CN108368105B - Substituted quinoline compound, and pharmaceutical composition and application thereof - Google Patents

Substituted quinoline compound, and pharmaceutical composition and application thereof Download PDF

Info

Publication number
CN108368105B
CN108368105B CN201780004823.3A CN201780004823A CN108368105B CN 108368105 B CN108368105 B CN 108368105B CN 201780004823 A CN201780004823 A CN 201780004823A CN 108368105 B CN108368105 B CN 108368105B
Authority
CN
China
Prior art keywords
compound
solution
added
synthesis
deuterium
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.)
Active
Application number
CN201780004823.3A
Other languages
Chinese (zh)
Other versions
CN108368105A (en
Inventor
王义汉
金剑
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shenzhen Targetrx Inc
Original Assignee
Shenzhen Targetrx Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shenzhen Targetrx Inc filed Critical Shenzhen Targetrx Inc
Publication of CN108368105A publication Critical patent/CN108368105A/en
Application granted granted Critical
Publication of CN108368105B publication Critical patent/CN108368105B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B59/00Introduction of isotopes of elements into organic compounds ; Labelled organic compounds per se
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic 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
    • C07D417/14Heterocyclic 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

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

Provided are a substituted quinoline compound, a pharmaceutical composition and an application thereof, wherein the quinoline compound is a compound shown as a formula (I), or a crystal form, a pharmaceutically acceptable salt, a hydrate or a solvate thereof. The compound can be used as a hepatitis C virus inhibitor, has better hepatitis C virus protein NS3/4A inhibition activity, better pharmacodynamics/pharmacokinetic performance, good applicability and high safety, can be used for preparing a medicament for treating hepatitis C virus infection, and has good market development prospect.

Description

Substituted quinoline compound, and pharmaceutical composition and application thereof
Technical Field
The invention belongs to the technical field of medicines, and particularly relates to a hepatitis C virus inhibitor, a pharmaceutical composition and application thereof.
Background
Hepatitis c is a blood-borne infectious liver disease that can cause significant damage to the liver if not treated in time. About 1.85 million people all over the world infect hepatitis C virus, the hepatitis C infection rate in China is about 3.2%, and the number of patients is about forty million; in the united states, about 320 million patients with hepatitis c die of the disease, about 1.5 million people per year, and much more of the disease is related to hepatitis c, such as cirrhosis and liver cancer.
HCV (Hepatitis C Virus) is an RNA Virus belonging to the genus Hepatitis C Virus (Hepacivirus genus) in the family Flaviviridae (Flaviviridae family). The encapsulated HCV virions comprise a positive strand 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 about 3000 amino acids. The polyprotein includes the core protein, the envelope proteins E1 and E2, the membrane bound protein P7, and the non-structural proteins NS2, NS3/4A, NS4A, NS4B, NS5A, and NS 5B.
Sofosbuvir is the first good medicine in the world at present and can completely cure hepatitis C in a short period. It directly reaches the focus by oral administration, has simple method and little side effect, and is deeply touted by patients. Sofosbuvir is produced by Gilidide, USA, is marketed in 2013, and can effectively treat type-1, type-2, type-3 and type-4 hepatitis C through clinical tests, including clinical tests on liver transplantation, liver cancer and HCV/HIV-1 combined infection. The breakthrough brings good news to hepatitis C patients all over the world.
HCV infection is associated with progressive liver disease symptoms, including cirrhosis and hepatocellular carcinoma. Simeprevir (Simeprevir) is a new generation of NS3/4A protease inhibitor, a once-daily oral drug, developed by Medivir corporation in combination with yanssen (Janssen) for the treatment of compensatory liver diseases including liver fibrosis at various stages in adult patients with chronic hepatitis c, and works by blocking the replication of HCV in liver cells by proteases. In 2013, 1 month and 22 days, a new hepatitis C drug, namely simetivir, is approved by FDA, and is combined with polyethylene glycol interferon and ribavirin (ribavirin) to be used for treating compensatory liver diseases (including liver cirrhosis) of adult patients with genotype 1 chronic hepatitis C.
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 NS 3/4A.
Disclosure of Invention
Aiming at the technical problems, the invention discloses a hepatitis C virus inhibitor, a pharmaceutical composition and application thereof, wherein the hepatitis C virus inhibitor has better hepatitis C virus protein NS3/4A inhibition activity and/or better pharmacodynamic/pharmacokinetic performance.
In contrast, the technical scheme adopted by the invention is as follows:
a hepatitis C virus inhibitor is a quinoline compound shown in formula (I) or a crystal form, a pharmaceutically acceptable salt, a hydrate or a solvent compound thereof,
Figure GPA0000258315050000031
wherein R is1、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、R45Each independently is hydrogen, deuterium, halogen or trifluoromethyl;
with the proviso that 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、R44And R45At least one of which is deuterated.
By adopting the technical scheme, the shape and the volume of deuterium in a drug molecule are basically the same as those of hydrogen, and if the hydrogen in the drug molecule is selectively replaced by deuterium, the original biological activity and selectivity of the deuterium-substituted drug can be generally kept. Meanwhile, the inventor proves that the combination of carbon and deuterium bonds is more stable than the combination of carbon and hydrogen bonds, and the absorption, distribution, metabolism, excretion and other properties of some medicines can be directly influenced, so that the curative effect, safety and tolerance of the medicines are improved.
Preferably, the deuterium isotope content of deuterium at the deuterated position is at least greater than the natural deuterium isotope content (0.015%), preferably greater than 30%, more preferably greater than 50%, more preferably greater than 75%, more preferably greater than 95%, more preferably greater than 99%.
Specifically, in the present invention 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、R44And R45The deuterium isotope content in each deuterated position is at least 5%, preferably greater than 10%, more preferably greater than 15%, more preferably greater than 20%, more preferably greater than 25%, more preferably greater than 30%, more preferably greater than 35%, more preferably greater than 40%, more preferably greater than 45%, more preferably greater than 50%, more preferably greater than 55%, more preferably greater than 60%, more preferably greater than 65%, more preferably greater than 70%, more preferably greater than 75%, more preferably greater than 80%, more preferably greater than 85%, more preferably greater than 90%, more preferably greater than 95%, more preferably greater than 99%.
Preferably, R of the compound of formula (I)1、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、R44And R45Preferably, at least one of R comprises deuterium, more preferably two of R comprises deuterium, more preferably three of R comprises deuterium, more preferably four of R comprises deuterium, more preferably five of R comprises deuterium, more preferably six of R comprises deuterium, more preferably seven of R comprises deuterium, more preferably eight of R comprises deuterium, more preferably nine of R comprises deuterium, more preferably ten of R comprises deuterium, more preferably eleven of R comprises deuterium, more preferably twelve of R comprises deuterium, more preferably thirteen of R comprises deuterium, more preferably fourteen of R comprises deuterium, more preferably fifteen of R comprises deuterium, more preferably sixteen of R comprises deuterium, more preferably seventeen of R comprises deuterium, more preferably eighteen of R comprises deuterium, more preferably nineteen of R comprises deuterium, more preferably twenty one of R comprises deuteriumMore preferably twenty two R contain deuterium, more preferably twenty three R contain deuterium, more preferably twenty four R contain deuterium, more preferably twenty five R contain deuterium, more preferably twenty six R contain deuterium, more preferably twenty seven R contain deuterium, more preferably twenty eight R contain deuterium, more preferably twenty nine R contain deuterium, more preferably thirty one R contain deuterium, more preferably thirty two R contain deuterium, more preferably thirty three R contain deuterium, more preferably thirty four R contain deuterium, more preferably thirty five R contain deuterium, more preferably thirty six R contain deuterium, more preferably thirty seven R contain deuterium, more preferably thirty eight R contain deuterium, more preferably thirty nine R contain deuterium, more preferably forty four R contain deuterium, more preferably forty one R contain deuterium, more preferably forty two R contain deuterium, more preferably forty three R contain deuterium, more preferably forty four R contain deuterium, more preferably five R contain deuterium.
As a further improvement of the invention, R19、R20And R21Each independently is deuterium or hydrogen.
In another preferred embodiment, R19、R20、R21Is deuterium.
As a further improvement of the invention, R32、R33And R34Each independently is deuterium or hydrogen.
In another preferred embodiment, R32、R33、R34Is deuterium.
As a further improvement of the invention, R35、R36And R37Each independently is deuterium or hydrogen.
In another preferred embodiment, R35、R36、R37Is deuterium.
As a further improvement of the present invention, the compound is selected from the following compounds or pharmaceutically acceptable salts thereof:
Figure GPA0000258315050000041
Figure GPA0000258315050000051
Figure GPA0000258315050000061
Figure GPA0000258315050000071
in another preferred embodiment, the compound does not include non-deuterated compounds.
The invention also discloses a pharmaceutical composition which contains a pharmaceutically acceptable carrier and the hepatitis C virus inhibitor, or a pharmaceutical composition of a crystal form, a pharmaceutically acceptable salt, a hydrate or a solvate, a stereoisomer, a prodrug or an isotopic variant thereof.
As a further improvement of the present invention, it also comprises other active compounds including, but not limited to, other HCV antivirals, anti-infectives, immunomodulators, antibiotics or vaccine combinations.
As a further improvement of the invention, the immunomodulator is an interferon drug compound.
As a further improvement of the invention, the quinoxaline macrocyclic compounds of the invention may be used in combination therapy 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 generally enhance immune system function and those that generate specific immune responses against HCV. Additional therapeutic agents targeting HCV include agents targeting NS3/4A and agents targeting other HCV activities, such as NS5A and NS5B, and agents targeting host cell activities involved in HCV replication.
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- α 2a (e.g., Roferon interferon available from Hoffmann-LaRoche, Nutley, NJ), pegylated interferon- α 2a (pegasys), interferon- α 2b (e.g., Intron-a interferon available from Schering corp., Kenilworth, NJ), pegylated interferon- α 2b (pegintron), recombinant consensus interferon (e.g., interferon alphacon-1), and purified interferon- α products. The individual components of the combination may be administered separately at different times during the course of therapy or concurrently in divided or single combination forms.
For the treatment of HCV infection, the compounds of the present invention may also be administered in combination with the antiviral agent amantadine (1-aminoadamantane). For a complete description of this agent, see J.Kirschbaum, 12 anal. profiles Drug Subs.1-36 (1983).
For the treatment of HCV infection, the compounds of the present invention may also be administered in combination with the antiviral agent polymerase inhibitor R7128 (Roche).
For the treatment of HCV infection, the compounds of the present invention may also be administered in combination with an HCV NS5B polymerase inhibitor. Such HCV NS5B polymerase inhibitors that may be used as combination therapies include, but are not limited to, international patent application publications WO 02/057287, WO 02/057425, WO 03/068244, WO 2004/000858, WO 04/003138, and WO 2004/007512; U.S. patent No.6,777,392 and U.S. patent application publication US2004/0067901 (the contents of each of which are incorporated herein by reference in their entirety). Other such HCV polymerase inhibitors include, but are not limited to, valopicitabine (NM-283; Idenix) and 2 '-F-2' - β -methylcytidine (see also WO 2005/003147).
As a further improvement of the invention, the pharmaceutically acceptable carrier comprises at least one of a glidant, a sweetener, a diluent, a preservative, a dye/colorant, a flavor enhancer, a surfactant, a wetting agent, a dispersing agent, a disintegrant, a suspending agent, a stabilizer, an isotonic agent, a solvent, or an emulsifier.
As a further improvement of the present invention, the pharmaceutical composition is a tablet, pill, capsule, powder, granule, paste, emulsion, suspension, solution, suppository, injection, inhalant, gel, microsphere or aerosol.
Typical routes of administration of the pharmaceutical compositions of the present 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 compositions of the present invention may be manufactured by methods well known in the art, such as conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, lyophilizing, and the like.
The present invention also provides a method of preparing a pharmaceutical composition comprising the steps of: mixing a pharmaceutically acceptable carrier and the hepatitis C virus inhibitor or the crystal form, the pharmaceutically acceptable salt, the hydrate or the solvate thereof to form the pharmaceutical composition.
The invention also discloses an application of the hepatitis C virus inhibitor, which is characterized in that: the application of the compound in preparing the medicine for treating hepatitis C virus infection.
The NS3/4A inhibitor may also be used 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 determine binding sites for other antiviral agents to HCV protease, for example, by competitive inhibition.
The compounds of the present invention, optionally in salt form, may be administered by contacting the active agent with the site of action of a drug for the purpose of inhibiting the HCV NS3/4A protease and treating HCV infection and/or reducing the likelihood or severity of symptoms of HCV infection. They may be administered as separate therapeutic agents or combinations of therapeutic agents by conventional means which may be used in conjunction with a drug. They may be administered alone, but will generally be administered with a pharmaceutical carrier selected in accordance with the chosen route of administration and standard pharmaceutical practice.
The HCV includes its various genotypes and various gene subtypes, e.g. 1a, 1b, 2a, 2b, 3a, 3b, 4a, 5a, 6 a.
It is to be understood that within the scope of the present invention, the above-described features of the present invention and those specifically described below (e.g., in the examples) may be combined with each other to form new or preferred embodiments. Not to be reiterated herein, but to the extent of space.
Herein, "halogen" means F, Cl, Br, and I, unless otherwise specified. More preferably, the halogen atom is selected from F, Cl and Br.
Herein, "deuterated", unless otherwise specified, means that one or more hydrogens of a compound or group are replaced with deuterium; deuterium can be mono-, di-, poly-, or fully substituted. The terms "deuterated one or more" and "deuterated one or more" are used interchangeably.
Herein, unless otherwise specified, "non-deuterated compound" means a compound containing deuterium at an atomic ratio of deuterium not higher than the natural deuterium isotope content (0.015%).
Pharmaceutically acceptable salts include inorganic and organic salts. One preferred class of salts is that formed by reacting a compound of the present invention with an acid. Suitable acids for forming the salts include, but are not limited to: inorganic acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid, and the like; organic acids such as formic acid, acetic acid, trifluoroacetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, 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 and the like; and amino acids such as proline, phenylalanine, aspartic acid, glutamic acid, etc. Another preferred class of salts are those of the compounds of the invention with bases, for example alkali metal salts (e.g., sodium or potassium salts), alkaline earth metal salts (e.g., magnesium or calcium salts), ammonium salts (e.g., lower alkanolammonium salts and other pharmaceutically acceptable amine salts), for example methylamine salts, ethylamine salts, propylamine salts, dimethylamine salts, trimethylamine salts, diethylamine salts, triethylamine salts, tert-butylamine salts, ethylenediamine salts, hydroxyethylamine salts, dihydroxyethylamine salts, triethanolamine salts, and amine salts formed from morpholine, piperazine, lysine, respectively.
The term "solvate" refers to a complex of a compound of the present invention coordinated to solvent molecules in a specific ratio. "hydrate" refers to a complex formed by the coordination of a compound of the present invention with water.
Compared with the prior art, the invention has the beneficial effects that: first, the compounds of the present invention have excellent inhibitory activity against the hepatitis C virus protein NS 3/4A. Second, the metabolism of compounds in organisms is altered by deuteration, which results in compounds with better pharmacokinetic parameters. In this case, the dosage can be varied and a long acting formulation formed, improving the applicability. Thirdly, deuterium is used for replacing hydrogen atoms in the compound, and due to the deuterium isotope effect, the medicine concentration of the compound in an animal body is improved, and the medicine curative effect is improved. Fourthly, the replacement of hydrogen atoms in the compound by deuterium can inhibit certain metabolites and improve the safety of the compound.
Detailed Description
The following describes more specifically the processes for the preparation of the compounds of formula (I) according to the invention, but these particular processes do not constitute any limitation of the invention. The compounds of the present invention may also be conveniently prepared by optionally combining various synthetic methods described in the present specification or known in the art, and such combinations may be readily carried out by those skilled in the art to which the present invention pertains.
In general, in the preparative schemes, each reaction is usually carried out in an inert solvent at a temperature ranging from room temperature to reflux temperature (e.g., from 0 ℃ to 100 ℃, preferably from 0 ℃ to 80 ℃). The reaction time is usually 0.1 to 60 hours, preferably 0.5 to 24 hours.
Example 1 synthesis of intermediate 5.
Figure GPA0000258315050000101
Step 1. Synthesis of Compound 2.
2.68g of benzyl bromide is dissolved in 100mL of acetonitrile, 4.19g of cinchonidine is added under nitrogen protection, and the mixture is stirred overnight in the dark. Filtration was carried out, and the residue was washed three times with acetonitrile, recrystallized from methylene chloride and petroleum ether, and vacuum-dried to obtain 4.22g of a pale red solid powder.
And 2, synthesizing a compound 5.
10g of glycine methyl ester hydrochloride and anhydrous sodium sulfate were dispersed in 200mL of methyl t-butyl ether, followed by addition of benzaldehyde and triethylamine, and stirred at room temperature overnight. Filtered and concentrated to give an oily liquid which was used directly in the next step.
435mg of Compound 2 was dispersed in 14mL of toluene, and 3.98g of the above oily liquid in toluene (16mL) was added dropwise in an ice-water bath, followed by reaction at room temperature for 15 minutes. A solution of 4.0g of 1, 4-dibromo-2-butene in toluene (16mL) was slowly added dropwise thereto, and the mixture was stirred in an ice-water bath for 45 minutes. Slowly adding 20% NaOH solution dropwise, and stirring at 10 ℃ overnight after finishing the addition. The mixture was allowed to stand for liquid separation, and the organic phase was filtered, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to obtain 3.14g of a pale yellow oily liquid.1H NMR(400MHz,CDCl3)δ5.71(ddd,J=17.1,10.3,9.2Hz,1H),5.22(ddd,J=17.2,1.9,0.7Hz,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=7.5,4.8Hz,1H),1.35(dd,J=9.3,4.7Hz,1H)。
Example 2 synthesis of intermediate 11.
Figure GPA0000258315050000102
Step 1. Synthesis of Compound 8.
Dissolving 5.0g of N-methyl trifluoroacetamide in 28mL of DMF, adding 1.656g of 60% NaH in batches under the cooling of an ice water bath, stirring for 1 hour at room temperature, dissolving 6.42g of 6-bromo-1-hexene in 25mL of DMF, dropwise adding the solution, and stirring at room temperature and overnight at 70 ℃ after dropwise adding. The reaction solution was poured into 200mL of water, extracted with diethyl ether, combined with diethyl ether, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to give a yellow oily liquid which was used directly in the next step.
The yellow oily liquid was dissolved in 50mL of methanol, 66mL of 25M KOH solution was added dropwise, and the mixture was stirred at room temperature overnight. Extracting the reaction solution with diethyl ether for three times, mixing diethyl ether, washing with saturated saline solution, and drying with anhydrous sodium sulfateFiltering and concentrating to obtain a light yellow oily liquid, distilling under reduced pressure by a water pump, and collecting 50-55 ℃ fractions to obtain 3.7g of a colorless liquid.1H NMR(400MHz,CDCl3)δ5.81(ddt,J=16.9,10.2,6.6Hz,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)。
Step 2. Synthesis of Compound 9.
Taking (1R, 4R, 5R) -3-oxo-2-oxabicyclo [2.2.1]Hexane-5-carboxylic acid cinchonidine salt 2.27g and compound 8 0.71g were dissolved in 25mL DMF and 2- (7-azobenzotriazol) -N, N, N ', N' -tetramethyluronium hexafluorophosphate (HATU, 2.30g), N, N-diisopropylethylamine (DIPEA, 0.78g) was added with cooling in an ice bath and stirred for 1 hour in the ice bath. Pouring the reaction solution into 100mL of water, extracting with ethyl acetate, combining ethyl acetate, washing with saturated saline solution, drying with anhydrous sodium sulfate, filtering, concentrating, and performing column chromatography to obtain 1.04g of yellow oily liquid, namely the compound 9.1H NMR(400MHz,CDCl3)δ6.01-5.58(m,1H),5.17-4.84(m,3H),3.51-3.17(m,2H),2.99(t,J=8.0Hz,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)+
And 3, synthesizing a compound 10.
1.04g of compound 9 was dissolved in 8mL of tetrahydrofuran, 4.5mL of 1M LiOH solution was added dropwise to the mixture in an ice-water bath, and the mixture was stirred for 1 hour after the addition of the solution in the ice-water bath. Adjusting the pH value of a 4N HCl solution to 1-2, extracting with ethyl acetate, combining ethyl acetate, washing with saturated saline solution, drying with anhydrous sodium sulfate, filtering and concentrating to obtain 0.99g of light yellow oily liquid. LC-MS (APCI): m/z 268.2(M +1)+
Step 4. Synthesis of Compound 11.
163mg of Compound 5 was dissolved in 3mL of anhydrous DMF, and 253mg of HATU, 90mg of Compound 10, 287. mu.L of DIPEA were added under ice-bath, and stirred for 2 hours under ice-bath. The reaction mixture was poured into 20mL of water, extracted with ethyl acetate, and the organic phases were combined, washed successively with 1N HCl solution, water, and saturated aqueous salt solution, dried over anhydrous sodium sulfate, filtered, concentrated, and subjected to column chromatography to give 209mg of compound 11 as a pale yellow oily liquid. LC-MS (APCI): m/z ═393.3(M+1)+
Example 3. Synthesis of intermediate 17.
Figure GPA0000258315050000121
Step 1. Synthesis of Compound 13.
2.6g of methyl isopropyl ketone is taken to be dissolved in absolute ethyl alcohol, 4.87g of liquid bromine is dripped under an ice salt bath, and the reaction is carried out for 3 hours at room temperature after the dripping is finished. 60mL of petroleum ether was added to the reaction system, and washed twice with water, the petroleum ether was used to back-extract the aqueous phase, the petroleum ether was combined, and washed twice with ice-cold saturated sodium carbonate solution, once with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to obtain 3.04g of colorless oily liquid.1H NMR(300MHz,CDCl3)δ3.99(s,2H),2.99(hept,J=6.9Hz,1H),2.44(s,OH),2.14(s,OH),1.94(s,OH),1.86(s,OH),1.17(d,J=6.9Hz,6H)。
Step 2. Synthesis of Compound 15.
0.75g of Compound 13 and 0.49g of ethyl thioaluminate (Compound 14) were dissolved in absolute ethanol and heated under reflux for 3 hours. Cooling in ice water bath, adding ethyl acetate and water to quench reaction, adjusting pH to 7 with concentrated ammonia water, separating liquid, extracting water phase with ethyl acetate, combining organic phases, washing with saturated saline solution, drying with anhydrous sodium sulfate, filtering, concentrating, and performing column chromatography to obtain 0.383g of light yellow oily liquid.1H NMR(300MHz,DMSO-d6)δ7.72(s,1H),4.35(q,J=7.1Hz,2H),3.17-3.05(m,1H),1.31(t,J=7.1Hz,3H),1.25(d,J=6.9Hz,6H)。
Step 3. Synthesis of Compound 16.
383mg of Compound 15 were dissolved in 1.1mL MeOH and 3.6mL THF (tetrahydrofuran), and LiOH H was added2O77 mg and 50. mu.L of water were reacted at 50 ℃ for 2 hours. Concentrating the reaction solution, adjusting the pH value to 2 with 4N HCl solution, adding ethyl acetate for extraction, combining ethyl acetate phases, washing with saturated saline solution, drying with anhydrous sodium sulfate, filtering and concentrating to obtain a brown yellow oily liquid 261mg, namely a compound 17, LC-MS (APCI): m/z 172.0(M +1)+
Step 4. Synthesis of Compound No. 17.
261mg of Compound 16 was dissolved in 3.6mL of dry dichloromethane, 7.2. mu.L of DMF (dimethylformamide) was added, 280. mu.L of oxalyl chloride was added under ice bath, and the mixture was stirred at room temperature for 1 hour. Concentration gave a tan oil which was used directly in the next step.
Example 4. synthesis of intermediate 21.
Figure GPA0000258315050000122
Step 1. Synthesis of Compound 19.
Dissolving 5.0g of 2-methyl-3-methoxyaniline in 55mL of xylene, and adding 1M BCl at 0 DEG C338.5mL of DCM solution reacts for 30 minutes at 5 ℃, 2.5mL of acetonitrile is added to react for 45 minutes at 5 ℃, the reaction solution is transferred to a dropping funnel, the solution is dropped into 25mL of dimethylbenzene solution containing 5.14g of aluminum trichloride at 0 ℃, the temperature is increased after the reaction is carried out for 45 minutes at 5 ℃, dichloromethane is separated by a water knockout drum, and the reaction is carried out overnight at 75 ℃. The reaction solution was poured into 10mL of ice water and refluxed for 2 hours. Cooling to room temperature, separating, extracting a water layer with dichloromethane, combining dimethylbenzene and dichloromethane, washing with a 1N NaOH solution and saturated saline solution in sequence, drying with anhydrous sodium sulfate, filtering and concentrating to obtain a brown oily liquid, adding ice-cold isopropyl ether at 0 ℃, stirring for 2 hours at 0 ℃, separating out a gray solid, filtering, washing filter residues with ice-cold isopropyl ether, collecting the filter residues, and drying in vacuum at 45 ℃ overnight to obtain 1.86g of gray solid powder, namely the compound 19.1H NMR(500MHz,CDCl3)δ7.66(d,J=9.0Hz,1H),6.31(d,J=9.0Hz,1H),3.88(s,3H),2.55(s,3H),2.02(s,3H)。
Step 2. Synthesis of Compound 20.
206mg of the above-prepared compound 17 was dissolved in 3mL of dioxane, and a solution of compound 19(245mg) in dioxane (3mL) was added dropwise thereto, followed by stirring at room temperature for 1 hour. The reaction was concentrated and 3mL of saturated NaHCO was added3The solution was separated from 3mL of dichloromethane and the aqueous phase was extracted with dichloromethane. The dichloromethane was combined and washed with saturated brine and anhydrous sulfuric acidSodium drying, filtering and concentrating to obtain 404mg of white solid powder which is the compound 20.1H NMR(300MHz,CDCl3)δ11.30(s,1H),7.76(d,J=8.7Hz,1H),7.17(d,J=0.9Hz,1H),6.79(d,J=8.7Hz,1H),3.93(s,3H),3.32-3.13(m,1H),2.59(s,3H),2.17(s,3H),1.41(d,J=6.9Hz,6H)。
Step 3. Synthesis of Compound 21.
255mg of Compound 20 was dissolved in 5mL of t-butanol, and 180mg of potassium t-butoxide was added thereto, followed by heating and refluxing for 6 hours. The reaction mixture was concentrated, Dichloromethane (DCM) and aqueous solution were added, the aqueous phase was extracted with DCM, the DCM phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to give 180mg of white solid powder which was compound 21.1H NMR(300MHz,CDCl3)δ9.54(s,1H),8.24(d,J=9.0Hz,1H),7.09(d,J=0.9Hz,1H),7.01(d,J=9.1Hz,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=6.9Hz,6H)。
EXAMPLE 5 preparation of substituted quinoline Compound A-1
Figure GPA0000258315050000141
The specific synthesis steps are as follows:
Figure GPA0000258315050000142
step 1. Synthesis of Compound 23.
Dissolving 5.0g of 2-methyl-3-hydroxyl nitrobenzene in 40mL of DMF, adding 6.30g of potassium carbonate solid, and adding 2.24mL of deuterated iodomethane (CD) in ice-water bath3I) Stirred at room temperature for 3 hours. The reaction solution was poured into 300mL of water, extracted three times with ethyl acetate (30 mL. times.3), the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to give 8.4g of a brown oily liquid.1H NMR(300MHz,CDCl3)δ7.40(dd,J=8.2,1.1Hz,1H),7.32-7.22(m,2H),7.04(dd,J=8.2,1.1Hz,1H)。
Step 2. Synthesis of Compound 24.
8.4g of Compound 23 are dissolved in 40mL ethanol (EtOH) and 10mL water (H)2O), 8.9g of reduced iron powder and 3.5g of ammonium chloride solid were added, and heated under reflux for 2 hours. Filtering, washing filter residue with ethyl acetate until the filtrate is colorless. The filtrate was concentrated and column-chromatographed to give 4.7g of a colorless oily liquid, LC-MS (APCI): m/z 170.2(M +1)+
Step 3. Synthesis of Compound 25.
4.7g of Compound 24 are dissolved in 55mL of xylene and 1M BCl is added at 0 DEG C338.85mL of DCM solution reacts for 30 minutes at 5 ℃, 4.01mL of acetonitrile is added to react for 45 minutes at 5 ℃, the reaction solution is transferred to a dropping funnel, the solution is dropped into 25mL of dimethylbenzene solution containing 4.56g of aluminum trichloride at 0 ℃, the temperature is increased after the reaction is carried out for 45 minutes at 5 ℃, dichloromethane is separated by a water knockout drum, and the reaction is carried out overnight at 75 ℃. The reaction solution was poured into 10mL of ice water and refluxed for 2 hours. Cooling to room temperature, separating liquid, extracting a water layer by using dichloromethane, combining dimethylbenzene and dichloromethane, washing by using a 1N NaOH solution and saturated saline solution sequentially, drying by using anhydrous sodium sulfate, filtering and concentrating to obtain brown oily liquid, adding ice-cold isopropyl ether at 0 ℃, stirring for 2 hours at 0 ℃, separating out gray solid, filtering, washing filter residue by using ice-cold isopropyl ether, collecting the filter residue, and drying in vacuum at 45 ℃ overnight to obtain 2.992g of gray solid powder, namely the compound 25.1H NMR(500MHz,CDCl3)δ7.65(d,J=9.0Hz,1H),6.30(d,J=9.0Hz,1H),2.55(s,3H),2.01(s,3H)。
Step 4. Synthesis of Compound 26.
500mg of Compound 17 was dissolved in 5mL of dioxane, and a solution of Compound 25(450mg) in dioxane (5mL) was added dropwise thereto, followed by stirring at room temperature for 1 hour. The reaction was concentrated and 3mL of saturated NaHCO was added3The solution was separated from 3mL of dichloromethane and the aqueous phase was extracted with dichloromethane. The dichloromethane was combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to obtain 413mg of a white solid powder, which was compound 26.
Step 5. Synthesis of Compound 27.
2.85g is taken to be dissolvedCompound 26 was dissolved in 30mL of t-butanol, and 2.37g of potassium t-butoxide was added thereto and the mixture was refluxed for 6 hours. The reaction mixture was concentrated, and DCM and water were added for liquid separation, the aqueous phase was extracted with DCM, the DCM phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to give 1.73g of white solid powder which was compound 27.1H NMR(300MHz,CDCl3)δ9.55(s,1H),8.25(d,J=9.0Hz,1H),7.10(d,J=0.9Hz,1H),7.01(d,J=9.1Hz,1H),6.72(d,J=1.8Hz,1H),3.28-3.12(m,1H),2.43(s,3H),1.39(d,J=6.9Hz,6H)。
Step 6. Synthesis of Compound 28.
500mg of Compound 11, 486mg of Compound 27 and 669mg of triphenylphosphine were dissolved in 10mL of freshly distilled anhydrous THF, 552. mu.L of diisolactone azodicarboxylate (DIAD) was added dropwise at-15 ℃ and stirred at-15 ℃ for 5 hours and at room temperature overnight. Adding ice water under ice salt bath to quench reaction, extracting water phase with Ethyl Acetate (EA), combining EA phases, washing with saturated saline solution, drying with anhydrous sodium sulfate, filtering, concentrating, and performing column chromatography to obtain 485mg of compound 28. LC-MS (APCI): m/z 692.4(M +1)+
Step 7. Synthesis of Compound 29.
485mg of Compound 28, 18mg of the first-generation Glabra catalyst (Hoveyda-Grubbs catalyst) were dissolved in 420mL of anhydrous dichloroethane, degassed with nitrogen for 30 minutes and stirred at 75 ℃ for 17 hours. The reaction solution was concentrated and subjected to column chromatography to obtain 234mg of a pale yellow solid, i.e., compound 29. LC-MS (APCI): m/z 665.4(M +1)+
Step 8. Synthesis of Compound 30.
477mg of compound 29 was dissolved in 15mL of methanol and 26mL of tetrahydrofuran, and 2.89g of lithium hydroxide monohydrate and 7.5mL of water were added under ice-water bath and stirred at room temperature overnight. Adjusting the pH value to 2-3 with 1N hydrochloric acid, extracting with EA, combining EA phases, washing with saturated saline solution, drying with anhydrous sodium sulfate, filtering and concentrating to obtain 315mg of pale yellow solid powder, LC-MS (APCI): 650.4(M +1) ═ M/z+
And 9, synthesizing a compound A-1.
496mg of compound 30 are dissolved in 15mL of freshly distilled anhydrous tetrahydrofuran, and 308mg of N, N' -carbonylbis are added under nitrogen protectionImidazole (CDI), heated at reflux for 2 hours. After cooling to 50 ℃ 400mg of cyclopropylsulfonamide and 286mg of 1, 8-diazabicyclo [5.4.0 ] are added]Undec-7-ene (DBU), stirred overnight at 50 ℃. Concentrating the reaction solution, adding 2mL of 1N hydrochloric acid and 5mL of dichloromethane for separating liquid, washing an organic phase with saturated saline solution, drying the organic phase with anhydrous sodium sulfate, filtering and concentrating, and performing column chromatography to obtain 277mg of light yellow solid powder, namely a compound A-1, LC-MS (APCI): m/z 753.4(M +1)+
EXAMPLE 6 preparation of substituted quinoline Compound A-2
Figure GPA0000258315050000161
The specific synthesis steps are as follows:
Figure GPA0000258315050000171
step 1. Synthesis of Compound 32.
4.0g of 2-chloro-6-nitrotoluene is dissolved in 40ml of MoeOD, 1.0g of MeONa is added, and the mixture is stirred for 2 hours at 100 ℃ by microwave. After cooling, adding concentrated deuterated hydrochloric acid to adjust the pH value to 3-4, pouring the reaction solution into 20mL saturated saline solution, extracting with ethyl acetate (10mL multiplied by 3), combining the ethyl acetate, drying with anhydrous sodium sulfate, filtering and concentrating to obtain 3.953g of light yellow solid, namely the compound 32.1H NMR(500MHz,CDCl3)δ7.71(dd,J=8.2,1.3Hz,1H),7.61(dd,J=8.0,1.3Hz,1H),7.29(d,J=8.2,1H)。
Step 2. Synthesis of Compound 33.
4.24g of the compound 32, 4.52g of sodium tetramethoxyboride, 223mg of tris (dibenzylideneacetone) dipalladium and 205mg of 2-di-tert-butylphosphino-2 ', 4 ', 6 ' -triisopropylbiphenyl were mixed, 22mL of anhydrous DMF was added under nitrogen protection, and the mixture was heated to 100 ℃ and stirred for 2 hours. The reaction was cooled, poured into 100mL of water, extracted three times with ethyl acetate (30 mL. times.3), the ethyl acetate combined, filtered, the filtrate washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to give 4.875g of a brown oily liquid.
Step 3. Synthesis of Compound 34.
3.25g of Compound 33, 6.35g of reduced iron powder, and 2.43g of ammonium chloride solid were mixed, 20mL of ethanol and 5mL of water were added, and the mixture was refluxed for 2 hours. Cooling the reaction liquid, filtering, washing filter residues with methanol until the filtrate is colorless, combining the filtrates, concentrating, and performing column chromatography to obtain colorless liquid 3.06 g.
Step 4. Synthesis of Compound 35.
4.7g of compound 34 are dissolved in 55mL of xylene and 1M BCl is added at 0 DEG C338.85mL of DCM solution reacts for 30 minutes at 5 ℃, 4.01mL of acetonitrile is added to react for 45 minutes at 5 ℃, the reaction solution is transferred to a dropping funnel, the solution is dropped into 25mL of dimethylbenzene solution containing 4.56g of aluminum trichloride at 0 ℃, the temperature is increased after the reaction is carried out for 45 minutes at 5 ℃, dichloromethane is separated by a water knockout drum, and the reaction is carried out overnight at 75 ℃. The reaction solution was poured into 10mL of ice water and refluxed for 2 hours. Cooling to room temperature, separating liquid, extracting a water layer by using dichloromethane, combining dimethylbenzene and dichloromethane, washing by using a 1N NaOH solution and saturated saline solution sequentially, drying by using anhydrous sodium sulfate, filtering and concentrating to obtain brown oily liquid, adding ice-cold isopropyl ether at 0 ℃, stirring for 2 hours at 0 ℃, separating out gray solid, filtering, washing filter residue by using ice-cold isopropyl ether, collecting the filter residue, and drying in vacuum at 45 ℃ overnight to obtain 2.992g of gray solid powder, namely the compound 35.1H NMR(500MHz,CDCl3)δ7.65(d,J=9.0Hz,1H),6.31(d,J=9.0Hz,1H),3.87(s,3H),2.55(s,3H)。
Step 5. Synthesis of Compound 36.
500mg of Compound 17 was dissolved in 5mL of dioxane, and a solution of Compound 35(450mg) in dioxane (5mL) was added dropwise thereto, followed by stirring at room temperature for 1 hour. The reaction was concentrated and 3mL of saturated NaHCO was added3The solution was separated from 3mL of dichloromethane and the aqueous phase was extracted with dichloromethane. The dichloromethane was combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to obtain 413mg of a white solid powder, which was compound 36.
Step 6. Synthesis of Compound 37.
451mg of Compound 36 was dissolved in 10mL of t-butanol, and 540mg of potassium t-butoxide was added thereto and the mixture was refluxed for 6 hours. Concentrating the reaction solution, addingThe mixture was separated with DCM and water, the aqueous phase was extracted with DCM, the DCM phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to give 413mg of a white solid powder which was compound 37.1H NMR(400MHz,CDCl3)δ9.53(s,1H),8.30-8.19(m,1H),7.10(d,J=0.9Hz,1H),7.02(d,J=9.1Hz,1H),6.72(d,J=1.8Hz,1H),3.98(s,3H),3.24-3.16(m,1H),1.40(d,J=6.9Hz,6H)。
Step 7. Synthesis of Compound 38.
500mg of Compound 11, 486mg of Compound 37 and 669mg of triphenylphosphine were dissolved in 10mL of freshly distilled anhydrous THF, 552. mu.L of DIAD was added dropwise at-15 ℃ and stirred at-15 ℃ for 5 hours, naturally warmed at room temperature and stirred overnight. Adding ice water under ice salt bath to quench reaction, extracting an aqueous phase by using EA, combining the EA phase, washing by using saturated saline solution, drying by using anhydrous sodium sulfate, filtering, concentrating, and carrying out column chromatography to obtain 485mg of the compound 38. LC-MS (APCI): m/z 692.4(M +1)+
Step 8. Synthesis of Compound 39.
481mg of Compound 38 and 18mg of Hoveyda-Grubbs first-generation catalyst are dissolved in 420mL of anhydrous dichloroethane, degassed with nitrogen for 30 minutes and stirred at 75 ℃ for 15 hours. The reaction solution was concentrated and subjected to column chromatography to obtain 220mg of a pale yellow solid, i.e., compound 39. LC-MS (APCI): m/z 664.4(M +1)+
Step 9. Synthesis of Compound 40.
220mg of compound 39 was dissolved in 20mL of methanol and 30mL of tetrahydrofuran, and 2.89g of lithium hydroxide monohydrate and 15mL of water were added in an ice-water bath and stirred at room temperature overnight. Adjusting the pH value to 2-3 with 1N hydrochloric acid, extracting with EA, combining EA phases, washing with saturated saline solution, drying with anhydrous sodium sulfate, filtering and concentrating to obtain 186mg of pale yellow solid powder, LC-MS (APCI): 650.4(M +1) ═ M/z+
Step 11. Synthesis of Compound A-2.
150mg of compound 40 was dissolved in 15mL of freshly distilled anhydrous tetrahydrofuran, and 120mg of CDI was added under nitrogen and heated under reflux for 2 hours. After cooling to 50 ℃ 145mg of cyclopropylsulfonamide and 83mg of DBU were added and stirring was carried out overnight at 50 ℃. The reaction mixture was concentrated, 2mL of 1N hydrochloric acid and 5mL of methylene chloride were added to separate the reaction mixture, and the organic phase was saturated with waterWashing with sodium chloride solution, drying with anhydrous sodium sulfate, filtering, concentrating, and performing column chromatography to obtain 102mg of light yellow solid powder, i.e. compound A-2. LC-MS (APCI): m/z 753.4(M +1)+
EXAMPLE 7 preparation of substituted quinoline Compounds A-3
Figure GPA0000258315050000191
The specific synthesis steps are as follows:
Figure GPA0000258315050000201
step 1. Synthesis of Compound 41.
0.749g of 6-bromo-1-ethene was dissolved in 7.4mL of DMSO, 448mg of sodium azide was added in portions, and the mixture was stirred at room temperature for 2 hours. The reaction mixture was poured into 25mL of water, extracted three times with ether (10 mL. times.3), the combined ether and washed with saturated brine. To the ether solution were added 2.41g of triphenylphosphine and 0.5mL of water, and the mixture was stirred at room temperature overnight. The reaction was washed three times with 1N hydrochloric acid (25 mL. times.3), the aqueous phases were combined and back-extracted once with ether. Adjusting the pH value of 8-9 with NaOH solid in an ice-water bath, extracting with diethyl ether, mixing the extracted diethyl ether, washing with saturated saline solution, drying with anhydrous sodium sulfate, filtering and concentrating to obtain 450mg of colorless oily liquid, and directly using the colorless oily liquid in the next step.
Step 2. Synthesis of Compound 42.
450mg of compound 41 was dissolved in 25mL of diethyl ether and Boc was added dropwise in an ice-water bath2O1.0 g, DIPEA 974. mu.L was added dropwise thereto, and the mixture was stirred at room temperature overnight. The reaction solution was concentrated, and column chromatography was performed to obtain 899mg of a colorless oily liquid.
Step 3. Synthesis of Compound 43.
899mg of Compound 42 are dissolved in 3mL of DMF, 130mg of 60% NaH are added in portions, and the mixture is stirred at room temperature for 30 minutes, 655mg of deuterated iodomethane is added dropwise, and the mixture is stirred at room temperature for 2 hours. The reaction mixture was poured into 20mL of water, extracted three times with ethyl acetate (5 mL. times.3), and the ethyl acetate phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated to give 756mg of a colorless oily liquid.1H NMR(400MHz,CDCl3)δ5.79(ddt,J=16.9,10.2,6.7Hz,1H),5.13-4.87(m,2H),3.19(s,2H),2.06(dt,J=8.6,4.3Hz,2H),1.51(dt,J=14.8,7.2Hz,2H),1.44(s,9H),1.36(dt,J=14.7,7.2Hz,2H)。
Step 4. Synthesis of Compound 44.
756mg of Compound 43 was dissolved in 10mL of a mixed solution of DCM/TFA (V/V, 8/2) and stirred at room temperature for 2 hours. The reaction mixture was concentrated, 3mL of a saturated sodium bicarbonate solution and 3mL of diethyl ether were added for liquid separation, the aqueous phase was extracted with diethyl ether (3 mL. times.2), the diethyl ether was combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to obtain 336g of a colorless oily liquid, which was Compound 44.
And 5, synthesizing a compound 45.
Taking (1R, 4R, 5R) -3-oxo-2-oxabicyclo [2.2.1]Hexane-5-carboxylic acid cinchonidine salt 3.51g and compound 44.09 g were dissolved in 10mL DMF, HATU3.57g, DIPEA10g were added with ice cooling, and stirred for 1 hour in ice bath. Pouring the reaction solution into 100mL of water, extracting with ethyl acetate, combining ethyl acetate, washing with saturated saline solution, drying with anhydrous sodium sulfate, filtering, concentrating, and performing column chromatography to obtain 1.86g of yellow oily liquid, namely the compound 45.1H NMR(500MHz,CDCl3)δ5.77(ttd,J=13.1,6.7,2.8Hz,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)+
Step 6. Synthesis of Compound 46.
1.86g of Compound 45 was dissolved in 13mL of tetrahydrofuran, 5.4mL of a 1M LiOH solution was added dropwise to the mixture in an ice-water bath, and the mixture was stirred for 1 hour after completion of the addition. Adjusting the pH value of a 4N HCl solution to 1-2, extracting with ethyl acetate (10X 3), combining ethyl acetate, washing with saturated saline, drying with anhydrous sodium sulfate, filtering and concentrating to obtain a light yellow oily liquid 1.26g, and performing LC-MS (APCI): 252.3(M +1) M/z+
Step 7. Synthesis of Compound 47.
1.26g of Compound 46 are dissolved in 10mL of anhydrous DMF and 1.94g of HATU and 789mg of Compound 5, 920. mu.L of DIPEA, ice are added under ice bathStir 1 hour under bath. The reaction mixture was poured into 100mL of water, EA extracted, and the EA phases were combined, washed successively with 1N HCl solution, water, and saturated aqueous salt solution, dried over anhydrous sodium sulfate, filtered, concentrated, and subjected to column chromatography to give 209mg of compound 47 as a pale yellow oily liquid. LC-MS (APCI): m/z 396.3(M +1)+
Step 8. Synthesis of Compound 48.
500mg of Compound 47, 477mg of Compound 21 and 664mg of triphenylphosphine were dissolved in 10mL of freshly distilled anhydrous THF, 552. mu.L of DIAD was added dropwise at-15 ℃ and stirred at-15 ℃ for 5 hours, naturally warmed at room temperature and stirred overnight. Adding ice water under ice salt bath to quench the reaction, extracting an aqueous phase by EA, combining the EA phases, washing with saturated saline solution, drying with anhydrous sodium sulfate, filtering, concentrating, and carrying out column chromatography to obtain 529mg of a compound 48. LC-MS (APCI): m/z 692.4(M +1)+
Step 9. Synthesis of Compound 49.
529mg of Compound 48 and 24mg of Hoveyda-Grubbs first-generation catalyst were dissolved in 480mL of anhydrous dichloroethane, degassed with nitrogen for 30 minutes, and stirred at 75 ℃ for 15 hours. The reaction solution was concentrated and subjected to column chromatography to obtain 327mg of a pale yellow solid, which was compound 49. LC-MS (APCI): m/z 664.4(M +1)+
Step 10. Synthesis of Compound 50.
689mg of Compound 49 was dissolved in 20mL of methanol and 30mL of tetrahydrofuran, and 2.89g of lithium hydroxide monohydrate and 15mL of water were added under ice-water bath and stirred at room temperature overnight. Adjusting the pH value to 2-3 with 1N hydrochloric acid, extracting with EA, combining EA phases, washing with saturated saline solution, drying with anhydrous sodium sulfate, filtering and concentrating to obtain 686mg of light yellow solid powder, LC-MS (APCI): 650.4(M +1) ═ M/z+
Step 11. Synthesis of Compound A-3.
686mg of compound 50 was dissolved in 17mL of freshly distilled anhydrous tetrahydrofuran, and 308mg of CDI was added under nitrogen protection, and the mixture was refluxed for 2 hours. After cooling to 50 ℃ 400mg of cyclopropylsulfonamide and 286mg of DBU were added and stirred at 50 ℃ overnight. Concentrating the reaction solution, adding 2mL of 1N hydrochloric acid and 5mL of dichloromethane for separating liquid, washing the organic phase with saturated saline solution, drying with anhydrous sodium sulfate, filtering, concentrating, and performing column chromatography to obtain 335mg of light yellow solid powder, namely compound A-3, LC-MS(APCI):m/z=753.4(M+1)+
EXAMPLE 8 preparation of substituted quinoline Compound A-4
Figure GPA0000258315050000221
The specific synthesis steps are as follows:
Figure GPA0000258315050000231
step 1. Synthesis of Compound 51.
4.41g of the compound 33 was dissolved in 30mL of dichloromethane, 4.69mL of boron trichloride was slowly added dropwise under an ice salt bath, and the reaction was carried out for 30 minutes while naturally raising the temperature. 10mL of ice water was added dropwise to the ice water bath. The organic phase was washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to give a pale yellow solid (3.80 g).
Step 2. Synthesis of Compound 52.
3.56 of the compound 51 was dissolved in 20ml of DMF, and 6.30g of solid potassium carbonate was added thereto, followed by stirring at room temperature for 30 minutes, and 3.97g of deuterated iodomethane was added dropwise thereto, followed by stirring at room temperature for 2 hours. The reaction mixture was poured into 150mL of water, extracted three times with ethyl acetate (20 mL. times.3), the ethyl acetate was combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to give 3.63g of a pale yellow oily liquid.
Step 3. Synthesis of Compound 53.
3.63g of Compound 52, 6.40g of reduced iron powder, and 2.41g of ammonium chloride solid were mixed, 20mL of ethanol and 5mL of water were added, and the mixture was refluxed for 2 hours. Cooling the reaction liquid, filtering, washing filter residues with methanol until the filtrate is colorless, combining the filtrates, concentrating, and performing column chromatography to obtain colorless liquid 3.36 g.
Step 4. Synthesis of Compound 54.
4.7g of compound 53 are dissolved in 55mL of xylene and 1M BCl is added at 0 DEG338.85mL of DCM solution was reacted at 5 ℃ for 30 minutes, 4.01mL of acetonitrile was added and the reaction was carried out at 5 ℃ for 45 minutes, the reaction mixture was transferred to a dropping funnel and added dropwise to 25mL of a xylene solution containing 4.56g of aluminum trichloride at 0 ℃,after the reaction at 5 ℃ for 45 minutes, the temperature was raised, and after separation of dichloromethane by a water separator, the reaction was carried out overnight at 75 ℃. The reaction solution was poured into 10mL of ice water and refluxed for 2 hours. Cooling to room temperature, separating, extracting a water layer with dichloromethane, combining dimethylbenzene and dichloromethane, washing with a 1N NaOH solution and saturated saline solution in sequence, drying with anhydrous sodium sulfate, filtering and concentrating to obtain a brown oily liquid, adding ice-cold isopropyl ether at 0 ℃, stirring for 2 hours at 0 ℃, separating out a gray solid, filtering, washing filter residues with ice-cold isopropyl ether, collecting the filter residues, and drying in vacuum at 45 ℃ overnight to obtain 2.992g of gray solid powder, namely the compound 54.1H NMR(400MHz,CDCl3)δ7.66(d,J=9.0Hz,1H),6.31(d,J=9.1Hz,1H),2.56(s,3H)。
Step 5. Synthesis of Compound 55.
500mg of Compound 54 was dissolved in 5mL of dioxane, and a solution of Compound 17(450mg) in dioxane (5mL) was added dropwise thereto, followed by stirring at room temperature for 1 hour. The reaction was concentrated and 3mL of saturated NaHCO was added3The solution was separated from 3mL of dichloromethane and the aqueous phase was extracted with dichloromethane. The dichloromethane was combined, washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to give 445mg of a white solid powder, which was compound 55.
Step 6. Synthesis of Compound 56.
445mg of Compound 55 were dissolved in 10mL of t-butanol, and 540mg of potassium t-butoxide was added thereto and the mixture was refluxed for 6 hours. The reaction solution was concentrated, and DCM and water were added for liquid separation, the aqueous phase was extracted with DCM, the DCM phases were combined and washed with saturated brine, dried over anhydrous sodium sulfate, filtered and concentrated to give 413mg of white solid powder, which was compound 56.1H NMR(400MHz,CDCl3)δ9.53(s,1H),8.30-8.19(m,1H),7.10(d,J=0.9Hz,1H),7.02(d,J=9.1Hz,1H),6.72(d,J=1.8Hz,1H),3.24-3.16(m,1H),1.40(d,J=6.9Hz,6H)。
Step 7. Synthesis of Compound 57.
500mg of Compound 11, 486mg of Compound 56 and 669mg of triphenylphosphine were dissolved in 10mL of freshly distilled anhydrous THF, 552. mu.L of DIAD was added dropwise at-15 ℃ and stirred at-15 ℃ for 5 hours, naturally warmed at room temperature and stirred overnight. Adding under ice salt bathQuenching the reaction with ice water, EA extracting the water phase, combining the EA phases, washing with saturated saline, drying with anhydrous sodium sulfate, filtering, concentrating, and performing column chromatography to obtain 485mg of compound 57. LC-MS (APCI): m/z 695.3(M +1)+
Step 8. Synthesis of Compound 58.
473mg of Compound 57 and 11mg of Hoveyda-Grubbs first-generation catalyst were dissolved in 400mL of anhydrous dichloroethane, degassed with nitrogen for 30 minutes, and stirred at 75 ℃ for 15 hours. The reaction solution was concentrated and subjected to column chromatography to obtain 288mg of a pale yellow solid, i.e., compound 58. LC-MS (APCI): m/z 666.4(M +1)+
Step 9. Synthesis of Compound 59.
250mg of compound 58 are dissolved in 20mL of methanol and 30mL of tetrahydrofuran, and 2.89g of lithium hydroxide monohydrate and 15mL of water are added in an ice-water bath and stirred at room temperature overnight. Adjusting the pH value to 2-3 with 1N hydrochloric acid, extracting with EA, combining EA phases, washing with saturated saline solution, drying with anhydrous sodium sulfate, filtering and concentrating to obtain 212mg of pale yellow solid powder, LC-MS (APCI): m/z 652.4(M +1)+
And 10, synthesizing a compound A-4.
200mg of compound 59 are dissolved in 15mL of freshly distilled anhydrous tetrahydrofuran, 210mg of CDI are added under nitrogen, and the mixture is refluxed for 2 hours. After cooling to 50 ℃ 235mg of cyclopropylsulfonamide and 189mg of DBU were added and stirred at 50 ℃ overnight. Concentrating the reaction solution, adding 2mL of 1N hydrochloric acid and 5mL of dichloromethane for separating liquid, washing an organic phase with saturated saline solution, drying with anhydrous sodium sulfate, filtering, concentrating, and carrying out column chromatography to obtain 168mg of light yellow solid powder, namely the compound A-4. LC-MS (APCI): m/z 755.3(M +1)+
Example 9 preparation of substituted quinoline Compound A-5.
Figure GPA0000258315050000251
The specific synthesis steps are as follows:
Figure GPA0000258315050000252
step 1. Synthesis of Compound 60.
500mg of Compound 26, 482mg of Compound 47 and 664mg of triphenylphosphine were dissolved in 10mL of freshly distilled anhydrous THF, 552. mu.L of DIAD was added dropwise at-15 ℃ and stirred at-15 ℃ for 5 hours, naturally warmed at room temperature and stirred overnight. Adding ice water under ice salt bath to quench reaction, extracting water phase with EA, mixing EA phases, washing with saturated saline solution, drying with anhydrous sodium sulfate, filtering, concentrating, and performing column chromatography to obtain 541mg of compound 60. LC-MS (APCI): m/z 695.4(M +1)+
Step 2. Synthesis of Compound 61.
534mg of compound 60, 11mg of Hoveyda-Grubbs first-generation catalyst are dissolved in 300mL of anhydrous dichloroethane, degassed with nitrogen for 30 minutes and stirred at 75 ℃ for 15 hours. The reaction solution was concentrated and subjected to column chromatography to obtain 365mg of a pale yellow solid, i.e., compound 61. LC-MS (APCI): m/z 666.4(M +1)+
Step 3. Synthesis of Compound 62.
689mg of Compound 61 was dissolved in 20mL of methanol and 30mL of tetrahydrofuran, and 2.89g of lithium hydroxide monohydrate and 15mL of water were added under ice-water bath and stirred at room temperature overnight. Adjusting the pH value to 2-3 with 1N hydrochloric acid, extracting with EA, combining EA phases, washing with saturated saline solution, drying with anhydrous sodium sulfate, filtering and concentrating to obtain 686mg of light yellow solid powder, LC-MS (APCI): m/z 653.4(M +1)+
And 4, synthesizing a compound A-5.
500mg of compound 62 are dissolved in 15mL of freshly distilled anhydrous tetrahydrofuran, 520mg of CDI are added under nitrogen, and the mixture is refluxed for 2 hours. After cooling to 50 ℃ 515mg of cyclopropylsulfonamide and 412mg of DBU were added and stirring was carried out overnight at 50 ℃. Concentrating the reaction solution, adding 2mL of 1N hydrochloric acid and 5mL of dichloromethane for separating liquid, washing an organic phase with saturated saline solution, drying with anhydrous sodium sulfate, filtering, concentrating, and carrying out column chromatography to obtain 450mg of light yellow solid powder, namely the compound A-5. LC-MS (APCI): m/z 755.3(M +1)+
And (4) evaluating the biological activity.
To verify the effect of the compounds described herein on HCV, the inventors used the HCV Replicon System (HCV replication System) as an evaluation model. Since the first report in Science1999, the HCV replicon system has become one of the most important tools for studying HCV RNA replication, pathogenicity, and virus persistence, for example, the minimal 5' -NCR region necessary for HCV RNA replication has been successfully demonstrated using the replicon, and the HCV replicon system has been successfully used as an evaluation model for antiviral drugs. The inventors of the present invention performed the verification according to the methods described in Science, 1999, 285(5424), 110-3, and j.virol, 2003, 77(5), 3007-19.
(1) Detection of Compound Activity against HCV 1a and 1b genotype replicons
HCV-1a and HCV-1b stably transfected replicon cells were used to detect the inhibitory activity of the compounds of hepatitis C virus genotype 1a and 1b replicons. This experiment will use the NS3/4A inhibitor Simeprevir as a positive control compound.
The method comprises the following steps: compounds were serially diluted 1: 3 for 8 concentration points, double-plated and added to 96-well plates. DMSO was set as no compound added control. The final concentration of DMSO in the cell culture broth was 0.5%.
Step two: HCV-1a and 1b cells were suspended in culture medium containing 10% FBS, respectively, and seeded into compound-containing 96-well plates at a density of 8,000 cells per well. Cells were in 5% CO2And cultured at 37 ℃ for 3 days.
Step three: cytotoxicity of compounds against GT1b replicon was determined using CellTiter-Fluor (Promega).
Step four: luciferase assay Compounds were assayed for anti-hepatitis C virus activity using Bright-glo (Promega).
Step five: data were analyzed using GraphPad Prism software, curves were fitted and EC calculated50Value sum CC50The value is obtained.
EC was calculated by analyzing the compounds A-1 to A-5 of examples 1 to 5 according to the above procedure50Value sum CC50The results are shown in Table 1.
TABLE 1 comparison of anti-HCV genotype replicon activity of example compounds A-1 to A-5 with control Simeprevir
Numbering GT1a EC50(nM) GT1b EC50(nM) CC50(nM)
Simeprevir 22.59 11.41 >1000
Compound A-1 8.07 3.79 >1000
Compound A-2 12.97 6.53 >1000
Compound A-3 9.48 3.96 >1000
Compound A-4 11.84 6.28 >1000
Compound A-5 10.29 8.43 >1000
The experimental result shows that the compound can inhibit multiple genotypes of HCV, and further can be used for inhibiting hepatitis C virus. Compared with the non-deuterated compound Simeprevir, the compound of the invention has better inhibitory activity on GT1a and GT1b replicons.
(2) Determination of Compound NS3/4A inhibitory Activity
See anal. biochem.373: 1-8, 2008 and international patent application publication WO 2006/102087, and the HCV NS3/4A protease Time Resolved Fluorescence (TRF) assay.
Figure GPA0000258315050000271
Figure GPA0000258315050000281
As shown in the table, the experimental results show that the compound can be used as an HCV NS3/4A protease inhibitor and further can be used in anti-hepatitis C virus medicines.
(3) Metabolic stability evaluation
Microsome experiment: human liver microsomes: 0.5mg/mL, Xenotech; rat liver microsomes: 0.5mg/mL, Xenotech; coenzyme (NADPH/NADH): 1mM, Sigma Life Science; magnesium chloride: 5mM, 100mM phosphate buffer (pH 7.4).
Preparing a stock solution: an amount of the compound examples, compounds A-1 to A-5 and positive control, Simeprevir, was finely weighed and dissolved in DMSO to 5mM, respectively.
Preparation of phosphate buffer (100mM, pH 7.4): 150mL of 0.5M potassium dihydrogenphosphate and 700mL of a 0.5M dipotassium hydrogenphosphate solution prepared in advance were mixed, the pH of the mixture was adjusted to 7.4 with the 0.5M dipotassium hydrogenphosphate solution, the mixture was diluted 5-fold with ultrapure water before use, and magnesium chloride was added to obtain a phosphate buffer (100mM) containing 100mM potassium phosphate and 3.3mM magnesium chloride at a pH of 7.4.
NADPH regenerating system solution (containing 6.5mM NADP, 16.5mM G-6-P, 3U/mL G-6-P D, 3.3mM magnesium chloride) was prepared and placed on wet ice before use.
Preparing a stop solution: acetonitrile solution containing 50ng/mL propranolol hydrochloride and 200ng/mL tolbutamide (internal standard). 25057.5 mu L of phosphate buffer solution (pH7.4) is taken to a 50mL centrifuge tube, 812.5 mu L of human liver microsome is respectively added and mixed evenly, and liver microsome dilution liquid with the protein concentration of 0.625mg/mL is obtained. 25057.5 mu L of phosphate buffer (pH7.4) is taken to a 50mL centrifuge tube, 812.5 mu L of SD rat liver microsome is respectively added, and the mixture is mixed evenly to obtain liver microsome dilution with the protein concentration of 0.625 mg/mL.
Incubation of the samples: the stock solutions of the corresponding compounds were diluted to 0.25mM each with an aqueous solution containing 70% acetonitrile, and used as working solutions. 398. mu.L of human liver microsome or rat liver microsome dilutions were added to a 96-well plate (N2), 2. mu.L of 0.25mM working solution was added, and mixed well.
Determination of metabolic stability: 300. mu.L 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 are placed in a 37 ℃ water bath box, shaken at 100 rpm and pre-incubated for 5 min. 80. mu.L of the incubation solution was taken out of each well of the incubation plate, added to the stop plate, mixed well, and supplemented with 20. mu.L of NADPH regenerating system solution as a 0min sample. Then 80. mu.L of NADPH regenerating system solution was added to each well of the incubation plate, the reaction was started, and the timer was started. The reaction concentration of the corresponding compound was 1. mu.M, and the protein concentration was 0.5 mg/mL. When the reaction was carried out for 10min, 30min and 90min, 100. mu.L of each reaction solution was added to the stop plate and vortexed for 3min to terminate the reaction. The stop plates were centrifuged at 5000 Xg for 10min at 4 ℃. And (3) taking 100 mu L of supernatant to a 96-well plate in which 100 mu L of distilled water is added in advance, mixing uniformly, and performing sample analysis by adopting LC-MS/MS.
And (3) data analysis: peak areas of corresponding compounds and internal standards were detected by LC-MS/MS systemAnd calculating the peak area ratio of the compound to the internal standard. The slope is determined by plotting the natural logarithm of the percentage of compound remaining against time and calculating t according to the following formula1/2And CLintWhere V/M is equal to 1/protein concentration.
Figure GPA0000258315050000291
The compounds of the invention and compounds without deuteration were tested simultaneously and compared to evaluate their metabolic stability in human and rat liver microsomes. The half-life and intrinsic hepatic clearance as indicators of metabolic stability are shown in table 2. The non-deuterated 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 non-deuterated compound Simeprevir, and thus is more suitable as a hepatitis c virus inhibitor.
TABLE 2 comparison of the metabolic stability of the example compounds A-1 to A-5 with the Simeprevir control
Figure GPA0000258315050000292
The compound of the embodiment is analyzed according to the steps, and the experimental result shows that the compound shows excellent metabolic stability in the human liver microsome and rat liver microsome experiments.
(4) Pharmacokinetic experiment of rat
Purpose of the experiment: study of the pharmacokinetic behavior of the compounds of the invention after administration of Simeprevir, the example compound, in rats was investigated.
Experimental animals:
species and strain: SD rat grade: SPF stage
Sex and amount: male, 6
Body weight range: 180 to 220g (actual weight range 187 to 197g)
The source is as follows: shanghai Xipulbikai laboratory animals Co., Ltd
Experimental and animal certification numbers: SCXK (Shanghai) 2013-0016.
The experimental process comprises the following steps:
before blood sample collection, 20L of 2M sodium fluoride solution (esterase inhibitor) was added to an EDTA-K2 anticoagulation tube, dried in an 80 ℃ oven, and stored in a 4 ℃ refrigerator.
Rats, males, weighing 187-197 g, were randomized into 2 groups, fasted overnight but with free access to water starting the afternoon of the day before the experiment, and given food 4h after administration. Simeprevir is given at 3mg/kg in group A, the compounds A-1 to A-53mg/kg of the formula example are given in group B, the blood is taken from orbital veins of rats for 15min, 30min, 1, 2, 3, 5, 8 and 10h after administration for about 100 plus 200L, the blood is placed in an Eppendorf tube of 0.5mL anticoagulated by EDTA-K2 and is mixed evenly immediately, after anticoagulation, the test tube is turned upside down and mixed evenly for 5 to 6 times as soon as possible, the blood is taken and is placed in an ice box, a blood sample is centrifugally separated at 4000rpm, 10min and 4 ℃ within 30min, and the plasma is immediately stored at-20 ℃ after all the plasma is collected. Plasma concentrations were determined in plasma at each time point after sample collection at all time points.
According to the mean plasma concentration-time data obtained after administration, the pharmacokinetic-related parameters of i.g. Simeprevir (3mg/kg) and example compounds A-1 to A-5(3mg/kg) after administration to male SD rats were calculated according to the theory of non-atrioventricular statistical moment by using Winnonin software.
The experimental result shows that compared with Simeprevir, the compound found by the inventor has better activity than that of Simeprevir and excellent pharmacokinetic property, so the compound is more suitable to be used as a compound for inhibiting the hepatitis C virus protein NS3/4A and is further suitable to be used for preparing a medicament for treating hepatitis C virus infection.
It is to be understood that these examples are intended to illustrate the invention and are not intended to limit the scope of the invention, and that experimental procedures not specifically identified in the examples will generally be performed under conventional conditions, or under conditions recommended by the manufacturer. Parts and percentages are parts and percentages by weight unless otherwise indicated.
The foregoing is a more detailed description of the invention in connection with specific preferred embodiments and it is not intended that the invention be limited to these specific details. For those skilled in the art to which the invention pertains, several simple deductions or substitutions can be made without departing from the spirit of the invention, and all shall be considered as belonging to the protection scope of the invention.

Claims (6)

1. A compound, wherein the compound is selected from the following compounds or a pharmaceutically acceptable salt thereof:
Figure FDA0003055503460000011
2. a pharmaceutical composition comprising a pharmaceutically acceptable carrier and a compound of claim 1 or a pharmaceutically acceptable salt thereof.
3. The pharmaceutical composition of claim 2, further comprising an additional active compound.
4. The pharmaceutical composition of claim 3 wherein said additional active compound is selected from the group consisting of HCV protease inhibitors, HCV NS5A inhibitors, and HCV NS5B polymerase inhibitors.
5. Use of a compound of claim 1 for the manufacture of a medicament for the treatment of hepatitis c virus infection.
6. Use of the pharmaceutical composition of any one of claims 2-4 in the manufacture of a medicament for inhibiting HCV NS3/4A protease activity.
CN201780004823.3A 2016-08-12 2017-08-07 Substituted quinoline compound, and pharmaceutical composition and application thereof Active CN108368105B (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN2016106621298 2016-08-12
CN201610662129 2016-08-12
PCT/CN2017/096198 WO2018028541A1 (en) 2016-08-12 2017-08-07 Substituted quinoline compound and pharmaceutical composition and use thereof

Publications (2)

Publication Number Publication Date
CN108368105A CN108368105A (en) 2018-08-03
CN108368105B true CN108368105B (en) 2021-06-29

Family

ID=61161778

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201780004823.3A Active CN108368105B (en) 2016-08-12 2017-08-07 Substituted quinoline compound, and pharmaceutical composition and application thereof

Country Status (2)

Country Link
CN (1) CN108368105B (en)
WO (1) WO2018028541A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
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

Patent Citations (2)

* Cited by examiner, † Cited by third party
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 (3)

* Cited by examiner, † Cited by third party
Title
"Deuterium Medicinal Chemistry:A New Approach to Durg Discovery and Development";Scott L. Harbeson et al.;《MEDCHEM NEWS》;20140531(第2期);第8-22页 *
"Structure–activity relationship study on a novel series of cyclopentane-containing macrocyclic inhibitors of the hepatitis C virus NS3/4A protease leading to the discovery of TMC435350";Pierre Raboisson et al.;《Bioorganic & Medicinal Chemistry Letters》;20080724;第18卷;第4853–4858页 *
"氘代作用在药物研究中的应用";江文峰 等;《齐鲁药事》;20101231;第29卷(第11期);第682-684页 *

Also Published As

Publication number Publication date
CN108368105A (en) 2018-08-03
WO2018028541A1 (en) 2018-02-15

Similar Documents

Publication Publication Date Title
DE60315420T2 (en) Heterocyclic tripeptides as hepatitis C inhibitors
EP1910378B1 (en) Hepatitis c inhibitor peptide analogs
TWI482766B (en) As a spiro compound for hepatitis C virus inhibitors
AU2003202347B2 (en) Tripeptides having a hydroxyproline ether of a substituted quinoline for the inhibition of NS3 (Hepatitis C)
US7119072B2 (en) Macrocyclic peptides active against the hepatitis C virus
TWI542585B (en) Hepatitis c virus inhibitors
AU2003202347A1 (en) Tripeptides having a hydroxyproline ether of a substituted quinoline for the inhibition of NS3 (Hepatitis C)
ZA200405639B (en) Macrocyclic peptides active against the hepatitis C virus.
AU2003202348A1 (en) Heterocyclic tripeptides as hepatitis c inhibitors
EP2076264A2 (en) Compounds and methods for treatment of hcv
CN104744444B (en) The fluoro- 9,10 dihydro phenanthrene class hepatitis C virus inhibitors of 9,9,10,10- tetra- and its application
EP1408031A1 (en) Pyrolidine derivatives useful in treatment of hepatitis C virus infection
WO2020001089A1 (en) Crystalline methyl carbamate compound
CN108368105B (en) Substituted quinoline compound, and pharmaceutical composition and application thereof
WO2017121188A1 (en) Hepatitis c virus inhibitor, pharmaceutical composition and application thereof
CN108368123B (en) Substituted imidazolyl compound and pharmaceutical composition thereof
CN108129366B (en) Antiviral compounds, methods of preparation and uses thereof
CN108368130B (en) Substituted macrocyclic quinoxaline compound, pharmaceutical composition and application thereof
CN108290844B (en) Substituted naphthalene ring compound, pharmaceutical composition and application thereof
CN108349907B (en) 1,4(1,4) -diphenylheterocycle hexa-tomato-12,43-diyl derivatives, process for their preparation and their use
WO2017121187A1 (en) Hepatitis c virus inhibitor, pharmaceutical composition and use thereof
CN108368128B (en) Substituted dicarbamate, pharmaceutical composition and application thereof
WO2017088730A1 (en) Silicone-containing compound for resisting hepatitis c virus infection
CA2474031C (en) Heterocyclic tripeptides as hepatitis c inhibitors
WO2017206688A1 (en) Substituted pyrrolidine compound, pharmaceutical composition, and applications thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant