WO2017101785A1 - Compound, preparation method therefor, pharmaceutical composition thereof and use thereof - Google Patents

Compound, preparation method therefor, pharmaceutical composition thereof and use thereof Download PDF

Info

Publication number
WO2017101785A1
WO2017101785A1 PCT/CN2016/109913 CN2016109913W WO2017101785A1 WO 2017101785 A1 WO2017101785 A1 WO 2017101785A1 CN 2016109913 W CN2016109913 W CN 2016109913W WO 2017101785 A1 WO2017101785 A1 WO 2017101785A1
Authority
WO
WIPO (PCT)
Prior art keywords
formula
compound
group
pharmaceutically acceptable
stereoisomer
Prior art date
Application number
PCT/CN2016/109913
Other languages
French (fr)
Chinese (zh)
Inventor
周星露
董晓武
刘兴国
Original Assignee
杭州和正医药有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from CN201510932904.2A external-priority patent/CN105348345A/en
Priority claimed from CN201610289337.8A external-priority patent/CN105777829B/en
Application filed by 杭州和正医药有限公司 filed Critical 杭州和正医药有限公司
Publication of WO2017101785A1 publication Critical patent/WO2017101785A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • A61K31/7072Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid having two oxo groups directly attached to the pyrimidine ring, e.g. uridine, uridylic acid, thymidine, zidovudine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H1/00Processes for the preparation of sugar derivatives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • C07H19/10Pyrimidine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Definitions

  • the invention belongs to the field of medicinal chemistry.
  • the present invention relates to a compound or a tautomer thereof, a stereoisomer, a mixture of stereoisomers, a prodrug, a pharmaceutically acceptable salt or a solvate thereof, a process for the preparation thereof, a pharmaceutical combination And its use as an anti-hepatitis C drug.
  • Hepatitis C is a liver disease caused by hepatitis C virus (HCV), which is a serious threat to human health.
  • HCV hepatitis C virus
  • HCV genome heterogeneity it can be divided into 6 types and 30 subtypes.
  • the type of infection has a strong regional nature, and China is mainly type 1.
  • type 1 accounted for 58.2%, of which gene type 1a was 1.4% and gene type 1b was 56.8%.
  • HCV-infected people there are more than 200 million HCV-infected people worldwide, accounting for 3.3% of the world's total population. There are about 3.2 million infected people in the United States. The number of HCV patients in China exceeds 40 million, ranking first in the world. Many HCV-infected patients are also hepatitis B and even AIDS patients, which increases the complexity of treatment. There is currently no officially approved hepatitis C vaccine, so preventing hepatitis C transmission remains a challenge. The existing chronically infected patients are developing, and it is expected that the peak incidence will be ushered in the next 10-20 years. The standard clinical regimen for the treatment of hepatitis C is peginterferon alfa combined with ribavirin.
  • the standard of cure is that the blood is not detected by HCV within 24 weeks after treatment.
  • This treatment has many side effects, including depression, fatigue, flu-like symptoms, and anemia; the cost of treatment is high, and the standard treatment takes 48 weeks and costs about $40,000.
  • the development of anti-hepatitis C virus small molecule compound drugs against the hepatitis C virus RNA gene sequence has rapidly become a hot spot, and antiviral drugs will enter the era of small molecule drugs from the interferon era.
  • HCV protease inhibitors have been extensively studied, among which Telaprevir (Terravir, VX-950, trade name Incivek) and Boceprevir (Bosipuvir, SCH-503034, trade name Victrelis) have been Approved for listing.
  • the inhibitor of the non-structural protein NS5A is also a specific antiviral drug acting on the hepatitis C virus RNA chain, and various genotypes of HCV virus have significant inhibitory effects.
  • drugs targeting NS5B polymerase are classified into two classes: nucleoside and non-nucleoside polymerase inhibitors. Among them, Sofosbuvir is by far the most potent anti-HCV drug, which is effective against HCV genotype 1, 2, 3, 4 and 6 infections.
  • the object of the present invention is to provide a novel anti-HCV virus and liver-protecting and liver-protecting bifunctional drugs, which can be used as a bifunctional drug, and has the characteristics of high oral bioavailability and good metabolic properties, and can be used for oral treatment. Viral hepatitis embolism.
  • Another object of the invention is to provide a process for the preparation of the compounds provided herein.
  • Still another object of the present invention is to provide a structural compound of the formula I or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable solvate thereof as an NS5B inhibitor, which protects liver cells, liver tissue, and improves The use of liver function and its application in the preparation of the treatment of viral hepatitis.
  • a further object of the present invention is to provide a pharmaceutical composition comprising one or more of the structural compounds of the formula I or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable solvate thereof.
  • X is hydroxy, F, Cl or Br
  • R, R' are independent of H, or And at least one of R and R' is Where Linker may or may not be present,
  • A is derived from a drug or derivative thereof for the treatment or adjuvant treatment of liver disease.
  • the drug or derivative thereof for treating or assisting in the treatment of liver disease may be a drug already on the market.
  • a group mentioned, such as the group I "from" the compound I generally means that the group I is obtained by reacting the compound I.
  • the invention protects a compound having the structure of formula I-1, or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable salt or solvate thereof Or a prodrug of the formula I-1:
  • X is a hydroxyl group or F, Cl, Br; R is Linker A is Or absent; G is derived from a drug or derivative thereof that has been marketed for the treatment or adjuvant treatment of liver disease; R 1 is H or C 1 -C 5 alkyl; n is an integer from 1 to 19.
  • a further preferred compound is a prodrug containing a nucleoside-like structure represented by the formula II-1A or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable salt thereof.
  • X is a hydroxyl group or F, Cl, Br; Linker A is or Or absent; R 1 is H or C 1 -C 5 alkyl; n is 1, 2, 3 , 4 or 5; and R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group.
  • a further preferred compound is a structure represented by the formula II-1Aa or the formula II-1Ab or a tautomer, an optical isomer, a stereoisomer thereof, a stereoisomer Body mixture, prodrug, pharmaceutically acceptable salt or solvate:
  • X is a hydroxyl group or F, Cl, Br; Linker A is or Or absent; R 1 is H or C 1 -C 5 alkyl; n is 1, 2, 3 , 4 or 5; and R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group.
  • a further preferred compound is a structure represented by the formula II-1B or a tautomer, an optical isomer, a stereoisomer, a mixture of stereoisomers, and a former Drug, pharmaceutically acceptable salt or solvate:
  • X is a hydroxyl group or F, Cl, Br; Linker A is Or absent; R 1 is H or C 1 -C 5 alkyl; n is 1, 2, 3 , 4 or 5; and R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group.
  • X is a hydroxyl group or F, Cl, Br; Linker A is Or absent; R 1 is H or C 1 -C 5 alkyl; n is 1, 2, 3 , 4 or 5; and R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group.
  • a prodrug containing a tiopronin structure represented by the formula II-1A-A(1) or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable substance thereof is further preferred.
  • X is hydroxyl or F, Cl, Br, and Linker A is or R 1 is H or C 1 -C 5 alkyl, and n is an integer from 1 to 19.
  • the following prodrug containing a tiopronin structure or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable thereof is further preferred Solvate:
  • the following prodrug containing a tiopronin structure or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable thereof is further preferred Solvate:
  • R 1 is H or C 1 -C 5 alkyl, and n is 1, 2, 3, 4 or 5.
  • Another object of the present invention is to provide a process for the preparation of a compound of the formula II-1A, which comprises: 1) reacting a compound of the formula III with a compound of the formula IV-1A to give a formula V-1A a compound represented by the formula V-1A is subjected to a deprotection reaction to obtain a compound of the formula II-1A;
  • X is a hydroxyl group or a halogen (F, Cl, Br); Linker A is Or absent; n is 1, 2, 3, 4 or 5; R 1 is H or C 1 -C 5 alkyl; R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group; The leaving group is preferably a halogen; Z is a mercapto protecting group, preferably trityl, 4-methoxytrityl, 4,4'-bismethoxytrityl.
  • Another object of the present invention is to provide a process for the preparation of a compound of the formula II-1B, which comprises: 1) reacting a compound of the formula III with a compound of the formula IV-1B to give a formula V-1B a compound of the formula V-1B is subjected to a deprotection reaction to give a compound of the formula II-1B;
  • X hydroxyl or halogen (F, Cl, Br); Linker A is Or absent; n is 1, 2, 3, 4 or 5; R 1 is H or C 1 -C 5 alkyl; R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group;
  • the leaving group is preferably a halogen;
  • Z is a mercapto protecting group, preferably trityl, 4-methoxytrityl, 4,4'-bismethoxytrityl.
  • a method for preparing a compound represented by the formula II-1A-Aa can be obtained by subjecting a compound represented by the formula IIIm to a compound represented by the formula IV-1 to obtain a formula V-1A. The compound shown is then obtained by deprotection.
  • X is a hydroxyl group or F, Cl or Br
  • R 1 is H or a C 1 -C 5 alkyl group
  • Y is a leaving group, preferably a halogen, such as fluorine, chlorine, bromine, iodine, etc.
  • Z is The thiol protecting group is preferably trityl, 4-methoxytrityl, 4,4'-bismethoxytrityl.
  • a method for producing a compound represented by the formula II-1A-Ab which can be obtained by subjecting a compound represented by the formula IIIm to a compound represented by the formula IV-1B to give a compound represented by the formula V-1B. Then through the deprotection reaction to get:
  • X is a hydroxyl group or a halogen (F, Cl, Br); R 1 is H or a C 1 -C 5 alkyl group; n is an integer from 1 to 19; Y is a leaving group, preferably a halogen, such as may be Fluorine, chlorine, bromine, iodine, etc.; Z is a mercapto protecting group, preferably trityl, 4-methoxytrityl, 4,4'-bismethoxytrityl.
  • Another object of the present invention is to provide a process for the preparation of a compound of the formula II-1A-B which can be subjected to a protective reaction by a compound represented by III and then reacted with an aldehyde to give a compound of the formula VII-1, and then Esterification or acylation with a compound represented by VIII-1A represented by the formula to give a compound of the formula IX-1A-B, followed by removal of a protecting group to give a compound of the formula II-1A-B .
  • the active site of the key intermediate of the preparation method is appropriately protected, which helps to reduce the occurrence of side reactions, has high reaction selectivity, and has high purity and easy purification of the obtained intermediate and final product.
  • the reaction involved in the method has the advantages of simple operation, convenient purification, and good controllability of the process, and is suitable for industrial production.
  • X is hydroxy or halogen (F, Cl, Br);
  • R 1 is H or C 1 -C 5 alkyl group;
  • R 3, R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group;
  • P 1 Is a hydroxy protecting group, preferably a trimethylsilyl group, a triethylsilyl group, a tert-butyldimethylsilyl group, a triphenylsilyl group;
  • P 2 is an amide NH protecting group, preferably a tert-butoxycarbonyl group , benzyloxycarbonyl, 4-methoxybenzyloxycarbonyl;
  • P 3 is a thiol protecting group, preferably trityl, 4-methoxytrityl, 4,4'-bismethoxytriphenyl methyl.
  • Another object of the present invention is to provide a process for the preparation of a compound of the formula II-1B-B which may be esterified with a compound of the formula VII-1 and then with a compound of the formula VIII-1B or
  • the acylation reaction gives the compound of the formula IX-1B-B, and then the protecting group is removed to give the compound of the formula II-1B-B.
  • the active site of the key intermediate of the preparation method is appropriately protected, which helps to reduce the occurrence of side reactions, has high reaction selectivity, and has high purity and easy purification of the obtained intermediate and final product.
  • the reaction involved in the method has the advantages of simple operation, convenient purification, and good controllability of the process, and is suitable for industrial production.
  • X, R 1 , R 3 , R 4 are as defined in the formula II-1B, and P 1 is a hydroxy protecting group, preferably trimethylsilyl, triethylsilyl, tert-butyl a dimethylsilyl group, a triphenylsilyl group; P 2 is an amide NH protecting group, preferably a tert-butoxycarbonyl group, a benzyloxycarbonyl group, a 4-methoxybenzyloxycarbonyl group; and P 3 is a fluorenyl protecting group. Preferred is trityl, 4-methoxytrityl, and 4,4'-bismethoxytrityl.
  • Another object of the present invention is to provide an intermediate represented by the following VII-1 and its use for preparing an anti-hepatitis C virus drug:
  • X is a hydroxyl group or a halogen (F, Cl, Br); R 1 is H or a C 1 -C 5 alkyl group; and P 1 is a hydroxy protecting group, preferably a trimethylsilyl group, a triethylsilyl group, Tert-butyldimethylsilyl, triphenylsilyl.
  • Another object of the present invention is to provide an intermediate represented by the following VIII-1A or a stereoisomer thereof or a mixture of stereoisomers thereof:
  • P 2 is an NH protecting group, preferably tert-butoxycarbonyl, benzyloxycarbonyl, 4-methoxybenzyloxycarbonyl;
  • P 3 is a fluorenyl protecting group, preferably trityl, 4-methoxy Tritylmethyl, 4,4'-bismethoxytrityl and the like.
  • Another object of the present invention is to provide a prodrug of the following IX-1A-B or a stereoisomer thereof, a mixture of stereoisomers:
  • X is a hydroxyl group or a halogen (F, Cl, Br);
  • R 1 is H or a C 1 -C 5 alkyl group; and
  • P 1 is a hydroxy protecting group, preferably a trimethylsilyl group, a triethylsilyl group, Tert-butyldimethylsilyl, triphenylsilyl;
  • P 2 is an NH protecting group, preferably tert-butoxycarbonyl, benzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, etc.;
  • P 3 is a fluorenyl protecting group
  • the group is preferably a trityl group, a 4-methoxytrityl group, a 4,4'-bismethoxytrityl group or the like.
  • Another object of the present invention is to provide an intermediate represented by the following VIII-1B or a stereoisomer thereof or a mixture of stereoisomers thereof:
  • R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group
  • P 2 is an amide NH protecting group, preferably a tert-butoxycarbonyl group, a benzyloxycarbonyl group or a 4-methoxybenzyloxycarbonyl group.
  • P 3 is a thiol protecting group, preferably trityl, 4-methoxytrityl, 4,4'-bismethoxytrityl.
  • Another object of the present invention is to provide a prodrug of the following IX-1B-B or a stereoisomer thereof, a mixture of stereoisomers:
  • X is hydroxy or halogen (F, Cl, Br);
  • R 1 is H or C 1 -C 5 alkyl group;
  • R 3, R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group;
  • P 1 Is a hydroxy protecting group, preferably a trimethylsilyl group, a triethylsilyl group, a tert-butyldimethylsilyl group, a triphenylsilyl group;
  • P 2 is an amide NH protecting group, preferably a tert-butoxycarbonyl group , benzyloxycarbonyl, 4-methoxybenzyloxycarbonyl;
  • P 3 is a thiol protecting group, preferably trityl, 4-methoxytrityl, 4,4'-bismethoxytriphenyl methyl.
  • the present invention provides a prodrug of the formula I-2, or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable salt or solvate thereof.
  • X is a hydroxyl group, F, Cl or Br
  • R' is Where Linker B may or may not be present, B is derived from a drug or derivative thereof that has been marketed for the treatment or adjuvant treatment of liver disease, F is linked to Linker B via a covalent bond, and then Linker B is covalently bonded to the nucleoside 4 The hydroxyl group is bonded to a hydroxyl group.
  • Linker B is absent, F is directly bonded to the hydroxy atom of the nucleoside at the 4-position by a covalent bond.
  • R 1 , R 2 is H or C 1 -C 5 alkyl, further preferably methyl or ethyl or propyl .
  • X is F, Br or Cl
  • Linker B is Or absent, when Linker B is absent, tiopronin or its derivative is directly linked to the hydroxy atom of the nucleoside 4 through an ester bond;
  • R 1 , R 2 are respectively H or C 1 -C 5 alkyl, further Preferred is a C 1 -C 5 alkyl group, still more preferably a methyl group or an ethyl group or a propyl group;
  • R 3 , R 4 is a hydrogen atom or a C 1 -C 5 alkyl group, and still more preferably a methyl group or an ethyl group. Or propyl.
  • Linker B is Or absent, when Linker B is absent, tiopronin or its derivative is directly linked to the hydroxy atom of the nucleoside 4 via an ester bond, and R 1 and R 2 are respectively H or C 1 -C 5 alkyl, R 3 , R 4 is a hydrogen atom or a C 1 - C 5 alkyl group, and still more preferably a methyl group or an ethyl group or a propyl group.
  • Linker B is Or absent, when Linker B is absent, acetylcysteine or its derivative is directly linked to the hydroxy atom of the nucleoside 4 via an ester bond, and R 1 and R 2 are respectively H or C 1 -C 5 alkyl Further, it is preferably a C 1 - C 5 alkyl group, still more preferably a methyl group or an ethyl group or a propyl group, and R 3 and R 4 are a hydrogen atom or a C 1 - C 5 alkyl group, and still more preferably a methyl group. Or ethyl or propyl.
  • Linker B is Or absent, when Linker B is absent, acetylcysteine or its derivative is directly linked to the hydroxy atom of the nucleoside 4 via an ester bond, and R 1 and R 2 are respectively H or C 1 -C 5 alkyl Further, it is preferably a methyl group or an ethyl group or a propyl group, and R 3 and R 4 are a hydrogen atom or a C 1 - C 5 alkyl group, and still more preferably a methyl group or an ethyl group or a propyl group.
  • the compound of the formula II-2 may also be selected from the following compounds of the formula III-2C, or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable solvate thereof:
  • Linker B is or R 3 and R 4 are a C 1 - C 5 hydrocarbon group, more preferably a C 1 - C 5 alkyl group, still more preferably a methyl group or an ethyl group or a propyl group.
  • the compounds of formula I may contain one or more chiral centers, as stereoisomers, ie, enantiomers, diastereomers or mixtures thereof, may be present.
  • the compounds of formula I- of the present invention may be individual isomers or mixtures of individual isomers.
  • the invention includes any prodrug form of the compound of Formula I.
  • the invention also includes pharmaceutically acceptable solvates of the compounds of formula I.
  • the invention also includes pharmaceutically acceptable oxides of the compounds of formula I, and pharmaceutically acceptable salts and pharmaceutically acceptable solvates thereof.
  • the invention also includes various crystalline forms of the compounds of formula I.
  • the present invention provides a compound of the formula I or a tautomer, a stereoisomer thereof, a mixture of stereoisomers, a prodrug, a pharmaceutically acceptable salt or a solvent thereof
  • a compound of the formula I or a tautomer, a stereoisomer thereof, a mixture of stereoisomers, a prodrug, a pharmaceutically acceptable salt or a solvent thereof Use of the compound as an NS5B inhibitor to simultaneously protect liver cells, liver tissue, improve liver function, and use in the preparation of a medicament for treating diseases such as viral hepatitis.
  • the present invention provides a therapeutically effective amount of a compound of the formula I or a tautomer thereof, a stereoisomer, a mixture of stereoisomers, a prodrug, a pharmacy
  • the present invention provides an NS5B inhibitor comprising a therapeutically effective amount of a compound of the formula I or a tautomer, a stereoisomer thereof, a mixture of stereoisomers
  • a prodrug a pharmaceutically acceptable salt or a solvate thereof
  • the inhibitor may optionally comprise a pharmaceutically acceptable carrier or excipient.
  • the composition consists of a therapeutically effective amount of one or more compounds of formula I or a tautomer, stereoisomer, stereoisomeric mixture, prodrug, pharmaceutically acceptable salt thereof or
  • the drug solvate is comprised of at least one pharmaceutically acceptable adjuvant.
  • the choice of pharmaceutical excipients varies depending on the route of administration and the characteristics of the action, and is usually a filler, a diluent, a binder, a wetting agent, a disintegrant, a lubricant, an emulsifier, a suspending agent, and the like.
  • the compound of the formula I, a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable solvate thereof, is present in the above composition in a proportion of from 0.1% to 99.9%, preferably from 1% to 99% by total weight. %.
  • the pharmaceutically acceptable carrier refers to a pharmaceutical carrier conventional in the pharmaceutical field, such as a diluent such as water; a filler such as starch, sucrose, etc.; a binder such as a cellulose derivative, an alginate, gelatin, Polyvinylpyrrolidone; wetting agent such as glycerin; disintegrants such as agar, calcium carbonate and sodium hydrogencarbonate; absorption enhancers such as quaternary ammonium compounds; surfactants such as cetyl alcohol; adsorption carriers such as kaolin and soap Clay; lubricants such as talc, calcium stearate and magnesium stearate, and polyethylene glycol.
  • a pharmaceutical carrier conventional in the pharmaceutical field, such as a diluent such as water; a filler such as starch, sucrose, etc.; a binder such as a cellulose derivative, an alginate, gelatin, Polyvinylpyrrolidone; wetting agent such as glycerin;
  • the present invention also provides a pharmaceutically acceptable compound of the formula I or a tautomer, a stereoisomer thereof, a mixture of stereoisomers, a prodrug, a pharmaceutically acceptable salt or a solvate thereof.
  • a method of preparing a composition A compound containing a tiopronin structure represented by Formula I or a tautomer thereof, a stereoisomer thereof, a mixture of stereoisomers, and a former
  • the drug, the pharmaceutically acceptable salt or the solvate thereof is mixed with a pharmaceutically acceptable adjuvant, and is prepared in a form suitable for administration by a conventional preparation method (dosage form).
  • Dosage forms include tablets, capsules, granules, pills, solutions, suspensions, emulsions, ointments, films, creams, aerosols, injections, suppositories, and the like. Preference is given to tablets and capsules.
  • the compound of the present invention is usually used in a dose of from 1 to 1000 mg per day, in single or multiple doses. However, if necessary, the above dosage can be appropriately deviated. Professionals can determine the optimal dose based on specific circumstances and expertise. These conditions include the severity of the disease, individual differences in the patient, characteristics of the formulation, and route of administration.
  • the present invention provides a compound of the formula I or a tautomer, a stereoisomer thereof, a mixture of stereoisomers, a prodrug, a pharmaceutically acceptable salt or a solvate thereof, or Use of a pharmaceutically acceptable composition as a human drug.
  • the present invention provides a method of treating viral hepatitis, the method comprising administering a therapeutically effective amount of a compound of the formula I or a tautomer thereof, a stereoisomer thereof One or more of a mixture of stereoisomers, prodrugs, pharmaceutically acceptable salts or solvates thereof or the pharmaceutical composition of the present invention is administered to a patient.
  • the compounds or compositions provided herein can be administered orally, by injection (intravenous, intramuscular, subcutaneous and intracoronary), sublingual, buccal, rectal, transurethral, vaginal, nasal, inhalation or topical routes.
  • the preferred route is oral.
  • it can be prepared into a conventional solid preparation such as a tablet, a powder, a granule, a capsule, etc., or as a liquid preparation, such as a water or oil suspension, or other liquid preparation, such as a syrup;
  • parenteral administration it may be prepared as a solution for injection, water or an oily suspension, or the like.
  • the present invention also provides a compound of the formula I or a tautomer, a stereoisomer thereof, a mixture of stereoisomers, a prodrug, a pharmaceutically acceptable salt or a pharmaceutically acceptable solvate thereof.
  • a compound of the formula I or a tautomer, a stereoisomer thereof, a mixture of stereoisomers, a prodrug, a pharmaceutically acceptable salt or a pharmaceutically acceptable solvate thereof Use in the preparation of a human NS5B inhibitor drug.
  • the present invention also provides a compound represented by Formula I, or a tautomer, a stereoisomer thereof, a mixture of stereoisomers, a prodrug, a pharmaceutically acceptable salt or a solvate thereof, and other therapeutic liver diseases.
  • a compound represented by Formula I or a tautomer, a stereoisomer thereof, a mixture of stereoisomers, a prodrug, a pharmaceutically acceptable salt or a solvate thereof, and other therapeutic liver diseases.
  • the combination of drugs is a compound represented by Formula I, or a tautomer, a stereoisomer thereof, a mixture of stereoisomers, a prodrug, a pharmaceutically acceptable salt or a solvate thereof, and other therapeutic liver diseases.
  • “Pharmaceutically acceptable” means a compound, material, composition, and/or dosage form that, within the scope of sound medical judgment, is suitable for contact with a patient's tissue without undue toxicity, irritation, allergies, or reasonable The benefits/risk ratios are commensurate with other problems and complications and are effectively used for the intended use.
  • halogen and “halo” are used interchangeably herein and refer to fluorine (F), chlorine (Cl), bromine (Br) or iodine (I).
  • alkyl or "alkyl group” as used herein, denotes a saturated straight or branched monovalent hydrocarbon group containing from 1 to 20 carbon atoms, wherein the alkyl group may be optionally selected The ground is replaced by one or more substituents described herein.
  • the alkyl group contains from 1 to 6 carbon atoms; in another embodiment, the alkyl group contains from 1 to 4 carbon atoms; and in one embodiment, the alkyl group contains 1 - 3 carbon atoms.
  • alkyl groups include, but are not limited to, methyl (Me, -CH 3 ), ethyl (Et, -CH 2 CH 3 ), n-propyl (n-Pr, -CH 2 CH 2 CH 3 ), isopropyl (i-Pr, -CH(CH 3 ) 2 ), n-butyl (n-Bu, -CH 2 CH 2 CH 2 CH 3 ), isobutyl (i-Bu, -CH 2 CH) (CH 3 ) 2 ), sec-butyl (s-Bu, -CH(CH 3 )CH 2 CH 3 ), tert-butyl (t-Bu, -C(CH 3 ) 3 ), and the like.
  • the "pharmaceutically acceptable salt” as used in the present invention means an organic salt and an inorganic salt of the compound of the present invention.
  • Pharmaceutically acceptable salts are well known in the art and are described in the literature: SMBerge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66: 1-19.
  • Salts formed by pharmaceutically acceptable non-toxic acids include, but are not limited to, mineral acid salts formed by reaction with amino groups, hydrochloride, hydrobromide, phosphate, sulfate, perchlorate, And organic acid salts such as acetates, oxalates, maleates, tartrates, citrates, succinates, malonates, or by other methods described in the literature, such as ion exchange These salts.
  • salts include adipate, alginate, ascorbate, aspartate, besylate, benzoate, disulfate, borate, butyrate, camphoric acid Salt, camphor sulfonate, cyclopentylpropionate, digluconate, lauryl sulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate Salt, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, Malate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, palmitate, pamoate, pectate, persulphate, 3-phenylpropionic acid Salt, picrate, pi
  • Salts obtained by appropriate bases include the alkali metal, alkaline earth metal, ammonium and N + (C 1-4 alkyl) 4 salts.
  • the present invention also contemplates quaternary ammonium salts formed from any of the compounds comprising a group of N. Water soluble or oil soluble or dispersed products can be obtained by quaternization.
  • Alkali metal or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Pharmaceutically acceptable salts further comprise suitable amine cation nontoxic ammonium, quaternary ammonium, and the counterion, such as halide, hydroxide, carboxylate, sulfated, phosphorylated compounds, nitrate compounds, C 1 -C 8 sulfonate and aromatic sulfonate.
  • suitable amine cation nontoxic ammonium, quaternary ammonium, and the counterion such as halide, hydroxide, carboxylate, sulfated, phosphorylated compounds, nitrate compounds, C 1 -C 8 sulfonate and aromatic sulfonate.
  • Solvent-forming solvents include, but are not limited to, water, isopropanol, ethanol, methanol, dimethyl sulfoxide, ethyl acetate, acetic acid, ethanolamine or mixtures thereof.
  • hydrate means that the solvent molecule is an association formed by water.
  • hydrate can be used.
  • a molecule of the compound of the invention may be combined with a water molecule, such as a monohydrate; in another embodiment, a molecule of the invention may be combined with more than one water molecule, such as dihydrate. In yet another embodiment, a molecule of the compound of the invention may be combined with less than one water molecule, such as a hemihydrate. It should be noted that the hydrates of the present invention retain the bioavailability of the compounds in a non-hydrated form.
  • stereochemistry of any particular chiral atom when the stereochemistry of any particular chiral atom is not indicated, all stereoisomers of the structure are contemplated within the invention, and as disclosed herein are included in the present invention. .
  • stereochemistry is indicated by a solid wedge or dashed line indicating a particular configuration, then the stereoisomers of the structure are defined and defined herein.
  • the compound of formula I can exist in the form of a salt.
  • the salt refers to a pharmaceutically acceptable salt.
  • pharmaceutically acceptable means that the substance or composition must be chemically and/or toxicologically compatible with the other ingredients comprising the formulation and/or the mammal treated therewith.
  • the salt is not necessarily a pharmaceutically acceptable salt, and may be an enantiomer for the preparation and/or purification of a compound of formula I and/or for isolation of a compound of formula I. Intermediates.
  • the pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound, basic or acidic moiety by conventional chemical methods.
  • such salts can be obtained by reacting the free acid form of these compounds with a stoichiometric amount of a suitable base such as a hydroxide, carbonate, bicarbonate, or the like of Na, Ca, Mg or K.
  • a suitable base such as a hydroxide, carbonate, bicarbonate, or the like of Na, Ca, Mg or K.
  • the free base form of these compounds is prepared by reaction with a stoichiometric amount of a suitable acid. This type of reaction is usually carried out in water or an organic solvent or a mixture of the two.
  • a non-aqueous medium such as diethyl ether, ethyl acetate, ethanol, isopropanol or acetonitrile.
  • a non-aqueous medium such as diethyl ether, ethyl acetate, ethanol, isopropanol or acetonitrile.
  • any structural formula given by the present invention is also intended to indicate that these compounds are not isotopically enriched and isotopically enriched.
  • Isotopically enriched compounds have the structure depicted by the general formula given herein, except that one or more atoms are replaced by an atom having a selected atomic mass or mass number.
  • Exemplary isotopes that may be incorporated into the compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, and chlorine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 17 O 18 O, 18 F, 31 P, 32 P, 35 S, 36 Cl and 125 I.
  • the invention relates to intermediates for the preparation of compounds of formula I.
  • the invention provides a pharmaceutical composition comprising a compound of the invention.
  • the pharmaceutical composition of the present invention further comprises a pharmaceutically acceptable carrier, excipient, adjuvant, vehicle, or a combination thereof.
  • the pharmaceutical composition can be in the form of a liquid, solid, semi-solid, gel or spray.
  • the compound of the present invention or a pharmaceutical composition thereof can be used for the treatment of hepatitis C, and has the advantages of high oral bioavailability and good metabolic properties, and has the functions of protecting liver tissue, liver cells, and improving liver function.
  • the compounds of the present invention can be prepared by the methods described herein, unless otherwise stated, wherein the substituents are as defined in Formula I.
  • the following reaction schemes and examples are provided to further illustrate the contents of the present invention.
  • Anhydrous tetrahydrofuran, dioxane, toluene and diethyl ether are obtained by refluxing with sodium metal.
  • Anhydrous dichloromethane and chloroform were obtained by reflux drying of calcium hydride.
  • Ethyl acetate, petroleum ether, n-hexane, N,N-dimethylacetamide and N,N-dimethylformamide were previously dried over anhydrous sodium sulfate.
  • reaction is generally carried out under a positive pressure of nitrogen or argon or on a dry solvent (unless otherwise indicated), the reaction bottle is stoppered with a suitable rubber stopper, and the substrate is driven through a syringe.
  • the glassware is dry.
  • the column is a silica gel column.
  • Silica gel 300-400 mesh was purchased from Qingdao Ocean Chemical Plant.
  • MS mass spectrometry
  • Step 1 5.0 g (9.5 mmol) of compound III-1 was dissolved in 25 mL of anhydrous N,N-dimethylformamide (DMF), and 1.6 g (23.5 mmol) of imidazole, 0.12 g (stepwise) was added under Ar protection.
  • DMAP 4-dimethylaminopyridine
  • TLCl triethylchlorosilane
  • Step 2 The product VII-1 freshly obtained in the previous step was dissolved in 40 mL of anhydrous dichloromethane (DCM), cooled in an ice bath under argon atmosphere, and then added 62 mg (0.5 mmol) of 4-dimethylaminopyridine (DMAP). And 2.36 g (12.3 mmol) of 1-ethyl-(3-dimethylaminopropyl)carbodiimide hydrochloride (EDC HCl), and 6.1 g (11.4 mmol) of compound VIII-A-1, The temperature was slowly raised to room temperature and the reaction was stirred for about 12 hours, and the reaction was completed by TLC. After adding 100 ml of dichloromethane, the mixture was washed with aq.
  • DCM anhydrous dichloromethane
  • Step 3 7.5 g (6.3 mmol) of compound IX-A-1 was dissolved in 50% water-acetone (20 mL) mixed solution, 30 mL of glacial acetic acid (HOAc) and 6 mL of trifluoroacetic acid (TFA) were added, and the reaction was stirred at room temperature. After about 6 hours, no remaining raw materials were detected by TLC.
  • HOAc glacial acetic acid
  • TFA trifluoroacetic acid
  • Compound VII-1 (3 mmol of compound III-1 was used as a starting material) was prepared by the method of Step B of Example 1, and dissolved in 12 mL of anhydrous dichloromethane (DCM), and cooled in an ice bath under argon atmosphere. 18 mg (0.15 mmol) of 4-dimethylaminopyridine (DMAP) and 0.75 g (3.9 mmol) of 1-ethyl-(3-dimethylaminopropyl)carbodiimide hydrochloride (EDC HCl) were added in sequence.
  • DMAP 4-dimethylaminopyridine
  • EDC HCl 1-ethyl-(3-dimethylaminopropyl)carbodiimide hydrochloride
  • the formaldehyde is replaced by propionaldehyde, and 2 g of the intermediate IX-A-3 can be prepared with 2.65 g (5 mmol) of the compound III-1 in a yield of 33% (two steps).
  • ESI-MS: m/z [M+H] + 121.
  • Step 1 529 mg (1.0 mmol) of III-1 was dissolved in 10 mL of anhydrous acetone, and 690 mg of IV-A-3 and potassium carbonate 414 mg (3.0 mmol) were added at room temperature, and the mixture was stirred under reflux for 6 hours, and the reaction was completely confirmed by TLC. After filtration, the filtrate was diluted with methylene chloride (30 mL). The residue was purified by silica gel column chromatography eluting with EtOAc
  • Step 1 529 mg (1.0 mmol) of III-1 was dissolved in 10 mL of anhydrous acetone, and 800 mg of IV-A-4 and potassium carbonate 414 mg (3.0 mmol) were added at room temperature, and the mixture was stirred under reflux for 12 h. After filtration, the filtrate was diluted with methylene chloride (30 mL). The residue was purified by silica gel column chromatography to yield 400 mg of V-A-5.
  • the second step of the method B VIII-A-2 is substituted for VIII-A-1, and 1.29 g of the intermediate IX-A-8 can be prepared by using 1.6 g (3 mmol) of the compound III-1.
  • the rate is 65% (two steps).
  • ESI-MS: m/z [M+H] + 1219.
  • VIII-B-1 obtained from (DL)-N-acetylcysteine
  • VIII-1 2.1 g (4 mmol) of compound III-1
  • ESI-MS: m/z [M+H] + 1191.
  • VIII-B-1a prepared by L-acetylcysteine, chiral purity >99%
  • VIII-B-1a is substituted for VIII-1, and 2.65 g (5 mmol) of compound III-1 is used.
  • Intermediate IX-B-1a can be prepared in a yield of 67% (two steps).
  • ESI-MS: m/z [M+H] + 1191.
  • the second step of the method of the first embodiment the VIII-B-2 (prepared by L-acetylcysteine, chiral purity >99%) is substituted for VIII-1, thereby giving 1.6 g (3 mmol) of the compound.
  • Intermediate IX-B-3 can be prepared in III-1 in a yield of 76% (two steps).
  • ESI-MS: m/z [M+H] + 1205.
  • the second step in the method B of the first embodiment the III-1 is replaced by III-2, and the VIII-B-1a (prepared by L-acetylcysteine, the chiral purity is >99%) is substituted for VIII.
  • III-1 2.64 g of Intermediate IX-B-4 can be obtained in 1.6 g (3 mmol) of compound III-2 in a yield of 72% (two steps).
  • Step 1 529 mg (1.0 mmol) of III-1 was dissolved in 10 mL of anhydrous dichloromethane, and 405 mg (1.0 mmol) of XA-1 was added at room temperature (preparation method refers to Polymer Chemistry, 2011, 2, 906-913) cyclohexyl carbon two
  • the imine 412 mg (2.0 mmol) and DMAP 12.2 mg (0.1 mmol) were stirred for 6 h, and the reaction was confirmed by TLC.
  • the solvent was removed by concentration under reduced pressure.
  • the residue was purified by silica gel column chromatography to give white solid XI-A-1, a yield of 43%, LC-ESI-MS : [M + H] + 916, was used directly in the next reaction.
  • Step 1 5.4 g (10 mmol) of compound III-1 was dissolved in 50 mL of anhydrous dichloromethane (DCM), cooled in an ice bath under Ar protection, and 61 mg (0.5 mmol) of 4-dimethylaminopyridine (DMAP) and 2.3 g (12 mmol) of 1-ethyl-(3-dimethylaminopropyl)carbodiimide hydrochloride (EDC HCl), after stirring for 5 minutes, 4.5 g (8.5 mmol) of compound VIII-A-1 was added. The reaction was stirred for about 6 hours and the reaction was complete by TLC. After adding 50 ml of dichloromethane, the mixture was washed with EtOAc EtOAc.
  • Step 2 4.7 g (4.5 mmol) of compound XII-A-1 was dissolved in 50 mL of dichloromethane, and 3.5 mL of trifluoroacetic acid (TFA) and 3.5 mL of triisopropylsilane ( i- Pr 3 SiH) were added at room temperature. After the reaction for 15 minutes, 3.5 mL of trifluoroacetic acid (TFA) was further added, and the reaction was continued for about 10 to 20 minutes. The reaction was completely confirmed by TLC, and the reaction was cautiously added with a saturated aqueous solution of sodium hydrogencarbonate. The organic layer was separated and dried over anhydrous sodium The residue was purified by silica gel column chromatography to give 2.5g white solid, yield 82%, LC-ESI-MS : [M + H] + 675.
  • TFA trifluoroacetic acid
  • i- Pr 3 SiH triisopropylsilane
  • Step 1 Dissolve 529 mg (1.0 mmol) of III-1 in 5 mL of dimethyl sulfoxide, add 4 mL of acetic anhydride and 1.5 mL of acetic acid, stir at room temperature for 48 hours, carefully add saturated brine and ethyl acetate, and separate the organic phase. The extract was washed with aq. The solvent was removed by concentration under reduced pressure. The residue was purified by silica gel column chromatography to give a pale yellow gum XIII-1, a yield of 25%, LC-ESI-MS : [M + H] + 590.
  • Step 2 118 mg (0.2 mmol) of XIII-1 was dissolved in anhydrous dichloromethane, and 0.3 mL of dichloromethane (1 M) of thionyl chloride was added thereto under ice-cooling. The reaction mixture was slowly warmed to room temperature and stirred for 2 h. The solvent was concentrated under reduced pressure to give a yellow gum. The above-mentioned gum XIII-1 was dissolved in 1 mL of anhydrous tetrahydrofuran, and 93 mg (0.2 mmol) of IX-A-1 and potassium carbonate 55 mg (0.4 mmol) were added at room temperature, and stirred for 6 hours, and the reaction was completely confirmed by TLC. The solvent was removed by concentration under reduced pressure.
  • Step 2 VIII-A-2 is substituted for VIII-A-1, and 5.3 g (10 mmol) of compound III-1 is used as a raw material to prepare 4.9 g of white solid product III-2A- 5, yield 71% (two steps).
  • ESI-MS: m/z [M+H] + 702.
  • Step 1 529 mg (1.0 mmol) of III-1 was dissolved in 10 mL of anhydrous dichloromethane, and 405 mg of XB-1 (1.0 mmol), cyclohexylcarbodiimide 412 mg (2.0 mmol) and DMAP 12.2 mg ( 0.1 mmol), stirred for 12 h, and the reaction was complete by TLC. The solvent was removed by concentration under reduced pressure. The residue was purified by silica gel column chromatography to give white solid XI-B-1, LC- ESI-MS: [M + H] + 916, was used directly in the next reaction.
  • VIII-B-1a prepared from L-acetylcysteine, L-form chiral purity 99%
  • VIII-A-1 was substituted for VIII-A-1 at 5.3 g (10 mmol).
  • Compound III-1 was used as a starting material to obtain 3.6 g of a white solid product II-A-1b in a yield of 51%.
  • Step 1 529 mg (1.0 mmol) of III-1 was dissolved in 10 mL of anhydrous dichloromethane, and 452 mg (1.0 mmol) of XV-1 was added under ice bath (preparation method refers to Chemistry-A European Journal, 2014, 20, 6526- 6531) and DMAP 122 mg (1.0 mmol), stirred for 1 h, filtered, EtOAc EtOAc (EtOAc)EtOAc. The solvent was concentrated under reduced pressure.
  • Example 28 Anti-hepatitis C virus activity (HCV, EC 50 ) and cytotoxicity (CC 50 )
  • HCV viral activity evaluation uses an enzyme that plays a key role in HCV RNA replication to establish an extracellular molecular replication model.
  • Huh7 cells containing HCV-replicon (1b genotype) were cultured in DMEM medium containing 10% fetal calf serum, 1X non-essential amino acid, Pen-Strep-Glu, G418. Antiviral screening tests were performed in different media without G418.
  • the cells were seeded in 96-well plates, and the test compounds were added immediately after inoculation of the cells, and incubated at 37 ° C in an incubator. The medium is then removed and the cells are used for whole nucleic acid extraction (including replicon RNA and host RNA).
  • the replicon RNA can be amplified in the Q-RT-PCR protocol and quantified accordingly.
  • the difference in the level of the observed replicon HCV RNA from the untreated control group was used as the manner of antiviral efficacy of the test compound, and the results are shown in Table 1.
  • the results of the inhibitory activity of the compound of intracellular replicon 1b of HCV (EC 50) was evaluated as an anti-hepatitis C virus activity, while cytotoxic activity (CC 50) for excluding a synchronization test false positive results due to the cytotoxicity caused.
  • inhibitory activity on intracellular HCV replicon (EC 50) was stronger than that in the positive control Sofosbuvir, indicating that the compound may be rapidly Sofosbuvir as intracellular degradation in the liver cells
  • the form that becomes an active metabolite exerts an anti-HCV effect.
  • another sub-thiopronin obtained by degradation in the molecule has a certain gain effect on antiviral activity, and this synergistic effect enables the compound provided by the present invention to be resistant to HCV.
  • the disease aspect has more application prospects.
  • Example 29 Study on the protective effect of mouse CCl 4 liver damage model
  • mice Male mice were taken before the experiment body weight of 24 ⁇ 28g, water, fasted 6 hours, were randomly divided into 3 groups, 5 mice per group, divided into blank control group, CCl 4 group, compound group (thio general Ning 50mg/kg, test compound 200mg/kg). The drug was administered once every 12 hours, and the perfusion volume was 10 mL/kg, which was administered 4 times. The blank control group and the model control group were simultaneously administered with an equal volume of physiological saline. One hour after the third administration, the other two groups of mice were intraperitoneally injected with 50% CCl 4 corn oil solution at 2 mg/kg bw of CCl 4 except for the blank control group. After 12 hours, each group was administered once more.
  • liver was cut into small pieces, fixed in formaldehyde, paraffin-embedded, stained with hematoxylin and eosin, and the liver cells were observed under a microscope.
  • III-2A-1 617.5 690.2 II-A-1a 596.3 672 II-A-1b 620.5 701.5 II-A-2 560.7 656.3 II-A-4 701.5 677.8 II-A-5 596.2 702.3 II-B-1 725.6 833.1 II-B-1a 699.1 795.3 III-2A-1a 663.5 701.5 III-2A-1b 682.3 679.2 III-2B-1a 710.3 695.8 III-2C-1 505.2 609.3
  • ALT and AST were abnormally elevated in the CCl 4 model group, and tiopronin significantly reversed the effect of this enzyme.
  • Sofosbuvir did not have this activity.
  • the compounds provided by the present invention have a significant enzyme-lowering effect, and the activity is comparable to or superior to that of tiopronin.
  • the reason may be that the thiopronin's carboxyl group is made into a prodrug, which improves its stability and its ability to be absorbed orally, thereby enhancing its activity.
  • the CCl4-induced liver injury leads to an increase in ALT and AST, and has significant inhibitory activity. And superior to the positive control tiopronin.
  • CCl 4 model group the central hepatic venous congestion, around lobular central or peripheral necrosis, liver cell swelling, ballooning deformation, II-A-1, II -Aa, II -A-2, II-A-5, III-2A-1a and other administration groups showed a small amount of punctate and fragmented necrosis, most of which showed balloon-like deformation, indicating that the toxicity caused by CCl 4 has been greatly reduced, indicating the present invention
  • the compounds involved do have a protective effect on liver damage caused by CCl 4 .
  • the compound provided by the present invention Due to the lack of an ideal animal model of HCV infection, it is not feasible to directly evaluate the protective effect of the compounds provided by the present invention on HCV-induced liver injury, but in theory and clinical application, the compound provided by the present invention has the function of protecting liver and protecting liver It also has a protective effect on the inflammatory response caused by HCV.
  • Example 30 Study on pharmacokinetic properties
  • the compounds of the present invention prepared using the above examples II-A-1, II-A-1a, II-A-1b, II-A-2, II-B-1, III-2A-1, III-2A- 1a (30mg/kg, the molar mass of the nucleoside moiety is consistent with the Sophie group), the positive control drug is Sophibuvir (25mg/kg), and each compound is added to 5% DMSO + 60% PEG400 + 35% physiological Saline was prepared by vortexing to a 10 mg/ml suspension for intragastric administration.
  • GS331007 was purchased from Shunyuan Technology (Shanghai) Co., Ltd.
  • SD rats were used as experimental animals (body weight 180-220 g), and 6 SD rats per test compound were randomly divided into 2 groups (gavage group and intravenous injection group), 3 in each group.
  • the time of blood collection from the tail vein of the intragastric administration was 0.17, 0.33, 0.5, 1, 1.5, 2, 4, 6, 8, 12, 24 hours; the time of blood collection by intravenous administration was 0.05, 0.1, 0.17, 0.5. 1,2,4,6,8,12,24 hours.
  • 0.3 ml of whole blood was taken, and 0.1 ml of plasma was taken after centrifugation and analyzed by LC-MS.
  • liver homogenate sample or standard curve sample was added to 100 ⁇ l of acetonitrile containing internal standard (100 ng/ml), vortexed for 2 min, centrifuged for 10 min (6000 rpm), and the supernatant was transferred to a sample vial;
  • GS331007 is the final metabolite and is a marker for studying the metabolic behavior of Sofosbuvir in vivo.
  • the blood concentration and metabolic behavior of GS331007 in rats II-A-1, II-A-1a, II-A-1b, II-A-2, II-B-1 and III-2A-1a were investigated.
  • the results showed that the blood concentration of GS331007 and the area under the curve of the test compound were significantly improved compared with the blood concentration of GS331007 in the sofosbuvir group. Oral bioavailability improved significantly.
  • the oral bioavailability was 3.38, 2.75, and 2.25 times of Soofibvir, respectively.
  • the half-lives of II-A-1 and II-A-2 reached 6.14 and 6.23 hours, respectively, which were 2.95 and 2.51 times that of Sophibuvir, respectively, and the drug-time curve was more moderate. Therefore, the compounds of the present invention are superior in pharmacokinetic properties and can be administered by oral absorption for the treatment of hepatitis C diseases.
  • Test compound III-2A-1 (1 mg/mL) was tested for stability in human simulated gastric juice, simulated intestinal fluid, human plasma, and human liver microsomes. The results showed that III-2A-1 has high stability in simulated gastric fluid (1 hour metabolic rate ⁇ 5%), III-2A-1 in simulated intestinal fluid (5 minutes metabolic rate is greater than 90%), plasma (5 minutes metabolism) Both rates are greater than 99%) and liver microsomes (more than 99% in 5 minutes) can rapidly degrade and release sofosbuvir and tiopronin. This result demonstrates that the test compound can be rapidly metabolized by the intestine, plasma, and liver by oral administration, and the prototype drug is released.
  • the absorption of the drug mainly occurs in the small intestine, it is derived from the nucleoside hydroxyl group of Sophibruvir compared to the pyrimidine nitrogen atom derivative of Sophibruvir (CN201510932904.2), and the resulting compound III-2A-
  • the PK properties of 1 and its analogues in vitro are more favorable for its in vivo transformation and absorption, and it is expected to be more effective as a bifunctional prodrug molecule for anti-hepatitis C "speculation and treatment".
  • test compound III-2A-1 test (100 mg) was taken and placed in a stable container of 40 ⁇ 2 ° C and 75 ⁇ 5% to carry out an accelerated stability test.
  • a one-month experiment showed that the degradation rate of III-2 was ⁇ 1% (new impurity content), and similar compound II-A-1 was derivatized with the pyrimidine nitrogen atom of Sofibvir previously found (CN201510932904.2). Compared with its chemical stability, it has obvious advantages (the degradation rate of II-A-1 is 4.5% under the same conditions).
  • the chemical stability of the obtained compound is higher by derivatization from the nucleoside hydroxyl group of Sophibuvir, which facilitates its further development into a dual function.
  • Anti-HC candidate drugs compared with a similar compound derivatized with the pyrimidine nitrogen atom of Sophibruvir, the chemical stability of the obtained compound is higher by derivatization from the nucleoside hydroxyl group of Sophibuvir, which facilitates its further development into a dual function.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Genetics & Genomics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Epidemiology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Disclosed in the present invention are a prodrug comprising a nucleoside analog structure as shown in formula I, a tautomer thereof, a stereisomer thereof, a stereisomer mixture thereof or a pharmaceutically acceptable solvate thereof. Also disclosed are a preparation method therefor, a pharmaceutical composition formed therefrom and a use thereof as an anti-hepatitis C drug. The present compound or the pharmaceutical composition thereof can be used for treating hepatitis C, and also has functions such as protecting liver tissue and liver cells, improving liver function and reducing aminotransferase.

Description

一种化合物、其制备方法、药物组合物及其用途Compound, preparation method thereof, pharmaceutical composition and use thereof 技术领域Technical field
本发明属于药物化学领域。具体而言,本发明涉及一种化合物或其互变异构体、立体异构体、立体异构体混合物、前药、药学上可接受的盐或其溶剂合物,其制备方法,药物组合物及其作为抗丙型肝炎药物的用途。The invention belongs to the field of medicinal chemistry. In particular, the present invention relates to a compound or a tautomer thereof, a stereoisomer, a mixture of stereoisomers, a prodrug, a pharmaceutically acceptable salt or a solvate thereof, a process for the preparation thereof, a pharmaceutical combination And its use as an anti-hepatitis C drug.
背景技术Background technique
丙型肝炎是丙型肝炎病毒(hepatitis C virus,以下简称HCV)引起的一种严重威胁人类健康的肝脏疾病。1978年,丙肝病毒被首次发现;1989年,完成基因序列测定,HCV被确认为非甲型、非乙型肝炎的又一种主要病原体。根据HCV基因组异质性特性,目前可以分为6种类型,30个亚型。感染的类型有较强的地域性,我国以1型为主。2011年的研究表明,中国HCV感染者中,1型占58.2%,其中基因1a型为1.4%,基因1b型为56.8%。目前,全球范围内HCV感染者超过2亿,占世界总人口的3.3%。美国约有320万感染者,中国HCV患者超过了4000万人次,居世界之首,很多HCV感染者同时是乙肝甚至艾滋病患者,这增加了治疗的复杂性。目前还没有正式获得批准的丙肝疫苗,所以防止丙肝传播仍面临挑战。已有的慢性感染者病情正在发展中,预计未来10-20年将会迎来发病高峰。临床上治疗丙肝的标准方案是聚乙二醇干扰素α联合利巴韦林,治愈标准是在治疗后24周内血液检测不到HCV的RNA。这种治疗方式副作用较大,包括抑郁、疲乏、流感样症状和贫血症等;治疗成本较高,标准疗程需要48周,费用约40,000美元。为解决上述问题,近年来针对丙肝病毒RNA基因序列的抗丙肝病毒的小分子化合物药物研发迅速成为热点,抗病毒药物将从干扰素时代进入小分子药物时代。目前,已有许多HCV蛋白酶抑制剂得到广泛研究,其中Telaprevir(替拉瑞韦,VX-950,商品名Incivek)和Boceprevir(波西普韦,SCH-503034,商品名Victrelis)已于2011年被批准上市。非结构蛋白NS5A的抑制剂也是一种作用于丙肝病毒RNA链的特异性抗病毒药物,多种基因型HCV病毒均具有显著的抑制作用。此外,以NS5B聚合酶为靶点的药物分为核苷类和非核苷聚合酶抑制剂两类。其中的Sofosbuvir是迄今为止最为高效的抗HCV药物,可有效对抗HCV基因型1、2、3、4和6感染。Hepatitis C is a liver disease caused by hepatitis C virus (HCV), which is a serious threat to human health. In 1978, hepatitis C virus was first discovered; in 1989, the completion of gene sequencing, HCV was confirmed as another major pathogen of non-A, non-B hepatitis. According to the HCV genome heterogeneity, it can be divided into 6 types and 30 subtypes. The type of infection has a strong regional nature, and China is mainly type 1. According to the 2011 study, among the HCV-infected people in China, type 1 accounted for 58.2%, of which gene type 1a was 1.4% and gene type 1b was 56.8%. At present, there are more than 200 million HCV-infected people worldwide, accounting for 3.3% of the world's total population. There are about 3.2 million infected people in the United States. The number of HCV patients in China exceeds 40 million, ranking first in the world. Many HCV-infected patients are also hepatitis B and even AIDS patients, which increases the complexity of treatment. There is currently no officially approved hepatitis C vaccine, so preventing hepatitis C transmission remains a challenge. The existing chronically infected patients are developing, and it is expected that the peak incidence will be ushered in the next 10-20 years. The standard clinical regimen for the treatment of hepatitis C is peginterferon alfa combined with ribavirin. The standard of cure is that the blood is not detected by HCV within 24 weeks after treatment. This treatment has many side effects, including depression, fatigue, flu-like symptoms, and anemia; the cost of treatment is high, and the standard treatment takes 48 weeks and costs about $40,000. In order to solve the above problems, in recent years, the development of anti-hepatitis C virus small molecule compound drugs against the hepatitis C virus RNA gene sequence has rapidly become a hot spot, and antiviral drugs will enter the era of small molecule drugs from the interferon era. At present, many HCV protease inhibitors have been extensively studied, among which Telaprevir (Terravir, VX-950, trade name Incivek) and Boceprevir (Bosipuvir, SCH-503034, trade name Victrelis) have been Approved for listing. The inhibitor of the non-structural protein NS5A is also a specific antiviral drug acting on the hepatitis C virus RNA chain, and various genotypes of HCV virus have significant inhibitory effects. In addition, drugs targeting NS5B polymerase are classified into two classes: nucleoside and non-nucleoside polymerase inhibitors. Among them, Sofosbuvir is by far the most potent anti-HCV drug, which is effective against HCV genotype 1, 2, 3, 4 and 6 infections.
发明内容Summary of the invention
本发明的目的在于提供一种新的抗HCV病毒和保肝、护肝双功能药物,使其发挥双功能药物的作用,具有口服生物利用度高,代谢性质好的特点,可以用于口服治疗病毒性肝炎等症。The object of the present invention is to provide a novel anti-HCV virus and liver-protecting and liver-protecting bifunctional drugs, which can be used as a bifunctional drug, and has the characteristics of high oral bioavailability and good metabolic properties, and can be used for oral treatment. Viral hepatitis embolism.
本发明的一个目的是提供一种通式I所示结构化合物或其立体异构体,立体异构体混合物或其药学上可接受的溶剂合物。It is an object of the present invention to provide a structural compound of the formula I or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable solvate thereof.
本发明的另一个目的是提供本发明提供的化合物的制备方法。Another object of the invention is to provide a process for the preparation of the compounds provided herein.
本发明的又一个目的是提供通式I所示结构化合物或其立体异构体,立体异构体混合物或其药学上可接受的溶剂合物作为NS5B抑制剂,保护肝细胞、肝脏组织,改善肝功能的用途,以及在制备治疗病毒性肝炎中的应用。Still another object of the present invention is to provide a structural compound of the formula I or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable solvate thereof as an NS5B inhibitor, which protects liver cells, liver tissue, and improves The use of liver function and its application in the preparation of the treatment of viral hepatitis.
本发明的再一个目的是提供包含通式I所示结构化合物或其立体异构体,立体异构体混合物或其药学上可接受的溶剂合物中的一种或多种的药物组合物。A further object of the present invention is to provide a pharmaceutical composition comprising one or more of the structural compounds of the formula I or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable solvate thereof.
本发明的再一个目的是提供一种治疗病毒性肝炎,同时保护肝细胞、肝脏组织,改善肝功能的方法。It is still another object of the present invention to provide a method for treating viral hepatitis while protecting liver cells, liver tissues, and improving liver function.
本发明采用如下所述的技术方案: The invention adopts the technical solution as described below:
一种化合物,其具有式I所示结构,或式I所示结构的互变异构体、光学异构体、立体异构体、立体异构体混合物、前药、药学上可接受的盐或溶剂合物:A compound having the structure of Formula I, or a tautomer, an optical isomer, a stereoisomer, a mixture of stereoisomers, a prodrug, a pharmaceutically acceptable salt of the structure of Formula I. Or solvate:
Figure PCTCN2016109913-appb-000001
Figure PCTCN2016109913-appb-000001
式I中,X为羟基、F、Cl或Br;In formula I, X is hydroxy, F, Cl or Br;
R、R’分别独立的为H、或者
Figure PCTCN2016109913-appb-000002
且R、R’中至少有一个为
Figure PCTCN2016109913-appb-000003
其中Linker可以存在或不存在,A来自于用于治疗或辅助治疗肝病的药物或其衍生物。所述治疗或辅助治疗肝病的药物或其衍生物可以是已经上市的药物。本发明中,提到的一个基团,比如基团I“来自于”化合物I,一般是指基团I是由化合物I反应得到。
R, R' are independent of H, or
Figure PCTCN2016109913-appb-000002
And at least one of R and R' is
Figure PCTCN2016109913-appb-000003
Where Linker may or may not be present, A is derived from a drug or derivative thereof for the treatment or adjuvant treatment of liver disease. The drug or derivative thereof for treating or assisting in the treatment of liver disease may be a drug already on the market. In the present invention, a group mentioned, such as the group I "from" the compound I, generally means that the group I is obtained by reacting the compound I.
根据本发明的一个方面,本发明保护一种化合物,其具有通式I-1所示的结构,或其立体异构体、立体异构体混合物或其药学上可接受的盐或溶剂合物,或者具有通式I-1所示的前药:According to one aspect of the invention, the invention protects a compound having the structure of formula I-1, or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable salt or solvate thereof Or a prodrug of the formula I-1:
Figure PCTCN2016109913-appb-000004
Figure PCTCN2016109913-appb-000004
其中,X为羟基或F、Cl、Br;R为
Figure PCTCN2016109913-appb-000005
Linker A为
Figure PCTCN2016109913-appb-000006
Figure PCTCN2016109913-appb-000007
或不存在;G来自于已经上市的用于治疗或辅助治疗肝病的药物或其衍生物;R1为H或C1-C5烷基;n为1-19的整数。
Wherein X is a hydroxyl group or F, Cl, Br; R is
Figure PCTCN2016109913-appb-000005
Linker A is
Figure PCTCN2016109913-appb-000006
Figure PCTCN2016109913-appb-000007
Or absent; G is derived from a drug or derivative thereof that has been marketed for the treatment or adjuvant treatment of liver disease; R 1 is H or C 1 -C 5 alkyl; n is an integer from 1 to 19.
在所述通式I-1中,进一步优选的化合物为通式II-1A所示含有类核苷结构的前药或其立体异构体、立体异构体混合物或其药学上可接受的盐或溶剂合物:In the above formula I-1, a further preferred compound is a prodrug containing a nucleoside-like structure represented by the formula II-1A or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable salt thereof. Or solvate:
Figure PCTCN2016109913-appb-000008
Figure PCTCN2016109913-appb-000008
其中,X为羟基或F、Cl、Br;Linker A为
Figure PCTCN2016109913-appb-000009
Figure PCTCN2016109913-appb-000010
或不存在;R1为H或C1-C5烷基;n为1、2、3、4或5;R3、R4各自独立地为氢原子或C1-C5烷基。
Wherein X is a hydroxyl group or F, Cl, Br; Linker A is
Figure PCTCN2016109913-appb-000009
or
Figure PCTCN2016109913-appb-000010
Or absent; R 1 is H or C 1 -C 5 alkyl; n is 1, 2, 3 , 4 or 5; and R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group.
在所述通式II-1A中,进一步优选的化合物为通式II-1Aa或者式II-1Ab所示的结构或其互变异构体、光学异构体、立体异构体、立体异构体混合物、前药、药学上可接受的盐或溶剂合物:In the above formula II-1A, a further preferred compound is a structure represented by the formula II-1Aa or the formula II-1Ab or a tautomer, an optical isomer, a stereoisomer thereof, a stereoisomer Body mixture, prodrug, pharmaceutically acceptable salt or solvate:
Figure PCTCN2016109913-appb-000011
Figure PCTCN2016109913-appb-000011
其中,X为羟基或F、Cl、Br;Linker A为
Figure PCTCN2016109913-appb-000012
Figure PCTCN2016109913-appb-000013
或不存在;R1为H或C1-C5烷基;n为1、2、3、4或5;R3、R4各自独立地为氢原子或C1-C5烷基。
Wherein X is a hydroxyl group or F, Cl, Br; Linker A is
Figure PCTCN2016109913-appb-000012
or
Figure PCTCN2016109913-appb-000013
Or absent; R 1 is H or C 1 -C 5 alkyl; n is 1, 2, 3 , 4 or 5; and R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group.
在所述通式I-1中,进一步优选为的化合物为通式II-1B所示的结构或其互变异构体、光学异构体、立体异构体、立体异构体混合物、前药、药学上可接受的盐或溶剂合物:In the above formula I-1, a further preferred compound is a structure represented by the formula II-1B or a tautomer, an optical isomer, a stereoisomer, a mixture of stereoisomers, and a former Drug, pharmaceutically acceptable salt or solvate:
Figure PCTCN2016109913-appb-000014
Figure PCTCN2016109913-appb-000014
其中,X为羟基或F、Cl、Br;Linker A为
Figure PCTCN2016109913-appb-000015
或不存在;R1为H或C1-C5烷基;n为1、2、3、4或5;R3、R4各自独立地为氢原子或C1-C5烷基。
Wherein X is a hydroxyl group or F, Cl, Br; Linker A is
Figure PCTCN2016109913-appb-000015
Or absent; R 1 is H or C 1 -C 5 alkyl; n is 1, 2, 3 , 4 or 5; and R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group.
在所述通式II-1B中,进一步优选为通式II-1Ba或式II-1Bb所示的结构或其光学异构体、药学上可接受的盐或溶剂合物:In the above formula II-1B, a structure represented by the formula II-1Ba or the formula II-1Bb or an optical isomer, a pharmaceutically acceptable salt or a solvate thereof is further preferred:
Figure PCTCN2016109913-appb-000016
Figure PCTCN2016109913-appb-000016
其中,X为羟基或F、Cl、Br;Linker A为
Figure PCTCN2016109913-appb-000017
或不存在;R1为H或C1-C5烷基;n为1、2、3、4或5;R3、R4各自独立地为氢原子或C1-C5烷基。
Wherein X is a hydroxyl group or F, Cl, Br; Linker A is
Figure PCTCN2016109913-appb-000017
Or absent; R 1 is H or C 1 -C 5 alkyl; n is 1, 2, 3 , 4 or 5; and R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group.
在所述通式I-1中,进一步优选为通式II-1A-A(1)所示含有硫普罗宁结构的前药或其立体异构体,立体异构体混合物或其药学上可接受的溶剂合物: In the above formula I-1, a prodrug containing a tiopronin structure represented by the formula II-1A-A(1) or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable substance thereof is further preferred. Accepted solvates:
Figure PCTCN2016109913-appb-000018
Figure PCTCN2016109913-appb-000018
其中,X为羟基或F、Cl、Br,Linker A为
Figure PCTCN2016109913-appb-000019
Figure PCTCN2016109913-appb-000020
R1为H或C1-C5烷基,n为1-19的整数。
Where X is hydroxyl or F, Cl, Br, and Linker A is
Figure PCTCN2016109913-appb-000019
or
Figure PCTCN2016109913-appb-000020
R 1 is H or C 1 -C 5 alkyl, and n is an integer from 1 to 19.
或者,作为优选,在所述通式I-1中,进一步优选为下述通式II-1A-A(2)所示含有硫普罗宁结构的前药或其立体异构体,立体异构体混合物或其药学上可接受的溶剂合物:Or, preferably, in the above formula I-1, a prodrug containing a tiopronin structure represented by the following formula II-1A-A(2) or a stereoisomer thereof, stereoisomerism a mixture or a pharmaceutically acceptable solvate thereof:
Figure PCTCN2016109913-appb-000021
Figure PCTCN2016109913-appb-000021
上式中:X、Linker A、R1、n定义同上式II-1A-A(1)。In the above formula: X, Linker A, R 1 , and n are as defined in the above formula II-1A-A(1).
在所述通式II-1A-A(1)中,进一步优选为以下含有硫普罗宁结构的前药或其立体异构体,立体异构体混合物或其药学上可接受的溶剂合物:In the above formula II-1A-A(1), further preferred are the following prodrugs containing a tiopronin structure or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable solvate thereof:
Figure PCTCN2016109913-appb-000022
Figure PCTCN2016109913-appb-000022
其中,X为羟基或F、Cl、Br;R1为H或C1-C5烷基,n为1、2、3、4或5。Wherein X is a hydroxyl group or F, Cl, Br; R 1 is H or a C 1 -C 5 alkyl group, and n is 1, 2, 3, 4 or 5.
或者,作为优选,在所述通式II-1A-A(1)中,进一步优选为以下含有硫普罗宁结构的前药或其立体异构体,立体异构体混合物或其药学上可接受的溶剂合物: Alternatively, preferably, in the above formula II-1A-A(1), the following prodrug containing a tiopronin structure or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable thereof is further preferred Solvate:
Figure PCTCN2016109913-appb-000023
Figure PCTCN2016109913-appb-000023
其中,X、R1、n定义同上。Where X, R 1 and n are as defined above.
或者,作为优选,在所述通式II-1A-A(1)中,进一步优选为以下含有硫普罗宁结构的前药或其立体异构体,立体异构体混合物或其药学上可接受的溶剂合物:Alternatively, preferably, in the above formula II-1A-A(1), the following prodrug containing a tiopronin structure or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable thereof is further preferred Solvate:
Figure PCTCN2016109913-appb-000024
Figure PCTCN2016109913-appb-000024
其中,X、R1、n定义同上。Where X, R 1 and n are as defined above.
在所述通式II-1A-A中,进一步优选为以下含有硫普罗宁结构的前药或其立体异构体,立体异构体混合物或其药学上可接受的溶剂合物:In the above formula II-1A-A, further preferred are the following prodrugs containing a tiopronin structure or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable solvate thereof:
Figure PCTCN2016109913-appb-000025
Figure PCTCN2016109913-appb-000025
Figure PCTCN2016109913-appb-000026
Figure PCTCN2016109913-appb-000026
其中,R1为H或C1-C5烷基,n为1、2、3、4或5。Wherein R 1 is H or C 1 -C 5 alkyl, and n is 1, 2, 3, 4 or 5.
在所述通式I-1所示的化合物中,进一步优选的具体化合物为下列化合物之一:Among the compounds represented by the formula I-1, further preferred specific compounds are one of the following compounds:
Figure PCTCN2016109913-appb-000027
Figure PCTCN2016109913-appb-000027
Figure PCTCN2016109913-appb-000028
Figure PCTCN2016109913-appb-000028
Figure PCTCN2016109913-appb-000029
Figure PCTCN2016109913-appb-000029
本发明的另一个目的是提供通式II-1A所示的化合物的制备方法,所述方法包括:1)式III所示的化合物与式IV-1A所示的化合物反应得到式V-1A所示的化合物,2)式V-1A所示的化合物经过脱保护反应得到式II-1A所示的化合物; Another object of the present invention is to provide a process for the preparation of a compound of the formula II-1A, which comprises: 1) reacting a compound of the formula III with a compound of the formula IV-1A to give a formula V-1A a compound represented by the formula V-1A is subjected to a deprotection reaction to obtain a compound of the formula II-1A;
Figure PCTCN2016109913-appb-000030
Figure PCTCN2016109913-appb-000030
其中,X为羟基或卤素(F、Cl、Br);Linker A为
Figure PCTCN2016109913-appb-000031
Figure PCTCN2016109913-appb-000032
或不存在;n为1、2、3、4或5;R1为H或C1-C5烷基;R3、R4各自独立地为氢原子或C1-C5烷基;LG为离去基团,优选为卤素;Z为巯基保护基团,优选为三苯甲基、4-甲氧基三苯甲基、4,4’-双甲氧基三苯甲基。
Wherein X is a hydroxyl group or a halogen (F, Cl, Br); Linker A is
Figure PCTCN2016109913-appb-000031
Figure PCTCN2016109913-appb-000032
Or absent; n is 1, 2, 3, 4 or 5; R 1 is H or C 1 -C 5 alkyl; R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group; The leaving group is preferably a halogen; Z is a mercapto protecting group, preferably trityl, 4-methoxytrityl, 4,4'-bismethoxytrityl.
本发明的另一个目的是提供通式II-1B所示的化合物的制备方法,所述方法包括:1)式III所示的化合物与式IV-1B所示的化合物反应得到式V-1B所示的化合物;2)式V-1B所示的化合物经脱保护反应得到式II-1B所示的化合物;Another object of the present invention is to provide a process for the preparation of a compound of the formula II-1B, which comprises: 1) reacting a compound of the formula III with a compound of the formula IV-1B to give a formula V-1B a compound of the formula V-1B is subjected to a deprotection reaction to give a compound of the formula II-1B;
Figure PCTCN2016109913-appb-000033
Figure PCTCN2016109913-appb-000033
其中,X羟基或卤素(F、Cl、Br);Linker A为
Figure PCTCN2016109913-appb-000034
Figure PCTCN2016109913-appb-000035
或不存在;n为1、2、3、4或5;R1为H或C1-C5烷基;R3、R4各自独立地为氢原子或C1-C5烷基;LG为离去基团,优选为卤素;Z为巯基保护基团,优选为三苯甲基、4-甲氧基三苯甲基、4,4’-双甲氧基三苯甲基。
Among them, X hydroxyl or halogen (F, Cl, Br); Linker A is
Figure PCTCN2016109913-appb-000034
Figure PCTCN2016109913-appb-000035
Or absent; n is 1, 2, 3, 4 or 5; R 1 is H or C 1 -C 5 alkyl; R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group; The leaving group is preferably a halogen; Z is a mercapto protecting group, preferably trityl, 4-methoxytrityl, 4,4'-bismethoxytrityl.
更进一步地,一种通式II-1A-Aa所示的化合物的制备方法,可以由通式IIIm所示的化合物与通式IV-1所示的化合物经过烃化反应得到通式V-1A所示的化合物,然后经过脱保护反应得到。 Further, a method for preparing a compound represented by the formula II-1A-Aa can be obtained by subjecting a compound represented by the formula IIIm to a compound represented by the formula IV-1 to obtain a formula V-1A. The compound shown is then obtained by deprotection.
Figure PCTCN2016109913-appb-000036
Figure PCTCN2016109913-appb-000036
其中,X为羟基或F、Cl或Br;R1为H或C1-C5烷基;Y为离去基团,优选为卤素,比如可以是氟、氯、溴、碘等,Z为巯基保护基团,优选为三苯甲基、4-甲氧基三苯甲基、4,4’-双甲氧基三苯甲基。Wherein X is a hydroxyl group or F, Cl or Br; R 1 is H or a C 1 -C 5 alkyl group; Y is a leaving group, preferably a halogen, such as fluorine, chlorine, bromine, iodine, etc., Z is The thiol protecting group is preferably trityl, 4-methoxytrityl, 4,4'-bismethoxytrityl.
一种式II-1A-Ab所示的化合物的制备方法,可以由通式IIIm所示的化合物与通式IV-1B所示的化合物经过烃化反应得到通式V-1B所示的化合物,然后经过脱保护反应得到:A method for producing a compound represented by the formula II-1A-Ab, which can be obtained by subjecting a compound represented by the formula IIIm to a compound represented by the formula IV-1B to give a compound represented by the formula V-1B. Then through the deprotection reaction to get:
Figure PCTCN2016109913-appb-000037
Figure PCTCN2016109913-appb-000037
其中,X为羟基或卤素(F、Cl、Br);R1为H或C1-C5烷基;n为1-19的整数;Y为离去基团,优选为卤素,比如可以是氟、氯、溴、碘等;Z为巯基保护基团,优选为三苯甲基、4-甲氧基三苯甲基、4,4’-双甲氧基三苯甲基。Wherein X is a hydroxyl group or a halogen (F, Cl, Br); R 1 is H or a C 1 -C 5 alkyl group; n is an integer from 1 to 19; Y is a leaving group, preferably a halogen, such as may be Fluorine, chlorine, bromine, iodine, etc.; Z is a mercapto protecting group, preferably trityl, 4-methoxytrityl, 4,4'-bismethoxytrityl.
本发明的另一个目的是提供通式II-1A-B所示的化合物的制备方法,可以由III所示的化合物经过保护反应,然后与醛反应得到通式VII-1所示的化合物,然后与通式所示的VIII-1A所示的化合物发生酯化或酰化反应得到通式IX-1A-B所示的化合物,然后脱去保护基得到通式II-1A-B所示的化合物。本制备方法的关键中间体的反应活泼位点被适当保护,有助于减少副反应的发生,反应选择性高,得到的中间体、最终产物纯度高,易于纯化。且本方法涉及的反应具有操作简单、纯化方便,以及工艺可控性好的优点,适合产业化生产。 Another object of the present invention is to provide a process for the preparation of a compound of the formula II-1A-B which can be subjected to a protective reaction by a compound represented by III and then reacted with an aldehyde to give a compound of the formula VII-1, and then Esterification or acylation with a compound represented by VIII-1A represented by the formula to give a compound of the formula IX-1A-B, followed by removal of a protecting group to give a compound of the formula II-1A-B . The active site of the key intermediate of the preparation method is appropriately protected, which helps to reduce the occurrence of side reactions, has high reaction selectivity, and has high purity and easy purification of the obtained intermediate and final product. Moreover, the reaction involved in the method has the advantages of simple operation, convenient purification, and good controllability of the process, and is suitable for industrial production.
Figure PCTCN2016109913-appb-000038
Figure PCTCN2016109913-appb-000038
其中,X为羟基或卤素(F、Cl、Br);R1为H或C1-C5烷基;R3、R4各自独立地为氢原子或C1-C5烷基;P1为羟基保护基团,优选为三甲基硅基、三乙基硅基、叔丁基二甲基硅基、三苯基硅基;P2为酰胺NH保护基团,优选为叔丁氧羰基、苄氧羰基、4-甲氧基苄氧羰基;P3为巯基保护基团,优选为三苯甲基、4-甲氧基三苯甲基、4,4’-双甲氧基三苯甲基。Wherein, X is hydroxy or halogen (F, Cl, Br); R 1 is H or C 1 -C 5 alkyl group; R 3, R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group; P 1 Is a hydroxy protecting group, preferably a trimethylsilyl group, a triethylsilyl group, a tert-butyldimethylsilyl group, a triphenylsilyl group; P 2 is an amide NH protecting group, preferably a tert-butoxycarbonyl group , benzyloxycarbonyl, 4-methoxybenzyloxycarbonyl; P 3 is a thiol protecting group, preferably trityl, 4-methoxytrityl, 4,4'-bismethoxytriphenyl methyl.
本发明的另一个目的是提供通式II-1B-B所示的化合物的制备方法,可以由通式VII-1所示的化合物,然后与通式VIII-1B所示的化合物发生酯化或酰化反应得到通式IX-1B-B所示的化合物,然后脱去保护基团得到通式II-1B-B所示的化合物。本制备方法的关键中间体的反应活泼位点被适当保护,有助于减少副反应的发生,反应选择性高,得到的中间体、最终产物纯度高,易于纯化。且本方法涉及的反应具有操作简单、纯化方便,以及工艺可控性好的优点,适合产业化生产。Another object of the present invention is to provide a process for the preparation of a compound of the formula II-1B-B which may be esterified with a compound of the formula VII-1 and then with a compound of the formula VIII-1B or The acylation reaction gives the compound of the formula IX-1B-B, and then the protecting group is removed to give the compound of the formula II-1B-B. The active site of the key intermediate of the preparation method is appropriately protected, which helps to reduce the occurrence of side reactions, has high reaction selectivity, and has high purity and easy purification of the obtained intermediate and final product. Moreover, the reaction involved in the method has the advantages of simple operation, convenient purification, and good controllability of the process, and is suitable for industrial production.
Figure PCTCN2016109913-appb-000039
Figure PCTCN2016109913-appb-000039
其中,X,R1,R3,R4的定义同其在通式II-1B中的定义,P1为羟基保护基团,优选为三甲基硅基、三乙基硅基、叔丁基二甲基硅基、三苯基硅基;P2为酰胺NH保护基团,优选为叔丁氧羰基、苄氧羰基、4-甲氧基苄氧羰基;P3为巯基保护基团,优选为三苯甲基、4-甲氧基三苯甲基、4,4’-双甲氧基三苯甲基。 Wherein X, R 1 , R 3 , R 4 are as defined in the formula II-1B, and P 1 is a hydroxy protecting group, preferably trimethylsilyl, triethylsilyl, tert-butyl a dimethylsilyl group, a triphenylsilyl group; P 2 is an amide NH protecting group, preferably a tert-butoxycarbonyl group, a benzyloxycarbonyl group, a 4-methoxybenzyloxycarbonyl group; and P 3 is a fluorenyl protecting group. Preferred is trityl, 4-methoxytrityl, and 4,4'-bismethoxytrityl.
本发明的另一目的是提供如下VII-1所示的中间体及其用于制备抗丙肝病毒药物的用途:Another object of the present invention is to provide an intermediate represented by the following VII-1 and its use for preparing an anti-hepatitis C virus drug:
Figure PCTCN2016109913-appb-000040
Figure PCTCN2016109913-appb-000040
其中,X为羟基或卤素(F、Cl、Br);R1为H或C1-C5烷基;P1为羟基保护基团,优选为三甲基硅基、三乙基硅基、叔丁基二甲基硅基、三苯基硅基。Wherein X is a hydroxyl group or a halogen (F, Cl, Br); R 1 is H or a C 1 -C 5 alkyl group; and P 1 is a hydroxy protecting group, preferably a trimethylsilyl group, a triethylsilyl group, Tert-butyldimethylsilyl, triphenylsilyl.
本发明的另一目的是提供如下VIII-1A所示中间体或其立体异构体或其立体异构体混合物:Another object of the present invention is to provide an intermediate represented by the following VIII-1A or a stereoisomer thereof or a mixture of stereoisomers thereof:
Figure PCTCN2016109913-appb-000041
Figure PCTCN2016109913-appb-000041
其中,P2为NH保护基团,优选为叔丁氧羰基、苄氧羰基、4-甲氧基苄氧羰基;P3为巯基基保护基团,优选为三苯甲基、4-甲氧基三苯甲基、4,4’-双甲氧基三苯甲基等。Wherein P 2 is an NH protecting group, preferably tert-butoxycarbonyl, benzyloxycarbonyl, 4-methoxybenzyloxycarbonyl; P 3 is a fluorenyl protecting group, preferably trityl, 4-methoxy Tritylmethyl, 4,4'-bismethoxytrityl and the like.
本发明的另一目的是提供如下IX-1A-B所示的前药或其立体异构体,立体异构体混合物:Another object of the present invention is to provide a prodrug of the following IX-1A-B or a stereoisomer thereof, a mixture of stereoisomers:
Figure PCTCN2016109913-appb-000042
Figure PCTCN2016109913-appb-000042
其中,X为羟基或卤素(F、Cl、Br);R1为H或C1-C5烷基;P1为羟基保护基团,优选为三甲基硅基、三乙基硅基、叔丁基二甲基硅基、三苯基硅基;P2为NH保护基团,优选为叔丁氧羰基、苄氧羰基、4-甲氧基苄氧羰基等;P3为巯基保护基团,优选为三苯甲基、4-甲氧基三苯甲基、4,4’-双甲氧基三苯甲基等。Wherein X is a hydroxyl group or a halogen (F, Cl, Br); R 1 is H or a C 1 -C 5 alkyl group; and P 1 is a hydroxy protecting group, preferably a trimethylsilyl group, a triethylsilyl group, Tert-butyldimethylsilyl, triphenylsilyl; P 2 is an NH protecting group, preferably tert-butoxycarbonyl, benzyloxycarbonyl, 4-methoxybenzyloxycarbonyl, etc.; P 3 is a fluorenyl protecting group The group is preferably a trityl group, a 4-methoxytrityl group, a 4,4'-bismethoxytrityl group or the like.
本发明的另一目的是提供如下VIII-1B所示中间体或其立体异构体或其立体异构体混合物:Another object of the present invention is to provide an intermediate represented by the following VIII-1B or a stereoisomer thereof or a mixture of stereoisomers thereof:
Figure PCTCN2016109913-appb-000043
Figure PCTCN2016109913-appb-000043
其中,R3、R4各自独立地为氢原子或C1-C5烷基;P2为酰胺NH保护基团,优选为叔丁氧羰基、苄氧羰基、4-甲氧基苄氧羰基;P3为巯基保护基团,优选为三苯甲基、4-甲氧基三苯甲基、4,4’-双甲氧基三苯甲基。Wherein R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group; and P 2 is an amide NH protecting group, preferably a tert-butoxycarbonyl group, a benzyloxycarbonyl group or a 4-methoxybenzyloxycarbonyl group. ; P 3 is a thiol protecting group, preferably trityl, 4-methoxytrityl, 4,4'-bismethoxytrityl.
本发明的另一目的是提供如下IX-1B-B所示的前药或其立体异构体,立体异构体混合物: Another object of the present invention is to provide a prodrug of the following IX-1B-B or a stereoisomer thereof, a mixture of stereoisomers:
Figure PCTCN2016109913-appb-000044
Figure PCTCN2016109913-appb-000044
其中,X为羟基或卤素(F、Cl、Br);R1为H或C1-C5烷基;R3、R4各自独立地为氢原子或C1-C5烷基;P1为羟基保护基团,优选为三甲基硅基、三乙基硅基、叔丁基二甲基硅基、三苯基硅基;P2为酰胺NH保护基团,优选为叔丁氧羰基、苄氧羰基、4-甲氧基苄氧羰基;P3为巯基保护基团,优选为三苯甲基、4-甲氧基三苯甲基、4,4’-双甲氧基三苯甲基。Wherein, X is hydroxy or halogen (F, Cl, Br); R 1 is H or C 1 -C 5 alkyl group; R 3, R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group; P 1 Is a hydroxy protecting group, preferably a trimethylsilyl group, a triethylsilyl group, a tert-butyldimethylsilyl group, a triphenylsilyl group; P 2 is an amide NH protecting group, preferably a tert-butoxycarbonyl group , benzyloxycarbonyl, 4-methoxybenzyloxycarbonyl; P 3 is a thiol protecting group, preferably trityl, 4-methoxytrityl, 4,4'-bismethoxytriphenyl methyl.
根据本发明的另一个方面,本发明同时提供了一种通式I-2所示的的前药或其立体异构体、立体异构体混合物或其药学上可接受的盐或溶剂合物:According to another aspect of the present invention, the present invention provides a prodrug of the formula I-2, or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable salt or solvate thereof. :
Figure PCTCN2016109913-appb-000045
Figure PCTCN2016109913-appb-000045
其中,X为羟基、F、Cl或Br;R’为
Figure PCTCN2016109913-appb-000046
其中Linker B可以存在或不存在,B来自于已经上市的用于治疗或辅助治疗肝病的药物或其衍生物,F通过共价键与Linker B连接,然后Linker B通过共价键与核苷4位羟基氧原子连接,当Linker B不存在时,F直接通过共价键与核苷4位羟基氧原子连接。
Wherein X is a hydroxyl group, F, Cl or Br; R' is
Figure PCTCN2016109913-appb-000046
Where Linker B may or may not be present, B is derived from a drug or derivative thereof that has been marketed for the treatment or adjuvant treatment of liver disease, F is linked to Linker B via a covalent bond, and then Linker B is covalently bonded to the nucleoside 4 The hydroxyl group is bonded to a hydroxyl group. When Linker B is absent, F is directly bonded to the hydroxy atom of the nucleoside at the 4-position by a covalent bond.
进一步优选为通式II-2(1)或者II-2(2)所示的化合物或其立体异构体、立体异构体混合物或其药学上可接受的盐或溶剂合物:Further preferred is a compound represented by the formula II-2(1) or II-2(2) or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable salt or solvate thereof:
Figure PCTCN2016109913-appb-000047
Figure PCTCN2016109913-appb-000047
其中,X为F、Cl或Br,R’为
Figure PCTCN2016109913-appb-000048
其中Linker B为
Figure PCTCN2016109913-appb-000049
Figure PCTCN2016109913-appb-000050
或不存在,F为已经上市的用于治疗或辅助治疗肝病的药物或其衍生物,R1,R2为H或C1-C5烷基,进一步优选为甲基或乙基或丙基。
Where X is F, Cl or Br, and R' is
Figure PCTCN2016109913-appb-000048
Where Linker B is
Figure PCTCN2016109913-appb-000049
Figure PCTCN2016109913-appb-000050
Or absent, F is a drug or derivative thereof which has been marketed for the treatment or adjuvant treatment of liver diseases, R 1 , R 2 is H or C 1 -C 5 alkyl, further preferably methyl or ethyl or propyl .
更进一步优选为以下通式III-2A化合物,或其立体异构体、立体异构体混合物或其药学上可接受的溶剂合物: Still more preferably a compound of the following formula III-2A, or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable solvate thereof:
Figure PCTCN2016109913-appb-000051
Figure PCTCN2016109913-appb-000051
其中,X为F、Br或Cl;Wherein X is F, Br or Cl;
Linker B为
Figure PCTCN2016109913-appb-000052
或不存在,当Linker B不存在时,硫普罗宁或其衍生物直接通过酯键与核苷4位羟基氧原子连接;R1,R2分别为H或C1-C5烷基,进一步优选为C1-C5的烷基,更进一步优选为甲基或乙基或丙基;R3,R4为氢原子或C1-C5烷基,更进一步优选为甲基或乙基或丙基。
Linker B is
Figure PCTCN2016109913-appb-000052
Or absent, when Linker B is absent, tiopronin or its derivative is directly linked to the hydroxy atom of the nucleoside 4 through an ester bond; R 1 , R 2 are respectively H or C 1 -C 5 alkyl, further Preferred is a C 1 -C 5 alkyl group, still more preferably a methyl group or an ethyl group or a propyl group; R 3 , R 4 is a hydrogen atom or a C 1 -C 5 alkyl group, and still more preferably a methyl group or an ethyl group. Or propyl.
更进一步优选为通式III-2Aa或者III-2Ab化合物,或其立体异构体、立体异构体混合物或其药学上可接受的溶剂合物:Still more preferably a compound of the formula III-2Aa or III-2 Ab, or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable solvate thereof:
Figure PCTCN2016109913-appb-000053
Figure PCTCN2016109913-appb-000053
其中,为F、Br或Cl;Linker B为
Figure PCTCN2016109913-appb-000054
或不存在,当Linker B不存在时,硫普罗宁或其衍生物直接通过酯键与核苷4位羟基氧原子连接,R1,R2分别为H或C1-C5烷基,R3,R4为氢原子或C1-C5的烷基,更进一步优选为甲基或乙基或丙基。
Where is F, Br or Cl; Linker B is
Figure PCTCN2016109913-appb-000054
Or absent, when Linker B is absent, tiopronin or its derivative is directly linked to the hydroxy atom of the nucleoside 4 via an ester bond, and R 1 and R 2 are respectively H or C 1 -C 5 alkyl, R 3 , R 4 is a hydrogen atom or a C 1 - C 5 alkyl group, and still more preferably a methyl group or an ethyl group or a propyl group.
更进一步优选为以下通式III-2B化合物,或其立体异构体、立体异构体混合物或其药学上可接受的溶剂合物:Still more preferably a compound of the following formula III-2B, or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable solvate thereof:
Figure PCTCN2016109913-appb-000055
Figure PCTCN2016109913-appb-000055
其中,X为F、Br或Cl;Linker B为
Figure PCTCN2016109913-appb-000056
或不存在,当Linker B不存在时,乙酰半胱氨酸或其衍生物直接通过酯键与核苷4位羟基氧原子连接,R1,R2分别为H或C1-C5烷基,进一步优选为C1-C5的烷基,更进一步优选 为甲基或乙基或丙基,R3,R4为氢原子或C1-C5的烷基,更进一步优选为甲基或乙基或丙基。
Where X is F, Br or Cl; Linker B is
Figure PCTCN2016109913-appb-000056
Or absent, when Linker B is absent, acetylcysteine or its derivative is directly linked to the hydroxy atom of the nucleoside 4 via an ester bond, and R 1 and R 2 are respectively H or C 1 -C 5 alkyl Further, it is preferably a C 1 - C 5 alkyl group, still more preferably a methyl group or an ethyl group or a propyl group, and R 3 and R 4 are a hydrogen atom or a C 1 - C 5 alkyl group, and still more preferably a methyl group. Or ethyl or propyl.
更进一步优选为通式III-2Ba或者III-2Bb化合物,或其立体异构体、立体异构体混合物或其药学上可接受的溶剂合物:Still more preferably a compound of the formula III-2Ba or III-2Bb, or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable solvate thereof:
Figure PCTCN2016109913-appb-000057
Figure PCTCN2016109913-appb-000057
其中,X为F、Br或Cl;Linker B为
Figure PCTCN2016109913-appb-000058
或不存在,当Linker B不存在时,乙酰半胱氨酸或其衍生物直接通过酯键与核苷4位羟基氧原子连接,R1,R2分别为H或C1-C5烷基,更进一步优选为甲基或乙基或丙基,R3,R4为氢原子或C1-C5烷基,更进一步优选为甲基或乙基或丙基。
Where X is F, Br or Cl; Linker B is
Figure PCTCN2016109913-appb-000058
Or absent, when Linker B is absent, acetylcysteine or its derivative is directly linked to the hydroxy atom of the nucleoside 4 via an ester bond, and R 1 and R 2 are respectively H or C 1 -C 5 alkyl Further, it is preferably a methyl group or an ethyl group or a propyl group, and R 3 and R 4 are a hydrogen atom or a C 1 - C 5 alkyl group, and still more preferably a methyl group or an ethyl group or a propyl group.
所述通式II-2化合物也可以选自以下通式III-2C化合物,或其立体异构体、立体异构体混合物或其药学上可接受的溶剂合物:The compound of the formula II-2 may also be selected from the following compounds of the formula III-2C, or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable solvate thereof:
Figure PCTCN2016109913-appb-000059
Figure PCTCN2016109913-appb-000059
其中,X为F或Cl;Linker B为
Figure PCTCN2016109913-appb-000060
Figure PCTCN2016109913-appb-000061
R3,R4为C1-C5烃基,进一步优选为C1-C5的烷基,更进一步优选为甲基或乙基或丙基。
Where X is F or Cl; Linker B is
Figure PCTCN2016109913-appb-000060
or
Figure PCTCN2016109913-appb-000061
R 3 and R 4 are a C 1 - C 5 hydrocarbon group, more preferably a C 1 - C 5 alkyl group, still more preferably a methyl group or an ethyl group or a propyl group.
更进一步优选为下列化合物,或其立体异构体、立体异构体混合物或其药学上可接受的溶剂合物:Still more preferably the following compounds, or stereoisomers thereof, mixtures of stereoisomers or pharmaceutically acceptable solvates thereof:
Figure PCTCN2016109913-appb-000062
Figure PCTCN2016109913-appb-000062
Figure PCTCN2016109913-appb-000063
Figure PCTCN2016109913-appb-000063
Figure PCTCN2016109913-appb-000064
Figure PCTCN2016109913-appb-000064
Figure PCTCN2016109913-appb-000065
Figure PCTCN2016109913-appb-000065
通式I所示的化合物可含有一个或多个手性中心,因可存在立体异构体,即对映异构体、非对映异构体或其混合物。因此,本发明式I-所示的化合物可以为单个异构体或各异构体的混合物。The compounds of formula I may contain one or more chiral centers, as stereoisomers, ie, enantiomers, diastereomers or mixtures thereof, may be present. Thus, the compounds of formula I- of the present invention may be individual isomers or mixtures of individual isomers.
本发明包括通式I所示的化合物的任何前药形式。The invention includes any prodrug form of the compound of Formula I.
本发明还包括通式I的化合物的可药用溶剂化物。The invention also includes pharmaceutically acceptable solvates of the compounds of formula I.
本发明也包括通式I所示的化合物的可药用氧化物,及其可药用盐和可药用溶剂化物。The invention also includes pharmaceutically acceptable oxides of the compounds of formula I, and pharmaceutically acceptable salts and pharmaceutically acceptable solvates thereof.
本发明还包括通式I所示的化合物的多种晶型。The invention also includes various crystalline forms of the compounds of formula I.
根据本发明的又一方面,本发明提供了通式I所示的化合物或其互变异构体、立体异构体,立体异构体混合物、前药、药学上可接受的盐或其溶剂合物的用途,其作为NS5B抑制剂同时保护保护肝细胞、肝脏组织,改善肝功能的用途,和在制备用于治疗病毒性肝炎等疾病的药物中的用途。According to still another aspect of the present invention, the present invention provides a compound of the formula I or a tautomer, a stereoisomer thereof, a mixture of stereoisomers, a prodrug, a pharmaceutically acceptable salt or a solvent thereof Use of the compound as an NS5B inhibitor to simultaneously protect liver cells, liver tissue, improve liver function, and use in the preparation of a medicament for treating diseases such as viral hepatitis.
根据本发明的再一方面,本发明还提供了一种包含治疗有效量的通式I所示的化合物或其互变异构体、立体异构体,立体异构体混合物、前药、药学上可接受的盐或其溶剂合物中的一种或多种的药物组合物,其可以作为NS5B抑制剂,以及该组合物可以任选包含药学上可接受的载体或赋形剂。According to a further aspect of the present invention, the present invention provides a therapeutically effective amount of a compound of the formula I or a tautomer thereof, a stereoisomer, a mixture of stereoisomers, a prodrug, a pharmacy A pharmaceutical composition of one or more of the above acceptable salts or solvates thereof, which may act as an NS5B inhibitor, and which composition may optionally comprise a pharmaceutically acceptable carrier or excipient.
根据本发明的另一方面,本发明还提供了一种NS5B抑制剂,其含治疗有效量的通式I所示的化合物或其互变异构体、立体异构体,立体异构体混合物、前药、药学上可接受的盐或其溶剂合物中的一种或多种,以及该抑制剂可以任选包含药学上可接受的载体或赋形剂。According to another aspect of the present invention, the present invention provides an NS5B inhibitor comprising a therapeutically effective amount of a compound of the formula I or a tautomer, a stereoisomer thereof, a mixture of stereoisomers One or more of a prodrug, a pharmaceutically acceptable salt or a solvate thereof, and the inhibitor may optionally comprise a pharmaceutically acceptable carrier or excipient.
该组合物由治疗有效量的一种或多种通式I所示的化合物或其互变异构体、立体异构体,立体异构体混合物、前药、药学上可接受的盐或其药溶剂合物与至少一种可药用辅料组成。药用辅料的选择因施用途径和作用特点而异,通常是填充剂、稀释剂、粘合剂、润湿剂、崩解剂、润滑剂、乳化剂、助悬剂等。式I化合物、其立体异构体,立体异构体混合物或其药学上可接受的溶剂合物在上述组合物中的所占的比例为总重量的0.1%~99.9%,优选1%~99%。The composition consists of a therapeutically effective amount of one or more compounds of formula I or a tautomer, stereoisomer, stereoisomeric mixture, prodrug, pharmaceutically acceptable salt thereof or The drug solvate is comprised of at least one pharmaceutically acceptable adjuvant. The choice of pharmaceutical excipients varies depending on the route of administration and the characteristics of the action, and is usually a filler, a diluent, a binder, a wetting agent, a disintegrant, a lubricant, an emulsifier, a suspending agent, and the like. The compound of the formula I, a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable solvate thereof, is present in the above composition in a proportion of from 0.1% to 99.9%, preferably from 1% to 99% by total weight. %.
所述药学上可接受的载体是指药学领域常规的药物载体,例如:稀释剂,如水等;填充剂,如淀粉、蔗糖等;粘合剂,如纤维素衍生物、藻酸盐、明胶、聚乙烯吡咯烷酮;湿润剂,如甘油;崩解剂,如琼脂、碳酸钙和碳酸氢钠;吸收促进剂,如季铵化合物;表面活性剂,如十六烷醇;吸附载体,如高岭土和皂粘土;润滑剂,如滑石粉、硬脂酸钙和硬脂酸镁、和聚乙二醇等。另外,还可以在所述药物组合物中加入其它辅剂,如香味剂和甜味剂等。The pharmaceutically acceptable carrier refers to a pharmaceutical carrier conventional in the pharmaceutical field, such as a diluent such as water; a filler such as starch, sucrose, etc.; a binder such as a cellulose derivative, an alginate, gelatin, Polyvinylpyrrolidone; wetting agent such as glycerin; disintegrants such as agar, calcium carbonate and sodium hydrogencarbonate; absorption enhancers such as quaternary ammonium compounds; surfactants such as cetyl alcohol; adsorption carriers such as kaolin and soap Clay; lubricants such as talc, calcium stearate and magnesium stearate, and polyethylene glycol. In addition, other adjuvants such as flavoring agents, sweeteners and the like may also be added to the pharmaceutical composition.
本发明还提供了通式I所示的化合物或其互变异构体、立体异构体,立体异构体混合物、前药、药学上可接受的盐或其溶剂合物的可药用的组合物的制备方法。通常将通式I所示的含有硫普罗宁结构的化合物或其互变异构体、立体异构体,立体异构体混合物、前 药、药学上可接受的盐或其溶剂合物与可药用辅料相混合,经常规的制备方法制成适于一定途径施用的形式(剂型)。剂型包括片剂、胶囊剂、颗粒剂、丸剂、溶液剂、混悬剂、乳剂、软膏、膜剂、霜剂、气雾剂、注射剂、栓剂等。优选片剂和胶囊剂。The present invention also provides a pharmaceutically acceptable compound of the formula I or a tautomer, a stereoisomer thereof, a mixture of stereoisomers, a prodrug, a pharmaceutically acceptable salt or a solvate thereof. A method of preparing a composition. A compound containing a tiopronin structure represented by Formula I or a tautomer thereof, a stereoisomer thereof, a mixture of stereoisomers, and a former The drug, the pharmaceutically acceptable salt or the solvate thereof is mixed with a pharmaceutically acceptable adjuvant, and is prepared in a form suitable for administration by a conventional preparation method (dosage form). Dosage forms include tablets, capsules, granules, pills, solutions, suspensions, emulsions, ointments, films, creams, aerosols, injections, suppositories, and the like. Preference is given to tablets and capsules.
本发明化合物的使用剂量一般为每天1~1000mg,分单次或多次使用。但在必要时,可适当偏离上述剂量。专业人员可根据具体情况和专业知识,确定最佳剂量。这些情况包括疾病的严重程度、患者的个体差异、制剂的特性和给药途径等。The compound of the present invention is usually used in a dose of from 1 to 1000 mg per day, in single or multiple doses. However, if necessary, the above dosage can be appropriately deviated. Professionals can determine the optimal dose based on specific circumstances and expertise. These conditions include the severity of the disease, individual differences in the patient, characteristics of the formulation, and route of administration.
此外,本发明还提供了通式I所示的化合物或其互变异构体、立体异构体,立体异构体混合物、前药、药学上可接受的盐或其溶剂合物,或其可药用的组合物作为人用药物的用途。Furthermore, the present invention provides a compound of the formula I or a tautomer, a stereoisomer thereof, a mixture of stereoisomers, a prodrug, a pharmaceutically acceptable salt or a solvate thereof, or Use of a pharmaceutically acceptable composition as a human drug.
根据本发明的又一方面,本发明还提供了治疗病毒性肝炎等症的方法,所述方法包括施用治疗有效量的通式I所示的化合物或其互变异构体、立体异构体,立体异构体混合物、前药、药学上可接受的盐或其溶剂合物中的一种或多种或者本发明的所述药物组合物给患者。According to still another aspect of the present invention, the present invention provides a method of treating viral hepatitis, the method comprising administering a therapeutically effective amount of a compound of the formula I or a tautomer thereof, a stereoisomer thereof One or more of a mixture of stereoisomers, prodrugs, pharmaceutically acceptable salts or solvates thereof or the pharmaceutical composition of the present invention is administered to a patient.
本发明提供的化合物或组合物可以口服、注射(静脉、肌肉、皮下和冠状动脉内)、舌下、经颊、经直肠、经尿道、经阴道、经鼻、吸入或局部途径施用。优选的途径是口服。用于口服时,可以将其制成常规的固体制剂,如片剂、粉剂、粒剂、胶囊等,或制成液体制剂,如水或油悬浮剂,或其它液体制剂,如糖浆等;用于肠外给药时,可将其制成注射用的溶液、水或油性悬浮剂等。The compounds or compositions provided herein can be administered orally, by injection (intravenous, intramuscular, subcutaneous and intracoronary), sublingual, buccal, rectal, transurethral, vaginal, nasal, inhalation or topical routes. The preferred route is oral. When used orally, it can be prepared into a conventional solid preparation such as a tablet, a powder, a granule, a capsule, etc., or as a liquid preparation, such as a water or oil suspension, or other liquid preparation, such as a syrup; For parenteral administration, it may be prepared as a solution for injection, water or an oily suspension, or the like.
本发明还提供了通式I所示的化合物或其互变异构体、立体异构体,立体异构体混合物、前药、药学上可接受的盐或其药学上可接受的溶剂合物,在制备NS5B抑制剂的人用药物中的用途。The present invention also provides a compound of the formula I or a tautomer, a stereoisomer thereof, a mixture of stereoisomers, a prodrug, a pharmaceutically acceptable salt or a pharmaceutically acceptable solvate thereof. Use in the preparation of a human NS5B inhibitor drug.
本发明还提供了通式I所示的化合物或其互变异构体、立体异构体,立体异构体混合物、前药、药学上可接受的盐或其溶剂合物进一步与其他治疗肝病的药物联用。至少一种选自下列的治疗:干扰素、干扰素β、干扰素γ和干扰素ω;白介素,包括白介素10和白介素12;利巴韦林;干扰素α或聚乙二醇化干扰素α与利巴韦林或者左旋韦林联合使用;左旋韦林;蛋白酶抑制剂,包括NS3抑制剂,NS3/4A抑制剂;NS5A抑制剂;解旋酶抑制剂;聚合酶抑制剂,包括HCV RNA聚合酶和NS5B聚合酶抑制剂;胶霉毒素;IRES抑制剂;反义寡核苷酸;噻唑烷衍生物;N-苯甲酰苯胺,核酶;另一种核苷,核苷前药或核苷衍生物;1-氨基-烷基环己烷;抗氧化剂,包括维生素E;角鲨烯;金刚胺;胆汁酸;N-(膦酰基乙酰基)-L-天冬氨酸;苯二羧酰胺;聚腺苷酸;苯并咪唑;胸腺素;预防疫苗;免疫调节剂,一种IMPDH抑制剂;水飞蓟素;水飞蓟素-磷脂酰胆碱内涵体;和麦考酚酸酯。The present invention also provides a compound represented by Formula I, or a tautomer, a stereoisomer thereof, a mixture of stereoisomers, a prodrug, a pharmaceutically acceptable salt or a solvate thereof, and other therapeutic liver diseases. The combination of drugs. At least one treatment selected from the group consisting of interferon, interferon beta, interferon gamma, and interferon ω; interleukins, including interleukin 10 and interleukin 12; ribavirin; interferon alpha or pegylated interferon alpha Combination of ribavirin or levovirin; levovirin; protease inhibitors, including NS3 inhibitors, NS3/4A inhibitors; NS5A inhibitors; helicase inhibitors; polymerase inhibitors, including HCV RNA polymerase And NS5B polymerase inhibitor; colloidal toxin; IRES inhibitor; antisense oligonucleotide; thiazolidine derivative; n-benzidine, ribozyme; another nucleoside, nucleoside prodrug or nucleoside Derivatives; 1-amino-alkylcyclohexane; antioxidants, including vitamin E; squalene; amantadine; bile acid; N-(phosphonoacetyl)-L-aspartic acid; benzene dicarboxamide Polyadenylation; benzimidazole; thymosin; prophylactic vaccine; immunomodulator, an IMPDH inhibitor; silymarin; silymarin-phosphatidylcholine endosome; and mycophenolate mofetil.
“药学上可接受的”是指这样一些化合物、原料、组合物和/或剂型,它们在合理医学判断的范围内,适用于与患者组织接触而无过度毒性、刺激性、变态反应或与合理的利益/风险比相对称的其他问题和并发症,并有效用于既定用途。"Pharmaceutically acceptable" means a compound, material, composition, and/or dosage form that, within the scope of sound medical judgment, is suitable for contact with a patient's tissue without undue toxicity, irritation, allergies, or reasonable The benefits/risk ratios are commensurate with other problems and complications and are effectively used for the intended use.
术语“卤素”和“卤代”在本发明中可互换使用,是指氟(F)、氯(Cl)、溴(Br)或碘(I)。The terms "halogen" and "halo" are used interchangeably herein and refer to fluorine (F), chlorine (Cl), bromine (Br) or iodine (I).
本发明使用的术语“烷基”或“烷基基团”,表示含有1-20个碳原子,饱和的直链或支链一价烃基基团,其中,所述烷基基团可以任选地被一个或多个本发明描述的取代基所取代。在一实施方案中,烷基基团含有1-6个碳原子;在另一实施方案中,烷基基团含有1-4个碳原子;还在一实施方案中,烷基基团含有1-3个碳原子。烷基基团的实例包含,但并不限于,甲基(Me、-CH3),乙基(Et、-CH2CH3),正丙基(n-Pr、-CH2CH2CH3),异丙基(i-Pr、-CH(CH3)2),正丁基(n-Bu、-CH2CH2CH2CH3),异丁基(i-Bu、-CH2CH(CH3)2),仲丁基(s-Bu、-CH(CH3)CH2CH3),叔丁基(t-Bu、-C(CH3)3),等等。The term "alkyl" or "alkyl group" as used herein, denotes a saturated straight or branched monovalent hydrocarbon group containing from 1 to 20 carbon atoms, wherein the alkyl group may be optionally selected The ground is replaced by one or more substituents described herein. In one embodiment, the alkyl group contains from 1 to 6 carbon atoms; in another embodiment, the alkyl group contains from 1 to 4 carbon atoms; and in one embodiment, the alkyl group contains 1 - 3 carbon atoms. Examples of alkyl groups include, but are not limited to, methyl (Me, -CH 3 ), ethyl (Et, -CH 2 CH 3 ), n-propyl (n-Pr, -CH 2 CH 2 CH 3 ), isopropyl (i-Pr, -CH(CH 3 ) 2 ), n-butyl (n-Bu, -CH 2 CH 2 CH 2 CH 3 ), isobutyl (i-Bu, -CH 2 CH) (CH 3 ) 2 ), sec-butyl (s-Bu, -CH(CH 3 )CH 2 CH 3 ), tert-butyl (t-Bu, -C(CH 3 ) 3 ), and the like.
本发明所使用的“药学上可接受的盐”是指本发明的化合物的有机盐和无机盐。药学上可接受的盐在所属领域是为我们所熟知的,如文献:S.M.Berge et al.,describe pharmaceutically acceptable salts in detail in J.Pharmaceutical Sciences,1977,66:1-19.所记 载的。药学上可接受的无毒的酸形成的盐包括,但并不限于,与氨基基团反应形成的无机酸盐有盐酸盐,氢溴酸盐,磷酸盐,硫酸盐,高氯酸盐,和有机酸盐如乙酸盐,草酸盐,马来酸盐,酒石酸盐,柠檬酸盐,琥珀酸盐,丙二酸盐,或通过书籍文献上所记载的其他方法如离子交换法来得到这些盐。其他药学上可接受的盐包括己二酸盐,藻酸盐,抗坏血酸盐,天冬氨酸盐,苯磺酸盐,苯甲酸盐,重硫酸盐,硼酸盐,丁酸盐,樟脑酸盐,樟脑磺酸盐,环戊基丙酸盐,二葡萄糖酸盐,十二烷基硫酸盐,乙磺酸盐,甲酸盐,反丁烯二酸盐,葡庚糖酸盐,甘油磷酸盐,葡萄糖酸盐,半硫酸盐,庚酸盐,己酸盐,氢碘酸盐,2-羟基-乙磺酸盐,乳糖醛酸盐,乳酸盐,月桂酸盐,月桂基硫酸盐,苹果酸盐,甲磺酸盐,2-萘磺酸盐,烟酸盐,硝酸盐,油酸盐,棕榈酸盐,扑酸盐,果胶酸盐,过硫酸盐,3-苯基丙酸盐,苦味酸盐,特戊酸盐,丙酸盐,硬脂酸盐,硫氰酸盐,对甲苯磺酸盐,十一酸盐,戊酸盐,等等。通过适当的碱得到的盐包括碱金属,碱土金属,铵和N+(C1-4烷基)4的盐。本发明也拟构思了任何所包含N的基团的化合物所形成的季铵盐。水溶性或油溶性或分散产物可以通过季铵化作用得到。碱金属或碱土金属盐包括钠,锂,钾,钙,镁,等等。药学上可接受的盐进一步包括适当的、无毒的铵,季铵盐和抗平衡离子形成的胺阳离子,如卤化物,氢氧化物,羧化物,硫酸化物,磷酸化物,硝酸化物,C1-C8磺酸化物和芳香磺酸化物。The "pharmaceutically acceptable salt" as used in the present invention means an organic salt and an inorganic salt of the compound of the present invention. Pharmaceutically acceptable salts are well known in the art and are described in the literature: SMBerge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66: 1-19. Salts formed by pharmaceutically acceptable non-toxic acids include, but are not limited to, mineral acid salts formed by reaction with amino groups, hydrochloride, hydrobromide, phosphate, sulfate, perchlorate, And organic acid salts such as acetates, oxalates, maleates, tartrates, citrates, succinates, malonates, or by other methods described in the literature, such as ion exchange These salts. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, besylate, benzoate, disulfate, borate, butyrate, camphoric acid Salt, camphor sulfonate, cyclopentylpropionate, digluconate, lauryl sulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate Salt, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, Malate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, palmitate, pamoate, pectate, persulphate, 3-phenylpropionic acid Salt, picrate, pivalate, propionate, stearate, thiocyanate, p-toluenesulfonate, undecanoate, valerate, and the like. Salts obtained by appropriate bases include the alkali metal, alkaline earth metal, ammonium and N + (C 1-4 alkyl) 4 salts. The present invention also contemplates quaternary ammonium salts formed from any of the compounds comprising a group of N. Water soluble or oil soluble or dispersed products can be obtained by quaternization. Alkali metal or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Pharmaceutically acceptable salts further comprise suitable amine cation nontoxic ammonium, quaternary ammonium, and the counterion, such as halide, hydroxide, carboxylate, sulfated, phosphorylated compounds, nitrate compounds, C 1 -C 8 sulfonate and aromatic sulfonate.
本发明的“溶剂化物”是指一个或多个溶剂分子与本发明的化合物所形成的缔合物。形成溶剂化物的溶剂包括,但并不限于,水,异丙醇,乙醇,甲醇,二甲亚砜,乙酸乙酯,乙酸、乙醇胺或其混合物。术语“水合物”是指溶剂分子是水所形成的缔合物。"Solvate" as used herein refers to an association of one or more solvent molecules with a compound of the invention. Solvent-forming solvents include, but are not limited to, water, isopropanol, ethanol, methanol, dimethyl sulfoxide, ethyl acetate, acetic acid, ethanolamine or mixtures thereof. The term "hydrate" means that the solvent molecule is an association formed by water.
当所述溶剂为水时,可以使用术语“水合物”。在一实施方案中,一个本发明化合物分子可以与一个水分子相结合,比如一水合物;在另一实施方案中,一个本发明化合物分子可以与多于一个的水分子相结合,比如二水合物,在又一实施方案中,一个本发明化合物分子可以与少于一个的水分子相结合,比如半水合物。应注意,本发明所述的水合物保留有非水合形式的所述化合物的生物有效性。When the solvent is water, the term "hydrate" can be used. In one embodiment, a molecule of the compound of the invention may be combined with a water molecule, such as a monohydrate; in another embodiment, a molecule of the invention may be combined with more than one water molecule, such as dihydrate. In yet another embodiment, a molecule of the compound of the invention may be combined with less than one water molecule, such as a hemihydrate. It should be noted that the hydrates of the present invention retain the bioavailability of the compounds in a non-hydrated form.
除非另作说明,本发明的化合物所有合适的同位素变化、立体异构体、互变异构体、溶剂化物、代谢产物、盐和药学上可接受的前药都包含在本发明范围内。All suitable isotopic variations, stereoisomers, tautomers, solvates, metabolites, salts and pharmaceutically acceptable prodrugs of the compounds of the invention are included within the scope of the invention unless otherwise stated.
在本发明公开的结构中,当任意特定的手性原子的立体化学未指明时,则该结构的所有立体异构体都考虑在本发明之内,并且作为本发明公开化合物包括在本发明中。当立体化学被表示特定构型的实楔形线(solid wedge)或虚线指明时,则该结构的立体异构体就此明确和定义。In the structures disclosed herein, when the stereochemistry of any particular chiral atom is not indicated, all stereoisomers of the structure are contemplated within the invention, and as disclosed herein are included in the present invention. . When stereochemistry is indicated by a solid wedge or dashed line indicating a particular configuration, then the stereoisomers of the structure are defined and defined herein.
式I所示化合物可以以盐的形式存在。在一实施方案中,所述盐是指药学上可接受的盐。术语“药学上可接受的”是指物质或组合物必须与包含制剂的其它成分和/或用其治疗的哺乳动物化学上和/或毒理学上相容。在另一实施方案中,所述盐不一定是药学上可接受的盐,可以是用于制备和/或提纯式I所示化合物和/或用于分离本式I所示化合物的对映体的中间体。The compound of formula I can exist in the form of a salt. In one embodiment, the salt refers to a pharmaceutically acceptable salt. The term "pharmaceutically acceptable" means that the substance or composition must be chemically and/or toxicologically compatible with the other ingredients comprising the formulation and/or the mammal treated therewith. In another embodiment, the salt is not necessarily a pharmaceutically acceptable salt, and may be an enantiomer for the preparation and/or purification of a compound of formula I and/or for isolation of a compound of formula I. Intermediates.
本发明的可药用盐可以用常规化学方法由母体化合物、碱性或酸性部分来合成。一般而言,该类盐可以通过使这些化合物的游离酸形式与化学计量量的适宜碱(如Na、Ca、Mg或K的氢氧化物、碳酸盐、碳酸氢盐等)反应,或者通过使这些化合物的游离碱形式与化学计量量的适宜酸反应来进行制备。该类反应通常在水或有机溶剂或二者的混合物中进行。一般地,在适当的情况中,需要使用非水性介质如乙醚、乙酸乙酯、乙醇、异丙醇或乙腈。在例如“Remington′s Pharmaceutical Sciences”,第20版,Mack Publishing Company,Easton,Pa.,(1985);和“药用盐手册:性质、选择和应用(Handbook of Pharmaceutical Salts:Properties,Selection,and Use)”,Stahl and Wermuth(Wiley-VCH,Weinheim,Germany,2002)中可找到另外一些适宜盐的列表。The pharmaceutically acceptable salts of the present invention can be synthesized from the parent compound, basic or acidic moiety by conventional chemical methods. In general, such salts can be obtained by reacting the free acid form of these compounds with a stoichiometric amount of a suitable base such as a hydroxide, carbonate, bicarbonate, or the like of Na, Ca, Mg or K. The free base form of these compounds is prepared by reaction with a stoichiometric amount of a suitable acid. This type of reaction is usually carried out in water or an organic solvent or a mixture of the two. Generally, where appropriate, it is desirable to use a non-aqueous medium such as diethyl ether, ethyl acetate, ethanol, isopropanol or acetonitrile. For example, "Remington's Pharmaceutical Sciences", 20th Edition, Mack Publishing Company, Easton, Pa., (1985); and "Handbook of Pharmaceutical Salts: Properties, Selection, and A list of additional suitable salts can be found in Use), Stahl and Wermuth (Wiley-VCH, Weinheim, Germany, 2002).
本发明给出的任何结构式也意欲表示这些化合物未被同位素富集的形式以及同位素 富集的形式。同位素富集的化合物具有本发明给出的通式描绘的结构,除了一个或多个原子被具有所选择原子量或质量数的原子替换。可引入本发明化合物中的示例性同位素包括氢、碳、氮、氧、磷、硫、氟和氯的同位素,如2H、3H、11C、13C、14C、15N、17O、18O、18F、31P、32P、35S、36Cl和125I。Any structural formula given by the present invention is also intended to indicate that these compounds are not isotopically enriched and isotopically enriched. Isotopically enriched compounds have the structure depicted by the general formula given herein, except that one or more atoms are replaced by an atom having a selected atomic mass or mass number. Exemplary isotopes that may be incorporated into the compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine, and chlorine, such as 2 H, 3 H, 11 C, 13 C, 14 C, 15 N, 17 O 18 O, 18 F, 31 P, 32 P, 35 S, 36 Cl and 125 I.
另一方面,本发明涉及制备式I所示化合物的中间体。In another aspect, the invention relates to intermediates for the preparation of compounds of formula I.
另一方面,本发明提供一种药物组合物,所述药物组合物包含本发明化合物。在一实施方案中,本发明所述药物组合物,更进一步包括药学上可接受的载体、赋形剂、佐剂、溶媒或它们的组合。在另一实施方案中,药物组合物可以是液体、固体、半固体、凝胶或喷雾剂型。In another aspect, the invention provides a pharmaceutical composition comprising a compound of the invention. In one embodiment, the pharmaceutical composition of the present invention further comprises a pharmaceutically acceptable carrier, excipient, adjuvant, vehicle, or a combination thereof. In another embodiment, the pharmaceutical composition can be in the form of a liquid, solid, semi-solid, gel or spray.
本发明的该类化合物或其药物组合物可用于丙型肝炎的治疗,具有口服生物利用度高、代谢性质好的优点,同时具有保护肝组织、肝细胞、改善肝功能的作用。The compound of the present invention or a pharmaceutical composition thereof can be used for the treatment of hepatitis C, and has the advantages of high oral bioavailability and good metabolic properties, and has the functions of protecting liver tissue, liver cells, and improving liver function.
具体实施方式detailed description
为描述本发明,以下列出了实施例。但需要理解,本发明不限于这些实施例,只是提供实践本发明的方法。For the purpose of describing the invention, the examples are set forth below. However, it is to be understood that the invention is not limited to the embodiments, but merely provides a method of practicing the invention.
一般地,本发明的化合物可以通过本发明所描述的方法制备得到,除非有进一步的说明,其中取代基的定义如式I所示。下面的反应方案和实施例用于进一步举例说明本发明的内容。In general, the compounds of the present invention can be prepared by the methods described herein, unless otherwise stated, wherein the substituents are as defined in Formula I. The following reaction schemes and examples are provided to further illustrate the contents of the present invention.
所属领域的专业人员将认识到:本发明所描述的化学反应可以用来合适地制备许多本发明的其他化合物,且用于制备本发明的化合物的其它方法都被认为是在本发明的范围之内。例如,根据本发明那些非例证的化合物的合成可以成功地被所属领域的技术人员通过修饰方法完成,如适当的保护干扰基团,通过利用其他已知的试剂除了本发明所描述的,或将反应条件做一些常规的修改。另外,本发明所公开的反应或已知的反应条件也公认地适用于本发明其他化合物的制备。Those skilled in the art will recognize that the chemical reactions described herein can be used to suitably prepare a number of other compounds of the invention, and that other methods for preparing the compounds of the invention are considered to be within the scope of the invention. Inside. For example, the synthesis of those non-exemplified compounds according to the present invention can be successfully accomplished by modifications by those skilled in the art, such as appropriate protection of the interfering group, by the use of other known reagents in addition to those described herein, or The reaction conditions are subject to some conventional modifications. Additionally, the reactions or known reaction conditions disclosed herein are also recognized to be suitable for the preparation of other compounds of the invention.
下面所描述的实施例,除非其他方面表明所有的温度定为摄氏度。试剂购买于商品供应商如Aldrich Chemical Company,Arco Chemical Company and Alfa Chemical Company,使用时都没有经过进一步纯化,除非其他方面表明。一般的试剂从汕头西陇化工厂,广东光华化学试剂厂,广州化学试剂厂,天津好寓宇化学品有限公司,天津市福晨化学试剂厂,武汉鑫华远科技发展有限公司,青岛腾龙化学试剂有限公司,和青岛海洋化工厂购买得到。The examples described below, unless otherwise indicated, all temperatures are set to degrees Celsius. The reagents were purchased from commercial suppliers such as Aldrich Chemical Company, Arco Chemical Company and Alfa Chemical Company and were used without further purification unless otherwise indicated. General reagents from Shantou Xiqiao Chemical Plant, Guangdong Guanghua Chemical Reagent Factory, Guangzhou Chemical Reagent Factory, Tianjin Haoyuyu Chemical Co., Ltd., Tianjin Fuchen Chemical Reagent Factory, Wuhan Xinhuayuan Technology Development Co., Ltd., Qingdao Tenglong Chemical Reagent Co., Ltd., and Qingdao Ocean Chemical Plant purchased it.
无水四氢呋喃,二氧六环,甲苯,乙醚是经过金属钠回流干燥得到。无水二氯甲烷和氯仿是经过氢化钙回流干燥得到。乙酸乙酯,石油醚,正己烷,N,N-二甲基乙酰胺和N,N-二甲基甲酰胺是经无水硫酸钠事先干燥使用。Anhydrous tetrahydrofuran, dioxane, toluene and diethyl ether are obtained by refluxing with sodium metal. Anhydrous dichloromethane and chloroform were obtained by reflux drying of calcium hydride. Ethyl acetate, petroleum ether, n-hexane, N,N-dimethylacetamide and N,N-dimethylformamide were previously dried over anhydrous sodium sulfate.
以下反应一般是在氮气或氩气正压下或在无水溶剂上套一干燥管(除非其他方面表明),反应瓶都塞上合适的橡皮塞,底物通过注射器打入。玻璃器皿都是干燥过的。The following reaction is generally carried out under a positive pressure of nitrogen or argon or on a dry solvent (unless otherwise indicated), the reaction bottle is stoppered with a suitable rubber stopper, and the substrate is driven through a syringe. The glassware is dry.
色谱柱是使用硅胶柱。硅胶(300-400目)购于青岛海洋化工厂。The column is a silica gel column. Silica gel (300-400 mesh) was purchased from Qingdao Ocean Chemical Plant.
1H NMR谱使用Bruker 400MHz或600MHz核磁共振谱仪记录。1H NMR谱以CDC13、DMSO-d6、CD3OD或丙酮-d6为溶剂(以ppm为单位),用TMS(0ppm)或氯仿(7.26ppm)作为参照标准。当出现多重峰的时候,将使用下面的缩写:s(singlet,单峰)、d(doublet,双峰)、t(triplet,三重峰)、m(multiplet,多重峰)、br(broadened,宽峰)、dd(doublet of doublets,双二重峰)、dt(doublet of triplets,双三重峰)。偶合常数,用赫兹(Hz)表示。 1 H NMR spectra were recorded using a Bruker 400 MHz or 600 MHz NMR spectrometer. The 1 H NMR spectrum was determined by using CDC1 3 , DMSO-d 6 , CD 3 OD or acetone-d 6 as a solvent (in ppm) using TMS (0 ppm) or chloroform (7.26 ppm) as a reference standard. When multiple peaks appear, the following abbreviations are used: s (singlet, single peak), d (doublet, doublet), t (triplet, triplet), m (multiplet, multiplet), br (broadened, wide) Peak), dd (doublet of doublets), dt (doublet of triplets). Coupling constant, expressed in Hertz (Hz).
低分辨率质谱(MS)数据的测定条件是:Agilent 6120四级杆HPLC-M(柱子型号:Zorbax SB-C18,2.1x 30mm,3.5微米,6min,流速为0.6mL/min。流动相:5%-95%(含0.1%甲酸的CH3CN)在(含0.1%甲酸的H2O)中的比例,采用电喷雾电离(ESI),在210nm/254nm下,用UV检测。The conditions for the determination of low resolution mass spectrometry (MS) data were: Agilent 6120 quadrupole HPLC-M (column model: Zorbax SB-C18, 2.1 x 30 mm, 3.5 micron, 6 min, flow rate 0.6 mL/min. Mobile phase: 5 ratio of 95% (0.1% formic acid in CH 3 CN) in (H containing 0.1% formic acid 2 O) in using electrospray ionization (ESI), at 210nm / 254nm, with UV detection.
纯的化合物使用Agilent 1260pre-HPLC或Calesep pump 250pre-HPLC(柱子型号: NOVASEP 50/80mm DAC),在210nm/254nm用UV检测。Pure compound using Agilent 1260 pre-HPLC or Calesep pump 250 pre-HPLC (column model: NOVASEP 50/80mm DAC), UV detection at 210nm/254nm.
下面简写词的使用贯穿本发明:The following abbreviations are used throughout the invention:
Figure PCTCN2016109913-appb-000066
Figure PCTCN2016109913-appb-000066
实施例1:化合物II-A-1的合成Example 1: Synthesis of Compound II-A-1
方法A:Method A:
Figure PCTCN2016109913-appb-000067
Figure PCTCN2016109913-appb-000067
步骤一:将529mg(1.0mmol)III-1溶于10mL无水丙酮中,室温下加入680mg(1.5mmol)IV-A-1及碳酸钾414mg(3.0mmol),加热搅拌回流6h,TLC检测反应完全。过滤,滤液用二氯甲烷(30mL)稀释,依次用水(10mL)及饱和食盐水(10mL)洗涤,分离有机相,无水硫酸钠干燥后过滤,减压浓缩除去溶剂。剩余物硅胶柱层析纯化得710mg白色固体V-A-1,收率75%,ESI-MS:m/z[M+H]+=947。Step 1: 529 mg (1.0 mmol) of III-1 was dissolved in 10 mL of anhydrous acetone, and 680 mg (1.5 mmol) of IV-A-1 and potassium carbonate 414 mg (3.0 mmol) were added at room temperature, and the mixture was stirred under reflux for 6 h. complete. After filtration, the filtrate was diluted with methylene chloride (30 mL). The residue was purified by silica gel column chromatography to give 710mg white solid VA-1, yield 75%, ESI-MS: m / z [M + H] + = 947.
步骤二:将200mg(0.21mmol)V-A-1溶于10mL无水二氯甲烷中,室温下加入2mL(体积比:二氯甲烷/三异丙基硅烷/三氟乙酸=5/1/1)溶液,反应约1h,TLC检测反应完全,小心加入饱和碳酸氢钠溶液,分离有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩。剩余物硅胶柱层析纯化得到96mg白色固体II-A-1,收率65%。1H NMR(400MHz,DMSO-d6)δ8.43(t,J=5.7Hz,1H),7.68(d,J=7.2Hz,1H),7.39(t,J=7.7Hz,2H),7.30–7.14(m,3H),6.14–5.96(m,2H),5.90(d,J=4.9Hz,1H),5.84(d,J=9.6Hz,1H),5.81(d,J=9.6Hz,1H),5.74(d,J=8.2Hz,1H),4.87(dt,J=12.4,6.2Hz,1H),4.47–4.34(m,1H),4.29–4.20(m,1H),4.08–4.01(m,1H),3.94–3.76(m,4H),3.57–3.46(m,1H),2.82(d,J=8.3Hz,1H),1.35(d,J=6.9Hz,3H),1.27(t,J=15.0Hz,6H),1.16(d,J=6.2Hz,6H).13C NMR(100MHz,DMSO-d6)δ173.7,173.1,173.0,169.2,161.1,151.2,151.1,150.6,130.1,125.1,120.6,120.5,101.9,101.6,99.8,80.2,72.1,71.9,68.5,65.1,64.9,50.3,41.0,36.3,22.4,22.3,21.9,21.8,20.3,20.2,17.2,16.9.31P NMR(162MHz,DMSO)δ3.83(s)ESI-MS:m/z[M+H]+=705。 Step 2: Dissolve 200 mg (0.21 mmol) of VA-1 in 10 mL of anhydrous dichloromethane, and add 2 mL at room temperature (volume ratio: dichloromethane / triisopropylsilane / trifluoroacetic acid = 5 / 1 / 1) The solution was reacted for about 1 h, and the reaction was completed by TLC. EtOAc (EtOAc m. The residue was purified by silica gel column chromatography toieliel 1 H NMR (400 MHz, DMSO-d 6 ) δ 8.43 (t, J = 5.7 Hz, 1H), 7.68 (d, J = 7.2 Hz, 1H), 7.39 (t, J = 7.7 Hz, 2H), 7.30 –7.14(m,3H), 6.14–5.96(m,2H), 5.90 (d, J=4.9Hz, 1H), 5.84 (d, J=9.6Hz, 1H), 5.81 (d, J=9.6Hz, 1H), 5.74 (d, J = 8.2 Hz, 1H), 4.87 (dt, J = 12.4, 6.2 Hz, 1H), 4.47 - 4.34 (m, 1H), 4.29 - 4.20 (m, 1H), 4.08 - 4.01 (m, 1H), 3.94 - 3.76 (m, 4H), 3.57 - 3.46 (m, 1H), 2.82 (d, J = 8.3 Hz, 1H), 1.35 (d, J = 6.9 Hz, 3H), 1.27 ( t, J = 15.0 Hz, 6H), 1.16 (d, J = 6.2 Hz, 6H). 13 C NMR (100 MHz, DMSO-d 6 ) δ 173.7, 173.1, 173.0, 169.2, 161.1, 151.2, 151.1, 150.6, 130.1 , 125.1, 120.6, 120.5, 101.9, 101.6, 99.8, 80.2, 72.1, 71.9, 68.5, 65.1, 64.9, 50.3, 41.0, 36.3, 22.4, 22.3, 21.9, 21.8, 20.3, 20.2, 17.2, 16.9. 31 P NMR (162 MHz, DMSO) δ 3.83 (s) ESI-MS: m/z [M+H] + = 705.
方法B:Method B:
Figure PCTCN2016109913-appb-000068
Figure PCTCN2016109913-appb-000068
步骤一:将5.0g(9.5mmol)化合物III-1溶于25mL无水N,N-二甲基甲酰胺(DMF)中,在Ar保护下依次加入1.6g(23.5mmol)咪唑,0.12g(1.0mmol)4-二甲氨基吡啶(DMAP)以及1.6mL(9.5mmol)三乙基氯硅烷(TESCl)。在室温下搅拌反应约10小时,TLC检测反应完全。然后往反应体系中加入7mL 37%HCHO溶液,加热至80度搅拌反应1小时。冷却至室温,加入80mL乙酸乙酯,依次用水、饱和食盐水洗涤,分离有机相用无水硫酸钠干燥,过滤后减压浓缩,得无色油状物VII-1,不经纯化直接用于下一步。Step 1: 5.0 g (9.5 mmol) of compound III-1 was dissolved in 25 mL of anhydrous N,N-dimethylformamide (DMF), and 1.6 g (23.5 mmol) of imidazole, 0.12 g (stepwise) was added under Ar protection. 1.0 mmol) 4-dimethylaminopyridine (DMAP) and 1.6 mL (9.5 mmol) of triethylchlorosilane (TESCl). The reaction was stirred at room temperature for about 10 hours, and the reaction was completed by TLC. Then, 7 mL of a 37% HCHO solution was added to the reaction system, and the mixture was heated to 80 ° C and stirred for 1 hour. After cooling to room temperature, 80 mL of ethyl acetate was added, and the mixture was washed with EtOAc. step.
步骤二:将上一步新鲜所得的产物VII-1溶于40mL无水二氯甲烷(DCM)中,在氩气保护下冰浴冷却,依次加入62mg(0.5mmol)4-二甲氨基吡啶(DMAP)和2.36g(12.3mmol)1-乙基-(3-二甲基氨基丙基)碳酰二亚胺盐酸盐(EDC HCl),及6.1g(11.4mmol)化合物VIII-A-1,慢慢升至室温并搅拌反应约12小时,TLC检测反应完全。加入100mL二氯甲烷,依次用饱和碳酸氢钠水溶液、饱和食盐水洗涤,分离有机相用无水硫酸钠干燥,过滤后减压浓缩。粗产物用二氯甲烷-正庚烷重结晶得8.5g白色固体产物IX-A-1,收率70%(二步)。1H NMR(400MHz,CDCl3)δ7.57(d,J=8.1Hz,1H),7.35(dd,J=17.2,7.6Hz,6H),7.31–7.12(m,12H),6.77(d,J=8.5Hz,2H),6.17(d,J=18.4Hz,1H),6.04–5.87(m,2H),5.68(d,J=8.2Hz,1H),5.00(dt,J=12.3,6.2Hz,1H),4.60(dd,J=11.4,6.4Hz,1H),4.35(q,J=6.9Hz,1H),4.29–4.20(m,2H),4.15–4.08(m,1H),4.02–3.85(m,3H),3.78(s,3H),3.71(t,J=10.0Hz,1H),1.39–1.32(m,6H),1.35(s,9H),1.27(d,J=11.9Hz,3H),1.23(d,J=6.2Hz,6H),0.92(s,9H),0.15(s,6H).ESI-MS:m/z[M+H]+=1191。Step 2: The product VII-1 freshly obtained in the previous step was dissolved in 40 mL of anhydrous dichloromethane (DCM), cooled in an ice bath under argon atmosphere, and then added 62 mg (0.5 mmol) of 4-dimethylaminopyridine (DMAP). And 2.36 g (12.3 mmol) of 1-ethyl-(3-dimethylaminopropyl)carbodiimide hydrochloride (EDC HCl), and 6.1 g (11.4 mmol) of compound VIII-A-1, The temperature was slowly raised to room temperature and the reaction was stirred for about 12 hours, and the reaction was completed by TLC. After adding 100 ml of dichloromethane, the mixture was washed with aq. The crude product was recrystallized from methylene chloride-n-heptane to afford 8.5 g of y. 1 H NMR (400MHz, CDCl 3 ) δ7.57 (d, J = 8.1Hz, 1H), 7.35 (dd, J = 17.2,7.6Hz, 6H), 7.31-7.12 (m, 12H), 6.77 (d, J = 8.5 Hz, 2H), 6.17 (d, J = 18.4 Hz, 1H), 6.04 - 5.87 (m, 2H), 5.68 (d, J = 8.2 Hz, 1H), 5.00 (dt, J = 12.3, 6.2 Hz, 1H), 4.60 (dd, J = 11.4, 6.4 Hz, 1H), 4.35 (q, J = 6.9 Hz, 1H), 4.29 - 4.20 (m, 2H), 4.15 - 4.08 (m, 1H), 4.02 – 3.85 (m, 3H), 3.78 (s, 3H), 3.71 (t, J = 10.0 Hz, 1H), 1.39 - 1.32 (m, 6H), 1.35 (s, 9H), 1.27 (d, J = 11.9) Hz, 3H), 1.23 (d, J = 6.2 Hz, 6H), 0.92 (s, 9H), 0.15 (s, 6H). ESI-MS: m/z [M+H] + =1191.
步骤三:将7.5g(6.3mmol)化合物IX-A-1溶于50%水-丙酮(20mL)混合溶液中,加入30mL冰醋酸(HOAc)及6mL三氟乙酸(TFA),室温下搅拌反应约6小时,至TLC检测不到剩余原料。减压旋蒸除去丙酮、水,然后依次加入60mL二氯甲烷,9ml三氟乙酸(TFA),6.5mL三异丙基硅烷(i-Pr3SiH),在室温下继续反应约15分钟,TLC检测反应完全,小心加入饱和碳酸氢钠水溶液萃灭反应。加入100mL二氯甲烷,依次用水、饱和食盐水洗涤,分离有机相用无水硫酸钠干燥,过滤后减压浓缩得粗产物。用乙酸乙酯-石油醚重结晶得3.9g白色固体II-A-1,收率88%。1H NMR(400MHz,DMSO)δ8.43(t,J=5.7Hz,1H),7.68(d,J=7.2Hz,1H),7.39(t,J=7.7Hz,2H),7.30–7.14(m,3H),6.14–5.96(m,2H),5.90(d,J=4.9Hz,1H),5.84(d,J=9.6Hz,1H),5.81(d,J=9.6Hz,1H),5.74(d,J=8.2Hz,1H),4.87(dt,J=12.4,6.2Hz,1H),4.47–4.34(m,1H),4.29–4.20(m,1H),4.08–4.01(m,1H),3.94–3.76(m,4H),3.57–3.46(m,1H),2.82(d,J=8.3Hz,1H),1.35(d,J=6.9Hz,3H),1.27(t,J=15.0Hz,6H),1.16(d,J=6.2Hz,6H).ESI-MS:m/z[M+H]+=705。Step 3: 7.5 g (6.3 mmol) of compound IX-A-1 was dissolved in 50% water-acetone (20 mL) mixed solution, 30 mL of glacial acetic acid (HOAc) and 6 mL of trifluoroacetic acid (TFA) were added, and the reaction was stirred at room temperature. After about 6 hours, no remaining raw materials were detected by TLC. The acetone and water were removed by vacuum distillation under reduced pressure, and then 60 mL of dichloromethane, 9 ml of trifluoroacetic acid (TFA), and 6.5 mL of triisopropylsilane ( i- Pr 3 SiH) were successively added, and the reaction was continued at room temperature for about 15 minutes, TLC The reaction was checked to completion and the reaction was quenched by carefully aqueous saturated sodium bicarbonate. After adding 100 ml of dichloromethane, the mixture was washed with water and aq. Recrystallization from ethyl acetate-petroleum ether gave 3.9 g of white solid II-A-1. 1 H NMR (400 MHz, DMSO) δ 8.43 (t, J = 5.7 Hz, 1H), 7.68 (d, J = 7.2 Hz, 1H), 7.39 (t, J = 7.7 Hz, 2H), 7.30 - 7.14 ( m, 3H), 6.14 - 5.96 (m, 2H), 5.90 (d, J = 4.9 Hz, 1H), 5.84 (d, J = 9.6 Hz, 1H), 5.81 (d, J = 9.6 Hz, 1H), 5.74 (d, J = 8.2 Hz, 1H), 4.87 (dt, J = 12.4, 6.2 Hz, 1H), 4.47 - 4.34 (m, 1H), 4.29 - 4.20 (m, 1H), 4.08 - 4.01 (m, 1H), 3.94–3.76 (m, 4H), 3.57–3.46 (m, 1H), 2.82 (d, J=8.3 Hz, 1H), 1.35 (d, J=6.9 Hz, 3H), 1.27 (t, J) =15.0 Hz, 6H), 1.16 (d, J = 6.2 Hz, 6H). ESI-MS: m/z [M+H] + = 705.
将重结晶母液浓缩,残余物使用硅胶柱层析纯化得到少量白色固体II-A-9,1H NMR(400MHz,CDCl3)δ7.51(d,J=7.9Hz,1H),7.35(t,J=7.8Hz,2H),7.20(dd,J=14.5,7.5Hz,3H),6.19(brs,1H),6.07(d,J=9.4Hz,1H),5.99(d,J=9.4Hz,1H),5.72(d,J=8.2Hz,1H), 5.00(dt,J=12.5,6.2Hz,1H),4.59–4.39(m,2H),4.37–4.22(m,2H),4.14(d,J=8.6Hz,1H),3.99–3.85(m,3H),3.65–3.58(m,1H),1.48(d,J=7.0Hz,1H),1.42–1.36(m,3H),1.38–1.26(m,6H),1.24(d,J=6.2Hz,6H).ESI-MS:m/z[M+H]+=705。The recrystallization mother liquor was concentrated and the residue was purified by silica gel column chromatography using a small amount of white solid to give II-A-9, 1 H NMR (400MHz, CDCl 3) δ7.51 (d, J = 7.9Hz, 1H), 7.35 (t , J = 7.8 Hz, 2H), 7.20 (dd, J = 14.5, 7.5 Hz, 3H), 6.19 (brs, 1H), 6.07 (d, J = 9.4 Hz, 1H), 5.99 (d, J = 9.4 Hz) , 1H), 5.72 (d, J = 8.2 Hz, 1H), 5.00 (dt, J = 12.5, 6.2 Hz, 1H), 4.59 - 4.39 (m, 2H), 4.37 - 4.22 (m, 2H), 4.14 ( d, J=8.6 Hz, 1H), 3.99–3.85 (m, 3H), 3.65–3.58 (m, 1H), 1.48 (d, J=7.0 Hz, 1H), 1.42–1.36 (m, 3H), 1.38 - 1.26 (m, 6H), 1.24 (d, J = 6.2 Hz, 6H). ESI-MS: m/z [M+H] + = 705.
实施例2:化合物II-A-1a的合成Example 2: Synthesis of Compound II-A-1a
Figure PCTCN2016109913-appb-000069
Figure PCTCN2016109913-appb-000069
依实施例1中方法B步骤一新鲜制得化合物VII-1(3mmol化合物III-1为起始原料),溶于12mL无水二氯甲烷(DCM)中,在氩气保护下冰浴冷却,依次加入18mg(0.15mmol)4-二甲氨基吡啶(DMAP)和0.75g(3.9mmol)1-乙基-(3-二甲基氨基丙基)碳酰二亚胺盐酸盐(EDC HCl),及1.93g(3.6mmol)化合物VIII-A-1a(由手性硫普罗宁制备得到,R构型手性纯度95%,分析方法:AD-H柱温:25C;检测波长:210nm;流速:1.0ml/min;流动相:n-Hex:EtOH:TFA=90:10:0.1等度洗脱),然后慢慢升至室温并搅拌反应约12小时,TLC检测反应完全。加入30mL二氯甲烷,依次用饱和碳酸氢钠水溶液、饱和食盐水洗涤,分离有机相用无水硫酸钠干燥,过滤后减压浓缩。粗产物用二氯甲烷-正庚烷重结晶得8.7g白色固体产物IX-A-1a,收率73%。Compound VII-1 (3 mmol of compound III-1 was used as a starting material) was prepared by the method of Step B of Example 1, and dissolved in 12 mL of anhydrous dichloromethane (DCM), and cooled in an ice bath under argon atmosphere. 18 mg (0.15 mmol) of 4-dimethylaminopyridine (DMAP) and 0.75 g (3.9 mmol) of 1-ethyl-(3-dimethylaminopropyl)carbodiimide hydrochloride (EDC HCl) were added in sequence. And 1.93 g (3.6 mmol) of compound VIII-A-1a (prepared by chiral tiopronin, R configuration chiral purity 95%, analytical method: AD-H column temperature: 25 C; detection wavelength: 210 nm; flow rate : 1.0 ml/min; mobile phase: n-Hex: EtOH: TFA = 90: 10: 0.1 isocratic elution), then slowly warmed to room temperature and stirred for about 12 hours, and the reaction was complete by TLC. The mixture was washed with aq. The crude product was recrystallized from methylene chloride-n-heptane to yield 8.7 g of y.
将2.1g(1.76mmol)化合物IX-A-1a溶于50%水-丙酮(5mL)混合溶液中,加入8mL冰醋酸(HOAc)及2mL三氟乙酸(TFA),室温下搅拌反应约6小时,至TLC检测不到剩余原料。减压旋蒸除去丙酮、水,然后依次加入15mL二氯甲烷,3ml三氟乙酸(TFA),2.2mL三异丙基硅烷(i-Pr3SiH),在室温下继续反应约15分钟,TLC检测反应完全,小心加入饱和碳酸氢钠水溶液萃灭反应。加入30mL二氯甲烷,依次用水、饱和食盐水洗涤,分离有机相用无水硫酸钠干燥,过滤后减压浓缩得粗产物。用乙酸乙酯-石油醚重结晶得1.7g白色固体II-A-1a,收率86%。ESI-MS:m/z[M+H]+=705。2.1 g (1.76 mmol) of compound IX-A-1a was dissolved in a 50% water-acetone (5 mL) mixed solution, 8 mL of glacial acetic acid (HOAc) and 2 mL of trifluoroacetic acid (TFA) were added, and the reaction was stirred at room temperature for about 6 hours. The remaining raw materials were not detected by TLC. The acetone and water were evaporated under reduced pressure, and then 15 mL of dichloromethane, 3 ml of trifluoroacetic acid (TFA), and 2.2 mL of triisopropylsilane (i-Pr 3 SiH) were successively added, and the reaction was continued at room temperature for about 15 minutes, TLC The reaction was checked to completion and the reaction was quenched by carefully aqueous saturated sodium bicarbonate. After adding 30 ml of dichloromethane, the mixture was washed with water and aq. Recrystallization from ethyl acetate-petroleum ether gave 1.7 g of white solid II-A-1a, yield 86%. ESI-MS: m/z [M+H] + = 705.
实施例3:化合物II-A-1b的合成 Example 3: Synthesis of Compound II-A-1b
Figure PCTCN2016109913-appb-000070
Figure PCTCN2016109913-appb-000070
依据实施例1方法B中步骤一、步骤二,以VIII-A-1b(由手性硫普罗宁制备得到,S构型手性纯度97%,分析方法:AD-H柱温:25C;检测波长:210nm;流速:1.0ml/min;流动相:n-Hex:EtOH:TFA=90:10:0.1等度洗脱)代替VIII-A-1,以5.3g(10mmol)化合物III-1可制备得2.57g中间体IX-A-1b,收率72%(二步)。According to the first step and the second step in the method B of Example 1, VIII-A-1b (prepared by chiral tiopronin, the chiral purity of the S configuration is 97%, analysis method: AD-H column temperature: 25C; detection Wavelength: 210 nm; flow rate: 1.0 ml/min; mobile phase: n-Hex: EtOH: TFA = 90: 10: 0.1 isocratic elution instead of VIII-A-1, 5.3 g (10 mmol) of compound III-1 2.57 g of intermediate IX-A-1b was obtained in a yield of 72% (two steps).
依据实施例1方法B中步骤三,化合物IX-A-1b(2.9mmol)继续脱去保护基,可制备得1.04g白色固体产物II-A-1b,收率84%。ESI-MS:m/z[M+H]+=705。According to the third step in the method B of Example 1, the compound IX-A-1b (2.9 mmol) was further deprotected, and 1.04 g of a white solid product II-A-1b was obtained in a yield of 84%. ESI-MS: m/z [M+H] + = 705.
实施例4:化合物II-A-2的合成Example 4: Synthesis of Compound II-A-2
方法A:Method A:
Figure PCTCN2016109913-appb-000071
Figure PCTCN2016109913-appb-000071
步骤一:将529mg(1.0mmol)III-1溶于10mL无水丙酮中,室温下加入680mg(1.5mmol)IV-A-2及碳酸钾414mg(3.0mmol),加热搅拌回流6h,TLC检测反应完全。过滤,滤液用二氯甲烷(30mL)稀释,依次用水(10mL)及饱和食盐水(10mL)洗涤,分离有机相,无水硫酸钠干燥后过滤,减压浓缩除去溶剂。剩余物硅胶柱层析纯化得306mg白色固体V-A-2。ESI-MS:m/z[M+H]+=961。Step 1: 529 mg (1.0 mmol) of III-1 was dissolved in 10 mL of anhydrous acetone, and 680 mg (1.5 mmol) of IV-A-2 and potassium carbonate 414 mg (3.0 mmol) were added at room temperature, and the mixture was stirred under reflux for 6 h. complete. After filtration, the filtrate was diluted with methylene chloride (30 mL). The residue was purified by silica gel column chromatography to afford 306 ESI-MS: m / z [ M + H] + = 961.
步骤二:将200mg(0.21mmol)V-A-2溶于10mL无水二氯甲烷中,室温下加入2mL(体积比:二氯甲烷/三异丙基硅烷/三氟乙酸=5/1/1)溶液,反应约1h,TLC检测反应完全,小心加入饱和碳酸氢钠溶液,分离有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩。剩余物硅胶柱层析纯化得到100mg白色固体II-A-2,收率67%。1H NMR(400MHz,CDCl3)δ9.64(s,1H),7.50(dd,J=8.1,2.3Hz,1H),7.36(t,J=7.8Hz,2H),7.30–7.22(m, 3H),7.19(d,J=7.6Hz,1H),6.65(q,J=6.5Hz,1H),6.19(d,J=17.9Hz,1H),5.10–4.94(m,1H),4.52(dd,J=11.4,5.7Hz,1H),4.39–4.21(m,3H),4.22–4.05(m,2H),3.98(tt,J=11.5,5.8Hz,1H),3.57–3.45(m,1H),2.17(dd,J=8.5,2.2Hz,1H),1.86(d,J=6.5Hz,3H),1.56(dd,J=7.1,1.0Hz,3H),1.37(dd,J=14.8,7.5Hz,6H),1.27(s,1H),1.25(d,J=6.3Hz,6H).ESI-MS:m/z[M+H]+=719。Step 2: 200 mg (0.21 mmol) of VA-2 was dissolved in 10 mL of anhydrous dichloromethane, and 2 mL (volume ratio: dichloromethane/triisopropylsilane/trifluoroacetic acid=5/1/1) was added at room temperature. The solution was reacted for about 1 h, and the reaction was completed by TLC. EtOAc (EtOAc m. The residue was purified by silica gel column chromatography eluting elut eluting 1 H NMR (400MHz, CDCl 3 ) δ9.64 (s, 1H), 7.50 (dd, J = 8.1,2.3Hz, 1H), 7.36 (t, J = 7.8Hz, 2H), 7.30-7.22 (m, 3H), 7.19 (d, J = 7.6 Hz, 1H), 6.65 (q, J = 6.5 Hz, 1H), 6.19 (d, J = 17.9 Hz, 1H), 5.10 - 4.94 (m, 1H), 4.52 ( Dd, J=11.4, 5.7 Hz, 1H), 4.39–4.21 (m, 3H), 4.22–4.05 (m, 2H), 3.98 (tt, J=11.5, 5.8 Hz, 1H), 3.57–3.45 (m, 1H), 2.17 (dd, J = 8.5, 2.2 Hz, 1H), 1.86 (d, J = 6.5 Hz, 3H), 1.56 (dd, J = 7.1, 1.0 Hz, 3H), 1.37 (dd, J = 14.8) , 7.5 Hz, 6H), 1.27 (s, 1H), 1.25 (d, J = 6.3 Hz, 6H). ESI-MS: m/z [M+H] + = 719.
方法B:Method B:
Figure PCTCN2016109913-appb-000072
Figure PCTCN2016109913-appb-000072
依据实施例1方法B步骤一、步骤二,以乙醛代替甲醛,以5.3g(10mmol)化合物III-1可制备得4.6g中间体IX-A-2,收率38%(二步)。ESI-MS:m/z[M+H]+=1205。According to the first step and the second step of the method B of Example 1, acetaldehyde was used instead of formaldehyde, and 4.6 g of the intermediate IX-A-2 was obtained in 5.3 g (10 mmol) of the compound III-1 in a yield of 38% (two steps). ESI-MS: m/z [M+H] + =1205.
依据实施例1方法B步骤三,化合物IX-A-2(2.9mmol)继续脱去保护基,可制备得1.7g白色固体产物II-A-2,收率82%。ESI-MS:m/z[M+H]+=719。According to the third step of the method B of Example 1, the compound IX-A-2 (2.9 mmol) was further deprotected, and 1.7 g of a white solid product II-A-2 was obtained in a yield of 82%. ESI-MS: m / z [ M + H] + = 719.
实施例5:化合物II-A-3的合成Example 5: Synthesis of Compound II-A-3
Figure PCTCN2016109913-appb-000073
Figure PCTCN2016109913-appb-000073
依据实施例1方法B步骤一、步骤二,以丙醛代替甲醛,以2.65g(5mmol)化合物III-1可制备得2g中间体IX-A-3,收率33%(二步)。ESI-MS:m/z[M+H]+=1219。According to the first step and the second step of the method of Example 1, the formaldehyde is replaced by propionaldehyde, and 2 g of the intermediate IX-A-3 can be prepared with 2.65 g (5 mmol) of the compound III-1 in a yield of 33% (two steps). ESI-MS: m/z [M+H] + =121.
依据实施例1方法B步骤三,化合物IX-A-3(0.8mmol)继续脱去保护基,可制备得445mg白色固体产物II-A-3,收率76%。ESI-MS:m/z[M+H]+=733。According to the third step of the method B of Example 1, the compound IX-A-3 (0.8 mmol) was further deprotected, and 445 mg of the white solid product II-A-3 was obtained in a yield of 76%. ESI-MS: m/z [M+H] + = 733.
实施例6:化合物II-A-4的合成 Example 6: Synthesis of Compound II-A-4
Figure PCTCN2016109913-appb-000074
Figure PCTCN2016109913-appb-000074
步骤一:将529mg(1.0mmol)III-1溶于10mL无水丙酮中,室温下加入690mgIV-A-3及碳酸钾414mg(3.0mmol),加热搅拌回流6h,TLC检测反应完全。过滤,滤液用二氯甲烷(30mL)稀释,依次用水(10mL)及饱和食盐水(10mL)洗涤,分离有机相,无水硫酸钠干燥后过滤,减压浓缩除去溶剂。剩余物硅胶柱层析纯化得300mg V-A-4,直接用于下一步反应。Step 1: 529 mg (1.0 mmol) of III-1 was dissolved in 10 mL of anhydrous acetone, and 690 mg of IV-A-3 and potassium carbonate 414 mg (3.0 mmol) were added at room temperature, and the mixture was stirred under reflux for 6 hours, and the reaction was completely confirmed by TLC. After filtration, the filtrate was diluted with methylene chloride (30 mL). The residue was purified by silica gel column chromatography eluting with EtOAc
步骤二:将200mg(0.21mmol)V-A-4溶于10mL无水二氯甲烷中,室温下加入2mL(体积比:二氯甲烷/三异丙基硅烷/三氟乙酸=5/1/1)溶液,反应约1h,TLC检测反应完全,小心加入饱和碳酸氢钠溶液,分离有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩。剩余物硅胶柱层析纯化得到100mg白色固体II-A-4。1H NMR(400MHz,CDCl3)δ9.62(s,1H),7.46(dd,J=8.1,2.2Hz,1H),7.34(t,J=7.8Hz,2H),7.29-7.22(m,3H),7.19(d,J=7.4Hz,1H),6.16(d,J=18.1Hz,1H),5.64-5.53(m,1H),5.37-5.17(m,1H),5.08-4.93(m,1H),4.53(dd,J=11.4,5.5Hz,1H),4.38-4.20(m,3H),4.40-4.08(m,6H),3.97(tt,J=11.7,5.9Hz,1H),3.57-3.43(m,1H),2.17(dd,J=8.4,2.2Hz,1H),1.55(dd,J=7.1,1.0Hz,3H),1.38(dd,J=14.8,7.7Hz,6H),1.27(s,1H),1.24(d,J=6.3Hz,6H).ESI-MS:749(M+1)。Step 2: Dissolve 200 mg (0.21 mmol) of VA-4 in 10 mL of anhydrous dichloromethane, and add 2 mL at room temperature (volume ratio: dichloromethane / triisopropylsilane / trifluoroacetic acid = 5 / 1 / 1) The solution was reacted for about 1 h, and the reaction was completed by TLC. EtOAc (EtOAc m. The residue was purified by silica gel column chromatography toield 1 H NMR (400MHz, CDCl3) δ9.62 (s, 1H), 7.46 (dd, J = 8.1,2.2Hz, 1H), 7.34 (t, J = 7.8Hz, 2H), 7.29-7.22 (m, 3H ), 7.19 (d, J = 7.4 Hz, 1H), 6.16 (d, J = 18.1 Hz, 1H), 5.64 - 5.53 (m, 1H), 5.37 - 5.17 (m, 1H), 5.08 - 4.93 (m, 1H), 4.53 (dd, J = 11.4, 5.5 Hz, 1H), 4.38-4.20 (m, 3H), 4.40-4.08 (m, 6H), 3.97 (tt, J = 11.7, 5.9 Hz, 1H), 3.57 -3.43 (m, 1H), 2.17 (dd, J = 8.4, 2.2 Hz, 1H), 1.55 (dd, J = 7.1, 1.0 Hz, 3H), 1.38 (dd, J = 14.8, 7.7 Hz, 6H), 1.27 (s, 1H), 1.24 (d, J = 6.3 Hz, 6H). ESI-MS: 749 (M+1).
实施例7:化合物II-A-5的合成Example 7: Synthesis of Compound II-A-5
Figure PCTCN2016109913-appb-000075
Figure PCTCN2016109913-appb-000075
步骤一:将529mg(1.0mmol)III-1溶于10mL无水丙酮中,室温下加入800mg IV-A-4及碳酸钾414mg(3.0mmol),加热搅拌回流12h,TLC检测反应完全。过滤,滤液用二氯甲烷(30mL)稀释,依次用水(10mL)及饱和食盐水(10mL)洗涤,分离有机相,无水硫酸钠干燥后过滤,减压浓缩除去溶剂。剩余物硅胶柱层析纯化得400mg V-A-5,直接用于下一步反应。Step 1: 529 mg (1.0 mmol) of III-1 was dissolved in 10 mL of anhydrous acetone, and 800 mg of IV-A-4 and potassium carbonate 414 mg (3.0 mmol) were added at room temperature, and the mixture was stirred under reflux for 12 h. After filtration, the filtrate was diluted with methylene chloride (30 mL). The residue was purified by silica gel column chromatography to yield 400 mg of V-A-5.
步骤二:将200mg(0.21mmol)V-A-5溶于10mL无水二氯甲烷中,室温下加入2mL (体积比:二氯甲烷/三异丙基硅烷/三氟乙酸=5/1/1)溶液,反应约1h,TLC检测反应完全,小心加入饱和碳酸氢钠溶液,分离有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩。剩余物硅胶柱层析纯化得到100mg白色固体II-A-5。1H NMR(400MHz,CDCl3)δ9.66(s,1H),7.44(dd,J=7.9,2.1Hz,1H),7.32(t,J=7.8Hz,2H),7.28–7.20(m,3H),7.16(d,J=7.5Hz,1H),6.13(d,J=17.6Hz,1H),5.15–5.03(m,1H),4.60(t,J=7.5Hz,2H),4.55(dd,J=11.5,5.7Hz,1H),4.48–4.25(m,3H),4.23–4.09(m,2H),3.95(tt,J=11.8,5.8Hz,1H),3.59–3.42(m,1H),3.25(t,J=7.5Hz,2H),2.16(dd,J=8.4,2.2Hz,1H),1.54(dd,J=7.1,1.0Hz,3H),1.39(dd,J=14.7,7.6Hz,6H),1.28(s,1H),1.25(d,J=6.5Hz,6H).ESI-MS:m/z[M+H]+=719。Step 2: Dissolve 200 mg (0.21 mmol) of VA-5 in 10 mL of anhydrous dichloromethane, and add 2 mL at room temperature (volume ratio: dichloromethane / triisopropylsilane / trifluoroacetic acid = 5 / 1 / 1) The solution was reacted for about 1 h, and the reaction was completed by TLC. EtOAc (EtOAc m. The residue was purified by silica gel column chromatography toield 1 H NMR (400MHz, CDCl 3 ) δ9.66 (s, 1H), 7.44 (dd, J = 7.9,2.1Hz, 1H), 7.32 (t, J = 7.8Hz, 2H), 7.28-7.20 (m, 3H), 7.16 (d, J = 7.5 Hz, 1H), 6.13 (d, J = 17.6 Hz, 1H), 5.15 - 5.03 (m, 1H), 4.60 (t, J = 7.5 Hz, 2H), 4.55 ( Dd, J = 11.5, 5.7 Hz, 1H), 4.48 - 4.25 (m, 3H), 4.23 - 4.09 (m, 2H), 3.95 (tt, J = 11.8, 5.8 Hz, 1H), 3.59 - 3.42 (m, 1H), 3.25 (t, J = 7.5 Hz, 2H), 2.16 (dd, J = 8.4, 2.2 Hz, 1H), 1.54 (dd, J = 7.1, 1.0 Hz, 3H), 1.39 (dd, J = 14.7) , 7.6 Hz, 6H), 1.28 (s, 1H), 1.25 (d, J = 6.5 Hz, 6H). ESI-MS: m/z [M+H] + = 719.
实施例8:化合物II-A-7的合成Example 8: Synthesis of Compound II-A-7
Figure PCTCN2016109913-appb-000076
Figure PCTCN2016109913-appb-000076
依据实施例1方法B步骤一、步骤二,仅以III-2代替代替III-1,以1.1g(2mmol)化合物III-2可制备得无色油状物IX-A-7,收率71%(二步)。ESI-MS:m/z[M+H]+=1207。According to the first step and the second step of the method of the first embodiment, only the III-2 is substituted for the III-1, and the colorless oil IX-A-7 can be prepared with 1.1 g (2 mmol) of the compound III-2, the yield is 71%. (two steps). ESI-MS: m/z [M+H] + =1207.
依据实施例1方法B步骤三,0.79g化合物IX-A-7(0.65mmol)继续脱去保护基,可制备得白色固体产物II-A-7,收率83%。ESI-MS:m/z[M+H]+=721。According to the third step of the method B of Example 1, 0.79 g of the compound IX-A-7 (0.65 mmol) was further deprotected, and the white solid product II-A-7 was obtained in a yield of 83%. ESI-MS: m/z [M+H] + = 721.
实施例9:化合物II-A-8的合成Example 9: Synthesis of Compound II-A-8
Figure PCTCN2016109913-appb-000077
Figure PCTCN2016109913-appb-000077
依据实施例1方法B步骤一,步骤二,以VIII-A-2代替VIII-A-1,从而以1.6g(3mmol)化合物III-1可制备得1.29g中间体IX-A-8,收率65%(二步)。ESI-MS:m/z[M+H]+=1219.According to the first step, the second step of the method B, VIII-A-2 is substituted for VIII-A-1, and 1.29 g of the intermediate IX-A-8 can be prepared by using 1.6 g (3 mmol) of the compound III-1. The rate is 65% (two steps). ESI-MS: m/z [M+H] + =1219.
依据实施例1方法B步骤三,化合物IX-A-8(0.8mmol)继续脱去保护基,可制备得0.5g白色固体产物II-A-8,收率83%。ESI-MS:m/z[M+H]+=733。According to the third step of the method B of Example 1, the compound IX-A-8 (0.8 mmol) was further deprotected, and 0.5 g of a white solid product II-A-8 was obtained in a yield of 83%. ESI-MS: m/z [M+H] + = 733.
实施例10:化合物II-B-1的合成 Example 10: Synthesis of Compound II-B-1
Figure PCTCN2016109913-appb-000078
Figure PCTCN2016109913-appb-000078
依据实施例1方法B,步骤二中以VIII-B-1(由(DL)-N-乙酰半胱氨酸制备获得)代替VIII-1,以2.1g(4mmol)化合物III-1可制备得3.38g中间体IX-B-1,收率71%(二步)。ESI-MS:m/z[M+H]+=1191。According to the method B of Example 1, in the second step, VIII-B-1 (obtained from (DL)-N-acetylcysteine) is substituted for VIII-1, and 2.1 g (4 mmol) of compound III-1 can be prepared. 3.38 g of intermediate IX-B-1, yield 71% (two steps). ESI-MS: m/z [M+H] + =1191.
依据实施例1方法B步骤三,化合物IX-B-1(2mmol)继续脱去保护基,可制备得1.17g白色固体产物II-B-1,收率83%。ESI-MS:m/z[M+H]+=705。According to the third step of the method B of Example 1, the compound IX-B-1 (2 mmol) was further deprotected, and 1.17 g of the white solid product II-B-1 was obtained in a yield of 83%. ESI-MS: m/z [M+H] + = 705.
实施例11:化合物II-B-1a的合成Example 11: Synthesis of Compound II-B-1a
Figure PCTCN2016109913-appb-000079
Figure PCTCN2016109913-appb-000079
依据实施例1方法B,步骤二中以VIII-B-1a(由L-乙酰半胱氨酸制备获得,手性纯度>99%)代替VIII-1,以2.65g(5mmol)化合物III-1可制备得中间体IX-B-1a,收率67%(二步)。ESI-MS:m/z[M+H]+=1191。According to the method B of Example 1, in the second step, VIII-B-1a (prepared by L-acetylcysteine, chiral purity >99%) is substituted for VIII-1, and 2.65 g (5 mmol) of compound III-1 is used. Intermediate IX-B-1a can be prepared in a yield of 67% (two steps). ESI-MS: m/z [M+H] + =1191.
依据实施例1方法B步骤三,化合物IX-B-1a(2.1mmol)继续脱去保护基,可制备得1.2g白色固体产物II-B-1a,收率85%。ESI-MS:m/z[M+H]+=705。According to the third step of the method B of Example 1, the compound IX-B-1a (2.1 mmol) was further deprotected, and 1.2 g of the white solid product II-B-1a was obtained in a yield of 85%. ESI-MS: m/z [M+H] + = 705.
实施例12:化合物II-B-1b的合成 Example 12: Synthesis of Compound II-B-1b
Figure PCTCN2016109913-appb-000080
Figure PCTCN2016109913-appb-000080
依据实施例1方法B,步骤二中以VIII-B-1b(由D-半胱氨酸制备获得,手性纯度95%)代替VIII-1,以2.65g(5mmol)化合物III-1可制备得4.2g中间体IX-B-1b,收率70%(二步)。ESI-MS:m/z[M+H]+=1191。According to the method B of the first embodiment, in the second step, VIII-B-1b (prepared by D-cysteine, the chiral purity is 95%) is substituted for VIII-1, and 2.65 g (5 mmol) of compound III-1 can be prepared. 4.2 g of intermediate IX-B-1b was obtained in a yield of 70% (two steps). ESI-MS: m/z [M+H] + =1191.
依据实施例1方法B步骤三,化合物IX-B-1b(2.1mmol)继续脱去保护基,可制备得1.2g白色固体产物II-B-1b,收率81%。ESI-MS:m/z[M+H]+=705。According to the third step of the method B of Example 1, the compound IX-B-1b (2.1 mmol) was further deprotected, and 1.2 g of the white solid product II-B-1b was obtained in a yield of 81%. ESI-MS: m/z [M+H] + = 705.
实施例13:化合物II-B-2的合成Example 13: Synthesis of Compound II-B-2
Figure PCTCN2016109913-appb-000081
Figure PCTCN2016109913-appb-000081
依据实施例1方法B步骤一,以乙醛代替甲醛,依据实施例1方法B步骤二,以VIII-B-1a(由L-乙酰半胱氨酸制备获得,手性纯度>99%)代替VIII-1,从而以1.6g(3mmol)化合物III-1可制备得1.3g中间体IX-B-2,收率36%(二步)。ESI-MS:m/z[M+H]+=1205。According to the first step of the method B of Example 1, acetaldehyde is substituted for formaldehyde, and according to the second step of the method B of the first embodiment, the VIII-B-1a (prepared by L-acetylcysteine, chiral purity >99%) is replaced. VIII-1, whereby 1.3 g of intermediate IX-B-2 can be prepared in 1.6 g (3 mmol) of compound III-1 in a yield of 36% (two steps). ESI-MS: m/z [M+H] + =1205.
依据实施例1方法B步骤三,化合物IX-B-2(1mmol)继续脱去保护基,可制备得0.57g白色固体产物II-B-2,收率79%。ESI-MS:m/z[M+H]+=719。According to the third step of the method B of Example 1, the compound IX-B-2 (1 mmol) was further deprotected, and 0.57 g of a white solid product II-B-2 was obtained in a yield of 79%. ESI-MS: m/z [M+H] + = 719.
实施例14:化合物II-B-3的合成 Example 14: Synthesis of Compound II-B-3
Figure PCTCN2016109913-appb-000082
Figure PCTCN2016109913-appb-000082
依据实施例1方法B步骤一,步骤二,以VIII-B-2(由L-乙酰半胱氨酸制备获得,手性纯度>99%)代替VIII-1,从而以1.6g(3mmol)化合物III-1可制备得中间体IX-B-3,收率76%(二步)。ESI-MS:m/z[M+H]+=1205。According to the first step, the second step of the method of the first embodiment, the VIII-B-2 (prepared by L-acetylcysteine, chiral purity >99%) is substituted for VIII-1, thereby giving 1.6 g (3 mmol) of the compound. Intermediate IX-B-3 can be prepared in III-1 in a yield of 76% (two steps). ESI-MS: m/z [M+H] + =1205.
依据实施例1方法B步骤三,化合物IX-B-3(1mmol)继续脱去保护基,可制备得0.57g白色固体产物II-B-3,收率82%。ESI-MS:m/z[M+H]+=719。According to the third step of the method B of Example 1, the compound IX-B-3 (1 mmol) was further deprotected, and 0.57 g of a white solid product II-B-3 was obtained in a yield of 82%. ESI-MS: m/z [M+H] + = 719.
实施例15:化合物II-B-4的合成Example 15: Synthesis of Compound II-B-4
Figure PCTCN2016109913-appb-000083
Figure PCTCN2016109913-appb-000083
依据实施例1方法B中步骤一,步骤二,以III-2代替III-1,并以VIII-B-1a(由L-乙酰半胱氨酸制备获得,手性纯度>99%)代替VIII-1,以1.6g(3mmol)化合物III-2可制备得2.64g中间体IX-B-4,收率72%(二步)。ESI-MS:m/z[M+H]+=1207。According to the first step, the second step in the method B of the first embodiment, the III-1 is replaced by III-2, and the VIII-B-1a (prepared by L-acetylcysteine, the chiral purity is >99%) is substituted for VIII. -1, 2.64 g of Intermediate IX-B-4 can be obtained in 1.6 g (3 mmol) of compound III-2 in a yield of 72% (two steps). ESI-MS: m/z [M+H] + =1207.
依据实施例6步骤三,化合物IX-B-4(1.5mmol)继续脱去保护基,可制备得0.93g白色固体产物II-B-4,收率84%。ESI-MS:m/z[M+H]+=721。According to the third step of Example 6, compound IX-B-4 (1.5 mmol) was further deprotected, and 0.93 g of white solid product II-B-4 was obtained in a yield of 84%. ESI-MS: m/z [M+H] + = 721.
实施例16:化合物III-2A-1的合成Example 16: Synthesis of Compound III-2A-1
方法A: Method A:
Figure PCTCN2016109913-appb-000084
Figure PCTCN2016109913-appb-000084
步骤一:将529mg(1.0mmol)III-1溶于10mL无水二氯甲烷中,室温下加入405mg(1.0mmol)X-A-1(制备方法参考Polymer Chemistry,2011,2,906-913)环己基碳二亚胺412mg(2.0mmol)及DMAP 12.2mg(0.1mmol),搅拌6h,TLC检测反应完全。减压浓缩除去溶剂。剩余物硅胶柱层析纯化得白色固体XI-A-1,收率43%,LC-ESI-MS:[M+H]+916,直接用于下一步反应。Step 1: 529 mg (1.0 mmol) of III-1 was dissolved in 10 mL of anhydrous dichloromethane, and 405 mg (1.0 mmol) of XA-1 was added at room temperature (preparation method refers to Polymer Chemistry, 2011, 2, 906-913) cyclohexyl carbon two The imine 412 mg (2.0 mmol) and DMAP 12.2 mg (0.1 mmol) were stirred for 6 h, and the reaction was confirmed by TLC. The solvent was removed by concentration under reduced pressure. The residue was purified by silica gel column chromatography to give white solid XI-A-1, a yield of 43%, LC-ESI-MS : [M + H] + 916, was used directly in the next reaction.
步骤二:将183mg(0.2mmol)XI-A-1溶于10mL无水二氯甲烷中,室温下加入2mL(体积比:二氯甲烷/三异丙基硅烷/三氟乙酸=5/1/1)溶液,反应约1h,TLC检测反应完全,小心加入饱和碳酸氢钠溶液,分离有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩。剩余物硅胶柱层析纯化得到白色固体III-2A-1,收率51%。1H NMR(400MHz,DMSO)δ11.57(s,1H),8.55(d,J=7.8Hz,1H),7.71(d,J=7.7Hz,1H),7.38(t,J=7.6Hz,2H),7.20(dd,J=18.6,7.6Hz,3H),6.18–5.88(m,2H),5.63(d,J=7.8Hz,1H),5.34(s,1H),4.86(dt,J=12.2,6.0Hz,1H),4.35–4.16(m,3H),4.08–3.88(m,2H),3.87–3.72(m,1H),3.54(dd,J=12.9,6.8Hz,1H),2.86(d,J=6.9Hz,1H),1.43–1.26(m,6H),1.23(d,J=6.9Hz,3H),1.16(d,J=6.1Hz,6H).LC-ESI-MS:[M+H]+675。Step 2: Dissolve 183 mg (0.2 mmol) of XI-A-1 in 10 mL of anhydrous dichloromethane, and add 2 mL at room temperature (volume ratio: dichloromethane/triisopropylsilane/trifluoroacetic acid=5/1/ 1) The solution was reacted for about 1 h, and the reaction was completed by TLC. EtOAc (EtOAc m. The residue was purified by silica gel column chromatography to afford white crystals, 1 H NMR (400MHz, DMSO) δ11.57 (s, 1H), 8.55 (d, J = 7.8Hz, 1H), 7.71 (d, J = 7.7Hz, 1H), 7.38 (t, J = 7.6Hz, 2H), 7.20 (dd, J = 18.6, 7.6 Hz, 3H), 6.18 - 5.88 (m, 2H), 5.63 (d, J = 7.8 Hz, 1H), 5.34 (s, 1H), 4.86 (dt, J =12.2, 6.0 Hz, 1H), 4.35–4.16 (m, 3H), 4.08–3.88 (m, 2H), 3.87–3.72 (m, 1H), 3.54 (dd, J = 12.9, 6.8 Hz, 1H), 2.86 (d, J = 6.9 Hz, 1H), 1.43 - 1.26 (m, 6H), 1.23 (d, J = 6.9 Hz, 3H), 1.16 (d, J = 6.1 Hz, 6H). LC-ESI-MS :[M+H] + 675.
Figure PCTCN2016109913-appb-000085
Figure PCTCN2016109913-appb-000085
方法B:Method B:
步骤一:将5.4g(10mmol)化合物III-1溶于50mL无水二氯甲烷(DCM)中,在Ar保护下冰浴冷却,加入61mg(0.5mmol)4-二甲氨基吡啶(DMAP)和2.3g(12mmol)1-乙基-(3-二甲基氨基丙基)碳酰二亚胺盐酸盐(EDC HCl),搅拌5分钟之后加入4.5g (8.5mmol)化合物VIII-A-1,继续搅拌反应约6小时,TLC检测反应完全。加入50mL二氯甲烷,依次用饱和碳酸氢钠水溶液、饱和食盐水洗涤,分离有机相用无水硫酸钠干燥,过滤后减压浓缩。剩余物硅胶柱层析纯化得到7.7g浅黄色油状物XII-A-1,收率87%。1H NMR(400MHz,CDCl3)δ9.66(s,1H),7.44(t,J=7.4Hz,1H),7.37(s,4H),7.34–7.09(m,14H),6.77(d,J=8.4Hz,2H),6.16(d,J=17.9Hz,1H),5.51–5.38(m,1H),5.21–5.05(m,1H),5.05–4.90(m,1H),4.52(s,1H),4.37–4.26(m,1H),4.25–4.08(m,4H),4.05–3.92(m,2H),3.78(s,3H),1.42(t,J=6.3Hz,3H),1.35(s,9H),1.37–1.23(m,6H),1.20(d,J=5.9Hz,6H)。31P NMR(162MHz,CDCl3)δ2.74(d,J=7.0Hz)。Step 1: 5.4 g (10 mmol) of compound III-1 was dissolved in 50 mL of anhydrous dichloromethane (DCM), cooled in an ice bath under Ar protection, and 61 mg (0.5 mmol) of 4-dimethylaminopyridine (DMAP) and 2.3 g (12 mmol) of 1-ethyl-(3-dimethylaminopropyl)carbodiimide hydrochloride (EDC HCl), after stirring for 5 minutes, 4.5 g (8.5 mmol) of compound VIII-A-1 was added. The reaction was stirred for about 6 hours and the reaction was complete by TLC. After adding 50 ml of dichloromethane, the mixture was washed with EtOAc EtOAc. The residue was purified by silica gel column chromatography to afford 177 g of pale yellow oily of 1 H NMR (400 MHz, CDCl 3 ) δ 9.66 (s, 1H), 7.44 (t, J = 7.4 Hz, 1H), 7.37 (s, 4H), 7.34 - 7.09 (m, 14H), 6.77 (d, J=8.4 Hz, 2H), 6.16 (d, J = 17.9 Hz, 1H), 5.51–5.38 (m, 1H), 5.21–5.05 (m, 1H), 5.05–4.90 (m, 1H), 4.52 (s) , 1H), 4.37–4.26 (m, 1H), 4.25–4.08 (m, 4H), 4.05–3.92 (m, 2H), 3.78 (s, 3H), 1.42 (t, J = 6.3 Hz, 3H), 1.35 (s, 9H), 1.37 - 1.23 (m, 6H), 1.20 (d, J = 5.9 Hz, 6H). 31 P NMR (162 MHz, CDCl 3 ) δ 2.74 (d, J = 7.0 Hz).
步骤二:将4.7g(4.5mmol)化合物XII-A-1溶于50mL二氯甲烷中,在室温下加入3.5mL三氟乙酸(TFA)及3.5mL三异丙基硅烷(i-Pr3SiH),反应15分钟后再加入3.5mL三氟乙酸(TFA),继续反应约10~20分钟,TLC检测反应完全,小心加入饱和碳酸氢钠水溶液崔灭反应。加入30mL二氯甲烷,依次用水、饱和食盐水洗涤,分离有机相用无水硫酸钠干燥,过滤后减压浓缩。剩余物硅胶柱层析纯化得到2.5g白色固体,收率82%,LC-ESI-MS:[M+H]+675。Step 2: 4.7 g (4.5 mmol) of compound XII-A-1 was dissolved in 50 mL of dichloromethane, and 3.5 mL of trifluoroacetic acid (TFA) and 3.5 mL of triisopropylsilane ( i- Pr 3 SiH) were added at room temperature. After the reaction for 15 minutes, 3.5 mL of trifluoroacetic acid (TFA) was further added, and the reaction was continued for about 10 to 20 minutes. The reaction was completely confirmed by TLC, and the reaction was cautiously added with a saturated aqueous solution of sodium hydrogencarbonate. The organic layer was separated and dried over anhydrous sodium The residue was purified by silica gel column chromatography to give 2.5g white solid, yield 82%, LC-ESI-MS : [M + H] + 675.
实施例17:化合物III-2A-1a的合成Example 17: Synthesis of Compound III-2A-1a
Figure PCTCN2016109913-appb-000086
Figure PCTCN2016109913-appb-000086
依据实施例16方法B步骤一、步骤二,以VIII-A-1a(由手性硫普罗宁制备得到,R构型手性纯度95%,分析方法:AD-H柱温:25C;检测波长:210nm;流速:1.0ml/min;流动相:n-Hex:EtOH:TFA=90:10:0.1等度洗脱)代替VIII-A-1,以5.3g(10mmol)化合物III-1为原料,可制备得白色固体产物III-2A-1a,收率78%(两步)。ESI-MS:m/z[M+H]+=675。According to the method of Example 16 Step B, Step 2, VIII-A-1a (prepared by chiral tiopronin, R configuration chiral purity 95%, analytical method: AD-H column temperature: 25C; detection wavelength : 210 nm; flow rate: 1.0 ml/min; mobile phase: n-Hex: EtOH: TFA = 90: 10: 0.1 isocratic elution instead of VIII-A-1, using 5.3 g (10 mmol) of compound III-1 as raw material The white solid product III-2A-1a was obtained in a yield of 78% (two steps). ESI-MS: m / z [ M + H] + = 675.
实施例18:化合物III-2A-1b的合成 Example 18: Synthesis of Compound III-2A-1b
Figure PCTCN2016109913-appb-000087
Figure PCTCN2016109913-appb-000087
依据实施例16方法B的步骤一、步骤二,以VIII-A-1b(由手性硫普罗宁制备得到,S构型手性纯度97%,分析方法:AD-H柱温:25C;检测波长:210nm;流速:1.0ml/min;流动相:n-Hex:EtOH:TFA=90:10:0.1等度洗脱)代替VIII-A-1,以5.3g(10mmol)化合物III-1为原料,可制备得5.0g白色固体产物III-2A-1b,收率75%(两步).ESI-MS:m/z[M+H]+=675。According to the first step and the second step of the method B of Example 16, VIII-A-1b (prepared by chiral tiopronin, the chiral purity of the S configuration is 97%, analytical method: AD-H column temperature: 25C; detection Wavelength: 210 nm; flow rate: 1.0 ml/min; mobile phase: n-Hex: EtOH: TFA = 90: 10: 0.1 isocratic elution instead of VIII-A-1, with 5.3 g (10 mmol) of compound III-1 Starting material, 5.0 g of a white solid product III-2A-1b was obtained in a yield of 75% (two steps). ESI-MS: m/z [M+H] + = 675.
实施例19:化合物III-2A-2的合成Example 19: Synthesis of Compound III-2A-2
Figure PCTCN2016109913-appb-000088
Figure PCTCN2016109913-appb-000088
步骤一:将529mg(1.0mmol)III-1溶于5mL二甲基亚砜中,加入4mL醋酐和1.5mL醋酸,室温搅拌48小时,小心加入饱和食盐水和乙酸乙酯,分离有机相依次用饱和NaHCO3和饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩。减压浓缩除去溶剂。剩余物硅胶柱层析纯化得淡黄色胶状物XIII-1,收率25%,LC-ESI-MS:[M+H]+590。Step 1: Dissolve 529 mg (1.0 mmol) of III-1 in 5 mL of dimethyl sulfoxide, add 4 mL of acetic anhydride and 1.5 mL of acetic acid, stir at room temperature for 48 hours, carefully add saturated brine and ethyl acetate, and separate the organic phase. The extract was washed with aq. The solvent was removed by concentration under reduced pressure. The residue was purified by silica gel column chromatography to give a pale yellow gum XIII-1, a yield of 25%, LC-ESI-MS : [M + H] + 590.
步骤二:将118mg(0.2mmol)XIII-1溶于无水二氯甲烷,冰浴条件下加入0.3mL二氯亚砜的二氯甲烷溶液(1M),反应液缓慢升温至室温,并搅拌2h,减压浓缩除去溶剂,得黄色胶状物直接用于下一步反应。将上述胶状物XIII-1溶于1mL无水四氢呋喃中,室温下加入93mg(0.2mmol)IX-A-1及碳酸钾55mg(0.4mmol),搅拌6h,TLC检测反应完全。减压浓缩除去溶剂。剩余物硅胶柱层析纯化得白色固体IX-A-1,收率45%,LC-ESI-MS:[M+H]+946。步骤三:将36.6mg(0.04mmol)IX-A-1溶于1mL无水二氯甲烷中,室温下加入0.5mL(体积比:二氯甲烷/三异丙基硅烷/三氟乙酸=5/1/1)溶液,反应约1h,TLC检测反应完全,小心加入饱和碳酸氢钠溶液,分离有机相用饱和食盐水洗涤,无水硫酸钠干 燥,过滤后减压浓缩。剩余物硅胶柱层析纯化得到白色固体III-2A-2,收率70%。1H NMR(400MHz,DMSO-d6)δ11.56(s,1H),8.53(d,J=7.8Hz,1H),7.70(d,J=7.5Hz,1H),7.28(t,J=7.5Hz,2H),7.21(dd,J=18.0,7.6Hz,3H),6.15–5.71(m,2H),5.61(d,J=7.8Hz,1H),5.55(d,J=6.2Hz,1H),5.49(d,J=6.2Hz,1H),5.34(s,1H),4.84(dt,J=12.0,6.0Hz,1H),4.31–4.14(m,3H),4.11–3.85(m,2H),3.85–3.73(m,1H),3.54(dd,J=12.0,6.5Hz,1H),2.84(d,J=7.0Hz,1H),1.45–1.23(m,6H),1.21(d,J=6.9Hz,3H),1.19(d,J=6.1Hz,6H).LC-ESI-MS:[M+H]+704。Step 2: 118 mg (0.2 mmol) of XIII-1 was dissolved in anhydrous dichloromethane, and 0.3 mL of dichloromethane (1 M) of thionyl chloride was added thereto under ice-cooling. The reaction mixture was slowly warmed to room temperature and stirred for 2 h. The solvent was concentrated under reduced pressure to give a yellow gum. The above-mentioned gum XIII-1 was dissolved in 1 mL of anhydrous tetrahydrofuran, and 93 mg (0.2 mmol) of IX-A-1 and potassium carbonate 55 mg (0.4 mmol) were added at room temperature, and stirred for 6 hours, and the reaction was completely confirmed by TLC. The solvent was removed by concentration under reduced pressure. The residue was purified by silica gel column chromatography to obtain white solid IX-A-1, a yield of 45%, LC-ESI-MS : [M + H] + 946. Step 3: 36.6 mg (0.04 mmol) of IX-A-1 was dissolved in 1 mL of anhydrous dichloromethane, and 0.5 mL was added at room temperature (volume ratio: dichloromethane / triisopropylsilane / trifluoroacetic acid = 5 / 1/1) The solution was reacted for about 1 h, and the reaction was completed by TLC. EtOAc (EtOAc m. The residue was purified by silica gel column chromatography to afford white crystal 1 H NMR (400MHz, DMSO- d6) δ11.56 (s, 1H), 8.53 (d, J = 7.8Hz, 1H), 7.70 (d, J = 7.5Hz, 1H), 7.28 (t, J = 7.5 Hz, 2H), 7.21 (dd, J = 18.0, 7.6 Hz, 3H), 6.15 - 5.71 (m, 2H), 5.61 (d, J = 7.8 Hz, 1H), 5.55 (d, J = 6.2 Hz, 1H) ), 5.49 (d, J = 6.2 Hz, 1H), 5.34 (s, 1H), 4.84 (dt, J = 12.0, 6.0 Hz, 1H), 4.31 - 4.14 (m, 3H), 4.11 - 3.85 (m, 2H), 3.85–3.73 (m, 1H), 3.54 (dd, J=12.0, 6.5 Hz, 1H), 2.84 (d, J=7.0 Hz, 1H), 1.45–1.23 (m, 6H), 1.21 (d) , J = 6.9 Hz, 3H), 1.19 (d, J = 6.1 Hz, 6H). LC-ESI-MS: [M+H] + 704.
实施例20:化合物III-2A-5的合成Example 20: Synthesis of Compound III-2A-5
Figure PCTCN2016109913-appb-000089
Figure PCTCN2016109913-appb-000089
依据实施例16方法B步骤一,步骤二,以VIII-A-2代替VIII-A-1,以5.3g(10mmol)化合物III-1为原料,可制备得4.9g白色固体产物III-2A-5,收率71%(两步)。ESI-MS:m/z[M+H]+=702。According to the method of Example 16 Step B, Step 2, VIII-A-2 is substituted for VIII-A-1, and 5.3 g (10 mmol) of compound III-1 is used as a raw material to prepare 4.9 g of white solid product III-2A- 5, yield 71% (two steps). ESI-MS: m/z [M+H] + = 702.
实施例21:化合物III-2A-6的合成Example 21: Synthesis of Compound III-2A-6
Figure PCTCN2016109913-appb-000090
Figure PCTCN2016109913-appb-000090
依据实施例16方法B步骤一,步骤二,以III-2代替III-1,以5.4g(10mmol)化合物III-2为原料,可制备得4.5g白色固体产物III-2A-6,收率65%(两步)。ESI-MS:m/z[M+H]+=691。According to the method of Example 16 Step B, Step 2, replacing III-1 with III-2, and preparing 5.4 g (10 mmol) of compound III-2 as raw material, 4.5 g of white solid product III-2A-6 can be prepared. 65% (two steps). ESI-MS: m/z [M+H] + = 691.
实施例22:化合物III-2B-1的合成Example 22: Synthesis of Compound III-2B-1
方法A: Method A:
Figure PCTCN2016109913-appb-000091
Figure PCTCN2016109913-appb-000091
步骤一:将529mg(1.0mmol)III-1溶于10mL无水二氯甲烷中,室温下加入405mg X-B-1(1.0mmol),环己基碳二亚胺412mg(2.0mmol)及DMAP 12.2mg(0.1mmol),搅拌12h,TLC检测反应完全。减压浓缩除去溶剂。剩余物硅胶柱层析纯化得白色固体XI-B-1,LC-ESI-MS:[M+H]+916,直接用于下一步反应。Step 1: 529 mg (1.0 mmol) of III-1 was dissolved in 10 mL of anhydrous dichloromethane, and 405 mg of XB-1 (1.0 mmol), cyclohexylcarbodiimide 412 mg (2.0 mmol) and DMAP 12.2 mg ( 0.1 mmol), stirred for 12 h, and the reaction was complete by TLC. The solvent was removed by concentration under reduced pressure. The residue was purified by silica gel column chromatography to give white solid XI-B-1, LC- ESI-MS: [M + H] + 916, was used directly in the next reaction.
步骤二:将183mg(0.2mmol)XI-B-1溶于10mL无水二氯甲烷中,室温下加入2mL(体积比:二氯甲烷/三异丙基硅烷/三氟乙酸=5/1/1)溶液,反应约1h,TLC检测反应完全,小心加入饱和碳酸氢钠溶液,分离有机相用饱和食盐水洗涤,无水硫酸钠干燥,过滤后减压浓缩。剩余物硅胶柱层析纯化得到白色固体III-2B-1,收率:62%。1H NMR(400MHz,DMSO-d6)δ11.60(s,1H),8.61(d,J=7.5Hz,1H),7.69(d,J=8.0Hz,1H),7.39(t,J=7.6Hz,2H),7.22(m,3H),6.19–5.90(m,2H),5.69(d,J=7.8Hz,1H),5.36(s,1H),5.12~5.06(m,1H),4.89(dt,J=12.2,6.0Hz,1H),4.37–4.15(m,3H),4.18–3.98(m,2H),3.57(dd,J=12.9,6.8Hz,1H),2.88(d,J=6.9Hz,1H),2.24(s,3H),1.48–1.23(m,6H),1.18(d,J=6.1Hz,6H).LC-ESI-MS:[M+H]+675。Step 2: Dissolve 183 mg (0.2 mmol) of XI-B-1 in 10 mL of anhydrous dichloromethane, and add 2 mL at room temperature (volume ratio: dichloromethane/triisopropylsilane/trifluoroacetic acid=5/1/ 1) The solution was reacted for about 1 h, and the reaction was completed by TLC. EtOAc (EtOAc m. The residue was purified by silica gel column chromatography to afford white crystals. 1 H NMR (400MHz, DMSO- d6) δ11.60 (s, 1H), 8.61 (d, J = 7.5Hz, 1H), 7.69 (d, J = 8.0Hz, 1H), 7.39 (t, J = 7.6 Hz, 2H), 7.22 (m, 3H), 6.19 - 5.90 (m, 2H), 5.69 (d, J = 7.8 Hz, 1H), 5.36 (s, 1H), 5.12 to 5.06 (m, 1H), 4.89 (dt, J = 12.2, 6.0 Hz, 1H), 4.37 - 4.15 (m, 3H), 4.18 - 3.98 (m, 2H), 3.57 (dd, J = 12.9, 6.8 Hz, 1H), 2.88 (d, J) = 6.9 Hz, 1H), 2.24 (s, 3H), 1.48 - 1.23 (m, 6H), 1.18 (d, J = 6.1 Hz, 6H). LC-ESI-MS: [M+H] + 675.
方法B:Method B:
Figure PCTCN2016109913-appb-000092
Figure PCTCN2016109913-appb-000092
依据实施例16方法B步骤一、步骤二,以VIII-B-1代替VIII-A-1,以5.3g(10mmol)化合物III-1为原料,可制备得5.7g白色固体产物III-2B-1,收率85%(两步)。ESI-MS:m/z[M+H]+=675。According to the method of Step B and Step 2 of Example 16, VIII-A-1 was replaced by VIII-B-1, and 5.3 g (10 mmol) of compound III-1 was used as a raw material to prepare 5.7 g of white solid product III-2B- 1, the yield of 85% (two steps). ESI-MS: m / z [ M + H] + = 675.
实施例23化合物III-2B-1a的合成Synthesis of Compound 23-2B-1a of Example 23
Figure PCTCN2016109913-appb-000093
Figure PCTCN2016109913-appb-000093
依据实施例16方法B步骤一、步骤二,以VIII-B-1a(由L-乙酰半胱氨酸制备得到,L构型手性纯度99%,分析方法:AD-H柱温:25C;检测波长:210nm;流速:1.0ml/min; 流动相:n-Hex:EtOH:TFA=90:10:0.1等度洗脱)代替VIII-A-1,以5.3g(10mmol)化合物III-1为原料,可制备得4.6g白色固体产物III-2B-1a,收率68%(两步).ESI-MS:m/z[M+H]+=675。According to the method of Example 16 Step B, Step 2, VIII-B-1a (prepared by L-acetylcysteine, L configuration chiral purity 99%, analytical method: AD-H column temperature: 25C; Detection wavelength: 210 nm; flow rate: 1.0 ml/min; mobile phase: n-Hex: EtOH: TFA = 90: 10: 0.1 isocratic elution instead of VIII-A-1, 5.3 g (10 mmol) of compound III-1 As a raw material, 4.6 g of a white solid product III-2B-1a was obtained in a yield of 68% (two steps). ESI-MS: m/z [M+H] + = 675.
实施例24化合物III-2B-1b的合成Synthesis of Example 14 Compound III-2B-1b
Figure PCTCN2016109913-appb-000094
Figure PCTCN2016109913-appb-000094
依据实施例22方法B步骤一、步骤二,以VIII-B-1b(由D-乙酰半胱氨酸制备得到,D构型手性纯度99%,分析方法:AD-H柱温:25C;检测波长:210nm;流速:1.0ml/min;流动相:n-Hex:EtOH:TFA=90:10:0.1等度洗脱)代替VIII-A-1,以5.3g(10mmol)化合物III-1为原料,可制备得5.2g白色固体产物II-A-1b,收率77%(两步).ESI-MS:m/z[M+H]+=675。According to the method of Example 22, Step B, Step 2, VIII-B-1b (prepared by D-acetylcysteine, D configuration chiral purity 99%, analytical method: AD-H column temperature: 25C; Detection wavelength: 210 nm; flow rate: 1.0 ml/min; mobile phase: n-Hex: EtOH: TFA = 90: 10: 0.1 isocratic elution instead of VIII-A-1, 5.3 g (10 mmol) of compound III-1 As a raw material, 5.2 g of a white solid product II-A-1b was obtained in a yield of 77% (two steps). ESI-MS: m/z [M+H] + = 675.
实施例25:化合物III-2B-2的合成Example 25: Synthesis of Compound III-2B-2
Figure PCTCN2016109913-appb-000095
Figure PCTCN2016109913-appb-000095
依据实施例19步骤一、二、三,以VIII-B-1a(由L-乙酰半胱氨酸制备得到,L构型手性纯度99%)代替VIII-A-1,以5.3g(10mmol)化合物III-1为原料,可制备得3.6g白色固体产物II-A-1b,收率51%。1H NMR(400MHz,DMSO-d6)δ11.59(s,1H),8.60(d,J=7.6Hz,1H),7.68(d,J=7.6Hz,1H),7.37(t,J=7.5Hz,2H),7.21(m,3H),6.17–5.95(m,2H),5.69(d,J=7.5Hz,1H),5.65(d,J=6.2Hz,1H),5.50(d,J=6.2Hz,1H),5.37(s,1H),5.22~5.16(m,1H),4.84(dt,J=12.2,6.0Hz,1H),4.35–4.17(m,3H),4.18–3.90(m,2H),3.53(m,1H),2.85(d,J=6.9Hz,1H),2.21(s,3H),1.44–1.22(m,6H),1.16(d,J=6.1Hz,6H).LC-ESI-MS:[M+H]+704。According to the first, second and third steps of Example 19, VIII-B-1a (prepared from L-acetylcysteine, L-form chiral purity 99%) was substituted for VIII-A-1 at 5.3 g (10 mmol). Compound III-1 was used as a starting material to obtain 3.6 g of a white solid product II-A-1b in a yield of 51%. 1 H NMR (400MHz, DMSO- d6) δ11.59 (s, 1H), 8.60 (d, J = 7.6Hz, 1H), 7.68 (d, J = 7.6Hz, 1H), 7.37 (t, J = 7.5 Hz, 2H), 7.21 (m, 3H), 6.17 - 5.95 (m, 2H), 5.69 (d, J = 7.5 Hz, 1H), 5.65 (d, J = 6.2 Hz, 1H), 5.50 (d, J) = 6.2 Hz, 1H), 5.37 (s, 1H), 5.22 to 5.16 (m, 1H), 4.84 (dt, J = 12.2, 6.0 Hz, 1H), 4.35 - 4.17 (m, 3H), 4.18 - 3.90 ( m, 2H), 3.53 (m, 1H), 2.85 (d, J = 6.9 Hz, 1H), 2.21 (s, 3H), 1.44 - 1.22 (m, 6H), 1.16 (d, J = 6.1 Hz, 6H) LC-ESI-MS: [M+H] + 704.
实施例26:化合物II-2B-5的合成 Example 26: Synthesis of Compound II-2B-5
Figure PCTCN2016109913-appb-000096
Figure PCTCN2016109913-appb-000096
依据实施例19步骤一、二、三,以VIII-B-1a(由L-乙酰半胱氨酸制备得到,L构型手性纯度99%)代替VIII-A-1,同时以III-2代替III-1,以5.4g(10mmol)化合物III-2为原料,可制备得4.2g白色固体产物III-2B-5,收率58%(两步)。LC-ESI-MS:[M+H]+721。Substituting VIII-A-1 (prepared from L-acetylcysteine, L-form chiral purity 99%) in place of VIII-A-1, in the presence of III-2, according to steps 19, 2 and 3 of Example 19. Instead of III-1, 5.4 g (10 mmol) of compound III-2 was used as a starting material to obtain 4.2 g of a white solid product III-2B-5 in a yield of 58% (two steps). LC-ESI-MS: [M+H] + 721.
实施例27:化合物III-2C-1的合成Example 27: Synthesis of Compound III-2C-1
Figure PCTCN2016109913-appb-000097
Figure PCTCN2016109913-appb-000097
步骤一:将529mg(1.0mmol)III-1溶于10mL无水二氯甲烷中,冰浴下加入452mg(1.0mmol)XV-1(制备方法参考Chemistry-A European Journal,2014,20,6526-6531)及DMAP 122mg(1.0mmol),搅拌1h,过滤,滤液用二氯甲烷(30mL)稀释,依次用水(10mL)及饱和食盐水(10mL)洗涤,分离有机相,无水硫酸钠干燥后过滤,减压浓缩除去溶剂。剩余物硅胶柱层析纯化得白色固体III-2C-1,收率:29%。1H NMR(400MHz,CDCl3)δ9.62(s,1H),7.46(dd,J=8.1,2.2Hz,1H),7.30-7.35(m,3H),7.29–7.22(m,3H),7.19(d,J=7.4Hz,1H),6.79(s,1H),6.16(d,J=18.1Hz,1H),6.06(d,J=8.3Hz,2H),6.00(s,2H),5.64–5.53(m,1H),4.50-4.60(m,3H),4.38–4.20(m,3H),4.20–4.04(m,1H),3.90-4.00(m,7H),3.77(s,3H),2.17(dd,J=8.4,2.2Hz,1H),1.46-1.55(m,3H),1.38(d,J=7.7Hz,3H),1.27(s,1H),1.24(d,J=6.3Hz,6H),LC-ESI-MS:[M+H]+946。Step 1: 529 mg (1.0 mmol) of III-1 was dissolved in 10 mL of anhydrous dichloromethane, and 452 mg (1.0 mmol) of XV-1 was added under ice bath (preparation method refers to Chemistry-A European Journal, 2014, 20, 6526- 6531) and DMAP 122 mg (1.0 mmol), stirred for 1 h, filtered, EtOAc EtOAc (EtOAc)EtOAc. The solvent was concentrated under reduced pressure. The residue was purified by silica gel column chromatography toield 1 H NMR (400MHz, CDCl 3 ) δ9.62 (s, 1H), 7.46 (dd, J = 8.1,2.2Hz, 1H), 7.30-7.35 (m, 3H), 7.29-7.22 (m, 3H), 7.19 (d, J = 7.4 Hz, 1H), 6.79 (s, 1H), 6.16 (d, J = 18.1 Hz, 1H), 6.06 (d, J = 8.3 Hz, 2H), 6.00 (s, 2H), 5.64–5.53 (m, 1H), 4.50-4.60 (m, 3H), 4.38–4.20 (m, 3H), 4.20–4.04 (m, 1H), 3.90-4.00 (m, 7H), 3.77 (s, 3H) ), 2.17 (dd, J = 8.4, 2.2 Hz, 1H), 1.46-1.55 (m, 3H), 1.38 (d, J = 7.7 Hz, 3H), 1.27 (s, 1H), 1.24 (d, J = 6.3 Hz, 6H), LC-ESI-MS: [M+H] + 946.
实施例28:抗丙肝病毒活性(HCV,EC50)以及细胞毒性(CC50)Example 28: Anti-hepatitis C virus activity (HCV, EC 50 ) and cytotoxicity (CC 50 )
由于缺乏理想的HCV体外感染细胞和动物模型,一般情况下,HCV病毒活性评价会采用HCV RNA复制中起关键作用的酶建立细胞外的分子复制模型。Due to the lack of ideal HCV in vitro infected cells and animal models, in general, HCV viral activity evaluation uses an enzyme that plays a key role in HCV RNA replication to establish an extracellular molecular replication model.
实验方法:在含有10%胎牛血清、1X非必需氨基酸、Pen-Strep-Glu、G418的DMEM培养基中,培养含HCV-复制子(1b基因型)的Huh7细胞。抗病毒筛选测试在不含G418的不同培养基中进行。细胞接种于96孔板,接种细胞后立即加入试验的化合物,并在培养箱中37℃孵育。然后除去培养基,细胞用于全核酸提取(包括复制子RNA和宿主RNA)。在Q-RT-PCR方案中可放大复制子RNA,并相应定量。所观察到的复制子HCV RNA与未处理对照组的水平差异作为试验化合物抗病毒效力的方式,其结果见表1。 Experimental method: Huh7 cells containing HCV-replicon (1b genotype) were cultured in DMEM medium containing 10% fetal calf serum, 1X non-essential amino acid, Pen-Strep-Glu, G418. Antiviral screening tests were performed in different media without G418. The cells were seeded in 96-well plates, and the test compounds were added immediately after inoculation of the cells, and incubated at 37 ° C in an incubator. The medium is then removed and the cells are used for whole nucleic acid extraction (including replicon RNA and host RNA). The replicon RNA can be amplified in the Q-RT-PCR protocol and quantified accordingly. The difference in the level of the observed replicon HCV RNA from the untreated control group was used as the manner of antiviral efficacy of the test compound, and the results are shown in Table 1.
表1化合物的抗丙肝病毒活性(HCV,EC50)以及细胞毒性(CC50)结果Anti-hepatitis C virus activity (HCV, EC 50 ) and cytotoxicity (CC 50 ) results for the compounds of Table 1
化合物编号Compound number EC50(μM)EC 50 (μM) CC50(μM)CC 50 (μM)
SofosbuvirSofosbuvir 0.180.18 >50>50
硫普罗宁Thiopronin >100>100 >50>50
II-A-1II-A-1 0.110.11 >50>50
II-A-1aII-A-1a 0.120.12 >50>50
II-A-1bII-A-1b 0.140.14 >50>50
II-A-2II-A-2 0.090.09 >50>50
II-A-4II-A-4 0.170.17 >50>50
II-A-5II-A-5 0.140.14 >50>50
II-A-8II-A-8 0.250.25 >50>50
II-B-1II-B-1 0.200.20 >50>50
II-B-1aII-B-1a 0.150.15 >50>50
II-B-2II-B-2 0.310.31 >50>50
II-B-4II-B-4 0.170.17 >50>50
III-2A-1III-2A-1 0.090.09 >50>50
III-2A-1aIII-2A-1a 0.130.13 >50>50
III-2A-1bIII-2A-1b 0.100.10 >50>50
III-2A-2III-2A-2 0.120.12 >50>50
III-2B-1III-2B-1 0.180.18 >50>50
III-2B-1aIII-2B-1a 0.150.15 >50>50
III-2B-2III-2B-2 0.160.16 >50>50
III-2C-1III-2C-1 0.130.13 >50>50
化合物对细胞内HCV复制子1b的抑制活性(EC50)的结果作为评价其抗丙肝病毒活性,而同时细胞毒活性(CC50)的同步测试用于排除由于细胞毒性造成的假阳性结果。治疗指数越高,代表化合物越有应用前景。结果表明,本发明提供的化合物表现出与Sofosbuvir相似或者更优的活性和治疗指数,以II-A-1、II-A-1a、II-A-1b、II-A-2、III-2A-1、III-2A-2为例,其对细胞内HCV复制子的抑制活性(EC50)明显强于比阳性对照Sofosbuvir,表明该化合物在肝细胞内可与Sofosbuvir一样能够在细胞内快速降解成为活性代谢产物的形式发挥抗HCV的作用,此外,分子中降解得到的另一份子硫普罗宁对于抗病毒活性有一定的增益作用,而这种协同效应使本发明提供的化合物,在抗HCV疾病方面更具有应用前景。The results of the inhibitory activity of the compound of intracellular replicon 1b of HCV (EC 50) was evaluated as an anti-hepatitis C virus activity, while cytotoxic activity (CC 50) for excluding a synchronization test false positive results due to the cytotoxicity caused. The higher the therapeutic index, the more promising the representative compound is. The results indicate that the compounds provided by the present invention exhibit similar or superior activity and therapeutic index to Sofosbuvir, as II-A-1, II-A-1a, II-A-1b, II-A-2, III-2A. -1, III-2A-2, for example, inhibitory activity on intracellular HCV replicon (EC 50) was stronger than that in the positive control Sofosbuvir, indicating that the compound may be rapidly Sofosbuvir as intracellular degradation in the liver cells The form that becomes an active metabolite exerts an anti-HCV effect. In addition, another sub-thiopronin obtained by degradation in the molecule has a certain gain effect on antiviral activity, and this synergistic effect enables the compound provided by the present invention to be resistant to HCV. The disease aspect has more application prospects.
实施例29:小鼠CCl4肝损模型的保护作用研究Example 29: Study on the protective effect of mouse CCl 4 liver damage model
实验方法:取体重24~28g的雄性小鼠实验前,禁食不禁水6小时,按体重随机分成3组,每组5只小鼠,分设空白对照组、CCl4组、化合物组(硫普罗宁50mg/kg、试验化合物200mg/kg)。每12h灌胃给药1次,每次灌胃容量均为10mL/kg,共给药4次。空白对照组和模型对照组同时灌服等容量的生理盐水。在第三次给药后1h,除空白对照组小鼠外,其它两组小鼠按2mg/kg b.w的CCl4腹腔注射50%CCl4玉米油溶液。于12小时后,各组再给药1次。于24小时后,各鼠取血,测定血清ALT和AST(表2),取肝脏切小块,甲醛固定后,石蜡切片,用苏木精和伊红染色,显微镜下观察肝细胞情况。Experimental Method: Male mice were taken before the experiment body weight of 24 ~ 28g, water, fasted 6 hours, were randomly divided into 3 groups, 5 mice per group, divided into blank control group, CCl 4 group, compound group (thio general Ning 50mg/kg, test compound 200mg/kg). The drug was administered once every 12 hours, and the perfusion volume was 10 mL/kg, which was administered 4 times. The blank control group and the model control group were simultaneously administered with an equal volume of physiological saline. One hour after the third administration, the other two groups of mice were intraperitoneally injected with 50% CCl 4 corn oil solution at 2 mg/kg bw of CCl 4 except for the blank control group. After 12 hours, each group was administered once more. After 24 hours, the rats were bled and serum ALT and AST (Table 2) were measured. The liver was cut into small pieces, fixed in formaldehyde, paraffin-embedded, stained with hematoxylin and eosin, and the liver cells were observed under a microscope.
表2本发明化合物的CCl4肝损模型的保护作用研究结果Table 2 Results of the protective effect of the CCl4 liver damage model of the compound of the present invention
组别Group ALT(U/L)ALT (U/L) AST(U/L)AST (U/L)
空白blank 58.258.2 125.4125.4
CCl4对照组CCl 4 control group 1423.21423.2 1214.31214.3
SofosbuvirSofosbuvir 1325.71325.7 1185.21185.2
硫普罗宁Thiopronin 689.3689.3 733.8733.8
II-A-1II-A-1 617.5617.5 690.2690.2
II-A-1aII-A-1a 596.3596.3 672672
II-A-1bII-A-1b 620.5620.5 701.5701.5
II-A-2II-A-2 560.7560.7 656.3656.3
II-A-4II-A-4 701.5701.5 677.8677.8
II-A-5II-A-5 596.2596.2 702.3702.3
II-B-1II-B-1 725.6725.6 833.1833.1
II-B-1aII-B-1a 699.1699.1 795.3795.3
III-2A-1aIII-2A-1a 663.5663.5 701.5701.5
III-2A-1bIII-2A-1b 682.3682.3 679.2679.2
III-2B-1aIII-2B-1a 710.3710.3 695.8695.8
III-2C-1III-2C-1 505.2505.2 609.3609.3
结果表明,CCl4模型组小鼠的ALT和AST异常升高,硫普罗宁能显著逆转这种升酶作用,相比较下,Sofosbuvir则没有这方面的活性。而本发明提供的化合物具有明显的降酶作用,活性与硫普罗宁相当或者更优。究其原因,可能是因为硫普罗宁的羧基被制成前药后,可提高了其稳定性和被口服吸收的能力,从而使其活性得到了增强。以II-A-1、II-A-a、II-A-2、II-A-5、III-2A-1a为例,其对CCl4诱导肝损伤导致ALT和AST升高作用,有明显的抑制活性,且优于阳性对照硫普罗宁。此外,肝脏组织病理切片观察结果为,CCl4模型组,肝中央静脉淤血,周围有小叶中央性或周围性坏死,肝细胞有肿胀、气球样变形,II-A-1、II-A-a、II-A-2、II-A-5、III-2A-1a等给药组呈现少量点状和碎片状坏死,大部分呈现气球样变形,显示由CCl4引起的毒性已大大减少,说明本发明涉及的化合物确实有保护CCl4引起的肝损作用。The results showed that ALT and AST were abnormally elevated in the CCl 4 model group, and tiopronin significantly reversed the effect of this enzyme. In comparison, Sofosbuvir did not have this activity. However, the compounds provided by the present invention have a significant enzyme-lowering effect, and the activity is comparable to or superior to that of tiopronin. The reason may be that the thiopronin's carboxyl group is made into a prodrug, which improves its stability and its ability to be absorbed orally, thereby enhancing its activity. Taking II-A-1, II-Aa, II-A-2, II-A-5, and III-2A-1a as examples, the CCl4-induced liver injury leads to an increase in ALT and AST, and has significant inhibitory activity. And superior to the positive control tiopronin. In addition, liver histopathology observation result, CCl 4 model group, the central hepatic venous congestion, around lobular central or peripheral necrosis, liver cell swelling, ballooning deformation, II-A-1, II -Aa, II -A-2, II-A-5, III-2A-1a and other administration groups showed a small amount of punctate and fragmented necrosis, most of which showed balloon-like deformation, indicating that the toxicity caused by CCl 4 has been greatly reduced, indicating the present invention The compounds involved do have a protective effect on liver damage caused by CCl 4 .
由于缺乏理想的HCV感染动物模型,直接评价本发明提供的化合物对HCV引起肝损伤的保护作用不可行,但理论上以及临床应用的角度来讲,本发明提供的化合物的保肝护肝的作用对于HCV引起的炎症反应同样具有保护作用。Due to the lack of an ideal animal model of HCV infection, it is not feasible to directly evaluate the protective effect of the compounds provided by the present invention on HCV-induced liver injury, but in theory and clinical application, the compound provided by the present invention has the function of protecting liver and protecting liver It also has a protective effect on the inflammatory response caused by HCV.
实施例30:药代动力学性质研究Example 30: Study on pharmacokinetic properties
1实验材料1 experimental material
1.1化合物1.1 compound
使用以上实施例制备的本发明的化合物II-A-1、II-A-1a、II-A-1b、II-A-2、II-B-1、III-2A-1、III-2A-1a(30mg/kg,其核苷部分的摩尔质量与索菲组一致),阳性对照药为索菲布韦(25mg/kg),将各化合物加至5%DMSO+60%PEG400+35%生理盐水,涡旋后制备成10mg/ml混悬液,用于灌胃给药。The compounds of the present invention prepared using the above examples II-A-1, II-A-1a, II-A-1b, II-A-2, II-B-1, III-2A-1, III-2A- 1a (30mg/kg, the molar mass of the nucleoside moiety is consistent with the Sophie group), the positive control drug is Sophibuvir (25mg/kg), and each compound is added to 5% DMSO + 60% PEG400 + 35% physiological Saline was prepared by vortexing to a 10 mg/ml suspension for intragastric administration.
GS331007购买自韶远科技(上海)有限公司。GS331007 was purchased from Shunyuan Technology (Shanghai) Co., Ltd.
1.2实验动物和采样1.2 Experimental animals and sampling
以SD大鼠为实验动物(体重180-220g),每个试验化合物6只SD大鼠,随机分成2组(灌胃组和静注组),每组3只。灌胃给药的尾静脉取血时间点为0.17,0.33,0.5,1,1.5,2,4,6,8,12,24小时;静脉给药取血时间点为0.05,0.1,0.17,0.5,1,2,4,6,8,12,24小时。取全血0.3ml,离心后取血浆0.1ml采用LC-MS进行分析。SD rats were used as experimental animals (body weight 180-220 g), and 6 SD rats per test compound were randomly divided into 2 groups (gavage group and intravenous injection group), 3 in each group. The time of blood collection from the tail vein of the intragastric administration was 0.17, 0.33, 0.5, 1, 1.5, 2, 4, 6, 8, 12, 24 hours; the time of blood collection by intravenous administration was 0.05, 0.1, 0.17, 0.5. 1,2,4,6,8,12,24 hours. 0.3 ml of whole blood was taken, and 0.1 ml of plasma was taken after centrifugation and analyzed by LC-MS.
1.3仪器1.3 instruments
Agilent 1200液相色谱系统,API4000QTRAP三重四极杆串联质谱仪Agilent 1200 LC System, API4000QTRAP Triple Quadrupole Mass Spectrometer
1.4样品处理1.4 sample processing
取30μl肝脏匀浆样品或标准曲线样品加入至100μl含内标(100ng/ml)的乙腈中,涡旋2min后离心10min(6000转/min),将上清液转移至进样瓶中;30 μl of liver homogenate sample or standard curve sample was added to 100 μl of acetonitrile containing internal standard (100 ng/ml), vortexed for 2 min, centrifuged for 10 min (6000 rpm), and the supernatant was transferred to a sample vial;
取5μL上清液,进样,采用LC-MS/MS检测样品中GS331007的浓度。根据所得的肝匀浆药物浓度-时间数据,使用药代动力学计算软件WinNonlin6.2.1非房室模型分别计算各测试化合物的代谢物GS331007的药代动力学参数,结果见表3。 5 μL of the supernatant was taken, injected, and the concentration of GS331007 in the sample was measured by LC-MS/MS. Based on the obtained liver homogenate drug concentration-time data, the pharmacokinetic parameters of the metabolite GS331007 of each test compound were calculated using the pharmacokinetic calculation software WinNonlin 6.2.1 non-compartmental model. The results are shown in Table 3.
表3SD大鼠口服给药后主要药动学参数汇总Table 3 Summary of main pharmacokinetic parameters of SD rats after oral administration
Figure PCTCN2016109913-appb-000098
Figure PCTCN2016109913-appb-000098
根据文献报道,其在体内的代谢途径如下图所示,其中GS331007是最终代谢产物,是研究Sofosbuvir在体内代谢行为的标志物。重点考察了化合物II-A-1、II-A-1a、II-A-1b、II-A-2、II-B-1、III-2A-1a在大鼠体内GS331007血药浓度和代谢行为,以考察受试化合物的药代动力学性质,结果表明,受试化合物的GS331007血药浓度和曲线下面积与索菲布韦组大鼠体内GS331007的血药浓度相比有明显改善,换算得口服生物利用度改善明显。以化合物II-A-1、II-A-2、II-B-1为例,其口服生物利用度分别是索菲布韦的3.38、2.75、2.25倍。另外,II-A-1和II-A-2的其半衰期分别达到了6.14和6.23小时,分别为索菲布韦的2.95和2.51倍,其药时曲线更为缓和。因此,本发明涉及的化合物在药动学性质上优势明显,可通过口服吸收给药用于丙肝疾病的治疗。According to the literature, its metabolic pathway in vivo is shown in the figure below, in which GS331007 is the final metabolite and is a marker for studying the metabolic behavior of Sofosbuvir in vivo. The blood concentration and metabolic behavior of GS331007 in rats II-A-1, II-A-1a, II-A-1b, II-A-2, II-B-1 and III-2A-1a were investigated. In order to investigate the pharmacokinetic properties of the test compound, the results showed that the blood concentration of GS331007 and the area under the curve of the test compound were significantly improved compared with the blood concentration of GS331007 in the sofosbuvir group. Oral bioavailability improved significantly. Taking the compounds II-A-1, II-A-2, and II-B-1 as examples, the oral bioavailability was 3.38, 2.75, and 2.25 times of Soofibvir, respectively. In addition, the half-lives of II-A-1 and II-A-2 reached 6.14 and 6.23 hours, respectively, which were 2.95 and 2.51 times that of Sophibuvir, respectively, and the drug-time curve was more moderate. Therefore, the compounds of the present invention are superior in pharmacokinetic properties and can be administered by oral absorption for the treatment of hepatitis C diseases.
实施例31:体外PK性质研究Example 31: In vitro PK properties study
取受试化合物III-2A-1(1mg/mL)测试其人模拟胃液、模拟肠液、人血浆、人肝微粒体中的稳定性。结果表明,III-2A-1在模拟胃液中稳定性高(1小时的代谢率<5%),III-2A-1在模拟肠液(5分钟代谢率均大于90%)、血浆(5分钟代谢率均大于99%)和肝微粒体(5分钟代谢率均大于99%)中可快速降解并释放出索菲布韦和硫普罗宁。该结果证明测试化合物经口服可经肠道、血浆、肝脏快速代谢,并释放出原型药物。与索菲布韦嘧啶氮原子衍生化的类似化合物II-A-1(CN201510932904.2)相比,其在模拟胃液中的稳定性更高,而在模拟肠液、血浆和肝微粒体中的代谢转化速率更快。考虑到药物的吸收主要发生在小肠,因此,与索菲布韦的嘧啶氮原子衍生物(CN201510932904.2)相比,从索菲布韦的核苷羟基上进行衍生,所得化合物III-2A-1及其类似物其体外的PK性质更有利于其在体内转化并实现吸收,预计其作为双功能的前药分子发挥抗丙肝“标本兼治”的疗效更佳。Test compound III-2A-1 (1 mg/mL) was tested for stability in human simulated gastric juice, simulated intestinal fluid, human plasma, and human liver microsomes. The results showed that III-2A-1 has high stability in simulated gastric fluid (1 hour metabolic rate <5%), III-2A-1 in simulated intestinal fluid (5 minutes metabolic rate is greater than 90%), plasma (5 minutes metabolism) Both rates are greater than 99%) and liver microsomes (more than 99% in 5 minutes) can rapidly degrade and release sofosbuvir and tiopronin. This result demonstrates that the test compound can be rapidly metabolized by the intestine, plasma, and liver by oral administration, and the prototype drug is released. Compared to the similar compound II-A-1 (CN201510932904.2) derivatized with the sofosbuvir pyrimidine nitrogen atom, it is more stable in simulated gastric fluid, but mimics metabolism in intestinal fluid, plasma and liver microsomes. The conversion rate is faster. Considering that the absorption of the drug mainly occurs in the small intestine, it is derived from the nucleoside hydroxyl group of Sophibruvir compared to the pyrimidine nitrogen atom derivative of Sophibruvir (CN201510932904.2), and the resulting compound III-2A- The PK properties of 1 and its analogues in vitro are more favorable for its in vivo transformation and absorption, and it is expected to be more effective as a bifunctional prodrug molecule for anti-hepatitis C "speculation and treatment".
实施例32:受试化合物的化学稳定性研究Example 32: Chemical stability study of test compounds
取受试化合物III-2A-1测试(100mg),放置于40±2℃、75±5%的稳定箱中,进行加速稳定性实验。1个月的实验表明,III-2的降解率<1%(新增杂质含量),而与之前发现的索菲布韦的嘧啶氮原子衍生化的类似化合物II-A-1(CN201510932904.2)相比,其化学稳定性具有明显优势(同等条件下,II-A-1的降解率为4.5%)。因此,与索菲布韦的嘧啶氮原子衍生化的类似化合物相比,从索菲布韦的核苷羟基上通过进行衍生,所得化合物的化学稳定性更高,有利于其进一步开发为双功能的抗丙肝候选药物。The test compound III-2A-1 test (100 mg) was taken and placed in a stable container of 40 ± 2 ° C and 75 ± 5% to carry out an accelerated stability test. A one-month experiment showed that the degradation rate of III-2 was <1% (new impurity content), and similar compound II-A-1 was derivatized with the pyrimidine nitrogen atom of Sofibvir previously found (CN201510932904.2). Compared with its chemical stability, it has obvious advantages (the degradation rate of II-A-1 is 4.5% under the same conditions). Therefore, compared with a similar compound derivatized with the pyrimidine nitrogen atom of Sophibruvir, the chemical stability of the obtained compound is higher by derivatization from the nucleoside hydroxyl group of Sophibuvir, which facilitates its further development into a dual function. Anti-HC candidate drugs.
以上所述仅为本发明的较佳实施例,并不用以限制本发明,凡在本发明的精神和原则之内所作的任何修改、等同替换和改进等,均包含在本发明的保护范围之内。 The above are only the preferred embodiments of the present invention, and are not intended to limit the present invention. Any modifications, equivalents, and improvements made within the spirit and scope of the present invention are included in the scope of the present invention. Inside.

Claims (34)

  1. 一种化合物,其具有式I所示结构,或式I所示结构的互变异构体、光学异构体、立体异构体、立体异构体混合物、前药、药学上可接受的盐或溶剂合物:A compound having the structure of Formula I, or a tautomer, an optical isomer, a stereoisomer, a mixture of stereoisomers, a prodrug, a pharmaceutically acceptable salt of the structure of Formula I. Or solvate:
    Figure PCTCN2016109913-appb-100001
    Figure PCTCN2016109913-appb-100001
    式I中,X为羟基、F、Cl或Br;In formula I, X is hydroxy, F, Cl or Br;
    R、R’分别独立的为H、或者
    Figure PCTCN2016109913-appb-100002
    且R、R’中至少有一个为
    Figure PCTCN2016109913-appb-100003
    其中Linker可以存在或不存在,D来自于用于治疗或辅助治疗肝病的药物或其衍生物。
    R, R' are independent of H, or
    Figure PCTCN2016109913-appb-100002
    And at least one of R and R' is
    Figure PCTCN2016109913-appb-100003
    Where Linker may or may not be present, and D is derived from a drug or derivative thereof for use in the treatment or adjuvant treatment of liver disease.
  2. 根据权利要求1所述的化合物,其具有式I-1所示结构,或式I-1所示结构的互变异构体、光学异构体、立体异构体、立体异构体混合物、前药、药学上可接受的盐或溶剂合物:The compound according to claim 1, which has a structure represented by Formula I-1, or a tautomer, an optical isomer, a stereoisomer, a mixture of stereoisomers of the structure represented by Formula I-1, Prodrug, pharmaceutically acceptable salt or solvate:
    Figure PCTCN2016109913-appb-100004
    Figure PCTCN2016109913-appb-100004
    其中,X为羟基或F、Cl、Br;Wherein X is a hydroxyl group or F, Cl, Br;
    R为
    Figure PCTCN2016109913-appb-100005
    R is
    Figure PCTCN2016109913-appb-100005
    Linker A为
    Figure PCTCN2016109913-appb-100006
    或不存在,G来自于用于治疗或辅助治疗肝病的药物或其衍生物;
    Linker A is
    Figure PCTCN2016109913-appb-100006
    Or absent, G is derived from a drug or a derivative thereof for treating or assisting in the treatment of liver disease;
    R1为H或C1-C5烷基;n为1-19的整数。R 1 is H or C 1 -C 5 alkyl; n is an integer from 1 to 19.
  3. 根据权利要求2所述的化合物,其具有式II-1A所示的结构或其互变异构体、光学异构体、立体异构体、立体异构体混合物、前药、药学上可接受的盐或溶剂合物:The compound according to claim 2, which has the structure of the formula II-1A or a tautomer, an optical isomer, a stereoisomer, a mixture of stereoisomers, a prodrug, and a pharmaceutically acceptable compound. Salt or solvate:
    Figure PCTCN2016109913-appb-100007
    Figure PCTCN2016109913-appb-100007
    其中,X为羟基或F、Cl、Br;Wherein X is a hydroxyl group or F, Cl, Br;
    Linker A为
    Figure PCTCN2016109913-appb-100008
    或不存在;
    Linker A is
    Figure PCTCN2016109913-appb-100008
    Or does not exist;
    R1为H或C1-C5烷基;n为1、2、3、4或5;R3、R4各自独立地为氢原子或C1-C5烷基。R 1 is H or C 1 -C 5 alkyl; n is 1, 2, 3 , 4 or 5; and R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group.
  4. 根据权利要求3所述的化合物,其具有式II-1Aa或者式II-1Ab所示的结构或其互变异构体、光学异构体、立体异构体、立体异构体混合物、前药、药学上可接受的盐或溶剂合物: The compound according to claim 3, which has the structure represented by Formula II-1Aa or Formula II-1Ab or a tautomer, optical isomer, stereoisomer, stereoisomer mixture, prodrug thereof a pharmaceutically acceptable salt or solvate:
    Figure PCTCN2016109913-appb-100009
    Figure PCTCN2016109913-appb-100009
    其中,X为羟基或F、Cl、Br;Wherein X is a hydroxyl group or F, Cl, Br;
    Linker A为
    Figure PCTCN2016109913-appb-100010
    或不存在;
    Linker A is
    Figure PCTCN2016109913-appb-100010
    Or does not exist;
    R1为H或C1-C5烷基;n为1、2、3、4或5;R3、R4各自独立地为氢原子或C1-C5烷基。R 1 is H or C 1 -C 5 alkyl; n is 1, 2, 3 , 4 or 5; and R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group.
  5. 根据权利要求2所述的化合物,其具有式II-1B所示的结构或其互变异构体、光学异构体、立体异构体、立体异构体混合物、前药、药学上可接受的盐或溶剂合物:The compound according to claim 2, which has the structure of the formula II-1B or a tautomer, an optical isomer, a stereoisomer, a mixture of stereoisomers, a prodrug, and a pharmaceutically acceptable compound. Salt or solvate:
    Figure PCTCN2016109913-appb-100011
    Figure PCTCN2016109913-appb-100011
    其中,X为羟基或F、Cl、Br;Wherein X is a hydroxyl group or F, Cl, Br;
    Linker A为
    Figure PCTCN2016109913-appb-100012
    或不存在;
    Linker A is
    Figure PCTCN2016109913-appb-100012
    Or does not exist;
    R1为H或C1-C5烷基;n为1、2、3、4或5;R3、R4各自独立地为氢原子或C1-C5烷基。R 1 is H or C 1 -C 5 alkyl; n is 1, 2, 3 , 4 or 5; and R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group.
  6. 根据权利要求5所述的化合物,其具有式II-1Ba或式II-1Bb所示的结构或其光学异构体、药学上可接受的盐或溶剂合物:The compound according to claim 5, which has the structure represented by Formula II-1Ba or Formula II-1Bb or an optical isomer, pharmaceutically acceptable salt or solvate thereof:
    Figure PCTCN2016109913-appb-100013
    Figure PCTCN2016109913-appb-100013
    其中,X为羟基或F、Cl、Br;Wherein X is a hydroxyl group or F, Cl, Br;
    Linker A为
    Figure PCTCN2016109913-appb-100014
    或不存在;
    Linker A is
    Figure PCTCN2016109913-appb-100014
    Or does not exist;
    R1为H或C1-C5烷基;n为1、2、3、4或5;R3、R4各自独立地为氢原子或C1-C5烷基。R 1 is H or C 1 -C 5 alkyl; n is 1, 2, 3 , 4 or 5; and R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group.
  7. 根据权利要求2所述的化合物,其为式II-1A-A所示含有硫普罗宁结构的前药或其立体异构体,立体异构体混合物或其药学上可接受的盐或溶剂合物: The compound according to claim 2, which is a prodrug of a tiopronin structure represented by the formula II-1A-A or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable salt or solvate thereof Object:
    Figure PCTCN2016109913-appb-100015
    Figure PCTCN2016109913-appb-100015
    其中,X为羟基或F、Cl、Br;Wherein X is a hydroxyl group or F, Cl, Br;
    Linker A为
    Figure PCTCN2016109913-appb-100016
    Linker A is
    Figure PCTCN2016109913-appb-100016
    R1为H或C1-C5烷基;n为1-19的整数。R 1 is H or C 1 -C 5 alkyl; n is an integer from 1 to 19.
  8. 根据权利要求7所述化合物,其为式II-1A-Aa、式II-1A-Ab、式II-1A-Ac、式II-1A-Ad分别所示的含有硫普罗宁结构的前药或其立体异构体,立体异构体混合物或其药学上可接受的盐或溶剂合物:a compound according to claim 7 which is a prostaglandin containing prodrug of formula II-1A-Aa, formula II-1A-Ab, formula II-1A-Ac, formula II-1A-Ad or a stereoisomer, a mixture of stereoisomers or a pharmaceutically acceptable salt or solvate thereof:
    Figure PCTCN2016109913-appb-100017
    Figure PCTCN2016109913-appb-100017
    其中,X为羟基或F、Cl、Br;R1为H或C1-C5烷基;n为1、2、3、4或5。Wherein X is a hydroxyl group or F, Cl, Br; R 1 is H or a C 1 -C 5 alkyl group; and n is 1, 2, 3, 4 or 5.
  9. 根据权利要求8所述的化合物,其为式II-1A-Aa1、式II-1A-Ab1、式II-1A-Ac1、式II-1A-Ad1分别所示的含有硫普罗宁结构的前药或其互变异构体、立体异构体,立体异构体混合物或其药学上可接受的盐或溶剂合物:The compound according to claim 8, which is a profenofin-containing prodrug represented by the formula II-1A-Aa1, the formula II-1A-Ab1, the formula II-1A-Ac1, and the formula II-1A-Ad1, respectively. Or a tautomer, a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable salt or solvate thereof:
    Figure PCTCN2016109913-appb-100018
    Figure PCTCN2016109913-appb-100018
    Figure PCTCN2016109913-appb-100019
    Figure PCTCN2016109913-appb-100019
    其中,R1为H或C1-C5烷基;n为1、2、3、4或5。Wherein, R 1 is H or C 1 -C 5 alkyl group; n is 3, 4 or 5.
  10. 根据权利要求3所述的化合物,其具有以下所示的结构或以下所示结构化合物的互变异构体、立体异构体、立体异构体混合物、药学上可接受的盐或溶剂合物:The compound according to claim 3, which has the structure shown below or a tautomer, a stereoisomer, a mixture of stereoisomers, a pharmaceutically acceptable salt or a solvate of the structural compound shown below. :
    Figure PCTCN2016109913-appb-100020
    Figure PCTCN2016109913-appb-100020
    Figure PCTCN2016109913-appb-100021
    Figure PCTCN2016109913-appb-100021
    Figure PCTCN2016109913-appb-100022
    Figure PCTCN2016109913-appb-100022
    Figure PCTCN2016109913-appb-100023
    Figure PCTCN2016109913-appb-100023
  11. 一种式II-1A所示的化合物的制备方法,步骤为:A method for preparing a compound of the formula II-1A, the steps are:
    1)式III所示的化合物与式IV-1A所示的化合物反应得到式V-1A所示化合物;1) a compound of formula III is reacted with a compound of formula IV-1A to give a compound of formula V-1A;
    2)式V-1A所示的化合物经过脱保护反应得到式II-1A所述的化合物;2) a compound of the formula V-1A is subjected to a deprotection reaction to give a compound of the formula II-1A;
    Figure PCTCN2016109913-appb-100024
    Figure PCTCN2016109913-appb-100024
    其中,X为羟基或F、Cl、Br;Wherein X is a hydroxyl group or F, Cl, Br;
    Linker A为
    Figure PCTCN2016109913-appb-100025
    或不存在;
    Linker A is
    Figure PCTCN2016109913-appb-100025
    Or does not exist;
    n为1、2、3、4或5;R1为H或C1-C5烷基;R3、R4各自独立地为氢原子或C1-C5烷基;LG为离去基团;Z为巯基保护基团。n is 1, 2, 3, 4 or 5; R 1 is H or C 1 -C 5 alkyl; R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group; LG is a leaving group. Group; Z is a thiol protecting group.
  12. 一种式II-1B所示的化合物的制备方法,步骤为:A method for preparing a compound of the formula II-1B, the steps are:
    1)式III所示的化合物与式IV-1B所示的化合物反应得到式V-1B所示的化合物;1) a compound of formula III is reacted with a compound of formula IV-1B to give a compound of formula V-1B;
    2)式V-1B所示的化合物经脱保护反应得到式II-1B所示的化合物;2) a compound represented by the formula V-1B is subjected to a deprotection reaction to obtain a compound represented by the formula II-1B;
    Figure PCTCN2016109913-appb-100026
    Figure PCTCN2016109913-appb-100026
    其中,X为羟基或F、Cl、Br; Wherein X is a hydroxyl group or F, Cl, Br;
    Linker A为
    Figure PCTCN2016109913-appb-100027
    或不存在;
    Linker A is
    Figure PCTCN2016109913-appb-100027
    Or does not exist;
    n为1、2、3、4或5;R1为H或C1-C5烷基;R3、R4各自独立地为氢原子或C1-C5烷基;LG为离去基团;Z为巯基保护基团。n is 1, 2, 3, 4 or 5; R 1 is H or C 1 -C 5 alkyl; R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group; LG is a leaving group. Group; Z is a thiol protecting group.
  13. 一种式II-1A-Aa所示的化合物的制备方法,由式IIIm所示的化合物与式IV-1所示的化合物经过烃化反应得到式V-1A所示的化合物,然后经过脱保护反应得到:A process for the preparation of a compound of the formula II-1A-Aa, wherein a compound of the formula IIIm and a compound of the formula IV-1 are subjected to an alkylation reaction to obtain a compound of the formula V-1A, which is then subjected to deprotection. The reaction is obtained:
    Figure PCTCN2016109913-appb-100028
    Figure PCTCN2016109913-appb-100028
    其中,X为羟基或F、Cl或Br;R1为H或C1-C5烷基;Y为离去基团;Z为巯基保护基团。Wherein X is a hydroxyl group or F, Cl or Br; R 1 is H or a C 1 -C 5 alkyl group; Y is a leaving group; and Z is a thiol protecting group.
  14. 一种式II-1A-Ab所示的化合物的制备方法,由式IIIm所示的化合物与式IV-1B所示的化合物经过烃化反应得到式V-1B所示的化合物,然后经过脱保护反应得到:A process for the preparation of a compound of the formula II-1A-Ab, wherein a compound of the formula IIIm and a compound of the formula IV-1B are subjected to an alkylation reaction to obtain a compound of the formula V-1B, which is then subjected to deprotection. The reaction is obtained:
    Figure PCTCN2016109913-appb-100029
    Figure PCTCN2016109913-appb-100029
    其中,X为羟基或F、Cl、Br;R1为H或C1-C5烷基;n为1、2、3、4或5;Y为离去基团;Z为巯基保护基团。Wherein X is hydroxy or F, Cl, Br; R 1 is H or C 1 -C 5 alkyl; n is 1, 2, 3, 4 or 5; Y is a leaving group; Z is a thiol protecting group .
  15. 一种式II-1A-B所示的化合物的制备方法,步骤为:A method for preparing a compound of the formula II-1A-B, the steps are:
    1)式III所示的化合物经过保护反应,然后与醛反应得到式VII-1所示的化合物;1) a compound of formula III is subjected to a protective reaction and then reacted with an aldehyde to give a compound of formula VII-1;
    2)式VII-1所示的化合物与VIII-1A所述的化合物发生反应得到式IX-1A-B所述的化合物;2) a compound of the formula VII-1 is reacted with a compound of VIII-1A to give a compound of the formula IX-1A-B;
    3)式IX-1A-B所示的化合物脱去保护基得到式II-1A-B所示的化合物; 3) a compound of formula IX-1A-B is deprotected to give a compound of formula II-1A-B;
    Figure PCTCN2016109913-appb-100030
    Figure PCTCN2016109913-appb-100030
    其中,X为羟基或F、Cl、Br;R1为H或C1-C5烷基;R3、R4各自独立地为氢原子或C1-C5烷基;P1为羟基保护基团;P2为酰胺NH保护基团;P3为巯基保护基团。Wherein X is a hydroxyl group or F, Cl, Br; R 1 is H or a C 1 -C 5 alkyl group; R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group; and P 1 is a hydroxy group. a group; P 2 is an amide NH protecting group; and P 3 is a thiol protecting group.
  16. 一种式II-1B-B所示的化合物的制备方法,步骤为:A method for preparing a compound of the formula II-1B-B, the steps are:
    1)式VII-1所示的化合物与VIII-1B所示的化合物发生反应得到式IX-1B-B所述的化合物;1) a compound of the formula VII-1 is reacted with a compound represented by VIII-1B to give a compound of the formula IX-1B-B;
    2)式IX-1B-B所示的化合物脱去保护基得到式II-1B-B所示的化合物;2) a compound of formula IX-1B-B is deprotected to give a compound of formula II-1B-B;
    Figure PCTCN2016109913-appb-100031
    Figure PCTCN2016109913-appb-100031
    其中,X为羟基或F、Cl、Br;R1为H或C1-C5烷基;R3、R4各自独立地为氢原子或C1-C5烷基;P1为羟基保护基团;P2为酰胺NH保护基团;P3为巯基保护基团。Wherein X is a hydroxyl group or F, Cl, Br; R 1 is H or a C 1 -C 5 alkyl group; R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group; and P 1 is a hydroxy group. a group; P 2 is an amide NH protecting group; and P 3 is a thiol protecting group.
  17. 一种化合物,其具有式VII-1所示的结构或其立体异构体、立体异构体混合物或药学上可接受的盐: A compound having the structure of formula VII-1 or a stereoisomer, a mixture of stereoisomers or a pharmaceutically acceptable salt thereof:
    Figure PCTCN2016109913-appb-100032
    Figure PCTCN2016109913-appb-100032
    其中,X为羟基或F、Cl、Br;R1为H或C1-C5烷基;P1为羟基保护基团。Wherein X is a hydroxyl group or F, Cl, Br; R 1 is H or a C 1 -C 5 alkyl group; and P 1 is a hydroxy protecting group.
  18. 一种化合物,其具有式VIII-1A所示的结构或其立体异构体、立体异构体混合物或药学上可接受的盐:A compound having the structure of Formula VIII-1A or a stereoisomer, a mixture of stereoisomers or a pharmaceutically acceptable salt thereof:
    Figure PCTCN2016109913-appb-100033
    Figure PCTCN2016109913-appb-100033
    其中,P2为NH保护基团;P3为巯基基保护基团。Wherein P 2 is an NH protecting group; and P 3 is a fluorenyl protecting group.
  19. 一种化合物,其具有式IX-1A-B所示的结构或其立体异构体、立体异构体混合物或药学上可接受的盐:A compound having the structure of the formula IX-1A-B or a stereoisomer, a mixture of stereoisomers or a pharmaceutically acceptable salt thereof:
    Figure PCTCN2016109913-appb-100034
    Figure PCTCN2016109913-appb-100034
    其中,X为羟基或F、Cl、Br;R1为H或C1-C5烷基;P1为羟基保护基团;P2为NH保护基团;P3为巯基保护基团。Wherein X is a hydroxyl group or F, Cl, Br; R 1 is H or a C 1 -C 5 alkyl group; P 1 is a hydroxy protecting group; P 2 is an NH protecting group; and P 3 is a thiol protecting group.
  20. 一种化合物,其具有式VIII-1B所示结构或其立体异构体、其立体异构体混合物或药学上可接受的盐:A compound having the structure of Formula VIII-1B or a stereoisomer thereof, a mixture of stereoisomers thereof or a pharmaceutically acceptable salt thereof:
    Figure PCTCN2016109913-appb-100035
    Figure PCTCN2016109913-appb-100035
    其中:R3、R4各自独立地为氢原子或C1-C5烷基;P2为酰胺NH保护基团;P3为巯基保护基团。Wherein: R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group; P 2 is an amide NH protecting group; and P 3 is a fluorenyl protecting group.
  21. 一种化合物,其具有式IX-1B-B所示的结构或其立体异构体、立体异构体混合物或药学上可接受的盐: A compound having the structure of the formula IX-1B-B or a stereoisomer, a mixture of stereoisomers or a pharmaceutically acceptable salt thereof:
    Figure PCTCN2016109913-appb-100036
    Figure PCTCN2016109913-appb-100036
    其中,X为羟基或F、Cl、Br;R1为H或C1-C5烷基;R3、R4各自独立地为氢原子或C1-C5烷基;P1为羟基保护基团;P2为酰胺NH保护基团;P3为巯基保护基团。Wherein X is a hydroxyl group or F, Cl, Br; R 1 is H or a C 1 -C 5 alkyl group; R 3 and R 4 are each independently a hydrogen atom or a C 1 -C 5 alkyl group; and P 1 is a hydroxy group. a group; P 2 is an amide NH protecting group; and P 3 is a thiol protecting group.
  22. 一种化合物,其具有式I-2所示结构,或I-2所示结构化合物的前药或其立体异构体、立体异构体混合物或其药学上可接受的盐或溶剂合物:A compound having a structure of the formula I-2, or a prodrug of a structural compound represented by the formula I-2, or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable salt or solvate thereof:
    Figure PCTCN2016109913-appb-100037
    Figure PCTCN2016109913-appb-100037
    其中,X为羟基、F、Cl或Br;Wherein X is a hydroxyl group, F, Cl or Br;
    R’为
    Figure PCTCN2016109913-appb-100038
    其中Linker B可以存在或不存在,G为用于治疗或辅助治疗肝病的药物或其衍生物。
    R' is
    Figure PCTCN2016109913-appb-100038
    Where Linker B may or may not be present, and G is a drug or a derivative thereof for treating or assisting in the treatment of liver diseases.
  23. 根据权利要求21所述的化合物,其具有式II-2所示结构,或式II-2所示结构的互变异构体、光学异构体、立体异构体、立体异构体混合物、前药、药学上可接受的盐或溶剂合物:The compound according to claim 21, which has a structure represented by formula II-2, or a tautomer, an optical isomer, a stereoisomer, a mixture of stereoisomers, or a structure of the formula Prodrug, pharmaceutically acceptable salt or solvate:
    Figure PCTCN2016109913-appb-100039
    Figure PCTCN2016109913-appb-100039
    其中,X为羟基、F、Cl或Br;Wherein X is a hydroxyl group, F, Cl or Br;
    R’为
    Figure PCTCN2016109913-appb-100040
    其中Linker B为
    Figure PCTCN2016109913-appb-100041
    Figure PCTCN2016109913-appb-100042
    或不存在,G来自于用于治疗或辅助治疗肝病的药物或其衍生物;R1,R2为H或C1-C5烷基。
    R' is
    Figure PCTCN2016109913-appb-100040
    Where Linker B is
    Figure PCTCN2016109913-appb-100041
    Figure PCTCN2016109913-appb-100042
    Or absent, G is derived from a drug or a derivative thereof for use in the treatment or adjuvant treatment of liver disease; R 1 , R 2 is H or a C 1 -C 5 alkyl group.
  24. 根据权利要求23所述的化合物,具有式III-2A所示结构,或其立体异构体、立体异构体混合物或其药学上可接受的盐或溶剂合物: A compound according to claim 23 having the structure of formula III-2A, or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable salt or solvate thereof:
    Figure PCTCN2016109913-appb-100043
    Figure PCTCN2016109913-appb-100043
    其中,X为羟基、F、Br或Cl;Wherein X is a hydroxyl group, F, Br or Cl;
    Linker B为
    Figure PCTCN2016109913-appb-100044
    或不存在;R1,R2分别为H或C1-C5烷基;R3,R4为氢原子或C1-C5烃基。
    Linker B is
    Figure PCTCN2016109913-appb-100044
    Or absent; R 1 , R 2 are each H or a C 1 -C 5 alkyl group; and R 3 , R 4 are a hydrogen atom or a C 1 -C 5 hydrocarbon group.
  25. 根据权利要求24所述的化合物,具有式III-2Aa或者式III-2Ab所示结构,或其立体异构体、立体异构体混合物或其药学上可接受的盐或溶剂合物:The compound according to claim 24, having the structure of the formula III-2Aa or the formula III-2Ab, or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable salt or solvate thereof:
    Figure PCTCN2016109913-appb-100045
    Figure PCTCN2016109913-appb-100045
    其中,X为羟基、F、Br或Cl;Wherein X is a hydroxyl group, F, Br or Cl;
    Linker B为
    Figure PCTCN2016109913-appb-100046
    或不存在;R1,R2分别为H或C1-C5烷基;R3,R4为氢原子或C1-C5烃基。
    Linker B is
    Figure PCTCN2016109913-appb-100046
    Or absent; R 1 , R 2 are each H or a C 1 -C 5 alkyl group; and R 3 , R 4 are a hydrogen atom or a C 1 -C 5 hydrocarbon group.
  26. 根据权利要求23所述的化合物,具有式III-2B所示结构,或其立体异构体、立体异构体混合物或其药学上可接受的盐或溶剂合物:A compound according to claim 23 having the structure of formula III-2B, or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable salt or solvate thereof:
    Figure PCTCN2016109913-appb-100047
    Figure PCTCN2016109913-appb-100047
    其中,X为羟基、F、Br或Cl;Wherein X is a hydroxyl group, F, Br or Cl;
    Linker B为
    Figure PCTCN2016109913-appb-100048
    或不存在;R1,R2分别为H或C1-C5烷基;R3,R4为氢原子或C1-C5烃基。
    Linker B is
    Figure PCTCN2016109913-appb-100048
    Or absent; R 1 , R 2 are each H or a C 1 -C 5 alkyl group; and R 3 , R 4 are a hydrogen atom or a C 1 -C 5 hydrocarbon group.
  27. 根据权利要求26所述的化合物,具有式III-2Ba或者式III-2Bb所示结构,或其立体异构体、立体异构体混合物或其药学上可接受的盐或溶剂合物: The compound according to claim 26, having the structure of the formula III-2Ba or the formula III-2Bb, or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable salt or solvate thereof:
    Figure PCTCN2016109913-appb-100049
    Figure PCTCN2016109913-appb-100049
    其中,X为羟基、F、Br或Cl;Wherein X is a hydroxyl group, F, Br or Cl;
    Linker B为
    Figure PCTCN2016109913-appb-100050
    或不存在;R1,R2分别为H或C1-C5烷基;R3,R4为氢原子或C1-C5烃基。
    Linker B is
    Figure PCTCN2016109913-appb-100050
    Or absent; R 1 , R 2 are each H or a C1-C5 alkyl group; and R 3 , R 4 are a hydrogen atom or a C1-C5 hydrocarbon group.
  28. 根据权利要求23所述的化合物,具有式III-2C所示结构,或其立体异构体、立体异构体混合物或其药学上可接受的盐或溶剂合物:A compound according to claim 23 having the structure of the formula III-2C, or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable salt or solvate thereof:
    Figure PCTCN2016109913-appb-100051
    Figure PCTCN2016109913-appb-100051
    其中,X为F或Cl;Wherein X is F or Cl;
    Linker B为
    Figure PCTCN2016109913-appb-100052
    R3,R4为C1-C5烃基。
    Linker B is
    Figure PCTCN2016109913-appb-100052
    R 3 and R 4 are a C1-C5 hydrocarbon group.
  29. 根据权利要求22所述的化合物,具有以下所示的结构,或其立体异构体、立体异构体混合物或其药学上可接受的盐或溶剂合物:The compound according to claim 22, which has the structure shown below, or a stereoisomer thereof, a mixture of stereoisomers or a pharmaceutically acceptable salt or solvate thereof:
    Figure PCTCN2016109913-appb-100053
    Figure PCTCN2016109913-appb-100053
    Figure PCTCN2016109913-appb-100054
    Figure PCTCN2016109913-appb-100054
    Figure PCTCN2016109913-appb-100055
    Figure PCTCN2016109913-appb-100055
    Figure PCTCN2016109913-appb-100056
    Figure PCTCN2016109913-appb-100056
  30. 一种包含权利要求1-10、22-29任一项所示化合物中的一种或多种的药物组合物。A pharmaceutical composition comprising one or more of the compounds of any one of claims 1-10, 22-29.
  31. 根据权利要求30所述的药物组合物,其中,所示式(I)所示结构或其互变异构体、光学异构体、立体异构体、立体异构体混合物、前药、药学上可接受的盐或溶剂合物的使用剂量为1~1000mg/天。The pharmaceutical composition according to claim 30, wherein the structure represented by the formula (I) or a tautomer, an optical isomer, a stereoisomer, a mixture of stereoisomers, a prodrug, and a pharmaceutically acceptable compound are shown. The acceptable salt or solvate is used in an amount of from 1 to 1000 mg/day.
  32. 一种权利要求1~10、22-29任一项所述化合物或权利要求30所述的组合物在制备作为治疗丙型肝炎药物中的用途。Use of a compound according to any one of claims 1 to 10, 22-29 or a composition according to claim 30 for the preparation of a medicament for the treatment of hepatitis C.
  33. 根据权利要求32所述的用途,其中,该化合物或组合物进一步与其他治疗肝病的药物联用。The use according to claim 32, wherein the compound or composition is further used in combination with other drugs for treating liver diseases.
  34. 根据权利要求31所述的用途,其中所述的药物联用中的药物为:干扰素、干扰素β、干扰素γ和干扰素ω;白介素,包括白介素10和白介素12;利巴韦林;干扰素α或聚乙二醇化干扰素α与利巴韦林或者左旋韦林联合使用;左旋韦林;蛋白酶抑制剂,包括NS3抑制剂,NS3/4A抑制剂;NS5A抑制剂;解旋酶抑制剂;聚合酶抑制剂,包括HCV RNA聚合酶和NS5B聚合酶抑制剂;胶霉毒素;IRES抑制剂;反义寡核苷酸;噻唑烷衍生物;N-苯甲酰苯胺,核酶;另一种核苷,核苷前药或核苷衍生物;1-氨基-烷基环己烷;抗氧化剂,包括维生素E;角鲨烯;金刚胺;胆汁酸;N-(膦酰基乙酰基)-L-天冬氨酸;苯二羧酰胺;聚腺苷酸;苯并咪唑;胸腺素;预防疫苗;免疫调节剂,一种IMPDH抑制剂;水飞蓟素;水飞蓟素-磷脂酰胆碱内涵体;和麦考酚酸酯。 The use according to claim 31, wherein the drug in combination is: interferon, interferon beta, interferon gamma, and interferon ω; interleukin, including interleukin 10 and interleukin 12; ribavirin; Interferon alpha or pegylated interferon alpha in combination with ribavirin or levovirin; levovirin; protease inhibitors, including NS3 inhibitors, NS3/4A inhibitors; NS5A inhibitors; helicase inhibition Polymerase inhibitors, including HCV RNA polymerase and NS5B polymerase inhibitor; colloidal toxin; IRES inhibitor; antisense oligonucleotide; thiazolidine derivative; n-benzidine, ribozyme; a nucleoside, a nucleoside prodrug or a nucleoside derivative; 1-amino-alkylcyclohexane; an antioxidant, including vitamin E; squalene; amantadine; bile acid; N-(phosphonoacetyl) -L-aspartic acid; phenyl dicarboxamide; polyadenylation; benzimidazole; thymosin; prophylactic vaccine; immunomodulator, an IMPDH inhibitor; silymarin; silymarin-phosphatidylcholine endosome; Mycophenolate mofetil.
PCT/CN2016/109913 2015-12-15 2016-12-14 Compound, preparation method therefor, pharmaceutical composition thereof and use thereof WO2017101785A1 (en)

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
CN201510932904.2A CN105348345A (en) 2015-12-15 2015-12-15 Prodrug containing tiopronin structure, preparation method of prodrug, pharmaceutical composition and application of pharmaceutical composition
CN201510932904.2 2015-12-15
CN201610289337.8 2016-05-05
CN201610289337.8A CN105777829B (en) 2016-05-05 2016-05-05 A kind of prodrug containing class nucleotide structure, preparation method, medical composition and its use
CN201611024004.9 2016-11-18
CN201611024004.9A CN106883279B (en) 2015-12-15 2016-11-18 A kind of prodrug, preparation method, medical composition and its use

Publications (1)

Publication Number Publication Date
WO2017101785A1 true WO2017101785A1 (en) 2017-06-22

Family

ID=59055839

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2016/109913 WO2017101785A1 (en) 2015-12-15 2016-12-14 Compound, preparation method therefor, pharmaceutical composition thereof and use thereof

Country Status (1)

Country Link
WO (1) WO2017101785A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210238577A1 (en) * 2020-02-04 2021-08-05 Microsoft Technology Licensing, Llc Electrochemically-cleavable linkers

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104470939A (en) * 2012-05-22 2015-03-25 埃迪尼克斯医药公司 D-amino acid compounds for liver disease
CN105294795A (en) * 2014-11-20 2016-02-03 南京曼杰生物科技有限公司 Novel nucleoside phosphoramidate derivatives and use thereof
CN105348345A (en) * 2015-12-15 2016-02-24 杭州和正医药有限公司 Prodrug containing tiopronin structure, preparation method of prodrug, pharmaceutical composition and application of pharmaceutical composition
CN105777829A (en) * 2016-05-05 2016-07-20 杭州和正医药有限公司 Prodrug with nucleoside-analog-structure, and preparation method, medicinal composition and application thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104470939A (en) * 2012-05-22 2015-03-25 埃迪尼克斯医药公司 D-amino acid compounds for liver disease
CN105294795A (en) * 2014-11-20 2016-02-03 南京曼杰生物科技有限公司 Novel nucleoside phosphoramidate derivatives and use thereof
CN105348345A (en) * 2015-12-15 2016-02-24 杭州和正医药有限公司 Prodrug containing tiopronin structure, preparation method of prodrug, pharmaceutical composition and application of pharmaceutical composition
CN105777829A (en) * 2016-05-05 2016-07-20 杭州和正医药有限公司 Prodrug with nucleoside-analog-structure, and preparation method, medicinal composition and application thereof

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210238577A1 (en) * 2020-02-04 2021-08-05 Microsoft Technology Licensing, Llc Electrochemically-cleavable linkers

Similar Documents

Publication Publication Date Title
US10280175B2 (en) Hepatitis B antiviral agents
CN108546277B (en) Novel nucleoside phosphoramidate compounds and uses thereof
TWI519515B (en) Hepatitis b antiviral agents
US10301255B2 (en) Hepatitis B antiviral agents
US8178531B2 (en) Antiviral agents
JP5406943B2 (en) Cyclosporine analogs for preventing or treating hepatitis C infection
TWI391403B (en) New cyclic peptide compounds
CN106883279B (en) A kind of prodrug, preparation method, medical composition and its use
CA3029688A1 (en) Oxadiazepinone derivatives and their use in the treatment of hepatitis b infections
WO2016161268A1 (en) Hepatitis b antviral agents
CA2928003A1 (en) Azepane derivatives and methods of treating hepatitis b infections
US20090060874A1 (en) Bicyclic pyrrolidine derivatives
JP2012514657A (en) Phosphoramidate derivatives of guanosine nucleoside compounds for the treatment of viral infections
WO2015101183A1 (en) Uracil nucleotide analogs, preparation methods therefor and uses thereof
KR20160045136A (en) Cyclosporin analogues for preventing or treating hepatitis c
WO2017101785A1 (en) Compound, preparation method therefor, pharmaceutical composition thereof and use thereof
US10640515B2 (en) Hepatitis C virus inhibitor and uses thereof
CN106967141B (en) Nucleoside phosphoramidate compounds and pharmaceutical compositions and uses thereof
US11008361B2 (en) Liver-specific delivery-based anti-hepatitis C prodrug nucleoside cyclo-phosphate compound and uses thereof
WO2021031997A1 (en) Dihydropyrimidine derivative and use thereof
WO2023230472A1 (en) Inhibitors of molluscum contagiosum infection and methods using the same
TW202106677A (en) Dicarbamate inhibitors of ns5a for the treatment of hepatitis c virus infections
WO2017076194A1 (en) Compounds with inhibitory activity against hcv, preparation method and use thereof
NZ724493B2 (en) Hepatitis B Antiviral Agents

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: 16874842

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: 16874842

Country of ref document: EP

Kind code of ref document: A1