WO2018028541A1 - Substituted quinoline compound and pharmaceutical composition and use thereof - Google Patents
Substituted quinoline compound and pharmaceutical composition and use thereof Download PDFInfo
- Publication number
- WO2018028541A1 WO2018028541A1 PCT/CN2017/096198 CN2017096198W WO2018028541A1 WO 2018028541 A1 WO2018028541 A1 WO 2018028541A1 CN 2017096198 W CN2017096198 W CN 2017096198W WO 2018028541 A1 WO2018028541 A1 WO 2018028541A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- compound
- hepatitis
- synthesis
- mixture
- added
- Prior art date
- Legal status (The legal status 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 status listed.)
- Ceased
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/435—Heterocyclic 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
- A61K31/47—Quinolines; Isoquinolines
- A61K31/4709—Non-condensed quinolines and containing further heterocyclic rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/14—Antivirals for RNA viruses
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07B—GENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
- C07B59/00—Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D417/00—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
- C07D417/14—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
Definitions
- the invention belongs to the technical field of medicine, and in particular relates to a hepatitis C virus inhibitor, a pharmaceutical composition and application thereof.
- Hepatitis C is a blood-borne infectious liver disease that, if left untreated, can cause significant damage to the liver. About 185 million people worldwide are infected with hepatitis C virus. The rate of hepatitis C infection in China is about 3.2%, and the number of patients is about 40 million. In the United States, there are about 3.2 million hepatitis C patients, and about 15,000 people die each year. Usually die from hepatitis C-related diseases such as cirrhosis and liver cancer.
- HCV Hepatitis C Virus
- the encapsulated HCV virion contains a positive-stranded RNA genome that encodes all known virus-specific proteins in a single uninterrupted open reading frame.
- the open reading frame comprises approximately 9500 nucleotides and encodes a single large polyprotein of approximately 3000 amino acids.
- Polyproteins include core proteins, envelope proteins E1 and E2, membrane-bound protein P7, and non-structural proteins NS2, NS3/4A, NS4A, NS4B, NS5A, and NS5B.
- Sofabru is the first medicine in the world that can completely cure hepatitis C in the short term. It takes the oral route directly to the lesion, and the method has simple side effects and is highly sought after by patients. Sofibuvir is produced by Gilead, USA, and is marketed in the United States in 2013. It has been clinically proven to be effective in treating hepatitis C, type 1, 2, 3, and 4, including liver transplantation, liver cancer, and HCV/HIV-1 co-infection. Clinical Trials. This breakthrough has brought the gospel to hepatitis C patients around the world.
- HCV infection is associated with progressive liver disease, including cirrhosis and hepatocellular carcinoma.
- Simeprevir is a new generation of NS3/4A protease inhibitors, a daily oral medication developed by Medivir and Janssen for the treatment of compensatory liver disease in adult patients with chronic hepatitis C. Including liver fibrosis at various stages, its working principle is to inhibit the replication of HCV in liver cells by blocking proteases.
- hepatitis C a new drug, dexamethavir was approved by the FDA, combined with peginterferon and ribavirin for genotype 1 chronic hepatitis C adult patients with compensatory liver disease ( Treatment including cirrhosis.
- the present invention discloses a hepatitis C virus inhibitor, a pharmaceutical composition and use thereof, which have better hepatitis C virus protein NS3/4A inhibitory activity and/or have better pharmacodynamics/pharmacokinetics. Kinetic performance.
- a hepatitis C virus inhibitor such as a quinoline compound of the formula (I), or a crystalline form thereof, a pharmaceutically acceptable salt, a hydrate or a solvent compound,
- R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 18 , R 19 , R 20 , R 21 , R 22 , R 23 , R 24 , R 25 , R 26 , R 27 , R 28 , R 29 , R 30 , R 31 , R 32 , R 33 And R 33 , R 34 , R 35 , R 36 , R 37 , R 38 , R 39 , R 40 , R 41 , R 42 , R 43 , R 44 , R 45 are each independently hydrogen, deuterium, halogen or trisole Fluoromethyl;
- Additional conditions are R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 , R 16 And R 17 , R 18 , R 19 , R 20 , R 21 , R 22 , R 23 , R 24 , R 25 , R 26 , R 27 , R 28 , R 29 , R 30 , R 31 , R 32 , R 33. At least one of R 33 , R 34 , R 35 , R 36 , R 37 , R 38 , R 39 , R 40 , R 41 , R 42 , R 43 , R 44 and R 45 is deuterated.
- the shape and volume of the ruthenium in the drug molecule are substantially the same as those of the hydrogen. If the hydrogen in the drug molecule is selectively replaced with hydrazine, the deuterated drug generally retains the original biological activity and selectivity. At the same time, the inventors have confirmed through experiments that the binding of carbon-germanium bonds is more stable than the combination of carbon-hydrogen bonds, which can directly affect the absorption, distribution, metabolism and excretion of some drugs, thereby improving the efficacy, safety and tolerability of the drugs.
- the strontium isotope content of the cerium in the deuterated position is at least greater than the natural strontium isotope content (0.015%), preferably greater than 30%, more preferably greater than 50%, more preferably greater than 75%, and even more preferably greater than 95. %, more preferably greater than 99%.
- R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 and R 14 , R 15 , R 16 , R 17 , R 18 , R 19 , R 20 , R 21 , R 22 , R 23 , R 24 , R 25 , R 26 , R 27 , R 28 , R 29 , R 30 , R 31 And R 32 , R 33 , R 33 , R 34 , R 35 , R 36 , R 37 , R 38 , R 39 , R 40 , R 41 , R 42 , R 43 , R 44 and R 45 are each in the metamorphic position
- the strontium isotope content is at least 5%, preferably more than 10%, more preferably more than 15%, more preferably more than 20%, more preferably more than 25%, more preferably more than 30%, more preferably more than 35%, More preferably
- R 19 , R 20 and R 21 are each independently hydrazine or hydrogen.
- R 19 , R 20 and R 21 are ⁇ .
- R 32 , R 33 and R 34 are each independently hydrazine or hydrogen.
- R 32 , R 33 , and R 34 are deuterium.
- R 35 , R 36 and R 37 are each independently hydrazine or hydrogen.
- R 35 , R 36 , and R 37 are ⁇ .
- the compound is selected from the group consisting of the following compounds or a pharmaceutically acceptable salt thereof:
- the compound does not include a non-deuterated compound.
- the present invention also discloses a pharmaceutical composition
- a pharmaceutical composition comprising a pharmaceutically acceptable carrier and the hepatitis C virus inhibitor as described above, or a crystalline form, a pharmaceutically acceptable salt, a hydrate or a solvent thereof
- a pharmaceutical composition of a compound, stereoisomer, prodrug or isotopic variation comprising a pharmaceutically acceptable carrier and the hepatitis C virus inhibitor as described above, or a crystalline form, a pharmaceutically acceptable salt, a hydrate or a solvent thereof.
- it further comprises other active compounds including, but not limited to, other HCV antiviral agents, anti-infective agents, immunomodulators, antibiotics or vaccine combinations.
- the immunomodulator is an interferon drug compound.
- the quinoxaline macrocycles of the invention may be used in combination therapies involving one or more additional therapeutic agents.
- Additional therapeutic agents include those that also target HCV, target different pathogenic agents, or enhance the immune system.
- Agents that enhance the immune system include those that normally enhance the function of the immune system and those that produce a specific immune response against HCV.
- Additional therapeutic agents that target HCV include agents that target NS3/4A and agents that target other HCV activities, such as NS5A and NS5B, and agents that target host cell activity involved in HCV replication.
- therapeutic agents that may be present in the combination include ribavirin, levovirin, viramidine, thymosin alpha-1, interferon-beta, interferon-alpha, PEGylated interferon-alpha (peg interferon-alpha) a combination of interferon-[alpha] and ribavirin, a combination of peg interferon-[alpha] and ribavirin, a combination of interferon-[alpha] and levovirin, and a combination of peg interferon-[alpha] and levovirin.
- peg interferon-alpha a combination of interferon-[alpha] and ribavirin
- peg interferon-[alpha] and ribavirin a combination of interferon-[alpha] and levovirin
- peg interferon-[alpha] and levovirin a combination of peg interferon-[alpha] and levovirin.
- Interferon- ⁇ includes recombinant interferon- ⁇ 2a (such as Roferon interferon available from Hoffmann-LaRoche, Nutley, NJ), PEGylated interferon- ⁇ 2a (Pegasys), interferon- ⁇ 2b (eg available from Schering Corp) .Kenilworth, NJ's Intron-A interferon), PEGylated interferon- ⁇ 2b (PegIntron), recombinant complex interferon (such as interferon alphacon-1), and purified interferon- ⁇ product.
- the individual components of the combination may be administered separately at different times during the course of the treatment or simultaneously in separate or individual combinations.
- the compounds of the invention may also be administered in combination with the antiviral agent amantadine (1-aminoadamantane).
- amantadine 1-aminoadamantane
- the compounds of the invention may also be administered in combination with the antiviral polymerase inhibitor R7128 (Roche).
- the compounds of the invention may also be administered in combination with an HCV NS5B polymerase inhibitor.
- HCV NS5B polymerase inhibitors that can be used as a combination therapy include, but are not limited to, International Patent Application Publication No. WO 02/057287, WO 02/057425, WO 03/068244, WO 2004/000858, WO 04/003138, and WO Those disclosed in U.S. Patent No. 6,777,392 and U.S. Patent Application Publication No.
- HCV polymerase inhibitors include, but are not limited to, valopicitabine (NM-283; Idenix) and 2'-F-2'-beta-methylcytidine (see also WO 2005/003147).
- the pharmaceutically acceptable carrier includes a glidant, a sweetener, a diluent, a preservative, a dye/colorant, a flavor enhancer, a surfactant, a wetting agent, a dispersant At least one of a disintegrant, a suspending agent, a stabilizer, an isotonic agent, a solvent or an emulsifier.
- the pharmaceutical composition is a tablet, a pill, a capsule, a powder, a granule, an ointment, an emulsion, a suspension, a solution, a suppository, an injection, an inhalant, a gel, a microsphere or Aerosol.
- Typical routes of administration of the pharmaceutical compositions of the invention include, but are not limited to, oral, rectal, transmucosal, enteral, or topical, transdermal, inhalation, parenteral, sublingual, intravaginal, intranasal, intraocular, intraperitoneal , intramuscular, subcutaneous, intravenous administration. Oral administration or injection administration is preferred.
- the pharmaceutical composition of the present invention can be produced by a method known in the art, such as a conventional mixing method, a dissolution method, a granulation method, a sugar-coating method, a pulverization method, an emulsification method, a freeze-drying method, and the like.
- the present invention also provides a method of preparing a pharmaceutical composition comprising the steps of: administering a pharmaceutically acceptable carrier to a hepatitis C virus inhibitor as described above, or a crystalline form thereof, a pharmaceutically acceptable salt, or a hydrate thereof Or the solvate is mixed to form a pharmaceutical composition.
- the invention also discloses the use of a hepatitis C virus inhibitor as described above, characterized in that it is used for the preparation of a medicament for the treatment of hepatitis C virus infection.
- NS3/4A inhibitors are also useful in the preparation and implementation of screening assays for antiviral compounds.
- such compounds can be used to isolate enzyme mutants, which are excellent screening tools for more potent antiviral compounds.
- these compounds can be used to establish or measure binding sites for other antiviral agents to HCV protease, for example by competitive inhibition.
- the compounds of the invention can be administered by contacting the active agent with the site of action of the drug. medicine. They can be administered as a separate therapeutic or combination of therapeutic agents by conventional means which can be used in combination with the drug. They can be administered alone, but they are It is often administered with a pharmaceutical carrier selected according to the chosen route of administration and standard pharmaceutical practice.
- the HCV includes a plurality of genotypes thereof and a plurality of gene subtypes, such as 1a, 1b, 2a, 2b, 3a, 3b, 4a, 5a, 6a.
- halogen means F, Cl, Br, and I unless otherwise specified. More preferably, the halogen atom is selected from the group consisting of F, Cl and Br.
- deuterated means that one or more hydrogens in the compound or group are replaced by deuterium; deuteration may be monosubstituted, disubstituted, polysubstituted or fully substituted.
- deuteration may be monosubstituted, disubstituted, polysubstituted or fully substituted.
- deuterated is used interchangeably with “one or more deuterated”.
- non-deuterated compound means a compound containing a proportion of germanium atoms not higher than the natural helium isotope content (0.015%).
- Pharmaceutically acceptable salts include inorganic and organic salts.
- a preferred class of salts are the salts of the compounds of the invention with acids.
- Suitable acids for forming salts include, but are not limited to, mineral acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid; formic acid, acetic acid, trifluoroacetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, Organic acids such as fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid, benzoic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, benzenesulfonic acid, naphthalenesulfonic acid; Amino acids such as amino acid, phenylalanine, aspartic acid, and glutamic acid.
- salts of the compounds of the invention with bases such as alkali metal salts (for example sodium or potassium salts), alkaline earth metal salts (for example magnesium or calcium salts), ammonium salts (for example lower alkanolammonium).
- bases such as alkali metal salts (for example sodium or potassium salts), alkaline earth metal salts (for example magnesium or calcium salts), ammonium salts (for example lower alkanolammonium).
- Salts and other pharmaceutically acceptable amine salts such as methylamine, ethylamine, propylamine, dimethylamine, trimethylamine, diethylamine, triethylamine, tert-butyl
- a base amine salt an ethylenediamine salt, a hydroxyethylamine salt, a dihydroxyethylamine salt, a trihydroxyethylamine salt, and an amine salt formed of morpholine, piperazine, and lysine, respectively.
- solvate refers to a complex of a compound of the invention that is coordinated to a solvent molecule to form a specific ratio.
- Hydrophilate means a complex formed by the coordination of a compound of the invention with water.
- the beneficial effects of the present invention compared to the prior art are: First, the compound of the present invention has excellent inhibitory effect on the hepatitis C virus protein NS3/4A. Second, by deuteration this technique changes the metabolism of the compound in the organism, giving the compound better pharmacokinetic parameter characteristics. In this case, the dosage can be changed and a long-acting preparation can be formed to improve the applicability. Third, replacing the hydrogen atom in the compound with hydrazine increases the drug concentration of the compound in the animal due to its strontium isotope effect, thereby improving the drug efficacy. Fourth, replacing the hydrogen atom in the compound with hydrazine can inhibit certain metabolites and improve the safety of the compound.
- the preparation of the structural compound of the formula (I) of the present invention is more specifically described below, but these specific methods do not constitute any limitation to the present invention.
- the compounds of the invention may also optionally combine various synthetic methods described in the specification or known in the art. It is convenient to make such a combination that can be easily carried out by those skilled in the art to which the present invention pertains.
- each reaction is usually carried out in an inert solvent at room temperature to reflux temperature (e.g., 0 ° C to 100 ° C, preferably 0 ° C to 80 ° C).
- the reaction time is usually from 0.1 to 60 hours, preferably from 0.5 to 24 hours.
- the inventors used the HCV Replicon System as an evaluation model. Since its first report in Science in 1999, the HCV replication system has become one of the most important tools for studying HCV RNA replication, pathogenicity and viral persistence, for example, the use of replicons has successfully demonstrated the 5' required for HCV RNA replication. - NCR minimum region, and the HCV replication subsystem has been successfully used as an evaluation model for antiviral drugs. The inventors of the present invention verified according to the methods described in Science, 1999, 285 (5424), 110-3, and J. Virol, 2003, 77(5), 3007-19.
- the inhibitory activities of the recombinant hepatitis C virus genotype 1a and 1b replicons were detected by stable transfection of replicon cells with HCV-1a and HCV-1b. This experiment will use the NS3/4A inhibitor Simeprevir as a positive control compound.
- Step 1 The compound was diluted 1:3 in 8 series points, double-replicated, and added to a 96-well plate.
- the DMSO was set to no compound control.
- the final concentration of DMSO in the cell culture was 0.5%.
- Step 2 HCV-1a and 1b cells were separately suspended in a culture medium containing 10% FBS, and seeded into a 96-well plate containing the compound at a density of 8,000 cells per well. The cells were cultured for 3 days at 5% CO 2 at 37 °C.
- Step 3 The cytotoxicity of the compound against GT1b replicon was determined using CellTiter-Fluor (Promega).
- Step 4 Detection of luciferase assay by Bright-Glo (Promega) for anti-hepatitis C virus activity.
- Step Five using GraphPad Prism data analysis software, the curve fitting and EC 50 values were calculated and the 50 value CC.
- the experimental results show that the compounds of the present invention can inhibit the multiple genotypes of HCV and can be used for the inhibition of hepatitis C virus.
- the compound of the present invention exhibited more excellent inhibitory activity against the GT1a and GT1b replicons than the non-deuterated compound Simeprevir.
- the experimental results indicate that the compound of the present invention can be used as an inhibitor of HCV NS3/4A protease, and can be used in a drug against hepatitis C virus.
- Microsomal experiments human liver microsomes: 0.5 mg/mL, Xenotech; rat liver microsomes: 0.5 mg/mL, Xenotech; coenzyme (NADPH/NADH): 1 mM, Sigma Life Science; magnesium chloride: 5 mM, 100 mM phosphate Buffer (pH 7.4).
- phosphate buffer 100 mM, pH 7.4.
- the pH of the solution was adjusted to 7.4, diluted 5 times with ultrapure water before use, and magnesium chloride was added to obtain a phosphate buffer (100 mM) containing 100 mM potassium phosphate, 3.3 mM magnesium chloride, and a pH of 7.4.
- NADPH regeneration system containing 6.5 mM NADP, 16.5 mM G-6-P, 3 U/mL G-6-P D, 3.3 mM magnesium chloride was prepared and placed on wet ice before use.
- Formulation stop solution acetonitrile solution containing 50 ng/mL propranolol hydrochloride and 200 ng/mL tolbutamide (internal standard). Take 25057.5 ⁇ L of phosphate buffer (pH 7.4) into a 50 mL centrifuge tube, add 812.5 ⁇ L of human liver microsomes, and mix to obtain a liver microsome dilution with a protein concentration of 0.625 mg/mL. Take 25057.5 ⁇ L of phosphate buffer (pH 7.4) into a 50 mL centrifuge tube, add 812.5 ⁇ L SD rat liver microsomes, and mix to obtain a liver microsome dilution with a protein concentration of 0.625 mg/mL.
- the corresponding compound had a reaction concentration of 1 ⁇ M and a protein concentration of 0.5 mg/mL.
- 100 ⁇ L of the reaction solution was taken at 10, 30, and 90 min, respectively, and added to the stopper plate, and the reaction was terminated by vortexing for 3 min.
- the plate was centrifuged at 5000 x g for 10 min at 4 °C.
- 100 ⁇ L of the supernatant was taken into a 96-well plate to which 100 ⁇ L of distilled water was previously added, mixed, and sample analysis was performed by LC-MS/MS.
- the metabolic stability of human and rat liver microsomes was evaluated by simultaneously testing the compounds of the present invention and their compounds without deuteration.
- the half-life and liver intrinsic clearance as indicators of metabolic stability are shown in Table 2.
- the undeuterated compound Simeprevir was used as a control sample in Table 2.
- the compound of the present invention can significantly improve metabolic stability by comparison with the undeuterated compound Simeprevir, and is thus more suitable as a hepatitis C virus inhibitor.
- the compounds of the examples were analyzed according to the above procedure, and the results of the experiments showed that the compounds of the present invention exhibited excellent metabolic stability in both human liver microsomes and rat liver microsomes.
- EXPERIMENTAL OBJECTIVE To investigate the pharmacokinetic behavior of the compounds of the present invention after administration of Simeprevir to the compounds of the examples.
- SD rat grade SPF grade
- Weight range 180 ⁇ 220g (actual weight range is 187 ⁇ 197g)
- the mouse eye vein was taken from a blood of about 100-200 L, placed in an EDTA-K2 anticoagulated 0.5 mL Eppendorf tube, and immediately mixed. After anticoagulation, the tube was gently inverted and mixed 5-6 times as soon as possible.
- Plasma concentrations in plasma at each time point were determined after sample collection at all time points.
- the experimental results show that compared with Simeprevir, the present inventors have found that the compound has superior activity than Simeprevir and has excellent pharmacokinetic properties, and thus is more suitable as a compound for inhibiting the hepatitis C virus protein NS3/4A, and is suitable for the compound. Preparation of a medicament for treating hepatitis C virus infection.
Landscapes
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Pharmacology & Pharmacy (AREA)
- Virology (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- General Health & Medical Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Medicinal Chemistry (AREA)
- Molecular Biology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Oncology (AREA)
- Communicable Diseases (AREA)
- Epidemiology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Description
本发明属于医药技术领域,尤其涉及一种丙型肝炎病毒抑制剂、药物组合物及其应用。The invention belongs to the technical field of medicine, and in particular relates to a hepatitis C virus inhibitor, a pharmaceutical composition and application thereof.
丙型肝炎是一种血源性传染性肝脏疾病,若不及时治疗,可能对肝脏造成重大损害。全世界约有1.85亿人感染丙肝病毒,中国丙肝感染率约为3.2%,患者数约四千万;在美国,约有320万丙型肝炎患者,每年约有1.5万人死于该病,大多死于丙型肝炎相关疾病,如肝硬化和肝癌。Hepatitis C is a blood-borne infectious liver disease that, if left untreated, can cause significant damage to the liver. About 185 million people worldwide are infected with hepatitis C virus. The rate of hepatitis C infection in China is about 3.2%, and the number of patients is about 40 million. In the United States, there are about 3.2 million hepatitis C patients, and about 15,000 people die each year. Mostly die from hepatitis C-related diseases such as cirrhosis and liver cancer.
HCV(Hepatitis C Virus,丙型肝炎病毒)是一种RNA病毒,其属于黄病毒科(Flaviviridae family)中的丙型肝炎病毒属(Hepacivirus genus)。包裹HCV病毒粒子包含正股RNA基因组,其在单个不间断的开放读码框中编码全部已知的病毒—特异的蛋白质。开放读码框包括大约9500个核苷酸并且编码单个约3000个氨基酸的巨大多蛋白。多蛋白包括芯蛋白,包裹蛋白E1和E2,膜结合蛋白P7,和非结构性蛋白NS2、NS3/4A、NS4A、NS4B、NS5A和NS5B。HCV (Hepatitis C Virus) is an RNA virus belonging to the genus Hepacivirus genus in the Flaviviridae family. The encapsulated HCV virion contains a positive-stranded RNA genome that encodes all known virus-specific proteins in a single uninterrupted open reading frame. The open reading frame comprises approximately 9500 nucleotides and encodes a single large polyprotein of approximately 3000 amino acids. Polyproteins include core proteins, envelope proteins E1 and E2, membrane-bound protein P7, and non-structural proteins NS2, NS3/4A, NS4A, NS4B, NS5A, and NS5B.
索非布韦是目前世界上第一个在短期内可以彻底治愈丙肝的良药。它以口服途径直达病灶,方法简单副作用很小,深受患者的追捧。索非布韦由美国吉利德公司生产,2013年在美国上市,经临床试验证实可有效治疗基因1、2、3、4型丙肝,包括对肝移植、肝癌以及HCV/HIV-1合并感染的临床试验。这一突破为全世界的丙肝患者带来了福音。Sofabru is the first medicine in the world that can completely cure hepatitis C in the short term. It takes the oral route directly to the lesion, and the method has simple side effects and is highly sought after by patients. Sofibuvir is produced by Gilead, USA, and is marketed in the United States in 2013. It has been clinically proven to be effective in treating hepatitis C, type 1, 2, 3, and 4, including liver transplantation, liver cancer, and HCV/HIV-1 co-infection. Clinical Trials. This breakthrough has brought the gospel to hepatitis C patients around the world.
HCV感染与进行性肝病状(包括肝硬化和肝细胞癌)有关。西美瑞韦(Simeprevir)是新一代NS3/4A蛋白酶抑制剂,为每日一次的口服药物,由Medivir公司和杨森(Janssen)联合开发,用于治疗慢性丙型肝炎成年患者的代偿性肝病,包括各个阶段的肝纤维化,其工作原理是通过阻断蛋白酶,来抑制HCV在肝脏细胞中的复制。2013年1月22日,丙肝新药西美瑞韦获FDA批准,联合聚乙二醇干扰素和利巴韦林(ribavirin),用于基因型1慢性丙型肝炎成人患者代偿性肝脏疾病(包括肝硬化)的治疗。HCV infection is associated with progressive liver disease, including cirrhosis and hepatocellular carcinoma. Simeprevir is a new generation of NS3/4A protease inhibitors, a daily oral medication developed by Medivir and Janssen for the treatment of compensatory liver disease in adult patients with chronic hepatitis C. Including liver fibrosis at various stages, its working principle is to inhibit the replication of HCV in liver cells by blocking proteases. On January 22, 2013, hepatitis C, a new drug, dexamethavir was approved by the FDA, combined with peginterferon and ribavirin for genotype 1 chronic hepatitis C adult patients with compensatory liver disease ( Treatment including cirrhosis.
因此,本领域仍需要开发对丙型肝炎病毒蛋白NS3/4A有抑制活性或更好药效学性能的化合物。Therefore, there is still a need in the art to develop compounds having inhibitory activity or better pharmacodynamic properties against the hepatitis C virus protein NS3/4A.
发明内容Summary of the invention
针对以上技术问题,本发明公开了一种丙型肝炎病毒抑制剂、药物组合物及其应用,其具有更好的丙肝病毒蛋白NS3/4A抑制活性和/或具有更好药效学/药代动力学性能。In view of the above technical problems, the present invention discloses a hepatitis C virus inhibitor, a pharmaceutical composition and use thereof, which have better hepatitis C virus protein NS3/4A inhibitory activity and/or have better pharmacodynamics/pharmacokinetics. Kinetic performance.
对此,本发明采用的技术方案为:In this regard, the technical solution adopted by the present invention is:
一种丙型肝炎病毒抑制剂,如式(I)所示的喹啉化合物,或其晶型、药学上可接受的盐、水合物或溶剂化合物, A hepatitis C virus inhibitor, such as a quinoline compound of the formula (I), or a crystalline form thereof, a pharmaceutically acceptable salt, a hydrate or a solvent compound,
其中,R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15、R16、R17、R18、R19、R20、R21、R22、R23、R24、R25、R26、R27、R28、R29、R30、R31、R32、R33、R33、R34、R35、R36、R37、R38、R39、R40、R41、R42、R43、R44、R45各自独立地为氢、氘、卤素或三氟甲基;Wherein R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 18 , R 19 , R 20 , R 21 , R 22 , R 23 , R 24 , R 25 , R 26 , R 27 , R 28 , R 29 , R 30 , R 31 , R 32 , R 33 And R 33 , R 34 , R 35 , R 36 , R 37 , R 38 , R 39 , R 40 , R 41 , R 42 , R 43 , R 44 , R 45 are each independently hydrogen, deuterium, halogen or trisole Fluoromethyl;
附加条件是R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15、R16、R17、R18、R19、R20、R21、R22、R23、R24、R25、R26、R27、R28、R29、R30、R31、R32、R33、R33、R34、R35、R36、R37、R38、R39、R40、R41、R42、R43、R44和R45中至少一个是氘代的。Additional conditions are R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 , R 16 And R 17 , R 18 , R 19 , R 20 , R 21 , R 22 , R 23 , R 24 , R 25 , R 26 , R 27 , R 28 , R 29 , R 30 , R 31 , R 32 , R 33. At least one of R 33 , R 34 , R 35 , R 36 , R 37 , R 38 , R 39 , R 40 , R 41 , R 42 , R 43 , R 44 and R 45 is deuterated.
采用此技术方案,氘在药物分子中的形状和体积与氢基本上相同,如果药物分子中氢被选择性替换为氘,氘代药物一般还会保留原来的生物活性和选择性。同时发明人经过实验证实,碳氘键的结合比碳氢键的结合更稳定,可直接影响一些药物的吸收、分布、代谢和排泄等属性,从而提高药物的疗效、安全性和耐受性。With this technical solution, the shape and volume of the ruthenium in the drug molecule are substantially the same as those of the hydrogen. If the hydrogen in the drug molecule is selectively replaced with hydrazine, the deuterated drug generally retains the original biological activity and selectivity. At the same time, the inventors have confirmed through experiments that the binding of carbon-germanium bonds is more stable than the combination of carbon-hydrogen bonds, which can directly affect the absorption, distribution, metabolism and excretion of some drugs, thereby improving the efficacy, safety and tolerability of the drugs.
优选的,氘在氘代位置的氘同位素含量至少是大于天然氘同位素含量(0.015%),较佳地大于30%,更佳地大于50%,更佳地大于75%,更佳地大于95%,更佳地大于99%。Preferably, the strontium isotope content of the cerium in the deuterated position is at least greater than the natural strontium isotope content (0.015%), preferably greater than 30%, more preferably greater than 50%, more preferably greater than 75%, and even more preferably greater than 95. %, more preferably greater than 99%.
具体地说,在本发明中R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15、R16、R17、R18、R19、R20、R21、R22、R23、R24、R25、R26、R27、R28、R29、R30、R31、R32、R33、R33、R34、R35、R36、R37、R38、R39、R40、R41、R42、R43、R44和R45各氘代位置中氘同位素含量至少是5%,较佳地大于10%,更佳地大于15%,更佳地大于20%,更佳地大于25%,更佳地大于30%,更佳地大于35%,更佳地大于40%,更佳地大于45%,更佳地大于50%,更佳地大于55%,更佳地大于60%,更佳地大于65%,更佳地大于70%,更佳地大于75%,更佳地大于80%,更佳地大 于85%,更佳地大于90%,更佳地大于95%,更佳地大于99%。Specifically, in the present invention, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 and R 14 , R 15 , R 16 , R 17 , R 18 , R 19 , R 20 , R 21 , R 22 , R 23 , R 24 , R 25 , R 26 , R 27 , R 28 , R 29 , R 30 , R 31 And R 32 , R 33 , R 33 , R 34 , R 35 , R 36 , R 37 , R 38 , R 39 , R 40 , R 41 , R 42 , R 43 , R 44 and R 45 are each in the metamorphic position The strontium isotope content is at least 5%, preferably more than 10%, more preferably more than 15%, more preferably more than 20%, more preferably more than 25%, more preferably more than 30%, more preferably more than 35%, More preferably more than 40%, more preferably more than 45%, more preferably more than 50%, more preferably more than 55%, more preferably more than 60%, more preferably more than 65%, more preferably more than 70%, more Preferably, it is greater than 75%, more preferably greater than 80%, more preferably greater than 85%, more preferably greater than 90%, more preferably greater than 95%, and even more preferably greater than 99%.
优选的,式(I)中化合物的R1、R2、R3、R4、R5、R6、R7、R8、R9、R10、R11、R12、R13、R14、R15、R16、R17、R18、R19、R20、R21、R22、R23、R24、R25、R26、R27、R28、R29、R30、R31、R32、R33、R33、R34、R35、R36、R37、R38、R39、R40、R41、R42、R43、R44和R45,至少其中一个R含氘,更佳地两个R含氘,更佳地三个R含氘,更佳地四个R含氘,更佳地五个R含氘,更佳地六个R含氘,更佳地七个R含氘,更佳地八个R含氘,更佳地九个R含氘,更佳地十个R含氘,更佳地十一个R含氘,更佳地十二个R含氘,更佳地十三个R含氘,更佳地十四个R含氘,更佳地十五个R含氘,更佳地十六个R含氘,更佳地十七个R含氘,更佳地十八个R含氘,更佳地十九个R含氘,更佳地二十个R含氘,更佳地二十一个R含氘,更佳地二十二个R含氘,更佳地二十三个R含氘,更佳地二十四个R含氘,更佳地二十五个R含氘,更佳地二十六个R含氘,更佳地二十七个R含氘,更佳地二十八个R含氘,更佳地二十九个R含氘,更佳地三十个R含氘,更佳地三十一个R含氘,更佳地三十二个R含氘,更佳地三十三个R含氘,更佳地三十四个R含氘,更佳地三十五个R含氘,更佳地三十六个R含氘,更佳地三十七个R含氘,更佳地三十八个R含氘,更佳地三十九个R含氘,更佳地四十个R含氘,更佳地四十一个R含氘,更佳地四十二个R含氘,更佳地四十三个R含氘,更佳地四十四个R含氘,更佳地四十五个R含氘。Preferably, R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R of the compound of formula (I) 14 , R 15 , R 16 , R 17 , R 18 , R 19 , R 20 , R 21 , R 22 , R 23 , R 24 , R 25 , R 26 , R 27 , R 28 , R 29 , R 30 , R 31 , R 32 , R 33 , R 33 , R 34 , R 35 , R 36 , R 37 , R 38 , R 39 , R 40 , R 41 , R 42 , R 43 , R 44 and R 45 , at least One R contains ruthenium, more preferably two R contains ruthenium, more preferably three R contains ruthenium, more preferably four R contains ruthenium, more preferably five R contains ruthenium, more preferably six R contains ruthenium, more preferably six R-containing ruthenium, More preferably, seven R contains ruthenium, more preferably eight R contains ruthenium, more preferably nine R contains ruthenium, more preferably ten R contains ruthenium, more preferably eleven R contains ruthenium, more preferably ten The two Rs contain ruthenium, more preferably thirteen R 氘, more preferably fourteen R 氘, more preferably fifteen R 氘, more preferably sixteen R 氘, more preferably ten The seven Rs contain ruthenium, more preferably eighteen R 氘, more preferably nineteen R 氘, more preferably twenty R 氘, more preferably twenty-one R 氘, more preferably Twenty-two R contains 氘, better two The three Rs contain 氘, preferably twenty-four R 氘, more preferably twenty-five R 氘, more preferably twenty-six R 氘, more preferably twenty-seven R 氘, More preferably twenty-eight R 氘, more preferably twenty-nine R 氘, more preferably thirty R 氘, more preferably thirty-one R 氘, more preferably thirty-two R contains 氘, more preferably thirty-three R 氘, more preferably thirty-four R 氘, more preferably thirty-five R 氘, more preferably thirty-six R 氘, better Thirty-seven R contains 氘, more preferably thirty-eight R contains 氘, more preferably thirty-nine R contains 氘, more preferably forty R contains 氘, more preferably forty-one R Preferably, forty-two R 氘, more preferably forty-three R 氘, more preferably forty four R 氘, more preferably forty-five R 氘.
作为本发明的进一步改进,R19、R20和R21各自独立地为氘或氢。As a further improvement of the present invention, R 19 , R 20 and R 21 are each independently hydrazine or hydrogen.
在另一优选例中,R19、R20、R21是氘。In another preferred embodiment, R 19 , R 20 and R 21 are 氘.
作为本发明的进一步改进,R32、R33和R34各自独立地为氘或氢。As a further improvement of the present invention, R 32 , R 33 and R 34 are each independently hydrazine or hydrogen.
在另一优选例中,R32、R33、R34是氘。In another preferred embodiment, R 32 , R 33 , and R 34 are deuterium.
作为本发明的进一步改进,R35、R36和R37各自独立地为氘或氢。As a further improvement of the present invention, R 35 , R 36 and R 37 are each independently hydrazine or hydrogen.
在另一优选例中,R35、R36、R37是氘。In another preferred embodiment, R 35 , R 36 , and R 37 are 氘.
作为本发明的进一步改进,所述化合物选自下述化合物或其药学上可接受的盐:As a further improvement of the present invention, the compound is selected from the group consisting of the following compounds or a pharmaceutically acceptable salt thereof:
在另一优选例中,所述化合物不包括非氘代化合物。In another preferred embodiment, the compound does not include a non-deuterated compound.
本发明还公开了一种药物组合物,其含有药学上可接受的载体和如上所述的所述的丙型肝炎病毒抑制剂,或其晶型、药学上可接受的盐、水合物或溶剂合物、立体异构体、前药或同位素变体的药物组合物。The present invention also discloses a pharmaceutical composition comprising a pharmaceutically acceptable carrier and the hepatitis C virus inhibitor as described above, or a crystalline form, a pharmaceutically acceptable salt, a hydrate or a solvent thereof A pharmaceutical composition of a compound, stereoisomer, prodrug or isotopic variation.
作为本发明的进一步改进,其还包含其他活性化合物,所述活性化合物包括但不限于,其它HCV抗病毒剂、抗感染药、免疫调节剂、抗生素或疫苗结合。As a further improvement of the present invention, it further comprises other active compounds including, but not limited to, other HCV antiviral agents, anti-infective agents, immunomodulators, antibiotics or vaccine combinations.
作为本发明的进一步改进,所述免疫调节剂为干扰素类药物化合物。As a further improvement of the present invention, the immunomodulator is an interferon drug compound.
作为本发明的进一步改进,本发明所述的喹喔啉大环化合物可用在涉及一种或多种附加治疗剂的联合治疗中。附加治疗剂包括也靶向HCV、靶向不同致病剂的那些或增强免疫系统的那些。增强免疫系统的药剂包括通常增强免疫系统功能的那些和针对HCV产生特异性免疫响应的那些。靶向HCV的附加治疗剂包括靶向NS3/4A的药剂和靶向其它HCV活性,如NS5A和NS5B的药剂,和靶向HCV复制中涉及的宿主细胞活性的药剂。As a further improvement of the invention, the quinoxaline macrocycles of the invention may be used in combination therapies involving one or more additional therapeutic agents. Additional therapeutic agents include those that also target HCV, target different pathogenic agents, or enhance the immune system. Agents that enhance the immune system include those that normally enhance the function of the immune system and those that produce a specific immune response against HCV. Additional therapeutic agents that target HCV include agents that target NS3/4A and agents that target other HCV activities, such as NS5A and NS5B, and agents that target host cell activity involved in HCV replication.
组合中可能存在的治疗剂的其它实例包括利巴韦林、levovirin、viramidine、胸腺素α-1、干扰素-β、干扰素-α、PEG化的干扰素-α(peg干扰素-α)、干扰素-α和利巴韦林的组合、peg干扰素-α和利巴韦林的组合、干扰素-α和levovirin的组合以及peg干扰素-α和levovirin的组合。干扰素-α包括重组干扰素-α2a(如可获自Hoffmann-LaRoche,Nutley,NJ的Roferon干扰素)、PEG化的干扰素-α2a(Pegasys)、干扰素-α2b(如可获自Schering Corp.,Kenilworth,NJ的Intron-A干扰素)、PEG化的干扰素-α2b(PegIntron)、重组复合干扰素(如干扰素alphacon-1)和纯化的干扰素–α产品。该组合的各个组分可以在治疗过程的不同时间分别给药或以分开的或单个的组合形式同时给药。Other examples of therapeutic agents that may be present in the combination include ribavirin, levovirin, viramidine, thymosin alpha-1, interferon-beta, interferon-alpha, PEGylated interferon-alpha (peg interferon-alpha) a combination of interferon-[alpha] and ribavirin, a combination of peg interferon-[alpha] and ribavirin, a combination of interferon-[alpha] and levovirin, and a combination of peg interferon-[alpha] and levovirin. Interferon-α includes recombinant interferon-α2a (such as Roferon interferon available from Hoffmann-LaRoche, Nutley, NJ), PEGylated interferon-α2a (Pegasys), interferon-α2b (eg available from Schering Corp) .Kenilworth, NJ's Intron-A interferon), PEGylated interferon-α2b (PegIntron), recombinant complex interferon (such as interferon alphacon-1), and purified interferon-α product. The individual components of the combination may be administered separately at different times during the course of the treatment or simultaneously in separate or individual combinations.
为了治疗HCV感染,本发明的化合物还可以与抗病毒剂金刚胺(1-氨基金刚烷)联合给药。关于该药剂的全面描述,参见J.Kirschbaum,12 Anal.Profiles Drug Subs.1-36(1983)。 For the treatment of HCV infection, the compounds of the invention may also be administered in combination with the antiviral agent amantadine (1-aminoadamantane). For a full description of the agent, see J. Kirschbaum, 12 Anal. Profiles Drug Subs. 1-36 (1983).
为了治疗HCV感染,本发明的化合物还可以与抗病毒剂聚合酶抑制剂R7128(Roche)联合给药。For the treatment of HCV infection, the compounds of the invention may also be administered in combination with the antiviral polymerase inhibitor R7128 (Roche).
为了治疗HCV感染,本发明的化合物还可以与HCV NS5B聚合酶抑制剂联合给药。可用作联合治疗的此类HCV NS5B聚合酶抑制剂包括,但不限于,国际专利申请公开WO 02/057287、WO 02/057425、WO 03/068244、WO 2004/000858、WO 04/003138和WO 2004/007512;美国专利No.6,777,392和美国专利申请公开US2004/0067901(它们各自的内容全部经此引用并入本文)中公开的那些。其它这样的HCV聚合酶抑制剂包括,但不限于,valopicitabine(NM-283;Idenix)和2’-F-2’-β-甲基胞苷(也参见WO 2005/003147)。For the treatment of HCV infection, the compounds of the invention may also be administered in combination with an HCV NS5B polymerase inhibitor. Such HCV NS5B polymerase inhibitors that can be used as a combination therapy include, but are not limited to, International Patent Application Publication No. WO 02/057287, WO 02/057425, WO 03/068244, WO 2004/000858, WO 04/003138, and WO Those disclosed in U.S. Patent No. 6,777,392 and U.S. Patent Application Publication No. 2004/0067901, the entire contents of each of each of Other such HCV polymerase inhibitors include, but are not limited to, valopicitabine (NM-283; Idenix) and 2'-F-2'-beta-methylcytidine (see also WO 2005/003147).
作为本发明的进一步改进,所述药学上可接受的载体包括助流剂、增甜剂、稀释剂、防腐剂、染料/着色剂、矫味增强剂、表面活性剂、润湿剂、分散剂、崩解剂、助悬剂、稳定剂、等渗剂、溶剂或乳化剂中的至少一种。As a further improvement of the present invention, the pharmaceutically acceptable carrier includes a glidant, a sweetener, a diluent, a preservative, a dye/colorant, a flavor enhancer, a surfactant, a wetting agent, a dispersant At least one of a disintegrant, a suspending agent, a stabilizer, an isotonic agent, a solvent or an emulsifier.
作为本发明的进一步改进,所述药物组合物为片剂、丸剂、胶囊剂、粉剂、颗粒剂、膏剂、乳剂、悬浮剂、溶液剂、栓剂、注射剂、吸入剂、凝胶剂、微球或气溶胶。As a further improvement of the present invention, the pharmaceutical composition is a tablet, a pill, a capsule, a powder, a granule, an ointment, an emulsion, a suspension, a solution, a suppository, an injection, an inhalant, a gel, a microsphere or Aerosol.
给予本发明药物组合物的典型途径包括但不限于口服、直肠、透黏膜、经肠给药,或者局部、经皮、吸入、肠胃外、舌下、阴道内、鼻内、眼内、腹膜内、肌内、皮下、静脉内给药。优选口服给药或注射给药。Typical routes of administration of the pharmaceutical compositions of the invention include, but are not limited to, oral, rectal, transmucosal, enteral, or topical, transdermal, inhalation, parenteral, sublingual, intravaginal, intranasal, intraocular, intraperitoneal , intramuscular, subcutaneous, intravenous administration. Oral administration or injection administration is preferred.
本发明的药物组合物可以采用本领域周知的方法制造,如常规的混合法、溶解法、制粒法、制糖衣药丸法、磨细法、乳化法、冷冻干燥法等。The pharmaceutical composition of the present invention can be produced by a method known in the art, such as a conventional mixing method, a dissolution method, a granulation method, a sugar-coating method, a pulverization method, an emulsification method, a freeze-drying method, and the like.
本发明还提供了一种制备药物组合物的方法,包括步骤:将药学上可接受的载体与如上所述的丙型肝炎病毒抑制剂,或其晶型、药学上可接受的盐、水合物或溶剂合物进行混合,形成药物组合物。The present invention also provides a method of preparing a pharmaceutical composition comprising the steps of: administering a pharmaceutically acceptable carrier to a hepatitis C virus inhibitor as described above, or a crystalline form thereof, a pharmaceutically acceptable salt, or a hydrate thereof Or the solvate is mixed to form a pharmaceutical composition.
本发明还公开了一种如上所述的丙型肝炎病毒抑制剂的用途,其特征在于:用于制备治疗丙型肝炎病毒感染的药物中的用途。The invention also discloses the use of a hepatitis C virus inhibitor as described above, characterized in that it is used for the preparation of a medicament for the treatment of hepatitis C virus infection.
NS3/4A抑制剂还可用于抗病毒化合物的筛选化验的准备和实施。例如,此类化合物可用于分离酶突变体,它们是更有力的抗病毒化合物的优异筛选工具。此外,这些化合物可用于建立或测定其它抗病毒剂与HCV蛋白酶的结合位点,例如通过竞争性抑制。NS3/4A inhibitors are also useful in the preparation and implementation of screening assays for antiviral compounds. For example, such compounds can be used to isolate enzyme mutants, which are excellent screening tools for more potent antiviral compounds. In addition, these compounds can be used to establish or measure binding sites for other antiviral agents to HCV protease, for example by competitive inhibition.
为了抑制HCV NS3/4A蛋白酶和治疗HCV感染和/或降低HCV感染症状的可能性或严重性,任选为盐形式的本发明的化合物可通过使活性剂与药物的作用位点接触的方式给药。它们可作为独立治疗剂或治疗剂的组合通过可与药物联合使用的常规方式给药。它们可独自给药,但通 常与根据所选给药途径和标准药物实践选择的药物载体一起给药。In order to inhibit the HCV NS3/4A protease and to treat HCV infection and/or reduce the likelihood or severity of HCV infection symptoms, the compounds of the invention, optionally in salt form, can be administered by contacting the active agent with the site of action of the drug. medicine. They can be administered as a separate therapeutic or combination of therapeutic agents by conventional means which can be used in combination with the drug. They can be administered alone, but they are It is often administered with a pharmaceutical carrier selected according to the chosen route of administration and standard pharmaceutical practice.
所述的HCV包括其多种基因型以及多种基因亚型,例如1a、1b、2a、2b、3a、3b、4a、5a、6a。The HCV includes a plurality of genotypes thereof and a plurality of gene subtypes, such as 1a, 1b, 2a, 2b, 3a, 3b, 4a, 5a, 6a.
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅,在此不再一一累述。It is to be understood that within the scope of the present invention, the various technical features of the present invention and the various technical features specifically described hereinafter (as in the embodiments) may be combined with each other to constitute a new or preferred technical solution. Due to space limitations, we will not repeat them here.
本文中,如无特别说明,“卤素”指F、Cl、Br、和I。更佳地,卤原子选自F、Cl和Br。Herein, "halogen" means F, Cl, Br, and I unless otherwise specified. More preferably, the halogen atom is selected from the group consisting of F, Cl and Br.
本文中,如无特别说明,“氘代”指化合物或基团中的一个或多个氢被氘所取代;氘代可以是一取代、二取代、多取代或全取代。术语“一个或多个氘代的”与“一次或多次氘代”可互换使用。As used herein, unless otherwise specified, "deuterated" means that one or more hydrogens in the compound or group are replaced by deuterium; deuteration may be monosubstituted, disubstituted, polysubstituted or fully substituted. The terms "one or more deuterated" are used interchangeably with "one or more deuterated".
本文中,如无特别说明,“非氘代的化合物”是指含氘原子比例不高于天然氘同位素含量(0.015%)的化合物。As used herein, unless otherwise specified, "non-deuterated compound" means a compound containing a proportion of germanium atoms not higher than the natural helium isotope content (0.015%).
药学上可接受的盐包括无机盐和有机盐。一类优选的盐是本发明化合物与酸形成的盐。适合形成盐的酸包括但并不限于:盐酸、氢溴酸、氢氟酸、硫酸、硝酸、磷酸等无机酸;甲酸、乙酸、三氟乙酸、丙酸、草酸、丙二酸、琥珀酸、富马酸、马来酸、乳酸、苹果酸、酒石酸、柠檬酸、苦味酸、苯甲酸、甲磺酸、乙磺酸、对甲苯磺酸、苯磺酸、萘磺酸等有机酸;以及脯氨酸、苯丙氨酸、天冬氨酸、谷氨酸等氨基酸。另一类优选的盐是本发明化合物与碱形成的盐,例如碱金属盐(例如钠盐或钾盐)、碱土金属盐(例如镁盐或钙盐)、铵盐(如低级的烷醇铵盐以及其它药学上可接受的胺盐),例如甲胺盐、乙胺盐、丙胺盐、二甲基胺盐、三甲基胺盐、二乙基胺盐、三乙基胺盐、叔丁基胺盐、乙二胺盐、羟乙胺盐、二羟乙胺盐、三羟乙胺盐,以及分别由吗啉、哌嗪、赖氨酸形成的胺盐。Pharmaceutically acceptable salts include inorganic and organic salts. A preferred class of salts are the salts of the compounds of the invention with acids. Suitable acids for forming salts include, but are not limited to, mineral acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid; formic acid, acetic acid, trifluoroacetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, Organic acids such as fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid, benzoic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, benzenesulfonic acid, naphthalenesulfonic acid; Amino acids such as amino acid, phenylalanine, aspartic acid, and glutamic acid. Another preferred class of salts are the salts of the compounds of the invention with bases, such as alkali metal salts (for example sodium or potassium salts), alkaline earth metal salts (for example magnesium or calcium salts), ammonium salts (for example lower alkanolammonium). Salts and other pharmaceutically acceptable amine salts), such as methylamine, ethylamine, propylamine, dimethylamine, trimethylamine, diethylamine, triethylamine, tert-butyl A base amine salt, an ethylenediamine salt, a hydroxyethylamine salt, a dihydroxyethylamine salt, a trihydroxyethylamine salt, and an amine salt formed of morpholine, piperazine, and lysine, respectively.
术语“溶剂合物”指本发明化合物与溶剂分子配位形成特定比例的配合物。“水合物”是指本发明化合物与水进行配位形成的配合物。The term "solvate" refers to a complex of a compound of the invention that is coordinated to a solvent molecule to form a specific ratio. "Hydrate" means a complex formed by the coordination of a compound of the invention with water.
与现有技术相比,本发明的有益效果为:第一,本发明的化合物对丙型肝炎病毒蛋白NS3/4A具有优异的抑制性。第二,通过氘化这一技术改变化合物在生物体中的代谢,使化合物具有更好的药代动力学参数特性。在这种情况下,可以改变剂量并形成长效制剂,改善适用性。第三,用氘取代化合物中的氢原子,由于其氘同位素效应,提高化合物在动物体内的药物浓度,提高了药物疗效。第四,用氘取代化合物中的氢原子,可以抑制某些代谢产物,提高了化合物的安全性。The beneficial effects of the present invention compared to the prior art are: First, the compound of the present invention has excellent inhibitory effect on the hepatitis C virus protein NS3/4A. Second, by deuteration this technique changes the metabolism of the compound in the organism, giving the compound better pharmacokinetic parameter characteristics. In this case, the dosage can be changed and a long-acting preparation can be formed to improve the applicability. Third, replacing the hydrogen atom in the compound with hydrazine increases the drug concentration of the compound in the animal due to its strontium isotope effect, thereby improving the drug efficacy. Fourth, replacing the hydrogen atom in the compound with hydrazine can inhibit certain metabolites and improve the safety of the compound.
下面更具体地描述本发明式(I)结构化合物的制备方法,但这些具体方法不对本发明构成任何限制。本发明化合物还可以任选将在本说明书中描述的或本领域已知的各种合成方法组合起来而 方便地制得,这样的组合可由本发明所属领域的技术人员容易地进行。The preparation of the structural compound of the formula (I) of the present invention is more specifically described below, but these specific methods do not constitute any limitation to the present invention. The compounds of the invention may also optionally combine various synthetic methods described in the specification or known in the art. It is convenient to make such a combination that can be easily carried out by those skilled in the art to which the present invention pertains.
通常,在制备流程中,各反应通常在惰性溶剂中,在室温至回流温度(如0℃~100℃,优选0℃~80℃)下进行。反应时间通常为0.1小时-60小时,较佳地为0.5-24小时。Usually, in the preparation scheme, each reaction is usually carried out in an inert solvent at room temperature to reflux temperature (e.g., 0 ° C to 100 ° C, preferably 0 ° C to 80 ° C). The reaction time is usually from 0.1 to 60 hours, preferably from 0.5 to 24 hours.
实施例1.中间体5的合成。Example 1. Synthesis of Intermediate 5.
步骤1.化合物2的合成。Step 1. Synthesis of Compound 2.
取2.68 g溴化苄溶于100 mL乙腈,氮气保护下加入4.19 g辛可尼丁,避光搅拌过夜。过滤,滤渣用乙腈洗涤三次,并用二氯甲烷和石油醚重结晶,真空干燥得淡红色固体粉末4.22 g。2.68 g of benzyl bromide was dissolved in 100 mL of acetonitrile, and 4.19 g of cinchonidine was added under nitrogen atmosphere, and the mixture was stirred overnight in the dark. Filtration, the residue was washed three times with EtOAc and EtOAc (EtOAc)EtOAc.
步骤2.化合物5的合成。Step 2. Synthesis of Compound 5.
取10 g甘氨酸甲酯盐酸盐和无水硫酸钠分散于200 mL甲基叔丁基醚,先后加入苯甲醛和三乙胺,室温搅拌过夜。过滤,浓缩,所得油状液体直接用于下一步。10 g of glycine methyl ester hydrochloride and anhydrous sodium sulfate were dispersed in 200 mL of methyl tert-butyl ether, and benzaldehyde and triethylamine were added successively, and the mixture was stirred at room temperature overnight. Filter and concentrate and the resulting oily liquid was used directly in the next step.
取435 mg化合物2分散于14 mL甲苯,冰水浴下滴加上述3.98 g油状液体的甲苯(16 mL)溶液,滴完室温反应15分钟。缓慢滴加4.0 g 1,4-二溴-2-丁烯的甲苯(16 mL)溶液,冰水浴下搅拌45分钟。缓慢滴加20%NaOH溶液,滴完后于10℃搅拌过夜。静置分液,有机相过滤,并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得3.14 g淡黄色油状液体。1H NMR(400 MHz,CDCl3)δ5.71(ddd,J=17.1,10.3,9.2 Hz,1H),5.22(ddd,J=17.2,1.9,0.7 Hz,1H),5.05(ddd,J=10.3,1.8,0.6Hz,1H),3.73(s,3H),2.08–1.99(m,1H),1.56(dd,J=7.5,4.8 Hz,1H),1.35(dd,J=9.3,4.7 Hz,1H)。435 mg of the compound 2 was dispersed in 14 mL of toluene, and the above-mentioned 3.98 g of an oily liquid in toluene (16 mL) was added dropwise thereto in an ice water bath, and the mixture was allowed to react at room temperature for 15 minutes. A solution of 4.0 g of 1,4-dibromo-2-butene in toluene (16 mL) was slowly added dropwise and stirred for 45 min. A 20% NaOH solution was slowly added dropwise, and after the completion of the dropwise addition, the mixture was stirred at 10 ° C overnight. The mixture was separated and the organic layer was filtered, washed with brine, dried 1 H NMR (400 MHz, CDCl 3 ) δ 5.71 (ddd, J = 17.1, 10.3, 9.2 Hz, 1H), 5.22 (ddd, J = 17.2, 1.9, 0.7 Hz, 1H), 5.05 (ddd, J = 10.3, 1.8, 0.6 Hz, 1H), 3.73 (s, 3H), 2.08–1.99 (m, 1H), 1.56 (dd, J=7.5, 4.8 Hz, 1H), 1.35 (dd, J=9.3, 4.7 Hz) , 1H).
实施例2.中间体11的合成。Example 2. Synthesis of Intermediate 11.
步骤1.化合物8的合成。 Step 1. Synthesis of Compound 8.
取N-甲基三氟乙酰胺5.0 g溶于28 mL DMF,冰水浴冷却下分批加入60%NaH 1.656 g,室温搅拌1小时,取6-溴-1-己烯6.42 g溶于25 mLDMF,滴加至上述溶液中,滴完后于70℃下室温搅拌过夜。反应液倒入200 mL水中,乙醚萃取,合并乙醚,并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得到黄色油状液体直接用于下一步。Take N-methyltrifluoroacetamide 5.0 g dissolved in 28 mL DMF, add 60% NaH 1.656 g in a cold water bath, and stir at room temperature for 1 hour. Take 6-bromo-1-hexene 6.42 g dissolved in 25 mL DMF. It was added dropwise to the above solution, and after the completion of the dropwise addition, it was stirred at room temperature at 70 ° C overnight. The reaction mixture was poured into water (200 mL), evaporated, evaporated, evaporated, evaporated
上述黄色油状液体用50 mL甲醇溶解,滴加25M KOH溶液66 mL,室温搅拌过夜。乙醚萃取反应液三次,合并乙醚,并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,得到淡黄色油状液体,水泵减压蒸馏,收集50~55℃馏分,得到无色液体3.7 g。1H NMR(400 MHz,CDCl3)δ5.81(ddt,J=16.9,10.2,6.6 Hz,1H),5.08–4.85(m,2H),2.62–2.53(m,2H),2.43(s,3H),2.13–2.01(m,2H),1.66–1.31(m,4H),1.26(br,1H)。The above yellow oily liquid was dissolved in 50 mL of methanol, and 66 mL of a 25 M KOH solution was added dropwise thereto, and stirred at room temperature overnight. The reaction mixture was extracted with EtOAc (EtOAc)EtOAc. 1 H NMR (400 MHz, CDCl 3 ) δ 5.81 (dd, J = 16.9, 10.2, 6.6 Hz, 1H), 5.08 - 4.85 (m, 2H), 2.62 - 2.53 (m, 2H), 2.43 (s, 3H), 2.13–2.01 (m, 2H), 1.66–1.31 (m, 4H), 1.26 (br, 1H).
步骤2.化合物9的合成。Step 2. Synthesis of Compound 9.
取(1R,4R,5R)-3-氧代-2-氧杂二环[2.2.1]己烷-5-羧酸辛可尼丁盐2.27 g和0.71 g化合物8溶于25mL DMF,冰浴冷却下加入2-(7-偶氮苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(HATU,2.30 g),N,N-二异丙基乙胺(DIPEA,0.78 g),冰浴搅拌1小时。反应液倒入100 mL水中,乙酸乙酯萃取,合并乙酸乙酯,并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,柱层析得达黄色油状液体1.04 g即为化合物9。1H NMR(400 MHz,CDCl3)δ6.01–5.58(m,1H),5.17–4.84(m,3H),3.51–3.17(m,2H),2.99(t,J=8.0 Hz,2H),2.93(s,1H),2.30–2.00(m,6H),1.70–1.31(m,4H).LC-MS(APCI):m/z=252.3(M+1)+。Take (1R,4R,5R)-3-oxo-2-oxabicyclo[2.2.1]hexane-5-carboxylic acid cinchonidine salt 2.27 g and 0.71 g of compound 8 dissolved in 25 mL DMF, ice bath 2-(7-Azobenzotriazole)-N,N,N',N'-tetramethyluronium hexafluorophosphate (HATU, 2.30 g), N,N-diisopropyl Ethylamine (DIPEA, 0.78 g) was stirred in an ice bath for 1 hour. The reaction mixture was poured into 100 mL of water, and ethyl acetate was evaporated, and ethyl acetate was evaporated. 1 H NMR (400 MHz, CDCl 3 ) δ 6.01 - 5.58 (m, 1H), 5.17 - 4.84 (m, 3H), 3.51 - 3.17 (m, 2H), 2.99 (t, J = 8.0 Hz, 2H) , 2.93 (s, 1H), 2.30 - 2.00 (m, 6H), 1.70 - 1.31 (m, 4H). LC-MS (APCI): m/z = 252.3 (M + 1) + .
步骤3.化合物10的合成。Step 3. Synthesis of Compound 10.
取1.04 g化合物9溶于8 mL四氢呋喃,冰水浴下滴加1M LiOH溶液4.5 mL,滴完冰水浴下搅拌1小时。4N HCl溶液调pH1~2,乙酸乙酯萃取,合并乙酸乙酯,并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,得淡黄色油状液体0.99 g。LC-MS(APCI):m/z=268.2(M+1)+。1.04 g of the compound 9 was dissolved in 8 mL of tetrahydrofuran, and 4.5 mL of a 1 M LiOH solution was added dropwise thereto in an ice water bath, and the mixture was stirred for 1 hour under ice-cold bath. 4N HCl solution was adjusted to pH 1-2, ethyl acetate was evaporated, and ethyl acetate was evaporated. LC-MS (APCI): m / z = 268.2 (M + 1) +.
步骤4.化合物11的合成。Step 4. Synthesis of Compound 11.
取163 mg化合物5溶于3 mL无水DMF,冰浴下加入253 mg HATU,90 mg化合物10,287μL DIPEA,冰浴下搅拌2小时。反应液倒入20 mL水中,乙酸乙酯萃取,合并有机相,依次用1N HCl溶液、水、饱和食盐水溶液洗涤,无水硫酸钠干燥,过滤浓缩,柱层析得到209 mg化合物11,为淡黄色油状液体。LC-MS(APCI):m/z=393.3(M+1)+。 163 mg of compound 5 was dissolved in 3 mL of anhydrous DMF, and 253 mg of HATU, 90 mg of compound 10, 287 μL of DIPEA was added under ice bath, and the mixture was stirred for 2 hours under ice bath. The reaction mixture was poured into 20 mL of water and extracted with EtOAc. EtOAc (EtOAc) Yellow oily liquid. LC-MS (APCI): m / z = 393.3 (M + 1) +.
实施例3.中间体17的合成。Example 3. Synthesis of Intermediate 17.
步骤1.化合物13的合成。Step 1. Synthesis of Compound 13.
取甲基异丙基酮2.6 g溶于无水乙醇,冰盐浴下滴加4.87 g液溴,滴完后室温反应3小时。取60 mL石油醚加入反应体系,并用水洗涤两次,石油醚反萃水相,合并石油醚,并用冰冷的饱和碳酸钠溶液洗涤两次,饱和食盐水洗涤一次,无水硫酸钠干燥,过滤浓缩,得无色油状液体3.04 g。1H NMR(300 MHz,CDCl3)δ3.99(s,2H),2.99(hept,J=6.9 Hz,1H),2.44(s,0H),2.14(s,0H),1.94(s,0H),1.86(s,0H),1.17(d,J=6.9 Hz,6H)。2.6 g of methyl isopropyl ketone was dissolved in absolute ethanol, and 4.87 g of liquid bromine was added dropwise to the ice salt bath, and the mixture was reacted at room temperature for 3 hours after the completion of the dropwise addition. Add 60 mL of petroleum ether to the reaction system, wash twice with water, strip the aqueous phase with petroleum ether, combine with petroleum ether, wash twice with ice-cold saturated sodium carbonate solution, wash once with saturated brine, dry over anhydrous sodium sulfate, filter Concentration gave a colorless oily liquid, 3.04 g. 1 H NMR (300 MHz, CDCl 3 ) δ 3.99 (s, 2H), 2.99 (hept, J = 6.9 Hz, 1H), 2.44 (s, 0H), 2.14 (s, 0H), 1.94 (s, 0H) ), 1.86 (s, 0H), 1.17 (d, J = 6.9 Hz, 6H).
步骤2.化合物15的合成。Step 2. Synthesis of Compound 15.
取0.75 g化合物13和0.49 g硫代草氨酸乙酯(化合物14)溶于无水乙醇,加热回流3小时。冰水浴降温,加乙酸乙酯和水淬灭反应,浓氨水调pH~7,分液,水相用乙酸乙酯萃取,合并有机相,饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,柱层析得淡黄色油状液体0.383 g。1H NMR(300MHz,DMSO-d6)δ7.72(s,1H),4.35(q,J=7.1 Hz,2H),3.17–3.05(m,1H),1.31(t,J=7.1 Hz,3H),1.25(d,J=6.9 Hz,6H)。0.75 g of Compound 13 and 0.49 g of thiooxalinate (Compound 14) were dissolved in absolute ethanol and heated to reflux for 3 hours. The ice water bath was cooled, and the reaction was quenched with ethyl acetate and water. The mixture was combined with ethyl acetate and water, and the mixture was evaporated. Column chromatography gave a pale yellow oily liquid 0.383 g. 1 H NMR (300MHz, DMSO- d6) δ7.72 (s, 1H), 4.35 (q, J = 7.1 Hz, 2H), 3.17-3.05 (m, 1H), 1.31 (t, J = 7.1 Hz, 3H ), 1.25 (d, J = 6.9 Hz, 6H).
步骤3.化合物16的合成。Step 3. Synthesis of Compound 16.
取383 mg化合物15溶于1.1 mL MeOH和3.6 mL THF(四氢呋喃),加入LiOH·H2O 77 mg和水50μL,50℃反应2小时。浓缩反应液,4N HCl溶液调pH~2,加入乙酸乙酯萃取,合并乙酸乙酯相,并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得到棕黄色油状液体261 mg即为化合物17,LC-MS(APCI):m/z=172.0(M+1)+。383 mg of Compound 15 was dissolved in 1.1 mL of MeOH and 3.6 mL of THF (tetrahydrofuran), and LiOH·H 2 O 77 mg and 50 μL of water were added, and the mixture was reacted at 50 ° C for 2 hours. The reaction mixture was concentrated, and the mixture was diluted with EtOAc EtOAc EtOAc EtOAc EtOAc. LC-MS (APCI): m / z = 172.0 (m + 1) +.
步骤4.化合物无17的合成。Step 4. Synthesis of the compound without 17.
取261 mg化合物16溶于3.6 mL干燥二氯甲烷,加入7.2μL DMF(二甲基甲酰胺),冰浴下加入草酰氯280μL,升温至室温搅拌1小时。浓缩,得到棕黄色油状液体直接用于下一步。261 mg of Compound 16 was dissolved in 3.6 mL of dry dichloromethane, 7.2 μL of DMF (dimethylformamide) was added, and 280 μL of oxalyl chloride was added thereto under ice-cooling, and the mixture was stirred at room temperature for 1 hour. Concentration gave a brownish yellow oily liquid which was taken directly to next.
实施例4.中间体21的合成。Example 4. Synthesis of intermediate 21.
步骤1.化合物19的合成。Step 1. Synthesis of Compound 19.
取2-甲基-3-甲氧基苯胺5.0 g溶于55 mL二甲苯,0℃下加入1M BCl3/DCM溶液38.5 mL,5℃反应30分钟,加入2.5 mL乙腈,5℃下反应45分钟,将反应液转移至滴液漏斗中,0℃下滴加至三氯化铝5.14 g的25 mL二甲苯溶液中,5℃反应45分钟后升温,分水器分离二氯甲烷后75℃反应过夜。冰水浴冷却,反应液倒入10 mL冰水中,回流反应2小时。冷却至室温后分液,水层用二氯甲烷萃取,合并二甲苯和二氯甲烷,并依次用1N NaOH溶液和饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得棕色油状液体,0℃下加入冰冷的异丙醚,并于0℃下搅拌2小时,析出灰色固体,过滤,滤渣用冰冷异丙醚洗涤,收集滤渣,45℃真空干燥过夜,得1.86 g灰色固体粉末即为化合物19。1H NMR(500 MHz,CDCl3)δ7.66(d,J=9.0 Hz,1H),6.31(d,J=9.0 Hz,1H),3.88(s,3H),2.55(s,3H),2.02(s,3H)。Take 5.0 g of 2-methyl-3-methoxyaniline in 55 mL of xylene, add 38.5 mL of 1M BCl 3 /DCM solution at 0 ° C, react for 5 minutes at 5 ° C, add 2.5 mL of acetonitrile, and react at 5 ° C. In a minute, the reaction solution was transferred to a dropping funnel, and added dropwise to a solution of 5.14 g of aluminum trichloride in 25 mL of xylene at 0 ° C, and reacted at 5 ° C for 45 minutes, then heated, and the separator was separated by dichloromethane at 75 ° C. The reaction was overnight. The mixture was cooled in an ice water bath, and the reaction solution was poured into 10 mL of ice water and refluxed for 2 hours. After cooling to room temperature, the mixture was separated with methylene chloride. EtOAc (EtOAc)EtOAc. The ice-cold isopropyl ether was added thereto, and the mixture was stirred at 0 ° C for 2 hours to precipitate a gray solid, which was filtered, and the residue was washed with ice-cold isopropyl ether. The residue was collected and dried under vacuum at 45 ° C overnight to obtain 1.86 g of a gray solid powder. . 1 H NMR (500 MHz, CDCl 3 ) δ 7.66 (d, J = 9.0 Hz, 1H), 6.31 (d, J = 9.0 Hz, 1H), 3.88 (s, 3H), 2.55 (s, 3H), 2.02 (s, 3H).
步骤2.化合物20的合成。Step 2. Synthesis of Compound 20.
取206 mg上述新制的化合物17溶于3 mL二氧六环,滴加化合物19(245 mg)的二氧六环(3mL)溶液,滴完室温搅拌1小时。浓缩反应液,加入3 mL饱和NaHCO3溶液和3 mL二氯甲烷分液,水相用二氯甲烷萃取。合并二氯甲烷,并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,得白色固体粉末404 mg即为化合物20。1H NMR(300 MHz,CDCl3)δ11.30(s,1H),7.76(d,J=8.7 Hz,1H),7.17(d,J=0.9 Hz,1H),6.79(d,J=8.7 Hz,1H),3.93(s,3H),3.32–3.13(m,1H),2.59(s,3H),2.17(s,3H),1.41(d,J=6.9 Hz,6H)。206 mg of the above-mentioned fresh compound 17 was dissolved in 3 mL of dioxane, and a solution of compound 19 (245 mg) in dioxane (3 mL) was added dropwise, and the mixture was stirred at room temperature for 1 hour. The reaction solution was concentrated, with 3 mL of saturated NaHCO 3 solution and 3 mL of dichloromethane, and the aqueous phase extracted with dichloromethane. The combined dichloromethane was washed with brine, dried over anhydrous sodium sulfate and filtered and evaporated 1 H NMR (300 MHz, CDCl 3 ) δ 11.30 (s, 1H), 7.76 (d, J = 8.7 Hz, 1H), 7.17 (d, J = 0.9 Hz, 1H), 6.79 (d, J = 8.7 Hz, 1H), 3.93 (s, 3H), 3.32 - 3.13 (m, 1H), 2.59 (s, 3H), 2.17 (s, 3H), 1.41 (d, J = 6.9 Hz, 6H).
步骤3.化合物21的合成。Step 3. Synthesis of Compound 21.
取255 mg化合物20溶于5 mL叔丁醇,加入180 mg叔丁醇钾,加热回流6小时。浓缩反应液,加入二氯甲烷(DCM)和水分液,水相用DCM萃取,合并DCM相,并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,得到白色固体粉末180 mg即为化合物21。1H NMR(300 MHz,CDCl3)δ9.54(s,1H),8.24(d,J=9.0 Hz,1H),7.09(d,J=0.9 Hz,1H),7.01(d,J=9.1 Hz,1H),6.72(d,J=1.8Hz,1H),3.97(s,3H),3.28–3.09(m,1H),2.42(s,3H),1.39(d,J=6.9 Hz,6H)。 255 mg of Compound 20 was dissolved in 5 mL of tert-butanol, and 180 mg of potassium t-butoxide was added thereto, followed by heating under reflux for 6 hours. The reaction mixture was concentrated, dichloromethane (DCM) and EtOAc (EtOAc) was evaporated. . 1 H NMR (300 MHz, CDCl 3 ) δ 9.54 (s, 1H), 8.24 (d, J = 9.0 Hz, 1H), 7.09 (d, J = 0.9 Hz, 1H), 7.01 (d, J = 9.1 Hz, 1H), 6.72 (d, J = 1.8 Hz, 1H), 3.97 (s, 3H), 3.28 - 3.09 (m, 1H), 2.42 (s, 3H), 1.39 (d, J = 6.9 Hz, 6H) ).
实施例5.制备取代的喹啉化合物A-1Example 5. Preparation of Substituted Quinoline Compound A-1
具体合成步骤如下:The specific synthesis steps are as follows:
步骤1.化合物23的合成。Step 1. Synthesis of Compound 23.
取2-甲基-3-羟基硝基苯5.0 g溶于40 mL DMF,加入6.30 g碳酸钾固体,冰水浴下加入2.24 mL氘代碘甲烷(CD3I),室温搅拌3小时。反应液倒入300 mL水中,乙酸乙酯萃取三次(30 mL×3),合并有机相,并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,得棕色油状液体8.4 g。1H NMR(300 MHz,CDCl3)δ7.40(dd,J=8.2,1.1 Hz,1H),7.32–7.22(m,2H),7.04(dd,J=8.2,1.1 Hz,1H)。5.0 g of 2-methyl-3-hydroxynitrobenzene was dissolved in 40 mL of DMF, 6.30 g of potassium carbonate solid was added, and 2.24 mL of deuterated iodomethane (CD 3 I) was added under ice water bath, and stirred at room temperature for 3 hours. The reaction mixture was poured into water (300 mL), EtOAc (EtOAc m. 1 H NMR (300 MHz, CDCl 3 ) δ 7.40 (dd, J = 8.2, 1.1 Hz, 1H), 7.32 - 7.22 (m, 2H), 7.04 (dd, J = 8.2, 1.1 Hz, 1H).
步骤2.化合物24的合成。Step 2. Synthesis of Compound 24.
取8.4 g化合物23溶于40 mL乙醇(EtOH)和10 mL水(H2O),加入8.9 g还原铁粉和3.5 g氯化铵固体,加热回流2小时。过滤,滤渣用乙酸乙酯洗涤至滤液呈无色。滤液浓缩,柱层析得无色油状液体4.7 g,LC-MS(APCI):m/z=170.2(M+1)+。 8.4 g of compound 23 was dissolved in 40 mL of ethanol (EtOH) and 10 mL of water (H 2 O), and 8.9 g of reduced iron powder and 3.5 g of ammonium chloride solid were added, and the mixture was heated under reflux for 2 hours. Filtration and filtration of the residue with ethyl acetate until the filtrate was colourless. The filtrate was concentrated to give a colorless oil by column chromatography, liquid 4.7 g, LC-MS (APCI ): m / z = 170.2 (M + 1) +.
步骤3.化合物25的合成。Step 3. Synthesis of Compound 25.
取4.7 g化合物24溶于55 mL二甲苯,0℃下加入1M BCl3/DCM溶液38.85 mL,5℃反应30分钟,加入4.01 mL乙腈,5℃下反应45分钟,将反应液转移至滴液漏斗中,0℃下滴加至三氯化铝4.56 g的25 mL二甲苯溶液中,5℃反应45分钟后升温,分水器分离二氯甲烷后75℃反应过夜。冰水浴冷却,反应液倒入10 mL冰水中,回流反应2小时。冷却至室温后分液,水层用二氯甲烷萃取,合并二甲苯和二氯甲烷,并依次用1N NaOH溶液和饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得棕色油状液体,0℃下加入冰冷的异丙醚,并于0℃下搅拌2小时,析出灰色固体,过滤,滤渣用冰冷异丙醚洗涤,收集滤渣,45℃真空干燥过夜,得2.992 g灰色固体粉末即为化合物25。1H NMR(500 MHz,CDCl3)δ7.65(d,J=9.0 Hz,1H),6.30(d,J=9.0 Hz,1H),2.55(s,3H),2.01(s,3H)。Take 4.7 g of compound 24 dissolved in 55 mL of xylene, add 38.85 mL of 1M BCl 3 /DCM solution at 0 ° C, react at 5 ° C for 30 minutes, add 4.01 mL of acetonitrile, react at 5 ° C for 45 minutes, transfer the reaction solution to the dropping solution. In a funnel, the solution was added dropwise to a solution of 4.56 g of aluminum trichloride in 25 mL of xylene at 0 ° C, and the mixture was heated at 5 ° C for 45 minutes, and then heated. The dichloromethane was separated by a water separator and reacted at 75 ° C overnight. The mixture was cooled in an ice water bath, and the reaction solution was poured into 10 mL of ice water and refluxed for 2 hours. After cooling to room temperature, the mixture was separated with methylene chloride. EtOAc (EtOAc)EtOAc. The ice-cold isopropyl ether was added thereto, and the mixture was stirred at 0 ° C for 2 hours to precipitate a gray solid, which was filtered, and the residue was washed with ice-cold isopropyl ether. The residue was collected and dried under vacuum at 45 ° C overnight to obtain 2.992 g of a gray solid powder. . 1 H NMR (500 MHz, CDCl 3 ) δ 7.65 (d, J = 9.0 Hz, 1H), 6.30 (d, J = 9.0 Hz, 1H), 2.55 (s, 3H), 2.01 (s, 3H).
步骤4.化合物26的合成。Step 4. Synthesis of Compound 26.
取500 mg化合物17溶于5 mL二氧六环,滴加化合物25(450 mg)的二氧六环(5 mL)溶液,滴完室温搅拌1小时。浓缩反应液,加入3 mL饱和NaHCO3溶液和3 mL二氯甲烷分液,水相用二氯甲烷萃取。合并二氯甲烷,并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,得白色固体粉末413 mg即为化合物26。500 mg of the compound 17 was dissolved in 5 mL of dioxane, and a solution of the compound 25 (450 mg) in dioxane (5 mL) was added dropwise, and the mixture was stirred at room temperature for 1 hour. The reaction solution was concentrated, with 3 mL of saturated NaHCO 3 solution and 3 mL of dichloromethane, and the aqueous phase extracted with dichloromethane. The combined dichloromethane was washed with brine, dried over anhydrous sodium sulfate
步骤5.化合物27的合成。Step 5. Synthesis of Compound 27.
取2.85 g化合物26溶于30 mL叔丁醇,加入2.37 g叔丁醇钾,加热回流6小时。浓缩反应液,加入DCM和水分液,水相用DCM萃取,合并DCM相,并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,得到白色固体粉末1.73 g即为化合物27。1H NMR(300 MHz,CDCl3)δ9.55(s,1H),8.25(d,J=9.0 Hz,1H),7.10(d,J=0.9 Hz,1H),7.01(d,J=9.1 Hz,1H),6.72(d,J=1.8 Hz,1H),3.28–3.12(m,1H),2.43(s,3H),1.39(d,J=6.9 Hz,6H)。2.85 g of Compound 26 was dissolved in 30 mL of tert-butanol, 2.37 g of potassium t-butoxide was added, and the mixture was heated under reflux for 6 hours. The reaction mixture was concentrated, DCM and EtOAc was evaporated, evaporated, evaporated, evaporated, evaporated. 1 H NMR (300 MHz, CDCl 3 ) δ 9.55 (s, 1H), 8.25 (d, J = 9.0 Hz, 1H), 7.10 (d, J = 0.9 Hz, 1H), 7.01 (d, J = 9.1 Hz, 1H), 6.72 (d, J = 1.8 Hz, 1H), 3.28 - 3.12 (m, 1H), 2.43 (s, 3H), 1.39 (d, J = 6.9 Hz, 6H).
步骤6.化合物28的合成。Step 6. Synthesis of Compound 28.
取500 mg化合物11、486 mg化合物27和669 mg三苯基膦溶于10 mL新蒸无水THF,-15℃下滴加552μL偶氮二甲酸二异丙酯(DIAD),并于-15℃搅拌5小时、自然升温室温搅拌过夜。冰盐浴下加入冰水淬灭反应,用乙酸乙酯(EA)萃取水相,合并EA相,饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,柱层析得到485 mg化合物28。LC-MS(APCI):m/z=692.4(M+1)+。500 mg of compound 11, 486 mg of compound 27 and 669 mg of triphenylphosphine were dissolved in 10 mL of freshly distilled anhydrous THF, and 552 μL of diisopropyl azodicarboxylate (DIAD) was added dropwise at -15 ° C, and at -15 The mixture was stirred at ° C for 5 hours, and naturally stirred at room temperature overnight. The reaction mixture was quenched with EtOAc (EtOAc)EtOAc. LC-MS (APCI): m / z = 692.4 (M + 1) +.
步骤7.化合物29的合成。Step 7. Synthesis of Compound 29.
取485 mg化合物28,18 mg第一代格拉布催化剂(Hoveyda-Grubbs催化剂)溶于420 mL无水二氯乙烷,氮气脱气30分钟,75℃搅拌17小时。浓缩反应液,柱层析得到234 mg淡黄色固体 即为化合物29。LC-MS(APCI):m/z=665.4(M+1)+。485 mg of compound 28, 18 mg of first-generation gerab catalyst (Hoveyda-Grubbs catalyst) was dissolved in 420 mL of anhydrous dichloroethane, degassed under nitrogen for 30 minutes, and stirred at 75 ° C for 17 hours. The reaction mixture was concentrated to give 234 mg (yield of pale yellow solid). LC-MS (APCI): m / z = 665.4 (M + 1) +.
步骤8.化合物30的合成。Step 8. Synthesis of Compound 30.
取477 mg化合物29溶于15 mL甲醇和26 mL四氢呋喃,冰水浴下加入2.89 g一水合氢氧化锂和7.5 mL水,室温搅拌过夜。1N盐酸调pH2~3,EA萃取,合并EA相并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得315 mg淡黄色色固体粉末,LC-MS(APCI):m/z=650.4(M+1)+。477 mg of compound 29 was dissolved in 15 mL of methanol and 26 mL of tetrahydrofuran, and 2.89 g of lithium hydroxide monohydrate and 7.5 mL of water were added thereto under ice water, and stirred at room temperature overnight. 1N Hydrochloric acid was adjusted to pH 2~3, EA was extracted, EA phase was combined and washed with saturated brine, dried over anhydrous sodium sulfate and filtered and evaporated to give 315 mg of pale yellow solid powder, LC-MS (APCI): m/z=650.4 (M +1) + .
步骤9.化合物A-1的合成。Step 9. Synthesis of Compound A-1.
取496 mg化合物30溶于15 mL新蒸无水四氢呋喃,氮气保护下加入308 mg N,N'-羰基二咪唑(CDI),加热回流2小时。冷却至50℃后加入400 mg环丙基磺酰胺和286 mg 1,8-二氮杂双环[5.4.0]十一碳-7-烯(DBU),50℃搅拌过夜。浓缩反应液,加入2 mL1N盐酸和5 mL二氯甲烷分液,有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,柱层析得277 mg淡黄色固体粉末即为化合物A-1,LC-MS(APCI):m/z=753.4(M+1)+。496 mg of compound 30 was dissolved in 15 mL of freshly distilled anhydrous tetrahydrofuran, and 308 mg of N,N'-carbonyldiimidazole (CDI) was added under a nitrogen atmosphere, and the mixture was heated under reflux for 2 hours. After cooling to 50 ° C, 400 mg of cyclopropylsulfonamide and 286 mg of 1,8-diazabicyclo [5.4.0]undec-7-ene (DBU) were added and stirred at 50 ° C overnight. The reaction mixture was concentrated, and 2 mL of 1N hydrochloric acid and 5 mL of dichloromethane was added. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate and filtered and evaporated to give 277 mg of pale yellow solid powder as compound A-1 , LC-MS (APCI): m/z = 753.4 (M + 1) + .
实施例6.制备取代的喹啉化合物A-2Example 6. Preparation of Substituted Quinoline Compound A-2
具体合成步骤如下: The specific synthesis steps are as follows:
步骤1.化合物32的合成。Step 1. Synthesis of Compound 32.
取2-氯-6-硝基甲苯4.0 g溶于40 mLMeOD,加入1.0 g MeONa,微波100℃搅拌2小时。冷却后加浓氘代盐酸调pH3~4,反应液倒入20 mL饱和食盐水中,乙酸乙酯萃取(10mL×3),合并乙酸乙酯,并用无水硫酸钠干燥,过滤浓缩得3.953 g淡黄色固体即为化合物32。1H NMR(500 MHz,CDCl3)δ7.71(dd,J=8.2,1.3 Hz,1H),7.61(dd,J=8.0,1.3 Hz,1H),7.29(d,J=8.2,1H)。4.0 g of 2-chloro-6-nitrotoluene was dissolved in 40 mL of MeOD, 1.0 g of MeONa was added, and the mixture was stirred at 100 ° C for 2 hours in a microwave. After cooling, the pH was adjusted to pH 3-4, and the reaction mixture was poured into 20 mL of brine. The yellow solid is compound 32. 1 H NMR (500 MHz, CDCl 3 ) δ 7.71 (dd, J = 8.2, 1.3 Hz, 1H), 7.61 (dd, J = 8.0, 1.3 Hz, 1H), 7.29 (d, J = 8.2, 1H) .
步骤2.化合物33的合成。Step 2. Synthesis of Compound 33.
取4.24 g化合物32,4.52 g四甲氧基硼化钠,223 mg三(二亚苄基丙酮)二钯,和205 mg 2-二叔丁膦基-2',4',6'-三异丙基联苯混合,氮气保护下加入22 mL无水DMF,加热100℃搅拌2小时。反应冷却后倒入100 mL水中,乙酸乙酯萃取三次(30mL×3),合并乙酸乙酯,过滤,滤液用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得4.875 g棕色油状液体。4.24 g of compound 32, 4.52 g of sodium tetramethoxyborate, 223 mg of tris(dibenzylideneacetone)dipalladium, and 205 mg of 2-di-tert-butylphosphino-2',4',6'-triisopropyl The phenylbenzene was mixed, and 22 mL of anhydrous DMF was added under nitrogen atmosphere, and the mixture was stirred at 100 ° C for 2 hours. After the reaction was cooled, the mixture was poured into EtOAc (3 mL).
步骤3.化合物34的合成。Step 3. Synthesis of Compound 34.
取3.25 g化合物33,6.35 g还原铁粉,2.43 g氯化铵固体混合,加入20 mL乙醇和5 mL水,加热回流2小时。反应液冷却后过滤,滤渣用甲醇洗涤至滤液无色,合并滤液,浓缩,柱层析得无色液体3.06 g。3.25 g of compound 33, 6.35 g of reduced iron powder, and 2.43 g of ammonium chloride solid were mixed, and 20 mL of ethanol and 5 mL of water were added, and the mixture was heated under reflux for 2 hours. The reaction mixture was cooled and filtered, and the residue was washed with methanol, and the filtrate was evaporated.
步骤4.化合物35的合成。Step 4. Synthesis of Compound 35.
取4.7 g化合物34溶于55 mL二甲苯,0℃下加入1M BCl3/DCM溶液38.85 mL,5℃反应30分钟,加入4.01 mL乙腈,5℃下反应45分钟,将反应液转移至滴液漏斗中,0℃下滴加至三氯 化铝4.56 g的25 mL二甲苯溶液中,5℃反应45分钟后升温,分水器分离二氯甲烷后75℃反应过夜。冰水浴冷却,反应液倒入10 mL冰水中,回流反应2小时。冷却至室温后分液,水层用二氯甲烷萃取,合并二甲苯和二氯甲烷,并依次用1N NaOH溶液和饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得棕色油状液体,0℃下加入冰冷的异丙醚,并于0℃下搅拌2小时,析出灰色固体,过滤,滤渣用冰冷异丙醚洗涤,收集滤渣,45℃真空干燥过夜,得2.992 g灰色固体粉末即为化合物35。1H NMR(500 MHz,CDCl3)δ7.65(d,J=9.0 Hz,1H),6.31(d,J=9.0 Hz,1H),3.87(s,3H),2.55(s,3H)。4.7 g of compound 34 was dissolved in 55 mL of xylene, and 38.85 mL of 1 M BCl 3 /DCM solution was added at 0 ° C, reacted at 5 ° C for 30 minutes, 4.01 mL of acetonitrile was added, and reacted at 5 ° C for 45 minutes to transfer the reaction solution to the dropping solution. In a funnel, the solution was added dropwise to a solution of 4.56 g of aluminum trichloride in 25 mL of xylene at 0 ° C, and the mixture was heated at 5 ° C for 45 minutes, and then heated. The dichloromethane was separated by a water separator and reacted at 75 ° C overnight. The mixture was cooled in an ice water bath, and the reaction solution was poured into 10 mL of ice water and refluxed for 2 hours. After cooling to room temperature, the mixture was separated with methylene chloride. EtOAc (EtOAc)EtOAc. The ice-cold isopropyl ether was added thereto, and the mixture was stirred at 0 ° C for 2 hours to precipitate a gray solid, which was filtered, and the residue was washed with ice-cold isopropyl ether. The residue was collected and dried under vacuum at 45 ° C overnight to obtain 2.992 g of a gray solid powder. . 1 H NMR (500 MHz, CDCl 3 ) δ 7.65 (d, J = 9.0 Hz, 1H), 6.31 (d, J = 9.0 Hz, 1H), 3.87 (s, 3H), 2.55 (s, 3H).
步骤5.化合物36的合成。Step 5. Synthesis of Compound 36.
取500 mg化合物17溶于5 mL二氧六环,滴加化合物35(450 mg)的二氧六环(5 mL)溶液,滴完室温搅拌1小时。浓缩反应液,加入3 mL饱和NaHCO3溶液和3 mL二氯甲烷分液,水相用二氯甲烷萃取。合并二氯甲烷,并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,得白色固体粉末413 mg即为化合物36。500 mg of the compound 17 was dissolved in 5 mL of dioxane, and a solution of the compound 35 (450 mg) in dioxane (5 mL) was added dropwise, and the mixture was stirred at room temperature for 1 hour. The reaction solution was concentrated, with 3 mL of saturated NaHCO 3 solution and 3 mL of dichloromethane, and the aqueous phase extracted with dichloromethane. The combined dichloromethane was washed with brine, dried over anhydrous sodium sulfate
步骤6.化合物37的合成。Step 6. Synthesis of Compound 37.
取451 mg化合物36溶于10 mL叔丁醇,加入540 mg叔丁醇钾,加热回流6小时。浓缩反应液,加入DCM和水分液,水相用DCM萃取,合并DCM相,并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,得到白色固体粉末413 mg即为化合物37。1H NMR(400 MHz,CDCl3)δ9.53(s,1H),8.30–8.19(m,1H),7.10(d,J=0.9 Hz,1H),7.02(d,J=9.1 Hz,1H),6.72(d,J=1.8 Hz,1H),3.98(s,3H),3.24–3.16(m,1H),1.40(d,J=6.9 Hz,6H)。451 mg of compound 36 was dissolved in 10 mL of t-butanol, and 540 mg of potassium t-butoxide was added thereto, followed by heating under reflux for 6 hours. The reaction mixture was concentrated, DCM and EtOAc was evaporated, evaporated, evaporated, evaporated, evaporated. 1 H NMR (400 MHz, CDCl 3 ) δ 9.53 (s, 1H), 8.30 - 8.19 (m, 1H), 7.10 (d, J = 0.9 Hz, 1H), 7.02 (d, J = 9.1 Hz, 1H) ), 6.72 (d, J = 1.8 Hz, 1H), 3.98 (s, 3H), 3.24 - 3.16 (m, 1H), 1.40 (d, J = 6.9 Hz, 6H).
步骤7.化合物38的合成。Step 7. Synthesis of Compound 38.
取500 mg化合物11、486 mg化合物37和669 mg三苯基膦溶于10 mL新蒸无水THF,-15℃下滴加552μL DIAD,并于-15℃搅拌5小时、自然升温室温搅拌过夜。冰盐浴下加入冰水淬灭反应,EA萃取水相,合并EA相,饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,柱层析得到485 mg化合物38。LC-MS(APCI):m/z=692.4(M+1)+。500 mg of compound 11, 486 mg of compound 37 and 669 mg of triphenylphosphine were dissolved in 10 mL of freshly distilled anhydrous THF, and 552 μL of DIAD was added dropwise at -15 ° C, and stirred at -15 ° C for 5 hours, and stirred at room temperature overnight. . The reaction was quenched by the addition of ice water, and the aqueous layer was evaporated. EtOAc was evaporated. LC-MS (APCI): m / z = 692.4 (M + 1) +.
步骤8.化合物39的合成。Step 8. Synthesis of Compound 39.
取481 mg化合物38,18 mg Hoveyda-Grubbs第一代催化剂溶于420 mL无水二氯乙烷,氮气脱气30分钟,75℃搅拌15小时。浓缩反应液,柱层析得到220 mg淡黄色固体即为化合物39。LC-MS(APCI):m/z=664.4(M+1)+。481 mg of compound 38, 18 mg of Hoveyda-Grubbs first generation catalyst was dissolved in 420 mL of anhydrous dichloroethane, degassed for 30 minutes under nitrogen, and stirred at 75 ° C for 15 hours. The reaction mixture was concentrated and purified by column chromatography to yieldd to afforded Compound Compound Compound LC-MS (APCI): m / z = 664.4 (M + 1) +.
步骤9.化合物40的合成。Step 9. Synthesis of Compound 40.
取220 mg化合物39溶于20 mL甲醇和30mL四氢呋喃,冰水浴下加入2.89 g一水合氢氧化锂 和15mL水,室温搅拌过夜。1N盐酸调pH2~3,EA萃取,合并EA相并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得186 mg淡黄色色固体粉末,LC-MS(APCI):m/z=650.4(M+1)+。220 mg of Compound 39 was dissolved in 20 mL of methanol and 30 mL of tetrahydrofuran, and 2.89 g of lithium hydroxide monohydrate and 15 mL of water were added under ice water, and stirred at room temperature overnight. 1N Hydrochloric acid was adjusted to pH 2~3, EA was extracted, EA phase was combined and washed with saturated brine, dried over anhydrous sodium sulfate and evaporated to give 186 mg of pale yellow solid powder, LC-MS (APCI): m/z=650.4 (M +1) + .
步骤11.化合物A-2的合成。Step 11. Synthesis of Compound A-2.
取150 mg化合物40溶于15 mL新蒸无水四氢呋喃,氮气保护下加入120 mg CDI,加热回流2小时。冷却至50℃后加入145 mg环丙基磺酰胺和83 mg DBU,50℃搅拌过夜。浓缩反应液,加入2mL1N盐酸和5mL二氯甲烷分液,有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,柱层析得102 mg淡黄色固体粉末即为化合物A-2。LC-MS(APCI):m/z=753.4(M+1)+。150 mg of compound 40 was dissolved in 15 mL of freshly distilled anhydrous tetrahydrofuran, and 120 mg of CDI was added under nitrogen atmosphere, and the mixture was heated under reflux for 2 hours. After cooling to 50 ° C, 145 mg of cyclopropylsulfonamide and 83 mg of DBU were added and stirred at 50 ° C overnight. The reaction mixture was concentrated, and 2 mL of 1N hydrochloric acid and 5 mL of dichloromethane was added, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, LC-MS (APCI): m / z = 753.4 (M + 1) +.
实施例7.制备取代的喹啉化合物A-3Example 7. Preparation of Substituted Quinoline Compound A-3
具体合成步骤如下: The specific synthesis steps are as follows:
步骤1.化合物41的合成。Step 1. Synthesis of Compound 41.
取6-溴-1-乙烯0.749 g溶于7.4 mL DMSO,分批缓慢加入448 mg叠氮化钠,室温搅拌2小时。反应液倒入25 mL水中,乙醚萃取三次(10 mL×3),合并乙醚,用饱和食盐水洗涤。向乙醚溶液里加入2.41 g三苯基膦,0.5 mL水,室温搅拌过夜。反应液用1N盐酸洗涤三次(25 mL×3),合并水相,并用乙醚反萃一次。冰水浴下用NaOH固体调节pH8~9,乙醚萃取,合并萃取乙醚,饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得450 mg无色油状液体,直接用于下一步。0.749 g of 6-bromo-1-ethene was dissolved in 7.4 mL of DMSO, and 448 mg of sodium azide was slowly added in portions, and the mixture was stirred at room temperature for 2 hours. The reaction solution was poured into 25 mL of water and extracted with EtOAc (3 mL, 3). To the ether solution, 2.41 g of triphenylphosphine, 0.5 mL of water was added, and the mixture was stirred at room temperature overnight. The reaction solution was washed three times with 1N hydrochloric acid (25 mL×3), The mixture was adjusted to pH 8 to 9 with NaOH solids, and extracted with diethyl ether. The mixture was combined with diethyl ether, washed with saturated brine, dried over anhydrous sodium sulfate and filtered to give 450 mg of colorless oil.
步骤2.化合物42的合成。Step 2. Synthesis of Compound 42.
取450 mg化合物41溶于25 mL乙醚,冰水浴下滴加Boc2O 1.0 g,滴加DIPEA 974μL,室温搅拌过夜。浓缩反应液,柱层析得899 mg无色油状液体。450 mg of compound 41 was dissolved in 25 mL of diethyl ether, Boc 2 O 1.0 g was added dropwise thereto in an ice water bath, and DIPEA 974 μL was added dropwise thereto, and stirred at room temperature overnight. The reaction mixture was concentrated and purified by column chromatography eluting 881
步骤3.化合物43的合成。Step 3. Synthesis of Compound 43.
取899 mg化合物42溶于3 mL DMF,分批加入130 mg 60%NaH,室温搅拌30分钟,滴加655mg氘代碘甲烷,室温搅拌2小时。将反应液倒入20 mL水,乙酸乙酯萃取三次(5mL×3),合并乙酸乙酯相,并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得无色油状液体756 mg。1H NMR(400MHz,CDCl3)δ5.79(ddt,J=16.9,10.2,6.7 Hz,1H),5.13–4.87(m,2H),3.19(s,2H),2.06(dt,J=8.6, 4.3 Hz,2H),1.51(dt,J=14.8,7.2 Hz,2H),1.44(s,9H),1.36(dt,J=14.7,7.2 Hz,2H)。899 mg of compound 42 was dissolved in 3 mL of DMF, and 130 mg of 60% NaH was added in portions, stirred at room temperature for 30 minutes, and 655 mg of deuterated iodomethane was added dropwise, and the mixture was stirred at room temperature for 2 hours. The reaction mixture was poured into water (20 mL), EtOAc (EtOAc)EtOAc. 1 H NMR (400MHz, CDCl 3 ) δ5.79 (ddt, J = 16.9,10.2,6.7 Hz, 1H), 5.13-4.87 (m, 2H), 3.19 (s, 2H), 2.06 (dt, J = 8.6 , 4.3 Hz, 2H), 1.51 (dt, J = 14.8, 7.2 Hz, 2H), 1.44 (s, 9H), 1.36 (dt, J = 14.7, 7.2 Hz, 2H).
步骤4.化合物44的合成。Step 4. Synthesis of Compound 44.
取756 mg化合物43,溶于10 mL DCM/TFA(V/V,8/2)混合溶液,室温搅拌2小时。浓缩反应液,加入3 mL饱和碳酸氢钠溶液和3 mL乙醚分液,水相用乙醚萃取(3mL×2),合并乙醚并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,得无色油状液体336 g即为化合物44。756 mg of compound 43 was dissolved in 10 mL of a DCM/TFA (V/V, 8/2) mixed solution and stirred at room temperature for 2 hours. The reaction mixture was concentrated, and the mixture was evaporated, evaporated, evaporated, evaporated. The oily liquid 336 g is the compound 44.
步骤5.化合物45的合成。Step 5. Synthesis of Compound 45.
取(1R,4R,5R)-3-氧代-2-氧杂二环[2.2.1]己烷-5-羧酸辛可尼丁盐3.51 g和1.09 g化合物44溶于10 mL DMF,冰浴冷却下加入HATU3.57 g,DIPEA10 g,冰浴搅拌1小时。反应液倒入100 mL水中,乙酸乙酯萃取,合并乙酸乙酯,并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,柱层析得达黄色油状液体1.86 g即为化合物45。1H NMR(500 MHz,CDCl3)δ5.77(ttd,J=13.1,6.7,2.8 Hz,1H),5.13–4.86(m,3H),3.43–3.23(m,2H),3.08–2.94(m,2H),2.29–2.02(m,6H),1.69–1.46(m,2H),1.46–1.31(m,2H).LC-MS(APCI):m/z=255.2(M+1)+。Take (1R,4R,5R)-3-oxo-2-oxabicyclo[2.2.1]hexane-5-carboxylic acid cinchonidine salt 3.51 g and 1.09 g of compound 44 dissolved in 10 mL DMF, ice HATU 3.57 g, DIPEA 10 g was added under cooling with a bath and stirred for 1 hour in an ice bath. The reaction mixture was poured into 100 mL of water, and ethyl acetate was evaporated, and ethyl acetate was evaporated. 1 H NMR (500 MHz, CDCl 3 ) δ 5.77 (ttd, J = 13.1, 6.7, 2.8 Hz, 1H), 5.13 - 4.86 (m, 3H), 3.43 - 3.23 (m, 2H), 3.08 - 2.94 ( m, 2H), 2.29–2.02 (m, 6H), 1.69–1.46 (m, 2H), 1.46–1.31 (m, 2H). LC-MS (APCI): m/z=255.2 (M+1) + .
步骤6.化合物46的合成。Step 6. Synthesis of Compound 46.
取1.86 g化合物45溶于13 mL四氢呋喃,冰水浴下滴加1M LiOH溶液5.4 mL,滴完冰水浴下搅拌1小时。4N HCl溶液调pH1~2,乙酸乙酯萃取(10×3),合并乙酸乙酯,并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,得淡黄色油状液体1.26g,LC-MS(APCI):m/z=252.3(M+1)+。1.86 g of the compound 45 was dissolved in 13 mL of tetrahydrofuran, and 5.4 mL of a 1 M LiOH solution was added dropwise thereto in an ice water bath, and the mixture was stirred for 1 hour under ice-cold bath. 4N HCl solution was adjusted to pH 1-2, ethyl acetate (10×3), and ethyl acetate was evaporated. APCI): m/z = 252.3 (M + 1) + .
步骤7.化合物47的合成。Step 7. Synthesis of Compound 47.
取1.26 g化合物46溶于10 mL无水DMF,冰浴下加入1.94 g HATU和789 mg化合物5,920μL DIPEA,冰浴下搅拌1小时。反应液倒入100 mL水中,EA萃取,合并EA相,依次用1N HCl溶液、水、饱和食盐水溶液洗涤,无水硫酸钠干燥,过滤浓缩,柱层析得到209 mg化合物47为淡黄色油状液体。LC-MS(APCI):m/z=396.3(M+1)+。1.26 g of compound 46 was dissolved in 10 mL of anhydrous DMF, and 1.94 g of HATU and 789 mg of compound 5, 920 μL of DIPEA were added under ice bath, and the mixture was stirred for 1 hour under ice bath. The reaction solution was poured into 100 mL of water, extracted with EA, and the EA phase was combined, washed with 1N HCl solution, water, saturated aqueous sodium chloride solution, dried over anhydrous sodium sulfate, and concentrated by filtration. . LC-MS (APCI): m / z = 396.3 (M + 1) +.
步骤8.化合物48的合成。Step 8. Synthesis of Compound 48.
取500 mg化合物47、477 mg化合物21和664 mg三苯基膦溶于10 mL新蒸无水THF,-15℃下滴加552μL DIAD,并于-15℃搅拌5小时、自然升温室温搅拌过夜。冰盐浴下加入冰水淬灭反应,EA萃取水相,合并EA相,饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,柱层析得到529 mg化合物48。LC-MS(APCI):m/z=692.4(M+1)+。500 mg of compound 47, 477 mg of compound 21 and 664 mg of triphenylphosphine were dissolved in 10 mL of freshly distilled anhydrous THF, and 552 μL of DIAD was added dropwise at -15 ° C, and stirred at -15 ° C for 5 hours, and stirred at room temperature overnight. . The reaction was quenched by the addition of ice water, and the mixture was evaporated. LC-MS (APCI): m / z = 692.4 (M + 1) +.
步骤9.化合物49的合成。Step 9. Synthesis of Compound 49.
取529 mg化合物48,24 mg Hoveyda-Grubbs第一代催化剂溶于480 mL无水二氯乙烷,氮气脱气30分钟,75℃搅拌15小时。浓缩反应液,柱层析得到327 mg淡黄色固体即为化合物49。 LC-MS(APCI):m/z=664.4(M+1)+。529 mg of compound 48, 24 mg of Hoveyda-Grubbs first generation catalyst was dissolved in 480 mL of anhydrous dichloroethane, degassed under nitrogen for 30 minutes, and stirred at 75 ° C for 15 hours. The reaction mixture was concentrated and purified by column chromatography to afford 327 g of pale yellow solid. LC-MS (APCI): m / z = 664.4 (M + 1) +.
步骤10.化合物50的合成。Step 10. Synthesis of Compound 50.
取689 mg化合物49溶于20 mL甲醇和30 mL四氢呋喃,冰水浴下加入2.89 g一水合氢氧化锂和15 mL水,室温搅拌过夜。1N盐酸调pH2~3,EA萃取,合并EA相并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得686mg淡黄色色固体粉末,LC-MS(APCI):m/z=650.4(M+1)+。689 mg of Compound 49 was dissolved in 20 mL of methanol and 30 mL of tetrahydrofuran, and 2.89 g of lithium hydroxide monohydrate and 15 mL of water were added thereto under ice water, and stirred at room temperature overnight. 1N Hydrochloric acid was adjusted to pH 2~3, EA was extracted, EA phase was combined and washed with saturated brine, dried over anhydrous sodium sulfate and filtered to give 686mg of pale yellow solid powder, LC-MS (APCI): m/z=650.4 (M+ 1) + .
步骤11.化合物A-3的合成。Step 11. Synthesis of Compound A-3.
取686 mg化合物50溶于17 mL新蒸无水四氢呋喃,氮气保护下加入308 mgCDI,加热回流2小时。冷却至50℃后加入400 mg环丙基磺酰胺和286 mgDBU,50℃搅拌过夜。浓缩反应液,加入2 mL 1N盐酸和5 mL二氯甲烷分液,有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,柱层析得335 mg淡黄色固体粉末即为化合物A-3,LC-MS(APCI):m/z=753.4(M+1)+。686 mg of compound 50 was dissolved in 17 mL of freshly distilled anhydrous tetrahydrofuran, and 308 mg of CDI was added under nitrogen atmosphere, and the mixture was heated under reflux for 2 hours. After cooling to 50 ° C, 400 mg of cyclopropylsulfonamide and 286 mg of DBU were added and stirred at 50 ° C overnight. The reaction mixture was concentrated, and 2 mL of 1N hydrochloric acid and 5 mL of dichloromethane was added, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give 335 mg of pale yellow solid powder as compound A- 3, LC-MS (APCI): m / z = 753.4 (M + 1) + .
实施例8.制备取代的喹啉化合物A-4Example 8. Preparation of Substituted Quinoline Compound A-4
具体合成步骤如下: The specific synthesis steps are as follows:
步骤1.化合物51的合成。Step 1. Synthesis of Compound 51.
取4.41 g化合物33溶于30 mL二氯甲烷,冰盐浴下缓慢滴加三氯化硼4.69 mL,自然升温反应30分钟。冰水浴下滴加冰水10 mL。分液,有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得淡黄色固体3.80 g。4.41 g of compound 33 was dissolved in 30 mL of dichloromethane, and 4.69 mL of boron trichloride was slowly added dropwise in an ice salt bath, and the reaction was naturally carried out for 30 minutes. 10 mL of ice water was added to the ice water bath. The organic layer was washed with brine, dried over anhydrous sodium sulfate
步骤2.化合物52的合成。Step 2. Synthesis of Compound 52.
取3.56化合物51溶于20 mLDMF,加入6.30 g碳酸钾固体,室温搅拌30分钟,滴加3.97 g氘代碘甲烷,室温搅拌2小时。反应液倒入150 mL水中,乙酸乙酯萃取三次(20mL×3),合并乙酸乙酯,饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得3.63 g淡黄色油状液体。3.56 Compound 51 was dissolved in 20 mL of DMF, and 6.30 g of potassium carbonate solid was added, stirred at room temperature for 30 minutes, and 3.97 g of deuterated iodomethane was added dropwise, and stirred at room temperature for 2 hours. The reaction mixture was poured into 150 mL of water and EtOAc (3 mL, EtOAc)
步骤3.化合物53的合成。Step 3. Synthesis of Compound 53.
取3.63 g化合物52,6.40 g还原铁粉,2.41g氯化铵固体混合,加入20 mL乙醇和5 mL水,加热回流2小时。反应液冷却后过滤,滤渣用甲醇洗涤至滤液无色,合并滤液,浓缩,柱层析得无色液体3.36 g。3.63 g of compound 52, 6.40 g of reduced iron powder, and 2.41 g of ammonium chloride solid were mixed, and 20 mL of ethanol and 5 mL of water were added, and the mixture was heated under reflux for 2 hours. The reaction mixture was cooled and filtered, and the residue was washed with methanol, and the filtrate was evaporated. The filtrate was concentrated and concentrated to give a colorless liquid 3.36 g.
步骤4.化合物54的合成。Step 4. Synthesis of Compound 54.
取4.7 g化合物53溶于55 mL二甲苯,0℃下加入1M BCl3/DCM溶液38.85 mL,5℃反应30分钟,加入4.01 mL乙腈,5℃下反应45分钟,将反应液转移至滴液漏斗中,0℃下滴加至三氯化铝4.56 g的25 mL二甲苯溶液中,5℃反应45分钟后升温,分水器分离二氯甲烷后75℃反应 过夜。冰水浴冷却,反应液倒入10 mL冰水中,回流反应2小时。冷却至室温后分液,水层用二氯甲烷萃取,合并二甲苯和二氯甲烷,并依次用1N NaOH溶液和饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得棕色油状液体,0℃下加入冰冷的异丙醚,并于0℃下搅拌2小时,析出灰色固体,过滤,滤渣用冰冷异丙醚洗涤,收集滤渣,45℃真空干燥过夜,得2.992 g灰色固体粉末即为化合物54。1H NMR(400 MHz,CDCl3)δ7.66(d,J=9.0 Hz,1H),6.31(d,J=9.1 Hz,1H),2.56(s,3H)。Take 4.7 g of compound 53 dissolved in 55 mL of xylene, add 38.85 mL of 1M BCl 3 /DCM solution at 0 ° C, react at 5 ° C for 30 minutes, add 4.01 mL of acetonitrile, react at 5 ° C for 45 minutes, transfer the reaction solution to the dropping solution. In a funnel, the solution was added dropwise to a solution of 4.56 g of aluminum trichloride in 25 mL of xylene at 0 ° C, and the mixture was heated at 5 ° C for 45 minutes, and then heated. The dichloromethane was separated by a water separator and reacted at 75 ° C overnight. The mixture was cooled in an ice water bath, and the reaction solution was poured into 10 mL of ice water and refluxed for 2 hours. After cooling to room temperature, the mixture was separated with methylene chloride. EtOAc (EtOAc)EtOAc. The ice-cold isopropyl ether was added thereto, and the mixture was stirred at 0 ° C for 2 hours to precipitate a gray solid, which was filtered, and the residue was washed with ice-cold isopropyl ether. The residue was collected and dried in vacuo at 45 ° C overnight to give the compound . 1 H NMR (400 MHz, CDCl 3 ) δ 7.66 (d, J = 9.0 Hz, 1H), 6.31 (d, J = 9.1 Hz, 1H), 2.56 (s, 3H).
步骤5.化合物55的合成。Step 5. Synthesis of Compound 55.
取500 mg化合物54溶于5 mL二氧六环,滴加化合物17(450 mg)的二氧六环(5 mL)溶液,滴完室温搅拌1小时。浓缩反应液,加入3 mL饱和NaHCO3溶液和3 mL二氯甲烷分液,水相用二氯甲烷萃取。合并二氯甲烷,并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,得白色固体粉末445 mg即为化合物55。500 mg of the compound 54 was dissolved in 5 mL of dioxane, and a solution of the compound 17 (450 mg) in dioxane (5 mL) was added dropwise, and the mixture was stirred at room temperature for 1 hour. The reaction solution was concentrated, with 3 mL of saturated NaHCO 3 solution and 3 mL of dichloromethane, and the aqueous phase extracted with dichloromethane. The combined dichloromethane was washed with brine, dried over anhydrous sodium sulfate
步骤6.化合物56的合成。Step 6. Synthesis of Compound 56.
取445 mg化合物55溶于10 mL叔丁醇,加入540 mg叔丁醇钾,加热回流6小时。浓缩反应液,加入DCM和水分液,水相用DCM萃取,合并DCM相,并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,得到白色固体粉末413 mg即为化合物56。1H NMR(400 MHz,CDCl3)δ9.53(s,1H),8.30–8.19(m,1H),7.10(d,J=0.9 Hz,1H),7.02(d,J=9.1 Hz,1H),6.72(d,J=1.8 Hz,1H),3.24–3.16(m,1H),1.40(d,J=6.9 Hz,6H)。445 mg of Compound 55 was dissolved in 10 mL of t-butanol, and 540 mg of potassium t-butoxide was added thereto, followed by heating under reflux for 6 hours. The reaction mixture was concentrated, DCM and EtOAc was evaporated. 1 H NMR (400 MHz, CDCl 3 ) δ 9.53 (s, 1H), 8.30 - 8.19 (m, 1H), 7.10 (d, J = 0.9 Hz, 1H), 7.02 (d, J = 9.1 Hz, 1H) ), 6.72 (d, J = 1.8 Hz, 1H), 3.24 - 3.16 (m, 1H), 1.40 (d, J = 6.9 Hz, 6H).
步骤7.化合物57的合成。Step 7. Synthesis of Compound 57.
取500 mg化合物11、486 mg化合物56和669 mg三苯基膦溶于10 mL新蒸无水THF,-15℃下滴加552μL DIAD,并于-15℃搅拌5小时、自然升温室温搅拌过夜。冰盐浴下加入冰水淬灭反应,EA萃取水相,合并EA相,饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,柱层析得到485 mg化合物57。LC-MS(APCI):m/z=695.3(M+1)+。500 mg of compound 11, 486 mg of compound 56 and 669 mg of triphenylphosphine were dissolved in 10 mL of freshly distilled anhydrous THF, and 552 μL of DIAD was added dropwise at -15 ° C, and stirred at -15 ° C for 5 hours, and stirred at room temperature overnight. . The reaction was quenched by the addition of ice water, and the mixture was evaporated. LC-MS (APCI): m/z = 695.3 (M+1 ) .
步骤8.化合物58的合成。Step 8. Synthesis of Compound 58.
取473 mg化合物57,11 mg Hoveyda-Grubbs第一代催化剂溶于400 mL无水二氯乙烷,氮气脱气30分钟,75℃搅拌15小时。浓缩反应液,柱层析得到288 mg淡黄色固体即为化合物58。LC-MS(APCI):m/z=666.4(M+1)+。473 mg of compound 57, 11 mg of Hoveyda-Grubbs first generation catalyst was dissolved in 400 mL of anhydrous dichloroethane, degassed under nitrogen for 30 minutes, and stirred at 75 ° C for 15 hours. The reaction mixture was concentrated and purified by column chromatography to yield 288 g of pale yellow solid. LC-MS (APCI): m / z = 666.4 (M + 1) +.
步骤9.化合物59的合成。Step 9. Synthesis of Compound 59.
取250 mg化合物58溶于20 mL甲醇和30 mL四氢呋喃,冰水浴下加入2.89 g一水合氢氧化锂和15 mL水,室温搅拌过夜。1N盐酸调pH2~3,EA萃取,合并EA相并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得212 mg淡黄色色固体粉末,LC-MS(APCI):m/z=652.4(M+1)+。 250 mg of Compound 58 was dissolved in 20 mL of methanol and 30 mL of tetrahydrofuran, and 2.89 g of lithium hydroxide monohydrate and 15 mL of water were added under ice water, and stirred at room temperature overnight. 1N Hydrochloric acid was adjusted to pH 2~3, EA was extracted, EA phase was combined and washed with saturated brine, dried over anhydrous sodium sulfate, and filtered and evaporated to give a pale yellow solid powder, LC-MS (APCI): m/z=652.4 (M +1) + .
步骤10.化合物A-4的合成。Step 10. Synthesis of Compound A-4.
取200 mg化合物59溶于15 mL新蒸无水四氢呋喃,氮气保护下加入210 mg CDI,加热回流2小时。冷却至50℃后加入235 mg环丙基磺酰胺和189 mg DBU,50℃搅拌过夜。浓缩反应液,加入2 mL 1N盐酸和5mL二氯甲烷分液,有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,柱层析得168 mg淡黄色固体粉末即为化合物A-4。LC-MS(APCI):m/z=755.3(M+1)+。200 mg of compound 59 was dissolved in 15 mL of freshly distilled anhydrous tetrahydrofuran, 210 mg of CDI was added under nitrogen, and the mixture was heated under reflux for 2 hours. After cooling to 50 ° C, 235 mg of cyclopropylsulfonamide and 189 mg of DBU were added and stirred at 50 ° C overnight. The reaction mixture was concentrated, and 2 mL of 1N hydrochloric acid and 5 mL of dichloromethane was added, and the organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, and filtered and concentrated to give 168 mg of pale yellow solid powder as compound A-4 . LC-MS (APCI): m / z = 755.3 (M + 1) +.
实施例9.制备取代的喹啉化合物A-5。Example 9. Preparation of substituted quinoline compound A-5.
具体合成步骤如下:The specific synthesis steps are as follows:
步骤1.化合物60的合成。Step 1. Synthesis of Compound 60.
取500 mg化合物26、482 mg化合物47和664 mg三苯基膦溶于10 mL新蒸无水THF,-15℃下滴加552μL DIAD,并于-15℃搅拌5小时、自然升温室温搅拌过夜。冰盐浴下加入冰水淬灭反应,EA萃取水相,合并EA相,饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,柱层析得到541 mg化合物60。LC-MS(APCI):m/z=695.4(M+1)+。500 mg of compound 26, 482 mg of compound 47 and 664 mg of triphenylphosphine were dissolved in 10 mL of freshly distilled anhydrous THF, and 552 μL of DIAD was added dropwise at -15 ° C, and stirred at -15 ° C for 5 hours, and stirred at room temperature overnight. . The reaction was quenched by the addition of ice water, and the aqueous layer was evaporated. EtOAc was evaporated. LC-MS (APCI): m / z = 695.4 (M + 1) +.
步骤2.化合物61的合成。Step 2. Synthesis of Compound 61.
取534 mg化合物60,11 mg Hoveyda-Grubbs第一代催化剂溶于300 mL无水二氯乙烷,氮气脱气30分钟,75 C搅拌15小时。浓缩反应液,柱层析得到365 mg淡黄色固体即为化合物61。LC-MS(APCI):m/z=666.4(M+1)+。534 mg of compound 60, 11 mg of Hoveyda-Grubbs first generation catalyst was dissolved in 300 mL of anhydrous dichloroethane, degassed under nitrogen for 30 minutes, and stirred at 75 C for 15 hours. The reaction mixture was concentrated and purified by column chromatography to afford 365 g of pale yellow solid. LC-MS (APCI): m / z = 666.4 (M + 1) +.
步骤3.化合物62的合成。Step 3. Synthesis of Compound 62.
取689 mg化合物61溶于20 mL甲醇和30 mL四氢呋喃,冰水浴下加入2.89 g一水合氢氧化锂和15 mL水,室温搅拌过夜。1N盐酸调pH2~3,EA萃取,合并EA相并用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩得686 mg淡黄色色固体粉末,LC-MS(APCI):m/z=653.4(M+1)+。689 mg of Compound 61 was dissolved in 20 mL of methanol and 30 mL of tetrahydrofuran, and 2.89 g of lithium hydroxide monohydrate and 15 mL of water were added under ice water, and stirred at room temperature overnight. 1N Hydrochloric acid was adjusted to pH 2~3, EA was extracted, EA phase was combined and washed with saturated brine, dried over anhydrous sodium sulfate, and filtered to give 686 mg of pale yellow solid powder, LC-MS (APCI): m/z=653.4 (M +1) + .
步骤4.化合物A-5的合成。Step 4. Synthesis of Compound A-5.
取500 mg化合物62溶于15 mL新蒸无水四氢呋喃,氮气保护下加入520 mg CDI,加热回流2小时。冷却至50℃后加入515 mg环丙基磺酰胺和412 mg DBU,50℃搅拌过夜。浓缩反应液,加入2 mL 1N盐酸和5 mL二氯甲烷分液,有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤浓缩,柱层析得450 mg淡黄色固体粉末即为化合物A-5。LC-MS(APCI):m/z=755.3(M+1)+。500 mg of compound 62 was dissolved in 15 mL of freshly distilled anhydrous tetrahydrofuran, and 520 mg of CDI was added under a nitrogen atmosphere, and the mixture was heated under reflux for 2 hours. After cooling to 50 ° C, 515 mg of cyclopropylsulfonamide and 412 mg of DBU were added and stirred at 50 ° C overnight. The reaction mixture was concentrated, and 2 ml of 1N hydrochloric acid and 5 mL of dichloromethane was added. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate 5. LC-MS (APCI): m / z = 755.3 (M + 1) +.
生物活性评价。Evaluation of biological activity.
为了验证本文所述的化合物对HCV的作用,发明人采用HCV复制子系统(HCV Replicon System)作为评价模型。自Science1999年首次报道以来,HCV复制子系统已经成为研究HCV RNA复制、致病性和病毒持续性的最重要的工具之一,例如已经利用复制子成功地证明了HCV RNA复制所必须的5'-NCR最小区域,并且HCV复制子系统已经成功地被用作抗病毒药物的评价模型。本发明的发明人按照Science,1999,285(5424),110-3,以及J.Virol,2003,77(5),3007-19所描述的方法进行验证。To verify the effect of the compounds described herein on HCV, the inventors used the HCV Replicon System as an evaluation model. Since its first report in Science in 1999, the HCV replication system has become one of the most important tools for studying HCV RNA replication, pathogenicity and viral persistence, for example, the use of replicons has successfully demonstrated the 5' required for HCV RNA replication. - NCR minimum region, and the HCV replication subsystem has been successfully used as an evaluation model for antiviral drugs. The inventors of the present invention verified according to the methods described in Science, 1999, 285 (5424), 110-3, and J. Virol, 2003, 77(5), 3007-19.
(1)检测化合物抗HCV 1a和1b基因型复制子活性(1) Detection of compound anti-HCV 1a and 1b genotype replicon activity
应用HCV-1a和HCV-1b稳定转染复制子细胞检测化合物丙型肝炎病毒基因型1a和1b复制子的抑制活性。本实验将以NS3/4A抑制剂Simeprevir作为阳性对照化合物。 The inhibitory activities of the recombinant hepatitis C virus genotype 1a and 1b replicons were detected by stable transfection of replicon cells with HCV-1a and HCV-1b. This experiment will use the NS3/4A inhibitor Simeprevir as a positive control compound.
步骤一:对化合物进行1:3系列稀释8个浓度点,双复孔,加入96孔板中。设置DMSO为无加化合物对照。细胞培养液中的DMSO最终浓度为0.5%。Step 1: The compound was diluted 1:3 in 8 series points, double-replicated, and added to a 96-well plate. The DMSO was set to no compound control. The final concentration of DMSO in the cell culture was 0.5%.
步骤二:将HCV-1a和1b细胞分别悬浮在含10%FBS的培养液中,以每孔8,000个细胞的密度种到含有化合物的96孔板中。细胞在5%CO2、37℃条件下培养3天。Step 2: HCV-1a and 1b cells were separately suspended in a culture medium containing 10% FBS, and seeded into a 96-well plate containing the compound at a density of 8,000 cells per well. The cells were cultured for 3 days at 5% CO 2 at 37 °C.
步骤三:用CellTiter-Fluor(Promega)测定化合物对GT1b复制子细胞毒性。Step 3: The cytotoxicity of the compound against GT1b replicon was determined using CellTiter-Fluor (Promega).
步骤四:用Bright-Glo(Promega)检测荧光素酶测定化合物抗丙型肝炎病毒活性。Step 4: Detection of luciferase assay by Bright-Glo (Promega) for anti-hepatitis C virus activity.
步骤五:采用GraphPad Prism软件分析数据,拟合曲线并计算EC50值和CC50值。Step Five: using GraphPad Prism data analysis software, the curve fitting and EC 50 values were calculated and the 50 value CC.
对实施例1~5的化合物A-1至A-5按照如上步骤进行分析计算EC50值和CC50值,结果如表1所示。The compounds A-1 to A-5 of Examples 1 to 5 were analyzed in the above procedure to calculate EC 50 values and CC 50 values, and the results are shown in Table 1.
表1实施例化合物A-1至A-5与对照品Simeprevir的抗HCV基因型复制子活性对比表Table 1 Comparison of anti-HCV genotype replicon activities of the compounds of Examples A-1 to A-5 and the control Simeprevir
实验结果表明,本发明化合物对抑制HCV的多个基因型,进而能够用于丙肝病毒的抑制。与未氘代的化合物Simeprevir相比,本发明的化合物对GT1a和GT1b复制子表现出更优良的抑制活性。The experimental results show that the compounds of the present invention can inhibit the multiple genotypes of HCV and can be used for the inhibition of hepatitis C virus. The compound of the present invention exhibited more excellent inhibitory activity against the GT1a and GT1b replicons than the non-deuterated compound Simeprevir.
(2)测定化合物NS 3/4A抑制活性(2) Determination of compound NS 3/4A inhibitory activity
试验方法参见Anal.Biochem.373:1-8,2008和国际专利申请公开WO 2006/102087中所述的HCV NS3/4A蛋白酶时间分辨荧光(TRF)化验法。For the test method, the HCV NS3/4A protease time-resolved fluorescence (TRF) assay described in Anal. Biochem. 373: 1-8, 2008 and International Patent Application Publication No. WO 2006/102087.
如上表所示,实验结果表明,本发明化合物可作为HCV NS3/4A蛋白酶抑制剂,进而可用于抗丙肝病毒的药物中。As shown in the above table, the experimental results indicate that the compound of the present invention can be used as an inhibitor of HCV NS3/4A protease, and can be used in a drug against hepatitis C virus.
(3)代谢稳定性评价(3) Metabolic stability evaluation
微粒体实验:人肝微粒体:0.5mg/mL,Xenotech;大鼠肝微粒体:0.5mg/mL,Xenotech;辅酶(NADPH/NADH):1mM,Sigma Life Science;氯化镁:5mM,100 mM磷酸盐缓冲剂(pH为7.4)。Microsomal experiments: human liver microsomes: 0.5 mg/mL, Xenotech; rat liver microsomes: 0.5 mg/mL, Xenotech; coenzyme (NADPH/NADH): 1 mM, Sigma Life Science; magnesium chloride: 5 mM, 100 mM phosphate Buffer (pH 7.4).
储备液的配制:精密称取一定量的化合物实施例化合物A-1至A-5和阳性对照物Simeprevir的粉末,并用DMSO分别溶解至5mM。Preparation of Stock Solution: A certain amount of Compound Example Compounds A-1 to A-5 and the positive control Simeprevir powder were accurately weighed and dissolved to 5 mM with DMSO, respectively.
磷酸盐缓冲液(100mM,pH7.4)的配制:取预先配好的0.5M磷酸二氢钾150 mL和700mL的0.5M磷酸氢二钾溶液混合,再用0.5M磷酸氢二钾溶液调节混合液pH值至7.4,使用前用超纯水稀释5倍,加入氯化镁,得到磷酸盐缓冲液(100 mM),其中含100 mM磷酸钾,3.3 mM氯化镁,pH为7.4。Preparation of phosphate buffer (100 mM, pH 7.4): Mix 150 mL of pre-formed 0.5 M potassium dihydrogen phosphate and 700 mL of 0.5 M potassium dihydrogen phosphate solution, and adjust the mixture with 0.5 M potassium dihydrogen phosphate solution. The pH of the solution was adjusted to 7.4, diluted 5 times with ultrapure water before use, and magnesium chloride was added to obtain a phosphate buffer (100 mM) containing 100 mM potassium phosphate, 3.3 mM magnesium chloride, and a pH of 7.4.
配制NADPH再生系统溶液(含有6.5mM NADP,16.5mM G-6-P,3U/mL G-6-P D,3.3mM氯化镁),使用前置于湿冰上。A solution of NADPH regeneration system (containing 6.5 mM NADP, 16.5 mM G-6-P, 3 U/mL G-6-P D, 3.3 mM magnesium chloride) was prepared and placed on wet ice before use.
配制终止液:含有50ng/mL盐酸普萘洛尔和200ng/mL甲苯磺丁脲(内标)的乙腈溶液。取25057.5μL磷酸盐缓冲液(pH7.4)至50 mL离心管中,分别加入812.5μL人肝微粒体,混匀,得到蛋白浓度为0.625 mg/mL的肝微粒体稀释液。取25057.5μL磷酸盐缓冲液(pH7.4)至50 mL离心管中,分别加入812.5μL SD大鼠肝微粒体,混匀,得到蛋白浓度为0.625 mg/mL的肝微粒体稀释液。Formulation stop solution: acetonitrile solution containing 50 ng/mL propranolol hydrochloride and 200 ng/mL tolbutamide (internal standard). Take 25057.5 μL of phosphate buffer (pH 7.4) into a 50 mL centrifuge tube, add 812.5 μL of human liver microsomes, and mix to obtain a liver microsome dilution with a protein concentration of 0.625 mg/mL. Take 25057.5 μL of phosphate buffer (pH 7.4) into a 50 mL centrifuge tube, add 812.5 μL SD rat liver microsomes, and mix to obtain a liver microsome dilution with a protein concentration of 0.625 mg/mL.
样品的孵育:用含70%乙腈的水溶液将相应化合物的储备液分别稀释至0.25 mM,作为工作液,备用。分别取398μL的人肝微粒体或者大鼠肝微粒体稀释液加入96孔孵育板中(N=2),分别加入2μL 0.25 mM的的工作液中,混匀。Incubation of the sample: The stock solution of the corresponding compound was diluted to 0.25 mM with an aqueous solution containing 70% acetonitrile, and used as a working solution. 398 μL of human liver microsomes or rat liver microsome dilutions were added to 96-well incubation plates (N=2), and 2 μL of 0.25 mM working solution was added and mixed.
代谢稳定性的测定:在96孔深孔板的每孔中加入300μL预冷的终止液,并置于冰上,作为终止板。将96孔孵育板和NADPH再生系统置于37℃水浴箱中,100转/分钟震荡,预孵5 min。从孵育板每孔取出80μL孵育液加入终止板,混匀,补充20μL NADPH再生系统溶液,作为0 min样品。再向孵育板每孔加入80μL的NADPH再生系统溶液,启动反应,开始计时。相应化合物的反应浓度为1μM,蛋白浓度为0.5 mg/mL。分别于反应10、30、90 min时,各取100μL反应液,加入终止板中,涡旋3 min终止反应。将终止板于5000×g,4℃条件下离心10 min。取100μL上清液至预先加入100μL蒸馏水的96孔板中,混匀,采用LC-MS/MS进行样品分析。Determination of metabolic stability: 300 μL of pre-cooled stop solution was added to each well of a 96-well deep well plate and placed on ice as a stop plate. The 96-well incubation plate and the NADPH regeneration system were placed in a 37 ° C water bath, shaken at 100 rpm, and pre-incubated for 5 min. 80 μL of the incubation solution was taken from each well of the incubation plate, added to the stopper plate, and mixed, and 20 μL of the NADPH regeneration system solution was added as a 0 min sample. Then, 80 μL of the NADPH regeneration system solution was added to each well of the incubation plate to start the reaction and start timing. The corresponding compound had a reaction concentration of 1 μM and a protein concentration of 0.5 mg/mL. 100 μL of the reaction solution was taken at 10, 30, and 90 min, respectively, and added to the stopper plate, and the reaction was terminated by vortexing for 3 min. The plate was centrifuged at 5000 x g for 10 min at 4 °C. 100 μL of the supernatant was taken into a 96-well plate to which 100 μL of distilled water was previously added, mixed, and sample analysis was performed by LC-MS/MS.
数据分析:通过LC-MS/MS系统检测相应化合物及内标的峰面积,计算化合物与内标峰面积 比值。通过化合物剩余量的百分率的自然对数与时间作图测得斜率,并根据以下公式计算t1/2和CLint,其中V/M即等于1/蛋白浓度。Data analysis: The peak area of the corresponding compound and the internal standard was detected by LC-MS/MS system, and the ratio of the peak area of the compound to the internal standard was calculated. The slope is measured by the natural logarithm of the percentage of the remaining amount of the compound versus time, and t 1/2 and CL int are calculated according to the following formula, where V/M is equal to 1/protein concentration.
对本发明化合物及其没有氘代的化合物同时测验比较,评价其在人和大鼠肝微粒体的代谢稳定性。作为代谢稳定性的指标的半衰期及肝固有清除率如表2所示。表2中采用未经氘代的化合物Simeprevir作为对照样品。如表2所示,通过与未经氘代的化合物Simeprevir对照,本发明化合物可以显著改善代谢稳定性,进而更适于作为丙型肝炎病毒抑制剂。The metabolic stability of human and rat liver microsomes was evaluated by simultaneously testing the compounds of the present invention and their compounds without deuteration. The half-life and liver intrinsic clearance as indicators of metabolic stability are shown in Table 2. The undeuterated compound Simeprevir was used as a control sample in Table 2. As shown in Table 2, the compound of the present invention can significantly improve metabolic stability by comparison with the undeuterated compound Simeprevir, and is thus more suitable as a hepatitis C virus inhibitor.
表2实施例化合物A-1至A-5与Simeprevir对照样的代谢稳定性对比表Table 2 Comparison of Metabolic Stability of Example Compounds A-1 to A-5 with Simeprevir Control
对实施例化合物按照上述步骤进行分析,实验结果表明,本发明化合物在人肝微粒体与大鼠肝微粒体实验中都表现出优异的代谢稳定性。The compounds of the examples were analyzed according to the above procedure, and the results of the experiments showed that the compounds of the present invention exhibited excellent metabolic stability in both human liver microsomes and rat liver microsomes.
(4)大鼠药代动力学实验(4) Rat pharmacokinetic experiments
实验目的:研究大鼠给予Simeprevir、实施例化合物后,考察本发明化合物的药代动力学行为。EXPERIMENTAL OBJECTIVE: To investigate the pharmacokinetic behavior of the compounds of the present invention after administration of Simeprevir to the compounds of the examples.
实验动物:Experimental animals:
种类及品系:SD大鼠等级:SPF级Type and strain: SD rat grade: SPF grade
性别及数量:雄性,6只Gender and quantity: male, 6
体重范围:180~220g(实际体重范围为187~197g)Weight range: 180 ~ 220g (actual weight range is 187 ~ 197g)
来源:上海西普尔必凯实验动物有限公司Source: Shanghai Xipuer Bikai Experimental Animal Co., Ltd.
实验及动物合格证号:SCXK(沪)2013-0016。 Laboratory and animal certificate number: SCXK (Shanghai) 2013-0016.
实验过程:experiment procedure:
在血样采集之前,预先在EDTA-K2抗凝管中加入20 L的2M氟化钠溶液(酯酶抑制剂),于80度烘箱内烘干后,置于4度冰箱存放。Before the blood sample was collected, 20 L of 2M sodium fluoride solution (esterase inhibitor) was added to the EDTA-K2 anticoagulant tube, dried in an 80 degree oven, and stored in a 4 degree refrigerator.
大鼠,雄性,体重187~197 g,随机分为2组,于实验前一天下午开始禁食过夜但可自由饮水,给药后4 h给食物。A组给予Simeprevir 3mg/kg,B组给予式实施例化合物A-1至A-53mg/kg,分别于给药后15 min、30 min、1、2、3、5、8、10 h从大鼠眼眶静脉取血100-200 L左右,置于经EDTA-K2抗凝的0.5 mL的Eppendorf管中,立即混匀,抗凝后,尽快将试管轻轻颠倒混匀5-6次后,血取好后放置在冰盒中,30 min内把血样本在4000 rpm,10 min,4℃条件下离心分离血浆,收集全部血浆后立即于-20℃保存。所有时间点样品采集后测定每个时间点的血浆中的血药浓度。Rats, males, weighing 187-197 g, were randomly divided into 2 groups. They were fasted overnight in the afternoon before the experiment but were free to drink water and given food 4 h after administration. Group A was given 3 mg/kg of Simeprevir, and group B was given compound A-1 to A-53 mg/kg of the formula, respectively, from 15 min, 30 min, 1, 2, 3, 5, 8, 10 h after administration. The mouse eye vein was taken from a blood of about 100-200 L, placed in an EDTA-K2 anticoagulated 0.5 mL Eppendorf tube, and immediately mixed. After anticoagulation, the tube was gently inverted and mixed 5-6 times as soon as possible. After taking it, place it in an ice box, centrifuge the blood sample at 4000 rpm, 10 min, 4 °C for 30 min, collect all the plasma and store at -20 °C immediately. Plasma concentrations in plasma at each time point were determined after sample collection at all time points.
根据上述所得的给药后平均血药浓度-时间数据,采用Winnonin软件,按非房室统计矩理论求算雄性SD大鼠分别i.g给予Simeprevir(3mg/kg)、实施例化合物A-1至A-5(3mg/kg)后的药代动力学相关参数。According to the average blood drug concentration-time data obtained after the above, male Sprague-Dawley rats were administered ig to Simeprevir (3 mg/kg) and Example Compounds A-1 to A according to the non-compartmental statistical moment theory using Winnonin software. Pharmacokinetic related parameters after -5 (3 mg/kg).
实验结果表明,与Simeprevir相比,本发明人发现化合物具有比Simeprevir更优的活性,并且具有优异的药代动力学性质,因此更适合作为抑制丙型肝炎病毒蛋白NS3/4A的化合物,进而适合制备治疗丙型肝炎病毒感染的药物。The experimental results show that compared with Simeprevir, the present inventors have found that the compound has superior activity than Simeprevir and has excellent pharmacokinetic properties, and thus is more suitable as a compound for inhibiting the hepatitis C virus protein NS3/4A, and is suitable for the compound. Preparation of a medicament for treating hepatitis C virus infection.
应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围,实施例中未注明具体条件的实验方法,通常按照常规条件,或按照制造厂商所建议的条件。除非另外说明,否则份数和百分比为重量份和重量百分比。It is to be understood that the examples are merely illustrative of the invention and are not intended to limit the scope of the invention, and the experimental methods in which the specific conditions are not indicated in the examples, usually in accordance with conventional conditions or in accordance with the conditions suggested by the manufacturer. Parts and percentages are parts by weight and percentage by weight unless otherwise stated.
以上内容是结合具体的优选实施方式对本发明所作的进一步详细说明,不能认定本发明的具体实施只局限于这些说明。对于本发明所属技术领域的普通技术人员来说,在不脱离本发明构思的前提下,还可以做出若干简单推演或替换,都应当视为属于本发明的保护范围。 The above is a further detailed description of the present invention in connection with the specific preferred embodiments, and the specific embodiments of the present invention are not limited to the description. It will be apparent to those skilled in the art that the present invention may be made without departing from the spirit and scope of the invention.
Claims (9)
根据权利要求1所述的丙型肝炎病毒抑制剂,其特征在于:所述化合物选自下述化合物或其药学上可接受的盐:
[Correct according to Rule 26 14.09.2017]
The hepatitis C virus inhibitor according to claim 1, wherein the compound is selected from the group consisting of the following compounds or a pharmaceutically acceptable salt thereof:
一种药物组合物,其特征在于:其含有药学上可接受的载体和如权利要求1~6任意一项所述的喹啉化合物,或其晶型、药学上可接受的盐、水合物或溶剂合物、立体异构体、前药或同位素变体的药物组合物。 [Correct according to Rule 26 14.09.2017]
A pharmaceutical composition comprising a pharmaceutically acceptable carrier and a quinoline compound according to any one of claims 1 to 6, or a crystalline form thereof, a pharmaceutically acceptable salt, a hydrate or A pharmaceutical composition of a solvate, stereoisomer, prodrug or isotopic variation.
根据权利要求7所述的药物组合物,其特征在于:其还包含其他活性化合物,所述活性化合物可选自HCV蛋白酶抑制剂、HCV NS5A抑制剂和HCV NS5B聚合酶抑制剂。 [Correct according to Rule 26 14.09.2017]
The pharmaceutical composition according to claim 7, characterized in that it further comprises other active compounds which may be selected from the group consisting of HCV protease inhibitors, HCV NS5A inhibitors and HCV NS5B polymerase inhibitors.
一种如权利要求1~6任意一项所述的喹啉化合物的用途,其特征在于:用于制备治疗丙型肝炎病毒感染的药物中的用途。 [Correct according to Rule 26 14.09.2017]
Use of a quinoline compound according to any one of claims 1 to 6 for use in the manufacture of a medicament for the treatment of hepatitis C virus infection.
根据权利要求7和8所述任一项的药物组合物的用途,其特征在于:用于制备用于在需要其的对象中抑制HCV NS3/4A蛋白酶活性的药物的用途。 [Correct according to Rule 26 14.09.2017]
Use of a pharmaceutical composition according to any one of claims 7 and 8, characterized in that it is used for the preparation of a medicament for inhibiting the activity of HCV NS3/4A protease in a subject in need thereof.
Priority Applications (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| CN201780004823.3A CN108368105B (en) | 2016-08-12 | 2017-08-07 | Substituted quinoline compound, and pharmaceutical composition and application thereof |
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| CN201610662129.8 | 2016-08-12 | ||
| CN201610662129 | 2016-08-12 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2018028541A1 true WO2018028541A1 (en) | 2018-02-15 |
Family
ID=61161778
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/CN2017/096198 Ceased WO2018028541A1 (en) | 2016-08-12 | 2017-08-07 | Substituted quinoline compound and pharmaceutical composition and use thereof |
Country Status (2)
| Country | Link |
|---|---|
| CN (1) | CN108368105B (en) |
| WO (1) | WO2018028541A1 (en) |
Cited By (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN117003697A (en) * | 2023-07-04 | 2023-11-07 | 武汉大学 | A kind of quinoline compound targeting angiotensin-converting enzyme 2 receptor and its preparation method and application |
Citations (2)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN101228169A (en) * | 2005-07-29 | 2008-07-23 | 泰博特克药品有限公司 | Macrocyclic Inhibitors of Hepatitis C Virus |
| WO2016089814A1 (en) * | 2014-12-02 | 2016-06-09 | Concert Pharmaceuticals, Inc. | Deuterated analogues of daclatasvir |
-
2017
- 2017-08-07 CN CN201780004823.3A patent/CN108368105B/en active Active
- 2017-08-07 WO PCT/CN2017/096198 patent/WO2018028541A1/en not_active Ceased
Patent Citations (2)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN101228169A (en) * | 2005-07-29 | 2008-07-23 | 泰博特克药品有限公司 | Macrocyclic Inhibitors of Hepatitis C Virus |
| WO2016089814A1 (en) * | 2014-12-02 | 2016-06-09 | Concert Pharmaceuticals, Inc. | Deuterated analogues of daclatasvir |
Non-Patent Citations (2)
| Title |
|---|
| HARBESON, S. L. ET AL.: "Deuterium Medicinal Chemistry: A New Approach to Drug Discovery and Development", MEDCHEM NEWS N.2, May 2014 (2014-05-01), pages 8 - 22, XP055246294, Retrieved from the Internet <URL:https://www.concertpharma.com/wp-content/uploads/2014/12/MedChemNews-0514.pdf> * |
| RABOISSON, P. ET AL.: "Structure - activity relationship study on a novel se- ries of cyclopentane-containing macrocyclic inhibitors of the hepatitis C virus NS3/4A protease leading to the discovery of TMC435350", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 18, no. 17, 24 July 2008 (2008-07-24), pages 4853 - 4858, XP029441726 * |
Cited By (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| CN117003697A (en) * | 2023-07-04 | 2023-11-07 | 武汉大学 | A kind of quinoline compound targeting angiotensin-converting enzyme 2 receptor and its preparation method and application |
Also Published As
| Publication number | Publication date |
|---|---|
| CN108368105B (en) | 2021-06-29 |
| CN108368105A (en) | 2018-08-03 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| US7091184B2 (en) | Hepatitis C inhibitor tri-peptides | |
| US7119072B2 (en) | Macrocyclic peptides active against the hepatitis C virus | |
| JP4060801B2 (en) | Tripeptides having hydroxyproline ethers of substituted quinolines for the suppression of NS3 (hepatitis C) | |
| JP4040578B2 (en) | Macrocyclic peptide active against hepatitis C virus | |
| EP1474423B1 (en) | Heterocyclic tripeptides as hepatitis c inhibitors | |
| EP1910378B1 (en) | Hepatitis c inhibitor peptide analogs | |
| US20030187018A1 (en) | Hepatitis C inhibitor tri-peptides | |
| TW201216979A (en) | Macrocyclic proline derived HCV serine protease inhibitors | |
| AU2003202348A1 (en) | Heterocyclic tripeptides as hepatitis c inhibitors | |
| AU2003202347A1 (en) | Tripeptides having a hydroxyproline ether of a substituted quinoline for the inhibition of NS3 (Hepatitis C) | |
| TW201019950A (en) | Therapeutic antiviral peptides | |
| WO2018028541A1 (en) | Substituted quinoline compound and pharmaceutical composition and use thereof | |
| WO2017121188A1 (en) | Hepatitis c virus inhibitor, pharmaceutical composition and application thereof | |
| WO2018006811A1 (en) | Substituted imidazolyl compound and pharmaceutical composition thereof | |
| CN108368130B (en) | Substituted macrocyclic quinoxaline compound, pharmaceutical composition and application thereof | |
| WO2017121187A1 (en) | Hepatitis c virus inhibitor, pharmaceutical composition and use thereof | |
| JP2016510746A (en) | Compounds for the treatment of hepatitis C | |
| CN108290844B (en) | A kind of substituted naphthalene ring compound and pharmaceutical composition and application thereof | |
| CA2474031C (en) | Heterocyclic tripeptides as hepatitis c inhibitors | |
| WO2017206688A1 (en) | Substituted pyrrolidine compound, pharmaceutical composition, and applications thereof | |
| WO2017181947A1 (en) | Substituted diamine carbamate, and pharmaceutical composition and application thereof | |
| HK1078884A (en) | Macrocyclic peptides active against the hepatitis c virus |
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: 17838671 Country of ref document: EP Kind code of ref document: A1 |
|
| NENP | Non-entry into the national phase |
Ref country code: DE |
|
| 122 | Ep: pct application non-entry in european phase |
Ref document number: 17838671 Country of ref document: EP Kind code of ref document: A1 |