(2×100mL)萃取。有机相以无水硫酸钠干燥。过滤后,滤液在减压下进行浓缩。产品经层析硅胶板纯化(石油醚:乙酸乙酯=3:1)得到产品暗黄色液体5g。
c)5-(3-羟丙基)噻吩-2-羧酸乙酯
将5-(3-羟基丙-1-炔-1-基)噻吩-2-羧酸乙酯(5g)溶于MeOH中,加入10%Pd/C(2g),氢气条件下室温搅拌16h。反应液过滤,旋干,粗品经层析硅胶柱纯化(石油醚:乙酸乙酯=3:1)得产品黄色液体4g。
d)4-溴-5-(3-羟丙基)噻吩-2-羧酸乙酯
在室温氮气保护下,将5-(3-羟丙基)噻吩-2-羧酸乙酯(2g)溶于二氯甲烷(5mL)中,加入三氯化铁(151.39mg),0℃下滴加液溴(0.7mL),20℃下搅拌16h。将反应液倒入饱和硫代硫酸钠溶液(50mL)搅拌10min,加入二氯甲烷(2×50mL),有机相用饱和氯化钠溶液洗涤(2×50mL),干燥,旋干,粗品经层析硅胶板纯化(石油醚:乙酸乙酯=3:1)得到产品淡黄色液体1.3g。
e)5-(3-羟丙基)-4-(1H-吡唑-3-基)噻吩-2-羧酸乙酯
将4-溴-5-(3-羟丙基)噻吩-2-羧酸乙酯(1.3g)溶于N,N二甲基甲酰胺(20mL)和水(4 mL)的混合溶液中,加入1H-吡唑-3-硼酸频哪酯(1.03g),[1,1'-双(二苯基膦)二茂铁]氯化钯(II)(324.5mg),碳酸铯(2.89g)。将反应混合物在100℃下搅拌8h。将反应液倾入水(50mL)中,再加入乙酸乙酯(2×50mL)萃取。有机相加入饱和氯化钠溶液(2×50mL)洗涤,无水硫酸钠干燥,旋干。产品经层析硅胶板纯化(石油醚:乙酸乙酯=1:1)得到产品淡黄色固体0.2g。
f)5-(3-溴丙基)-4-(1H-吡唑-3-基)噻吩-2-羧酸乙酯
在20℃下将5-(3-羟丙基)-4-(1H-吡唑-3-基)噻吩-2-羧酸乙酯(200mg)溶于二氯甲烷(5mL)中,加入三苯基膦(243.35mg)和四溴化碳(236.59mg),室温搅拌16h。反应液旋干,粗品经层析硅胶柱纯化(石油醚:乙酸乙酯=3:1)得到产品淡黄色液体0.2g。
g)6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂9-羧酸乙酯
在20℃下将5-(3-溴丙基)-4-(1H-吡唑-3-基)噻吩-2-羧酸乙酯(0.2g)溶于N,N二甲基甲酰胺(5mL)中,加入碳酸铯(379.90mg)和碘化钾(145.09mg),室温搅拌16h。将反应液倾入水(100mL)中,再加入乙酸乙酯(2×100mL)萃取。有机相加入饱和氯化钠溶液(2×100mL)洗涤,无水硫酸钠干燥,旋干。产品经层析硅胶板纯化(石油醚:乙酸乙酯=3:1)得到产品白色固体0.10g。
流程B:
Figure PCTCN2021135266-appb-000012
反应条件:a)3,4-二氟苯甲醛,(甲酰基亚甲基)三苯基膦,N,N二甲基甲酰胺;b)(2S)-2-[二苯基[(三甲基硅酯)氧基]甲基]-吡咯烷,2-硝基乙基氨基甲酸叔丁酯,苯甲酸,二氯甲烷;c)(4S,5S)4-(3,4-二氟苯基)-2-羟基-5-硝基哌啶-1-甲酸叔丁酯,三乙基硅烷,三氟乙酸,二氯甲烷;d)二碳酸二叔丁酯,三乙胺,二氯甲烷;e)氢气,10%氢氧化钯,乙醇。
中间体2 (3S,4S)3-氨基-4-(3,4-二氟苯基)哌啶-1-甲酸叔丁酯
Figure PCTCN2021135266-appb-000013
a)(E)-3-(3,4-二氟苯基)丙烯醛
在20℃下将3,4二氟苯甲醛(1g)溶于N,N二甲基甲酰胺(20mL)中,加入(甲酰基亚甲基)三苯基膦(2.14g),80℃搅拌8h。将反应液倾入水(100mL)中,再加入乙酸乙酯(2×100mL)萃取。有机相加入饱和氯化钠溶液(2×100mL)洗涤,无水硫酸钠干燥,旋干。产品经层析硅胶板纯化(石油醚:乙酸乙酯=5:1)得到产品白色固体0.65g。
b)(4S,5S)4-(3,4-二氟苯基)-2-羟基-5-硝基哌啶-1-甲酸叔丁酯
将(2S)-2-[二苯基[(三甲基硅酯)氧基]甲基]-吡咯烷(58.08mg),2-硝基乙基氨基甲酸叔丁酯(407.22mg)和苯甲酸(43.58mg),溶入二氯甲烷(10mL)中,0℃加入(E)-3-(3,4-二氟苯基)丙烯醛(300mg),室温搅拌16h。反应液加入饱和碳酸氢钠溶液(100mL)中,再加入乙酸乙酯(2×100mL)萃取。有机相加入饱和氯化钠溶液(2×100mL)洗涤,无水硫酸钠干燥,旋干。产品经层析硅胶板纯化(石油醚:乙酸乙酯=5:1)得到产品白色固体0.5g。
c)(3S,4S)-4-(3,4-二氟苯基)-3-硝基哌啶
20℃氮气保护下,将(4S,5S)4-(3,4-二氟苯基)-2-羟基-5-硝基哌啶-1-甲酸叔丁酯(800mg)溶于二氯甲烷(50mL)中,加入三乙基硅烷(519.18mg),0℃加入三氟乙酸(2.5g),室温搅拌16h。反应液旋干,得粗品无色油状物(600mg)。
d)(3S,4S)4-(3,4-二氟苯基)-3-硝基哌啶-1-甲酸叔丁酯
20℃氮气保护下,将(3S,4S)-4-(3,4-二氟苯基)-3-硝基哌啶(600mg)溶于四氢呋喃(10mL)中,加入三乙胺(751.96mg),0℃下加入二碳酸二叔丁酯(2.70g)。20℃下搅拌16h。加入的混合物用水(100mL)稀释。混合物用乙酸乙酯(2×100mL)萃取。有机相用饱和氯化钠溶液洗涤(2×100mL)。有机相以无水硫酸钠干燥。过滤后,滤液在减压下进行浓缩。产品经层析硅胶板纯化(石油醚:乙酸乙酯=5:1)得到产品暗黄色液体600mg。
e)(3S,4S)3-氨基-4-(3,4-二氟苯基)哌啶-1-甲酸叔丁酯
将(3S,4S)4-(3,4-二氟苯基)-3-硝基哌啶-1-甲酸叔丁酯(200mg),溶入乙醇(5mL)中,加入10%氢氧化钯(200mg),氢气条件下50℃搅拌8h。反应液过滤,旋干,得到产品白色固体0.18g。
流程C:
Figure PCTCN2021135266-appb-000014
反应条件:a)N-氯代丁二酰亚胺,四氢呋喃;b)氢氧化钠,甲醇,水;c)1-氯-6,7-二氢-5氢-吡唑[1,5-a]噻吩[3,2-c]氮杂卓-9-甲酸,N,N-二异丙基乙胺,2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐,二氯甲烷;d)氯化氢二氧六环溶液,二氯甲烷。
实施例1 1-氯-N-(((3S,4S)-4-(3,4-二氟苯基)哌啶-3-基)-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酰胺盐酸盐
Figure PCTCN2021135266-appb-000015
a)1-氯-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酸乙酯
在20℃下将6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酸乙酯(0.1g)溶于四氢呋喃(5mL)中,加入N-氯代丁二酰亚胺(61.08mg),30℃搅拌4h。将反应液倾入水(100mL)中,再加入乙酸乙酯(2×100mL)萃取。有机相加入饱和氯化钠溶液(2×100mL)洗涤,无水硫酸钠干燥,旋干。产品经层析硅胶板纯化(石油醚:乙酸乙酯=3:1)得到产品白色固体0.05g。
b)1-氯-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酸
在20℃下将1-氯-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酸乙酯(50mg)溶于甲醇(10mL)和水(4mL)的混合溶液中,加入氢氧化钠(26.95mg)。80℃下搅拌2h。反应液冷却至室温,搅拌下加入2M盐酸(3mL),将反应液倾入水(30mL)中,再加入乙酸乙酯(2×30mL)萃取。有机相加入饱和氯化钠溶液(2×30mL)洗涤,无水硫酸钠干燥,旋干。得到产品白色固体0.04g。
c)((3S,4S)-叔丁基3-(1-氯-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酰胺基)-4-(3,4-二氟苯基)哌啶-1-甲酸
将(3S,4S)3-氨基-4-(3,4-二氟苯基)哌啶-1-甲酸叔丁酯(30mg),溶入二氯甲烷(5mL)中,加入1-氯-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酸(30.97mg),N,N-二异丙基乙胺(37.24mg)和2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(43.82mg),室温搅拌1h。反应液加入水(100mL)中,再加入乙酸乙酯(2×100mL)萃取。有机相加入饱和氯化钠溶液(2×100mL)洗涤,无水硫酸钠干燥,旋干。产品经层析硅胶板纯化(二氯甲烷:甲醇=10:1)得到产品白色固体40mg。
d)1-氯-N-(((3S,4S)-4-(3,4-二氟苯基)哌啶-3-基)-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酰胺盐酸盐
将((3S,4S)-叔丁基3-(1-氯-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓--9-甲酰胺基)-4-(3,4-二氟苯基)哌啶-1-甲酸(40mg),溶入二氯甲烷(5mL)中,加入氯化氢二氧六环溶液(1mL),20℃搅拌4h。反应液旋干,得到产品0.01g。LC-MS(ESI)m/z:463.0(M+H). 1HNMR(400MHz,MeOD)δ8.50(s,2H),7.94(s,1H),7.46(s,1H),7.04–7.27(m,3H),4.42(td,J=11.5,4.2Hz,1H),4.24–4.33(m,2H),3.50–3.58(m,1H),3.39–3.49(m,1H),3.13(t,J=7.0Hz,2H),2.91–3.09(m,3H),2.23–2.34(m,2H),2.07–2.17(m,1H),1.90–2.04(m,1H).
流程D:
Figure PCTCN2021135266-appb-000016
反应条件:a)6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酸乙酯,N-溴代丁二酰亚胺,四氢呋喃;b)氢氧化钠,甲醇,水;c)(3S,4S)3-氨基-4-(3,4-二氟苯基)哌啶-1-甲酸叔丁酯,N,N-二异丙基乙胺,2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐,二氯甲烷;d)氯化氢二氧六环溶液,二氯甲烷;
实施例2 1-溴-N-(((3S,4S)-4-(3,4-二氟苯基)哌啶-3-基)-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酰胺盐酸盐
Figure PCTCN2021135266-appb-000017
a)1-溴-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酸乙酯
在20℃下将6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酸乙酯(0.9g)溶于四氢呋喃(10mL)中,加入N-溴代丁二酰亚胺(732.76mg),15℃搅拌1h。将反应液倾入水(100mL)中,再加入乙酸乙酯(2×100mL)萃取。有机相加入饱和氯化钠溶液(2×100mL)洗涤,无水硫酸钠干燥,旋干。产品经层析硅胶板纯化(石油醚:乙酸乙酯=3:1)得到产品白色固体1.0g。
b)1-溴-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-羧酸
在20℃下将1-溴-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酸乙酯(100mg)溶于甲醇(10mL)和水(2mL)的混合溶液中,加入氢氧化钠(46.89mg)。80℃下搅拌2h。反应液冷却至室温,搅拌下加入2M盐酸(3mL),将反应液倾入水(30mL)中,再加入乙酸乙酯(2×30mL)萃取。有机相加入饱和氯化钠溶液(2×30mL)洗涤,无水硫酸钠干燥,旋干。得到产品白色固体0.08g。
c)((3S,4S)-叔丁基3-(1-溴-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酰胺基)-4-(3,4-二氟苯基)哌啶-1-甲酸
将(3S,4S)3-氨基-4-(3,4-二氟苯基)哌啶-1-甲酸叔丁酯(50mg),溶入二氯甲烷(5mL)中,加入1-溴-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂9-羧酸(50.13mg),N,N-二异丙基乙胺(62.07mg)和2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(73.04mg),室温搅拌1h。反应液加入水(100mL)中,再加入乙酸乙酯(2×100mL)萃取。有机相加入饱和氯化钠溶液(2×100mL)洗涤,无水硫酸钠干燥,旋干。产品经制备分离得到产品白色固体50mg。
d)1-溴-N-(((3S,4S)-4-(3,4-二氟苯基)哌啶-3-基)-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酰胺盐酸盐
将((3S,4S)-叔丁基3-(1-溴-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酰胺基)-4-(3,4-二氟苯基)哌啶-1-甲酸(50mg),溶入二氯甲烷(5mL)中,加入氯化氢二氧六环溶液(1mL),20℃搅拌4h。反应液旋干,得到产品26mg。LC-MS(ESI)m/z:507.0(M+H). 1H NMR(400MHz,MeOD)δ7.94(s,1H),7.50(s,1H),7.10–7.28(m,3H),4.44–4.55(m,1H),4.26(td,J=6.4,2.4Hz,2H),3.61–3.69(m,1H),3.49–3.56(m,1H),3.00–3.16(m,5H),2.27–2.38(m,2H),2.15–2.22(m,1H),1.98–2.05(m,1H).
流程E:
Figure PCTCN2021135266-appb-000018
反应条件:a)1-溴-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酸乙酯,甲基硼酸,[1,1'-双(二苯基膦基)二茂铁]二氯化钯,碳酸钾,N,N-二甲基甲酰胺;b)氢氧化钠,甲醇,水;c)(3S,4S)3-氨基-4-(3,4-二氟苯基)哌啶-1-甲酸叔丁酯,N,N-二异丙基乙胺,2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐,二氯甲烷;d)氯化氢二氧六环溶液,二氯甲烷。
实施例3 N-((3S,4S)-4-(3,4-二氟苯基)哌啶-3-基)-1-甲基-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酰胺盐酸盐
Figure PCTCN2021135266-appb-000019
a)1-甲基-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酸乙酯
在20℃下将1-溴-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酸乙酯(100mg),甲基硼酸(139mg),碳酸钾(340mg)溶于N,N-二甲基甲酰胺(10mL)中,加入[1,1'-双(二苯基膦基)二茂铁]二氯化钯(47mg),110℃搅拌8h。将反应液倾入水(100mL)中,再加入乙酸乙酯(2×100mL)萃取。有机相加入饱和氯化钠溶液(2×100mL)洗涤,无水硫酸钠干燥,旋干。产品经层析硅胶板纯化(石油醚:乙酸乙酯=3:1)得到产品白色固体100mg。
b)1-甲基-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酸
在20℃下将1-甲基-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酸乙酯(100mg)溶于甲醇(10mL)和水(2mL)的混合溶液中,加入氢氧化钠(57.89mg)。80℃下搅拌2h。反应液冷却至室温,搅拌下加入2M盐酸(3mL),将反应液倾入水(30mL)中,再加入乙酸乙酯(2×30mL)萃取。有机相加入饱和氯化钠溶液(2×30mL)洗涤,无水硫酸钠干燥,旋干。得到产品白色固体0.08g。
c)(3S,4S)-叔丁基4-(3,4-二氟苯基)-3-(1-甲基-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酰胺基)哌啶-1-羧酸盐
将(3S,4S)3-氨基-4-(3,4-二氟苯基)哌啶-1-甲酸叔丁酯(80mg),溶入二氯甲烷(5mL)中,加入1-甲基-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酸(63.59mg),N,N-二异丙基乙胺(99.03mg)和2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(116.86mg),室温搅拌1h。反应液加入水(100mL)中,再加入乙酸乙酯(2×100mL)萃取。有机相加入饱和氯化钠溶液(2×100mL)洗涤,无水硫酸钠干燥,旋干。产品经制备分离得到产品白色固体80mg。
d)N-((3S,4S)-4-(3,4-二氟苯基)哌啶-3-基)-1-甲基-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酰胺盐酸盐
将(3S,4S)-叔丁基4-(3,4-二氟苯基)-3-(1-甲基-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酰胺基)哌啶-1-羧酸盐(80mg),溶入二氯甲烷(5mL)中,加入氯化氢二氧六环溶液(1mL),20℃搅拌4h。反应液旋干,得到产品69mg。LC-MS(ESI)m/z:433.1(M+H). 1H NMR(400MHz,MeOD)δ7.79(s,1H),7.58(s,1H),7.08–7.33(m,3H),4.46–4.56(m,1H),4.18–4.32(m,2H),3.49–3.63(m,2H),3.00–3.23(m,5H),2.27–2.39(m,2H),2.23(s,3H),1.93–2.12(m,2H).
流程F:
Figure PCTCN2021135266-appb-000020
反应条件:a)烯丙基三甲基硅烷,三氟化硼乙醚,二氯甲烷;b)三氯化钌,高碘酸钠,二氯甲烷,乙腈,水;c)2M甲胺四氢呋喃溶液;N,N-二异丙基乙胺,2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐,二氯甲烷;d)氢气,10%氢氧化钯,乙醇。
中间体3 (4S,5S)-5-氨基-4-(3,4-二氟苯基)-2-(2-(甲基氨基)-2-氧乙基)哌啶-1-甲酸叔丁酯
Figure PCTCN2021135266-appb-000021
a)(4S,5S)2-2-烯丙基-4-(3,4-二氟苯基)-5-硝基哌啶-1-甲酸叔丁酯
将(4S,5S)4-(3,4-二氟苯基)-2-羟基-5-硝基哌啶-1-甲酸叔丁酯(500mg),溶入二氯甲烷(20mL)中,加入烯丙基三甲基硅烷(1mL),-78℃滴加三氟化硼乙醚(0.5mL),室温搅拌16h。反应液加入饱和碳酸氢钠溶液(100mL)中,再加入乙酸乙酯(2×100mL)萃取。有机相加入饱和氯化钠溶液(2×100mL)洗涤,无水硫酸钠干燥,旋干。产品经层析硅胶板纯化(石油醚:乙酸乙酯=3:1)得到产品白色固体0.09g。
b)(2-((4S,5S)-1-(叔丁氧羰基)-4-(3,4-二氟苯基)-5-硝基哌啶-2-基)乙酸
将(4S,5S)2-2-烯丙基-4-(3,4-二氟苯基)-5-硝基哌啶-1-甲酸叔丁酯(90mg),溶入二氯甲烷(2mL),乙腈(2mL)和水(4mL)的混合溶剂中,加入三氯化钌(9.76mg),高碘酸钠(251.7mg),室温搅拌16h。反应液过滤,滤液加入2M盐酸溶液调至PH=5,再加入二氯甲烷(2×50mL)萃取。有机相加入饱和氯化钠溶液(2×50mL)洗涤,无水硫酸钠干燥,旋干。得到产品白色固体0.1g。
c)(4S,5S)4-(3,4-二氟苯基)-2-(2-(甲基氨基)-2-氧乙基)-5-硝基哌啶-1-甲酸叔丁酯
将(2-((4S,5S)-1-(叔丁氧羰基)-4-(3,4-二氟苯基)-5-硝基哌啶-2-基)乙酸(100mg),溶入二氯甲烷(20mL)中,加入2M甲胺四氢呋喃溶液(0.19mL),N,N-二异丙基乙胺(96.84mg)和2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(113.96mg),室温搅拌1h。反应液加入水(100mL)中,再加入乙酸乙酯(2×100mL)萃取。有机相加入饱和氯化钠溶液(2×100mL)洗涤,无水硫酸钠干燥,旋干。产品经层析硅胶板纯化(二氯甲烷:甲醇=10:1)得到产品白色固体0.09g。
d)(4S,5S)-叔丁基5-氨基-4-(3,4-二氟苯基)-2-(2-(甲基氨基)-2-氧乙基)哌啶-1-甲酸
将(4S,5S)4-(3,4-二氟苯基)-2-(2-(甲基氨基)-2-氧乙基)-5-硝基哌啶-1-甲酸叔丁酯(90mg),溶入乙醇(5mL)中,加入10%氢氧化钯(113.96mg),氢气条件下70℃搅拌8h。反应液过滤,旋干,得到产品白色固体0.07g。
流程G:
Figure PCTCN2021135266-appb-000022
反应条件:a)(4S,5S)-5-氨基-4-(3,4-二氟苯基)-2-(2-(甲基氨基)-2-氧乙基)哌啶-1-甲酸叔丁酯,N,N-二异丙基乙胺,2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐,二氯甲烷;b)氯化氢二氧六环溶液,二氯甲烷;
实施例4 1-氯-N-((3S,4S)-4-(3,4-二氟苯基)-6-(2-(甲基氨基)-2-氧乙基)哌啶-3-基)-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酰胺盐酸盐
Figure PCTCN2021135266-appb-000023
a)(4S,5S)-叔丁基5-(1-氯-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酰胺基)-4-(3,4-二氟苯基)-2-(2-(甲基氨基)-2-氧乙基)哌啶-1-甲酸
将(4S,5S)-叔丁基5-氨基-4-(3,4-二氟苯基)-2-(2-(甲基氨基)-2-氧乙基)哌啶-1-甲酸(30mg),溶入二氯甲烷(20mL)中,加入1-氯-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酸(25.23mg),N,N-二异丙基乙胺(30.34mg)和2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐(35.7mg),室温搅拌1h。反应液加入水(100mL)中,再加入乙酸乙酯(2×100mL)萃取。有机相加入饱和氯化钠溶液(2×100mL)洗涤,无水硫酸钠干燥,旋干。产品经层析硅胶板纯化(二氯甲烷:甲醇=10:1)得到产品白色固体30mg。
b)1-氯-N-((3S,4S)-4-(3,4-二氟苯基)-6-(2-(甲基氨基)-2-氧乙基)哌啶-3-基)-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酰胺盐酸盐
将(4S,5S)-叔丁基5-(1-氯-6,7-二氢-5H-吡唑并[1,5-a]噻吩并[3,2-c]氮杂卓-9-甲酰胺基) -4-(3,4-二氟苯基)-2-(2-(甲基氨基)-2-氧乙基)哌啶-1-甲酸(30mg),溶入二氯甲烷(5mL)中,加入氯化氢二氧六环溶液(1mL),20℃搅拌4h。反应液旋干,得到产品0.07g。LC-MS(ESI)m/z:534.1(M+H). 1H NMR(400MHz,MeOD)δ7.97(s,1H),7.47(s,1H),7.12–7.29(m,3H),4.38–4.47(m,1H),4.26–4.34(m,2H),4.04–4.12(m,1H),3.57–3.75(m,2H),3.42–3.49(m,1H),3.14(t,J=6.8Hz,2H),2.80(d,J=10.4Hz,4H),2.25–2.33(m,2H),2.02–2.10(m,1H),1.28–1.38(m,2H).
流程H:
Figure PCTCN2021135266-appb-000024
反应条件:a)碘甲烷,碳酸钾,N,N-二甲基甲酰胺;b)叔丁基二甲基(2-丙炔氧基)硅烷,[1,1’-双(二苯基膦)二茂铁]二氯化钯,碘化亚铜,三乙胺;c)四丁基氟化铵/四氢呋喃溶液(1.0M);d)氢气,10%湿钯碳,无水乙醇;e)液溴,三氯化铝,二氯甲烷;f)1H-吡唑-3-硼酸频哪酯,二(三叔丁基膦)钯,碳酸氢钠,二氧六环,水;g)四溴化碳,三苯基膦,二氯甲烷;h)碳酸铯,碘化钾,N,N-二甲基甲酰胺。
中间体4 6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酸甲酯
Figure PCTCN2021135266-appb-000025
a)5-溴呋喃-2-甲酸甲酯
将5-溴呋喃-2-甲酸(20.0g,105.3mmol)溶解于N,N-二甲基甲酰胺(150mL)中,随后加入碘甲烷(5.7mL,126.4mmol)和碳酸钾(58.2g,421.2mmol),25℃下反应24小时。反应完全后,向反应液中加入水(300mL),以乙酸乙酯(100mL×3)萃取,饱和食盐水洗涤有机相(10mL×3),减压蒸馏除去有机相,得到白色固体17.8g,无需纯化直接用于下 一步反应。
b)5-(3-((叔丁基二甲基硅基)氧基)丙-1-炔-1-基)呋喃-2-甲酸甲酯
将5-溴呋喃-2-甲酸甲酯(10.0g,49.0mmol)、叔丁基二甲基(2-丙炔氧基)硅烷(10.0g,58.8mmol)、[1,1’-双(二苯基膦)二茂铁]二氯化钯(1.79g,2.45mmol)和碘化亚铜(0.93g,4.90mmol)悬浮于三乙胺(50mL)中,氮气保护下搅拌,反应液在110℃下反应3小时后,反应完毕。将反应液冷却至室温,减压蒸馏除去有机溶剂后,残余物通过柱色谱纯化得到黄色油状液体11.5g。
c)5-(3-羟丙-1-炔-1-基)呋喃-2-甲酸甲酯
将5-(3-((叔丁基二甲基硅基)氧基)丙-1-炔-1-基)呋喃-2-甲酸甲酯(11.5g,46.2mmol)溶于四氢呋喃(30mL)中,随后加入四丁基氟化铵/四氢呋喃溶液(1.0M)(46mL),氮气保护下搅拌,反应液在室温下反应10小时后,反应完毕。减压蒸馏除去有机溶剂,用二氯甲烷(100mL)重新溶解,用2N盐酸洗涤(100mL x 2),无水硫酸钠干燥后,减压蒸馏后浓缩,残余物通过柱色谱纯化得到黄色油状液体5.8g。
d)5-(3-羟丙基)呋喃-2-甲酸甲酯
氮气保护下,往5-(3-羟丙-1-炔-1-基)呋喃-2-甲酸甲酯(5.8g,32.1mmol)的乙醇(40mL)溶液中,加入钯碳(1.2g,10%)。混悬液用氢气置换三次,室温常压下搅拌反应12小时后,反应完全。抽滤除去钯碳,减压蒸馏浓缩,得到黄色油状液体4.9g。LC-MS(ESI)m/z:185.1(M+H). 1H NMR(400MHz,CDCl 3)δ(ppm)7.10(d,J=3.4Hz,1H),6.16(d,J=3.4Hz,1H),3.88(s,3H),3.70(t,J=6.3Hz,2H),2.82(t,J=7.5Hz,2H),1.99-1.91(m,2H).
e)4-溴-5-(3-羟丙基)呋喃-2-甲酸甲酯
0℃氮气保护下,往5-(3-羟丙基)呋喃-2-甲酸甲酯(5.8g,31.5mmol)和三氯化铝(14.7g,110.3mmol)的二氯甲烷(80mL)混悬物中滴加液溴(10.1g,63.0mmol)。滴加完毕后,移至室温下搅拌反应12小时后,反应完全。加入饱和硫代硫酸钠溶液(30mL)淬灭,用二氯甲烷(100mL)萃取,用饱和氯化钠洗涤(100mL x 2),无水硫酸钠干燥后,减压蒸馏后浓缩,残余物通过柱色谱纯化得到黄色油状液体5.0g。LC-MS(ESI)m/z:263.0(M+H). 1H NMR(400MHz,CDCl 3)δ(ppm)7.12(s,1H),3.88(s,3H),3.68(t,J=6.2Hz,2H),2.85(t,J=7.4Hz,2H),2.05(s,1H),1.99-1.91(m,2H).
f)5-(3-羟丙基)-4-(1H-吡唑-3-基)呋喃-2-甲酸甲酯
将4-溴-5-(3-羟丙基)呋喃-2-甲酸甲酯(3.0g,11.4mmol)、1H-吡唑-3-硼酸频哪酯(2.7g,13.7mmol)、二(三叔丁基膦)钯(0.88g,1.7mmol)和碳酸氢钠(2.4g,28.6mmol)混合在1,4-二氧六环(10mL)和水(2.5mL)溶液中,110℃氮气保护下,搅拌反应4小时后,反应 完毕。冷却至室温,抽滤除去不溶性杂质,滤液加入2N盐酸调节pH至酸性,加入二氯甲烷(50mL x 2)萃取,合并有机层用饱和氯化钠(20mL x 2)洗涤,无水硫酸钠干燥,减压蒸馏浓缩,残余物通过柱色谱纯化得到黄色固体1.1g。LC-MS(ESI)m/z:251.1(M+H). 1HNMR(400MHz,CDCl 3)δ(ppm)7.61(d,J=2.4Hz,1H),7.33(s,1H),6.43(d,J=2.5Hz,1H),3.91(s,3H),3.63(t,J=5.8Hz,2H),3.20-3.13(m,2H),2.06-1.99(m,2H).
g)5-(3-溴丙基)-4-(1H-吡唑-3-基)呋喃-2-甲酸甲酯
氮气保护下,往5-(3-羟丙基)-4-(1H-吡唑-3-基)呋喃-2-甲酸甲酯(1.1g,4.3mmol)的二氯甲烷(20mL)溶液中加入四溴化碳(1.4g,4.3mmol)和三苯基膦(1.5g,5.6mmol)。室温下搅拌反应6小时后,停止反应。减压蒸馏除去有机溶剂,用二氯甲烷(20mL)重新溶解,用饱和氯化钠洗涤(20mL x 2),无水硫酸钠干燥后,减压蒸馏后浓缩,残余物通过柱色谱纯化得到白色固体310mg。LC-MS(ESI)m/z:313.0(M+H). 1H NMR(400MHz,CDCl 3)δ(ppm)7.64(d,J=2.4Hz,1H),7.41(s,1H),6.47(d,J=2.4Hz,1H),3.91(s,3H),3.46(t,J=6.6Hz,2H),3.18(t,J=6.6Hz,2H),2.38-2.28(m,2H).
h)6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酸甲酯
将5-(3-溴丙基)-4-(1H-吡唑-3-基)呋喃-2-甲酸甲酯(310mg,1.0mmol)溶于N,N-二甲基甲酰胺(10mL)中,加入碳酸铯(647mg,2.0mmol)和碘化钾(247mg,1.5mmol),室温下搅拌4小时后,反应完全。减压蒸馏浓缩至干,残余物通过柱色谱纯化得到白色固体240mg。LC-MS(ESI)m/z:233.1(M+H). 1H NMR(400MHz,CDCl 3)δ(ppm)7.42(d,J=1.9Hz,1H),7.30(s,1H),6.39(d,J=2.0Hz,1H),4.52-4.43(m,2H),3.92(s,3H),3.16(t,J=6.4Hz,2H),2.33-2.21(m,2H).
流程I:
Figure PCTCN2021135266-appb-000026
反应条件:a)N-氯代丁二酰亚胺,四氢呋喃;b)氢氧化钠,甲醇,水;c)(3S,4S)-3-氨基-4-(3,4-二氟苯基)哌啶-1-羧酸叔丁酯,2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯, 二异丙基乙胺,二氯甲烷;d)氯化氢/1,4-二氧六环溶液(4.0M)。
实施例5 1-氯-N-((3S,4S)-4-(3,4-二氟苯基)哌啶-3-基)-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酰胺甲酸盐
Figure PCTCN2021135266-appb-000027
a)1-氯-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酸甲酯
将6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酸甲酯(240mg,1.0mmol)溶于四氢呋喃(10mL)中,加入N-氯代丁二酰亚胺(152mg,1.1mmol),65℃下搅拌反应4小时后,反应完毕。加水(10mL)淬灭,用乙酸乙酯(20mL x 2)萃取,有机相用饱和氯化钠(20mL x 2)洗涤,无水硫酸钠干燥,减压蒸馏后浓缩,残余物通过柱色谱纯化得到白色固体170mg。LC-MS(ESI)m/z:267.0(M+H). 1H NMR(400MHz,CDCl 3)δ(ppm)7.89(s,1H),7.39(s,1H),4.46-4.39(m,2H),3.93(s,3H),3.18(t,J=6.6Hz,2H),2.27-2.21(m,2H).
b)1-氯-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酸
往1-氯-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酸甲酯(170mg,0.64mmol)的甲醇(10mL)和水(2mL)混合溶液中加入氢氧化钠(256mg,6.4mmol),80℃下搅拌反应4小时后,反应完全。用2N盐酸调节反应液pH至酸性,加入乙酸乙酯(40mL)萃取,有机相用饱和氯化钠(20mL x 2)洗涤,无水硫酸钠干燥,减压蒸馏浓缩,得到白色固体140mg。LC-MS(ESI)m/z:253.0(M+H). 1H NMR(400MHz,MeOD)δ(ppm)7.84(s,1H),7.43(s,1H),4.47-4.40(m,2H),3.19(t,J=6.6Hz,2H),2.25-2.19(m,2H).
c)(3S,4S)-3-(1-氯-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酰胺)-4-(3,4-二氟苯基)哌啶-1-羧酸叔丁酯
0℃氮气保护下,将1-氯-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酸(40mg,0.16mmol)和(3S,4S)-3-氨基-4-(3,4-二氟苯基)哌啶-1-羧酸叔丁酯(50mg,0.16mmol)溶于二氯甲烷(5mL)中,加入二异丙基乙胺(82mg,0.63mmol)和2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(72mg,0.19mmol),室温下搅拌反应2小时后,反应完全。加入2N盐酸(50mL),用乙酸乙酯(10mL x 2)萃取,有机相用饱和氯化钠(10mL x 2)洗涤,无水硫酸钠干燥,减压蒸馏浓缩,残余物通过薄层制备色谱纯化得到白色固体50mg。
d)1-氯-N-((3S,4S)-4-(3,4-二氟苯基)哌啶-3-基)-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酰胺甲酸盐
将(3S,4S)-3-(1-氯-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酰胺)-4-(3,4-二氟苯基)哌啶-1-羧酸叔丁酯(50mg,0.09mmol)溶于甲醇(5mL)中,加入氯化氢/1,4-二氧六环溶液(4.0M)(1mL),室温下搅拌反应4小时后,反应完全。减压蒸馏除去溶液,残余物通过制备型HPLC纯化得到目标产品25mg。制备条件:色谱柱:Kromasil 10μm C18 30×150mm,流动相A:水(含0.1%甲酸),流动相B:乙腈。梯度:时间0-10min,B相20-40%;10-12min,B相95%;12-14min,B相20%(体积比),RT=7.56min。LC-MS(ESI)m/z:427.1(M+H). 1H NMR(400MHz,MeOD)δ(ppm)8.51(s,1H),7.35(s,1H),7.27-7.22(m,1H),7.21(s,1H),7.20-7.13(m,1H),7.12-7.07(m,1H),4.52-4.44(m,1H),4.35-4.29(m,2H),3.53-3.39(m,3H),3.11(t,J=6.6Hz,2H),3.08-2.92(m,3H),2.19(s,3H),2.17-2.08(m,3H),1.93(q,J=12.9Hz,1H).
流程J:
Figure PCTCN2021135266-appb-000028
反应条件:a)N-溴代丁二酰亚胺,四氢呋喃;b)氢氧化钠,甲醇,水;c)(3S,4S)-3-氨基-4-(3,4-二氟苯基)哌啶-1-羧酸叔丁酯,2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯,二异丙基乙胺,二氯甲烷;d)氯化氢/1,4-二氧六环溶液(4.0M)。
实施例6 1-溴-N-((3S,4S)-4-(3,4-二氟苯基)哌啶-3-基)-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酰胺甲酸盐
Figure PCTCN2021135266-appb-000029
a)1-溴-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酸甲酯
将6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酸甲酯(820mg,3.5mmol)溶于四氢呋喃(10mL)中,加入N-溴代丁二酰亚胺(690mg,3.9mmol),65℃下搅拌反应4小时后,反应完毕。加水(10mL)淬灭,用乙酸乙酯(20mL x 2)萃取,有机相用饱和氯化钠(20mL x 2)洗涤,无水硫酸钠干燥,减压蒸馏后浓缩,残余物通过柱色谱纯化得到白色固体800mg。b)1-溴-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酸
往1-溴-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酸甲酯(210mg,0.67mmol)的甲醇(10mL)和水(2mL)混合溶液中加入氢氧化钠(269mg,6.7mmol),80℃下搅拌反应4小时后,反应完全。用2N盐酸调节反应液pH至酸性,加入乙酸乙酯(40mL)萃取,有机相用饱和氯化钠(20mL x 2)洗涤,无水硫酸钠干燥,减压蒸馏浓缩,得到白色固体140mg。c)(3S,4S)-3-(1-溴-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酰胺)-4-(3,4-二氟苯基)哌啶-1-羧酸叔丁酯
0℃氮气保护下,将1-溴-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酸(47mg,0.16mmol)和(3S,4S)-3-氨基-4-(3,4-二氟苯基)哌啶-1-羧酸叔丁酯(50mg,0.16mmol)溶于二氯甲烷(5mL)中,加入二异丙基乙胺(82mg,0.63mmol)和2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(72mg,0.19mmol),室温下搅拌反应2小时后,反应完全。加入2N盐酸(50mL),用乙酸乙酯(10mL x 2)萃取,有机相用饱和氯化钠(10mL x 2)洗涤,无水硫酸钠干燥,减压蒸馏浓缩,残余物通过薄层制备色谱纯化得到白色固体70mg。
d)1-溴-N-((3S,4S)-4-(3,4-二氟苯基)哌啶-3-基)-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酰胺甲酸盐
将(3S,4S)-3-(1-溴-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酰胺)-4-(3,4-二氟苯基)哌啶-1-羧酸叔丁酯(70mg,0.12mmol)溶于甲醇(5mL)中,加入氯化氢/1,4-二氧六环溶液(4.0M)(1mL),室温下搅拌反应4小时后,反应完全。减压蒸馏除去溶液,残余物通过制备型HPLC纯化得到产物30mg。制备条件:色谱柱:Kromasil 10μm C18 30×150mm,流动相A:水(含0.1%甲酸),流动相B:乙腈。梯度:时间0-10min,B相25-45%;10-12min,B 相95%;12-14min,B相25%(体积比),RT=6.22min。LC-MS(ESI)m/z:491.1(M+H). 1H NMR(400MHz,MeOD)δ(ppm)7.81(s,1H),7.42(s,1H),7.27-7.06(m,3H),4.47(s,1H),4.42-4.35(m,2H),3.56-3.39(m,2H),3.16-3.01(m,5H),2.24-2.04(m,3H),1.97-1.84(m,1H).
流程K:
Figure PCTCN2021135266-appb-000030
反应条件:a)甲基硼酸,[1,1'-双(二苯基膦基)二茂铁]二氯化钯,碳酸钾,N,N-二甲基甲酰胺;b)氢氧化钠,甲醇,水;c)(3S,4S)-3-氨基-4-(3,4-二氟苯基)哌啶-1-羧酸叔丁酯,2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯,二异丙基乙胺,二氯甲烷;d)氯化氢/1,4-二氧六环溶液(4.0M)。
实施例7 N-((3S,4S)-4-(3,4-二氟苯基)哌啶-3-基)-1-甲基-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酰胺甲酸盐
Figure PCTCN2021135266-appb-000031
a)1-甲基-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酸甲酯
在20℃下将1-溴-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酸甲酯(300mg,0.96mmol),甲基硼酸(0.46g,7.71mmol),碳酸钾(0.80g,5.78mmol)溶于N,N-二甲基甲酰胺(10mL)中,加入[1,1'-双(二苯基膦基)二茂铁]二氯化钯(106mg,0.14mmol),110℃搅拌3h。将反应液倾入水(100mL)中,再加入乙酸乙酯(100mL x 2)萃取。有机相加入饱和氯化钠溶液(100mL x 2)洗涤,无水硫酸钠干燥,旋干。产品经层析硅胶板纯化(石油醚:乙酸乙酯=3:1)得到产品白色固体210mg。
b)1-甲基-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酸
往1-甲基-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酸甲酯(210mg,0.85mmol)的甲醇(10mL)和水(2mL)混合溶液中加入氢氧化钠(341mg,8.5mmol),80℃下搅拌反应4小时后,反应完全。用2N盐酸调节反应液pH至酸性,加入乙酸乙酯(40mL)萃取,有机相用饱和氯化钠(20mL x 2)洗涤,无水硫酸钠干燥,减压蒸馏浓缩,得到白色固体190mg。c)(3S,4S)-3-(1-甲基-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酰胺)-4-(3,4-二氟苯基)哌啶-1-羧酸叔丁酯
0℃氮气保护下,将1-甲基-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酸(37mg,0.16mmol)和(3S,4S)-3-氨基-4-(3,4-二氟苯基)哌啶-1-羧酸叔丁酯(50mg,0.16mmol)溶于二氯甲烷(5mL)中,加入二异丙基乙胺(82mg,0.63mmol)和2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(72mg,0.19mmol),室温下搅拌反应2小时后,反应完全。加入2N盐酸(50mL),用乙酸乙酯(10mL x 2)萃取,有机相用饱和氯化钠(10mL x 2)洗涤,无水硫酸钠干燥,减压蒸馏浓缩,残余物通过薄层制备色谱纯化得到白色固体50mg。
d)N-((3S,4S)-4-(3,4-二氟苯基)哌啶-3-基)-1-甲基-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酰胺甲酸盐
将(3S,4S)-3-(1-甲基-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酰胺)-4-(3,4-二氟苯基)哌啶-1-羧酸叔丁酯(50mg,0.09mmol)溶于甲醇(5mL)中,加入氯化氢/1,4-二氧六环溶液(4.0M)(1mL),室温下搅拌反应4小时后,反应完全。减压蒸馏除去溶液,残余物通过制备型HPLC纯化得到产物20mg。制备条件:色谱柱:Kromasil 10μm C18 30×150mm,流动相A:水(含0.1%甲酸),流动相B:乙腈。梯度:时间0-10min,B相20-40%;10-12min,B相95%;12-14min,B相20%(体积比),RT=7.88min。LC-MS(ESI)m/z:427.1(M+H). 1H NMR(400MHz,MeOD)δ(ppm)8.51(s,1H),7.35(s,1H),7.27-7.22(m,1H),7.21(s,1H),7.20-7.13(m,1H),7.12-7.07(m,1H),4.52-4.44(m,1H),4.35-4.29(m,2H),3.53-3.39(m,3H),3.11(t,J=6.6Hz,2H),3.08-2.92(m,3H),2.19(s,3H),2.17-2.08(m,3H),1.93(q,J=12.9Hz,1H).
流程L:
Figure PCTCN2021135266-appb-000032
反应条件:a)(4S,5S)-5-氨基-4-(3,4-二氟苯基)-2-(2-(甲基氨基)-2-氧乙基)哌啶-1-甲酸叔丁酯,N,N-二异丙基乙胺,2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸盐, 二氯甲烷;b)氯化氢/1,4-二氧六环溶液(4.0M)。
实施例8 1-氯-N-((3S,4S)-4-(3,4-二氟苯基)-6-(2-(甲基氨基)-2-氧乙基)哌啶-3-基)-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酰胺甲酸盐
Figure PCTCN2021135266-appb-000033
a)(4S,5S)-5-(1-氯-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酰胺基)-4-(3,4-二氟苯基)-2-(2-(甲基氨基)-2-氧乙基)哌啶-1-甲酸叔丁酯
0℃氮气保护下,将1-氯-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酸(20mg,0.08mmol)和(4S,5S)-5-氨基-4-(3,4-二氟苯基)-2-(2-(甲基氨基)-2-氧乙基)哌啶-1-甲酸叔丁酯(30mg,0.08mmol)溶于二氯甲烷(5mL)中,加入二异丙基乙胺(40mg,0.31mmol)和2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(36mg,0.09mmol),室温下搅拌反应2小时后,反应完全。加入2N盐酸(50mL),用乙酸乙酯(10mL x 2)萃取,有机相用饱和氯化钠(10mL x 2)洗涤,无水硫酸钠干燥,减压蒸馏浓缩,残余物通过薄层制备色谱纯化得到产物30mg。
b)1-氯-N-((3S,4S)-4-(3,4-二氟苯基)-6-(2-(甲基氨基)-2-氧乙基)哌啶-3-基)-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酰胺甲酸盐
将(4S,5S)-5-(1-氯-6,7-二氢-5H-呋喃[3,2-c]吡唑[1,5-a]氮杂卓-9-甲酰胺基)-4-(3,4-二氟苯基)-2-(2-(甲基氨基)-2-氧乙基)哌啶-1-甲酸叔丁酯(30mg,0.05mmol)溶于甲醇(5mL)中,加入氯化氢/1,4-二氧六环溶液(4.0M)(1mL),室温下搅拌反应4小时后,反应完全。减压蒸馏除去溶液,残余物通过制备型HPLC纯化得到产物18mg。制备条件:色谱柱:Kromasil 10μm C18 30×150mm,流动相A:水(含0.1%甲酸),流动相B:乙腈。梯度:时间0-10min,B相20-40%;10-12min,B相95%;12-14min,B相20%(体积比),RT=7.99min。LC-MS(ESI)m/z:518.2(M+H). 1H NMR(400MHz,MeOD)δ(ppm)8.48(s,1H),7.71(s,1H),7.40(s,1H),7.28-7.07(m,3H),4.44-4.31(m,3H),3.81(s,1H),3.27-3.10(m,6H),2.91-2.82(m,1H),2.77(s,3H),2.63-2.56(m,1H),2.24-2.05(m,3H),1.95(d,J=14.4Hz,1H).
实验例9体外酶活性测试
1.材料和试剂
Envision型号读板仪(Molecular Devices)
白色384孔板(货号#264706,Thermo)
HTRF kinEASE TK试剂盒包含的主要试剂(货号#62TKOPEC,Cisbio)
TK-生物素底物
链霉亲和素-XL665
铕标记的酪氨酸激酶底物抗体
5x酶反应缓冲液
SEB
HTRF检测缓冲液
AKT1(货号#01-101,Carna)
AKT2(货号#01-102,Carna)
AKT3(货号#PV3185,Invitrogen)
ATP 10mM(货号#PV3227,Invitrogen)
DTT 1M(货号#D5545,Sigma)
MgCl 21M(货号#M8266,Sigma)
本发明化合物
阳性对照物:GDC-0068
2.实验步骤
2.1试剂配制
终浓度为1x激酶反应缓冲液:14mL激酶AKT1,2,3的1x激酶反应缓冲液中由含有2800μL 5x激酶反应缓冲液、140μL 100mM MgCl 2、140μL 100mM DTT、10920L超纯水配制而成。
表1、激酶的反应缓冲液的配制
Figure PCTCN2021135266-appb-000034
首先用DMSO将化合物储液(10mM的DMSO溶液)稀释至100μM化合物溶液,然后用1x激酶反应缓冲液稀释至2.5μM化合物工作液(含2.5%的DMSO)。使用1x激酶反应缓冲液配制2.5%的DMSO溶液,然后用含有2.5%的DMSO的缓冲反应溶液稀释2.5μM化合物工作液,以3.16的稀释比稀释7次,共8个浓度(如表2所示)。
表2化合物浓度梯度
浓度/nM 2500 791.14 250.36 79.23 25.07 7.93 2.51 0.79
用1x激酶反应缓冲液稀释AKT1,2,3激酶及其底物底物和ATP至表3浓度。
表3、酶、底物及ATP浓度(终浓度)
  酶浓度/ng/μL 底物/μM ATP/μM
AKT1 0.008 1 26.0
AKT2 0.05 1 40.0
AKT2 0.008 1 33.0
用检测缓冲液配制SA-XL665和STK Antibody-Cryptate至上表浓度。
表4、XL665及抗体浓度(终浓度)
  SA-XL665 STKAntibody-Cryptate
AKT1/2/3 62.5nM
2.2实验过程:
a)化合物浓度梯度设置:见表2,按浓度梯度,在化合物滴定仪设置浓度梯度;
b)酶配制:见表3,分别1.67×终浓度的酶溶液,使用自动分液仪每孔加入6μL,后使用化合物滴定仪喷入化合物;预混15min;
c)ATP和底物配制:见表3,分别配制5×ATP和5×底物,两者混匀后,使用自动分液仪4μL加入每孔;
d)2500转离心30s,25℃孵育30分钟;
e)XL665和抗体配制:见表4,分别配制4×XL665,和抗体等体积混匀,使用自动分液仪10μL加入每孔,2500转离心30s,封膜25℃孵育1h,后多功能读板仪HTRF模块检测数值;
2.3数据处理:
Figure PCTCN2021135266-appb-000035
ER=665nm荧光值/615nm荧光值
3.实验结果
实验结果如表5所示。
表5、化合物对AKT1/2/3酶抑制IC50结果
化合物编号 AKT1/nM AKT2/nM AKT3/nM
实施例1 <0.01 0.71 1.12
实施例2 <0.01 1.06 0.66
实施例3 0.24 1.12 4.29
实施例4 0.24 1.18 1.60
实施例5 1.00 15.35 10.45
实施例6 1.20 11.04 5.09
实施例7 2.40 26.35 23.96
实施例8 2.11 20.22 12.63
GDC-0068 5.96 43.04 51.23
实施例10本发明化合物对LNCaP细胞增殖抑制实验
1.实验步骤
LNCaP(人前列腺癌细胞,购自南京科佰生物科技有限公司),培养基:1640+15%FBS(购自Gibco),置于37℃,5%CO 2的培养箱中培养。取对数生长期的上述细胞分别以6000个/孔的细胞密度铺在96孔板中,并同时设置空白对照组。
将待测化合物溶解在二甲基亚砜(DMSO)中以制备10mM的储液,并置于-80℃冰箱中长期保存。细胞铺板24h后,用DMSO稀释10mM的化合物储液得到200倍工作液的溶液(工作液最高浓度30μM,3倍梯度,共10个浓度),每个浓度各取3μl加入到197μl的完全培养基中,稀释得到3倍工作液的溶液,然后取50μl加入到100μl的细胞培养液中(DMSO终浓度为0.5%,v/v),每个浓度设置两个复孔。加药处理72h后,每孔加入50μl的
Figure PCTCN2021135266-appb-000036
(购自Promega),按照说明书的操作流程在Envision(PerkinElmer)上测定荧光信号,使用GraphPad Prism 5软件log(inhibitor)vs.response-Variable slope拟合量效曲线,得到化合物对细胞增殖抑制的IC50值,如表6所示。抑制率计算公式:
Figure PCTCN2021135266-appb-000037
其中:
受试物信号值:细胞+培养基+化合物组荧光信号均值;
空白组信号值:培养基组(含0.5%DMSO)荧光信号均值;
阴性对照组信号值:细胞+培养基组(含0.5%DMSO)荧光信号均值。
2.实验结果
化合物对LNCaP细胞增殖抑制的IC 50如表6所示。
实施例11本发明化合物对TOV21G细胞增殖抑制实验
1.实验步骤
TOV21G(人卵巢癌细胞,购自南京科佰生物科技有限公司),培养基:1640+15%FBS(购自Gibco),置于37℃,5%CO 2的培养箱中培养。取对数生长期的上述细胞分别以2000个/孔的细胞密度铺在96孔板中,并同时设置空白对照组。
将待测化合物溶解在二甲基亚砜(DMSO)中以制备10mM的储液,并置于-80℃冰箱中长期保存。细胞铺板24h后,用DMSO稀释10mM的化合物储液得到200倍工作液的溶液(工作液最高浓度30μM,3倍梯度,共10个浓度),每个浓度各取3μl加入到197μl的完全培养基中,稀释得到3倍工作液的溶液,然后取50μl加入到100μl的细胞培养液中(DMSO终浓度为0.5%,v/v),每个浓度设置两个复孔。加药处理72h后,每孔加入50μl的
Figure PCTCN2021135266-appb-000038
(购自Promega),按照说明书的操作流程在Envision(PerkinElmer)上测定荧光信号,使用GraphPad Prism 5软件log(inhibitor)vs.response-Variable slope拟合量效曲线,得到化合物对细胞增殖抑制的IC50值,如表6所示。抑制率计算公式:
Figure PCTCN2021135266-appb-000039
其中:
受试物信号值:细胞+培养基+化合物组荧光信号均值;
空白组信号值:培养基组(含0.5%DMSO)荧光信号均值;
阴性对照组信号值:细胞+培养基组(含0.5%DMSO)荧光信号均值。
2.实验结果
化合物对TOV21G细胞增殖抑制的IC 50如表6所示。
表6化合物对TOV21G和LNCaP细胞增殖抑制的IC 50
序号 IC 50(LNCaP,μM) IC 50(TOV21G,μM)
GDC0068 0.99 0.65
实施例1 0.08 0.09
实施例2 0.05 0.05
实施例3 0.12 0.10
实施例4 0.26 0.24
实施例5 0.94 0.55
实施例6 0.71 0.47
实施例7 0.72 0.62
实施例8 1.89 1.25

Claims (10)

  1. 一种结构如式I所示的化合物或其药学上可接受的盐,
    Figure PCTCN2021135266-appb-100001
    其中:
    R 1选自H或2-(C 1-C 4烷基)氨基-2-氧代乙基;
    R 2选自H、卤素或C 1-C 4烷基;
    R 3选自卤素;
    X选自S、O或NH。
  2. 根据权利要求1所述的化合物或其药学上可接受的盐,其特征在于:R 1选自H或2-(甲基氨基)-2-氧代乙基;
    优选的,R 1选自H。
  3. 根据权利要求1所述的化合物或其药学上可接受的盐,其特征在于:R 2选自氯、溴或甲基;
    优选的,R 2选自氯或溴;
    更优选的,R 2选自溴。
  4. 根据权利要求1所述的化合物或其药学上可接受的盐,其特征在于:R 3选自氟、氯或溴;
    优选的,R 3选自氟。
  5. 根据权利要求1所述的化合物或其药学上可接受的盐,其特征在于:X选自S、O;
    优选的,X选自S。
  6. 根据权利要求1所述的化合物或其药学上可接受的盐,其具有如式II所示的结构:
    Figure PCTCN2021135266-appb-100002
    其中,R 1、R 2、R 3和X定义如式I化合物中所定义的。
  7. 下列化合物或其药学上可接受的盐,
    Figure PCTCN2021135266-appb-100003
  8. 下列化合物,
    Figure PCTCN2021135266-appb-100004
  9. 药物组合物,其包含权利要求1-8任一项所述化合物或其药学上可接受的盐;优选地,所述组合物包含权利要求1-8任一项所述化合物或其药学上可接受的盐以及一种或多种药学上可接受的载体。
  10. 权利要求1-8任一项所述化合物或其药学上可接受的盐或权利要求9所述药物组合物在制备用于预防和/或治疗AKT蛋白激酶介导的疾病或疾病状态的药物中的用途;优选地,所述疾病或疾病状态为癌症;更优选地,所述疾病或疾病状态为乳腺癌、前列腺癌或卵巢癌。
PCT/CN2021/135266 2020-12-07 2021-12-03 一种吡唑并氮杂卓类akt抑制剂 WO2022121788A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202180079206.6A CN116583525A (zh) 2020-12-07 2021-12-03 一种吡唑并氮杂卓类akt抑制剂

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202011415872.6 2020-12-07
CN202011415872 2020-12-07

Publications (1)

Publication Number Publication Date
WO2022121788A1 true WO2022121788A1 (zh) 2022-06-16

Family

ID=81974198

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/135266 WO2022121788A1 (zh) 2020-12-07 2021-12-03 一种吡唑并氮杂卓类akt抑制剂

Country Status (2)

Country Link
CN (1) CN116583525A (zh)
WO (1) WO2022121788A1 (zh)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017215588A1 (zh) * 2016-06-16 2017-12-21 南京明德新药研发股份有限公司 作为Akt抑制剂的二氢吡唑氮杂卓类化合物
EP3725791A1 (en) * 2017-12-13 2020-10-21 Harbin Zhenbao Pharmaceutical Co., Ltd. Salt serving as akt inhibitor and crystal thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017215588A1 (zh) * 2016-06-16 2017-12-21 南京明德新药研发股份有限公司 作为Akt抑制剂的二氢吡唑氮杂卓类化合物
EP3725791A1 (en) * 2017-12-13 2020-10-21 Harbin Zhenbao Pharmaceutical Co., Ltd. Salt serving as akt inhibitor and crystal thereof

Also Published As

Publication number Publication date
CN116583525A (zh) 2023-08-11

Similar Documents

Publication Publication Date Title
TWI749404B (zh) 作為mcl-1抑制劑的大環吲哚
WO2021218110A1 (zh) 一类苯并噻唑基联芳基类化合物、制备方法和用途
AU2020223687A1 (en) Pyrazolopyridine Derivative Having GLP-1 Receptor Agonist Effect
JP2022523981A (ja) 抗癌剤として有用な縮合三環式化合物
EP3131897B1 (en) Factor ixa inhibitors
WO2021121397A1 (zh) 取代的炔基杂环化合物
US20170066766A1 (en) Ring-fused bicyclic pyridyl derivatives as FGFR4 inhibitors
AU2012228090A1 (en) Pyrrolopyridineamino derivatives as Mps1 inhibitors
JP2018521025A (ja) Tnf活性のモジュレーターとしてのベンゾオキサジノン誘導体およびその類似体
CN107108644A (zh) 二氟甲基‑氨基吡啶和二氟甲基‑氨基嘧啶
TWI605048B (zh) Novel pyrrolopyrimidine compounds or salts thereof, and pharmaceutical compositions containing the same, in particular, prophylactic and / or therapeutic agents for tumors and the like based on the inhibition of NAE
CN104011052A (zh) 新的化合物
CN108349996B (zh) 三环pi3k抑制剂化合物及其使用方法
JP2023513854A (ja) 大環状化合物およびその使用
WO2020156437A1 (zh) Akt抑制剂
CN113754682B (zh) 具有大环结构的化合物及其用途
KR20230002721A (ko) Egfr 억제제로서의 삼환계 화합물
WO2021032004A9 (zh) 氮杂芳基化合物及其应用
CN111655689B (zh) 吡唑并吡啶酮化合物
CA3103055A1 (en) Erk inhibitor and use thereof
WO2023178928A1 (zh) 2-氨基-4-吲哚基嘧啶类化合物及其制备方法与应用
WO2022121788A1 (zh) 一种吡唑并氮杂卓类akt抑制剂
WO2021228223A1 (zh) 氘代akt激酶抑制剂
WO2015110092A1 (zh) 4-取代吡咯并[2,3-d]嘧啶化合物及其用途
CN117897384A (zh) Cdk2抑制剂

Legal Events

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

Ref document number: 21902495

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 202180079206.6

Country of ref document: CN

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21902495

Country of ref document: EP

Kind code of ref document: A1