WO2024022095A1 - Axl&c-met dual inhibitor, and preparation method and use - Google Patents

Axl&c-met dual inhibitor, and preparation method and use Download PDF

Info

Publication number
WO2024022095A1
WO2024022095A1 PCT/CN2023/106523 CN2023106523W WO2024022095A1 WO 2024022095 A1 WO2024022095 A1 WO 2024022095A1 CN 2023106523 W CN2023106523 W CN 2023106523W WO 2024022095 A1 WO2024022095 A1 WO 2024022095A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
cancer
add
pharmaceutically acceptable
prodrug
Prior art date
Application number
PCT/CN2023/106523
Other languages
French (fr)
Chinese (zh)
Inventor
谢军
姜春阳
李惠
裴欣宇
舒海英
Original Assignee
上海博悦生物科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 上海博悦生物科技有限公司 filed Critical 上海博悦生物科技有限公司
Publication of WO2024022095A1 publication Critical patent/WO2024022095A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/444Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a six-membered ring with nitrogen as a ring heteroatom, e.g. amrinone
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • 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
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings

Definitions

  • the invention relates to the field of medicinal chemistry, relates to a substituted pyridine amide compound, and specifically relates to a type of Axl&c-Met dual inhibitor and its preparation method and use.
  • c-MET is a transmembrane receptor tyrosine kinase encoded by the MET gene and has high affinity to hepatocyte growth factor (HGF) structurally. Dysregulation of HGF/c-MET signaling leads to the activation of downstream pathways, including the RAS/MAPK, PI3K/AkT, and Rac/Rho pathways, which are involved in cell proliferation, survival, and metastasis. High-level MET gene amplification, protein overexpression or gene mutation are the main mechanisms leading to abnormal activation of the HGF/c-MET pathway. Increasing evidence shows that c-Met receptor tyrosine kinase plays a role in tumor development and metastasis progression. role.
  • HGF hepatocyte growth factor
  • dysregulation of MET tyrosine kinases is associated with resistance to targeted therapies in cancer patients and often occurs in patients with non-small cell lung cancer (NSCLC) who are resistant to EGFR inhibitors.
  • Drugs targeting MET signaling have the potential to improve treatment of patients with MET disorders.
  • c-Met/HGF namely Crizotinib, Cabozantinib, Capmatinib and Tepotinib. The indications are non-small cell lung cancer. host.
  • Receptor tyrosine kinases are transmembrane proteins that connect the extracellular and intracellular environments. As a mediator of signal transduction, it plays an important role in normal cellular processes, including differentiation, adhesion, migration, apoptosis, and metabolism.
  • Axl also known as Ufo, Ark or Tyro7
  • Tyro3 and Mer constitute the TAM subfamily of receptor tyrosine kinases.
  • Overexpression of Axl kinase was first discovered in chronic myelogenous leukemia and chronic myeloproliferation. Subsequently, Paccez et al.
  • Axl kinase overexpression Abnormal expression of Axl activates and antagonizes tumor cell apoptosis, promotes tumor cell invasion and metastasis, promotes tumor angiogenesis, and promotes the occurrence and development of tumors in many aspects.
  • Axl kinase overexpression Abnormal expression of Axl activates and antagonizes tumor cell apoptosis, promotes tumor cell invasion and metastasis, promotes tumor angiogenesis, and promotes the occurrence and development of tumors in many aspects.
  • Dimer produced by overexpression of Axl and binding to EGFR is an important reason for the acquired resistance of tumor cells to EGFR inhibitors; preclinical research on the combination of Axl inhibitors can Effectively overcome resistance to EGFR inhibitors.
  • Axl abnormal activation of Axl overexpression is also closely related to resistance to other targeted inhibitors and chemotherapy drugs, suggesting that Axl may have a wide range of application space for combination drugs.
  • Axl is highly expressed in macrophages and dendritic cells in the tumor microenvironment, and can interact with tumor cells and Other stromal cells interact and synergistically promote tumor progression. Therefore, in recent years, the development of Axl-targeted inhibitors has become the forefront and hot spot of anti-tumor drug research. Small molecule inhibitors developed for it have shown effects in tumor treatment.
  • the present invention provides a new class of Axl&c-Met dual inhibitors, which have excellent effects in treating various tumors. Moreover, this series of inhibitors has strong ability to penetrate the blood-brain barrier and is expected to be used to treat the brain. Tumor.
  • the purpose of the present invention is to develop a class of Axl&c-Met dual inhibitors, preparation methods and uses thereof.
  • This new type of Axl&c-Met dual inhibitor has a completely new structure different from existing compounds and shows excellent anti-tumor activity.
  • the invention provides a series of compounds, including compounds represented by formula (I) or pharmaceutically acceptable salts thereof, prodrugs thereof, hydrates or solvates thereof:
  • R 1 is hydrogen, halogen, substituted or unsubstituted alkyl, alkoxy or haloalkoxy, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl;
  • R 2 is halogen, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted alkoxy, or hydroxyalkyl;
  • n 1 or 2.
  • the alkyl group is a C 1 -C 6 alkyl group
  • the cycloalkyl group is a C 3 -C 4 cycloalkyl group
  • the alkoxy group is a C 1 -C 6 Alkoxy group
  • the hydroxyalkyl group is a C 1 -C 6 hydroxyalkyl group
  • the substituent is fluorine or chlorine.
  • This compound has dual inhibitory effects on Axl&c-Met and is a new dual inhibitor of Axl&c-Met.
  • R 1 is selected from hydrogen, methyl, methoxy, trifluoromethyl, trifluoromethoxy, fluorine or chlorine;
  • R 2 is selected from fluorine, chlorine, methyl.
  • R1 is selected from hydrogen, methyl, fluorine or chlorine
  • R2 is selected from fluorine, chlorine, methyl
  • any one of the following compounds is selected:
  • the invention also provides a method for preparing an Axl&c-Met dual inhibitor compound, which method includes the following steps: compound and compounds
  • the reaction produces a compound represented by formula (I), and the reaction is preferably carried out in the presence of an organic solvent, HATU and triethylamine.
  • the organic solvent includes N,N-dimethylacetamide, N-methylpyrrolidone, dichloromethane, chloroform, tetrahydrofuran, methyl acetate, ethyl acetate, isopropyl acetate, 1,2-dichloro Ethane, acetonitrile, etc.
  • R 1 , R 2 and n are as defined in the present invention.
  • the present invention also provides the use of the compound represented by formula (I) or a pharmaceutically acceptable salt thereof, a prodrug thereof, a hydrate or a solvate thereof for the preparation of a medicament for treating related diseases caused by Axl kinase and/or c-Met kinase. use in.
  • the related diseases caused by the Axl kinase and/or c-Met kinase include brain tumors, gastric cancer, bladder cancer, breast cancer, colorectal cancer, head and neck cancer, kidney cancer, liver cancer, lung cancer, ovarian cancer, pancreatic cancer, and pancreatic cancer.
  • the brain tumors include brain gliomas, meningiomas, brain metastases, etc.
  • the present invention also provides the use of the compound represented by formula (I) or its pharmaceutically acceptable salt, its prodrug, its hydrate or solvate in the preparation of Axl&c-Met inhibitors.
  • the invention also provides the use of a series of Axl&c-Met inhibitors in the preparation of drugs for treating tumors.
  • the tumor Includes: brain tumors, stomach cancer, bladder cancer, breast cancer, colorectal cancer, head and neck cancer, kidney cancer, liver cancer, lung cancer, ovarian cancer, pancreatic adenocarcinoma/gallbladder cancer, prostate cancer, thyroid cancer, osteosarcoma, rhabdomyosarcoma , MFH/fibrosarcoma, malignant glioma/astrocytoma, melanoma or mesothelioma, etc.
  • the tumor drug is a brain tumor drug, including brain glioma drugs, meningioma drugs, and brain metastasis drugs.
  • Figure 1 is a graph showing the changes in plasma concentration over time after intragastric administration of Compound 1 to mice (dose 10 mg/kg);
  • Figure 2 is a graph showing the changes in brain concentration over time after compound 1 was administered to mice by gavage (dose 10 mg/kg);
  • Figure 3 is a comparison chart of the effects of compounds on the body weight of nude mice
  • Figure 4 is a comparison chart of the effects of compounds on tumor volume in the human gastric cancer cell MKN45 subcutaneous transplant tumor model
  • Figure 5 is a comparison chart of the effects of compounds on the relative tumor volume of the human gastric cancer cell MKN45 subcutaneous transplant tumor model
  • Figure 6 is a comparison chart of the effects of compounds on subcutaneous xenografts of human gastric cancer cells MKN45.
  • intermediate B1A Add 15 g of intermediate B1A, 150 mL of ethanol, 10 g of 4-methoxybut-3-en-2-one, and 50 mL of sodium ethoxide ethanol solution into the 500 mL reaction bottle, and raise the temperature to reflux at 70-80°C and incubate the reaction for 6 hours. Cool to room temperature, add 150 mL each of 1 mol/L hydrochloric acid and methylene chloride, stir and separate the liquids. The dichloromethane phase was concentrated to dryness. The concentrated residue was beaten with 50 mL of ethyl acetate at 5°C, filtered, and dried to obtain 8.8 g of intermediate B1 with a yield of 53.9%.
  • intermediate B4A Add 4g of intermediate B4A, 40mL of ethanol, 2.48g of 4-methoxybut-3-en-2-one, and 12.5mL of sodium ethoxide ethanol solution into the 100mL reaction bottle, and heat to reflux and incubate for 7 hours. Cool to room temperature, add 60 mL each of 1 mol/L hydrochloric acid and methylene chloride, stir and separate the liquids. The dichloromethane phase was concentrated to dryness. The concentrated residue was beaten with 20 mL of ethyl acetate at 5°C, filtered, and dried to obtain 2.49 g of intermediate B4 with a yield of 57.1%.
  • the inhibitory effect of the compound of the present invention on MET and AXL kinase was detected by using fluorescence microfluidic mobility detection technology (Mobility-Shift Assay).
  • Kinase catalyzes the removal of a phosphate group from ATP to generate ADP, and transfers the phosphate group to the substrate peptide.
  • the substrate peptide is fluorescently labeled, and the charge of the product changes due to the addition of a phosphate group.
  • the substrate and phosphorylated products are separated due to different mobilities and are detected respectively, and their amounts are proportional to the fluorescence signal.
  • Use the Caliper instrument to measure the amounts of substrate and product, calculate the conversion rate of the product, and then calculate the inhibition rate.
  • the MET kinase reaction system is 25uL, which includes 5nM MET, concentration gradient small molecule inhibitor, 10mM MgCl2, 1M DTT, 26uM ATP (measured Km value), 3uM Peptide2 (5-FAM-EAIYAAPFAKKK-CONH2), 0.0015% Brij-35 and pH 7.5 50mM HEPES, 2mM DTT, 10mM MgCl2;
  • AXL kinase reaction system is 25uL, including 6nM AXL, concentration gradient small molecule inhibitor, 10mM MgCl2, 1M DTT, 81uM ATP (measured Km value ), 3uM Peptide22 (5-FAM-EEPLYWSFPAKKK-CONH2), 0.0015% Brij-35 and 50mM HEPES, pH 7.5, 2mM DTT, 10mM MgCl2.
  • Experimental tumor cell lines were cultured in RPMI-1640 and DMEM containing 10-20% Gibco serum at 37°C in a 5% CO2 incubator. According to the background data of the laboratory, 4000 tumor cells/well were seeded in a 96-well culture plate. The cells were in the logarithmic growth phase throughout the experiment.
  • the inhibition rate is calculated according to the following formula:
  • Inhibition rate (%) 1-OD administration/OD control ⁇ 100%. If the inhibition rate ⁇ 0, it is recorded as 0.
  • the tested compounds have varying degrees of inhibitory effects on U87-MG and MKN45, two human tumor cells derived from different tissues.
  • compound 1, compound 2, compound 5, compound 6, compound 9, compound 11, compound 12 and compound 15 have inhibitory effects on U87 -MG cells have obvious inhibitory effects, especially compound 11 is more sensitive to U87-MG cells, with an IC50 value of 2.71 ⁇ M (see Table 2 for details).
  • Compound 11 has a significant inhibitory effect on MKN45 cells, with an IC50 value of 8.71 ⁇ M, which is significantly better than the 19.37 ⁇ M of the control drug BMS777607.
  • Samples were taken from plasma and brain at 0.25h, 0.5h, 1h, 2h, 4h, 8h, and 24h, and then the amounts of chemical components in plasma and brain tissue were detected by LC-MS/MS to obtain a series of drugs.
  • Kinetic parameters, the results are shown in Table 3 and Figures 1 and 2 below.
  • compound 1 shows excellent oral pharmacokinetic properties and can pass through the blood-brain barrier with a permeability of 0.24.
  • the drug has high brain tissue exposure and can be developed to treat brain gliomas, meningiomas, and brain metastases. drugs for brain tumors such as tumors.
  • Example 18 Experimental therapeutic effect of compound 1 on subcutaneous transplantation of human gastric cancer cell MKN45 in nude mice.
  • mice 5 ⁇ 106 human gastric cancer MKN45 cells were injected into the left armpit of nude mice. Cut out the tumor pieces from MKN45 mice and place them in a glass dish filled with physiological saline. Peel off the surface blood vessels and remove the necrotic area. Cut the tumor pieces into 1-2 mm 3 and insert them into the nude mice with a trocar. After the tumor in the left armpit grew to an average volume of about 100 mm 3 , the animals were randomly divided into groups according to tumor volume and then administered. 32 mice were divided into 4 groups: G1 blank vehicle group (Control), G2BMS777607 30mg/kg group, G3 compound 1 15mg/kg group and G4 compound 1 30mg/kg group. There were 8 rats in each group, and each group was intragastrically administered the test substance of the corresponding concentration at a dosage volume of 10 mL/kg, once a day, with a 21-day dosing cycle.
  • G1 blank vehicle group Control
  • G2BMS777607 30mg
  • the tumor volumes of the G2BMS777607 30mg/kg group, G3 compound 1 15mg/kg group and G4 compound 1 30mg/kg group were 1600 ⁇ 135 and 900 ⁇ 112 respectively (P ⁇ 0.01 ) and 859 ⁇ 96mm3 (P ⁇ 0.01).
  • the tumor mass weight of the G2BMS777607 30mg/kg group, G3 compound 1 15mg/kg group and G4 compound 1 30mg/kg group were 1.1525 ⁇ 0.1271 and 0.6556 ⁇ 0.0907 respectively (P ⁇ 0.05) and 0.5886 ⁇ 0.0840g (P ⁇ 0.05), IR were 27.55%, 58.79% and 63.00% respectively.
  • the effect of compound 1 on subcutaneous xenografts of human gastric cancer cells MKN45 (see Figure 6 for details).
  • Example 19 uses human liver microsomes to evaluate the inhibitory effect of Compound 1 of the present invention on 7 CYP450 isoforms (CYP 1A2, 2B6, 2C8, 2C9, 2C19, 2D6 and 3A4).
  • the inhibitors are CYP1A2: a-Naphthoflavone; CYP2C9: Sulfaphenazole; CYP2C19: Omeprazole; CYP3A4: Ketoconazole; CYP2D6: quinidine; CYP2C8: Nicardipine; CYP2B6: Clopidogrel;
  • the matrix is CYP1A2: Phenacetin at 30 ⁇ M; CYP2C9: Diclofenac at 10 ⁇ M; CYP2C19: S-Mephenytoin at 35 ⁇ M; CYP3A4: Midazolam at 5 ⁇ M; CYP2D6: Bufuralol at 10 ⁇ M; CYP2C8: Paclitaxel at 10 ⁇ M; CYP2B6: Bupropion at 70 ⁇ M.
  • Detection system Human liver microsomes from Corning, incubation conditions CYP1A2, 2C9, 2D6, 2C8, 2B6: 10 minutes, 37°C; CYP2C19: 45 minutes, 37°C; CYP3A4: 5 minutes, 37°C. Sample size 2 copies (n 2 ), the bioanalytical method is LC-MS/MS.
  • the present invention has the following beneficial effects:
  • the Axl&c-Met dual inhibitor provided by the present invention has strong inhibitory activity against both AXL and c-Met. It can inhibit both GAS6/AXL and HGF/c-Met signaling pathways at the same time, and is expected to produce more significant effects. anti-tumor effect,
  • the inhibitory effect of the compound of the present invention on Axl and c-Met kinase is better than that of the positive control drug BMS777607.
  • the Axl&c-Met dual inhibitor of the present invention showed significant anti-tumor activity, was significantly better than the positive control drug BMS777607, and was well tolerated by animals.
  • the Axl&c-Met dual inhibitor of the present invention has excellent pharmacokinetic properties and is expected to be developed into an anti-tumor drug; in particular, this series of inhibitors has a strong ability to penetrate the blood-brain barrier and in brain tissue With good exposure, it can be used to develop drugs for brain tumors such as glioma, brain metastases, and meningiomas.
  • the present invention also provides a simple preparation method for a series of compounds with dual Axl&c-Met inhibitory effects.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Public Health (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Diabetes (AREA)
  • Cardiology (AREA)
  • Urology & Nephrology (AREA)
  • Rheumatology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Vascular Medicine (AREA)
  • Dermatology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Emergency Medicine (AREA)
  • Endocrinology (AREA)
  • Obesity (AREA)
  • Epidemiology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Hematology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Pain & Pain Management (AREA)
  • Ophthalmology & Optometry (AREA)
  • Oncology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Disclosed in the present invention are a class of Axl&c-Met dual inhibitors, and a preparation method therefor and the use thereof. The inhibitor is a compound as represented by formula (I) or a pharmaceutically acceptable salt, prodrug, hydrate or solvate thereof, wherein R1 is hydrogen, halogen, substituted or unsubstituted alkyl, alkoxy or haloalkoxy, substituted or unsubstituted cycloalkyl, or substituted or unsubstituted aryl; R2 is halogen, or substituted or unsubstituted alkyl; and the value of n is 1 or 2. The Axl&c-Met dual inhibitor of the present invention has excellent pharmacokinetic properties and is expected to be developed into an anti-tumor drug. In particular, the series of inhibitors have an effective blood-brain barrier penetration ability, have good exposure in brain tissues, and can be used for developing drugs for brain tumors such as brain glioma and brain metastatic tumors.

Description

Axl&c-Met双重抑制剂、制备方法以及用途Axl&c-Met dual inhibitor, preparation method and use 技术领域Technical field
本发明涉及医药化学领域,涉及一种取代吡啶酰胺类化合物,具体涉及一类Axl&c-Met双重抑制剂及其制备方法和用途。The invention relates to the field of medicinal chemistry, relates to a substituted pyridine amide compound, and specifically relates to a type of Axl&c-Met dual inhibitor and its preparation method and use.
背景技术Background technique
c-MET是一种由MET基因编码的跨膜受体酪氨酸激酶,在结构上与肝细胞生长因子(HGF)具有高亲和力。HGF/c-MET信号传导的失调导致下游通路的激活,包括RAS/MAPK、PI3K/AkT和Rac/Rho通路,它们参与细胞增殖、存活和转移。高水平MET基因扩增、蛋白过表达或基因突变是导致HGF/c-MET通路异常激活的主要机制,越来越多的证据表明c-Met受体酪氨酸激酶在肿瘤发展和转移进展中的作用。此外MET酪氨酸激酶的失调与癌症患者对靶向治疗的耐药性相关,并且经常发生在对EGFR抑制剂耐药的非小细胞肺癌(NSCLC)患者中。靶向MET信号传导的药物有望改善对MET失调患者群体的治疗。当前针对于c-Met/HGF靶点开发,已上市药物已有4个,分别为克唑替尼(Crizotinib)、卡博替尼(Cabozantinib)、Capmatinib和Tepotinib,适应症以非小细胞肺癌为主。c-MET is a transmembrane receptor tyrosine kinase encoded by the MET gene and has high affinity to hepatocyte growth factor (HGF) structurally. Dysregulation of HGF/c-MET signaling leads to the activation of downstream pathways, including the RAS/MAPK, PI3K/AkT, and Rac/Rho pathways, which are involved in cell proliferation, survival, and metastasis. High-level MET gene amplification, protein overexpression or gene mutation are the main mechanisms leading to abnormal activation of the HGF/c-MET pathway. Increasing evidence shows that c-Met receptor tyrosine kinase plays a role in tumor development and metastasis progression. role. In addition, dysregulation of MET tyrosine kinases is associated with resistance to targeted therapies in cancer patients and often occurs in patients with non-small cell lung cancer (NSCLC) who are resistant to EGFR inhibitors. Drugs targeting MET signaling have the potential to improve treatment of patients with MET disorders. Currently, there are four drugs currently on the market targeting c-Met/HGF, namely Crizotinib, Cabozantinib, Capmatinib and Tepotinib. The indications are non-small cell lung cancer. host.
受体酪氨酸激酶(RTKs)是连接细胞外和细胞内环境的跨膜蛋白。作为信号转导的调停者,其在正常的细胞过程中扮演了重要的角色,包括分化、粘附、迁移、凋亡、代谢。Axl(又名Ufo,Ark或Tyro7)是一种受体酪氨酸激酶,其与Tyro3及Mer共同组成受体酪氨酸激酶TAM亚家族。Axl激酶的过表达最初是在慢性粒细胞白血病和慢性骨髓增生症中发现的,随后,Paccez等人发现在乳腺癌、肺癌、前列腺癌、结肠癌、食管癌、肝癌等多种癌症中也存在Axl激酶过表达。Axl异常表达激活拮抗肿瘤细胞凋亡、促进肿瘤细胞侵袭、转移,促进肿瘤血管新生,多环节推动了肿瘤的发生发展。尤其值得关注的是,近年研究显示,Axl过表达并且同EGFR结合所产生的二聚体是肿瘤细胞对EGFR抑制剂产生获得性耐药性的重要原因;临床前研究Axl抑制剂的联合用药可以有效克服EGFR抑制剂耐药。此外,Axl过表达异常激活与其他靶向抑制剂以及化疗药物的耐药也密切相关,提示了Axl可能具有广泛的联合用药的应用空间。与其他激酶不同的是,Axl在肿瘤微环境的巨噬细胞、树突状细胞中高表达,可以通过与肿瘤细胞以及 其他基质细胞交互作用,协同促进肿瘤进展。因此,近年来,靶向Axl抑制剂的研发已成为抗肿瘤药物研究的前沿和热点。针对其开发的小分子抑制剂已经在肿瘤治疗中显示效应。Receptor tyrosine kinases (RTKs) are transmembrane proteins that connect the extracellular and intracellular environments. As a mediator of signal transduction, it plays an important role in normal cellular processes, including differentiation, adhesion, migration, apoptosis, and metabolism. Axl (also known as Ufo, Ark or Tyro7) is a receptor tyrosine kinase, which together with Tyro3 and Mer constitute the TAM subfamily of receptor tyrosine kinases. Overexpression of Axl kinase was first discovered in chronic myelogenous leukemia and chronic myeloproliferation. Subsequently, Paccez et al. found that it also exists in various cancers such as breast cancer, lung cancer, prostate cancer, colon cancer, esophageal cancer, and liver cancer. Axl kinase overexpression. Abnormal expression of Axl activates and antagonizes tumor cell apoptosis, promotes tumor cell invasion and metastasis, promotes tumor angiogenesis, and promotes the occurrence and development of tumors in many aspects. What is particularly noteworthy is that recent studies have shown that the dimer produced by overexpression of Axl and binding to EGFR is an important reason for the acquired resistance of tumor cells to EGFR inhibitors; preclinical research on the combination of Axl inhibitors can Effectively overcome resistance to EGFR inhibitors. In addition, abnormal activation of Axl overexpression is also closely related to resistance to other targeted inhibitors and chemotherapy drugs, suggesting that Axl may have a wide range of application space for combination drugs. Unlike other kinases, Axl is highly expressed in macrophages and dendritic cells in the tumor microenvironment, and can interact with tumor cells and Other stromal cells interact and synergistically promote tumor progression. Therefore, in recent years, the development of Axl-targeted inhibitors has become the forefront and hot spot of anti-tumor drug research. Small molecule inhibitors developed for it have shown effects in tumor treatment.
同时,研究显示Axl以及c-MET在脑胶质瘤等多种恶性肿瘤中均有过表达。脑胶质瘤等脑部肿瘤治疗的主要障碍之一是药物无法有效透过血脑屏障,急需发现有血脑屏障透过能力的小分子药物。At the same time, studies have shown that Axl and c-MET are overexpressed in a variety of malignant tumors such as glioma. One of the main obstacles in the treatment of brain tumors such as glioma is that drugs cannot effectively penetrate the blood-brain barrier. There is an urgent need to discover small molecule drugs with the ability to penetrate the blood-brain barrier.
本发明提供了一类新型Axl&c-Met双重抑制剂,在治疗各种肿瘤都体现了优异的效果,而且,该系列抑制剂有较强的透过血脑屏障的能力,有望用于治疗脑部肿瘤。The present invention provides a new class of Axl&c-Met dual inhibitors, which have excellent effects in treating various tumors. Moreover, this series of inhibitors has strong ability to penetrate the blood-brain barrier and is expected to be used to treat the brain. Tumor.
发明内容Contents of the invention
本发明的目的在于开发一类Axl&c-Met双重抑制剂、制备方法及其用途。该类新型Axl&c-Met双重抑制剂具有不同于现有化合物的全新结构,显示出优异的抗肿瘤活性。The purpose of the present invention is to develop a class of Axl&c-Met dual inhibitors, preparation methods and uses thereof. This new type of Axl&c-Met dual inhibitor has a completely new structure different from existing compounds and shows excellent anti-tumor activity.
本发明的目的是通过以下技术方案来实现的:The purpose of the present invention is achieved through the following technical solutions:
<第一方面><First aspect>
本发明提供了一系列化合物,包括式(I)所示化合物或其药学上可接受的盐、其前药、其水合物或溶剂合物:
The invention provides a series of compounds, including compounds represented by formula (I) or pharmaceutically acceptable salts thereof, prodrugs thereof, hydrates or solvates thereof:
其中,R1为氢、卤素、取代或未取代的烷基、烷氧基或者卤代烷氧基、取代或未取代的环烷基、取代或未取代的芳基;Wherein, R 1 is hydrogen, halogen, substituted or unsubstituted alkyl, alkoxy or haloalkoxy, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl;
R2为卤素、取代或未取代的烷基、取代或未取代的环烷基、取代或未取代的烷氧基、羟烷基;R 2 is halogen, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted alkoxy, or hydroxyalkyl;
n的数值为1或者2。The value of n is 1 or 2.
作为本发明的一个实施方案,所述烷基为C1-C6的烷基,所述环烷基为C3-C4的环烷基,所述烷氧基为C1-C6的烷氧基,所述羟烷基为C1-C6的羟烷基,取代基为氟或者氯。As an embodiment of the present invention, the alkyl group is a C 1 -C 6 alkyl group, the cycloalkyl group is a C 3 -C 4 cycloalkyl group, and the alkoxy group is a C 1 -C 6 Alkoxy group, the hydroxyalkyl group is a C 1 -C 6 hydroxyalkyl group, and the substituent is fluorine or chlorine.
该化合物具有Axl&c-Met双重抑制作用,是一种新型Axl&c-Met双重抑制剂。This compound has dual inhibitory effects on Axl&c-Met and is a new dual inhibitor of Axl&c-Met.
作为本发明的一个实施方案,R1选自氢、甲基、甲氧基、三氟甲基、三氟甲氧基、氟或氯;R2选自氟、氯、甲基。 As an embodiment of the present invention, R 1 is selected from hydrogen, methyl, methoxy, trifluoromethyl, trifluoromethoxy, fluorine or chlorine; R 2 is selected from fluorine, chlorine, methyl.
作为本发明的一个实施方案,R1选自氢、甲基、氟或氯,而且R2选自氟、氯、甲基。As an embodiment of the invention, R1 is selected from hydrogen, methyl, fluorine or chlorine, and R2 is selected from fluorine, chlorine, methyl.
作为本发明的一个实施方案,选自以下化合物中的任意一个:
As an embodiment of the present invention, any one of the following compounds is selected:
若化学命名和图示的化学结构有差异,则图示的化学结构优选于给出的化学命名。If there is a discrepancy between the chemical nomenclature and the illustrated chemical structure, the illustrated chemical structure is preferred over the given chemical nomenclature.
<第二方面><Second aspect>
本发明还提供了Axl&c-Met双重抑制剂化合物的制备方法,所述方法包括如下步骤: 化合物和化合物反应生成如式(I)所示化合物,优选有机溶剂、HATU和三乙胺存在的条件下进行反应。所述有机溶剂包括N,N-二甲基乙酰胺,N-甲基吡咯烷酮,二氯甲烷,三氯甲烷,四氢呋喃,乙酸甲酯,乙酸乙酯,乙酸异丙酯,1,2-二氯乙烷,乙腈等。The invention also provides a method for preparing an Axl&c-Met dual inhibitor compound, which method includes the following steps: compound and compounds The reaction produces a compound represented by formula (I), and the reaction is preferably carried out in the presence of an organic solvent, HATU and triethylamine. The organic solvent includes N,N-dimethylacetamide, N-methylpyrrolidone, dichloromethane, chloroform, tetrahydrofuran, methyl acetate, ethyl acetate, isopropyl acetate, 1,2-dichloro Ethane, acetonitrile, etc.
反应式如下:
The reaction formula is as follows:
其中,R1、R2和n如本发明所定义。Wherein, R 1 , R 2 and n are as defined in the present invention.
化合物的制备包括如下步骤:compound The preparation includes the following steps:
A1、化合物在二氯甲烷和三乙胺存在的条件下与氯甲酰乙酸乙酯反应,生成中间体化合物 A1. Compound React with ethyl chloroformyl acetate in the presence of dichloromethane and triethylamine to generate an intermediate compound
A2、中间体化合物和4-甲氧基丁-3-烯-2-酮在乙醇和乙醇钠乙醇溶液存在条件下反应,即得。 A2. Intermediate compounds It can be obtained by reacting with 4-methoxybut-3-en-2-one in the presence of ethanol and sodium ethoxide ethanol solution.
化合物的制备包括如下步骤:compound The preparation includes the following steps:
B1、化合物与化合物在DMAc、叔丁醇钾存在的条件下,在氮气保护下于80-90℃反应,生成中间体化合物 B1, compound with compounds In the presence of DMAc and potassium tert-butoxide, react at 80-90°C under nitrogen protection to generate an intermediate compound
B2、中间体化合物与1-甲基吡唑-4-硼酸频哪醇酯在二氧六环、碳酸钾和Pd(PPh3)4存在条件下,90~95℃保温反应,即得。B2, intermediate compounds It can be obtained by reacting with 1-methylpyrazole-4-boronic acid pinacol ester at 90-95℃ in the presence of dioxane, potassium carbonate and Pd(PPh 3 ) 4 .
<第三方面><Third aspect>
本发明还提供了式(I)所示化合物或其药学上可接受的盐、其前药、其水合物或溶剂合物在制备治疗Axl激酶和/或c-Met激酶引起的相关疾病的药物中的用途。The present invention also provides the use of the compound represented by formula (I) or a pharmaceutically acceptable salt thereof, a prodrug thereof, a hydrate or a solvate thereof for the preparation of a medicament for treating related diseases caused by Axl kinase and/or c-Met kinase. use in.
其中,所述Axl激酶和/或c-Met激酶引起的相关疾病包括脑部肿瘤、胃癌、膀肮癌、乳腺癌、结肠直肠癌、头颈癌、肾癌、肝癌、肺癌、卵巢癌、膜腺癌/胆囊癌、前列腺癌、甲状腺癌、骨肉瘤、横纹肌肉瘤、MFH/纤维肉瘤、恶性胶质瘤/星形细胞瘤、黑色素瘤或间皮瘤、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肾病等。Wherein, the related diseases caused by the Axl kinase and/or c-Met kinase include brain tumors, gastric cancer, bladder cancer, breast cancer, colorectal cancer, head and neck cancer, kidney cancer, liver cancer, lung cancer, ovarian cancer, pancreatic cancer, and pancreatic cancer. Cancer/gallbladder cancer, prostate cancer, thyroid cancer, osteosarcoma, rhabdomyosarcoma, MFH/fibrosarcoma, glioblastoma/astrocytoma, melanoma or mesothelioma, psoriasis, cirrhosis, diabetes, angiogenesis, regeneration Stenosis, eye diseases, rheumatoid arthritis and other inflammatory diseases, immune diseases, cardiovascular diseases such as arteriosclerosis and kidney disease, etc.
其中,所述脑部肿瘤包括脑胶质瘤、脑膜瘤、脑转移瘤等。Wherein, the brain tumors include brain gliomas, meningiomas, brain metastases, etc.
<第四方面><The fourth aspect>
本发明还提供了式(I)所示化合物或其药学上可接受的盐、其前药、其水合物或溶剂合物在制备Axl&c-Met抑制剂中的应用。The present invention also provides the use of the compound represented by formula (I) or its pharmaceutically acceptable salt, its prodrug, its hydrate or solvate in the preparation of Axl&c-Met inhibitors.
本发明还提供了一系列Axl&c-Met抑制剂在制备治疗肿瘤药物中的用途。所述肿瘤 包括:脑部肿瘤、胃癌、膀肮癌、乳腺癌、结肠直肠癌、头颈癌、肾癌、肝癌、肺癌、卵巢癌、膜腺癌/胆囊癌、前列腺癌、甲状腺癌、骨肉瘤、横纹肌肉瘤、MFH/纤维肉瘤、恶性胶质瘤/星形细胞瘤、黑色素瘤或间皮瘤等。The invention also provides the use of a series of Axl&c-Met inhibitors in the preparation of drugs for treating tumors. The tumor Includes: brain tumors, stomach cancer, bladder cancer, breast cancer, colorectal cancer, head and neck cancer, kidney cancer, liver cancer, lung cancer, ovarian cancer, pancreatic adenocarcinoma/gallbladder cancer, prostate cancer, thyroid cancer, osteosarcoma, rhabdomyosarcoma , MFH/fibrosarcoma, malignant glioma/astrocytoma, melanoma or mesothelioma, etc.
作为本发明的一个实施方案,所述肿瘤药物为脑部肿瘤药物,包括脑胶质瘤药物脑膜瘤药物、脑转移瘤药物。As an embodiment of the present invention, the tumor drug is a brain tumor drug, including brain glioma drugs, meningioma drugs, and brain metastasis drugs.
附图说明Description of drawings
通过阅读参照以下附图对非限制性实施例所作的详细描述,本发明的其它特征、目的和优点将会变得更明显:Other features, objects and advantages of the present invention will become more apparent by reading the detailed description of the non-limiting embodiments with reference to the following drawings:
图1为化合物1给小鼠灌胃给药后血浆中浓度随时间变化图(剂量10mg/kg);Figure 1 is a graph showing the changes in plasma concentration over time after intragastric administration of Compound 1 to mice (dose 10 mg/kg);
图2为化合物1给小鼠灌胃给药后脑部浓度随时间变化图(剂量10mg/kg);Figure 2 is a graph showing the changes in brain concentration over time after compound 1 was administered to mice by gavage (dose 10 mg/kg);
图3为化合物对裸小鼠体重的影响对比图;Figure 3 is a comparison chart of the effects of compounds on the body weight of nude mice;
图4为化合物对人胃癌细胞MKN45皮下移植瘤模型瘤体积的影响对比图;Figure 4 is a comparison chart of the effects of compounds on tumor volume in the human gastric cancer cell MKN45 subcutaneous transplant tumor model;
图5为化合物对人胃癌细胞MKN45皮下移植瘤模型相对肿瘤体积的影响对比图;Figure 5 is a comparison chart of the effects of compounds on the relative tumor volume of the human gastric cancer cell MKN45 subcutaneous transplant tumor model;
图6为化合物对人胃癌细胞MKN45皮下移植瘤的作用对比图。Figure 6 is a comparison chart of the effects of compounds on subcutaneous xenografts of human gastric cancer cells MKN45.
具体实施方式Detailed ways
下面结合实施例对本发明进行详细说明。以下实施例将有助于本领域的技术人员进一步理解本发明,但不以任何形式限制本发明。应当指出的是,对本领域的普通技术人员来说,在不脱离本发明构思的前提下,还可以做出若干调整和改进。这些都属于本发明的保护范围。所有提及到的出版物通过引用的方式以其全文并入。The present invention will be described in detail below with reference to examples. The following examples will help those skilled in the art to further understand the present invention, but do not limit the present invention in any form. It should be noted that, for those of ordinary skill in the art, several adjustments and improvements can be made without departing from the concept of the present invention. These all belong to the protection scope of the present invention. All publications mentioned are incorporated by reference in their entirety.
实施例1、化合物1合成Example 1, synthesis of compound 1
1.1中间体B1合成
1.1 Synthesis of intermediate B1
向1000mL反应瓶中加入4-氟苯胺20g,二氯甲烷200mL,三乙胺23.6g,搅拌降温至0℃。将氯甲酰乙酸乙酯35.2g溶于100mL二氯甲烷,控温0~5℃滴加入反应瓶中。滴完后升至20℃反应2小时,加入200mL水,搅拌分液。二氯甲烷相用200mL饱和食盐水洗,浓缩干。残余物用石油醚90mL和乙酸乙酯30mL打浆,过滤,干燥,得到中间体B1A 30.0g,收率74.1%。 Add 20g of 4-fluoroaniline, 200mL of methylene chloride, and 23.6g of triethylamine into the 1000mL reaction bottle, stir and cool to 0°C. Dissolve 35.2g of ethyl chloroformyl acetate in 100 mL of methylene chloride, and add it dropwise into the reaction bottle at a temperature of 0 to 5°C. After dropping, raise to 20°C and react for 2 hours, add 200 mL of water, stir and separate. Wash the dichloromethane phase with 200 mL of saturated brine and concentrate to dryness. The residue was slurried with 90 mL of petroleum ether and 30 mL of ethyl acetate, filtered, and dried to obtain 30.0 g of intermediate B1A with a yield of 74.1%.
MS:m/z=226.35[M+H]+ MS: m/z=226.35[M+H] +
1H NMR(400MHz,Chloroform-d)δ9.29(s,1H),7.54(m,2H),7.04(m,2H),4.28(q,J=7.1Hz,2H),3.49(s,2H),1.35(t,J=7.1Hz,3H). 1 H NMR (400MHz, Chloroform-d) δ9.29 (s, 1H), 7.54 (m, 2H), 7.04 (m, 2H), 4.28 (q, J = 7.1Hz, 2H), 3.49 (s, 2H ),1.35(t,J=7.1Hz,3H).
向500mL反应瓶中加入中间体B1A 15g,乙醇150mL,4-甲氧基丁-3-烯-2-酮10g,乙醇钠乙醇溶液50mL,升温至回流70-80℃保温反应6小时。降至室温,加入1mol/L盐酸和二氯甲烷各150mL,搅拌,分液。二氯甲烷相浓缩干。浓缩残余物用50mL乙酸乙酯5℃打浆,过滤,干燥,得到中间体B1 8.8g,收率53.9%。Add 15 g of intermediate B1A, 150 mL of ethanol, 10 g of 4-methoxybut-3-en-2-one, and 50 mL of sodium ethoxide ethanol solution into the 500 mL reaction bottle, and raise the temperature to reflux at 70-80°C and incubate the reaction for 6 hours. Cool to room temperature, add 150 mL each of 1 mol/L hydrochloric acid and methylene chloride, stir and separate the liquids. The dichloromethane phase was concentrated to dryness. The concentrated residue was beaten with 50 mL of ethyl acetate at 5°C, filtered, and dried to obtain 8.8 g of intermediate B1 with a yield of 53.9%.
MS:m/z=248.33[M+H]+ MS: m/z=248.33[M+H] +
1H NMR(400MHz,Chloroform-d)δ13.87(s,1H),8.53(d,J=7.5Hz,1H),7.36–7.30(m,2H),7.28–7.23(m,2H),6.57(d,J=7.5Hz,1H),2.18(s,3H). 1 H NMR (400MHz, Chloroform-d) δ13.87 (s, 1H), 8.53 (d, J = 7.5Hz, 1H), 7.36–7.30 (m, 2H), 7.28–7.23 (m, 2H), 6.57 (d,J=7.5Hz,1H),2.18(s,3H).
1.2中间体A1合成
1.2 Synthesis of intermediate A1
向500mL反应瓶中加入4-氨基-2-氟苯酚3.07g,DMAc 50mL,叔丁醇钾2.71g,氮气保护下搅拌0.5小时。加入2,3-二氯-4-碘吡啶5.5g,升温至85℃反应4小时。降至室温,反应液减压浓缩干。向浓缩残余物中加入乙酸乙酯150mL,搅拌,过滤。乙酸乙酯相减压浓缩干,得到中间体A1B。Add 3.07g of 4-amino-2-fluorophenol, 50mL of DMAc, and 2.71g of potassium tert-butoxide into the 500mL reaction bottle, and stir for 0.5 hours under nitrogen protection. Add 5.5g of 2,3-dichloro-4-iodopyridine, raise the temperature to 85°C and react for 4 hours. After cooling to room temperature, the reaction solution was concentrated to dryness under reduced pressure. 150 mL of ethyl acetate was added to the concentrated residue, stirred, and filtered. The ethyl acetate phase was concentrated to dryness under reduced pressure to obtain intermediate A1B.
MS:m/z=273.21[M+H]+ MS: m/z=273.21[M+H] +
1H NMR(400MHz,Chloroform-d)δ8.09(d,J=5.6Hz,1H),6.99(t,J=8.6Hz,1H),6.59–6.52(m,2H),6.49(ddd,J=8.7,2.7,1.3Hz,1H),3.86(s,2H). 1 H NMR(400MHz,Chloroform-d)δ8.09(d,J=5.6Hz,1H),6.99(t,J=8.6Hz,1H),6.59–6.52(m,2H),6.49(ddd,J =8.7,2.7,1.3Hz,1H),3.86(s,2H).
向250mL反应瓶中加入中间体A1B,二氧六环100mL,1-甲基吡唑-4-硼酸频哪醇酯5.25g,碳酸钾8.37g,水20mL,氮气置换,加入Pd(PPh3)4 1.5g,氮气置换,升温至90~95℃保温反应16小时。降温至室温,加入水和乙酸乙酯各200mL,搅拌分液。水相再用乙酸乙酯100mL萃取两次。合并乙酸乙酯相,减压浓缩干,通过硅胶柱层析分离(二氯甲烷:乙醇=100:1,v/v),得到中间体A1 4.36g,收率67.6%。Add intermediate A1B, 100 mL of dioxane, 5.25 g of 1-methylpyrazole-4-boronic acid pinacol ester, 8.37 g of potassium carbonate, and 20 mL of water into a 250 mL reaction bottle, replace with nitrogen, and add Pd (PPh 3 ) 4 1.5g, replace with nitrogen, raise the temperature to 90~95℃ and keep the reaction for 16 hours. Cool to room temperature, add 200 mL each of water and ethyl acetate, stir and separate the liquids. The aqueous phase was extracted twice with 100 mL of ethyl acetate. The ethyl acetate phases were combined, concentrated to dryness under reduced pressure, and separated by silica gel column chromatography (dichloromethane: ethanol = 100:1, v/v) to obtain 4.36 g of intermediate A1 with a yield of 67.6%.
MS:m/z=319.37[M+H]+ MS: m/z=319.37[M+H] +
1H NMR(400MHz,Chloroform-d)δ8.29(s,1H),8.27(d,J=5.5Hz,1H),8.21(s,1H),7.01(t,J=8.7Hz,1H),6.55(dd,J=11.9,2.7Hz,1H),6.49(ddd,J=8.7,2.8,1.3Hz,1H),6.46(dd,J=5.5,1.3Hz,1H),4.01(s,3H),3.83(s,2H). 1 H NMR (400MHz, Chloroform-d) δ8.29 (s, 1H), 8.27 (d, J = 5.5Hz, 1H), 8.21 (s, 1H), 7.01 (t, J = 8.7Hz, 1H), 6.55(dd,J=11.9,2.7Hz,1H),6.49(ddd,J=8.7,2.8,1.3Hz,1H),6.46(dd,J=5.5,1.3Hz,1H),4.01(s,3H) ,3.83(s,2H).
1.3化合物1合成
1.3 Synthesis of Compound 1
向500mL反应瓶中加入中间体A1 9g,中间体B1 6.98g,DMF 200mL,HATU 12.88g,三乙胺5.70g,搅拌过夜反应。滴加400mL水,过滤,滤饼干燥得化合物1粗品10.53g,收率68.0%。取500mg样品TLC纯化(展开剂为二氯甲烷:乙醇15:1),得产物289mg,纯度98.18%。Add 9g of intermediate A1, 6.98g of intermediate B1, 200mL of DMF, 12.88g of HATU, and 5.70g of triethylamine to the 500mL reaction bottle, and stir overnight for reaction. 400 mL of water was added dropwise, filtered, and the filter cake was dried to obtain 10.53 g of crude compound 1, with a yield of 68.0%. Take 500 mg of the sample and perform TLC purification (the developing solvent is methylene chloride: ethanol 15:1) to obtain 289 mg of product with a purity of 98.18%.
MS:m/z=548.45[M+H]+ MS: m/z=548.45[M+H] +
1H NMR(400MHz,Chloroform-d)δ11.94(s,1H),8.66(d,J=7.5Hz,1H),8.28(d,J=5.5Hz,2H),8.21(s,1H),7.97(dd,J=12.4,2.5Hz,1H),7.38–7.31(m,3H),7.28–7.24(m,2H),7.15(t,J=8.7Hz,1H),6.54(dd,J=7.6,0.9Hz,1H),6.47(dd,J=5.5,1.3Hz,1H),4.01(s,3H),2.15(s,3H). 1 H NMR (400MHz, Chloroform-d) δ11.94 (s, 1H), 8.66 (d, J = 7.5Hz, 1H), 8.28 (d, J = 5.5Hz, 2H), 8.21 (s, 1H), 7.97(dd,J=12.4,2.5Hz,1H),7.38–7.31(m,3H),7.28–7.24(m,2H),7.15(t,J=8.7Hz,1H),6.54(dd,J= 7.6,0.9Hz,1H),6.47(dd,J=5.5,1.3Hz,1H),4.01(s,3H),2.15(s,3H).
实施例2、化合物2合成Example 2, synthesis of compound 2
2.1中间体A2合成
2.1 Synthesis of intermediate A2
向250mL反应瓶中加入4-氨基-2-氟苯酚2.37g,DMAc 50mL,叔丁醇钾2.09g,氮气保护下搅拌0.5小时。加入2-氯-3-氟-4-碘吡啶4g,升温至85℃反应4小时。降至室温,加入水和乙酸乙酯各200mL,搅拌,静置分液。水相再用100mL乙酸乙酯萃取两次。合并乙酸乙酯相,减压浓缩干,得到中间体A2B。Add 2.37g of 4-amino-2-fluorophenol, 50mL of DMAc, and 2.09g of potassium tert-butoxide into the 250mL reaction bottle, and stir for 0.5 hours under nitrogen protection. Add 4g of 2-chloro-3-fluoro-4-iodopyridine, raise the temperature to 85°C and react for 4 hours. Cool to room temperature, add 200 mL each of water and ethyl acetate, stir, and let stand for liquid separation. The aqueous phase was extracted twice with 100 mL of ethyl acetate. Combine the ethyl acetate phases and concentrate to dryness under reduced pressure to obtain intermediate A2B.
向250mL反应瓶中加入上一步得到的中间体A2B,二氧六环100mL,1-甲基吡唑-4-硼酸频哪醇酯4.03g,碳酸钾6.43g,水20mL,氮气置换,加入Pd(PPh3)4 2.5g,氮气置换,升温至90~95℃保温反应20小时。降温至室温,反应液减压浓缩干。浓缩残余物用乙酸乙酯打浆,通过硅藻土过滤。滤液浓缩后通过硅胶柱层析分离(二氯甲烷:乙醇=100:1,v/v),得到中间体A2 2.5g,收率53.4%。Add the intermediate A2B obtained in the previous step, 100 mL of dioxane, 4.03 g of 1-methylpyrazole-4-boronic acid pinacol ester, 6.43 g of potassium carbonate, and 20 mL of water into a 250 mL reaction bottle. Replace with nitrogen and add Pd. (PPh 3 ) 4 2.5g, replaced with nitrogen, heated to 90-95°C and kept for 20 hours. The temperature was cooled to room temperature, and the reaction solution was concentrated to dryness under reduced pressure. The concentrated residue was slurried with ethyl acetate and filtered through Celite. The filtrate was concentrated and separated by silica gel column chromatography (dichloromethane: ethanol = 100:1, v/v) to obtain 2.5 g of intermediate A2 with a yield of 53.4%.
2.2化合物2合成
2.2 Synthesis of Compound 2
向100mL反应瓶中加入中间体A2 0.6g,中间体B1 0.49g,DMF 15mL,HATU 0.91g,三乙胺0.40g,搅拌过夜反应。滴加30mL水,过滤,滤饼干燥得化合物2粗品0.70g,收率66.5%。取360mg样品TLC纯化,得产物118mg,纯度95.28%。Add 0.6g of intermediate A2, 0.49g of intermediate B1, 15mL of DMF, 0.91g of HATU, and 0.40g of triethylamine to the 100mL reaction bottle, and stir overnight for reaction. 30 mL of water was added dropwise, filtered, and the filter cake was dried to obtain 0.70 g of crude compound 2, with a yield of 66.5%. Take 360 mg of the sample and perform TLC purification to obtain 118 mg of product with a purity of 95.28%.
MS:m/z=532.47[M+H]+ MS: m/z=532.47[M+H] +
1H NMR(400MHz,Chloroform-d)δ11.94(s,1H),8.66(d,J=7.6Hz,1H),8.19–8.16(m,2H),8.04(d,J=2.0Hz,1H),7.96(dd,J=12.5,2.5Hz,1H),7.37–7.31(m,3H),7.27–7.24(m,2H),7.15(t,J=8.7Hz,1H),6.56–6.51(m,2H),4.01(s,3H),2.15(s,3H). 1 H NMR(400MHz,Chloroform-d)δ11.94(s,1H),8.66(d,J=7.6Hz,1H),8.19–8.16(m,2H),8.04(d,J=2.0Hz,1H ),7.96(dd,J=12.5,2.5Hz,1H),7.37–7.31(m,3H),7.27–7.24(m,2H),7.15(t,J=8.7Hz,1H),6.56–6.51( m,2H),4.01(s,3H),2.15(s,3H).
实施例3、化合物3合成Example 3, synthesis of compound 3
3.1中间体B2合成
3.1 Synthesis of intermediate B2
向100mL反应瓶中加入4-甲苯胺2g,二氯甲烷20mL,三乙胺2.64g,搅拌降温至0℃。将氯甲酰乙酸乙酯3.65g溶于10mL二氯甲烷,控温0~5℃滴加入反应瓶中。滴完后升至20℃反应2小时,加入20mL水,搅拌分液。二氯甲烷相用20mL饱和食盐水洗,浓缩干。残余物用石油醚12mL和乙酸乙酯3mL打浆,过滤,干燥,得到中间体B2A 3.8g,收率92.0%。Add 2g of 4-toluidine, 20mL of methylene chloride, and 2.64g of triethylamine into the 100mL reaction bottle, stir and cool down to 0°C. Dissolve 3.65g of ethyl chloroformyl acetate in 10 mL of methylene chloride, and add it dropwise into the reaction bottle while controlling the temperature at 0 to 5°C. After dropping, raise to 20°C and react for 2 hours, add 20 mL of water, stir and separate. Wash the dichloromethane phase with 20 mL of saturated brine and concentrate to dryness. The residue was beaten with 12 mL of petroleum ether and 3 mL of ethyl acetate, filtered, and dried to obtain 3.8 g of intermediate B2A with a yield of 92.0%.
向100mL反应瓶中加入中间体B2A 3.8g,乙醇38mL,4-甲氧基丁-3-烯-2-酮2.58g,乙醇钠乙醇溶液12.9mL,升温至回流保温反应6小时。降至室温,加入1mol/L盐酸和二氯甲烷各60mL,搅拌,分液。二氯甲烷相浓缩干。浓缩残余物用20mL乙酸乙酯5℃打浆,过滤,干燥,得到中间体B1 1.73g,收率41.6%。Add 3.8g of intermediate B2A, 38mL of ethanol, 2.58g of 4-methoxybut-3-en-2-one, and 12.9mL of sodium ethoxide ethanol solution into the 100mL reaction bottle, and heat to reflux and incubate for 6 hours. Cool to room temperature, add 60 mL each of 1 mol/L hydrochloric acid and methylene chloride, stir and separate the liquids. The dichloromethane phase was concentrated to dryness. The concentrated residue was beaten with 20 mL of ethyl acetate at 5°C, filtered, and dried to obtain 1.73 g of intermediate B1 with a yield of 41.6%.
3.2化合物3合成
3.2 Synthesis of compound 3
向100mL反应瓶中加入中间体A2 0.6g,中间体B2 0.48g,DMF 15mL,HATU 0.91g,三乙胺0.40g,搅拌过夜反应。滴加30mL水,过滤,滤饼干燥得化合物3粗品0.50g,收率48.2%。取360mg样品TLC纯化,得产物168mg,纯度98.95%。Add 0.6g of intermediate A2, 0.48g of intermediate B2, 15mL of DMF, 0.91g of HATU, and 0.40g of triethylamine to the 100mL reaction bottle, and stir overnight for reaction. Add 30 mL of water dropwise, filter, and dry the filter cake to obtain 0.50 g of crude compound 3, with a yield of 48.2%. 360 mg of the sample was taken and purified by TLC to obtain 168 mg of product with a purity of 98.95%.
MS:m/z=528.50[M+H]+ MS:m/z=528.50[M+H] +
1H NMR(400MHz,Chloroform-d)δ12.05(s,1H),8.64(d,J=7.5Hz,1H),8.18(d,J=5.5Hz,2H),8.04(s,1H),7.96(dd,J=12.5,2.4Hz,1H),7.44(d,J=8.0Hz,2H),7.35(d,J =8.9Hz,1H),7.18–7.10(m,3H),6.56–6.49(m,2H),4.01(s,3H),2.49(s,3H),2.15(s,3H). 1 H NMR (400MHz, Chloroform-d) δ12.05 (s, 1H), 8.64 (d, J = 7.5Hz, 1H), 8.18 (d, J = 5.5Hz, 2H), 8.04 (s, 1H), 7.96(dd,J=12.5,2.4Hz,1H),7.44(d,J=8.0Hz,2H),7.35(d,J =8.9Hz,1H),7.18–7.10(m,3H),6.56–6.49(m,2H),4.01(s,3H),2.49(s,3H),2.15(s,3H).
实施例4、化合物4合成
Example 4, synthesis of compound 4
向100mL反应瓶中加入中间体A1 1g,中间体B2 0.76g,DMF 25mL,HATU 1.43g,三乙胺0.63g,搅拌过夜反应。滴加50mL水,过滤,滤饼干燥得化合物4粗品0.98g,收率57.1%。取360mg样品TLC纯化,得产物214mg,纯度97.86%。Add 1g of intermediate A1, 0.76g of intermediate B2, 25mL of DMF, 1.43g of HATU, and 0.63g of triethylamine to the 100mL reaction bottle, and stir overnight for reaction. 50 mL of water was added dropwise, filtered, and the filter cake was dried to obtain 0.98 g of crude compound 4, with a yield of 57.1%. 360 mg of the sample was taken and purified by TLC to obtain 214 mg of product with a purity of 97.86%.
MS:m/z=544.47[M+H]+ MS: m/z=544.47[M+H] +
1H NMR(400MHz,Chloroform-d)δ12.06(s,1H),8.64(d,J=7.5Hz,1H),8.28(d,J=5.5Hz,2H),8.21(s,1H),7.97(dd,J=12.5,2.4Hz,1H),7.44(d,J=8.0Hz,2H),7.35(d,J=9.4Hz,1H),7.17–7.11(m,3H),6.49(dd,J=19.6,6.2Hz,2H),4.01(s,3H),2.49(s,3H),2.15(s,3H). 1 H NMR (400MHz, Chloroform-d) δ12.06 (s, 1H), 8.64 (d, J = 7.5Hz, 1H), 8.28 (d, J = 5.5Hz, 2H), 8.21 (s, 1H), 7.97(dd,J=12.5,2.4Hz,1H),7.44(d,J=8.0Hz,2H),7.35(d,J=9.4Hz,1H),7.17–7.11(m,3H),6.49(dd ,J=19.6,6.2Hz,2H),4.01(s,3H),2.49(s,3H),2.15(s,3H).
实施例5、化合物5合成Example 5, synthesis of compound 5
5.1中间体B3合成
5.1 Synthesis of intermediate B3
向100mL反应瓶中加入苯胺3g,二氯甲烷30mL,三乙胺4.56g,搅拌降温至0℃。将氯甲酰乙酸乙酯6.30g溶于15mL二氯甲烷,控温0~5℃滴加入反应瓶中。滴完后升至20℃反应2小时,加入30mL水,搅拌分液。二氯甲烷相用30mL饱和食盐水洗,浓缩干,得到中间体B3A。Add 3g of aniline, 30mL of methylene chloride, and 4.56g of triethylamine into a 100mL reaction bottle, stir and cool down to 0°C. Dissolve 6.30 g of ethyl chloroformyl acetate in 15 mL of methylene chloride, and add it dropwise into the reaction bottle while controlling the temperature at 0 to 5°C. After dropping, raise to 20°C and react for 2 hours, add 30 mL of water, stir and separate. The dichloromethane phase was washed with 30 mL of saturated brine, and concentrated to dryness to obtain intermediate B3A.
向100mL反应瓶中加入上一步得到的中间体B3A,乙醇30mL,4-甲氧基丁-3-烯-2-酮4.84g,乙醇钠乙醇溶液24.1mL,升温至回流保温反应7小时。降至室温,加入1mol/L盐酸和二氯甲烷各60mL,搅拌,分液。二氯甲烷相浓缩干。浓缩残余物用20mL乙酸乙酯5℃打浆,过滤,干燥,得到中间体B3 3.76g,收率50.9%。Add the intermediate B3A obtained in the previous step, 30 mL of ethanol, 4.84 g of 4-methoxybut-3-en-2-one, and 24.1 mL of sodium ethoxide ethanol solution into the 100 mL reaction bottle, and heat to reflux and incubate for 7 hours. Cool to room temperature, add 60 mL each of 1 mol/L hydrochloric acid and methylene chloride, stir and separate the liquids. The dichloromethane phase was concentrated to dryness. The concentrated residue was beaten with 20 mL of ethyl acetate at 5°C, filtered, and dried to obtain 3.76 g of intermediate B3 with a yield of 50.9%.
5.2化合物5合成
5.2 Synthesis of compound 5
向100mL反应瓶中加入中间体A2 0.6g,中间体B3 0.46g,DMF 15mL,HATU 0.91g,三乙胺0.40g,搅拌过夜反应。滴加30mL水,过滤,滤饼干燥得化合物5粗品0.52g,收率51.1%。取360mg样品TLC纯化,得产物210mg,纯度99.01%。Add 0.6g of intermediate A2, 0.46g of intermediate B3, 15mL of DMF, 0.91g of HATU, and 0.40g of triethylamine to the 100mL reaction bottle, and stir overnight for reaction. 30 mL of water was added dropwise, filtered, and the filter cake was dried to obtain 0.52 g of crude compound 5, with a yield of 51.1%. 360 mg of the sample was taken and purified by TLC to obtain 210 mg of product with a purity of 99.01%.
MS:m/z=514.47[M+H]+ MS: m/z=514.47[M+H] +
1H NMR(400MHz,Chloroform-d)δ12.00(s,1H),8.66(d,J=7.5Hz,1H),8.18(d,J=5.5Hz,2H),8.04(d,J=1.6Hz,1H),7.96(dd,J=12.5,2.4Hz,1H),7.69–7.53(m,3H),7.35(dd,J=8.8,1.4Hz,1H),7.27(s,1H),7.14(t,J=8.7Hz,1H),6.58–6.49(m,2H),4.01(s,3H),2.15(s,3H). 1 H NMR (400MHz, Chloroform-d) δ12.00 (s, 1H), 8.66 (d, J = 7.5Hz, 1H), 8.18 (d, J = 5.5Hz, 2H), 8.04 (d, J = 1.6 Hz,1H),7.96(dd,J=12.5,2.4Hz,1H),7.69–7.53(m,3H),7.35(dd,J=8.8,1.4Hz,1H),7.27(s,1H),7.14 (t,J=8.7Hz,1H),6.58–6.49(m,2H),4.01(s,3H),2.15(s,3H).
实施例6、化合物6合成
Example 6, synthesis of compound 6
向100mL反应瓶中加入中间体A1 1g,中间体B3 0.72g,DMF 25mL,HATU 1.43g,三乙胺0.63g,搅拌过夜反应。滴加50mL水,过滤,滤饼干燥得化合物6粗品0.95g,收率56.9%。取360mg样品TLC纯化,得产物169mg,纯度99.01%。Add 1g of intermediate A1, 0.72g of intermediate B3, 25mL of DMF, 1.43g of HATU, and 0.63g of triethylamine to the 100mL reaction bottle, and stir overnight for reaction. 50 mL of water was added dropwise, filtered, and the filter cake was dried to obtain 0.95 g of crude compound 6, with a yield of 56.9%. 360 mg of the sample was taken and purified by TLC to obtain 169 mg of product with a purity of 99.01%.
MS:m/z=530.45[M+H]+ MS: m/z=530.45[M+H] +
1H NMR(400MHz,Chloroform-d)δ12.02(s,1H),8.66(d,J=7.5Hz,1H),8.28(d,J=5.4Hz,2H),8.21(s,1H),7.97(dd,J=12.5,2.3Hz,1H),7.69–7.55(m,3H),7.36(d,J=8.3Hz,1H),7.27(s,2H),7.14(t,J=8.7Hz,1H),6.50(dd,J=25.7,6.5Hz,2H),4.01(s,3H),2.14(s,3H). 1 H NMR (400MHz, Chloroform-d) δ12.02 (s, 1H), 8.66 (d, J = 7.5Hz, 1H), 8.28 (d, J = 5.4Hz, 2H), 8.21 (s, 1H), 7.97(dd,J=12.5,2.3Hz,1H),7.69–7.55(m,3H),7.36(d,J=8.3Hz,1H),7.27(s,2H),7.14(t,J=8.7Hz ,1H),6.50(dd,J=25.7,6.5Hz,2H),4.01(s,3H),2.14(s,3H).
实施例7、化合物7合成Example 7, synthesis of compound 7
7.1中间体B4合成
7.1 Synthesis of intermediate B4
向100mL反应瓶中加入4-氯苯胺3g,二氯甲烷30mL,三乙胺3.33g,搅拌降温至0℃。将氯甲酰乙酸乙酯4.6g溶于15mL二氯甲烷,控温0~5℃滴加入反应瓶中。滴完后升至20℃反应2小时,加入30mL水,搅拌分液。二氯甲烷相用30mL饱和食盐水洗, 浓缩干,浓缩残余物用石油醚13.5mL乙酸乙酯4.5mL打浆,过滤,得到中间体B4A 4.52g,收率79.5%。Add 3g of 4-chloroaniline, 30mL of methylene chloride, and 3.33g of triethylamine into a 100mL reaction bottle, stir and cool to 0°C. Dissolve 4.6 g of ethyl chloroformyl acetate in 15 mL of methylene chloride, and add it dropwise into the reaction bottle while controlling the temperature at 0 to 5°C. After dropping, raise to 20°C and react for 2 hours, add 30 mL of water, stir and separate. Wash the methylene chloride phase with 30 mL saturated brine. Concentrate to dryness, and the concentrated residue is slurried with 13.5 mL of petroleum ether and 4.5 mL of ethyl acetate, and filtered to obtain 4.52 g of intermediate B4A with a yield of 79.5%.
向100mL反应瓶中加入中间体B4A 4g,乙醇40mL,4-甲氧基丁-3-烯-2-酮2.48g,乙醇钠乙醇溶液12.5mL,升温至回流保温反应7小时。降至室温,加入1mol/L盐酸和二氯甲烷各60mL,搅拌,分液。二氯甲烷相浓缩干。浓缩残余物用20mL乙酸乙酯5℃打浆,过滤,干燥,得到中间体B4 2.49g,收率57.1%。Add 4g of intermediate B4A, 40mL of ethanol, 2.48g of 4-methoxybut-3-en-2-one, and 12.5mL of sodium ethoxide ethanol solution into the 100mL reaction bottle, and heat to reflux and incubate for 7 hours. Cool to room temperature, add 60 mL each of 1 mol/L hydrochloric acid and methylene chloride, stir and separate the liquids. The dichloromethane phase was concentrated to dryness. The concentrated residue was beaten with 20 mL of ethyl acetate at 5°C, filtered, and dried to obtain 2.49 g of intermediate B4 with a yield of 57.1%.
7.2化合物7合成
7.2 Synthesis of Compound 7
向100mL反应瓶中加入中间体A2 0.6g,中间体B4 0.52g,DMF 15mL,HATU 0.91g,三乙胺0.40g,搅拌过夜反应。滴加30mL水,过滤,滤饼干燥得化合物7粗品0.60g,收率55.3%。取360mg样品TLC纯化,得产物144mg,纯度98.59%。Add 0.6g of intermediate A2, 0.52g of intermediate B4, 15mL of DMF, 0.91g of HATU, and 0.40g of triethylamine to the 100mL reaction bottle, and stir overnight for reaction. 30 mL of water was added dropwise, filtered, and the filter cake was dried to obtain 0.60 g of crude compound 7, with a yield of 55.3%. 360 mg of the sample was taken and purified by TLC to obtain 144 mg of product with a purity of 98.59%.
MS:m/z=548.46[M+H]+ MS: m/z=548.46[M+H] +
1H NMR(400MHz,Chloroform-d)δ11.91(s,1H),8.66(d,J=7.5Hz,1H),8.18(d,J=5.6Hz,2H),8.04(s,1H),7.96(dd,J=12.5,2.4Hz,1H),7.63(d,J=8.6Hz,2H),7.34(d,J=8.8Hz,1H),7.23(d,J=8.6Hz,2H),7.15(t,J=8.7Hz,1H),6.56–6.51(m,2H),4.01(s,3H),2.16(s,3H). 1 H NMR (400MHz, Chloroform-d) δ11.91 (s, 1H), 8.66 (d, J = 7.5Hz, 1H), 8.18 (d, J = 5.6Hz, 2H), 8.04 (s, 1H), 7.96(dd,J=12.5,2.4Hz,1H),7.63(d,J=8.6Hz,2H),7.34(d,J=8.8Hz,1H),7.23(d,J=8.6Hz,2H), 7.15(t,J=8.7Hz,1H),6.56–6.51(m,2H),4.01(s,3H),2.16(s,3H).
实施例8、化合物8合成
Example 8, synthesis of compound 8
向100mL反应瓶中加入中间体A1 1g,中间体B4 0.83g,DMF 25mL,HATU 1.43g,三乙胺0.63g,搅拌过夜反应。滴加50mL水,过滤,滤饼干燥得化合物8粗品1.18g,收率66.7%。取360mg样品TLC纯化,得产物160mg,纯度97.92%。Add 1g of intermediate A1, 0.83g of intermediate B4, 25mL of DMF, 1.43g of HATU, and 0.63g of triethylamine to the 100mL reaction bottle, and stir overnight for reaction. 50 mL of water was added dropwise, filtered, and the filter cake was dried to obtain 1.18 g of crude compound 8, with a yield of 66.7%. Take 360 mg of the sample and perform TLC purification to obtain 160 mg of product with a purity of 97.92%.
MS:m/z=564.39[M+H]+ MS:m/z=564.39[M+H] +
1H NMR(400MHz,Chloroform-d)δ11.92(s,1H),8.66(d,J=7.6Hz,1H),8.28(d,J=5.4Hz,2H),8.21(s,1H),7.97(dd,J=12.5,2.4Hz,1H),7.63(d,J=8.8Hz,2H),7.35(ddd,J=8.7,2.4,1.3Hz,1H),7.25–7.20(m,2H),7.15(t,J=8.7Hz,1H),6.57–6.43(m,2H),4.01(s,3H),2.16(d,J=0.7Hz,3H). 1 H NMR (400MHz, Chloroform-d) δ11.92 (s, 1H), 8.66 (d, J = 7.6Hz, 1H), 8.28 (d, J = 5.4Hz, 2H), 8.21 (s, 1H), 7.97(dd,J=12.5,2.4Hz,1H),7.63(d,J=8.8Hz,2H),7.35(ddd,J=8.7,2.4,1.3Hz,1H),7.25–7.20(m,2H) ,7.15(t,J=8.7Hz,1H),6.57–6.43(m,2H),4.01(s,3H),2.16(d,J=0.7Hz,3H).
实施例9、化合物9合成 Example 9, synthesis of compound 9
9.1中间体A3合成
9.1 Synthesis of Intermediate A3
向50mL反应瓶中加入4-氨基-3-氟苯酚0.84g,DMAc 9mL,叔丁醇钾0.74g,氮气保护下搅拌0.5小时。加入2,3-二氯-4-碘吡啶1.5g,升温至85℃反应9时。降至室温,反应液减压浓缩干。向浓缩残余物中加入乙酸乙酯60mL,搅拌,过滤。乙酸乙酯相减压浓缩干,得到中间体A3B。Add 0.84g of 4-amino-3-fluorophenol, 9mL of DMAc, and 0.74g of potassium tert-butoxide into the 50mL reaction bottle, and stir for 0.5 hours under nitrogen protection. Add 1.5g of 2,3-dichloro-4-iodopyridine, raise the temperature to 85°C and react for 9 hours. After cooling to room temperature, the reaction solution was concentrated to dryness under reduced pressure. 60 mL of ethyl acetate was added to the concentrated residue, stirred, and filtered. The ethyl acetate phase was concentrated to dryness under reduced pressure to obtain intermediate A3B.
向100mL反应瓶中加入中间体A3B,二氧六环30mL,1-甲基吡唑-4-硼酸频哪醇酯1.43g,碳酸钾2.27g,水6mL,氮气置换,加入Pd(PPh3)4 0.5g,氮气置换,升温至90~95℃保温反应30小时。降温至室温,加入水和乙酸乙酯各20mL,搅拌分液。水相再用乙酸乙酯20mL萃取两次。合并乙酸乙酯相,减压浓缩干,通过硅胶柱层析分离(二氯甲烷:乙醇=100:1,v/v),得到中间体A3 1.03g,收率59.2%。Add intermediate A3B, 30 mL of dioxane, 1.43 g of 1-methylpyrazole-4-boronic acid pinacol ester, 2.27 g of potassium carbonate, 6 mL of water into a 100 mL reaction bottle, replace with nitrogen, and add Pd (PPh 3 ) 4 0.5g, replace with nitrogen, raise the temperature to 90~95℃ and keep the reaction for 30 hours. Cool to room temperature, add 20 mL each of water and ethyl acetate, stir and separate the liquids. The aqueous phase was extracted twice with 20 mL of ethyl acetate. The ethyl acetate phases were combined, concentrated to dryness under reduced pressure, and separated by silica gel column chromatography (dichloromethane: ethanol = 100:1, v/v) to obtain 1.03 g of intermediate A3 with a yield of 59.2%.
9.1化合物9合成
9.1 Synthesis of Compound 9
向100mL反应瓶中加入中间体A3 1.03g,中间体B1 0.80g,DMF 15mL,HATU 1.47g,三乙胺0.65g,搅拌过夜反应。滴加30mL水,过滤,滤饼干燥得化合物9粗品1.05g,收率59.3%。取360mg样品TLC纯化,得产物125mg,纯度98.14%。Add 1.03g of intermediate A3, 0.80g of intermediate B1, 15mL of DMF, 1.47g of HATU, and 0.65g of triethylamine to the 100mL reaction bottle, and stir overnight for reaction. 30 mL of water was added dropwise, filtered, and the filter cake was dried to obtain 1.05 g of crude compound 9, with a yield of 59.3%. 360 mg of the sample was taken and purified by TLC to obtain 125 mg of product with a purity of 98.14%.
MS:m/z=548.43[M+H]+ MS: m/z=548.43[M+H] +
1H NMR(400MHz,Chloroform-d)δ11.98(s,1H),8.64(d,J=7.5Hz,1H),8.59(t,J=9.0Hz,1H),8.31(d,J=5.4Hz,1H),8.29(s,1H),8.21(s,1H),7.31(dd,J=9.8,7.2Hz,2H),7.28–7.23(m,2H),6.97–6.90(m,2H),6.58(d,J=5.4Hz,1H),6.51(d,J=7.9Hz,1H),4.01(s,3H),2.13(s,3H). 1 H NMR (400MHz, Chloroform-d) δ11.98 (s, 1H), 8.64 (d, J = 7.5Hz, 1H), 8.59 (t, J = 9.0Hz, 1H), 8.31 (d, J = 5.4 Hz,1H),8.29(s,1H),8.21(s,1H),7.31(dd,J=9.8,7.2Hz,2H),7.28–7.23(m,2H),6.97–6.90(m,2H) ,6.58(d,J=5.4Hz,1H),6.51(d,J=7.9Hz,1H),4.01(s,3H),2.13(s,3H).
实施例10、化合物10合成Example 10, synthesis of compound 10
10.1中间体A4合成
10.1 Synthesis of Intermediate A4
向50mL反应瓶中加入4-氨基-2,3-二氟苯酚0.64g,DMAc 6mL,叔丁醇钾0.50g,氮气保护下搅拌0.5小时。加入2,3-二氯-4-碘吡啶1.0g,升温至85℃反应4时。降至室温,加入乙酸乙酯和水各30mL,搅拌,分液。乙酸乙酯相减压浓缩干,得到中间体A4B。向100mL反应瓶中加入上一步所得的中间体A4B,二氧六环30mL,1-甲基吡唑-4-硼酸频哪醇酯1.15g,碳酸钾1.83g,水6mL,氮气置换,加入Pd(PPh3)4 0.5g,氮气置换,升温至90~95℃保温反应20小时。降温至室温,加入水和乙酸乙酯各20mL,搅拌分液。水相再用乙酸乙酯20mL萃取两次。合并乙酸乙酯相,减压浓缩干,通过硅胶柱层析分离(二氯甲烷:乙醇=100:1,v/v),得到中间体A4 1.30g,收率87.5%。Add 0.64g of 4-amino-2,3-difluorophenol, 6mL of DMAc, and 0.50g of potassium tert-butoxide into the 50mL reaction bottle, and stir for 0.5 hours under nitrogen protection. Add 1.0g of 2,3-dichloro-4-iodopyridine, raise the temperature to 85°C and react for 4 hours. Cool to room temperature, add 30 mL each of ethyl acetate and water, stir, and separate the liquids. The ethyl acetate phase was concentrated to dryness under reduced pressure to obtain intermediate A4B. Add the intermediate A4B obtained in the previous step, 30 mL of dioxane, 1.15 g of 1-methylpyrazole-4-boronic acid pinacol ester, 1.83 g of potassium carbonate, and 6 mL of water into a 100 mL reaction bottle. Replace with nitrogen and add Pd. (PPh 3 ) 4 0.5g, replaced with nitrogen, heated to 90-95°C and kept for 20 hours. Cool to room temperature, add 20 mL each of water and ethyl acetate, stir and separate the liquids. The aqueous phase was extracted twice with 20 mL of ethyl acetate. The ethyl acetate phases were combined, concentrated to dryness under reduced pressure, and separated by silica gel column chromatography (dichloromethane: ethanol = 100:1, v/v) to obtain 1.30 g of intermediate A4 with a yield of 87.5%.
10.2化合物10合成
10.2 Synthesis of compound 10
向100mL反应瓶中加入中间体A4 1.30g,中间体B1 0.95g,DMF 15mL,HATU 1.76g,三乙胺0.78g,搅拌过夜反应。滴加30mL水,过滤,滤饼干燥得化合物10粗品1.82g,收率83.3%。取400mg样品TLC纯化,得产物160mg,纯度96.17%。Add 1.30g of intermediate A4, 0.95g of intermediate B1, 15mL of DMF, 1.76g of HATU, and 0.78g of triethylamine to the 100mL reaction bottle, and stir overnight for reaction. 30 mL of water was added dropwise, filtered, and the filter cake was dried to obtain 1.82 g of crude compound 10, with a yield of 83.3%. 400 mg of the sample was taken and purified by TLC to obtain 160 mg of product with a purity of 96.17%.
MS:m/z=566.46[M+H]+ MS: m/z=566.46[M+H] +
1H NMR(400MHz,Chloroform-d)δ12.10(s,1H),8.64(d,J=7.5Hz,1H),8.40–8.33(m,1H),8.31(d,J=5.5Hz,1H),8.30(d,J=0.7Hz,1H),8.22(s,1H),7.35–7.30(m,2H),7.28–7.24(m,2H),7.03–6.97(m,1H),6.55–6.48(m,2H),4.01(s,3H),2.15(s,3H). 1 H NMR(400MHz,Chloroform-d)δ12.10(s,1H),8.64(d,J=7.5Hz,1H),8.40–8.33(m,1H),8.31(d,J=5.5Hz,1H ),8.30(d,J=0.7Hz,1H),8.22(s,1H),7.35–7.30(m,2H),7.28–7.24(m,2H),7.03–6.97(m,1H),6.55– 6.48(m,2H),4.01(s,3H),2.15(s,3H).
实施例11、化合物11合成Example 11, synthesis of compound 11
11.1中间体A5合成
11.1 Synthesis of intermediate A5
向50mL反应瓶中加入4-氨基-2,5-二氟苯酚0.93g,DMAc 9mL,叔丁醇钾0.72g,氮气保护下搅拌0.5小时。加入2,3-二氯-4-碘吡啶1.46g,升温至85℃反应4时。降至室温,加入乙酸乙酯和水各30mL,搅拌,分液。乙酸乙酯相减压浓缩干,得到中间体A5B。Add 0.93g of 4-amino-2,5-difluorophenol, 9mL of DMAc, and 0.72g of potassium tert-butoxide into the 50mL reaction bottle, and stir for 0.5 hours under nitrogen protection. Add 1.46g of 2,3-dichloro-4-iodopyridine, raise the temperature to 85°C and react for 4 hours. Cool to room temperature, add 30 mL each of ethyl acetate and water, stir, and separate the liquids. The ethyl acetate phase was concentrated to dryness under reduced pressure to obtain intermediate A5B.
向100mL反应瓶中加入中间体A5B,二氧六环30mL,1-甲基吡唑-4-硼酸频哪醇酯1.67g,碳酸钾2.66g,水6mL,氮气置换,加入Pd(PPh3)4 0.5g,氮气置换,升温至90~95℃ 保温反应20小时。降温至室温,加入水和乙酸乙酯各20mL,搅拌分液。水相再用乙酸乙酯20mL萃取两次。合并乙酸乙酯相,减压浓缩干,通过硅胶柱层析分离(二氯甲烷:乙醇=100:1,v/v),得到中间体A5 0.97g,收率44.9%。Add intermediate A5B, 30 mL of dioxane, 1.67 g of 1-methylpyrazole-4-boronic acid pinacol ester, 2.66 g of potassium carbonate, 6 mL of water into a 100 mL reaction bottle, replace with nitrogen, and add Pd (PPh 3 ) 4 0.5g, nitrogen replacement, heating to 90~95℃ The reaction was kept warm for 20 hours. Cool to room temperature, add 20 mL each of water and ethyl acetate, stir and separate the liquids. The aqueous phase was extracted twice with 20 mL of ethyl acetate. The ethyl acetate phases were combined, concentrated to dryness under reduced pressure, and separated by silica gel column chromatography (dichloromethane: ethanol = 100:1, v/v) to obtain 0.97g of intermediate A5 with a yield of 44.9%.
11.2化合物11合成
11.2 Synthesis of compound 11
向100mL反应瓶中加入中间体A5 0.97g,中间体B1 0.71g,DMF 15mL,HATU 1.31g,三乙胺0.58g,搅拌过夜反应。滴加30mL水,过滤,滤饼干燥得化合物11粗品1.13g,收率69.6%。取400mg样品TLC纯化,得产物143mg,纯度99.00%。Add 0.97g of intermediate A5, 0.71g of intermediate B1, 15mL of DMF, 1.31g of HATU, and 0.58g of triethylamine to the 100mL reaction bottle, and stir overnight for reaction. 30 mL of water was added dropwise, filtered, and the filter cake was dried to obtain 1.13 g of crude compound 11, with a yield of 69.6%. 400 mg of the sample was taken and purified by TLC to obtain 143 mg of product with a purity of 99.00%.
MS:m/z=566.46[M+H]+ MS: m/z=566.46[M+H] +
1H NMR(400MHz,Chloroform-d)δ12.13(s,1H),8.67–8.62(m,2H),8.32(d,J=5.4Hz,1H),8.29(s,1H),8.21(s,1H),7.36–7.32(m,2H),7.28–7.22(m,2H),7.01(dd,J=10.3,7.0Hz,1H),6.52(t,J=6.9Hz,2H),4.01(s,3H),2.14(s,3H). 1 H NMR (400MHz, Chloroform-d) δ12.13 (s, 1H), 8.67–8.62 (m, 2H), 8.32 (d, J = 5.4Hz, 1H), 8.29 (s, 1H), 8.21 (s ,1H),7.36–7.32(m,2H),7.28–7.22(m,2H),7.01(dd,J=10.3,7.0Hz,1H),6.52(t,J=6.9Hz,2H),4.01( s,3H),2.14(s,3H).
实施例12、化合物12合成Example 12, synthesis of compound 12
12.1中间体A6合成
12.1 Synthesis of intermediate A6
向50mL反应瓶中加入4-氨基-2,6-二氟苯酚1.25g,DMAc 12mL,叔丁醇钾0.97g,氮气保护下搅拌0.5小时。加入2,3-二氯-4-碘吡啶1.96g,升温至85℃反应5时。降至室温,加入乙酸乙酯和水各50mL,搅拌,分液。乙酸乙酯相减压浓缩干,得到中间体A6B。Add 1.25g of 4-amino-2,6-difluorophenol, 12mL of DMAc, and 0.97g of potassium tert-butoxide into the 50mL reaction bottle, and stir for 0.5 hours under nitrogen protection. Add 1.96g of 2,3-dichloro-4-iodopyridine, raise the temperature to 85°C and react for 5 hours. Cool to room temperature, add 50 mL each of ethyl acetate and water, stir and separate the liquids. The ethyl acetate phase was concentrated to dryness under reduced pressure to obtain intermediate A6B.
向100mL反应瓶中加入中间体A6B,二氧六环30mL,1-甲基吡唑-4-硼酸频哪醇酯2.24g,碳酸钾3.57g,水6mL,氮气置换,加入Pd(PPh3)4 0.5g,氮气置换,升温至90~95℃保温反应20小时。降温至室温,加入水和乙酸乙酯各20mL,搅拌分液。水相再用乙酸乙酯20mL萃取两次。合并乙酸乙酯相,减压浓缩干,通过硅胶柱层析分离(二氯甲烷:乙醇=100:1,v/v),得到中间体A6 2.05g,收率70.7%。Add intermediate A6B, 30 mL of dioxane, 2.24 g of 1-methylpyrazole-4-boronic acid pinacol ester, 3.57 g of potassium carbonate, and 6 mL of water into a 100 mL reaction flask, replace with nitrogen, and add Pd (PPh 3 ) 4 0.5g, replace with nitrogen, raise the temperature to 90~95℃ and keep the reaction for 20 hours. Cool to room temperature, add 20 mL each of water and ethyl acetate, stir and separate the liquids. The aqueous phase was extracted twice with 20 mL of ethyl acetate. The ethyl acetate phases were combined, concentrated to dryness under reduced pressure, and separated by silica gel column chromatography (dichloromethane: ethanol = 100:1, v/v) to obtain 2.05g of intermediate A6 with a yield of 70.7%.
12.2化合物12合成
12.2 Synthesis of compound 12
向100mL反应瓶中加入中间体A6 2.05g,中间体B1 1.51g,DMF 15mL,HATU 2.78g,三乙胺1.23g,搅拌过夜反应。滴加30mL水,过滤,滤饼干燥得化合物12粗品2.40g,收率69.6%。取450mg样品TLC纯化,得产物184mg,纯度99.52%。Add 2.05g of intermediate A6, 1.51g of intermediate B1, 15mL of DMF, 2.78g of HATU, and 1.23g of triethylamine to the 100mL reaction bottle, and stir overnight for reaction. 30 mL of water was added dropwise, filtered, and the filter cake was dried to obtain 2.40 g of crude compound 12, with a yield of 69.6%. 450 mg of the sample was taken for TLC purification to obtain 184 mg of product with a purity of 99.52%.
MS:m/z=566.43[M+H]+ MS: m/z=566.43[M+H] +
1H NMR(400MHz,Chloroform-d)δ12.04(s,1H),8.65(d,J=7.5Hz,1H),8.33–8.28(m,2H),8.22(s,1H),7.57–7.50(m,2H),7.39–7.31(m,2H),7.28–7.23(m,2H),6.55(d,J=7.9Hz,1H),6.48(d,J=5.4Hz,1H),4.01(s,3H),2.16(s,3H). 1 H NMR (400MHz, Chloroform-d) δ12.04 (s, 1H), 8.65 (d, J = 7.5Hz, 1H), 8.33–8.28 (m, 2H), 8.22 (s, 1H), 7.57–7.50 (m,2H),7.39–7.31(m,2H),7.28–7.23(m,2H),6.55(d,J=7.9Hz,1H),6.48(d,J=5.4Hz,1H),4.01( s,3H),2.16(s,3H).
实施例13、化合物13合成Example 13, synthesis of compound 13
13.1中间体A7合成
13.1 Synthesis of Intermediate A7
向50mL反应瓶中加入4-氨基-3,5-二氟苯酚0.50g,DMAc 5mL,叔丁醇钾0.39g,氮气保护下搅拌0.5小时。加入2,3-二氯-4-碘吡啶0.79g,升温至85℃反应5时。降至室温,加入乙酸乙酯和水各50mL,搅拌,分液。乙酸乙酯相减压浓缩干,得到中间体A7B。Add 0.50g of 4-amino-3,5-difluorophenol, 5mL of DMAc, and 0.39g of potassium tert-butoxide into the 50mL reaction bottle, and stir for 0.5 hours under nitrogen protection. Add 0.79g of 2,3-dichloro-4-iodopyridine, raise the temperature to 85°C and react for 5 hours. Cool to room temperature, add 50 mL each of ethyl acetate and water, stir, and separate the liquids. The ethyl acetate phase was concentrated to dryness under reduced pressure to obtain intermediate A7B.
向100mL反应瓶中加入上一步反应获得的中间体A7B,二氧六环30mL,1-甲基吡唑-4-硼酸频哪醇酯0.90g,碳酸钾1.43g,水6mL,氮气置换,加入Pd(PPh3)4 0.5g,氮气置换,升温至90~95℃保温反应20小时。降温至室温,加入水和乙酸乙酯各20mL,搅拌分液。水相再用乙酸乙酯20mL萃取两次。合并乙酸乙酯相,减压浓缩干,通过硅胶柱层析分离(二氯甲烷:乙醇=100:1,v/v),得到中间体A7 1.05g,收率90.2%。Add the intermediate A7B obtained in the previous reaction, 30 mL of dioxane, 0.90 g of 1-methylpyrazole-4-boronic acid pinacol ester, 1.43 g of potassium carbonate, 6 mL of water, nitrogen replacement, and add to the 100 mL reaction bottle. Pd(PPh 3 ) 4 0.5g, replaced with nitrogen, heated to 90-95°C and kept for 20 hours. Cool to room temperature, add 20 mL each of water and ethyl acetate, stir and separate the liquids. The aqueous phase was extracted twice with 20 mL of ethyl acetate. The ethyl acetate phases were combined, concentrated to dryness under reduced pressure, and separated by silica gel column chromatography (dichloromethane: ethanol = 100:1, v/v) to obtain 1.05 g of intermediate A7 with a yield of 90.2%.
13.2化合物13合成
13.2 Synthesis of compound 13
向100mL反应瓶中加入中间体A7 1.05g,中间体B1 0.77g,DMF 15mL,HATU 1.42g, 三乙胺0.63g,搅拌过夜反应。滴加30mL水,过滤,滤饼干燥得化合物13粗品1.06g,收率60.2%。取500mg样品TLC纯化,得产物151mg,纯度96.61%。Add 1.05g of intermediate A7, 0.77g of intermediate B1, 15mL of DMF, and 1.42g of HATU into the 100mL reaction bottle. Triethylamine 0.63g, stir overnight for reaction. 30 mL of water was added dropwise, filtered, and the filter cake was dried to obtain 1.06 g of crude compound 13, with a yield of 60.2%. Take 500 mg of the sample and perform TLC purification to obtain 151 mg of product with a purity of 96.61%.
MS:m/z=566.45[M+H]+ MS: m/z=566.45[M+H] +
1H NMR(400MHz,Chloroform-d)δ11.17(s,1H),8.65(d,J=7.5Hz,1H),8.39(d,J=5.4Hz,1H),8.29(d,J=0.7Hz,1H),8.22(s,1H),7.35–7.30(m,3H),7.25(t,J=5.0Hz,1H),6.77–6.71(m,3H),6.51(d,J=8.3Hz,1H),4.01(s,3H),2.15(s,3H). 1 H NMR (400MHz, Chloroform-d) δ11.17 (s, 1H), 8.65 (d, J = 7.5Hz, 1H), 8.39 (d, J = 5.4Hz, 1H), 8.29 (d, J = 0.7 Hz,1H),8.22(s,1H),7.35–7.30(m,3H),7.25(t,J=5.0Hz,1H),6.77–6.71(m,3H),6.51(d,J=8.3Hz ,1H),4.01(s,3H),2.15(s,3H).
实施例14、化合物15合成Example 14, synthesis of compound 15
14.1中间体A9合成
14.1 Synthesis of Intermediate A9
向50mL反应瓶中加入4-氨基-2-氟苯酚0.90g,DMAc 9mL,叔丁醇钾0.80g,氮气保护下搅拌0.5小时。加入2-氯-4-碘-3-甲基吡啶1.5g,升温至85℃反应3时。降至室温,加入乙酸乙酯和水各30mL,搅拌,分液。乙酸乙酯相减压浓缩干,得到中间体A9B。向100mL反应瓶中加入中间体A9B,二氧六环30mL,1-甲基吡唑-4-硼酸频哪醇酯1.54g,碳酸钾2.45g,水6mL,氮气置换,加入Pd(PPh3)4 0.5g,氮气置换,升温至90~95℃保温反应20小时。降温至室温,加入水和乙酸乙酯各20mL,搅拌分液。水相再用乙酸乙酯20mL萃取两次。合并乙酸乙酯相,减压浓缩干,通过硅胶柱层析分离(二氯甲烷:乙醇=100:1,v/v),得到中间体A9 0.96g,收率54.4%。Add 0.90g of 4-amino-2-fluorophenol, 9mL of DMAc, and 0.80g of potassium tert-butoxide into a 50mL reaction bottle, and stir for 0.5 hours under nitrogen protection. Add 1.5g of 2-chloro-4-iodo-3-methylpyridine, raise the temperature to 85°C and react for 3 hours. Cool to room temperature, add 30 mL each of ethyl acetate and water, stir, and separate the liquids. The ethyl acetate phase was concentrated to dryness under reduced pressure to obtain intermediate A9B. Add intermediate A9B, 30 mL of dioxane, 1.54 g of 1-methylpyrazole-4-boronic acid pinacol ester, 2.45 g of potassium carbonate, 6 mL of water into a 100 mL reaction bottle, replace with nitrogen, and add Pd (PPh 3 ) 4 0.5g, replace with nitrogen, raise the temperature to 90~95℃ and keep the reaction for 20 hours. Cool to room temperature, add 20 mL each of water and ethyl acetate, stir and separate the liquids. The aqueous phase was extracted twice with 20 mL of ethyl acetate. The ethyl acetate phases were combined, concentrated to dryness under reduced pressure, and separated by silica gel column chromatography (dichloromethane: ethanol = 100:1, v/v) to obtain 0.96 g of intermediate A9 with a yield of 54.4%.
14.2化合物15合成
14.2 Synthesis of compound 15
向100mL反应瓶中加入中间体A9 0.96g,中间体B1 0.79g,DMF 15mL,HATU 1.46g,三乙胺0.65g,搅拌过夜反应。滴加30mL水,过滤,滤饼干燥得化合物15粗品1.25g,收率73.9%。取360mg样品TLC纯化,得产物191mg,纯度99.16%。Add 0.96g of intermediate A9, 0.79g of intermediate B1, 15mL of DMF, 1.46g of HATU, and 0.65g of triethylamine to the 100mL reaction bottle, and stir overnight for reaction. 30 mL of water was added dropwise, filtered, and the filter cake was dried to obtain 1.25 g of crude compound 15, with a yield of 73.9%. 360 mg of the sample was taken and purified by TLC to obtain 191 mg of product with a purity of 99.16%.
MS:m/z=528.50[M+H]+ MS:m/z=528.50[M+H] +
1H NMR(400MHz,Chloroform-d)δ11.91(s,1H),8.66(d,J=7.5Hz,1H),8.27(d,J=5.7Hz,1H),7.97–7.90(m,2H),7.86(s,1H),7.37–7.31(m,3H),7.26(dt,J=6.8,2.3Hz, 2H),7.09(t,J=8.7Hz,1H),6.54(dd,J=7.5,0.9Hz,1H),6.43(d,J=5.5Hz,1H),4.00(s,3H),2.51(s,3H). 1 H NMR (400MHz, Chloroform-d) δ11.91 (s, 1H), 8.66 (d, J = 7.5Hz, 1H), 8.27 (d, J = 5.7Hz, 1H), 7.97–7.90 (m, 2H ),7.86(s,1H),7.37–7.31(m,3H),7.26(dt,J=6.8,2.3Hz, 2H),7.09(t,J=8.7Hz,1H),6.54(dd,J=7.5,0.9Hz,1H),6.43(d,J=5.5Hz,1H),4.00(s,3H),2.51( s,3H).
实施例15激酶活性测试Example 15 Kinase Activity Test
本实验通过利用荧光微流体迁移率检测技术(Mobility-Shift Assay),检测本发明化合物对MET和AXL激酶的抑制作用。激酶催化ATP脱去一个磷酸基团生成ADP,并将该磷酸基团转移到底物肽上,该底物肽带有荧光标记,其产物因增加了一个磷酸基团,所带的电荷发生了变化,在电泳泳动过程中,底物和磷酸化的产物因迁移率不同被分开,并分别被检测到,其量与荧光信号成正比。利用Caliper仪器测定底物与产物的量,并计算出产物的转化率,进而计算出抑制率。In this experiment, the inhibitory effect of the compound of the present invention on MET and AXL kinase was detected by using fluorescence microfluidic mobility detection technology (Mobility-Shift Assay). Kinase catalyzes the removal of a phosphate group from ATP to generate ADP, and transfers the phosphate group to the substrate peptide. The substrate peptide is fluorescently labeled, and the charge of the product changes due to the addition of a phosphate group. , during the electrophoresis process, the substrate and phosphorylated products are separated due to different mobilities and are detected respectively, and their amounts are proportional to the fluorescence signal. Use the Caliper instrument to measure the amounts of substrate and product, calculate the conversion rate of the product, and then calculate the inhibition rate.
MET激酶反应体系为25uL,其中包括5nM MET、浓度梯度的小分子抑制剂、10mM MgCl2、1M DTT、26uM ATP(测得的Km值)、3uM Peptide2(5-FAM-EAIYAAPFAKKK-CONH2)、0.0015%Brij-35和pH 7.5的50mM HEPES,2mM DTT,10mM MgCl2;AXL激酶反应体系为25uL,其中包括6nM AXL、浓度梯度的小分子抑制剂、10mM MgCl2、1M DTT、81uM ATP(测得的Km值)、3uM Peptide22(5-FAM-EEPLYWSFPAKKK-CONH2)、0.0015%Brij-35和pH 7.5的50mM HEPES,2mM DTT,10mM MgCl2。The MET kinase reaction system is 25uL, which includes 5nM MET, concentration gradient small molecule inhibitor, 10mM MgCl2, 1M DTT, 26uM ATP (measured Km value), 3uM Peptide2 (5-FAM-EAIYAAPFAKKK-CONH2), 0.0015% Brij-35 and pH 7.5 50mM HEPES, 2mM DTT, 10mM MgCl2; AXL kinase reaction system is 25uL, including 6nM AXL, concentration gradient small molecule inhibitor, 10mM MgCl2, 1M DTT, 81uM ATP (measured Km value ), 3uM Peptide22 (5-FAM-EEPLYWSFPAKKK-CONH2), 0.0015% Brij-35 and 50mM HEPES, pH 7.5, 2mM DTT, 10mM MgCl2.
将酶与抑制剂加入384孔板中室温孵育10分钟,然后加入底物与ATP,反应开始,30分钟后加入25uL Stop Buffer终止反应,Caliper仪器数据,以抑制剂的Log浓度为X轴,抑制率为Y轴绘制曲线,根据公式Y=Bottom+(Top-Bottom)/(1+(IC50/X)^HillSlope)得出IC50。Add the enzyme and inhibitor to the 384-well plate and incubate at room temperature for 10 minutes, then add the substrate and ATP to start the reaction. After 30 minutes, add 25uL Stop Buffer to terminate the reaction. Caliper instrument data, with the Log concentration of the inhibitor as the X-axis, inhibition Draw a curve with the rate on the Y-axis, and get IC50 according to the formula Y=Bottom+(Top-Bottom)/(1+(IC50/X)^HillSlope).
测得各数据如下表1:The measured data are as follows in Table 1:
表1

Table 1

实施例16体外抑瘤活性检测Example 16 In vitro anti-tumor activity assay
检测本发明化合物对不同组织来源的人肿瘤细胞的体外增殖抑制作用。The in vitro proliferation inhibitory effect of the compound of the present invention on human tumor cells derived from different tissues was detected.
实验方法experimental method
1)细胞培养和接种1) Cell culture and inoculation
实验用肿瘤细胞株培养于含10~20%Gibco血清的RPMI-1640、DMEM中,于37℃,5%CO2孵箱中培养。根据实验室的背景数据,接种4000个/孔的肿瘤细胞于96孔培养板,整个实验中细胞处于对数生长期。Experimental tumor cell lines were cultured in RPMI-1640 and DMEM containing 10-20% Gibco serum at 37°C in a 5% CO2 incubator. According to the background data of the laboratory, 4000 tumor cells/well were seeded in a 96-well culture plate. The cells were in the logarithmic growth phase throughout the experiment.
2)给药2) Administration
细胞接种于96孔板后,贴壁过夜,然后根据不同化合物设置5个浓度梯度(0.625~10μM/6.25~100μM),每个浓度两个复孔。After the cells were seeded in a 96-well plate and allowed to adhere overnight, 5 concentration gradients (0.625-10 μM/6.25-100 μM) were set according to different compounds, with two duplicate wells for each concentration.
3)供试品配制3) Preparation of test products
分别取受试物,每管加入DMSO溶解,分装保存于-20℃。临用前用新鲜培养液稀释成工作浓度。具体浓度设置见实验结果部分。给药72小时后,测细胞增殖抑制作用。Take the test substances separately, add DMSO to each tube to dissolve, aliquot and store at -20°C. Dilute to working concentration with fresh culture medium before use. See the experimental results section for specific concentration settings. 72 hours after administration, the cell proliferation inhibitory effect was measured.
4)SRB法4)SRB method
受试物作用细胞72h后,弃去培养液,每孔加入预冷的10%三氯乙酸(TCA)溶液固定细胞,置于4℃冰箱进行固定2h,培养板各孔以去离子水洗涤5遍,以去除三氯乙酸溶液,干燥后每孔加入1%乙酸配制的SRB溶液(4mg/ml),室温下放置20分钟,弃去各孔内液体后用1%乙酸洗涤5遍,洗净未结合的SRB染料,干燥后每孔加入适当体积的10mM Tris-base(三羟甲基胺基甲烷)溶液进行溶解,完全溶解后,酶标仪515nm波长下测定吸光度OD值。After the test substance acts on the cells for 72 hours, discard the culture medium, add pre-cooled 10% trichloroacetic acid (TCA) solution to each well to fix the cells, and place them in a 4°C refrigerator for 2 hours. Each well of the culture plate is washed with deionized water for 5 seconds. times to remove the trichloroacetic acid solution. After drying, add SRB solution (4mg/ml) prepared with 1% acetic acid to each well and leave it at room temperature for 20 minutes. Discard the liquid in each well and wash it 5 times with 1% acetic acid. For unbound SRB dye, add an appropriate volume of 10mM Tris-base (trishydroxymethylaminomethane) solution to each well after drying. After complete dissolution, measure the absorbance OD value with a microplate reader at a wavelength of 515nm.
5)结果处理 5) Result processing
根据酶标仪测定的OD值,按下列公式计算抑制率:According to the OD value measured by the microplate reader, the inhibition rate is calculated according to the following formula:
抑制率(%)=1-OD给药/OD对照×100%,若抑制率≤0,记为0。Inhibition rate (%) = 1-OD administration/OD control × 100%. If the inhibition rate ≤ 0, it is recorded as 0.
计算IC50。Calculate IC50.
6)实验结果6)Experimental results
受试化合物对2种不同组织来源的人肿瘤细胞U87-MG和MKN45具有不同程度抑制作用,其中化合物1、化合物2、化合物5、化合物6、化合物9、化合物11和化合物12和化合物15对U87-MG细胞均有明显抑制作用,尤其是化合物11对U87-MG细胞较敏感,IC50值为2.71μM,(详见表2)。化合物11对MKN45细胞有明显抑制作用,IC50值达到8.71μM,明显优于对照药BMS777607的19.37μM。The tested compounds have varying degrees of inhibitory effects on U87-MG and MKN45, two human tumor cells derived from different tissues. Among them, compound 1, compound 2, compound 5, compound 6, compound 9, compound 11, compound 12 and compound 15 have inhibitory effects on U87 -MG cells have obvious inhibitory effects, especially compound 11 is more sensitive to U87-MG cells, with an IC50 value of 2.71 μM (see Table 2 for details). Compound 11 has a significant inhibitory effect on MKN45 cells, with an IC50 value of 8.71 μM, which is significantly better than the 19.37 μM of the control drug BMS777607.
表2化合物对肿瘤细胞的增殖抑制作用
Table 2: Inhibitory effects of compounds on tumor cell proliferation
实施例17透过血脑屏障能力的评价Example 17 Evaluation of blood-brain barrier penetration ability
将化合物1溶解于DMSO:PEG400:Water=1:6:3(V/V/V)的溶液中,制备成1mg/mL的制剂溶液,按照10mg/kg的剂量给CD1小鼠灌胃给药。于0.25h,0.5h,1h,2h,4h,8h,24h在血浆中和脑部中分别依次取样,然后通过LC-MS/MS检测血浆和脑组织的化学成分的量,得到一系列的药动学参数,结果见下表3和图1、图2。Compound 1 was dissolved in a solution of DMSO:PEG400:Water=1:6:3 (V/V/V) to prepare a preparation solution of 1 mg/mL, and administered to CD1 mice by gavage at a dose of 10 mg/kg. . Samples were taken from plasma and brain at 0.25h, 0.5h, 1h, 2h, 4h, 8h, and 24h, and then the amounts of chemical components in plasma and brain tissue were detected by LC-MS/MS to obtain a series of drugs. Kinetic parameters, the results are shown in Table 3 and Figures 1 and 2 below.
表3

table 3

结果显示,化合物1显示了优秀的口服药代动力学性质,而且能通过血脑屏障,透过率为0.24,药物脑组织暴露量高,可以开发为治疗脑胶质瘤、脑膜瘤、脑转移瘤等脑部肿瘤的药物。The results show that compound 1 shows excellent oral pharmacokinetic properties and can pass through the blood-brain barrier with a permeability of 0.24. The drug has high brain tissue exposure and can be developed to treat brain gliomas, meningiomas, and brain metastases. drugs for brain tumors such as tumors.
实施例18、化合物1对人胃癌细胞MKN45裸小鼠皮下移植瘤的实验治疗作用Example 18. Experimental therapeutic effect of compound 1 on subcutaneous transplantation of human gastric cancer cell MKN45 in nude mice.
方法:将5×106个人胃癌细胞MKN45细胞注射入裸小鼠左腋下。剖取MKN45种鼠瘤块,放入盛有生理盐水的玻璃皿内,剥弃表面血管,切开去除坏死区域后,将瘤块切成1-2mm3,用套管针接入裸小鼠左腋下待肿瘤生长至平均体积100mm3左右后,将动物按瘤体积随机分组后给药。32只小鼠分为4组:G1空白溶媒组(Control)、G2BMS777607 30mg/kg组、G3化合物1 15mg/kg组和G4化合物1 30mg/kg组。每组8只,各组按10mL/kg的给药容量灌胃给予相应浓度的受试物,每天1次,给药周期21天。Methods: 5×106 human gastric cancer MKN45 cells were injected into the left armpit of nude mice. Cut out the tumor pieces from MKN45 mice and place them in a glass dish filled with physiological saline. Peel off the surface blood vessels and remove the necrotic area. Cut the tumor pieces into 1-2 mm 3 and insert them into the nude mice with a trocar. After the tumor in the left armpit grew to an average volume of about 100 mm 3 , the animals were randomly divided into groups according to tumor volume and then administered. 32 mice were divided into 4 groups: G1 blank vehicle group (Control), G2BMS777607 30mg/kg group, G3 compound 1 15mg/kg group and G4 compound 1 30mg/kg group. There were 8 rats in each group, and each group was intragastrically administered the test substance of the corresponding concentration at a dosage volume of 10 mL/kg, once a day, with a 21-day dosing cycle.
每周称重和测量肿瘤体积2次,于第21天称量体重,测量肿瘤大小后处死小鼠取瘤块称重,计算相对肿瘤体积(RTV)、相对肿瘤增值率(T/C)、肿瘤生长抑制率(TGI)和肿瘤抑制百分率(IR),并用SPSS进行统计学分析。Weigh and measure the tumor volume twice a week. Weigh the body weight on the 21st day. After measuring the tumor size, the mice are sacrificed and the tumor blocks are taken out and weighed. The relative tumor volume (RTV), relative tumor proliferation rate (T/C), and Tumor growth inhibition rate (TGI) and tumor inhibition percentage (IR), and SPSS was used for statistical analysis.
结果:result:
实验结束时,与G1空白溶媒组相比,各给药组小鼠体重未见明显变化。详见表4和图3。At the end of the experiment, compared with the G1 blank vehicle group, there was no significant change in the body weight of mice in each administration group. See Table 4 and Figure 3 for details.
表4化合物1对裸小鼠体重的影响
Table 4 Effect of compound 1 on body weight of nude mice
与G1空白溶媒组相比,各组无显著性差异。Compared with the G1 blank vehicle group, there was no significant difference between each group.
与G1空白溶媒组肿瘤体积2220±248mm3相比,G2BMS777607 30mg/kg组、G3化合物1 15mg/kg组和G4化合物1 30mg/kg组肿瘤体积分别为1600±135、900±112(P<0.01)和859±96mm3(P<0.01)。与G1空白溶媒组RTV值24.25±3.24相比,G2BMS777607 30mg/kg组、G3化合物1 15mg/kg组和G4化合物1 30mg/kg组RTV 值分别为16.92±1.17、9.72±1.28(P<0.05)和8.94±0.72(P<0.05);T/C值分别为69.78%、40.07%和36.87%,TGI值分别为30.22%、59.93%和63.13%。(详见图4和图5)。 Compared with the tumor volume of the G1 blank vehicle group of 2220±248mm3, the tumor volumes of the G2BMS777607 30mg/kg group, G3 compound 1 15mg/kg group and G4 compound 1 30mg/kg group were 1600±135 and 900±112 respectively (P<0.01 ) and 859±96mm3 (P<0.01). Compared with the RTV value of G1 blank solvent group of 24.25±3.24, the RTV of G2BMS777607 30mg/kg group, G3 compound 1 15mg/kg group and G4 compound 1 30mg/kg group The values were 16.92±1.17, 9.72±1.28 (P<0.05) and 8.94±0.72 (P<0.05) respectively; the T/C values were 69.78%, 40.07% and 36.87% respectively, and the TGI values were 30.22%, 59.93% and 63.13%. (See Figures 4 and 5 for details).
与G1空白溶媒组瘤块重量1.5907±0.2323g相比,G2BMS777607 30mg/kg组、G3化合物1 15mg/kg组和G4化合物1 30mg/kg瘤块重量分别为1.1525±0.1271、0.6556±0.0907(P<0.05)和0.5886±0.0840g(P<0.05),IR分别为27.55%、58.79%和63.00%。化合物1对人胃癌细胞MKN45皮下移植瘤的作用(详见图6)。Compared with the tumor mass weight of the G1 blank vehicle group of 1.5907±0.2323g, the tumor mass weight of the G2BMS777607 30mg/kg group, G3 compound 1 15mg/kg group and G4 compound 1 30mg/kg group were 1.1525±0.1271 and 0.6556±0.0907 respectively (P< 0.05) and 0.5886±0.0840g (P<0.05), IR were 27.55%, 58.79% and 63.00% respectively. The effect of compound 1 on subcutaneous xenografts of human gastric cancer cells MKN45 (see Figure 6 for details).
结论:本实验条件下,化合物1 15~30mg/kg(q.d.共21天)灌胃给予裸小鼠能够显著剂量依赖性抑制人胃癌细胞MKN45裸小鼠皮下移植瘤的生长,且效果大大优于阳性对照药物BMS777607。Conclusion: Under the conditions of this experiment, Compound 1 administered intragastrically to nude mice at 15-30 mg/kg (q.d. for 21 days) can significantly inhibit the growth of human gastric cancer cell MKN45 subcutaneous transplanted tumors in nude mice in a dose-dependent manner, and the effect is much better than Positive control drug BMS777607.
实施例19使用人肝微粒体评估本发明化合物1对7种CYP450亚型(CYP 1A2、2B6、2C8、2C9、2C19、2D6和3A4)的抑制作用。Example 19 uses human liver microsomes to evaluate the inhibitory effect of Compound 1 of the present invention on 7 CYP450 isoforms (CYP 1A2, 2B6, 2C8, 2C9, 2C19, 2D6 and 3A4).
抑制剂为CYP1A2:a-Naphthoflavone;CYP2C9:Sulfaphenazole;CYP2C19:Omeprazole;CYP3A4:Ketoconazole;CYP2D6:quinidine;CYP2C8:Nicardipine;CYP2B6:Clopidogrel;The inhibitors are CYP1A2: a-Naphthoflavone; CYP2C9: Sulfaphenazole; CYP2C19: Omeprazole; CYP3A4: Ketoconazole; CYP2D6: quinidine; CYP2C8: Nicardipine; CYP2B6: Clopidogrel;
基质为CYP1A2:Phenacetin at 30μM;CYP2C9:Diclofenac at 10μM;CYP2C19:S-Mephenytoin at 35μM;CYP3A4:Midazolam at 5μM;CYP2D6:Bufuralol at 10μM;CYP2C8:Paclitaxel at 10μM;CYP2B6:Bupropion at 70μM.The matrix is CYP1A2: Phenacetin at 30μM; CYP2C9: Diclofenac at 10μM; CYP2C19: S-Mephenytoin at 35μM; CYP3A4: Midazolam at 5μM; CYP2D6: Bufuralol at 10μM; CYP2C8: Paclitaxel at 10μM; CYP2B6: Bupropion at 70μM.
检测系统Human liver microsomes from Corning,孵化条件CYP1A2,2C9,2D6,2C8,2B6:10分钟,37℃;CYP2C19:45分钟,37℃;CYP3A4:5分钟,37℃.样本量2份(n=2),生物分析方法为LC-MS/MS。Detection system Human liver microsomes from Corning, incubation conditions CYP1A2, 2C9, 2D6, 2C8, 2B6: 10 minutes, 37℃; CYP2C19: 45 minutes, 37℃; CYP3A4: 5 minutes, 37℃. Sample size 2 copies (n=2 ), the bioanalytical method is LC-MS/MS.
计算方式:基于使用以下公式的数据计算,使用Sigmoidal(非线性)剂量反应模型(GraphPad Prism 5.0或Xlfit模型205)执行曲线拟合以计算IC50:Y=Bottom+(Top-Bottom)/(1+10^((LogIC50-X)*HillSlope))。其中X是浓度的对数。Y是响应抑制剂浓度从高到低的S形从底部到顶部的响应。结果见表5。Calculation method: Perform curve fitting using a Sigmoidal (nonlinear) dose-response model (GraphPad Prism 5.0 or Xlfit Model 205) to calculate IC50 based on data calculations using the following formula: Y=Bottom+(Top-Bottom)/(1+10 ^((LogIC50-X)*HillSlope)). where X is the logarithm of concentration. Y is the S-shaped response from bottom to top in response to inhibitor concentration from high to low. The results are shown in Table 5.
表5本发明化合物1对7种CYP450亚型的抑制作用
Table 5 Inhibitory effects of compound 1 of the present invention on 7 CYP450 subtypes
与现有技术相比,本发明具有如下有益效果:Compared with the prior art, the present invention has the following beneficial effects:
1)本发明提供的Axl&c-Met双重抑制剂对AXL,c-Met均有较强的抑制活性化合物,可同时抑制GAS6/AXL以及HGF/c-Met两种信号通路,预期将产生更加显著的抗肿瘤效果,1) The Axl&c-Met dual inhibitor provided by the present invention has strong inhibitory activity against both AXL and c-Met. It can inhibit both GAS6/AXL and HGF/c-Met signaling pathways at the same time, and is expected to produce more significant effects. anti-tumor effect,
2)本发明化合物的Axl和c-Met激酶抑制效果优于阳性对照药BMS777607。在人源化胃癌MKN45模型中,本发明的Axl&c-Met双重抑制剂显示了显著的抗肿瘤活性,且明显优于阳性对照药BMS777607,动物耐受性好。2) The inhibitory effect of the compound of the present invention on Axl and c-Met kinase is better than that of the positive control drug BMS777607. In the humanized gastric cancer MKN45 model, the Axl&c-Met dual inhibitor of the present invention showed significant anti-tumor activity, was significantly better than the positive control drug BMS777607, and was well tolerated by animals.
3)本发明的Axl&c-Met双重抑制剂具有出色的药代动力学性质,有望开发成抗肿瘤药物;尤其是,该系列抑制剂有较强的透过血脑屏障的能力,在脑部组织具有良好的暴露量,可以用于开发脑胶质瘤、脑转移瘤、脑膜瘤等脑部肿瘤药物。3) The Axl&c-Met dual inhibitor of the present invention has excellent pharmacokinetic properties and is expected to be developed into an anti-tumor drug; in particular, this series of inhibitors has a strong ability to penetrate the blood-brain barrier and in brain tissue With good exposure, it can be used to develop drugs for brain tumors such as glioma, brain metastases, and meningiomas.
4)本发明的Axl&c-Met双重抑制作用有望克服肿瘤药的耐药性。4) The dual inhibitory effect of Axl&c-Met of the present invention is expected to overcome the resistance of tumor drugs.
5)本发明还提供的一系列Axl&c-Met双重抑制作用的化合物的简便制备方法。5) The present invention also provides a simple preparation method for a series of compounds with dual Axl&c-Met inhibitory effects.
以上对本发明的具体实施例进行了描述。需要理解的是,本发明并不局限于上述特定实施方式,本领域技术人员可以在权利要求的范围内做出各种变形或修改,这并不影响本发明的实质内容。 Specific embodiments of the present invention have been described above. It should be understood that the present invention is not limited to the specific embodiments described above. Those skilled in the art can make various variations or modifications within the scope of the claims, which does not affect the essence of the present invention.

Claims (10)

  1. 如式(I)所示化合物或其药学上可接受的盐、其前药、其水合物或溶剂合物:
    For example, the compound represented by formula (I) or its pharmaceutically acceptable salt, its prodrug, its hydrate or solvate:
    其中,R1为氢、卤素、取代或未取代的烷基、烷氧基或者卤代烷氧基、取代或未取代的环烷基、取代或未取代的芳基;Wherein, R 1 is hydrogen, halogen, substituted or unsubstituted alkyl, alkoxy or haloalkoxy, substituted or unsubstituted cycloalkyl, substituted or unsubstituted aryl;
    R2为卤素、取代或未取代的烷基;R 2 is halogen, substituted or unsubstituted alkyl;
    n的数值为1或者2。The value of n is 1 or 2.
  2. 如权利要求1所述化合物或其药学上可接受的盐、其前药、其水合物或溶剂合物,其特征在于,所述烷基为C1-C6的烷基,所述环烷基为C3-C4的环烷基,所述烷氧基为C1-C6的烷氧基,所述羟烷基为C1-C6的羟烷基,取代基为氟或者氯。The compound of claim 1 or a pharmaceutically acceptable salt thereof, a prodrug thereof, a hydrate or a solvate thereof, wherein the alkyl group is a C1-C6 alkyl group, and the cycloalkyl group is C 3 -C 4 cycloalkyl group, the alkoxy group is a C 1 -C 6 alkoxy group, the hydroxyalkyl group is a C 1 -C 6 hydroxyalkyl group, and the substituent is fluorine or chlorine.
  3. 如权利要求1所述化合物或其药学上可接受的盐、其前药、其水合物或溶剂合物,其特征在于,R1选自氢、甲基、甲氧基、三氟甲基、三氟甲氧基、氟或氯;R2选自氟、氯、甲基。The compound of claim 1 or a pharmaceutically acceptable salt thereof, a prodrug thereof, a hydrate or a solvate thereof, wherein R 1 is selected from hydrogen, methyl, methoxy, trifluoromethyl, Trifluoromethoxy, fluorine or chlorine; R 2 is selected from fluorine, chlorine, methyl.
  4. 如权利要求1所述化合物或其药学上可接受的盐、其前药、其水合物或溶剂合物,其特征在于,所述化合物为以下化合物中的任意一个:

    The compound of claim 1 or a pharmaceutically acceptable salt thereof, a prodrug thereof, a hydrate or a solvate thereof, wherein the compound is any one of the following compounds:

  5. 一种药物组合物,其特征在于,包含药学上可接受的载体和一种或多种治疗有效量的如权利要求1所述的化合物或其药学上可接受的盐、其前药、其水合物或溶剂合物。A pharmaceutical composition, characterized by comprising a pharmaceutically acceptable carrier and one or more therapeutically effective amounts of the compound of claim 1 or a pharmaceutically acceptable salt thereof, a prodrug thereof, or a hydrate thereof substance or solvate.
  6. 一种如权利要求1所述的化合物或其药学上可接受的盐、其前药、其水合物或溶剂合物的制备方法,其特征在于,所述方法包括如下步骤:A method for preparing the compound of claim 1 or a pharmaceutically acceptable salt thereof, a prodrug thereof, a hydrate or a solvate thereof, characterized in that the method includes the following steps:
    反应生成如式(I)所示化合物。 The reaction produces a compound represented by formula (I).
  7. 一种如权利要求1所述的化合物或其药学上可接受的盐、其前药、其水合物或溶剂合物在制备抗肿瘤药物中的用途。The use of a compound as claimed in claim 1 or a pharmaceutically acceptable salt thereof, a prodrug thereof, a hydrate or a solvate thereof in the preparation of anti-tumor drugs.
  8. 一种如权利要求1所述的化合物或其药学上可接受的盐、其前药、其水合物或溶剂合物在制备治疗Axl激酶和/或c-Met激酶引起的相关疾病的药物中的用途。 A compound as claimed in claim 1 or a pharmaceutically acceptable salt thereof, a prodrug thereof, a hydrate or a solvate thereof in the preparation of a medicament for treating related diseases caused by Axl kinase and/or c-Met kinase use.
  9. 如权利要求8所述的用途,其特征在于所述Axl激酶和/或c-Met激酶引起的相关疾病包括脑部肿瘤、胃癌、膀肮癌、乳腺癌、结肠直肠癌、头颈癌、肾癌、肝癌、肺癌、卵巢癌、膜腺癌/胆囊癌、前列腺癌、甲状腺癌、骨肉瘤、横纹肌肉瘤、MFH/纤维肉瘤、恶性胶质瘤/星形细胞瘤、黑色素瘤或间皮瘤、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肾病。The use according to claim 8, characterized in that the related diseases caused by the Axl kinase and/or c-Met kinase include brain tumors, gastric cancer, bladder cancer, breast cancer, colorectal cancer, head and neck cancer, and kidney cancer. , liver cancer, lung cancer, ovarian cancer, pancreatic adenocarcinoma/gallbladder cancer, prostate cancer, thyroid cancer, osteosarcoma, rhabdomyosarcoma, MFH/fibrosarcoma, glioblastoma/astrocytoma, melanoma or mesothelioma, psoriasis , liver cirrhosis, diabetes, angiogenesis, restenosis, ophthalmic diseases, rheumatoid arthritis and other inflammatory diseases, immune diseases, cardiovascular diseases such as arteriosclerosis and nephropathy.
  10. 如权利要求9所述的用途,其特征在于,所述脑部肿瘤包括脑胶质瘤、脑膜瘤、脑转移瘤。 The use according to claim 9, wherein the brain tumors include gliomas, meningiomas, and brain metastases.
PCT/CN2023/106523 2022-07-25 2023-07-10 Axl&c-met dual inhibitor, and preparation method and use WO2024022095A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210879471.9A CN117486860A (en) 2022-07-25 2022-07-25 Axl & c-Met dual inhibitor, preparation method and application
CN202210879471.9 2022-07-25

Publications (1)

Publication Number Publication Date
WO2024022095A1 true WO2024022095A1 (en) 2024-02-01

Family

ID=89680546

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2023/106523 WO2024022095A1 (en) 2022-07-25 2023-07-10 Axl&c-met dual inhibitor, and preparation method and use

Country Status (2)

Country Link
CN (1) CN117486860A (en)
WO (1) WO2024022095A1 (en)

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101128199A (en) * 2004-04-23 2008-02-20 布里斯托尔-迈尔斯斯奎布公司 Monocyclic heterocycles as kinase inhibitors
WO2011139891A1 (en) * 2010-04-29 2011-11-10 Deciphera Pharmaceuticals, Llc Pyridone amides and analogs exhibiting anti-cancer and anti-proliferative activites
US20140343103A1 (en) * 2013-03-15 2014-11-20 Deciphera Pharmaceuticals, Llc Pyridone amides and analogs exhibiting anti-cancer and anti-proliferative activities
WO2016104617A1 (en) * 2014-12-25 2016-06-30 小野薬品工業株式会社 Quinoline derivative
CN110330479A (en) * 2019-07-19 2019-10-15 南京华威医药科技集团有限公司 A kind of antitumoral compounds and application thereof as AXL inhibitor
WO2020042618A1 (en) * 2018-08-27 2020-03-05 北京越之康泰生物医药科技有限公司 Multi-substituted pyridone derivatives and medical use thereof
WO2021198709A1 (en) * 2020-04-03 2021-10-07 Kinsensus Limited Naphthyridine compounds as inhibitors of mer tyrosine kinase and axl tyrosine kinase

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101128199A (en) * 2004-04-23 2008-02-20 布里斯托尔-迈尔斯斯奎布公司 Monocyclic heterocycles as kinase inhibitors
WO2011139891A1 (en) * 2010-04-29 2011-11-10 Deciphera Pharmaceuticals, Llc Pyridone amides and analogs exhibiting anti-cancer and anti-proliferative activites
US20140343103A1 (en) * 2013-03-15 2014-11-20 Deciphera Pharmaceuticals, Llc Pyridone amides and analogs exhibiting anti-cancer and anti-proliferative activities
WO2016104617A1 (en) * 2014-12-25 2016-06-30 小野薬品工業株式会社 Quinoline derivative
WO2020042618A1 (en) * 2018-08-27 2020-03-05 北京越之康泰生物医药科技有限公司 Multi-substituted pyridone derivatives and medical use thereof
CN110330479A (en) * 2019-07-19 2019-10-15 南京华威医药科技集团有限公司 A kind of antitumoral compounds and application thereof as AXL inhibitor
WO2021198709A1 (en) * 2020-04-03 2021-10-07 Kinsensus Limited Naphthyridine compounds as inhibitors of mer tyrosine kinase and axl tyrosine kinase

Also Published As

Publication number Publication date
CN117486860A (en) 2024-02-02

Similar Documents

Publication Publication Date Title
US8436011B2 (en) Pyridinopyridinone derivatives, preparation thereof and therapeutic use thereof
CA2720552C (en) Acylthiourea compound or salt thereof, and use thereof
TW201716415A (en) Chiral diaryl macrocycles as modulators of protein kinases
JP2018514559A (en) Pyrimidopyrroles compound, preparation method thereof, pharmaceutical composition and application thereof
WO2014040549A1 (en) Alkynyl heteroaromatic ring compound and application thereof
KR20140016889A (en) Diarylacetylene hydrazide containing tyrosine kinase inhibitors
WO2020038460A1 (en) Novel quinoline derivative inhibitor
KR20120099732A (en) Pyridine-pyridinone derivatives, preparation and therapeutic use thereof
CN107531683A (en) USP7 inhibitor compounds and application method
Xu et al. Synthesis and biological evaluation of 4-(pyridin-4-oxy)-3-(3, 3-difluorocyclobutyl)-pyrazole derivatives as novel potent transforming growth factor-β type 1 receptor inhibitors
WO2015117551A1 (en) Pyrrole-substituted indolone derivative, preparation method therefor, composition comprising same and use thereof
WO2023078451A1 (en) Compound used as cdk7 kinase inhibitor and use thereof
WO2016041472A1 (en) Preparation method for aromatic heterocyclic compound used as selective jak3 and/or jak1 kinase inhibitor and application of aromatic heterocyclic compound
WO2002002550A1 (en) Novel pyrazinone derivatives
WO2018157730A1 (en) Urea-substituted aromatic ring-linked dioxane-quinazoline and -linked dioxane-quinoline compounds, preparation method therefor and use thereof
Dai et al. Design, synthesis and biological evaluation of 4-(4-aminophenoxy) picolinamide derivatives as potential antitumor agents
WO2017144025A1 (en) Pyrimido[4,5-d][1,3]oxazin-2-one derivative serving as egfr inhibitor, and application thereof
WO2024022095A1 (en) Axl&c-met dual inhibitor, and preparation method and use
CN110467616B (en) Preparation and application of triazolopyrazine compound containing heteroaryl substituted pyridazinone structure
CN102688234B (en) Indolone derivatives is as the Synthesis and application of RSK2 inhibitor
TWI546304B (en) Protein tyrosine kinase inhibitors and their use
Xi et al. Modification of osimertinib to discover new potent EGFRC797S-TK inhibitors
WO2019157959A1 (en) Pyrimidine compound, preparation method therefor and medical use thereof
CN115322158A (en) As KRAS G12C Substituted quinazoline compounds of protein inhibitor
CN110845476B (en) High-selectivity CSF1R inhibitor, preparation method and pharmaceutical application thereof

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: 23845308

Country of ref document: EP

Kind code of ref document: A1