WO2019233458A1 - Vegfr抑制剂及其制备方法和应用 - Google Patents

Vegfr抑制剂及其制备方法和应用 Download PDF

Info

Publication number
WO2019233458A1
WO2019233458A1 PCT/CN2019/090223 CN2019090223W WO2019233458A1 WO 2019233458 A1 WO2019233458 A1 WO 2019233458A1 CN 2019090223 W CN2019090223 W CN 2019090223W WO 2019233458 A1 WO2019233458 A1 WO 2019233458A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
cycloalkyl
group
deuterium
hydroxyl
Prior art date
Application number
PCT/CN2019/090223
Other languages
English (en)
French (fr)
Inventor
吴勇
彭宇然
龚彦春
史谦
邓洁
吴小东
刘永强
Original Assignee
江苏威凯尔医药科技有限公司
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
Priority claimed from CN201910469158.6A external-priority patent/CN110577546B/zh
Application filed by 江苏威凯尔医药科技有限公司 filed Critical 江苏威凯尔医药科技有限公司
Publication of WO2019233458A1 publication Critical patent/WO2019233458A1/zh

Links

Images

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
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4741Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having oxygen as a ring hetero atom, e.g. tubocuraran derivatives, noscapine, bicuculline
    • 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
    • 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
    • C07D491/02Heterocyclic 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 two hetero rings
    • C07D491/04Ortho-condensed systems
    • C07D491/044Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring
    • C07D491/048Ortho-condensed systems with only one oxygen atom as ring hetero atom in the oxygen-containing ring the oxygen-containing ring being five-membered
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Definitions

  • the present application belongs to the technical field of medicine, and particularly relates to quinoline or quinazoline derivatives and uses thereof for preparing a medicament for treating malignant tumor diseases.
  • Receptor tyrosine kinases are a class of enzymes that span cell membranes. They have extracellular binding regions, transmembrane structural regions, and intracellular parts that bind growth factors. Residues phosphorylate and affect cell proliferation.
  • VEGF Vascular endothelial growth factor
  • VPF vascular permeability factor
  • VEGFR-1, VEGFR-2, VEGFR-3 vascular endothelial growth factor receptors
  • vascular endothelial cell growth factor vascular endothelial cell growth factor
  • VEGFR-1 and VEGFR-2 are mainly distributed on the surface of tumor vascular endothelial cells and regulate tumor angiogenesis; VEGFR-3 is mainly distributed on the surface of lymphatic endothelium and regulate tumor angiogenesis.
  • Most human tumors express VEGF and its receptor at high levels, which has led to the hypothesis that VEGF released by tumor cells stimulates capillary growth and tumor endothelial proliferation in a paracrine manner, and promotes tumor growth by increasing blood supply.
  • VEGF vascular endothelial growth factor
  • inhibition of angiogenesis especially capillaries, the balance between cell death and proliferation makes the tumor without net growth; due to the lack of blood inflow and outflow from the tumor, Inhibit the metastasis of tumor cells; inhibit the proliferation of endothelial cells and avoid the effect of endothelial cells that arrange blood vessels on the paracrine growth-stimulation of surrounding tissues.
  • the object of the present invention is to provide a class of quinoline or quinazoline VEGFR inhibitors having excellent activity.
  • Another object of the present invention is to provide the use of the quinoline or quinazoline VEGFR inhibitor in the preparation of a medicament for preventing or treating angiogenesis-related diseases, in particular to prevent or treat tumor diseases related to protein tyrosine kinase .
  • R 1 is selected from C 1 -C 8 alkyl or C 3 -C 8 cycloalkyl, optionally further selected from one or more of deuterium, halogen, hydroxyl, cyano, nitro, -NR 8 R 9 ,- NR 8 COR 7 , -COR 7 , -SO 2 R 7 , -SOR 7 , C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl, C 1 -C 8 alkoxy or 4-10 membered heterocyclic
  • the C 3 -C 6 cycloalkyl or 4- to 10-membered heterocyclic group may be substituted by an amino group, a hydroxyl group, a-(CH 2 ) n CN, a carboxyl group, or a C 1 -C 4 alkyl group. Or C 1 -C 4 alkoxy.
  • n is selected from 0, 1, 2 or 3;
  • R 2 is selected from H, deuterium or halogen
  • X is selected from N or CH
  • Y is selected from O or CR 3 R 4 ;
  • R 3 and R 4 are each independently selected from H, deuterium, halogen, C 1 -C 8 alkyl, C 3 -C 8 cycloalkyl, or C 1 -C 8 alkoxy;
  • R 5 is selected from H, C 1 -C 8 alkyl or C 3 -C 8 cycloalkyl
  • R 6 is selected from H, C 1 -C 8 alkyl or C 3 -C 8 cycloalkyl
  • R 7 is selected from H, C 1 -C 8 alkyl, C 3 -C 8 cycloalkyl, -NR 8 R 9 or C 3 -C 6 heterocyclyl.
  • the C 3 -C 6 heterocyclyl may be Further substituted with hydroxy, carboxy or C 1 -C 4 alkyl;
  • R 8 and R 9 are each independently selected from H, C 1 -C 8 alkyl or C 3 -C 8 cycloalkyl.
  • R 1 is C 1 -C 4 alkyl, optionally further substituted with one or more substituents selected from deuterium, hydroxyl, methyl, ethyl, cyclopropyl, or a 4-6 membered heterocyclic group, said The cyclopropyl or 4- to 6-membered heterocyclic group may be further substituted with an amino group, a hydroxyl group, a carboxyl group, a C 1 to C 4 alkyl group, or a C 1 to C 4 alkoxy group. Is selected from pyrrolidinyl, morpholinyl, piperazinyl or piperidinyl.
  • R 2 is selected from H, deuterium or halogen
  • X is CH
  • Y is CH 2 ;
  • R 3 and R 4 are each independently selected from H, deuterium, halogen, C 1 -C 8 alkyl, C 3 -C 8 cycloalkyl, or C 1 -C 8 alkoxy;
  • R 5 is selected from H, C 1 -C 8 alkyl or C 3 -C 8 cycloalkyl
  • R 6 is selected from H, C 1 -C 8 alkyl or C 3 -C 8 cycloalkyl.
  • An embodiment of the present invention is a compound described by the general formula (II), a stereoisomer thereof, a pharmaceutically acceptable salt or ester, or a solvate:
  • R 1 is C 1 -C 8 alkyl, optionally further selected from one or more of deuterium, halogen, hydroxyl, cyano, -COR 7 , C 1 -C 6 alkyl, C 3 -C 6 cycloalkyl Or a 4-6 membered heterocyclyl substituent, the C 3 -C 6 cycloalkyl or 4-6 membered heterocyclyl may be further substituted with amino, hydroxyl,-(CH 2 ) n CN, carboxyl, C 1 -C 4 alkyl or C 1 -C 4 alkoxy;
  • n is selected from 0, 1, 2 or 3;
  • R 7 is selected from the group consisting of H, C 1 -C 8 alkyl, C 3 -C 8 cycloalkyl, or C 3 -C 6 heterocyclyl.
  • the C 3 -C 6 heterocyclyl may be further substituted by hydroxy, carboxy or C 1 -C 4 alkyl.
  • R 1 is C 1 -C 4 alkyl, optionally further selected from one or more of deuterium, halogen, hydroxyl, cyano, C 1 -C 2 alkyl, C 3 -C 6 cycloalkyl, or 4-6 membered heterocyclic group substituents, the C 3 -C 6 cycloalkyl or 4-6 membered heterocyclic group may be further substituted by amino, hydroxy, - (CH 2) n CN , carboxy, C 1 -C 4 alkyl or C 1 -C 4 alkoxy substituted; preferably, R 1 is methyl, ethyl, which is further substituted with a hydroxy, methyl, cyclopropyl or pyrrolidinyl substituent, said Cyclopropyl or pyrrolidinyl may be further substituted with amino, hydroxyl,-(CH 2 ) n CN, carboxyl, methyl or methoxy;
  • n is selected from 0, 1 or 2, preferably n is 0 or 1.
  • R 1 is C 1 -C 4 alkyl, optionally further substituted with one or more substituents selected from deuterium, hydroxyl, methyl, ethyl, cyclopropyl, or a 4-6 membered heterocyclic group, said The cyclopropyl or 4- to 6-membered heterocyclic group may be further substituted with an amino group, a hydroxyl group, a carboxyl group, a C 1 to C 4 alkyl group, or a C 1 to C 4 alkoxy group. Is selected from pyrrolidinyl, morpholinyl, piperazinyl or piperidinyl.
  • the compound is selected from:
  • Another aspect of the present invention provides a method for preparing a compound of formula (I) or a pharmaceutically acceptable salt or ester or solvate thereof, comprising the following steps:
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , X, Y are as defined for the compound of formula (I).
  • the compound of formula (I) or a pharmaceutically acceptable salt or ester or solvate thereof according to the present invention is a novel VEGFR inhibitor, and thus can be used to prepare a medicament for preventing or treating angiogenesis-related diseases, especially for preventing or treating proteins. Tyrosine kinase-associated malignancies.
  • the tumor is selected from the group consisting of ovarian cancer, cervical cancer, colorectal cancer, breast cancer, pancreatic cancer, glioma, glioblastoma, melanoma, prostate cancer, leukemia, lymphoma, non-cancer Hodgkin's lymphoma, gastric cancer, lung cancer, hepatocellular carcinoma, gastrointestinal stromal tumor, thyroid cancer, bile duct cancer, endometrial cancer, kidney cancer, anaplastic large cell lymphoma, acute myeloid leukemia, multiple bone marrow Tumor, melanoma or mesothelioma, soft tissue sarcoma.
  • Another aspect of the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a therapeutically effective amount of the aforementioned compound of formula (I) or a pharmaceutically acceptable salt or ester or solvate thereof as an active ingredient and a pharmaceutically acceptable carrier.
  • Another aspect of the present invention provides the application of the aforementioned pharmaceutical composition in the preparation of a medicament for preventing or treating malignant tumors.
  • C 1 -C 8 alkyl refers to a straight-chain alkyl group and a branched alkyl group including 1 to 8 carbon atoms.
  • the alkyl group refers to a saturated aliphatic hydrocarbon group, such as methyl, ethyl, n- Propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 1,2-dimethylpropyl, 2,2 -Dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2-methylpropyl, 1,1,2-tris Methylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3-dimethylbutyl, 2-ethylbutyl
  • cycloalkyl refers to a saturated monocyclic hydrocarbon substituent
  • C 3 -C 8 cycloalkyl refers to a monocyclic cycloalkyl including 3 to 8 carbon atoms
  • Restrictive examples include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, and the like.
  • heterocyclyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent, wherein one or more ring atoms are selected from nitrogen, oxygen, or S (O) r (where r is an integer of 0, Heteroatoms of 1, 2), but excluding the ring part of -OO-, -OS- or -SS-, the remaining ring atoms are carbon.
  • S (O) r where r is an integer of 0, Heteroatoms of 1, 2
  • 4-10 membered heterocyclyl refers to a cyclic group containing 4 to 10 ring atoms.
  • Non-limiting examples of monocyclic heterocyclyl include azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, homopiperazinyl, and the like.
  • Polycyclic heterocyclic groups include spiro, fused and bridged heterocyclic groups.
  • alkoxy means -O- (alkyl), wherein alkyl is as defined above.
  • C 1 -C 8 alkoxy refers to an alkyl group having 1 to 8 carbon atoms, non-limiting embodiments include methoxy, ethoxy, propoxy, butoxy and the like.
  • Halogen means fluorine, chlorine, bromine or iodine.
  • “Pharmaceutical composition” means a mixture containing one or more of the compounds described herein or a physiologically pharmaceutically acceptable salt or prodrug thereof with other chemical components, as well as other components such as physiologically pharmaceutically acceptable carriers and excipients. ⁇ ⁇ Shape agent.
  • the purpose of the pharmaceutical composition is to promote the administration to the organism, which is beneficial to the absorption of the active ingredient and then exerts the biological activity.
  • N-methoxy-N-methyl-2,3-dihydrobenzofuran-7-carboxamide 16.37 g, 78.99 mmol
  • THF 450 ml
  • N 2 N 2
  • a solution of methyl magnesium bromide in THF 17.0.00 ml, 170.00 mmol
  • the reaction system was moved to an ice bath, and the pH was adjusted to 3 to 4 with 1N hydrochloric acid.
  • TFAA (200ml) was added to a single-necked flask, and H 2 O 2 (50ml) was added dropwise at -10 ° C. After the drop was completed, 1- (5-nitro-2,3-dihydrogen) was added dropwise after stirring at this temperature for 20min.
  • a solution of benzofuran-7-yl) ethanone (19.00 g, 91.70 mmol) in DCM (45 ml) was added, and the reaction was terminated at room temperature for 3 h. Extracted with EA, washed twice with saturated brine, dried the organic phase over anhydrous sodium sulfate, filtered, and concentrated to give a yellow solid, which was directly used in the next reaction without further purification.
  • Step 9 Synthesis of 5- (ethoxymethylene) -2,2-dimethyl-1,3-dioxane-4,6-dione
  • Step 10 5-((7-benzyloxy-2,3-dihydrobenzofuran-5-ylamino) methylene) -2,2-dimethyl-1,3-dioxane-4 Synthesis of 1,6-dione
  • Step 12 Synthesis of 4-benzyloxy-9-chloro-1,2-dihydrofuro [3,2-f] quinoline
  • Step 14 Synthesis of 9- (4-fluoro-2methyl-1-hydroindole-5-yloxy) -1,2-dihydrofuro [3,2-f] quinolin-4-ol
  • Step 15 Synthesis of (1- (tert-butoxycarbonylamino) cyclopropyl) methyl mesylate
  • Step 16 1-((9- (4-fluoro-2methyl-1-hydroindole-5-yloxy) -1,2-dihydrofuro [3,2-f] quinoline-4 -Yloxy) methyl) cyclopropylcarbamic acid tert-butyl ester synthesis
  • Step 17 1-((9- (4-fluoro-2-methyl-1-hydroindole-5-yloxy) -1,2-dihydrofuro [3,2-f] quinoline- Synthesis of 4-yloxy) methyl) -cyclopropane-1-aminohydrochloride
  • Step 1 Referring to steps 1-14 of Example 1, 9- (4-fluoro-2methyl-1-hydroindole-5-yloxy) -1,2-dihydrofuro [3,2- f] quinolin-4-ol.
  • Step 1 Synthesis of tert-butyl (3R, 4S) -3,4-dimethoxypyrrolidine-1-carboxylic acid ester
  • Step 3 4- (2-Bromoethoxy) -9-((4-fluoro-2-methyl-1H-indol-5-yl) oxy) -1,2-dihydrofuran [3, Synthesis of 2-f] quinoline
  • Step 4 4- (2-((3S, 4R) -3,4-dimethoxypyrrolidin-1-yl) ethoxy) -9-((4-fluoro-2-methyl-1H- Synthesis of indole-5-yl) oxy) -1,2-dihydrofuran [3,2-f] quinoline
  • Step 1 Synthesis of tert-butyl (3S, 4S) -3,4-dimethoxypyrrolidine-1-carboxylic acid ester
  • Step 3 4- (2-((3R, 4R) -3,4-dimethoxypyrrolidin-1-yl) ethoxy) -9-((4-fluoro-2-methyl-1H- Synthesis of indole-5-yl) oxy) -1,2-dihydrofuran [3,2-f] quinoline
  • This test uses ⁇ - 33 p-ATP isotope test compound on kinase assays VEGFR / VEGFR2 / VEGFR3, and to obtain the compound inhibiting the activity of half inhibitory concentration IC 50.
  • the positive control drug Anlotinib was purchased from Suzhou Hengning Pharmaceutical Technology Co., Ltd., batch number 2016120201, and Lenvatinib was purchased from Hubei Weidley Chemical Technology Co., Ltd., batch number HBW171008.
  • the compound was dissolved to a specific concentration with 100% DMSO, and then it was gradient diluted to a different concentration of the test sample (DMSO solution) using an automatic sampling device.
  • the reaction solution was subjected to ion exchange filtration system to remove unreacted ATP and ADP plasma generated by the reaction, and then measured the 33 P isotope radiation in the substrate.
  • the inhibitory effect of different concentrations of compounds on kinase activity was calculated based on the amount of kinase added to the inhibitor system at different concentrations, and the IC 50 was inhibited by graphpad prism fitting.
  • the compound of the present invention has better kinase inhibitory activity, especially VEGFR2 inhibitory activity.
  • This test method by MTT test compound effect of VEGF-induced HUVEC cell activity and derived compounds inhibit VEGF-induced proliferation of HUVEC cells half maximal inhibitory concentration IC 50 activity.
  • HUVEC cell lines were cultured under the conditions of ECM + 5% FBS + 1% ECGS (endothelial cell growth factor additive) + 1% P / S (penicillin streptomycin mixed solution). The day before the experiment, the cells were cultured with ECM + 5% FBS. A 96-well cell culture plate was inoculated with 100 ⁇ L of HUVEC cell suspension in logarithmic growth phase, with a density of 5 ⁇ 10 4 / ml. The culture plate was cultured in an incubator for 24 hours to make the cells adhere (37 ° C, 5% CO 2 ).
  • Each compound has been dissolved in DMSO to prepare a 10 mM stock solution, which is diluted 400 times the target concentration with DMSO and diluted to 2 times the target concentration with serum-free medium to maintain the DMSO concentration in the drug solution at 0.5. %.
  • DMSO fetal sulfate
  • concentrations of drug solution and 10 ng / mL of VEGF165, 100 ⁇ L / well were sequentially added. Three replicates were set for each concentration, and blank control, normal control, and VEGF-induced groups were set up, and the culture was continued at 37 ° C and 5% CO 2 for 72 hours.
  • the compound 2 of the present invention has better activity of inhibiting the proliferation of HUVEC cells induced by VEGF.
  • the total volume of the incubation system was 250 ⁇ L.
  • 2.5 ⁇ L of 100 ⁇ mol / L test compound and The above incubation solution was mixed with 197.5 ⁇ L, and pre-incubated in a 37 ° C water bath for 5 min.
  • 50 ⁇ L of a reduced coenzyme II solution (5 mmol / L) which was also pre-incubated for 5 min was added to start the reaction.
  • test compound 1 ⁇ mol / L
  • incubate with shaking in a 37 ° C water bath, and take them out at 0, 5, 15, 30, and 60 min respectively, and immediately add the internal standard as Terfenadine (positive ion internal standard, 25ng / mL) and Tolbutamide (anion internal standard, 50ng / mL) positive and negative internal standard mixed methanol solution 600 ⁇ L to stop the reaction.
  • the incubation solution was shaken for 2 minutes, centrifuged (4 ° C, 16000 r / min) for 10 minutes, and the supernatant was taken for LC-MS / MS detection to quantitatively analyze the remaining amount of the parent drug. (DMSO ⁇ 0.1%).
  • liver microsomal test data is shown in Table 3:
  • the compound of the present invention Compared with the positive control drug, the compound of the present invention has considerable human liver microsomal metabolic stability, and therefore has good drug-making properties.

Abstract

本发明涉及具有式(I)结构的喹啉或喹唑啉衍生物、含有式(I)化合物的药物组合物及所述化合物在制备预防或治疗血管生成相关疾病的药物的用途,特别是用于预防或治疗与蛋白质酪氨酸激酶相关的肿瘤。其中式(I)中的各取代基与说明书中的定义相同。

Description

VEGFR抑制剂及其制备方法和应用 技术领域
本申请属于医药技术领域,具体涉及喹啉或喹唑啉衍生物及其用于制备治疗恶性肿瘤疾病的药物的用途。
本申请要求中国专利CN201810597224.3(申请日2018年6月8日,发明名称VEGFR抑制剂及其制备方法和应用)的优先权。
背景技术
受体酪氨酸激酶是横跨细胞膜的一类酶,具有结合生长因子的胞外结合区、跨膜结构区和胞内部分,胞内部分的功能是作为激酶将蛋白质中的特定酪氨酸残基磷酸化并影响细胞的增殖。
血管内皮生长因子(VEGF),又称血管通透因子(VPF)是一种高度特异性的促血管内皮细胞生长因子,与血管内皮生长因子受体(包括VEGFR-1、VEGFR-2、VEGFR-3)特异性结合并激活受体酪氨酸激酶发挥血管调节作用,促进血管通透性增加、细胞外基质变性、血管内皮细胞迁移、增殖和血管形成。
正常的血管生成在多种过程中起着重要作用,包括胚胎发育、创伤愈合等;不良的或者病理的血管生成与疾病状态相关,包括糖尿病视网膜病变、银屑癣、癌症、风湿性关节炎、动脉粥样化。肿瘤血管生成,新血管的形成及其通透性主要通过(源于肿瘤的)血管内皮细胞生长因子(VEGF)调节,其通过至少两种不同的受体发挥作用:VEGFR-1和VEGFR-2,受体对血管内皮细胞具有高度特异性(Endocr.Rev.1992,13,18;FASEB J.1999,13,9)。VEGFR-1和VEGFR-2主要分布在肿瘤血管内皮表面,调节肿瘤血管的生成;VEGFR-3主要分布在淋巴内皮表面,调节肿瘤***的生成。大多数人类肿瘤高水平表达VEGF及其受体,由此产生了以下假设,肿瘤细胞释放的VEGF以旁泌性方式刺激毛细血管的生长和肿瘤内皮增殖,并通过提高血液供应,促进肿瘤生长。
VEGF在体内作为肿瘤血管生成因子的作用表现在通过VEGF抗体、VEGFR-2负性突变体、VEGF反义RNA抑制VEGF表达或者VEGF活性,可使得体内神经胶质瘤细胞系或者其它肿瘤细胞系生长减缓。几种主要机制在抗肿瘤的血管生成抑制活性中起重要作用:抑制血管生长特别是毛细血管,细胞死亡和增殖之间实现平衡使得肿瘤无净增长;由于缺乏血液从肿瘤的流入和流出,因此抑制肿瘤细胞的转移;抑制内皮细胞增殖,避免排布血管的内皮细胞对周围组织的旁分泌生长-刺激的作用。
目前已公开了一系列VEGFR抑制剂及其在血管相关疾病方面应用的专利申请,其中包括 WO 2016091165、WO 2017177962、CN103382206等,但仍需要开发新的具有更好的药效的新型VEGFR抑制剂。
发明内容
本发明的目的是提供一类具有优异活性的喹啉或喹唑啉类VEGFR抑制剂。
本发明的另一个目的是提供所述喹啉或喹唑啉类VEGFR抑制剂在制备预防或治疗血管生成相关疾病的药物方面的用途,特别是预防或治疗与蛋白质酪氨酸激酶相关的肿瘤疾病。
为实现本发明的目的,本发明的技术方案如下:
本发明所述的如下式(I)所示的化合物、其立体异构体、药学上可接受的盐或酯或溶剂化物:
Figure PCTCN2019090223-appb-000001
R 1选自C 1-C 8烷基或C 3-C 8环烷基,任选进一步被一个或多个选自氘、卤素、羟基、氰基、硝基、-NR 8R 9、-NR 8COR 7、-COR 7、-SO 2R 7、-SOR 7、C 1-C 6烷基、C 3-C 6环烷基、C 1-C 8烷氧基或4-10元杂环基的取代基所取代,所述的C 3-C 6环烷基或4-10元杂环基可进一步被氨基、羟基、-(CH 2) nCN、羧基、C 1-C 4烷基或C 1-C 4烷氧基所取代。
n选自0、1、2或3;
R 2选自H、氘或卤素;
X选自N或CH;
Y选自O或CR 3R 4
R 3和R 4各自独立选自H、氘、卤素、C 1-C 8烷基、C 3-C 8环烷基或C 1-C 8烷氧基;
R 5选自H、C 1-C 8烷基或C 3-C 8环烷基;
R 6选自H、C 1-C 8烷基或C 3-C 8环烷基;
R 7选自H、C 1-C 8烷基、C 3-C 8环烷基、-NR 8R 9或C 3-C 6杂环基,所述的C 3-C 6杂环基可进一步被羟基、羧基或C 1-C 4烷基所取代;
R 8和R 9各自独立选自H、C 1-C 8烷基或C 3-C 8环烷基。
本发明实施方案,其特征在于:
R 1为C 1-C 4烷基,任选进一步被一个或多个选自氘、羟基、甲基、乙基、环丙基或4-6元杂环基的取代基所取代,所述的环丙基或4-6元杂环基可进一步被氨基、羟基、羧基、C 1-C 4烷基或C 1-C 4烷氧基所取代,所述的4-6元杂环基选自吡咯烷基、吗啉基、哌嗪基或哌啶基。
R 2选自H、氘或卤素;
X为CH;
Y为CH 2
R 3和R 4各自独立选自H、氘、卤素、C 1-C 8烷基、C 3-C 8环烷基或C 1-C 8烷氧基;
R 5选自H、C 1-C 8烷基或C 3-C 8环烷基;
R 6选自H、C 1-C 8烷基或C 3-C 8环烷基。
本发明实施方案,其为通式(II)所述的化合物、其立体异构体、药学上可接受的盐或酯或溶剂化物:
R 1为C 1-C 8烷基,任选进一步被一个或多个选自氘、卤素、羟基、氰基、-COR 7、C 1-C 6烷基、C 3-C 6环烷基或4-6元杂环基的取代基所取代,所述的C 3-C 6环烷基或4-6元杂环基可进一步被氨基、羟基、-(CH 2) nCN、羧基、C 1-C 4烷基或C 1-C 4烷氧基所取代;
n选自0、1、2或3;
R 7选自H、C 1-C 8烷基、C 3-C 8环烷基或C 3-C 6杂环基,所述的C 3-C 6杂环基可进一步被羟基、羧基或C 1-C 4烷基所取代。
本发明实施方案,其特征在于:
R 1为C 1-C 4烷基,任选进一步被一个或多个选自氘、卤素、羟基、氰基、C 1-C 2烷基、C 3-C 6环烷基或4-6元杂环基的取代基所取代,所述的C 3-C 6环烷基或4-6元杂环基可进一步被氨基、羟基、-(CH 2) nCN、羧基、C 1-C 4烷基或C 1-C 4烷氧基所取代;优选地,R 1为甲基、乙基,其进一步被羟基、甲基、环丙基或吡咯烷基的取代基所取代,所述的环丙基或吡咯烷基可进一步被氨基、羟基、-(CH 2) nCN、羧基、甲基或甲氧基所取代;
n选自0、1或2,优选n为0或1。
本发明实施方案,其特征在于:
R 1为C 1-C 4烷基,任选进一步被一个或多个选自氘、羟基、甲基、乙基、环丙基或4-6元杂环基的取代基所取代,所述的环丙基或4-6元杂环基可进一步被氨基、羟基、羧基、C 1-C 4烷基或C 1-C 4烷氧基所取代,所述的4-6元杂环基选自吡咯烷基、吗啉基、哌嗪基或哌啶基。
本发明优选方案,化合物选自:
Figure PCTCN2019090223-appb-000003
本发明另一方面提供式(I)化合物或其药学上可接受的盐或酯或溶剂化物的制备方法,包括如下步骤:
Figure PCTCN2019090223-appb-000004
其中,R 1、R 2、R 3、R 4、R 5、R 6、X、Y如式(I)化合物所定义。
本发明所述的式(I)化合物或其药学上可接受的盐或酯或溶剂化物是新型VEGFR抑制剂,因而可用于制备预防或治疗血管生成相关疾病的药物,特别是预防或治疗与蛋白质酪氨酸激酶相关的恶性肿瘤疾病。
作为进一步优选的方案,所述肿瘤选自:卵巢癌、***、结肠直肠癌、乳腺癌、胰腺癌、胶质瘤、胶质母细胞瘤、黑色素瘤、***癌、白血病、淋巴瘤、非霍奇金淋巴瘤、胃癌、肺癌、肝细胞癌、胃肠道间质瘤、甲状腺癌、胆管癌、子宫内膜癌、肾癌、间变性大细胞淋巴瘤、急性髓细胞白血病、多发性骨髓瘤、黑色素瘤或间皮瘤、软组织肉瘤。
本发明另一方面提供一种药物组合物,其包括治疗有效剂量的前述式(I)化合物或其药学上可接受的盐或酯或溶剂化物作为活性成份和可药用的载体。
本发明另一方面提供了前述药物组合物在制备预防或治疗恶性肿瘤药物中的应用。
除非有相反陈述,下列用在说明书和权利要求书中的术语具有下述含义。
本发明中“C 1-C 8烷基”指包括1至8个碳原子的直链烷基和含支链烷基,烷基指饱和的脂族烃基团,例如甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基、正庚基、2-甲基己基、3-甲基己基、4-甲基己基、5-甲基己基、2,3-二甲基戊基、2,4-二甲基戊基、2,2-二甲基戊基、3,3-二甲基戊基、2-乙基戊基、3-乙基戊基、正辛基、2,3-二甲基己基、2,4-二甲基己基、2,5-二甲基己基、2,2-二甲基己基、3,3-二甲基己基、4,4-二甲基己基、2-乙基己基、3-乙基己基、4-乙基己基、2-甲基-2-乙基戊基、2-甲基-3-乙基戊基或其各种支链异构体等。
本发明中“环烷基”指饱和单环烃取代基,“C 3-C 8环烷基”指包括3至8个碳原子的单环环烷基,例如:单环环烷基的非限制性实施例包含环丙基、环丁基、环戊基、环己基、环庚基、环辛基等。
本发明中“杂环基”指饱和或部分不饱和单环或多环环状烃取代基,其中一个或多个环原子选自氮、氧或S(O)r(其中r是整数0、1、2)的杂原子,但不包括-O-O-、-O-S-或-S-S-的环部分,其余环原子为碳。“4-10元杂环基”指包含4至10个环原子的环基。单环杂环基的非限制性实施例包含氮杂环丁基、吡咯烷基、哌啶基、哌嗪基、吗啉基、硫代吗啉基、高哌嗪基等。多环杂环基包括螺环、稠环和桥环的杂环基。
本发明中“烷氧基”指-O-(烷基),其中烷基的定义如上所述。“C 1-C 8烷氧基”指含1-8个碳的烷基氧基,非限制性实施例包含甲氧基、乙氧基、丙氧基、丁氧基等。
“卤素”指氟、氯、溴或碘。
“药物组合物”表示含有一种或多种本文所述化合物或其生理学上可药用的盐或前体药物与其他化学组分的混合物,以及其他组分例如生理学可药用的载体和赋形剂。药物组合物的目的是促进对生物体的给药,利于活性成分的吸收进而发挥生物活性。
本发明制备步骤中,所用试剂的缩写分别表示:
DCM               二氯甲烷
DIEA              N,N-二异丙基乙胺
MTBE              甲基叔丁基醚
EA                乙酸乙酯
PE                石油醚
THF               四氢呋喃
TFAA              三氟乙酸酐
EtOH              乙醇
BnBr              溴化苄
DMAP              4-二甲氨基吡啶
TFA               三氟乙酸
MsCl              对甲基苯磺酰氯
DMAC              二甲基乙酰胺
TBDPSCl           叔丁基二苯基氯硅烷
TBAB              四丁基溴化铵
附图说明
图1实施例1化合物的核磁共振氢谱;
图2实施例2化合物的核磁共振氢谱;
图3实施例3化合物的核磁共振氢谱;
具体实施方式
以下参照具体的实施例来说明本发明。本领域技术人员能够理解,这些实施例仅用于说明本发明,其不以任何方式限制本发明的范围。
下述实施例中的实验方法,如无特殊说明,均为常规方法。下述实施例中所用的药材原料、试剂材料等,如无特殊说明,均为市售购买产品。
实施例1
1-(((9-((4-氟-2-甲基-1-氢吲哚-5-基)氧基)-1,2-二氢呋喃并[3,2-f]喹啉-4-基)氧基)甲基)-环丙烷-1-氨基盐酸盐
Figure PCTCN2019090223-appb-000005
步骤1:2,3-二氢苯并呋喃-7-甲酰氯的合成
Figure PCTCN2019090223-appb-000006
冰浴下,向单口瓶中依次加入2,3-二氢苯并呋喃-7-甲酸(13.65g,83.15mmol)、SOCl 2(70ml),60℃下反应1h终止。降至室温后减压浓缩得类白色固体。未做进一步纯化,直接投入下一步反应。
步骤2:N-甲氧基-N-甲基-2,3-二氢苯并呋喃-7-甲酰胺的合成
Figure PCTCN2019090223-appb-000007
室温下,向单口瓶中加入2,3-二氢苯并呋喃-7-甲酰氯(15.18g,83.13mmol),溶于DCM(150ml),冰浴下滴加DIEA(30ml)并加入N,O-二甲基羟胺盐酸盐(9.73g,99.75mmol),室温反应1h终止。用EA萃取,饱和食盐水洗涤两次,有机相经无水硫酸钠干燥,过滤,浓缩,柱层析纯化(EA/PE体系)得黄色固体16.37g,两步收率95%。 1H NMR(400MHz,DMSO-d 6) δ7.30(dd,J=7.3,0.9Hz,1H),7.09(d,J=7.5Hz,1H),6.86(t,J=7.5Hz,1H),4.56(t,J=8.7Hz,2H),3.55(s,3H),3.20(t,J=8.8Hz,2H),3.18(s,3H)。
步骤3:1-(2,3-二氢苯并呋喃-7-基)乙酮的合成
Figure PCTCN2019090223-appb-000008
室温下,向三口瓶中依次加入N-甲氧基-N-甲基-2,3-二氢苯并呋喃-7-甲酰胺(16.37g,78.99mmol)、THF(450ml),N 2置换三次,冰浴下滴加甲基溴化镁的THF溶液(170.00ml,170.00mmol),滴毕后于室温反应1h终止。将反应体系移至冰浴下,用1N的盐酸调节pH至3~4。随后用EA萃取,饱和食盐水洗涤两次,有机相经无水硫酸钠干燥,过滤,浓缩,柱层析纯化(EA/PE体系)得类白色固体11.11g,收率85%。 1H NMR(400MHz,DMSO-d 6)δ7.53(d,J=7.9Hz,1H),7.46(dd,J=7.2,1.1Hz,1H),6.91(t,J=7.5Hz,1H),4.68(t,J=8.8Hz,2H),3.23(t,J=8.8Hz,2H),2.52(s,3H)。
步骤4:1-(5-硝基-2,3-二氢苯并呋喃-7-基)乙酮的合成
Figure PCTCN2019090223-appb-000009
冰浴下,向三口瓶中加入H 2SO 4(50ml),搅拌下分批加入1-(2,3-二氢苯并呋喃-7-基)乙酮(11.10g,68.44mmol)和KNO 3(11.76g,116.32mmol),0℃下反应1.5h终止。将反应液缓慢倒入冰水中搅拌10min,用EA萃取分液,有机相再用饱和碳酸氢钠溶液和饱和食盐水各洗涤一次,有机相经无水硫酸钠干燥,过滤,浓缩,柱层析纯化(EA/PE体系)得黄色固体12.20g,收率86%。 1H NMR(400MHz,DMSO-d 6)δ8.39(d,J=2.2Hz,1H),8.33-8.29(m,1H),4.90(t,J=8.8Hz,2H),3.35(t,J=8.9Hz,2H),2.58(s,3H)。
步骤5:(5-硝基-2,3-二氢苯并呋喃-7-基)乙酸酯的合成
Figure PCTCN2019090223-appb-000010
在单口瓶中加入TFAA(200ml),-10℃下滴加H 2O 2(50ml),滴毕,在该温度下搅拌20min后滴加1-(5-硝基-2,3-二氢苯并呋喃-7-基)乙酮(19.00g,91.70mmol)的DCM溶液(45ml),滴毕后于室温反应3h终止。用EA萃取,饱和食盐水洗涤两次,有机相经无水硫酸钠干燥,过滤,浓缩得黄色固体,未做进一步纯化,直接投入下一步反应。
步骤6:5-硝基-2,3-二氢苯并呋喃-7-醇的合成
Figure PCTCN2019090223-appb-000011
室温下在单口瓶中依次加入(5-硝基-2,3-二氢苯并呋喃-7-基)乙酸酯(20.47g,91.71mmol)、EtOH(250ml),冰浴下滴加40%NaOH溶液(25ml),滴毕后于室温反应0.5h终止。冰浴下用2N盐酸调节pH至3~4,用EA萃取,饱和食盐水洗涤两次,有机相经无水硫酸钠干燥,过滤,浓缩,柱层析纯化(EA/PE体系)得黄色固体15.00g,两步收率90%。 1H NMR(400MHz,DMSO-d 6)δ7.70–7.64(m,1H),7.54(d,J=2.3Hz,1H),4.70(t,J=8.9Hz,2H),3.26(t,J=8.8Hz,2H)。MS(ESI)m/z:180.0[M-H] -
步骤7:7-苄氧基-5-硝基-2,3-二氢苯并呋喃的合成
Figure PCTCN2019090223-appb-000012
室温下,向单口瓶中依次加入5-硝基-7-羟基-2,3-二氢苯并呋喃(15.0g,82.80mmol)、DMF(300ml)、K 2CO 3(171.66g,1242.02mmol)、BnBr(17.00g,99.40mmol),80℃反应0.5h终止。用EA萃取,饱和食盐水洗涤两次,有机相经无水硫酸钠干燥,过滤,浓缩,再用PE(250ml)于室温下打浆30min,抽滤得黄色固体19.42g,收率87%。 1H NMR(400MHz,DMSO-d 6)δ7.87–7.83(m,1H),7.80(d,J=2.0Hz,1H),7.47–7.33(m,5H),5.24(s,2H),4.74(t,J=8.9Hz,2H),3.30(t,J=8.9Hz,2H)。
步骤8:7-苄氧基-2,3-二氢苯并呋喃-5-胺的合成的合成
Figure PCTCN2019090223-appb-000013
室温下,向单口瓶中依次加入7-苄氧基-5-硝基-2,3-二氢苯并呋喃(19.00g,70.04mmol)、EtOH(450ml)、H 2O(112ml)、NH 4Cl(15.69g,293.33mmol),升温至80℃,加入Fe粉(30.00g,537.20mmol),并在该温度下反应1.5h终止。抽滤,滤液中加入H 2O,用EA萃取三次,有机相合并经无水硫酸钠干燥,过滤,浓缩,柱层析纯化(EA/PE体系)得黑色固体14.71g,收率87%。 1H NMR(400MHz,DMSO-d 6)δ7.42–7.29(m,5H),6.14(s,1H),6.11(s,1H),5.00(s,2H),4.37(t,J=8.6Hz,2H),3.01(t,J=8.5Hz,2H)。MS(ESI)m/z:242.2[M+H] +
步骤9:5-(乙氧基亚甲基)-2,2-二甲基-1,3-二噁烷-4,6-二酮的合成
Figure PCTCN2019090223-appb-000014
室温下,向单口瓶中依次加入环-丙二酸异亚丙酯(8.36g,58.00mmol)、原甲酸三乙酯(29.37ml,176.58mmol),80℃反应1h终止。未做进一步处理,直接用于下一步反应。
步骤10:5-((7-苄氧基-2,3-二氢苯并呋喃-5-基氨基)亚甲基)-2,2-二甲基-1,3-二噁烷-4,6-二酮的合成
Figure PCTCN2019090223-appb-000015
室温下,向单口瓶中依次加入7-苄氧基-2,3-二氢苯并呋喃-5-胺(13.99g,57.98mmol)、5-(乙氧基亚甲基)-2,2-二甲基-1,3-二噁烷-4,6-二酮(11.61g,57.99mmol)、异丙醇(350ml),80℃反应0.5h终止。降至室温,抽滤得深黄色固体19.05g,两步收率83%。 1H NMR(400MHz,DMSO-d 6)δ8.50(d,J=16.0Hz,1H),7.46–7.32(m,5H),7.26(d,J=1.1Hz,1H),7.08(s,1H),5.15(s,2H),4.56(t,J=8.8Hz,2H),3.19(t,J=8.7Hz,2H),1.66(s,6H)。MS(ESI)m/z:394.2[M-H] -
步骤11:4-苄氧基-1,2-二氢呋喃并[3,2-f]喹啉-9-醇的合成
Figure PCTCN2019090223-appb-000016
室温下,向微波反应瓶中依次加入5-((7-苄氧基-2,3-二氢苯并呋喃-5-基氨基)亚甲基)-2,2-二甲基-1,3-二噁烷-4,6-二酮(1.5g,3.79mmol)、二苯醚(18ml),70℃水浴搅拌15min后,移至微波中220℃反应0.5h终止。冷却至室温,加入PE打浆1h,抽滤得黄色固体1.02g,收率92%。未做进一步纯化,直接投入下一步反应。 1H NMR(400MHz,DMSO-d 6)δ7.67(t,J=4.0Hz,1H),7.49–7.33(m,5H),6.92(s,1H),5.83(d,J=7.2Hz,1H),5.19(s,2H),4.58(t,J=9.1Hz,2H),3.66(t,J=9.0Hz,2H)。MS(ESI)m/z:294.2[M+H] +
步骤12:4-苄氧基-9-氯-1,2-二氢呋喃并[3,2-f]喹啉的合成
Figure PCTCN2019090223-appb-000017
室温下,向三口瓶中依次加入4-苄氧基-1,2-二氢呋喃并[3,2-f]喹啉-9-醇(6.17g,21.04mmol)、三氯氧磷(60ml),N 2置换三次,107℃反应0.5h终止。浓缩得黑色粗品,经柱层析纯化(MeOH/DCM体系)得黄色固体3.62g,收率55%。 1H NMR(400MHz,DMSO-d 6)δ8.51(d,J=4.8Hz,1H),7.51–7.45(m,4H),7.44–7.33(m,3H),5.32(s,2H),4.76(t,J=9.2Hz,2H),3.88(t,J=9.2Hz,2H)。MS(ESI)m/z:312.2[M+H] +
步骤13:4-苄氧基-9-(4-氟-2甲基-1-氢吲哚-5-基氧基)-1,2-二氢呋喃并[3,2-f]喹啉的合成
Figure PCTCN2019090223-appb-000018
室温下,向微波反应瓶中依次加入4-苄氧基-9-氯-1,2-二氢呋喃并[3,2-f]喹啉(3.62g,11.61mmol)、2-甲基-4-氟-5-羟基-1-氢吲哚(3.84g,23.25mmol)、DMAP(2.41g,19.73mmol)、2,6-二甲基吡啶(72ml),N 2置换三次,微波200℃反应2.5h终止。浓缩后用EA萃取,饱和食盐水洗涤两次,有机相经无水硫酸钠干燥,过滤,浓缩,柱层析纯化(THF/DCM体系)得黄色固体1.29g,收率25%。 1H NMR(400MHz,DMSO-d 6)δ8.35(d,J=5.2Hz,1H),7.52(d,J=7.2Hz,2H),7.45–7.34(m,4H),7.20(d,J=8.6Hz,1H),6.98(t,J=8.1Hz,1H),6.31(d,J=5.1Hz,1H),6.27(s,1H),5.31(s,2H),4.75(t,J=9.1Hz,2H),3.81(t,J=9.1Hz,2H),2.41(s,3H)。MS(ESI)m/z:441.2[M+H] +
步骤14:9-(4-氟-2甲基-1-氢吲哚-5-基氧基)-1,2-二氢呋喃并[3,2-f]喹啉-4-醇的合成
Figure PCTCN2019090223-appb-000019
室温下,向三口瓶中加入4-苄氧基-9-(4-氟-2甲基-1-氢吲哚-5-基氧基)-1,2-二氢呋喃并[3,2-f]喹啉(0.62g,1.41mmol),N 2置换三次,加入TFA(12ml)、H 2O(3ml),85℃反应3h终止。用EA与THF混合溶液萃取,饱和食盐水洗涤两次,水相用EA反萃一次,有机相合并经无水硫酸钠干燥,过滤,浓缩,柱层析纯化(EA/MeOH体系)得黄色固体0.31g,收率50%。 1H NMR(400MHz,DMSO-d 6)δ8.28(d,J=5.2Hz,1H),7.19(d,J=8.6Hz,1H),7.17(s,1H),6.97(t,J=8.1Hz,1H),6.26(s,1H),6.22(d,J=5.1Hz,1H),4.73(t,J=9.1Hz,2H),3.79(t,J=9.1Hz,2H),2.41(s,3H)。MS(ESI)m/z:351.2[M+H] +
步骤15:(1-(叔丁氧基羰基氨基)环丙基)甲磺酸甲酯的合成
Figure PCTCN2019090223-appb-000020
冰浴下,向单口瓶中加入(1-羟甲基环丙基)-叔丁氧羰基氨基(1.00g,5.34mmol)、DCM(10ml)、DIEA(1.38g,10.68mmol),搅拌15min后滴加MsCl(0.673g,5.85mmol), 滴毕后室温反应0.5h终止。用EA萃取,饱和碳酸氢钠溶液洗涤两次,有机相经无水硫酸钠干燥,过滤,浓缩得淡黄色固体1.29g,收率91%。 1H NMR(400MHz,DMSO-d 6)δ4.18(s,2H),3.12(s,3H),1.37(s,9H),0.79(t,J=4.0Hz,2H),0.73(t,J=4.0Hz,2H)。
步骤16:1-((9-(4-氟-2甲基-1-氢吲哚-5-基氧基)-1,2-二氢呋喃并[3,2-f]喹啉-4-基氧基)甲基)环丙基氨基甲酸叔丁酯的合成
Figure PCTCN2019090223-appb-000021
室温下,向三口瓶中加入9-(4-氟-2-甲基-1-氢吲哚-5-基氧基)-1,2-二氢呋喃并[3,2-f]喹啉-4-醇(0.10g,0.29mmol)、(1-(叔丁氧基羰基氨基)环丙基)甲磺酸甲酯(0.30g,1.13mmol)、碳酸铯(0.46g,1.41mmol)、DMAC(6ml),N 2置换三次,100℃反应1h终止。用EA萃取,纯化水洗涤两次,有机相经无水硫酸钠干燥,过滤,浓缩,先正向柱层析纯化(MeOH/DCM),再反相柱纯化(0.05%磷酸水溶液/乙腈体系)得黄色固体0.07g,收率50%。 1H NMR(400MHz,DMSO-d 6)δ7.35(s,1H),7.27(s,1H),7.20(d,J=8.6Hz,1H),6.98(t,J=8.1Hz,1H),6.30(d,J=5.0Hz,1H),6.27(s,1H),4.75(t,J=9.1Hz,2H),4.17(s,2H),3.81(t,J=9.2Hz,2H),2.41(s,3H),1.36(s,9H),0.85(t,J=5.6Hz,2H),0.76(t,J=5.6Hz,2H)。MS(ESI)m/z:520.2[M+H] +
步骤17:1-((9-(4-氟-2-甲基-1-氢吲哚-5-基氧基)-1,2-二氢呋喃并[3,2-f]喹啉-4-基氧基)甲基)-环丙烷-1-氨基盐酸盐的合成
Figure PCTCN2019090223-appb-000022
室温下,向三口瓶中依次加入1-((9-(4-氟-2甲基-1-氢吲哚-5-基氧基)-1,2-二氢呋喃并[3,2-f]喹啉-4-基氧基)甲基)环丙基氨基甲酸叔丁酯(0.04g,0.08mmol)、THF(2ml),N 2置换三次,冰浴下滴加盐酸四氢呋喃溶液,滴毕后40℃反应3h终止。移至冰浴下搅拌0.5h后抽滤得黄色固体0.03g,收率70%。 1H NMR(400MHz,DMSO-d 6)δ8.72(d,J=6.3Hz,1H),7.72(s,1H),7.29(d,J=8.4Hz,1H),7.11(t,J=7.9Hz,1H),6.79(d,J=6.0Hz,1H),6.32(s,1H),4.90(t,J=9.1Hz,2H),4.45(s,2H),3.93(t,J=9.0Hz,2H),2.43(s,3H),1.19(t,J=3.0Hz,2H),1.07(t,J=3.0Hz,2H)。MS(ESI)m/z:418.2[M-H] -
实施例2
9-(4-氟-2-甲基-1-氢吲哚-5-基氧基)-4-(2-(吡咯烷-1-基)-羟乙基)-1,2-二氢呋喃并[3,2-f]喹啉
Figure PCTCN2019090223-appb-000023
步骤1:参照实施例1的步骤1-14合成9-(4-氟-2甲基-1-氢吲哚-5-基氧基)-1,2-二氢呋喃并[3,2-f]喹啉-4-醇。
步骤2:2-(吡咯烷-1-基乙基)甲磺酸酯的合成
Figure PCTCN2019090223-appb-000024
室温下,向单口瓶中依次加入N-(2-羟乙基)-吡咯烷(1.00g,8.68mmol)、NaHCO 3(1.75g,20.83mmol)、DCM(15ml)、MsCl(0.99g,8.64mmol),室温反应3h终止。在反应液中加入无水硫酸钠搅拌干燥,过滤,浓缩,得黄色固体,经DCM打浆,抽滤,得白色固体与滤液,将滤液浓缩,柱层析纯化(氨甲醇溶液/DCM体系)得淡黄色固体0.22g,收率13%。
步骤3:9-(4-氟-2-甲基-1-氢吲哚-5-基氧基)-4-(2-(吡咯烷-1-基)-羟乙基)-1,2-二氢呋喃并[3,2-f]喹啉的合成
Figure PCTCN2019090223-appb-000025
室温下,向三口瓶中依次加入9-(4-氟-2甲基-1-氢吲哚-5-基氧基)-1,2-二氢呋喃并[3,2-f]喹啉-4-醇(0.05g,0.14mmol)、2-(吡咯烷-1-基乙基)甲磺酸酯(0.06g,0.31mmol)、Cs 2CO 3(0.12g,0.37mmol)、DMAC(3ml),N 2置换三次,100℃反应1h终止。用EA萃取,纯化水洗涤两次,有机相经无水硫酸钠干燥,过滤,浓缩,正向柱层析纯化(氨甲醇溶液/MeOH),再反相柱纯化(0.1%甲酸水溶液/乙腈体系),最后正向柱层析纯化(氨甲醇溶液/MeOH)得类白色固体0.01g,收率15%。 1H NMR(400MHz,DMSO-d 6)δ8.36(d,J=5.2Hz,1H),7.33(s,1H),7.20(d,J=8.6Hz,1H),6.98(t,J=8.0Hz,1H),6.30(d,J=5.1Hz,1H), 6.27(s,1H),4.75(t,J=9.0Hz,2H),4.28(t,J=4.9Hz,2H),3.81(t,J=9.1Hz,2H),2.92(t,J=4.0Hz,2H),2.67–2.57(m,4H),2.41(s,3H),1.75–1.69(m,4H)。MS(ESI)m/z:448.2[M+H] +
实施例3
2-(9-(4-氟-2-甲基-1H-吲哚-5-基)氧基)-1,2-二氢呋喃[3,2-f]喹啉-4-基)乙烷氧基)-1-醇
Figure PCTCN2019090223-appb-000026
室温下,在三口瓶中加入9-(4-氟-2甲基-1-氢吲哚-5-基氧基)-1,2-二氢呋喃并[3,2-f]喹啉-4-醇(0.20g,0.57mmol)、2-溴乙醇(0.29g,2.28mmol)、Cs 2CO 3(0.93g,2.85mmol)与DMAC(5mL),氮气保护,60℃反应2.5h终止。EA萃取,饱和食盐水水洗两次,有机相经无水硫酸钠干燥、过滤、浓缩,柱层析纯化(MeOH/DCM体系)得黄色固体产物0.06g,收率28%。 1H NMR(400MHz,DMSO-d 6)δ8.35(d,J=5.1Hz,1H),7.31(s,1H),7.20(d,J=8.5Hz,1H),6.98(t,J=8.0Hz,1H),6.30(d,J=4.9Hz,1H),6.27(s,1H),4.74(t,J=9.0Hz,2H),4.18(t,J=4.2Hz,2H),3.81(t,J=7.0Hz,4H),2.41(s,3H)。MS(ESI)m/z:395.0[M+H] +
实施例4
(S)-1-((9-((4-氟-2-甲基-1H-吲哚-5-基)氧基)-1,2-二氢呋喃[3,2-f]喹啉-4-基)氧基)丙烷-2-醇
Figure PCTCN2019090223-appb-000027
室温下,向三口瓶中加入9-(4-氟-2-甲基-1-氢吲哚-5-基氧基)-1,2-二氢呋喃并[3,2-f]喹啉-4-醇(0.20g,0.57mmol)、Cs 2CO 3(0.93g,2.85mmol)、(S)-1-氯-2-丙醇(0.22g,2.28mmol)与DMAC(3mL),氮气保护,80℃反应2h终止。加入EA萃取,饱和食盐水水洗两次,有机相经无水硫酸钠干燥,过滤,浓缩,柱层析纯化(MeOH/DCM体系)得黄色固体产物0.98g,收率70%。MS(ESI)m/z:409.2[M+H] +
实施例5
(R)-1-((9-((4-氟-2-甲基-1H-吲哚-5-基)氧基)-1,2-二氢呋喃[3,2-f]喹啉-4-基)氧基)丙烷-2-醇
Figure PCTCN2019090223-appb-000028
室温下,向三口瓶中加入9-((4-氟-2甲基-1-氢吲哚-5-基)氧基)-1,2-二氢呋喃并[3,2-f]喹啉-4-醇(0.20g,0.57mmol)、Cs 2CO 3(0.93g,2.85mmol)、(R)-1-氯-2-丙醇(0.22g,2.28mmol)与DMAC(3mL),氮气保护,80℃反应2h终止。加入EA萃取,饱和食盐水水洗两次,有机相经无水硫酸钠干燥,过滤,浓缩,柱层析纯化(MeOH/DCM体系)得黄色固体产物0.13g,收率55%。MS(ESI)m/z:409.2[M+H] +
实施例6
(R)-3-((9-((4-氟-2-甲基-1H-吲哚-5-基)氧基)-1,2-二氢呋喃[3,2-f]喹啉-4-基)氧基)丙烷-1,2-二醇
Figure PCTCN2019090223-appb-000029
室温下,向三口瓶中加入9-(4-氟-2-甲基-1-氢吲哚-5-基氧基)-1,2-二氢呋喃并[3,2-f]喹啉-4-醇(0.20g,0.57mmol)、Cs 2CO 3(0.93g,2.85mmol)、(R)-3-氯-1,2-丙二醇(0.25g,2.28mmol)与DMF(3mL),氮气保护,80℃反应4h终止。加入EA萃取,饱和食盐水水洗两次,有机相经无水硫酸钠干燥,过滤,浓缩,柱层析纯化(MeOH/DCM体系)得黄色固体产物0.09g,收率37%。MS(ESI)m/z:425.1[M+H] +
实施例7
(S)-3-((9-((4-氟-2-甲基-1H-吲哚-5-基)氧基)-1,2-二氢呋喃[3,2-f]喹啉-4-基)氧基)丙烷-1,2-二醇
Figure PCTCN2019090223-appb-000030
室温下,向三口瓶中加入9-(4-氟-2-甲基-1-氢吲哚-5-基氧基)-1,2-二氢呋喃并[3,2-f]喹啉-4-醇(0.20g,0.57mmol)、Cs 2CO 3(0.93g,2.85mmol)、(S)-3-氯-1,2-丙二醇(0.25g,2.28mmol)与DMF(3mL),氮气保护,80℃反应4h终止。加入EA萃取,饱和食盐水水洗两次,有机相经无水硫酸钠干燥,过滤,浓缩,柱层析纯化(MeOH/DCM体系)得黄色固体产物0.10g,收率41%。MS(ESI)m/z:425.1[M+H] +
实施例8
2-(2-((3S,4R)-3,4-二甲氧基吡咯烷-1-基)乙氧基)-9-((4-氟-2-甲基-1H-吲哚-5-基)氧基)-1,2-二氢呋喃[3,2-f]喹啉
Figure PCTCN2019090223-appb-000031
步骤1:叔丁基(3R,4S)-3,4-二甲氧基吡咯烷-1-羧酸酯的合成
Figure PCTCN2019090223-appb-000032
冰浴下,向三口瓶中加入叔丁基(3R,4S)-3,4-二羟基吡咯烷-1-羧酸酯(1.00g,4.92mmol)、NaH(0.30g,12.30mmol)与DMF(10mL),氮气保护,0℃搅拌15min后滴加碘甲烷(1.54g,10.83mmol),滴毕后0℃反应3h终止。加入EA萃取,饱和食盐水水洗两次,有机相经无水硫酸钠干燥、过滤、浓缩,柱层析纯化(PE/EA体系)得淡黄色油状物0.91g,收率80%。
步骤2:(3R,4S)-3,4-二甲氧基吡咯烷盐酸盐的合成
Figure PCTCN2019090223-appb-000033
室温下,向单口瓶中加入叔丁基(3R,4S)-3,4-二甲氧基吡咯烷-1-羧酸酯(0.91g,3.93mmol)与DCM(5mL),冰浴下滴加盐酸乙酸乙酯溶液(1mL),滴毕后20℃反应1h终止。浓缩得黄色固体0.65g,收率98%。
步骤3:4-(2-溴乙氧基)-9-((4-氟-2-甲基-1H-吲哚-5-基)氧基)-1,2-二氢呋喃[3,2-f]喹啉的合成
Figure PCTCN2019090223-appb-000034
室温下,向三口瓶中加入9-(4-氟-2-甲基-1-氢吲哚-5-基氧基)-1,2-二氢呋喃并[3,2-f]喹啉-4-醇(1.00g,2.85mmol)、K 2CO 3(1.18g,8.56mmol)、1,2-二溴乙烷(1.61g,8.56mmol)与DMAC(5mL),氮气保护,90℃反应6h终止。加入EA萃取,饱和食盐水水洗两次,有机相经无水硫酸钠干燥,过滤,浓缩,柱层析纯化(MeOH/DCM体系)得黄色固体0.98g,收率70%。 1H NMR(400MHz,DMSO-d 6)δ8.41(d,J=5.3Hz,1H),7.35(s,1H),7.21(d,J=8.6Hz,1H),7.00(t,J=8.1Hz,1H),6.38(d,J=5.2Hz,1H),6.27(s,1H),4.78(t,J=9.2Hz,2H),4.54(t,J=5.3Hz,2H),3.89(t,J=5.3Hz,2H),3.83(t,J=9.1Hz,2H),2.41(s,3H).MS(ESI)m/z:456.8,458.8[M+H] +
步骤4:4-(2-((3S,4R)-3,4-二甲氧基吡咯烷-1-基)乙氧基)-9-((4-氟-2-甲基-1H-吲哚-5-基)氧基)-1,2-二氢呋喃[3,2-f]喹啉的合成
Figure PCTCN2019090223-appb-000035
室温下,向三口瓶中加入4-(2-溴乙氧基)-9-((4-氟-2-甲基-1H-吲哚-5-基)氧基)-1,2-二氢呋喃[3,2-f]喹啉(0.20g,0.44mmol)、(3R,4S)-3,4-二甲氧基吡咯烷盐酸盐(0.11g,0.66mmol)、Cs 2CO 3(0.43g,1.31mmol)与DMF(8mL),氮气保护,80℃反应2h终止。加入EA萃取,饱和食盐水水洗两次,有机相经无水硫酸钠干燥、过滤、浓缩,反相柱纯化(0.05%磷酸水溶液/乙腈体系)得类白色固体产物0.14g,收率64%。MS(ESI)m/z:508.1[M+H] +
实施例9
4-(2-((3R,4R)-3,4-二甲氧基吡咯烷-1-基)乙氧基)-9-((4-氟-2-甲基-1H-吲哚-5-基)氧基)-1,2-二氢呋喃[3,2-f]喹啉
Figure PCTCN2019090223-appb-000036
步骤1:叔丁基(3S,4S)-3,4-二甲氧基吡咯烷-1-羧酸酯的合成
Figure PCTCN2019090223-appb-000037
冰浴下,向三口瓶中加入叔丁基(3S,4S)-3,4-二羟基吡咯烷-1-羧酸酯(1.00g,4.92mmol)、NaH(0.30g,12.30mmol)与DMF(10mL),氮气保护,0℃搅拌15min后滴加碘甲烷(1.54g,10.83mmol),滴毕后0℃反应3h终止。加入EA萃取,饱和食盐水水洗两次,有机相经无水硫酸钠干燥、过滤、浓缩,柱层析纯化(PE/EA体系)得淡黄色油状物0.80g,收率70%。
步骤2:(3S,4S)-3,4-二甲氧基吡咯烷盐酸盐的合成
Figure PCTCN2019090223-appb-000038
室温下,向单口瓶中加入叔丁基(3S,4S)-3,4-二甲氧基吡咯烷-1-羧酸酯(0.80g,3.93mmol)与DCM(5mL),冰浴下滴加盐酸乙酸乙酯溶液(1mL),滴毕后20℃反应1h终止。浓缩得黄色固体0.55g,收率95%。
步骤3:4-(2-((3R,4R)-3,4-二甲氧基吡咯烷-1-基)乙氧基)-9-((4-氟-2-甲基-1H-吲哚-5-基)氧基)-1,2-二氢呋喃[3,2-f]喹啉的合成
Figure PCTCN2019090223-appb-000039
室温下,向三口瓶中加入4-(2-溴乙氧基)-9-((4-氟-2-甲基-1H-吲哚-5-基)氧基)-1,2-二氢呋喃[3,2-f]喹啉(0.40g,0.88mmol)、(3S,4S)-3,4-二甲氧基吡咯烷盐酸盐(0.22g,1.31mmol)、Cs 2CO 3(0.86g,2.62mmol)与DMF(20mL),氮气保护,80℃反应2h终止。加入EA萃取,饱和食盐水水洗两次,有机相经无水硫酸钠干燥、过滤、浓缩,反相柱纯化(0.05%磷酸水溶液/乙腈体系)得类白色固体产物0.26g,收率60%。MS(ESI)m/z:508.1[M+H] +
实施例10
VEGFR激酶活性测试
本试验采用γ- 33p-ATP同位素测试法测试化合物对激酶VEGFR/VEGFR2/VEGFR3的抑制 作用,并得出化合物对该酶抑制活性的半数抑制浓度IC 50。阳性对照药Anlotinib购于苏州衡宁医药科技有限公司,批号2016120201,Lenvatinib购于湖北威德利化学科技有限公司,批号HBW171008。
1.基础反应缓冲液
20mM Hepes(pH 7.5),10mM MgCl 2,1mM EGTA,0.02%Brij35,0.02mg/ml BSA,0.1mM Na3VO4,2mM DTT,1%DMSO。
2.化合物配制
化合物采用100%DMSO溶解至特定的浓度,之后采用自动加样装置梯度稀释成不同浓度的待测样品(DMSO溶解液)。
3.反应步骤
3.1使用基础反应缓冲液稀释反应底物;
3.2将激酶加入底物溶液中,轻柔混匀;
3.3采用自动加样***将100%DMSO稀释的不同浓度化合物加入激酶溶液中,室温下孵育20min;
3.4室温下加入 33P-ATP(10μM,10μCi/μl)启动激酶反应,反应2h。
4.检测
反应液经离子交换过滤***除去未反应的ATP及反应产生的ADP等离子后检测底物中 33P同位素放射量。
5.数据处理
依据放射量计算加入不同浓度抑制剂体系中的激酶活性从而得到不同浓度化合物对激酶活性的抑制作用,采用graphpad prism拟合得化合物抑制IC 50
本发明化合物的生物化学活性通过以上的试验进行测定,测得的IC 50值参见表1:
表1 VEGFR激酶活性测试结果
Figure PCTCN2019090223-appb-000040
注:表格中“-”代表未测试。
结论:本发明化合物与阳性对照药Anlotinib相比具有更好的激酶抑制活性,特别是VEGFR2抑制活性。
实施例11
VEGF诱导HUVEC细胞增殖抑制试验
本实验采用MTT的方法测试化合物对VEGF诱导的HUVEC细胞活性作用,并得出化合物抑制VEGF诱导的HUVEC细胞增殖活性的半数抑制浓度IC 50
1.HUVEC细胞株在ECM+5%FBS+1%ECGS(内皮细胞生长因子添加剂)+1%P/S(青霉素链霉素混合液)的条件下进行培养。实验前一天用ECM+5%FBS培养。在96孔细胞培养板中接种100μL的处于对数生长期的HUVEC细胞悬液,密度为5X 10  4/ml,将培养板于培养箱中培养24h令细胞贴壁(37℃,5%CO 2)。
2.各化合物已事先溶解在DMSO中配制成10mM的储存液,采用DMSO梯度稀释为目标浓度的400倍,采用无血清培养基稀释到目的浓度的2倍,维持药液中DMSO浓度均为0.5%。在接种细胞的96孔板中依次加入不同浓度药液以及10ng/mL的VEGF165,100μL/孔。每个浓度设3个复孔,并设空白对照、正常对照以及VEGF诱导组,继续在37℃、5%CO 2中继续培养72h。
3.终止培养,每孔加入20μL MTT溶液(5mg/ml),继续在37℃、5%CO 2中继续培养4h后弃除培养基,加入DMSO 150μL/孔,室温震荡10min,在570nM、620nM双波长测OD值(OD 570-OD 620),并经Graphpad Prism 6.0数据处理计算IC 50值。
本发明化合物的生物化学活性通过以上的试验进行测定,测得的IC 50值参见表2:
表2 VEGF诱导HUVEC细胞增殖抑制试验
Figure PCTCN2019090223-appb-000041
结论:本发明化合物2与阳性对照药相比具有更好的抑制VEGF诱导的HUVEC细胞增殖的活性。
实施例12
肝微粒体测试
孵育体系总体积为250μL,用50mmol/LPBS缓冲液(pH=7.4)配制其中含有蛋白浓度为0.5mg/mL的人源肝微粒体孵育液,孵育开始前将100μmol/L待测化合物2.5μL与上述孵育液197.5μL混合,在37℃水浴中预孵育5min后加入同样经预孵育5min的还原性辅酶Ⅱ溶液(5mmol/L)50μL启动反应,(反应体系中种属肝微粒体蛋白含量为0.5g/L、待测化合物终浓度为1μmol/L),在37℃水浴中振荡孵育,并分别于0,5,15,30,60min取出,立即加入内标为Terfenadine(正离子内标,25ng/mL)和Tolbutamide(负离子内标,50ng/mL)的正负内标混合甲醇溶液600μL以终止反应。将终止后的孵育液振荡2min、离心(4℃、16000r/min)10min后取上清液进行LC-MS/MS检测,定量分析母药的剩余量。(DMSO<0.1%)。
将孵育0min化合物的浓度作为100%,其他孵育时间点的浓度转换为百分剩余量,将各时间点的百分剩余量的自然对数对孵育时间作线性回归,求算得斜率k,根据公式,T 1/2=-0.693/k计算得到体外半衰期。肝微粒体中的清除率(CLint(μL/min/mg protein)=Ln(2)*1000/T 1/2(min)/Protein Conc(mg/ml))。
肝微粒体测试数据详见表3:
表3肝微粒体测试结果
Figure PCTCN2019090223-appb-000042
结论:本发明化合物与阳性对照药相比具有相当的人肝微粒体代谢稳定性,因而具有较好的成药性。

Claims (10)

  1. 式(I)所示的化合物、其立体异构体、药学上可接受的盐或酯或溶剂化物:
    Figure PCTCN2019090223-appb-100001
    R 1选自C 1-C 8烷基或C 3-C 8环烷基,任选进一步被一个或多个选自氘、卤素、羟基、氰基、硝基、-NR 8R 9、-NR 8COR 7、-COR 7、-SO 2R 7、-SOR 7、C 1-C 6烷基、C 3-C 6环烷基、C 1-C 8烷氧基或4-10元杂环基的取代基所取代,所述的C 3-C 6环烷基或4-10元杂环基可进一步被氨基、羟基、-(CH 2) nCN、羧基、C 1-C 4烷基或C 1-C 4烷氧基所取代。
    n选自0、1、2或3;
    R 2选自H、氘或卤素;
    X选自N或CH;
    Y选自O或CR 3R 4
    R 3和R 4各自独立选自H、氘、卤素、C 1-C 8烷基、C 3-C 8环烷基或C 1-C 8烷氧基;
    R 5选自H、C 1-C 8烷基或C 3-C 8环烷基;
    R 6选自H、C 1-C 8烷基或C 3-C 8环烷基;
    R 7选自H、C 1-C 8烷基、C 3-C 8环烷基、-NR 8R 9或C 3-C 6杂环基,所述的C 3-C 6杂环基可进一步被羟基、羧基或C 1-C 4烷基所取代;
    R 8和R 9各自独立选自H、C 1-C 8烷基或C 3-C 8环烷基。
  2. 根据权利要求1所述的化合物,其特征在于:
    R 1为C 1-C 4烷基,任选进一步被一个或多个选自氘、羟基、甲基、乙基、环丙基或4-6元杂环基的取代基所取代,所述的环丙基或4-6元杂环基可进一步被氨基、羟基、羧基、C 1-C 4烷基或C 1-C 4烷氧基所取代,所述的4-6元杂环基选自吡咯烷基、吗啉基、哌嗪基或哌啶基。
    R 2选自H、氘或卤素;
    X为CH;
    Y为CH 2
    R 3和R 4各自独立选自H、氘、卤素、C 1-C 8烷基、C 3-C 8环烷基或C 1-C 8烷氧基;
    R 5选自H、C 1-C 8烷基或C 3-C 8环烷基;
    R 6选自H、C 1-C 8烷基或C 3-C 8环烷基。
  3. 根据权利要求1通式(I)所述的化合物、其立体异构体、药学上可接受的盐或酯或溶剂 化物,其为通式(II)所述的化合物、其立体异构体、药学上可接受的盐或酯或溶剂化物:
    Figure PCTCN2019090223-appb-100002
    R 1为C 1-C 8烷基,任选进一步被一个或多个选自氘、卤素、羟基、氰基、-COR 7、C 1-C 6烷基、C 3-C 6环烷基或4-6元杂环基的取代基所取代,所述的C 3-C 6环烷基或4-6元杂环基可进一步被氨基、羟基、-(CH 2) nCN、羧基、C 1-C 4烷基或C 1-C 4烷氧基所取代;
    n选自0、1、2或3;
    R 7选自H、C 1-C 8烷基、C 3-C 8环烷基或C 3-C 6杂环基,所述的C 3-C 6杂环基可进一步被羟基、羧基或C 1-C 4烷基所取代。
  4. 根据权利要求3所述的化合物,其特征在于:
    R 1为C 1-C 4烷基,任选进一步被一个或多个选自氘、卤素、羟基、氰基、C 1-C 2烷基、C 3-C 6环烷基或4-6元杂环基的取代基所取代,所述的C 3-C 6环烷基或4-6元杂环基可进一步被氨基、羟基、-(CH 2) nCN、羧基、C 1-C 4烷基或C 1-C 4烷氧基所取代;
    n选自0、1或2。
  5. 根据权利要求4所述的化合物,其特征在于:
    R 1为C 1-C 4烷基,任选进一步被一个或多个选自氘、羟基、甲基、乙基、环丙基或4-6元杂环基的取代基所取代,所述的环丙基或4-6元杂环基可进一步被氨基、羟基、羧基、C 1-C 4烷基或C 1-C 4烷氧基所取代,所述的4-6元杂环基选自吡咯烷基、吗啉基、哌嗪基或哌啶基。
  6. 化合物、其立体异构体、药学上可接受的盐或酯或溶剂,其特征在于所述的化合物选自:
    Figure PCTCN2019090223-appb-100003
    Figure PCTCN2019090223-appb-100004
  7. 一种药物组合物,其包含如权利要求1~6中任一项所述的化合物、其立体异构体、药学上可接受的盐或酯或溶剂化物和可药用的载体。
  8. 如权利要求7所述的药物组合物,其特征在于,所述药物组合物是胶囊剂、散剂、片剂、颗粒剂、丸剂、注射剂、糖浆剂、口服液、吸入剂、软膏剂、栓剂或贴剂。
  9. 权利要求1~6中任一项所述的化合物或权利要求7~8任一项所述的药物组合物在制备预防或治疗由血管生成介导疾病的治疗药物中的应用。
  10. 权利要求1~6中任一项所述的化合物或权利要求7~8任一项所述的药物组合物在制备预防或治疗恶性肿瘤的药物中的应用。
PCT/CN2019/090223 2018-06-08 2019-06-06 Vegfr抑制剂及其制备方法和应用 WO2019233458A1 (zh)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
CN201810597224.3 2018-06-08
CN201810597224 2018-06-08
CN201910469158.6A CN110577546B (zh) 2018-06-08 2019-05-31 Vegfr抑制剂及其制备方法和应用
CN201910469158.6 2019-05-31

Publications (1)

Publication Number Publication Date
WO2019233458A1 true WO2019233458A1 (zh) 2019-12-12

Family

ID=68769504

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2019/090223 WO2019233458A1 (zh) 2018-06-08 2019-06-06 Vegfr抑制剂及其制备方法和应用

Country Status (1)

Country Link
WO (1) WO2019233458A1 (zh)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101087788A (zh) * 2004-12-22 2007-12-12 拜耳先灵医药股份有限公司 喹啉衍生物、其应用、制备以及包含该化合物的药物
WO2007147578A1 (en) * 2006-06-21 2007-12-27 Bayer Schering Pharma Aktiengesellschaft Quinoline derivatives, their preparation, their use, and medicaments comprising them
CN101809012A (zh) * 2007-03-14 2010-08-18 南京爱德程医药科技有限公司 作为血管生成抑制剂的螺取代化合物
CN103382206A (zh) * 2012-05-04 2013-11-06 上海恒瑞医药有限公司 喹啉或喹唑啉类衍生物、其制备方法及其在医药上的应用
CN106535896A (zh) * 2015-04-27 2017-03-22 江苏恒瑞医药股份有限公司 蛋白激酶抑制剂在制备治疗纤维化疾病的药物中的用途

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101087788A (zh) * 2004-12-22 2007-12-12 拜耳先灵医药股份有限公司 喹啉衍生物、其应用、制备以及包含该化合物的药物
WO2007147578A1 (en) * 2006-06-21 2007-12-27 Bayer Schering Pharma Aktiengesellschaft Quinoline derivatives, their preparation, their use, and medicaments comprising them
CN101809012A (zh) * 2007-03-14 2010-08-18 南京爱德程医药科技有限公司 作为血管生成抑制剂的螺取代化合物
CN103382206A (zh) * 2012-05-04 2013-11-06 上海恒瑞医药有限公司 喹啉或喹唑啉类衍生物、其制备方法及其在医药上的应用
CN106535896A (zh) * 2015-04-27 2017-03-22 江苏恒瑞医药股份有限公司 蛋白激酶抑制剂在制备治疗纤维化疾病的药物中的用途

Similar Documents

Publication Publication Date Title
RU2656591C2 (ru) Модуляторы протеин-тирозинкиназы и способы их применения
TWI751163B (zh) Fgfr4抑制劑、其製備方法和應用
WO2020253860A1 (zh) 芳基磷氧化物类衍生物抑制剂、其制备方法和应用
CA2720552C (en) Acylthiourea compound or salt thereof, and use thereof
CN105121443B (zh) 蛋白激酶抑制剂
BR112016006692A2 (zh) Quinazoline derivatives and method of preparation
CN105315285A (zh) 2,4-二取代7H-吡咯并[2,3-d]嘧啶衍生物、其制法与医药上的用途
EP2740729B1 (en) Quinazoline derivative, composition having the derivative, and use of the derivative in preparing medicament
WO2019233459A1 (zh) 人表皮生长因子受体抑制剂及其制备方法和应用
TW201934546A (zh) 嘧啶類化合物、其製備方法及其醫藥用途
WO2020233669A1 (zh) 含吲哚类衍生物抑制剂、其制备方法和应用
CN108530426A (zh) 含喹喔啉酮的4-苯氧基取代喹啉类化合物及其应用
WO2020001351A1 (zh) Egfr抑制剂及其制备和应用
CN116322697A (zh) 一种喹唑啉化合物及其药物组合物
JP7190755B2 (ja) オキサジノキナゾリンおよびオキサジノキノリン系化合物、ならびに調製方法およびその使用
CN110577546B (zh) Vegfr抑制剂及其制备方法和应用
WO2019233458A1 (zh) Vegfr抑制剂及其制备方法和应用
WO2022100738A1 (zh) 含二并环类衍生物抑制剂自由碱的晶型及其制备方法和应用
CN107235931B (zh) 新型嘧啶类抗肿瘤化合物及其制备方法与用途
WO2019223777A1 (zh) 一种含有芳胺基取代的吡咯并嘧啶类化合物、制备方法及其应用
CN106866642B (zh) 含芳基酰腙结构的喹唑啉类化合物及其应用
WO2022007841A1 (zh) 一种egfr抑制剂、其制备方法和在药学上的应用
JP7022454B2 (ja) ジオキシノキノリン系化合物、その調製方法および使用
CN113979995A (zh) Mek抑制剂及其在医药上的应用
CN112010864B (zh) 血管内皮生长因子受体抑制剂及其制备方法和应用

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: 19814042

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19814042

Country of ref document: EP

Kind code of ref document: A1