WO2018064852A1 - 萘啶化合物和药物组合物及它们的应用 - Google Patents

萘啶化合物和药物组合物及它们的应用 Download PDF

Info

Publication number
WO2018064852A1
WO2018064852A1 PCT/CN2016/105012 CN2016105012W WO2018064852A1 WO 2018064852 A1 WO2018064852 A1 WO 2018064852A1 CN 2016105012 W CN2016105012 W CN 2016105012W WO 2018064852 A1 WO2018064852 A1 WO 2018064852A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
cancer
compound
alkyl
mmol
Prior art date
Application number
PCT/CN2016/105012
Other languages
English (en)
French (fr)
Inventor
黄伟
Original Assignee
南京纳丁菲医药科技有限公司
武汉诺菲科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 南京纳丁菲医药科技有限公司, 武汉诺菲科技有限公司 filed Critical 南京纳丁菲医药科技有限公司
Priority to JP2019540476A priority Critical patent/JP6862563B2/ja
Priority to PL16918185T priority patent/PL3421038T3/pl
Priority to AU2016425880A priority patent/AU2016425880B2/en
Priority to ES16918185T priority patent/ES2784525T3/es
Priority to EP16918185.6A priority patent/EP3421038B1/en
Publication of WO2018064852A1 publication Critical patent/WO2018064852A1/zh

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • Multi-targeted kinase inhibitors can inhibit or block multiple cell growth signal transduction pathways at the same time, which has become a hot spot in tumor therapy and new drug development.
  • Tumor cells can stimulate the secretion of HGF by adjacent fibroblasts by releasing cytokines such as IL-1, FGF-2, and PDGF. Some tumor cells can simultaneously overexpress c-Met and HGF through an autocrine pathway. Overexpression of c-Met can be found in human liver cancer, cholangiocarcinoma, pancreatic cancer, lung cancer, thyroid cancer, pleural stromal tumor, and the like. In metastatic tumors, HGF/c-Met signaling pathway may affect adhesion between tumor cells, promote extracellular matrix degradation, induce angiogenesis, and promote cell proliferation. Targeting the HGF/c-Met signaling pathway, it is relatively easy to achieve simultaneous interference with multiple pathways.
  • VEGFR Vascular endothelial growth factor
  • VEGFR-2 in malignant colorectal cancer, lung cancer, breast cancer and other tumors is also significantly higher than non-tumor tissue.
  • Some drugs target VEGFR signaling responses, either alone or in combination with other chemotherapeutic drugs, and are effective in patients with advanced malignancies.
  • Axl kinase belongs to the TAM family of receptor tyrosine kinases. Binding to the ligand Gas6 activates the tyrosine kinase activity of Axl, thereby activating its downstream signal transduction pathway and participating in cell growth, migration, aggregation and apoptosis. .
  • Recent studies have shown that Axl kinase is overexpressed or activated in a variety of cancers, especially in cancer cells that develop resistance after chemotherapy and receptor tyrosine kinase inhibitor (TKI) treatment. Particularly serious is one of the important reasons for drug resistance. Therefore, Axl kinase inhibitor is a new strategy for the treatment of cancer. (Axl Kinase as a Key Target for Oncology: Focus on Small Molecule Inhibitors. Mol. Cancer Ther. 2014, 13, 2141-2148).
  • RET is a receptor tyrosine kinase that acts as a cell surface molecule capable of converting signals for cell growth and differentiation. This gene plays an important role in the development of neural ridges, and due to cytogenetic rearrangements, in vivo and Carcinogenic activation occurs in vitro. Mutations in the RET gene are associated with multiple endocrine neoplasia, Hirschsprung's disease, and medullary thyroid tumors. Mutations in RET-enhanced mutations result in: medullary thyroid carcinoma, multiple endocrine neoplasia (types 2A, and 2B), pheochromocytoma and parathyroid hyperplasia.
  • RET rearrangement also frequently occurs in non-small cell lung cancer, which is closely related to the occurrence and development of lung cancer. (Development of RET kinase inhibitors for targeted cancer therapy. Curr. Med. Chem. 2011, 18, 162-175).
  • pathological conditions associated with protein kinases include psoriasis, cirrhosis, diabetes, angiogenesis, restenosis, ophthalmic diseases, rheumatoid arthritis and other inflammatory diseases, immune diseases, cardiovascular diseases such as arteriosclerosis and various kidney diseases.
  • Naphthyridine derivatives have a wide range of biological activities and have important applications in the field of medicine.
  • many naphthyridine small molecule compounds have been used as protein kinase inhibitors, widely used in the treatment of a variety of diseases associated with abnormal kinase activity, such as tumors, psoriasis, cirrhosis, diabetes, angiogenesis, ophthalmic diseases, rheumatoid Arthritis and other inflammatory diseases, immune diseases, cardiovascular diseases such as arteriosclerosis and various kidney diseases.
  • 2,7-naphthyridines (WO2013033981, WO0192256, WO0242264), 1,5-naphthyridines (WO2006106046), 1,6-naphthyridines (WO2007060028, WO2010037249, WO2010088177), 2,6-naphthyridines (WO2008122614), heterocyclic fused naphthyridines (WO2009148887, WO2009148916), 2,7-naphthyridones (WO2008109613, WO2009097287, WO201303398), 1,8- Naphthyridinone compounds (WO2010002779) and the like are all used for protein kinase inhibitors.
  • 1,6-Naphthyridine-1(2H)-one is one of the important naphthyridine compounds having a molecular formula of C 8 H 6 N 2 O having a molecular weight of 146.1 and having the chemical structure shown above.
  • 1,6-naphthyridin-1(2H)-keto compounds for the treatment of protein kinase inhibitors has rarely been reported, and only a class of 1,6-naphthyridine-1 as a c-Met kinase inhibitor is disclosed in WO2013097753 patent.
  • (2H)-ketone compound The patent focuses on a series of compounds in which the A fragment is a quinazoline substitution.
  • the present invention has attempted to develop a naphthyridine compound having a more structural type, more excellent kinase inhibitory activity and disease treatment effect.
  • the inventors of the present invention have prepared a naphthyridine compound having a structure represented by the formula (I) by a large amount of scientific research, and found that the naphthyridine compound exhibits excellent inhibitory activity against c-Met and VEGFR-2, which is remarkably superior.
  • the present invention relates to a compound of formula (I):
  • R 3 is selected from the group consisting of H, C 1 -C 3 alkoxy
  • R 5 is selected from the group consisting of H, F, Cl, Br, I, CN, C 1-3 alkyl or C 1-3 haloalkyl.
  • Alkyl represents an unsubstituted or substituted straight or branched chain saturated hydrocarbon group having a certain number of carbon atoms.
  • the alkyl group in the present invention generally has 1 to 6 carbon atoms, further may have 1 to 4 carbon atoms, and most preferably 1 to 3 carbon atoms.
  • Typical alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl , isohexyl, 2,2-dimethylbutyl and 2,3-dimethylbutyl.
  • alkylene group means itself or as a moiety of another substituent, and refers to a linear saturated or unsaturated alkanediyl group having two terminal monovalent group centers, which are derived from two terminal carbon atoms of a linear parent, alkene or alkyne. Each removes a hydrogen atom.
  • Typical alkylene groups include, but are not limited to, methylene, ethylene, vinylidene, ethynylene, propylene, butylene, and the like.
  • Alkoxy represents -O-alkyl wherein alkyl is as defined herein. Typically it is a C 1 -C 6 alkoxy group, preferably a C 1 -C 3 alkoxy group, including but not limited to a methoxy group, an ethoxy group.
  • Thiomorpholino refers to a group having the following chemical structure.
  • Heteroaryl means a monocyclic or fused ring radical of 5 to 10 ring atoms containing one, two, three or four ring heteroatoms selected from N, O or S, the remaining ring atoms being C In addition, it has a fully conjugated ⁇ -electron system.
  • Preferred examples of such groups are pyrrole, furan, thiophene, imidazole, oxazole, isoxazole, thiazole, pyrazole, triazole, pyridine, pyridone, pyrimidine, oxazole, anthrone, quinoline.
  • R 3 is at the 6-position, the 7-position of the quinoline ring or at the 7-position or the 6-position of the quinoline ring, respectively.
  • R 1 and R 2 are selected from a methyl group.
  • R 3 is selected from the group consisting of H, methoxy.
  • R 5 is selected from H, F, Cl, Br, I or CN; further, R 5 is selected from H, F, Cl or Br.
  • the present invention relates to a compound of any one of the following, or a stereoisomer, geometric isomer, tautomer, oxynitride, hydrate, solvate, metabolite, pharmaceutically acceptable salt thereof or the like Prodrugs, but are by no means limited to these compounds:
  • the present invention also encompasses the use of the compound of the present invention and a pharmaceutically acceptable salt thereof, that is, for the treatment of pharmaceutical products sexual angiogenesis-mediated diseases, including those described in the present invention.
  • the compounds of the invention are also useful in the manufacture of a medical article to alleviate, arrest, control or treat a disease mediated by c-Met, VEGFR-2, Axl or RET.
  • the invention comprises a pharmaceutical composition comprising a therapeutically effective amount of a combination of a compound represented by formula (I) and at least one pharmaceutically acceptable carrier, adjuvant or diluent.
  • the invention likewise encompasses a method of treating, or responsive to, an angiogenesis-mediated disease in a patient comprising treating a patient with a therapeutically effective amount of a compound represented by formula (I).
  • the salt is a pharmaceutically acceptable salt.
  • pharmaceutically acceptable includes that the substance or composition must be chemically or toxicologically relevant to the other components of the formulation and to the mammal being treated.
  • organic acids such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid and salicylic acid; pyranoic acid such as glucuronic acid and galactose Aldehydic acid; alpha-hydroxy acids such as citric acid and tartaric acid; amino acids such as aspartic acid and glutamic acid; aromatic acids such as benzoic acid and cinnamic acid; sulfonic acids such as p-toluenesulfonic acid, ethanesulfonic acid, and many more.
  • compositions, formulations and administration of the compounds of the invention are compositions, formulations and administration of the compounds of the invention:
  • the pharmaceutical composition of the present invention comprises a compound of formula (I), a compound of the present invention, or a compound of Examples 1-18, and a pharmaceutically acceptable carrier, adjuvant, or excipient.
  • the amount of the compound in the composition of the present invention is effective to detectably inhibit protein kinases in biological specimens or patients.
  • compositions of the present invention further comprise a pharmaceutically acceptable carrier, adjuvant, or excipient, as used herein, including any solvent, diluent, or other Liquid excipients, dispersing or suspending agents, surfactants, isotonic agents, thickeners, emulsifiers, preservatives, solid binders or lubricants, etc., are suitable for the particular target dosage form.
  • a pharmaceutically acceptable carrier including any solvent, diluent, or other Liquid excipients, dispersing or suspending agents, surfactants, isotonic agents, thickeners, emulsifiers, preservatives, solid binders or lubricants, etc.
  • the pharmaceutical composition of the present invention is characterized by comprising a compound represented by the formula (I) or a compound of the present invention, and a pharmaceutically acceptable carrier, adjuvant or excipient.
  • the amount of the compound in the composition of the present invention is effective to detectably inhibit the activity of a protein kinase such as c-Met, VEGFR-2, Axl or RET.
  • the compounds of the invention or compositions thereof will treat a patient as an anti-tumor drug or reduce the deleterious effects of c-Met, VEGFR-2, Axl or RET signaling.
  • the compounds of the invention will be, but are not limited to, administered to a patient using an effective amount of a compound or composition of the invention to prevent or treat a proliferative disorder in a patient.
  • diseases include cancer, especially metastatic cancer, atherosclerosis and pulmonary fibrosis.
  • the compounds of the invention are also useful in the treatment of ophthalmic conditions such as corneal transplant rejection, neovascularization of the eye, neovascularization of the retina including neovascularization following injury or infection; diabetic retinopathy; posterior lens hyperplasia, and neovascularization Glaucoma; retinal ischemia; vitreous hemorrhage; ulcerative diseases such as gastric ulcer; pathological but non-malignant conditions such as hemangioma, including infantile vascular endothelial cell tumor, aneurysm of nasopharyngeal and avascular necrosis; female reproduction System disorders such as endometriosis. These compounds are also used to treat conditions of edema and vascular hyperpermeability.
  • the compounds of the invention may be used to treat conditions associated with diabetes such as diabetic retinopathy and microangiopathy.
  • the compounds of the invention are also useful in the reduction of blood flow in cancer patients.
  • the compounds of the invention also have beneficial effects on the reduction of tumor metastasis in patients.
  • the compounds of the present invention are also useful in veterinary treatment of pets, introduced species of animals, and farm animals, including mammals, rodents, and the like. Other examples of animals include horses, dogs, and cats.
  • the compounds of the invention include pharmaceutically acceptable derivatives thereof.
  • Figure 1 is a U87MG tumor growth curve of Example 4 and Comparative Compound A.
  • the invention discloses a compound, a preparation method thereof, an intermediate of the compound and a preparation method thereof, and the use of the compound as a c-Met, VEGFR-2, Axl or RET inhibitor, and those skilled in the art can learn from the paper Content, appropriate improvement of process parameters. It is to be understood that all such alternatives and modifications are obvious to those skilled in the art and are considered to be included in the present invention.
  • the method and the application of the present invention have been described by the preferred embodiments, and it is obvious that the method and application described herein may be modified or appropriately modified and combined without departing from the scope of the present invention. The technique of the present invention is applied.
  • Step 3) 5-(((3-benzyloxy)-4-methoxyphenyl) methine)-2,2-dimethyl-1,3-dioxane-4,6- Synthesis of diketones:
  • Step 7) Synthesis of 1-((4-chloro-6-methoxyquinolin-7-yl)oxo)-2-methylpropan-2-ol:
  • Step 8) Synthesis of 1-((4-(2-fluoro-4-nitrophenoxy-6-methoxyquinolin-7-yl)oxo)2-methylpropan-2-ol:
  • Step 9) Synthesis of 1-((4-(4-amino-2-fluorophenyl)-6-methoxyquinolin-7-yl)oxo)2-methylpropan-2-ol:
  • Step 2) Synthesis of 4-(2-fluoro-4-nitrophenoxy)-6-methoxyquinolin-7-ol
  • Step 3) Synthesis of 4-(2-fluoro-4-nitrophenoxy)-6-methoxy-7-(oxiran-2-ylmethoxy)quinoline:
  • Step 4) Synthesis of 1-((4-(4-amino-2-fluorophenyl)quinolin-7-yl)oxo)propan-2-ol:
  • Step 5) 5-((3-Fluoro-4-((7-(2-hydroxypropyl)-6-methoxyquinolin-4-yl)oxy)phenyl)amino)-3-( Synthesis of 4-fluorophenyl)-1,6-naphthyridin-4(1H)-one:
  • the preparation method was the same as in Example 1, except that 5-chloro-3-(4-fluorophenyl)-1,6-naphthyridin-4(1H)-one was changed to 5-chloro-3-phenyl- 1,6-naphthyridin-4(1H)-one.
  • Step 2) Synthesis of 1-(4-chloroquinolin-7-yl)oxo)-2-methylpropan-2-ol:
  • Step 3) Synthesis of 1-((4-(2-fluoro-4-nitrophenyl)quinolin-7-yl)oxo)-2-methylpropan-2-ol:
  • Step 4) Synthesis of 1-((4-(4-amino-2-fluorophenyl)quinolin-7-yl)oxo)-2-methylpropan-2-ol:
  • Step 5) 5-((3-Fluoro-4-((7-(2-hydroxy-2-methylpropyl)quinolin-4-yl)oxy)phenyl)amino)-3-(4) Of -fluorophenyl)-1,6-naphthyridin-4(1H)-one
  • the preparation method was the same as in Example 4 except that 5-chloro-3-(4-fluorophenyl)-1,6-naphthyridin-4(1H)-one was changed to 5-chloro-3-(2- Fluorophenyl)-1,6-naphthyridin-4(1H)-one.
  • the preparation method was the same as in Example 4 except that 5-chloro-3-(4-fluorophenyl)-1,6-naphthyridin-4(1H)-one was changed to 5-chloro-3-(3- Fluorophenyl)-1,6-naphthyridin-4(1H)-one.
  • the preparation method was the same as in Example 4 except that 5-chloro-3-(4-fluorophenyl)-1,6-naphthyridin-4(1H)-one was changed to 5-chloro-3-phenyl- 1,6-naphthyridin-4(1H)-one.
  • the preparation method was the same as in Example 1, except that 2-methoxy-5-nitrophenol was changed to 2-methoxy-4-nitrophenol, and 5-chloro-3-(4-fluorophenyl group was used.
  • The-1,6-naphthyridin-4(1H)-one was changed to 5-chloro-3-phenyl-1,6-naphthyridin-4(1H)-one.
  • Example 14 1-Cyclopropylmethyl-5-((3-fluoro-4-((7-(2-hydroxypropyl)-6-methoxyquinolin-4-yl)oxo)benzene Of amino)amino)-3-(4-fluorophenyl)-1,6-naphthyridin-4(1H)-one
  • Example 16 5-((3-Fluoro-4-((7-(2-hydroxypropyl)-6-methoxyquinolin-4-yl)oxy)phenyl)amino)-3-( Preparation of 4-fluorophenyl)-1-(2-methoxyethyl)-1,6-naphthyridin-4(1H)-one
  • Example 17 5-((3-Fluoro-4-((7-(2-hydroxypropyl)-6-methoxyquinolin-4-yl)oxy)phenyl)amino)-3-( Preparation of 4-fluorophenyl)-1-(2-hydroxyethyl)-1,6-naphthyridin-4(1H)-one
  • Comparative Compound C To further reveal the effect of the 7-position branched alkyl group and the linear alkyl group of the quinoline ring on the kinase inhibitory activity, we also synthesized Comparative Compound C.
  • the preparation method was the same as in Example 4 except that methyl propylene oxide was changed to 2-iodoethanol.
  • Step 1) Synthesis of 3-fluoro-4-((7-(2-methoxy-2-methylpropoxy)quinolin-4-yl)oxo)aniline
  • Example 21 In vitro biochemical level inhibition protein kinase (PK) activity assay
  • test compound was formulated into a mother liquor of 0.5-10 mmol/L with dimethyl sulfoxide (DMSO), and stored at -20 ° C after dispensing;
  • DMSO dimethyl sulfoxide
  • HTRF reagent detection step adding 8 ⁇ L of detection working solution to the well to terminate the reaction; 25 ° C reaction for 1 h;
  • the ratio 665 nm / 620 nm
  • the present invention provides structural formula I as concentration of compounds to inhibit c-Met and VEGFR-2 half (IC 50) shown in Table 1:
  • Example 4 and Example 7 showed good inhibitory activity against Axl kinase with IC 50 of 15.7 and 14.9 nM, respectively; and also showed good inhibitory activity against RET with IC 50 of 62.0 and 53.0 nM, respectively.
  • Comparative Example 4 and Comparative Compound C were administered to rats in the form of a polyethylene glycol 400 aqueous solution (70%). For intravenous administration, rats were given a dose of 1 mg/kg. For oral administration, rats were given a dose of 5 mg/kg.
  • Example 4 and Comparative Compound C Oral group were collected from blood samples of about 0.3 mL to heparinized Eppendorf tubes at 15, 30, 45 min, 1, 2, 4, 6, 8, 10, 24 h after administration, Example 4 and comparison. Approximately 0.3 mL of blood samples were taken from heparinized Eppendorf tubes at 5, 15, 30, 1, 2, 4, 6, 8, 10, and 24 h after administration of Compound C intravenously, and temporarily stored on ice until centrifugation. Whole blood was centrifuged at 8000 rpm for 5 min, and plasma was collected. The plasma was transferred to a 96-well plate and stored at -20 ° C until LC-MS/MS.
  • the pharmacokinetic parameters of the rats after administration were calculated using a non-compartmental model of the software WinNonlin software.
  • Peak concentration Cmax measured value
  • Example 4 has good pharmacokinetic properties including ideal clearance (CL), half-life (t 1/2 ), and exposure (AUC 0-t ).
  • CL ideal clearance
  • t 1/2 half-life
  • AUC 0-t exposure
  • Human tumor cells U87MG glioma cells, MKN45 gastric adenocarcinoma cells, Caki-1 kidney cancer cells, HUH 7 liver cancer cells, NCI-H441 lung adenocarcinoma epithelial cells, MDA-MB-231 breast cancer cells, SMMC-7721 liver cancer cells
  • ATCC ATCC
  • the animals were randomly divided into a solvent control group (70% aqueous solution of PEG-400) and a compound group (6 animals per group). Subsequent administration of the compound to the animal by intragastric administration (3-10 mpk/dose, dissolved in 70% PEG-400 in water), starting anywhere in the 0 to 22 days after tumor cell inoculation, and usually in the trial Once a day.
  • TGI Tumor growth inhibition
  • the evolution of tumors is evaluated by the relationship between tumor volume and time.
  • the long axis (L) and the short axis (W) of the subcutaneous tumor were measured twice a week by a caliper, and the volume of the tumor (TV) was calculated by the formula (L ⁇ W2) / 2).
  • TGI was calculated from the median tumor volume of the solvent group and the median tumor volume of the drug group, expressed as a percentage of the median tumor volume of the solvent control group, and calculated by the following formula:
  • Tumor partial regression Tumor volume is considered to be regression when tumor volume is less than the tumor volume at the end of dosing at the end of the last dose.
  • the original statistical analysis was performed by repeated variance analysis (RMANOVA). Next, multiple comparisons were performed by the Scheffe psot hoc test method. A separate solvent (70% PEG-400, etc.) was used as a negative control.
  • Figure 1 shows the effect of Example 4 on inhibiting tumor growth in the U87MG glioblastoma model.
  • Example 4 was orally administered (p.o.) at a dose of 3 (10D) and 10 mg/kg per day for 22 days. All doses were statistically significant and dose-dependently inhibited the growth of subcutaneous U87MG tumors in nude mice. On the last day of dosing (Day 22), 87.0% and 105.9% (TGI) of the mean tumor volume were inhibited at doses of 3 and 10 mg/kg, respectively, compared to the mean tumor volume of the solvent group.
  • TGI 105.9%

Landscapes

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

Abstract

本发明涉及生物医药领域,公开了萘啶化合物和药物组合物及它们的应用,该萘啶化合物具有式(I)所示的结构或其立体异构体,几何异构体,互变异构体,氮氧化物,水合物,溶剂化物,代谢产物,药学上可接受的盐或它的前药。本发明的萘啶化合物具有明显优于现有技术的抗肿瘤效果,并且本发明的萘啶化合物能够治疗经蛋白激酶中介的疾病。

Description

萘啶化合物和药物组合物及它们的应用 技术领域
本发明涉及生物医药领域,特别涉及一类萘啶化合物,本发明还涉及该类萘啶化合物作为蛋白激酶抑制剂的应用。
背景技术
蛋白激酶是真核细胞中最大的基因家族,对多种细胞过程都起着至关重要的调节作用,如:细胞增殖、细胞死亡、细胞周期进程、分化和细胞存活等。而蛋白酪氨酸激酶(Protein Tyrosine Kinases,PTKs)则是蛋白激酶家族中最为重要的一类。蛋白酪氨酸激酶在正常细胞的信号转导机制中具有重要作用,它们的异常表达将导致许多疾病,特别是肿瘤的产生,因而抑制酪氨酸激酶的过度表达,以恢复生理平衡,将成为一种新的治疗手段。
过去十余年里,人们成功的研发了多个基于酪氨酸激酶信号通路的全新癌症治疗药物。同时酪氨酸激酶抑制剂(Tyrosine Kinases Inhibitors,TKIs)具有分子量小、口服有效和耐受性好的特点,已被批准用于多类肿瘤的治疗,如:肺癌、乳腺癌、肾癌、胰腺癌,以及胃肠道癌症和慢性白血病等。
越来越多的基础研究和临床研究表明:肿瘤是一种多因素,多种信号影响的疾病,发病机制非常复杂(Giamas G.,Man Y.L.,Hirner H.Bischof J,Kramer K,Khan K,Ahmed SS,Stebbing J,Knippschild U.Kinases as targets in the treatment of solid tumors.Cell Signal 2010,22(7),984-1002.)。多靶点激酶抑制剂能够同时抑制或阻断多个细胞生长信号转导通路,已经成为肿瘤治疗和新药研发的热点。
c-Met是酪氨酸激酶家族中的一个重要成员,属于受体酪氨酸激酶(RTK),c-Met最初被认为是致瘤融合蛋白(TPR-MET),现今证明c-Met是由原癌基因MET编码的一种酪氨酸激酶受体,它是肝细胞生长因子(hepatocyte growth factor,HGF)的唯一高亲和受体。在肿瘤发生及进展中,尤其针对具有侵袭和转移潜能的肿瘤,HGF/c-Met信号通路起了至关重要的作用。肿瘤细胞可以通过释放IL-1、FGF-2、和PDGF等细胞因子,刺激邻近的成纤维细胞分泌HGF。有些肿瘤细胞可通过自分泌途径同时过表达c-Met和HGF。c-Met的过表达可见于人肝癌、胆管癌、胰腺癌、肺癌、甲状腺癌、胸膜间质瘤等。在发生转移的肿瘤中,HGF/c-Met信号通路可能影响肿瘤细胞间的黏附、促进细胞外基质降解、诱导血管发生以及促进细胞增殖等。以HGF/c-Met信号通路为靶标,可以相对容易的实现对多条通路的同时干扰,一旦在肿瘤细胞中异常活化并过度表达的HGF/c-Met信号通路被阻断,肿瘤细胞就会出现细胞形态改变、增值减缓、成瘤性降低、侵袭能力下降等一系列变化。(The MET oncogene drives a genetic programme linking cancer to haemostasis.Nature 2005,434,396-400;Drug development of MET inhibitors:targeting oncogene addiction and expedience.Nat.Rev.Drug Discov.2008,7,504-516;Targeting receptor tyrosine kinase MET in cancer:small molecule inhibitors and clinical  progress.J.Med.Chem.2014,57,4427–4453.)
血管内皮生长因子(VEGF)是目前发现的作用最强、特异性最高的促血管生成因子。VEGF在脉管生成、血管形成和血管迁移过程中起重要调节作用,并在多种恶性肿瘤中过度表达,与肿瘤的生长、转移、预后关系密切。VEGFR是一类酪氨酸激酶跨膜糖蛋白VEGFR主要包括VEGFR-1(Flt-1)、VEGFR-2(KDR/Flk-1)、VEGFR-3(Flt-4)这3种受体,其中VEGFR-2是一种特异性糖蛋白,相对分子质量为210000~230000,主要分布在血管内皮细胞和造血干细胞中,可以与VEGF-A、VEGF-C、VEGF-D、VEGF-E结合,主要调节VEGF在血管内皮细胞中的生理反应,包括通透性、增殖和迁移,是生理性和病理性血管生成过程中的一个关键信号传感器。VEGFR-2在卵巢癌、甲状腺癌、黑色素瘤和髓母细胞瘤中过度表达,主要通过调控肿瘤脉管系统(包括血液和淋巴)给大部分肿瘤组织提供营养。此外,VEGFR-2在恶性肠癌、肺癌、乳腺癌等肿瘤中的表达水平亦显著高于非肿瘤组织。一些药物靶向作用于VEGFR信号响应,无论是单独给药,抑或与其它化学治疗药物联用,均对晚期恶性肿瘤患者有效。(An overview of smallmolecule inhibitors of VEGFR signalling.Nat.Rev.Clin.Oncol.2009,6,569-579;Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases.Nat.Rev.Drug Discovery.2011,10,417-427;Vascular Endothelial Growth Factor(VEGF)Receptors:Drugs and New Inhibitors.J.Med.Chem.2012,55,10797-10822)。
Axl激酶属于受体酪氨酸激酶TAM家族,与配体Gas6结合可激活Axl的酪氨酸激酶活性,从而活化其下游的信号转导通路,参与细胞的生长、迁移、聚集和凋亡等进程。近期研究表明,Axl激酶在多种癌症中都存在超表达或激活,特别是在经过化疗和受体酪氨酸激酶抑制剂(TKI)治疗后产生抗药性的癌细胞中,Axl的超表达现象尤为严重,是产生耐药性的重要原因之一,因此,Axl激酶抑制剂是治疗癌症的一个新策略。(Axl Kinase as a Key Target for Oncology:Focus on Small Molecule Inhibitors.Mol.Cancer Ther.2014,13,2141-2148)。
RET是一种受体酪氨酸激酶,作为细胞表面分子能够转换用于细胞生长和分化的信号,此基因在神经脊发育中起重要作用,并且由于细胞遗传学的重排原因,在体内和体外均产生致癌性的激活。RET基因所产生的突变与多发性内分泌肿瘤,先天性巨结肠病,以及甲状腺髓样瘤均相关。RET功能增强性的突变会导致:甲状腺髓样癌,多发性内分泌瘤形成(2A型,和2B型),嗜铬细胞瘤和甲状旁腺增生。RET重排也频繁发生在非小细胞肺癌中,与肺癌的发生发展密切相关。(Development of RET kinase inhibitors for targeted cancer therapy.Curr.Med.Chem.2011,18,162-175)。
与蛋白激酶有关的其它病理状况还包括牛皮癣、肝硬化、糖尿病、血管发生、再狭窄、眼科疾病、类风湿关节炎和其它的炎症疾病、免疫疾病、心血管疾病如动脉硬化和多种肾病。
萘啶衍生物具有广泛的生物活性,在医药领域具有重要的应用。近年来,许多萘啶类小分子化合物已被作为蛋白激酶抑制剂,广泛用于治疗多种与异常激酶活性相关的疾病,如肿瘤、牛皮癣、肝硬化、糖尿病、血管发生、眼科疾病、类风湿关节炎和其它的炎症疾病、免疫疾病、心血管疾病如动脉硬化和多种肾病。其中,2,7-萘啶类化合物(WO2013033981、WO0192256、WO0242264)、1,5-萘啶类化合物(WO2006106046)、1,6-萘啶类化合物(WO2007060028、WO2010037249、 WO2010088177)、2,6-萘啶类化合物(WO2008122614)、杂环稠合萘啶类化合物(WO2009148887、WO2009148916)、2,7-萘啶酮类化合物(WO2008109613、WO2009097287、WO201303398)、1,8-萘啶酮类化合物(WO2010002779)等均用于蛋白激酶抑制剂。
Figure PCTCN2016105012-appb-000001
1,6-萘啶-1(2H)-酮是其中一类重要的萘啶化合物,分子式为C8H6N2O,分子量为146.1,具有如上所示的化学结构。
Figure PCTCN2016105012-appb-000002
WO2013097753公开的化合物的结构通式
1,6-萘啶-1(2H)-酮类化合物用于治疗蛋白激酶抑制剂鲜见报道,仅WO2013097753专利中公开了一类作为c-Met激酶抑制剂的1,6-萘啶-1(2H)-酮类化合物。该专利重点研究的为A片段为喹唑啉取代的一系列化合物。但分析已上市的一些激酶抑制剂的结构特点,我们发现A片段为喹啉环时,化合物的成药性可能更为理想。因此,本发明尝试开发结构类型更丰富,激酶抑制活性和疾病治疗效果更为优异的萘啶类化合物。
发明内容
本发明的目的在于提供一类新型萘啶化合物,用于治疗各种类型的肿瘤。
本发明的发明人通过大量的科学研究,制备出具有式(I)所示结构的萘啶化合物,并发现该萘啶化合物对c-Met和VEGFR-2表现出优异的抑制活性,明显优于WO2013097753专利中公开的典型化合物A和B。更为重要的是,该类萘啶化合物在动物水平上的抗肿瘤活性同样明显优于典型化合物A,从而表现出更为优异的肿瘤治疗效果。
Figure PCTCN2016105012-appb-000003
本发明涉及一种如式(I)所示的化合物:
Figure PCTCN2016105012-appb-000004
或其立体异构体,几何异构体,互变异构体,氮氧化物,水合物,溶剂化物,代谢产物,药学上可接受的盐或它的前药,其中:
R1和R2选自H,C1-C3烷基;
R3选自H,C1-C3烷氧基;
R4选自H、C1-C6烷基、3至8元全碳单环环烷基、3至8元杂脂环基,其中C1-C6烷基、3至8元全碳单环环烷基、3至8元杂脂环基可选进一步被一个或多个选自C1-C6烷基、C1-C6烷氧基、3至8元杂脂环基、C6-C10芳基、C5-C10杂芳基或羟基的取代基所取代;
R5选自H,F,Cl,Br,I,CN,C1-3烷基或C1-3卤代烷基。
烷基表示具有一定数目之碳原子的未经取代或经取代的直链或支链饱和烃基。本发明中的烷基一般是具有1-6个碳原子数,进一步可具有1-4个碳原子数,最优选1-3个碳原子数。典型的烷基包括(但不限于)甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、正戊基、异戊基、新戊基、正己基、异己基、2,2-二甲基丁基和2,3-二甲基丁基。
亚烷基表示本身或作为另一取代基的部分,系指具有两个终端单价基中心之直链饱和或不饱和烷二基,其从直链母烷、烯或炔的两个终端碳原子各移除一个氢原子而得。典型的亚烷基包括(但不局限于)亚甲基、亚乙基、亚乙烯基、亚乙炔基、亚丙基、亚丁基等。
烷氧基表示-O-烷基,其中烷基如本文定义。典型地为C1-C6烷氧基,优选C1-C3烷氧基,包括但不局限于甲氧基、乙氧基。
卤素或卤素基团为氟、氯、溴或碘,优选为氟、氯、溴。C1-3卤代烷基表示其中1个或多个氢被卤素置换的烷基,优选含一个、两个或三个卤素基团。
3至8元全碳单环环烷基包括(但不限于)环丙烷、环丁烷、环戊烷、环己烷。
杂脂环基表示含1个或多个N、O或S的杂原子的单环或稠合的环。典型地为含1个或多个N、O或S的杂原子的3-8原杂环基,优选含1个或多个N、O或S的杂原子的3-6元杂环基,例如环氧丙烷基、环氧丁烷、哌嗪子基、吗啉代基、哌啶子基、吡咯烷基及其衍生物等。
哌嗪子基指的是具有以下化学结构的基团。
Figure PCTCN2016105012-appb-000005
吗啉代基指的是具有以下化学结构的基团。
Figure PCTCN2016105012-appb-000006
硫代吗啉代基指的是具有以下化学结构的基团。
Figure PCTCN2016105012-appb-000007
哌啶子基指的是具有以下化学结构的基团。
Figure PCTCN2016105012-appb-000008
吡咯烷基指的是具有以下化学结构的基团。
Figure PCTCN2016105012-appb-000009
“芳基”表示6至10个碳原子的全碳单环或稠合多环基团,具有完全共轭的π电子系统。“芳基”包括:苯;萘等,芳基可以是取代的或未取代的。
“杂芳基”表示5至10个环原子的单环或稠合环基团,含有一个、两个、三个或四个选自N、O或S的环杂原子,其余环原子是C,另外具有完全共轭的π电子系统。杂芳基包括但不限于,吡咯、呋喃、噻吩、咪唑、噁唑、异噁唑、噻唑、吡唑、三氮唑、吡啶、吡啶酮、嘧啶、吡嗪、哒嗪、吲哚、吲唑、氮杂吲哚、苯并咪唑、吲哚啉、吲哚酮、喹啉、异喹啉、喹唑啉、苯并呋喃、苯并咪唑、苯并噁唑、噻吩并吡啶、噻吩并嘧啶等。此类基团的优选实施例为吡咯、呋喃、噻吩、咪唑、噁唑、异噁唑、噻唑、吡唑、三氮唑、吡啶、吡啶酮、嘧啶、吲唑、吲哚酮、喹啉。
在一种优选方案中,
Figure PCTCN2016105012-appb-000010
R3分别在喹啉环6-位、7-位或者分别在喹啉环7-位、6-位。
进一步优选的,R1和R2选自H,C1-C3烷基,且R1和R2不同时为氢。
更进一步的,R1和R2选自甲基。R3选自H,甲氧基。
在一种优选方案中,R4选自H、C1-C6烷基、3至8元全碳单环环烷基、3至8元杂脂环基C1-C6亚烷基、C1-C6烷氧基C1-C6亚烷基或C6-C10芳基C1-C6亚烷基。
更进一步的,R4选自氢、甲基、乙基、丙基、异丙基、环丙基、
Figure PCTCN2016105012-appb-000011
2-甲氧基乙基、2-羟基乙基或苄基。
在一种优选方案中,R5选自H,F,Cl,Br,I或CN;更进一步的,R5选自H,F,Cl或Br。
喹啉环的6-位和7-位所指的取代位置如下所示:
Figure PCTCN2016105012-appb-000012
本发明涉及到以下其中之一的化合物或其立体异构体,几何异构体,互变异构体,氮氧化物,水合物,溶剂化物,代谢产物,药学上可接受的盐或它的前药,但绝不限于这些化合物:
Figure PCTCN2016105012-appb-000013
Figure PCTCN2016105012-appb-000014
本发明还包含本发明的化合物及其药学上可接受的盐的应用,即用于生产医药产品治疗急慢 性血管发生介导的疾病,包括那些本发明所描述的。本发明的化合物在生产抗癌药物中的应用。本发明的化合物同样用于生产一种医药用品来减轻,阻止,控制或治疗由c-Met、VEGFR-2、Axl或RET所介导的疾病。本发明包含药物组合物,该药物组合物包括式(I)所代表的化合物与至少一个药学上可接受的载体,辅剂或稀释剂的结合所需的有效治疗用量。
本发明同样包含治疗患者血管发生介导的疾病,或对此病症敏感的方法,该方法包含使用式(I)所代表化合物的治疗有效量对患者进行治疗。
除非其他方面表明,本发明的化合物所有的立体异构体,几何异构体,互变异构体,氮氧化物,水合物,溶剂化物,代谢产物和药学上可接受的前药都属于本发明的范围。
具体地说,盐是药学上可接受的盐。术语“药学上可接受的”包括物质或组合物必须是适合化学或毒理学地,与组成制剂的其他组分和用于治疗的哺乳动物有关。
如果本发明的化合物是碱性的,则想得到的盐可以通过文献上提供的任何合适的方法制备得到,例如,使用无机酸,如盐酸,氢溴酸,硫酸,硝酸和磷酸等等。或者使用有机酸,如乙酸,马来酸,琥珀酸,扁桃酸,富马酸,丙二酸,丙酮酸,草酸,羟乙酸和水杨酸;吡喃糖酸,如葡萄糖醛酸和半乳糖醛酸;α-羟酸,如柠檬酸和酒石酸;氨基酸,如天门冬氨酸和谷氨酸;芳香族酸,如苯甲酸和肉桂酸;磺酸,如对甲苯磺酸,乙磺酸,等等。
如果本发明的化合物是酸性的,则想得到的盐可以通过合适的方法制备得到,如,使用无机碱或有机碱,如氨(伯氨,仲氨,叔氨),碱金属氢氧化物或碱土金属氢氧化物,等等。合适的盐包括,但并不限于,从氨基酸得到的有机盐,如甘氨酸和精氨酸,氨,如伯氨、仲氨和叔氨,和环状氨,如哌啶,吗啉和哌嗪等,和从钠,钙,钾,镁,锰,铁,铜,锌,铝和锂得到无机盐。
本发明的化合物的组合物,制剂和给药:
根据另一方面,本发明的药物组合物的特点包括式(I)的化合物,本发明所列出的化合物,或实施例1-18的化合物,和药学上可接受的载体,辅剂,或赋形剂。本发明的组合物中化合物的量能有效地可探测地抑制生物标本或患者体内的蛋白激酶。
本发明的化合物存在自由形态,或合适的、作为药学上可接受的衍生物。根据本发明,药学上可接受的衍生物包括,但并不限于,药学上可接受的前药,盐,酯,酯类的盐,或能直接或间接地根据患者的需要给药的其他任何加合物或衍生物,本发明其他方面所描述的化合物,其代谢产物或他的残留物。
像本发明所描述的,本发明药学上可接受的组合物进一步包含药学上可接受的载体,辅剂,或赋形剂,这些像本发明所应用的,包括任何溶剂,稀释剂,或其他液体赋形剂,分散剂或悬浮剂,表面活性剂,等渗剂,增稠剂,乳化剂,防腐剂,固体粘合剂或润滑剂,等等,适合于特有的目标剂型。如以下文献所描述的:In Remington:The Science and Practice of Pharmacy,21st edition,2005,ed.D.B.Troy,Lippincott Williams&Wilkins,Philadelphia,and Encyclopedia of Pharmaceutical Technology,eds.J.Swarbrick and J.C.Boylan,1988-1999,Marcel  Dekker,New York,综合此处文献的内容,表明不同的载体可应用于药学上可接受的组合物的制剂和它们公知的制备方法。除了任何常规的载体媒介与本发明的化合物不相容的范围,例如所产生的任何不良的生物效应或与药学上可接受的组合物的任何其他组分以有害的方式产生的相互作用,它们的用途也是本发明所考虑的范围。
本发明的化合物和组合物的用途:
本发明的药物组合物的特征包括式(I)所示的化合物或本发明所列出的化合物,以及药学上可接受的载体,辅剂或赋形剂。本发明的组合物中化合物的量可以有效地可探测地抑制蛋白激酶如c-Met、VEGFR-2、Axl或RET的活性。本发明化合物或其组合物将作为抗肿瘤药物对患者进行治疗或减小c-Met、VEGFR-2、Axl或RET信号响应的有害作用。
本发明的化合物将应用于,但绝不限于,使用本发明的化合物或组合物的有效量对患者给药来预防或治疗患者增殖性疾病。这样的疾病包括癌症,尤其是转移癌,动脉粥样硬化和肺纤维化。
本发明的化合物将应用于瘤的治疗包括癌症和转移癌,进一步包括但并不限于,癌症如膀胱癌,乳腺癌,结肠癌,肾癌,肝癌,肺癌(包括小细胞肺癌),食道癌,胆囊癌,卵巢癌,胰腺癌,胃癌,宫颈癌,甲状腺癌,前列腺癌,和皮肤癌(包括鳞状细胞癌);淋巴系统造血肿瘤(包括白血病,急性淋巴囊肿性白血病,急性成淋巴细胞性白血病,B细胞淋巴瘤,T细胞淋巴瘤,何杰金(氏)淋巴瘤,非何杰金(氏)淋巴瘤,多毛细胞白血病和伯基特淋巴瘤);骨髓系统造血肿瘤(包括急慢性骨髓性粒细胞性白血病,骨髓增生异常综合症,和前髓细胞白血病);间充质细胞起源的肿瘤(包括纤维肉瘤和横纹肌肉瘤,和其他肉瘤,如软组织和软骨);中枢末梢神经系统瘤(包括星形细胞瘤,成神经细胞瘤,神经胶质瘤,和神经鞘瘤);和其他肿瘤(包括黑素瘤,精原细胞瘤,畸胎癌,骨肉瘤,萎缩性着色血管瘤(xenoderoma pigmentosum),角化棘皮瘤(keratoctanthoma),甲状腺滤泡瘤和卡波济(氏)肉瘤)。
本发明的化合物还可用于治疗眼科病症例如角膜移植排斥,眼的新生血管形成,视网膜新生血管形成包括损伤或感染后的新生血管形成;糖尿病性视网膜病;晶状体后纤维组织增生症,和新生血管性青光眼;视网膜缺血;玻璃体出血;溃疡性疾病如胃溃疡;病理学的但非恶性状况如血管瘤,包括婴儿血管内皮细胞瘤,鼻咽和无血管性骨坏死的血管纤维瘤;雌性生殖系统紊乱如子宫内膜异位。这些化合物同样也用于治疗水肿和脉管通透性过高的状况。
本发明的化合物可以用于处理与糖尿病相关的情况如糖尿病性视网膜病和微血管病。本发明的化合物同样用于癌症患者血流量减少的情况。本发明的化合物对患者肿瘤转移减少也有有益效果。
本发明的化合物除了对人类治疗有益以外,还可应用于兽医治疗宠物、引进品种的动物和农场的动物,包括哺乳动物,啮齿类动物等等。另外一些动物的实例包括马、狗和猫。在此,本发明的化合物包括其药学上可接受的衍生物。
本发明的其它特征和优点将在随后的具体实施方式部分予以详细说明。
附图说明
附图是用来提供对本发明的进一步理解,并且构成说明书的一部分,与下面的具体实施方式一起用于解释本发明,但并不构成对本发明的限制。在附图中:
图1是实施例4与对比化合物A的U87MG肿瘤生长曲线。
具体实施方式
以下对本发明的具体实施方式进行详细说明。应当理解的是,此处所描述的具体实施方式仅用于说明和解释本发明,并不用于限制本发明。
在本文中所披露的范围的端点和任何值都不限于该精确的范围或值,这些范围或值应当理解为包含接近这些范围或值的值。对于数值范围来说,各个范围的端点值之间、各个范围的端点值和单独的点值之间,以及单独的点值之间可以彼此组合而得到一个或多个新的数值范围,这些数值范围应被视为在本文中具体公开。
本发明公开了一种化合物及其制备方法、该化合物的中间体及其制备方法,及其该化合物作为c-Met、VEGFR-2、Axl或RET抑制剂的应用,本领域技术人员可以借鉴本文内容,适当改进工艺参数实现。特别需要指出的是,所有类似的替换和改动对本领域技术人员来说是显而易见的,它们都被视为包括在本发明。本发明的方法及应用已经通过较佳实施例进行了描述,相关人员明显能在不脱离本发明内容、精神和范围内对本文所述的方法和应用进行改动或适当变更与组合,来实现和应用本发明技术。
下面结合实施例,进一步阐述本发明:
实施例1:5-((3-氟-4-((7-(2-羟基-2-甲基丙基)-6-甲氧基喹啉-4-基)氧代)苯基)氨基)-3-(4-氟苯基)-1,6-萘啶-4(1H)-酮的制备
Figure PCTCN2016105012-appb-000015
步骤1):2-苄氧基-1-甲氧基-4-硝基苯的合成:
在室温条件下,将2-甲氧基-5-硝基苯酚(5g,29.6mmol)溶于60mL N,N-二甲基甲酰胺(DMF),磁力搅拌,加入无水K2CO3(6.1g,44.2mmol)。然后向体系中缓慢加入溴化苄(5.56g,32.5mmol),升温到40℃,反应过夜,TLC监测(石油醚:丙酮=20:5,Rf=0.4)。减压蒸馏除去大部分的DMF,在搅拌的条件下,将剩余体系倒入冰水中,有大量固体析出。过滤,滤饼用碱溶液洗,直至滤液为无色,滤饼干燥,得暗红色固体,收率:99%。
步骤2):3-苄氧基-4-甲氧基苯胺的合成:
将2-苄氧基-1-甲氧基-4-硝基苯(7.1g,23.3mmol),NH4Cl(4.4g,82.2mmol)加入到200mL的梨形瓶中,向其中加入乙醇(72mL),水(24mL)。在室温,磁力搅拌条件下,加入铁粉(12.3g,219mmol)。回流反应1h,TLC监测(石油醚:丙酮=20:7,Rf=0.4)。冷却至50℃,硅藻土过滤,滤液减压浓缩尽量除去乙醇,加水并用乙酸乙酯萃取。有机相用饱和K2CO3洗两次,水洗两次,用无水硫酸钠干燥,浓缩得黑色固体。收率:84%。
步骤3):5-(((3-苄氧基)-4-甲氧基苯基)次甲基)-2,2-二甲基-1,3-二氧六环-4,6-二酮的合成:
将3-苄氧基-4-甲氧基苯胺(5g,21.7mmol),丙二酸环亚异丙酯(3.756g,26mmol)溶于无水乙醇(55mL)。在磁力搅拌的条件下,向其中缓慢加入原甲酸三乙酯(3.86g,26mmol),然后加热回流1h,有大量茶绿色固体析出。在冰浴下继续搅拌2h,过滤,滤饼用冷无水乙醇洗,得到茶绿色固体。收率:92%。
步骤4):7-苄氧基-6-甲氧基-喹啉-4-醇的合成:
将5-(((3-苄氧基)-4-甲氧基苯基)次甲基)-2,2-二甲基-1,3-二氧六环-4,6-二酮(5g,13mmol)加入到邻二氯苯(40mL)中,得到悬浮液。加热到180℃,反应7h,TLC监测(二氯甲烷:甲醇=20:1,Rf=0.2)。冷却至室温,然后冰浴下继续搅拌2h,过滤,滤饼用冷邻二氯苯洗,再用乙醚洗,干燥得土黄色固体。收率:54%。
步骤5):7-苄氧基-4-氯-6-甲氧基-喹啉的合成:
将7-苄氧基-6-甲氧基-喹啉-4-醇(2g,7.1mmol),加入到重蒸甲苯(10mL)中,得到悬浮液,缓慢加入重蒸POCl3(1.1g,7.2mmol)。加热到120℃,反应1.5h,TLC监测(石油醚:丙酮=20:6,Rf=0.4)。冷却至室温,加水,用3M NaOH溶液调节pH=8,过滤,水洗,干燥得土黄色固体。收率:90%。
步骤6)4-氯-6-甲氧基喹啉-7-醇的合成:
将7-苄氧基-4-氯-6-甲氧基-喹啉(1.675,5.58mmol),冰醋酸(10mL)加入到50mL梨形瓶中,磁力搅拌下出现大量白色固体。再向体系中加入溴化氢水溶液(40%,10mL),升温至80℃反应3h。反应体系冷却至45℃左右,倒入80mL乙醚中搅拌,有大量白色固体析出,抽滤,乙醚洗涤,真空干燥得白色固体。收率:80%。
步骤7):1-((4-氯6-甲氧基喹啉-7-基)氧代)-2-甲基丙-2-醇的合成:
将4-氯-6-甲氧基喹啉-7-醇(50mg,0.24mmol)溶于THF/H2O混合溶剂(3mL,THF/H2O=1:1,V/V)中,依次向其中加入NaOH(30mg,0.75mmol)和甲基环氧丙烷(172mg,2.4mmol)。在 45℃下,搅拌72h,用乙酸乙酯稀释,然后用1N NaOH(10mL×4)洗母液,再用饱和食盐水洗,有机相用无水硫酸钠干燥。经柱层析纯化(TLC,石油醚:丙酮=20:5,Rf=0.45)得白色固体。收率:42.4%。
步骤8):1-((4-(2-氟-4-硝基苯氧基-6-甲氧基喹啉-7-基)氧代)2-甲基丙-2醇的合成:
将1-((4-氯6-甲氧基喹啉-7-基)氧代)-2-甲基丙-2-醇(570mg,2.02mmol)和2-氟-4-硝基苯酚(475mg,3.02mmol)悬浮在重蒸甲苯(10mL)中,然后向反应体系中加入N,N-二异丙基乙胺(DIPEA,520mg,4.03mmol)。在油浴温度120℃条件下,反应48h。用乙酸乙酯稀释反应体系,然后用1N NaOH洗有机相,饱和食盐水洗,无水硫酸钠干燥。经柱层析纯化(TLC,石油醚:丙酮=20:8,Rf=0.3)得灰色固体。收率:53%。
步骤9):1-((4-(4-氨基-2-氟苯基)-6-甲氧基喹啉-7-基)氧代)2-甲基丙-2醇的合成:
将1-((4-(2-氟-4-硝基苯氧基-6-甲氧基喹啉-7-基)氧代)2-甲基丙-2醇(410mg,1.02mmol),氯化铵(163mg,3.05mmol),乙醇:水=3:1(12mL)加入到25mL的梨形瓶,加入铁粉(399mg,7.125mmol),油浴温度80℃,反应1h。冷却到50℃左右,硅藻土抽滤,滤饼用乙酸乙酯洗。母液用乙酸乙酯萃取,合并有机相,无水硫酸钠干燥,经柱层析纯化得土黄色固体。收率:84%。
步骤10):5-((3-氟-4-((7-(2-羟基-2-甲基丙基)-6-甲氧基喹啉-4-基)氧代)苯基)氨基)-3-(4-氟苯基)-1,6-萘啶-4(1H)-酮的合成:
将1-((4-(4-氨基-2-氟苯基)-6-甲氧基喹啉-7-基)氧代)2-甲基丙-2醇(60mg,0.16mmol)和5-氯-3-(4-氟苯基)-1,6-萘啶-4(1H)-酮(44mg,0.16mmol)加入到25mL的梨形瓶中,然后加入异丙醇(6mL),在磁力搅拌下加入一水合对甲苯磺酸(PTSA·H2O,37mg,0.195mmol),升温至90℃反应1h。过滤不溶物,滤饼用异丙醇洗涤后干燥得白色固体。1H NMR(600MHz,DMSO-d6):δ13.40(s,1H),12.96(s,1H),8.88(d,J=6.3Hz,1H),8.23–8.33(m,2H),8.11(d,J=6.0Hz,1H),7.79(s,1H),7.77–7.71(m,2H),7.69(s,1H),7.68–7.63(m,1H),7.61(d,J=8.4Hz,1H),7.29(t,J=8.4Hz,2H),7.17(d,J=4.2Hz,1H),7.01(d,J=5.7Hz,1H),4.07(s,3H),3.99(s,2H),1.29(s,6H).
实施例2:5-((3-氟-4-((7-(2-羟基丙基)-6-甲氧基喹啉-4-基)氧代)苯基)氨基)-3-(4-氟苯基)-1,6-萘啶-4(1H)-酮的制备
Figure PCTCN2016105012-appb-000016
步骤1):7-苄氧基-4-(2-氟4-硝基苯氧基)-6-甲氧基喹啉的合成:
向7-苄氧基-4-氯-6-甲氧基喹啉(2.5g,8.3mmol),2-氟-4-硝基苯酚(2.6g,16.6mmol)中加入重蒸甲苯(25mL),在磁力搅拌下,加入DIPEA(2.7g,20.9mmol),加热到120℃回流48h,TLC监测(石油醚:丙酮=20:8,Rf=0.4)。冷却至室温加入200mL乙酸乙酯,用1M NaOH溶液洗有机相,直到水相无色,有机相用无水硫酸钠干燥,浓缩有机相,加乙醚重结晶,得到土黄色固体。收率:80%。
步骤2):4-(2-氟4-硝基苯氧基)-6-甲氧基喹啉-7-醇的合成;
向7-苄氧基-4-(2-氟4-硝基苯氧基)-6-甲氧基喹啉(1.5g,3.6mmol)中加入10mL冰醋酸,搅拌,出现大量白色固体。再向其中加入溴化氢水溶液(40%,10mL),加热至80℃,反应4h,TLC监测(二氯甲烷:甲醇=40:1,Rf=0.2)。冷却至室温,将体系倾入100mL乙醚中,搅拌2h。过滤,滤饼用乙醚洗,得白色固体。收率:90%。
步骤3):4-(2-氟4-硝基苯氧基)-6-甲氧基-7-(环氧乙烷-2-基甲氧基)喹啉的合成:
将4-(2-氟4-硝基苯氧基)-6-甲氧基喹啉-7-醇(500mg,1.51mmol),无水碳酸钾(627mg,4.54mmol)加入到25mL的梨形瓶中,然后加入DMF(13mL),环氧氯丙烷(700mg,7.56mmol),升温至80℃,反应10小时。冷却至室温,倒入水中,用乙酸乙酯萃取,合并有机相,干燥,柱层析得到化合物。收率:30%。
步骤4):1-((4-(4-氨基-2-氟苯基)喹啉-7-基)氧代)丙-2醇的合成:
将4-(2-氟4-硝基苯氧基)-6-甲氧基-7-(环氧乙烷-2-基甲氧基)喹啉(146mg,0.378mmol)溶于甲醇(10mL)和二氯甲烷(15mL)的混合溶剂中。在搅拌状态下,向其中加入Pd/C(30mg),氢气氛下室温反应过夜。反应液除去溶剂后经柱层析纯化得浅黄色固体。收率:63%。
步骤5):5-((3-氟-4-((7-(2-羟基丙基)-6-甲氧基喹啉-4-基)氧代)苯基)氨基)-3-(4-氟苯基)-1,6-萘啶-4(1H)-酮的合成:
将1-((4-(4-氨基-2-氟苯基)喹啉7-基)氧代)丙-2醇和5-氯-3-(4-氟苯基)-1,6-萘啶-4(1H)-酮(44mg,0.16mmol)加入到10mL的梨形瓶中,然后加入异丙醇(4mL),在磁力搅拌下加入PTSA·H2O(38mg,0.2mmol),升温至90℃反应1小时。冷却至室温无固体析出,除去异丙醇,经柱层析纯化得黄色固体。收率:47%。1H NMR(400MHz,DMSO-d6):δ13.22(s,1H),12.49(s,1H),8.67(d,J=6.0Hz,1H),8.44(d,J=13.6Hz,1H),8.17(d,J=5.6Hz,2H),7.74–7.68(m,2H),7.67(s,1H),7.56(d,J=8.6Hz,1H),7.50(s,1H),7.47(d,J=6.8Hz,1H),7.24(t,J=8.0Hz,2H),6.88(d,J=5.4Hz,1H),6.79(d,J=6.0Hz,1H),5.01(s,1H),4.07–4.08–4.00(m,5H),1.95–1.87(m,1H),1.23(d,J=5.2Hz,3H).
实施例3:5-((3-氟-4-((7-(2-羟基-2-甲基丙基)-6-甲氧基喹啉-4-基)氧代)苯基)氨基)-3-苯基-1,6-萘啶-4(1H)-酮的制备
制备方法同实施例1,不同之处在于将5-氯-3-(4-氟苯基)-1,6-萘啶-4(1H)-酮改为5-氯-3-苯基-1,6-萘啶-4(1H)-酮。1H NMR(600MHz,DMSO-d6):δ13.45(s,1H),12.93(s,1H),8.89(d,J=6.6Hz,1H),8.25–8.34(m,2H),8.11(d,J=4.8Hz,1H),7.79(s,1H),7.67–7.72(m,3H),7.65(d,J=8.4Hz,1H),7.62(d,J=8.4Hz,1H),7.48(d,J=7.8Hz,2H),7.38(t,J=7.2Hz,1H),7.17(d,J=4.2Hz,1H), 7.01(d,J=5.7Hz,1H),4.07(s,3H),3.99(s,2H),1.29(s,6H).
实施例4:5-((3-氟-4-((7-(2-羟基-2-甲基丙基)喹啉-4-基)氧代)苯基)氨基)-3-(4-氟苯基)-1,6-萘啶-4(1H)-酮的制备
Figure PCTCN2016105012-appb-000017
步骤1):4-氯喹啉-7-醇的合成:
在氮气保护,-70℃的条件下,向4-氯-7-甲氧基喹啉(0.95g,4.9mmol)的二氯甲烷(45mL)溶液中缓慢滴加BBr3(4.9g,19.6mmol)的二氯甲烷(15mL)稀释溶液。然后将反应体系升温到室温,向其中加入苄基三乙基氯化铵(TEBA,0.19g 0.83mmol)的二氯甲烷(5mL)稀释溶液,室温搅拌20h。TLC监测(石油醚:丙酮=20:5,Rf=0.4)。在冰浴冷却he搅拌下,向体系中缓慢加入冰水(25mL)淬灭BBr3。除去绝大部分二氯甲烷,用1N NaOH调节剩余溶液的PH=7,有大量白色固体析出,抽滤,真空(45℃)干燥24小时得化合物。收率:90%。1H NMR(600MHz,DMSO-d6):δ10.54(s,1H),8.69(d,J=4.8Hz,1H),8.06(d,J=9.6Hz,1H),7.51(d,J=4.8Hz,1H),7.30–7.36(m,2H).
步骤2):1-(4-氯喹啉7-基)氧代)-2-甲基丙-2醇的合成:
将4-氯喹啉-7-醇(100mg,0.56mmol)溶于THF/H2O混合溶剂(8mL,THF/H2O=1:1,V/V)中,依次向其中加入NaOH(66.6mg,1.67mmol)和甲基环氧丙烷(400mg,5.56mmol)。在45℃下,搅拌24小时,用乙酸乙酯稀释,然后用1N NaOH(10mL×4)洗母液,再用饱和食盐水洗,有机相用无水硫酸钠干燥。经柱层析纯化(TLC,石油醚:丙酮=20:5,Rf=0.45),得到白色固体。收率:63%。1H NMR(600MHz,DMSO-d6):δ7.88(d,J=4.8Hz,1H),7.22(d,J=9.0Hz,1H),6.70(d,J=4.8Hz,1H),6.52–6.57(m,2H),3.88(s,1H),3.06(s,2H),0.41(s,6H).
步骤3):1-((4-(2-氟-4-硝基苯基)喹啉7-基)氧代)-2-甲基丙-2醇的合成:
将1-(4-氯喹啉7-基)氧代)-2-甲基丙-2醇(0.456g,1.8mmol)和2-氟-4-硝基苯酚(0.568g,3.6mmol)悬浮在重蒸甲苯(20mL)中,然后向反应体系中加入N,N-二异丙基乙胺(0.583g,4.5mmol)。在油浴温度120℃条件下,反应30小时。用乙酸乙酯稀释反应体系,然后用1N NaOH洗有机相,饱和食盐水洗,无水硫酸钠干燥。经过柱层析纯化(TLC,石油醚:丙酮=20:8,Rf=0.3),得灰色固体。收率:90%。1H NMR(600MHz,DMSO-d6):δ8.72(d,J=4.8Hz,1H),8.48(d,J=10.2Hz,1H),8.20(d,J=9.0Hz,1H),8.16(d,J=9.0Hz,1H),7.64(t,J=8.4Hz,1H),7.44(s,1H),7.36(d,J=9.0Hz,1H),6.80(d,J=4.8Hz,1H),4.76(s,1H),3.93(s,2H),1.27(s,6H).
步骤4):1-((4-(4-氨基-2-氟苯基)喹啉7-基)氧代)-2-甲基丙-2醇的合成:
将1-((4-(2-氟-4-硝基苯基)喹啉7-基)氧代)-2-甲基丙-2醇(54mg,0.145mmol)溶于甲醇(10mL)。在搅拌状态下,向其中加入Pd/C(11mg),氢气氛下室温反应5h。除去溶经柱层析纯化(TLC,石油醚:丙酮=20:10,Rf=0.3)得灰色固体。收率:90%。1H NMR(400MHz,DMSO-d6):δ8.55(d,J=4.2Hz,1H),8.18(d,J=9.0Hz,1H),7.33(s,1H),7.28(d,J=9.0Hz,1H),7.06(t,J=12.0Hz,1H),6.53(d,J=12.8Hz,1H),6.44(d,J=8.4Hz,1H),6.38(d,J=4.8Hz,1H),5.51(s,2H),4.72(s,1H),3.89(s,2H),1.26(s,6H).
步骤5):5-((3-氟-4-((7-(2-羟基-2-甲基丙基)喹啉-4-基)氧代)苯基)氨基)-3-(4-氟苯基)-1,6-萘啶-4(1H)-酮的制备
将1-((4-(4-氨基-2-氟苯基)喹啉7-基)氧代)-2-甲基丙-2醇(40mg,0.117mmol)和5-氯-3-(4-氟苯基)-1,6-萘啶-4(1H)-酮(33mg,0.12mmol)加入到25mL的梨形瓶中,然后加入异丙醇(8mL),在磁力搅拌下加入浓盐酸(1滴),升温至90℃反应1h。过滤不溶物,滤饼溶解于10mL二氯甲烷和甲醇的混合溶剂中,加入等当量的三乙胺,室温搅拌0.5h,析出的固体抽滤后干燥得白色固体。收率:88%。1H NMR(600MHz,DMSO-d6):δ13.41(s,2H),9.03(d,J=6.0Hz,1H),8.54(d,J=9.0Hz,1H),8.30(d,J=3.6Hz,1H),8.21(s,1H),8.06(d,J=4.8Hz,1H),7.78(s,1H),7.77–7.72(m,2H),7.72–7.63(m,2H),7.59(d,J=8.4Hz,1H),7.33(s,1H),7.29(t,J=8.4Hz,2H),7.10(d,J=5.4Hz,1H),4.00(s,2H),1.29(s,6H).
实施例5:5-((3-氟-4-((7-(2-羟基-2-甲基丙基)喹啉-4-基)氧代)苯基)氨基)-3-(2-氟苯基)-1,6-萘啶-4(1H)-酮的制备
制备方法同实施例4,不同之处在于将5-氯-3-(4-氟苯基)-1,6-萘啶-4(1H)-酮改为5-氯-3-(2-氟苯基)-1,6-萘啶-4(1H)-酮。1H NMR(600MHz,DMSO-d6):δ13.22(s,1H),13.08(s,1H),8.98(d,J=3.0Hz,1H),8.53(d,J=8.4Hz,1H),8.43(d,J=11.4Hz,1H),8.15–8.24(m,2H),7.79(s,1H),7.64(d,J=7.8Hz,1H),7.55–7.75(m,2H),7.48–7.54(m 1H),7.43–7.47(m,1H),7.26–7.33(m,2H),7.07–7.12(m,1H),7.01–7.06(m,1H),4.00(s,2H),1.29(s,6H).
实施例6:5-((3-氟-4-((7-(2-羟基-2-甲基丙基)喹啉-4-基)氧代)苯基)氨基)-3-(3-氟苯基)-1,6-萘啶-4(1H)-酮的制备
制备方法同实施例4,不同之处在于将5-氯-3-(4-氟苯基)-1,6-萘啶-4(1H)-酮改为5-氯-3-(3-氟苯基)-1,6-萘啶-4(1H)-酮。1H NMR(600MHz,DMSO-d6):δ13.35(s,2H),9.03(d,J=6.6Hz,1H),8.54(d,J=9.0Hz,1H),8.37(d,J=5.4Hz,1H),8.25(d,J=12.6Hz,1H),8.09(d,J=6.0Hz,1H),7.77(s,1H),7.63–7.72(m,2H),7.57–7.66(m,2H),7.57(d,J=7.8Hz,1H),7.49(dd,J=14.4,7.2Hz,1H),7.25–7.33(m,1H),7.21(t,J=8.4Hz,1H),7.10(d,J=6.3Hz,1H),4.00(s,2H),1.29(s,6H).
实施例7:5-((3-氟-4-((7-(2-羟基-2-甲基丙基)喹啉-4-基)氧代)苯基)氨基)-3-苯基-1,6-萘啶-4(1H)-酮的制备
制备方法同实施例4,不同之处在于将5-氯-3-(4-氟苯基)-1,6-萘啶-4(1H)-酮改为5-氯-3-苯基-1,6-萘啶-4(1H)-酮。1H NMR(600MHz,DMSO-d6):δ13.45(s,1H),13.27(s,1H),9.03(d,J=6.6Hz,1H),8.54(d,J=9.3Hz,1H),8.28(d,J=5.4Hz,1H),8.24(d,J=9.3Hz,1H),8.07(d,J=6.0Hz,1H), 7.77(s,1H),7.68–7.73(m,3H),7.66(d,J=9.6Hz,1H),7.60(d,J=8.4Hz,1H),7.46(t,J=7.5,7.5Hz,2H),7.38(t,J=7.2,1H),7.24–7.33(m,1H),7.08(d,J=6.0Hz,1H),4.00(s,2H),1.29(s,6H).
实施例8:5-((3-氟-4-((6-(2-羟基-2-甲基丙基)-7-甲氧基喹啉-4-基)氧代)苯基)氨基)-3-(4-氟苯基)-1,6-萘啶-4(1H)-酮的制备
制备方法同实施例1,不同之处在于将2-甲氧基-5-硝基苯酚改为2-甲氧基-4-硝基苯酚。1H NMR(600MHz,DMSO-d6):δ13.38(s,1H),12.90(s,1H),8.87(d,J=6.0Hz,1H),8.31(d,J=9.0Hz,1H),8.26(d,J=3.6Hz,1H),8.12(d,J=4.2Hz,1H),7.81(s,1H),7.71–7.76(m,1H),7.68(s,1H),7.58–7.64(m,1H),7.49(d,J=7.2Hz 1H),7.28(t,J=8.4Hz,2H),7.10–7.16(m,2H),7.00(d,J=4.8Hz,1H),4.07(s,3H),4.02(s,2H),1.27(s,6H).
实施例9:5-((3-氟-4-((6-(2-羟基-2-甲基丙基)-7-甲氧基喹啉-4-基)氧代)苯基)氨基)-3-苯基-1,6-萘啶-4(1H)-酮的制备
制备方法同实施例1,不同之处在于将2-甲氧基-5-硝基苯酚改为2-甲氧基-4-硝基苯酚,将5-氯-3-(4-氟苯基)-1,6-萘啶-4(1H)-酮改为5-氯-3-苯基-1,6-萘啶-4(1H)-酮。1H NMR(400MHz,DMSO-d6):δ13.39(s,1H),12.85(s,1H),8.85(d,J=6.8Hz,1H),8.03–8.23(m,2H),8.08(d,J=6.4Hz,1H),7.78(s,1H),7.67–7.59(m,4H),7.46–7.40(m,2H),7.35(t,J=7.2Hz,1H),7.15–7.09(m,2H),6.97(d,J=6.4Hz,1H),4.06(s,3H),4.01(s,2H),1.27(s,6H).
实施例10:5-((3-氟-4-((7-(2-羟基丙基)-6-甲氧基喹啉-4-基)氧代)苯基)氨基)-3-(4-氟苯基)-1-甲基-1,6-萘啶-4(1H)-酮的制备
将5-((3-氟-4-((7-(2-羟基丙基)-6-甲氧基喹啉-4-基)氧代)苯基)氨基)-3-(4-氟苯基)-1,6-萘啶-4(1H)-酮(100mg,0.13mmol)和无水碳酸钾(95mg,0.69mmol)加入到10mL的梨形瓶中,加入DMF(3mL),磁力搅拌30分钟,然后加入碘甲烷(71mg,0.52mmol),继续搅拌4h。将体系倾入水(12mL)中,搅拌,过滤不溶物,滤饼用水洗涤,真空干燥,得到淡黄色固体。收率:73.5%。1H NMR(400MHz,DMSO-d6):δ13.57(s,1H),8.97(d,J=6.8Hz,1H),8.50(d,J=9.2Hz,1H),8.40(s,1H),8.35(d,J=12.8Hz,1H),8.23(d,J=6.4Hz,1H),7.83–7.69(m,3H),7.68–7.54(m,3H),7.38–7.23(m,2H),7.14(d,J=5.2Hz,1H),7.04(d,J=6.4Hz,1H),3.98(s,2H),3.89(s,3H),1.28(s,6H).
实施例11:1-乙基-5-((3-氟-4-((7-(2-羟基丙基)-6-甲氧基喹啉-4-基)氧代)苯基)氨基)-3-(4-氟苯基)-1,6-萘啶-4(1H)-酮的制备
制备方法同实施例10,不同之处在于将碘甲烷改为碘乙烷。1H NMR(400MHz,DMSO-d6):δ13.47(s,1H),8.58(d,J=5.2Hz,1H),8.37(dd,J=13.6,2.0Hz,1H),8.34(s,1H),8.27(d,J=6.4Hz,1H),8.22(d,J=8.8Hz,1H),7.74(d,J=5.6Hz,1H),7.72(d,J=5.6Hz,1H),7.50(d,J=8.0Hz,1H),7.41–7.36(m,2H),7.32(dd,J=9.0,2.4Hz,1H),7.26(t,J=8.8Hz,2H),7.04(d,J=6.4Hz,1H),6.48(d,J=5.2Hz,1H),4.74(s,1H),4.32(q,J=7.2Hz,2H),3.91(s,2H),1.39(t,J=7.2Hz,3H),1.27(s,6H).
实施例12:5-((3-氟-4-((7-(2-羟基丙基)-6-甲氧基喹啉-4-基)氧代)苯基)氨基)-3-(4-氟苯基)-1-丙基-1,6-萘啶-4(1H)-酮的制备
制备方法同实施例10,不同之处在于将碘甲烷改为碘丙烷。1H NMR(400MHz,DMSO-d6):δ13.45(s,1H),8.58(d,J=3.6Hz,1H),8.30–8.40(m,2H),8.18–8.28(m,2H),7.66–7.76(m,2H),7.49 (d,J=7.0Hz,1H),7.44–7.34(m,2H),7.20–7.33(m,3H),7.04(d,J=4.8Hz,1H),6.48(d,J=5.2Hz,1H),4.71(s,1H),4.17–4.30(m,2H),3.90(s,2H),1.74–1.88(m,2H),1.27(s,6H),0.90–1.02(m,3H).
实施例13:5-((3-氟-4-((7-(2-羟基丙基)-6-甲氧基喹啉-4-基)氧代)苯基)氨基)-3-(4-氟苯基)-1-异丙基-1,6-萘啶-4(1H)-酮的制备
制备方法同实施例10,不同之处在于将碘甲烷改为2-碘丙烷。1H NMR(400MHz,DMSO-d6):δ13.56(s,1H),8.58(d,J=5.2Hz,1H),8.36(dd,J=13.6,2.0Hz,1H),8.26(d,J=6.4Hz,1H),8.19–8.24(m,2H),7.75–7.66(m,2H),7.49(d,J=8.0Hz,1H),7.43–7.34(m,2H),7.31(dd,J=9.2,2.2Hz,1H),7.23–7.28(m,2H),7.18(d,J=6.6Hz,1H),6.47(d,J=5.0Hz,1H),5.06–4.95(m,1H),4.72(s,1H),3.90(s,2H),1.53(d,J=6.4Hz,6H),1.27(s,6H).
实施例14:1-环丙基甲基-5-((3-氟-4-((7-(2-羟基丙基)-6-甲氧基喹啉-4-基)氧代)苯基)氨基)-3-(4-氟苯基)-1,6-萘啶-4(1H)-酮的制备
制备方法同实施例10,不同之处在于将碘甲烷改为溴代环丙烷。1H NMR(400MHz,DMSO-d6):δ13.46(s,1H),8.57(d,J=5.2Hz,1H),8.40–8.32(m,2H),8.26(d,J=6.0Hz,1H),8.21(d,J=9.0Hz,1H),7.71(d,J=5.6Hz,1H),7.69(d,J=5.8Hz,1H),7.50(d,J=8.2Hz,1H),7.40(d,J=9.0Hz,1H),7.36(d,J=2.0Hz,1H),7.30(dd,J=9.2,2.0Hz,1H),7.20–7.28(m,2H),7.13(d,J=6.2Hz,1H),6.47(d,J=5.0Hz,1H),4.71(s,1H),4.17(d,J=6.8Hz,1H),3.90(s,2H),1.41–1.32(m,1H),1.26(s,6H),0.53–0.60(m,,2H),0.48–0.52(m,2H).
实施例15:5-((3-氟-4-((7-(2-羟基丙基)-6-甲氧基喹啉-4-基)氧代)苯基)氨基)-3-(4-氟苯基)-1-(环氧乙烷-2-基甲基)-1,6-萘啶-4(1H)-酮的制备
制备方法同实施例10,不同之处在于将碘甲烷改为环氧溴丙烷。1H NMR(400MHz,DMSO-d6):δ13.14(s,1H),8.55(d,J=5.2Hz,1H),8.34–8.16(m,3H),7.56(s,1H),7.54(d,J=5.4Hz,1H),7.52(d,J=5.4Hz,1H),7.46–7.40(m,1H),7.38(d,J=2.4Hz,1H),7.28–7.21(m,2H),7.20–7.14(m,1H),7.13–7.09(m,1H),6.63(d,J=6.2Hz,1H),6.44(d,J=5.2Hz,1H),4.51(dd,J=15.6,5.6Hz,1H),4.03(dd,J=15.6,5.6Hz,1H),3.96(s,2H),3.42–3.35(m,1H),2.97–2.92(m,1H),2.62–2.56(m,1H),1.40(s,6H).
实施例16:5-((3-氟-4-((7-(2-羟基丙基)-6-甲氧基喹啉-4-基)氧代)苯基)氨基)-3-(4-氟苯基)-1-(2-甲氧基乙基)-1,6-萘啶-4(1H)-酮的制备
制备方法同实施例10,不同之处在于将碘甲烷改为2-溴乙基甲基醚。1H NMR(400MHz,DMSO-d6):δ13.44(s,1H),8.59(d,J=5.2Hz,1H),8.37(dd,J=13.6,2.0Hz,1H),8.26(d,J=6.0Hz,1H),8.24–8.19(m,2H),7.71(d,J=5.6Hz,1H),7.69(d,J=6.0Hz,1H),7.51(dd,J=9.2,1.2Hz,1H),7.41(d,J=9.0Hz,1H),7.37(d,J=2.4Hz,1H),7.31(dd,J=9.2,2.4Hz,1H),7.23–7.29(m,2H),7.08(d,J=6.4Hz,1H),6.48(d,J=5.2Hz,1H),4.73(s,1H),4.49(t,J=4.2,4.0Hz,2H),3.91(s,2H),3.70(t,J=4.2,4.0Hz,2H),3.26(s,3H),1.27(s,6H).
实施例17:5-((3-氟-4-((7-(2-羟基丙基)-6-甲氧基喹啉-4-基)氧代)苯基)氨基)-3-(4-氟苯基)-1-(2-羟基乙基)-1,6-萘啶-4(1H)-酮的制备
制备方法同实施例10,不同之处在于将碘甲烷改为溴乙醇。1H NMR(400MHz,DMSO-d6):δ13.65(s,1H),8.99(d,J=6.4Hz,1H),8.50(d,J=9.2Hz,1H),8.37–8.26(m,2H),8.16(d,J=6.2Hz, 1H),7.73(d,J=6.2Hz,1H),7.71(d,J=5.6Hz,1H),7.67(s,1H),7.62(d,J=8.8Hz,2H),7.59(d,J=4.8Hz,1H),7.32–7.23(m,2H),7.19(d,J=6.4Hz,1H),7.14(d,J=6.0Hz,1H),5.15(br),4.51–4.37(m,2H),3.98(s,2H),3.82–3.71(m,2H),1.27(s,6H).
实施例18:1-苄基-5-((3-氟-4-((7-(2-羟基丙基)-6-甲氧基喹啉-4-基)氧代)苯基)氨基)-3-(4-氟苯基)-1,6-萘啶-4(1H)-酮的制备
制备方法同实施例10,不同之处在于将碘甲烷改为溴化苄。1H NMR(400MHz,DMSO-d6):δ13.37(s,1H),8.61–8.53(m,2H),8.32(d,J=13.6Hz,1H),8.21(d,J=9.0Hz,1H),8.16(d,J=6.0Hz,1H),7.80–7.71(m,2H),7.49(d,J=8.2Hz,1H),7.42–7.32(m,4H),7.32–7.22(m,6H),6.86(d,J=5.4Hz,1H),6.46(d,J=5.0Hz,1H),5.56(s,2H),4.70(s,1H),3.90(s,2H),1.26(s,6H).
实施例19:对比化合物C的合成
Figure PCTCN2016105012-appb-000018
为了进一步揭示喹啉环7-位支链烷基和直链烷基对激酶抑制活性的影响,我们还合成了对比化合物C。制备方法同实施例4,不同之处在于将甲基环氧丙烷改为2-碘乙醇。1H NMR(400MHz,DMSO-d6):δ13.33(s,1H),12.91(s,1H),8.83(d,J=6.4Hz,1H),8.23–8.29(m,2H),8.07(d,J=6.4Hz,1H),7.69–7.75(m,2H),7.56–7.63(m,1H),7.46(d,J=8.0Hz,2H),7.25(t,J=7.2Hz,2H),7.15(d,J=6.4Hz,1H),7.09(d,J=8.0Hz,2H),6.98(d,J=9.0Hz,1H),4.26(t,J=4.8Hz,2H),3.86(t,J=4.8Hz,2H).
实施例20:对比化合物D的合成
Figure PCTCN2016105012-appb-000019
为了进一步揭示喹啉环7-位羟基支链烷基取代基中羟基对激酶抑制活性的影响,我们还合成了对比化合物D。
步骤1):3-氟-4-((7-(2-甲氧基-2-甲基丙氧基)喹啉-4-基)氧代)苯胺的合成
将1-((4-(4-氨基-2-氟苯基)喹啉7-基)氧代)-2-甲基丙-2醇(324mg,0.84mmol)和无水碳酸钾(170mg,1.23mmol)加入到25mL的梨形瓶中,加入DMF(10mL),磁力搅拌,加入碘甲烷(310mg,2.18mmol),继续搅拌2.5小时。将体系倒入水(40mL)中,搅拌,过滤不溶物,滤饼用水洗涤,真空干燥,得到淡黄色固体。
步骤2):对比化合物D的合成
将3-氟-4-((7-(2-甲氧基-2-甲基丙氧基)喹啉-4-基)氧代)苯胺(42mg,0.117mmol)和5-氯-3-(4- 氟苯基)-1,6-萘啶-4(1H)-酮(33mg,0.12mmol)加入到25mL的梨形瓶中,然后加入异丙醇(8mL),在磁力搅拌下加入浓盐酸(1滴),升温至90℃反应1小时。过滤不溶物,滤饼溶解于10mL二氯甲烷和甲醇的混合溶剂中,加入等当量的三乙胺,室温搅拌0.5h,析出的固体抽滤后干燥得到白色固体。1H NMR(600MHz,DMSO-d6):δ13.34(s,1H),12.86(s,1H),8.84(d,J=6.4Hz,1H),8.24–8.29(m,2H),8.08(d,J=6.4Hz,1H),7.67–7.76(m,3H),7.56–7.62(m,2H),7.46(d,J=8.0Hz,1H),7.26(t,J=8.8Hz,2H),7.13(d,J=6.8Hz,1H),7.09(d,J=8.0Hz,1H),6.97(d,J=6.0Hz,1H),4.10(s,2H),3.27(s,3H),1.30(s,6H).
实施例21:体外生化水平抑制蛋白激酶(PK)活性实验
材料与方法:c-Met、VEGFR-2、Axl和RET等激酶,来源于Invitrogen;HTRF KinEASE;TK kit(Cisbio公司);384孔板(Greiner公司);ATP(sigma公司),MgCl2(sigma)公司;PHERAstar FS多功能酶标仪(BMG公司);低速离心机(StaiteXiangyi公司);恒温箱(Binder公司)。选取的对照化合物为WO2013097753专利中公开的典型化合物A和B,结构如下:
Figure PCTCN2016105012-appb-000020
化合物溶解及保存:视溶解性用二甲基亚砜(DMSO)将受试化合物配置成0.5-10mmol/L的母液,分装后-20℃保存;
化合物工作液的配制:测试前将分装的化合物从冰箱取出,用纯DMSO稀释到50×所需浓度;然后用去离子水将化合物稀释至4×所需浓度;
1.33×酶缓冲液(Enzymatic buffer)的配制:将5×酶缓冲液(来源于HTRF试剂盒)用去离子水稀释到1.33×,并且加入1.33×终浓度的相应成分:1.33mmol/L二硫苏糖醇(DTT)和1.33mmol/L MgCl2
激酶工作液的配制:用1.33×酶缓冲液将Met稀释到2×所需终浓度0.2ng/μL;
底物工作液的配制:用1.33×酶缓冲液将生物素标记的底物(来源于HTRF试剂盒)和ATP(10mM)稀释为4×所需终浓度的混合液;
检测工作液的配制:用HTRF测试缓冲液将16.67μmol/L的抗生蛋白链菌-XL665(Streptavidin-XL665)稀释到4×所需终浓度,然后与等体积的抗体-铕隐酸盐(Antibody-Cryptate)混合(均来源于HTRF试剂盒)。
酶反应步骤:向低体积384微孔板的每个孔中加入4μLμl的激酶工作液,同时加入4μL的1.33×酶缓冲液作为阴性对照(Negative);向孔加入2μl的化合物工作液,同时加入2μL的8%DMSO 水溶液作为零化合物浓度对照(即阳性对照,Positive);于25℃(或30℃)孵育5-10min;向孔中加入2μL底物工作液启动酶反应,于25℃(或30℃)振荡反应15-60min。
HTRF试剂检测步骤:向孔加入8μL的检测工作液终止反应;25℃反应1h;
HTRF信号的读取:采用PHERAstar FS读数检测信号,仪器相应设置如下:
Optic module 
Figure PCTCN2016105012-appb-000021
积分延迟(Integration delay,lag time)50μs
积分时间(Integration time)400μs
闪光次数(Number of flashes)200
对于每孔读出的原始数据,比值=665nm/620nm;
抑制率的计算:
Figure PCTCN2016105012-appb-000022
IC50值的计算:以化合物浓度的对数为横坐标,抑制率为纵坐标,在GraphPad Prism 5中,拟合非线性曲线:log(inhibitor)vs.response--Variable slope,求出酶活抑制率为50%时的待测化合物浓度即IC50
实验结果:c-Met、VEGFR-2、Axl和RET激酶活性半数抑制浓度(IC50,nM)
本发明提供结构如式Ⅰ所示化合物对c-Met和VEGFR-2的半数抑制浓度(IC50)见表1:
表1:化合物的c-Met和VEGFR-2激酶抑制活性
Figure PCTCN2016105012-appb-000023
Figure PCTCN2016105012-appb-000024
进一步的测试还表明,实施例4和实施例7对Axl激酶表现出良好的抑制活性,IC50分别为15.7和14.9nM;对RET也表现出良好的抑制活性,IC50分别为62.0和53.0nM
实施例21:大鼠体内药物代谢研究
对比将实施例4和对比化合物C以聚乙二醇400水溶液(70%)形式大鼠给药。对于静脉注射给药,大鼠给予1mg/kg的剂量。对于口服给药,大鼠给予5mg/kg的剂量。实施例4和对比化合物C口服组给药后15、30、45min、1、2、4、6、8、10、24h各采集血样约0.3mL至肝素化的Eppendorf管中,实施例4和对比化合物C静脉组给药后5、15、30min、1、2、4、6、8、10和24h各采集血样约0.3mL至肝素化Eppendorf管中,于冰上暂存至离心。全血经8000rpm离心5min后收集血浆,转移血浆至96孔板中,于-20℃保存至LC-MS/MS检测。
采用软件WinNonlin软件的非室模型计算大鼠给药后的药代动力学参数。
达峰浓度Cmax:采用实测值;
药时曲线下面积AUC0-t值:采用梯形法计算;AUC0-∞=AUC0-t+Ct/ke,Ct为最后一个可测得时间点的血药浓度,ke为消除速率常数;
消除半衰期t1/2=0.693/ke;
绝度生物利用度F=Doseiv*AUC0-t,ig/Doseig*AUC0-t,iv×100%。
表2列出了静注或口服给药后,实施例4与对比化合物C在大鼠体内的药代动力学参数。结果表明,实施例4具有良好的药代动力学性质,包括理想的清除率(CL)、半衰期(t1/2)和暴露量(AUC0-t)。在相同剂量下,实施例4的口服给药暴露量是对比化合物C的22倍,并且生物利用度也明显高于对比化合物C。
表2:实施例4与对比化合物C在大鼠体内的药代动力学数据
Figure PCTCN2016105012-appb-000025
实施例22:异体移植瘤模型
本发明化合物的药效是通过移植肿瘤的标准鼠类模型来进行评价的。人肿瘤细胞(U87MG胶质瘤细胞,MKN45胃腺癌细胞,Caki-1肾癌细胞,HUH 7肝癌细胞,NCI-H441肺腺癌上皮细胞, MDA-MB-231乳腺癌细胞,SMMC-7721肝癌细胞,ATCC)培养、收集后,于后腹侧皮下接种于6-7周龄的雌性裸小鼠体内(BALB/cA nu/nu,上海SLAC动物实验室)。当肿瘤体积达到150mm3时,动物随机地分为溶剂对照组(70%PEG-400的水溶液)和化合物组(每组6只动物)。后续采用化合物对动物进行灌胃给药(3-10mpk/dose,溶解在70%PEG-400的水溶液中),从肿瘤细胞接种后的0到22天中的任何地方开始,并且通常在试验中每天进行一次。
肿瘤生长抑制(TGI)分析
肿瘤的演化生长是通过肿瘤体积与时间的关系来进行评价的。皮下肿瘤的长轴(L)和短轴(W)通过测径器每周测定两次,肿瘤的体积(TV)通过公式(L×W2)/2)进行计算。TGI由溶剂组小鼠肿瘤体积的中值和药物组小鼠肿瘤体积中值的差值来进行计算,以溶剂对照组肿瘤体积中值的百分比来表示,通过下述公式进行计算:
Figure PCTCN2016105012-appb-000026
肿瘤部分消退(PR):当末次给药结束时肿瘤体积小于给药开始时肿瘤体积,被视为肿瘤发生消退。
原始统计分析是通过重复方差测定分析(RMANOVA)来完成的。接下来通过Scheffe psot hoc试验方法进行多重比较。单独溶剂(70%PEG-400等等)为阴性对照。
图1显示了实施例4在U87MG胶质母细胞瘤模型中的抑制肿瘤生长效果。将实施例4以每天(QD)3和10mg/kg的剂量口服给药(p.o.),持续22天。所有的剂量均具有统计学上的意义,可剂量依赖性的抑制裸鼠皮下U87MG肿瘤的生长。给药的最后一天(第22天),与溶剂组的平均肿瘤体积比较,3和10mg/kg剂量下分别抑制平均肿瘤体积的87.0%和105.9%(TGI)。
表3:U87MG模型肿瘤抑制效果
Figure PCTCN2016105012-appb-000027
以上详细描述了本发明的优选实施方式,但是,本发明并不限于上述实施方式中的具体细节,在本发明的技术构思范围内,可以对本发明的技术方案进行多种简单变型,这些简单变型均属于本发明的保护范围。
另外需要说明的是,在上述具体实施方式中所描述的各个具体技术特征,在不矛盾的情况下,可以通过任何合适的方式进行组合,为了避免不必要的重复,本发明对各种可能的组合方式不再另行说明。
此外,本发明的各种不同的实施方式之间也可以进行任意组合,只要其不违背本发明的思想,其同样应当视为本发明所公开的内容。

Claims (10)

  1. 一种萘啶化合物,或其立体异构体,几何异构体,互变异构体,氮氧化物,水合物,溶剂化物,代谢产物,药学上可接受的盐或它的前药,该化合物的结构如式(I)所示:
    Figure PCTCN2016105012-appb-100001
    其中:
    R1和R2选自H、C1-C3烷基;
    R3选自H、C1-C3烷氧基;
    R4选自H、C1-C6烷基、3至8元全碳单环环烷基、3至8元杂脂环基,其中C1-C6烷基、3至8元全碳单环环烷基、3至8元杂脂环基可选进一步被一个或多个选自C1-C6烷基、C1-C6烷氧基、3至8元杂脂环基、C6-C10芳基、C5-C10杂芳基或羟基的取代基所取代;
    R5选自H、F、Cl、Br、I、CN、C1-3烷基或C1-3卤代烷基。
  2. 根据权利要求1所述的化合物,其中,
    Figure PCTCN2016105012-appb-100002
    R3分别在喹啉环6-位、7-位或者分别在喹啉环7-位、6-位。
  3. 根据权利要求2所述的化合物,其中,R1和R2选自H,C1-C3烷基,且R1和R2不同时为氢。
  4. 根据权利要求1所述的化合物,其中,R4选自H、C1-C6烷基、3至8元全碳单环环烷基、3至8元杂脂环基C1-C6亚烷基、C1-C6烷氧基C1-C6亚烷基或C6-C10芳基C1-C6亚烷基。
  5. 根据权利要求4所述的化合物,其中,R4选自氢、甲基、乙基、丙基、异丙基、环丙基、
    Figure PCTCN2016105012-appb-100003
    2-甲氧基乙基、2-羟基乙基或苄基。
  6. 根据权利要求1所述的化合物,其中,所述萘啶化合物具有以下其中之一的结构:
    Figure PCTCN2016105012-appb-100004
    Figure PCTCN2016105012-appb-100005
    或其立体异构体,几何异构体,互变异构体,氮氧化物,水合物,溶剂化物,代谢产物,药学上可接受的盐或它的前药。
  7. 一种药物组合物,其包含药学上可接受的载体、赋形剂或稀释剂,以及作为活性成分的权利要求1至6中任一项所定义的化合物。
  8. 权利要求1-6任一项所述的化合物或权利要求7所述的组合物在制备治疗经蛋白激酶中介的疾病的药物中的应用。
  9. 根据权利要求8所述的应用,其特征在于,所述蛋白激酶中介的疾病选自与c-Met、VEGFR-2、Axl或RET相关的疾病。
  10. 根据权利要求9所述的应用,其特征在于,所述疾病选自结肠直肠癌、膀胱癌、乳腺癌、肝癌、肺癌、胰腺癌、胃肠癌、白血病、卵巢癌、头和颈癌、前列腺癌、肾癌、鼻咽癌、成胶质细胞瘤、鳞状细胞癌、星形细胞癌、卡波济氏肉瘤、黑素瘤、神经胶质瘤、泌尿生殖道癌,骨髓增殖性疾患;动脉粥样硬化或肺纤维化。
PCT/CN2016/105012 2016-10-09 2016-11-08 萘啶化合物和药物组合物及它们的应用 WO2018064852A1 (zh)

Priority Applications (5)

Application Number Priority Date Filing Date Title
JP2019540476A JP6862563B2 (ja) 2016-10-09 2016-11-08 ナフチリジン化合物、薬物組成物およびそれらの使用
PL16918185T PL3421038T3 (pl) 2016-10-09 2016-11-08 Związek naftyrydyny, kompozycja farmaceutyczna i ich zastosowanie
AU2016425880A AU2016425880B2 (en) 2016-10-09 2016-11-08 Naphthyridine compound, pharmaceutical composition and use thereof
ES16918185T ES2784525T3 (es) 2016-10-09 2016-11-08 Compuestos de naftiridina, combinaciones médicas y uso de las mismas
EP16918185.6A EP3421038B1 (en) 2016-10-09 2016-11-08 Naphthyridine compound, pharmaceutical composition and use thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201610880102.6A CN106749231B (zh) 2016-10-09 2016-10-09 萘啶化合物和药物组合物及它们的应用
CN201610880102.6 2016-10-09

Publications (1)

Publication Number Publication Date
WO2018064852A1 true WO2018064852A1 (zh) 2018-04-12

Family

ID=58972188

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2016/105012 WO2018064852A1 (zh) 2016-10-09 2016-11-08 萘啶化合物和药物组合物及它们的应用

Country Status (9)

Country Link
US (1) US9938274B1 (zh)
EP (1) EP3421038B1 (zh)
JP (2) JP6862563B2 (zh)
CN (1) CN106749231B (zh)
AU (1) AU2016425880B2 (zh)
ES (1) ES2784525T3 (zh)
PL (1) PL3421038T3 (zh)
PT (1) PT3421038T (zh)
WO (1) WO2018064852A1 (zh)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11273160B2 (en) 2018-04-03 2022-03-15 Blueprint Medicines Corporation RET inhibitor for use in treating cancer having a RET alteration
US11279688B2 (en) 2015-11-02 2022-03-22 Blueprint Medicines Corporation Inhibitors of RET

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019143977A1 (en) 2018-01-18 2019-07-25 Array Biopharma Inc. Substituted pyrrolo[2,3-d]pyrimidines compounds as ret kinase inhibitors
CA3087972C (en) 2018-01-18 2023-01-10 Array Biopharma Inc. Substituted pyrazolyl[4,3-c]pyridinecompounds as ret kinase inhibitors
CN110862397A (zh) * 2018-08-27 2020-03-06 北京赛特明强医药科技有限公司 二噁烷并喹唑啉与二噁烷并喹啉类化合物及其制备方法与应用
CN111961045A (zh) * 2020-08-07 2020-11-20 南京纳丁菲医药科技有限公司 1,6-萘啶-4(1h)-酮化合物的合成方法
CN113520999B (zh) * 2021-07-30 2022-08-09 石家庄四药有限公司 含萘啶类化合物的口服液及其制备方法
CN113633623A (zh) * 2021-08-25 2021-11-12 石家庄四药有限公司 一种含萘啶类化合物的胶囊及其制备方法

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103183674A (zh) * 2011-12-31 2013-07-03 江苏先声药物研究有限公司 一类稠杂环衍生物及其应用

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6809097B1 (en) 1996-09-25 2004-10-26 Zeneca Limited Quinoline derivatives inhibiting the effect of growth factors such as VEGF
US9155792B2 (en) * 2006-02-13 2015-10-13 Trustees Of Boston University RecA inhibitors with antibiotic activity, compositions and methods of use
MX352844B (es) * 2011-02-28 2017-12-11 Calitor Sciences Llc Compuestos de quinolina sustituidos y metodos de uso.

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103183674A (zh) * 2011-12-31 2013-07-03 江苏先声药物研究有限公司 一类稠杂环衍生物及其应用

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11279688B2 (en) 2015-11-02 2022-03-22 Blueprint Medicines Corporation Inhibitors of RET
US11273160B2 (en) 2018-04-03 2022-03-15 Blueprint Medicines Corporation RET inhibitor for use in treating cancer having a RET alteration
US11872192B2 (en) 2018-04-03 2024-01-16 Blueprint Medicines Corporation RET inhibitor for use in treating cancer having a RET alteration
US11963958B2 (en) 2018-04-03 2024-04-23 Rigel Pharmaceuticals, Inc. RET inhibitor for use in treating cancer having a RET alteration

Also Published As

Publication number Publication date
EP3421038A1 (en) 2019-01-02
JP2021054871A (ja) 2021-04-08
JP6862563B2 (ja) 2021-04-21
CN106749231B (zh) 2019-03-01
AU2016425880B2 (en) 2020-09-17
PT3421038T (pt) 2020-04-22
EP3421038A4 (en) 2019-03-20
US20180099962A1 (en) 2018-04-12
JP2019534323A (ja) 2019-11-28
CN106749231A (zh) 2017-05-31
JP7085032B2 (ja) 2022-06-15
PL3421038T3 (pl) 2020-06-29
AU2016425880A1 (en) 2019-05-30
ES2784525T3 (es) 2020-09-28
EP3421038B1 (en) 2020-01-15
US9938274B1 (en) 2018-04-10

Similar Documents

Publication Publication Date Title
WO2018064852A1 (zh) 萘啶化合物和药物组合物及它们的应用
JP5950978B2 (ja) カリウムイオンチャネルインヒビターとしてのキナゾリン
KR102007056B1 (ko) 과증식성 질환 치료시 Bub1 키나제 저해제로 사용하기 위한 치환된 벤질인다졸
US9890168B2 (en) 2,4-disubstituted 7H-pyrrolo[2,3-d]pyrimidine derivative, preparation method and medicinal use thereof
TWI628179B (zh) [1,2,4]三唑並[4,3-a]吡啶類衍生物,其製備方法或其在醫藥上的應用
WO2015127872A1 (zh) 2,4-二取代苯-1,5-二胺衍生物及其应用以及由其制备的药物组合物和药用组合物
WO2022206723A1 (zh) 杂环类衍生物、其制备方法及其医药上的用途
WO2013185353A1 (en) Compound as wnt signaling inhibitor, composition, and use thereof
TW201418254A (zh) 作為c-Met酪氨酸激酶抑制劑的新型稠合吡啶衍生物
CA3050239A1 (en) Pyrimidine compound and pharmaceutical use thereof
WO2022037631A1 (zh) 杂环类衍生物及其制备方法和用途
JP2023511302A (ja) 新規なピラゾール誘導体
WO2018157730A1 (zh) 脲取代的芳环连二噁烷并喹唑啉与连二噁烷并喹啉类化合物及其制备方法与应用
CN108264511B (zh) 杂环类衍生物及其制备方法和其在医药上的用途
WO2019154133A1 (zh) 二噁烷并喹啉类化合物及其制备方法与应用
WO2022089219A1 (zh) 芳基酰胺化合物、包含其的药物组合物及其制备方法和用途
WO2019170086A1 (zh) 一种酰基取代的噁嗪并喹唑啉类化合物、制备方法及其应用
WO2023020512A1 (zh) 取代的吡啶类似物、其制备方法及作为ahr调节剂的用途
WO2022184000A1 (zh) 咪唑类化合物、其药物组合物及其用途
WO2021105317A1 (en) Benzylamide derivatives as inhibitors of transforming growth factor-beta receptor i/alk5
EA045526B1 (ru) Производные бензиламида в качестве ингибиторов рецептора i/alk5 трансформирующего фактора роста-бета
TW201914999A (zh) 1,2-二氫-1,6-萘啶類衍生物、其製備方法、其藥物組合物及其在醫藥上的用途

Legal Events

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

Ref document number: 2016918185

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2016918185

Country of ref document: EP

Effective date: 20180925

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

Ref document number: 16918185

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2019540476

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2016425880

Country of ref document: AU

Date of ref document: 20161108

Kind code of ref document: A