CN108602842A - Heterocyclic compound and its preparation method and application - Google Patents

Heterocyclic compound and its preparation method and application Download PDF

Info

Publication number
CN108602842A
CN108602842A CN201780005344.3A CN201780005344A CN108602842A CN 108602842 A CN108602842 A CN 108602842A CN 201780005344 A CN201780005344 A CN 201780005344A CN 108602842 A CN108602842 A CN 108602842A
Authority
CN
China
Prior art keywords
alkyl
compound
group
ester
pharmaceutically acceptable
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN201780005344.3A
Other languages
Chinese (zh)
Other versions
CN108602842B (en
Inventor
蔡家强
宋帅
邓汉文
曾宏
宋宏梅
唐祖建
刘瑶
田强
黄海涛
王利春
王晶翼
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Original Assignee
Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Sichuan Kelun Biotech Biopharmaceutical Co Ltd filed Critical Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Publication of CN108602842A publication Critical patent/CN108602842A/en
Application granted granted Critical
Publication of CN108602842B publication Critical patent/CN108602842B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/683Diesters of a phosphorus acid with two hydroxy compounds, e.g. phosphatidylinositols
    • A61K31/685Diesters of a phosphorus acid with two hydroxy compounds, e.g. phosphatidylinositols one of the hydroxy compounds having nitrogen atoms, e.g. phosphatidylserine, lecithin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings
    • 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
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

The present invention relates to the heterocyclic compound with antiviral activity, the pharmaceutical composition comprising it, preparation method and its purposes in preventing or treating the viral disease for including but not limited to viral hepatitis type A, virus B hepatitis, viral hepatitis type C, influenza, bleb and acquired immunodeficiency syndrome (AIDS).

Description

Heterocyclic compound and its preparation method and application Invention field
The present invention relates to the heterocyclic compound with antiviral activity, the pharmaceutical composition comprising it, preparation method and its preventing or treating the purposes in the viral disease for including but is not limited to viral hepatitis type A, virus B hepatitis, viral hepatitis type C, influenza, bleb and acquired immunodeficiency syndrome (AIDS).
Background of invention
Virus is made of a kind of nucleic acid molecules (DNA or RNA) and protein or is only made of (such as prion) protein.Virus can cause a variety of communicable diseases, and common disease caused by virus includes but is not limited to viral hepatitis type A, virus B hepatitis, viral hepatitis type C, influenza, bleb and acquired immunodeficiency syndrome (AIDS).
The antiviral drugs of clinical use is played a role by inhibiting viral attachment, shelling, viral gene duplication, maturation or release, or the immune system by influencing host at present, mainly includes that reverse transcriptase inhibitor and capsid protein assemble inhibitor etc..
Hepatitis type B virus (HBV) is a kind of common addicted to liver property DNA virus pathogen.This viroid can cause the diseases such as oxyhepatitis, chronic hepatitis, liver fibrosis, cirrhosis and liver cancer.
Virus B hepatitis can be used following nucleoside analog and be treated:
Wherein, tenofovir (Tenofovir) is nucleoside analog reverse transcriptase inhibitor, to human immunodeficiency virus (HIV) and hepatitis type B virus (HBV) infect it is significant in efficacy (Antivir Ther, 2004,9,57-65;J Viral Hepat,2000,7,161-165).Its action principle is to terminate the synthesis of DNA chain after the DNA of HBV by being integrated into natural substrate desoxyadenossine -5 '-triphosphoric acid competitiveness, to inhibit reverse transcriptase activity.But tenofovir is hardly absorbed by gastrointestinal tract.And its esterified prodrug tenofovir dipivoxil fumarate (TDF) and amidation prodrug tenofovir Chinese mugwort draw phenol amine fumarate (TAF) water-soluble good (Nucleosides Nucleotides Nucleic Acids.2001,20,1085-1090;Antimicrob Agents Chemother.2005,49,1898-1906;Antimicrob Agents Chemother.2015,59,3563-3569;Journal of Hepatology.2015,62,533-540).
In addition, virus B hepatitis can also use non-nucleoside to be treated like object.Deres et al. research discovery heteroaryl Dihydropyrimidines (Bay41-4109) can be by inhibiting viral capsid proteins assembly to prevent HBV virus replication (Science, 2003,299,893-896).Its specific mechanism of action is Dihydropyrimidines induction core protein mistake assembly, to form unstable capsid protein, accelerates the degradation (Biochem.Pharmacol., 2003,66,2273-2279) of core protein.The heteroaryl Dihydropyrimidines HAP1 (Proc.Natl.Acad.Sci. of Zlotnick et al. discovery, 2005,102,8138-8143) and Guangdong Dongyang Guang Pharmaceutical Co., Ltd report heteroaryl Dihydropyrimidines (GLS4) (Antimicrob.Agents Chemother., 2013,57,5344-5354;WO2015078391, US2016206616) also there is Anti-HBV effect.
Although above-mentioned two classes compound shows antiviral activity to some extent, their activity can't reach satisfactory degree.Therefore, it finds more effectively and the smaller drug of toxic side effect has urgent demand and important meaning.
Summary of the invention
The present invention provides a kind of heterocyclic antiviral compounds, can play antivirus action by the inhibiting effect assembled to reverse transcriptase and/or capsid protein.Particularly, preferred compounds of the invention can show the inhibiting effect assembled to reverse transcriptase and capsid protein simultaneously, to obtain superior antiviral effect under smaller dosage.In addition, the compound of the present invention also has physically better chemical property (such as solubility, physically and/or chemically stability), improved pharmacokinetic profile (such as improved bioavilability, suitable half-life period and acting duration), improved safety (lower toxicity and/or less side effect), the superior property such as be less also easy to produce drug resistance.
One aspect of the present invention provides compound or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorphic Object, solvate, metabolin or prodrug, wherein the compound has the structure of formula (I):
Wherein:
B is selected from:
A is selected from optionally by one or more RbSubstituted C1-6Alkylidene, C2-6Alkenylene, C2-6Alkynylene, the alkylidene, alkenylene or alkynylene are optionally interrupted by one or more-O- ,-NR- or-S-;
Or A is selected from following groups:
WhereinIt indicates singly-bound or double bond, and is connect at 1 position with B, is connect at 2 positions with phosphorus atoms (P);
X, Y and Z are each independently selected from CH at each occurrence2, O, S and NR;
RaAnd RbIt is each independently selected from halogen ,-OH ,-CN ,-NO at each occurrence2、-N(R)2、-N3、C1-6Alkyl and C3-6Naphthenic base;
R2Selected from H, C1-12Alkyl, C3-6Naphthenic base, C6-14Aryl, 5-14 unit's heteroaryl, C6-20Aralkyl ,-C1-6Alkylidene-COOH ,-C1-6Alkylidene-C (=O) O-C1-6Alkyl ,-C1-6Alkylidene-OC (=O)-C1-6Alkyl ,-C1-6Alkylidene-OC (=O) O-C1-6Alkyl and-((CH2)iO)m-(CH2)q-O-C1-6Alkyl, above-mentioned group are respectively optionally selected from halogen ,-OH ,-CN ,-NO by one or more2、-N(R)2、C1-6Alkyl, halogenated C1-6Alkyl, C1-6Alkyl sulfenyl and C3-6The substituent group of naphthenic base replaces;
I and q is each independently 1,2,3,4,5 or 6 at each occurrence;
M is the arbitrary integer in 0-50, preferably 0-20, particularly preferred 0-6;
R1It is selected from:
3 to 14 yuan of azepine ring systems are indicated, optionally in addition containing independently selected from N, O, C=O, S, S=O and S (=O)21,2 or 3 ring members;
Ar1And Ar2It is each independently selected from C6-14Aryl and 5-14 unit's heteroaryl are optionally selected from halogen ,-OH ,-CN ,-NO by one or more2、-N(R)2、C1-6Alkyl, halogenated C1-6Alkyl, C1-6Alkyl sulfenyl and C3-6The substituent group of naphthenic base replaces;
L is not present or selected from-O- ,-S- and-NR-;
R3And R4It is each independently selected from H, C1-4Alkyl and C3-6Naphthenic base;
R5At each occurrence with the connection of the rest part of singly-bound or double bond and molecule;
R5And R6It is each independently selected from halogen ,-OH ,-COOH ,-CN ,-NO at each occurrence2、-N(R)2、C1-6Alkyl, halogenated C1-6Alkyl ,-W-C1-6Alkyl ,-C1-6Alkylidene-W-R ,-W-C1-6Alkylidene-W '-R ,-W-C2-6Alkenyl ,-C2-6Alkenylene-W-R ,-W-C2-6Alkenylene-W '-R and C3-6Naphthenic base, wherein the alkylidene and alkenylene are optionally further by one or more intervals W;
W and W ' is each independently selected from O, C (=O), C (=O) O, NR, S, S=O and S (=O) at each occurrence2
R is each independently selected from H, C at each occurrence1-6Alkyl and C3-6Naphthenic base;
N is each independently 0,1,2,3,4 or 5 at each occurrence, condition be n be not more than on corresponding group can substituted position number;And
When n is greater than 1, each RaCan be identical or different, each RbCan be identical or different, each R5Can be identical or different, each R6It can be identical or different.
Another aspect of the present invention provide pharmaceutical composition, it includes prevention or therapeutically effective amount the compound of the present invention or its pharmaceutically may be used Salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or the prodrug of receiving and one or more pharmaceutically acceptable carriers.
Another aspect of the present invention provides the method for preparation pharmaceutical composition, and the method includes combining the compound of the present invention or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug with one or more pharmaceutically acceptable carriers.
Another aspect of the present invention provides the compound of the present invention or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug or pharmaceutical composition of the invention and is preparing the purposes in the drug for preventing or treating viral disease.
Another aspect of the present invention provides the compound of the present invention or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug or pharmaceutical composition of the invention, and it is used to prevent or treat viral diseases.
The method that another aspect of the present invention provides prevention or treatment viral disease, the method includes to needing its individual that a effective amount of the compound of the present invention or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug or pharmaceutical composition of the invention is administered.
Above-mentioned viral disease includes but is not limited to viral hepatitis type A, virus B hepatitis, viral hepatitis type C, influenza, bleb and acquired immunodeficiency syndrome (AIDS).
Another aspect of the present invention provides the method for preparation the compound of the present invention, the described method comprises the following steps:
Or
Wherein:
R12、R13And R14It is each independently selected from F, Cl, Br, I ,-NHR, hydroxyl, trifluoromethanesulfonic acid ester group, p-methyl benzenesulfonic acid ester group and boric acid ester group;
PG is selected from tertbutyloxycarbonyl, benzyloxycarbonyl group, 9-fluorenylmethyloxycarbonyl, allyloxycarbonyl, trimethylsilyl ethoxycarbonyl, methoxycarbonyl group, carbethoxyl group, p-toluenesulfonyl, ortho-nitrophenyl sulfonyl, p-nitrophenyl sulfonyl, formoxyl, acetyl group, trifluoroacetyl group, propiono, pivaloyl group, phenyl, benzoyl, trityl, benzyl, 2,4- dimethoxy-benzyl and to methoxy-benzyl;
Remaining each group is as defined above;
The first step carries out in non-protonic solvent or under condition of no solvent in the presence of organic base or inorganic base and/or condensation reagent (particularly preferred DCC, DIC, EDC, BOP, PyAOP or PyBOP);
Second step carries out under conditions of being suitable for removing PG group;
Third step carries out in the presence of organic base or inorganic base in non-protonic solvent;And
4th step preferably carries out in the presence of organic base or inorganic base and/or condensation reagent (particularly preferred DCC, DIC, EDC, BOP, PyAOP or PyBOP) in non-protonic solvent.
Another aspect of the present invention provides the method for preparation the compound of the present invention, the described method comprises the following steps:
Wherein:
R12Selected from F, Cl, Br, I ,-NHR, hydroxyl, trifluoromethanesulfonic acid ester group, p-methyl benzenesulfonic acid ester group and boric acid ester group;
LG is leaving group, is preferably pentafluorophenyl group and p-nitrophenyl;
Remaining each group is as defined above;
The first step carries out in non-protonic solvent in the presence of organic base or inorganic base;
Second step carries out in non-protonic solvent in the presence of organic base or inorganic base;And
Third step carries out in non-protonic solvent in the presence of organic base or inorganic base.
Definition
Unless hereinafter defined otherwise, the meaning intention of all technical terms and scientific terms used herein is generally understood identical with those skilled in the art.Refer to that technology intention used herein refers to the technology being generally understood in the art, the replacement of variation or equivalence techniques including those technologies that will be apparent to those skilled in the art.While it is believed that following term is for those skilled in the art it is well understood that but still illustrating defined below preferably to explain the present invention.
The terms "include", "comprise", " having ", " containing " or " being related to " and its other variant forms herein are the (inclusive) or open of inclusive, and are not excluded for other unlisted elements or method and step.
As used herein, term " alkylidene " indicates saturated divalent hydrocarbon radical, preferably indicates the saturated divalent hydrocarbon radical with 1,2,3,4,5 or 6 carbon atom, such as methylene, ethylidene, propylidene or butylidene.
As used herein, term " alkenylene " indicates the bivalent hydrocarbon radical comprising one or more double bonds, preferably has 2,3,4,5 or 6 carbon atoms, such as ethenylidene, allylidene or acrol.When the compound of the present invention contains alkenylene, the compound can be in the form of pure E (heteropleural (entgegen)), the presence of pure Z (ipsilateral (zusammen)) form or its any mixture form.
As used herein, term " alkynylene " indicates the bivalent hydrocarbon radical comprising one or more three keys, preferably has 2,3,4,5 or 6 carbon atoms, such as ethynylene or sub- propinyl.
As used herein, term " alkyl " is defined as linear chain or branched chain saturated aliphatic hydrocarbons.In some embodiments, alkyl has 1 to 12 carbon atom, such as 1 to 6 carbon atom.For example, as used herein, term " C1-6Alkyl " refers to the linear chain or branched chain saturated aliphatic hydrocarbons (such as methyl, ethyl, n-propyl, isopropyl, normal-butyl, isobutyl group, sec-butyl, tert-butyl, n-pentyl or n-hexyl) with 1 to 6 carbon atom, optionally replaces (group is referred to as " halogenated alkyl " at this time) (such as CF by the substituent group such as halogen that 1 or multiple (such as 1 to 3) is suitble to3、C2F5、CHF2、CH2F、CH2CF3、CH2Cl or-CH2CH2CF3Deng).Term " C1-4Alkyl " refers to the linear chain or branched chain saturated aliphatic hydrocarbons (i.e. methyl, ethyl, n-propyl, isopropyl, normal-butyl, isobutyl group, sec-butyl or tert-butyl) with 1 to 4 carbon atom.
As used herein, term " naphthenic base " refers to saturated or unsaturated non-aromatic monocyclic or polycyclic (such as bicyclic) hydrocarbon ring (such as monocycle, such as cyclopropyl, cyclobutyl, cyclopenta, cyclohexyl, suberyl, cyclooctyl, cyclononyl, or it is bicyclic, including loop coil, condensed or bridging system (bicyclic [1.1.1] amyl, bicyclic [2.2.1] heptyl, bicyclic [3.2.1] octyl or bicyclic [5.2.0] nonyl, decahydronaphthalene naphthalene etc.)), optionally replaced by the substituent group that 1 or multiple (such as 1 to 3) is suitble to.The naphthenic base has 3 to 15 carbon atoms.For example, term " C3-6Naphthenic base " refers to saturated or unsaturated non-aromatic monocyclic or polycyclic (such as bicyclic) hydrocarbon ring (such as cyclopropyl, cyclobutyl, cyclopenta or cyclohexyl) with 3 to 6 ring carbons; it is optionally replaced by the substituent group that 1 or multiple (such as 1 to 3) is suitble to, such as methyl substituted cyclopropyl.
As used herein, term " aryl " refers to full carbon monocycle or fused rings polycyclic aromatic group with conjugated pi electron system.For example, as used herein, term " C6-14Aryl " means the aromatic group containing 6 to 14 carbon atoms, such as phenyl or naphthyl.Aryl is optional Substituent group (such as halogen ,-OH ,-the CN ,-NO that ground is suitble to by 1 or multiple (such as 1 to 3)2、C1-6Alkyl etc.) replace.
As used herein, term " aralkyl " indicates the alkyl being substituted with aryl, wherein the aryl and the alkyl are as defined herein.In general, the aryl can have 6-14 carbon atom, and the alkyl can have 1-6 carbon atom.Exemplary aralkyl includes but is not limited to benzyl, phenylethyl, phenyl propyl, phenyl butyl.
As used herein, term " heteroaryl " refers to monocyclic, bicyclic or tricyclic aromatics ring system, it has 5,6,8,9,10,11,12,13 or 14 annular atoms, especially 1 or 2 or 3 or 4 or 5 or 6 or 9 or 10 carbon atom, and it includes the hetero atoms that at least one can be identical or different (hetero atom is such as oxygen, nitrogen or sulphur), it also, at each occurrence can be in addition benzo-fused.Particularly, heteroaryl is selected from thienyl, furyl, pyrrole radicals, oxazolyl, thiazolyl, imidazole radicals, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazoles base, triazolyl, thiadiazolyl group etc. and their benzo derivative;Or pyridyl group, pyridazinyl, pyrimidine radicals, pyrazinyl, triazine radical etc. and their benzo derivative.
As used herein, term " halogenated " or " halogen " group definition be include F, Cl, Br or I.
As used herein, term " alkyl sulfenyl " refers to the alkyl as defined above that parent molecular moiety is connected to by sulphur atom.C1-6The representative example of alkyl sulfenyl includes but is not limited to methyl mercapto, ethylmercapto group, tertiary butylthio and own sulfenyl.
As used herein, term " azepine ring system " refers to saturated or unsaturated monocycle or bicyclic radicals, it has 2,3,4,5,6,7,8,9,10,11,12 or 13 carbon atoms and at least one nitrogen-atoms in ring, is also selected from N, O, C=O, S, S=O and S (=O) optionally including one or more (such as one, two, three or four)2Ring members;The azepine ring system is connected by the rest part of nitrogen-atoms and molecule.Particularly, 3 to 14 yuan of azepine ring systems are the cyclic group in ring with 3-14 carbon atom and hetero atom (wherein at least one is nitrogen-atoms) comprising but be not limited to '-aziridino, azetidinyl, pyrrole radicals, pyrrolidinyl, pyrrolinyl, pyrrolidone-base, imidazole radicals, imidazolidinyl, imidazolinyl, pyrazolyl, pyrazolinyl, piperidyl, morpholinyl, thiomorpholine base, piperazinyl, indyl, indoline base etc..
Term " substitution " refers to that one or more (such as one, two, the three or four) hydrogen on specified atom is replaced from the selection of pointed group, and condition is the normal atom valency being less than specified atom in the current situation and the substitution forms stable compound.The combination of substituent group and/or variable is only just allowed when this combination forms stable compound.
If substituent group is described as " being optionally substituted ", substituent group can (1) it is unsubstituted or (2) be substituted.If the carbon of substituent group is described as being optionally substituted one or more substitutions in base list, the optional substituent group that one or more hydrogen (to the degree of existing any hydrogen) on carbon can be independently selected individually and/or together is substituted.If the nitrogen of substituent group is described as being optionally substituted one or more substitutions in base list, the optional substituent group that one or more hydrogen (to the degree of existing any hydrogen) on nitrogen can be respectively independently selected is substituted.
If substituent group is described as one group of " independently selected from ", each substituent group is selected independently of another one.Therefore, each substituent group can be identical or different with another (other) substituent group.
As used herein, term " one or more " means 1 under reasonable terms or more than 1, such as 2,3,4,5 or 10.
Unless indicated, otherwise as used herein, the tie point of substituent group may be from any suitable location of substituent group.
When the key of substituent group is shown as pass through the key for connecting two atoms in ring, then such substituent group can be bonded to any ring member nitrogen atoms in the substitutive ring.
The invention also includes the compounds of all pharmaceutically acceptable isotope labellings, it is identical as the compound of the present invention, in addition to one or more atoms are by with same atoms ordinal number but atomic mass or mass number are different from the atom of dominant in nature atomic mass or mass number and substitute.Be suitble to include isotope in the compound of the present invention example include but is not limited to hydrogen isotope (such as2H、3H);Carbon isotope (such as11C、13C and14C);Chlorine isotope (such as36Cl);Fluorine isotope (such as18F);Iodine isotope (such as123I and125I);Nitrogen isotope (such as13N and15N);Oxygen isotope (such as15O、17O and18O);Phosphorus isotope (such as32P);And sulphur isotope (such as35S).The compound of the present invention (such as mix radioisotopic those) of certain isotope labellings can be used in drug and/or substrate tissue distribution research (such as analysis).Radioactive isotope tritium is (i.e.3H) and carbon-14 (i.e.14C) it is particularly useful for the purpose because being easily incorporate into and being easy detection.With Positron emitting isotopes (such as11C、18F、15O and13N) carrying out replacing can be used to examine substrate receptor occupancy in positron emission tomography art (PET) research.The compound of the present invention being isotopically labeled can be by preparing with the reagent that those of is described in accompanying route and/or embodiment and preparation the non-marked used before similar method is replaced by using the reagent appropriate that is isotopically labeled.Pharmaceutically acceptable solvate of the invention include wherein recrystallisation solvent those of can be replaced by isotope, for example, D2O, acetone-d6Or DMSO-d6
Term " stereoisomer " indicates the isomers formed due at least one asymmetric center.In the compound with one or more (such as one, two, three or four) asymmetric centers, racemic mixture, single enantiomter, non-enantiomer mixture and individual diastereoisomer can produce.Specific individual molecules can also exist with geometric isomer (cis/trans).Similarly, the mixture (commonly referred to as tautomer) of the different form of the structure that the compound of the present invention can be in Fast-Balance with two or more exists.The representative example of tautomer includes ketone-enol tautomers, phenol -one tautomer, nitroso-oxime tautomer, imine-enamine tautomers etc..Exist for example, dihydro-pyrimidin group can be balanced in the solution with following tautomeric form:It is appreciated that scope of the present application covers all such or mixtures thereof isomers with arbitrary proportion (such as 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%).
Usable solid line herein, real wedge shapeOr empty wedge shapeDescribe the carbon-carbon bond of the compound of the present invention.It is bonded to the key of asymmetric carbon atom to describe using solid line and is intended to show include all possible stereoisomer (for example, specific enantiomter, racemic mixture etc.) at the carbon atom.Using real or imaginary wedge shape with describe be bonded to asymmetric carbon atom key be intended to show to exist shown in stereoisomer.When being present in racemic mixture, using real and imaginary wedge shape to define relative stereochemistry, rather than absolute stereochemistry.Unless otherwise specified, the compound of the present invention is intended to exist in the form of stereoisomer (it includes cis- and transisomer, optical isomer (such as R and S enantiomter), diastereoisomer, geometric isomer, rotational isomer, conformer, atropisomer and its mixture).The compound of the present invention can express the isomerism of more than one types, and be made of its mixture (such as racemic mixture and diastereoisomer to).
The present invention covers all possible crystal form or polymorph of the compound of the present invention, can be single polycrystalline type thing or the mixture of the arbitrary proportion of more than one polymorph.
It is also understood that certain compounds of the invention can exist for treating in a free form, or where appropriate, exist in the form of its pharmaceutically acceptable derivates.In the present invention, pharmaceutically acceptable derivates include but is not limited to, pharmaceutically acceptable salt, ester, solvate, metabolin or prodrug to after needing its patient to be administered, can directly or indirectly provide them to the compound of the present invention or its metabolin or residue.Therefore, when referenced herein " the compound of the present invention ", the above-mentioned various derivative forms for covering compound are also intended to.
The pharmaceutically acceptable salt of the compound of the present invention includes its acid-addition salts and base addition salts.
Suitable acid-addition salts are formed by the acid of formation pharmaceutically acceptable salt.Example includes aspartate, bicarbonate/carbonate, fumarate, gluceptate, gluconate, glucuronate, hexafluorophosphate, hydrobromate/bromide, hydriodate/iodide, naphthoate (naphthylate), nicotinate, nitrate, Orotate, palmitate and other similar salt.
Suitable base addition salts are formed by the alkali of formation pharmaceutically acceptable salt.Example includes aluminium salt, arginine salt, choline salt, lysine salt, magnesium salts, meglumine salt, amino butanetriol salt and other similar salt.
The summary of suitable salt referring to Stahl and Wermuth " Handbook of Pharmaceutical Salts:Properties, Selection, and Use " (Wiley-VCH, 2002).The method of pharmaceutically acceptable salt for the preparation of the compounds of the present invention is well known by persons skilled in the art.
As used herein, term " ester " means the ester derived from each general formula compound in the application comprising physiologically hydrolyzable ester (can hydrolyze in physiological conditions to discharge the compound of the present invention in the form of free acid or alcohol).The compound of the present invention itself is also possible to ester.
The compound of the present invention can exist in the form of solvate (preferably hydrate), and wherein the compound of the present invention includes the polar solvent of the structural element as the compound lattice, especially such as water, methanol or ethyl alcohol.The amount of polar solvent especially water can exist with stoichiometric ratio or non-stoichiometric.
It within the scope of the invention further include the metabolin of the compound of the present invention, i.e., the substance formed in body when the compound of the present invention is administered.Such product can be generated by the oxidation for the compound being for example administered, reduction, hydrolysis, amidation, desamidization, esterification, degreasing, enzymatic hydrolysis etc..Therefore, the present invention includes the metabolin of the compound of the present invention, including as making the compound of the present invention and mammalian animal be enough to generate compound made from the method for the time of its metabolite.
The present invention further comprises the prodrug of the compound of the present invention within its scope, it is expected active the compound of the present invention for that itself can have smaller pharmacological activity or certain derivatives of the compound of the present invention without pharmacological activity that can be converted to have for example, by hydrolytic rupture when being administered in body or thereon.Usually such prodrug can be the functional group derivant of the compound, be easy to be converted to desired therapeutical active compound in vivo.The other information used about prodrug can be found in " Pro-drugs as Novel Delivery Systems "; volume 14; ACS Symposium Series (T.Higuchi and V.Stella) and " Bioreversible Carriers in Drug Design; " Pergamon Press, 1987 (E.B.Roche is edited, American Pharmaceutical Association).Prodrug of the invention can be for example by using certain parts known to those skilled in the art as " preceding-part (pro-moiety) (such as " Design of Prodrugs "; described in H.Bundgaard (Elsevier, 1985)) " to substitute appropriate functional group present in the compound of the present invention to prepare.
Present invention also contemplates that the compound of the present invention containing protecting group.Prepare the compound of the present invention it is any during, protection may be necessary and/or desired in any sensitive group in relation on molecule or reactive group, and the form of the chemoproection of the compound of the present invention is consequently formed.This can realize by conventional protecting group, for example, in Protective Groups in Organic Chemistry, ed.J.F.W.McOmie, Plenum Press, 1973;And T.W.Greene&P.G.M.Wuts, Protective Groups in Organic Synthesis, John Wiley&Sons, protecting group those of described in 1991, these bibliography are by quoting addition herein.Using methods known in the art, protecting group can be removed in follow-up phase appropriate.
Term " about " refers in ± 10% range of the numerical value, in preferably ± 5% range, in more preferable ± 2% range.
Detailed description of the Invention
Compound and preparation method thereof
In one embodiment, the present invention provides compound or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug, wherein the compound has the structure of formula (I):
Wherein:
B is selected from:
A is selected from optionally by one or more RbSubstituted C1-6Alkylidene, C2-6Alkenylene, C2-6Alkynylene, the alkylidene, alkenylene or alkynylene are optionally interrupted by one or more-O- ,-NR- or-S-;
Or A is selected from following groups:
WhereinIt indicates singly-bound or double bond, and is connect at 1 position with B, is connect at 2 positions with phosphorus atoms (P);
X, Y and Z are each independently selected from CH at each occurrence2, O, S and NR;
RaAnd RbIt is each independently selected from halogen ,-OH ,-CN ,-NO at each occurrence2、-N(R)2、-N3、C1-6Alkyl and C3-6Naphthenic base;
R2Selected from H, C1-12Alkyl, C3-6Naphthenic base, C6-14Aryl, 5-14 unit's heteroaryl, C6-20Aralkyl ,-C1-6Alkylidene-COOH ,-C1-6Alkylidene-C (=O) O-C1-6Alkyl ,-C1-6Alkylidene-OC (=O)-C1-6Alkyl ,-C1-6Alkylidene-OC (=O) O-C1-6Alkyl and-((CH2)iO)m-(CH2)q-O-C1-6Alkyl, above-mentioned group are respectively optionally selected from halogen ,-OH ,-CN ,-NO by one or more2、-N(R)2、C1-6Alkyl, halogenated C1-6Alkyl, C1-6Alkyl sulfenyl and C3-6The substituent group of naphthenic base replaces;
I and q is each independently 1,2,3,4,5 or 6 at each occurrence;
M is the arbitrary integer in 0-50, preferably 0-20, particularly preferred 0-6;
R1It is selected from:
3 to 14 yuan of azepine ring systems are indicated, optionally in addition containing independently selected from N, O, C=O, S, S=O and S (=O)2 1,2 or 3 ring members;
Ar1And Ar2It is each independently selected from C6-14Aryl and 5-14 unit's heteroaryl are optionally selected from halogen ,-OH ,-CN ,-NO by one or more2、-N(R)2、C1-6Alkyl, halogenated C1-6Alkyl, C1-6Alkyl sulfenyl and C3-6The substituent group of naphthenic base replaces;
L is not present or selected from-O- ,-S- and-NR-;
R3And R4It is each independently selected from H, C1-4Alkyl and C3-6Naphthenic base;
R5At each occurrence with the connection of the rest part of singly-bound or double bond and molecule;
R5And R6It is each independently selected from halogen ,-OH ,-COOH ,-CN ,-NO at each occurrence2、-N(R)2、C1-6Alkyl, halogenated C1-6Alkyl ,-W-C1-6Alkyl ,-C1-6Alkylidene-W-R ,-W-C1-6Alkylidene-W '-R ,-W-C2-6Alkenyl ,-C2-6Alkenylene-W-R ,-W-C2-6Alkenylene-W '-R and C3-6Naphthenic base, wherein the alkylidene and alkenylene are optionally further by one or more intervals W;
W and W ' is each independently selected from O, C (=O), C (=O) O, NR, S, S=O and S (=O) at each occurrence2
R is each independently selected from H, C at each occurrence1-6Alkyl and C3-6Naphthenic base;
N is each independently 0,1,2,3,4 or 5 at each occurrence, condition be n be not more than on corresponding group can substituted position number, and
When n is greater than 1, each RaCan be identical or different, each RbCan be identical or different, each R5Can be identical or different, each R6It can be identical or different.
In preferred embodiments, X, Y and Z are each independently selected from CH at each occurrence2, O and NH.
In particularly preferred embodiments, X, Y and Z are each independently selected from O and NH at each occurrence.
In preferred embodiments, RaAnd RbIt is each independently selected from halogen ,-OH ,-N (R) at each occurrence2、-N3And C1-6Alkyl.
In particularly preferred embodiments, RaAnd RbIt is each independently selected from F ,-OH, amino, cyclopropylamino and methyl at each occurrence.
In preferred embodiments, B is selected from:
In particularly preferred embodiments, B is
In preferred embodiments, A is selected from:
It wherein connect at 1 position with B, is connect at 2 positions with phosphorus atoms (P).
In a more preferred embodiment, A isIt wherein connect at 1 position with B, is connect at 2 positions with phosphorus atoms (P).
In particularly preferred embodiments, A isIt wherein connect at 1 position with B, is connect at 2 positions with phosphorus atoms (P).
In preferred embodiments,Indicate 3 yuan, 4 yuan, 5- or 6-membered azepine ring system.
In particularly preferred embodiments,Selected from azetidinyl, pyrrolidinyl, piperidyl, morpholinyl and piperazinyl.
In preferred embodiments, R1It is selected from:
In preferred embodiments, Ar1And Ar2It is each independently selected from imidazole radicals, oxazolyl, thiazolyl, pyridyl group, pyrimidine radicals, piperazinyl and phenyl, respectively optionally by one or more identical or different halogens, C1-6Alkyl, halogenated C1-6Alkyl and C3-6Naphthenic base replaces.
In a more preferred embodiment, Ar1And Ar2It is each independently selected from imidazole radicals, oxazolyl, thiazolyl, pyridyl group, pyrimidine radicals, piperazinyl and phenyl, respectively optionally by one or more identical or different halogens, C1-6Alkyl and C3-6Naphthenic base replaces.
In preferred embodiments, Ar1It is selected from:
Wherein RcIt is each independently selected from F, Cl, Br, I, C at each occurrence1-6Alkyl, halogenated C1-6Alkyl and C3-6Naphthenic base;
Preferably RcIt is each independently selected from F, Cl, Br, I, C at each occurrence1-6Alkyl and C3-6Naphthenic base;
Ar1It preferably is selected from:
Ar1More preferably certainly:
In preferred embodiments, Ar2It is selected from:
The above group is optionally selected from halogen, C by one or more1-6Alkyl, halogenated C1-6Alkyl and C3-6The group of naphthenic base replaces.
In a more preferred embodiment, Ar2It is selected from:
The above group is optionally selected from halogen, C by one or more1-6Alkyl and C3-6The group of naphthenic base replaces.
In preferred embodiments, L is-O-.
In preferred embodiments, R3And R4It is each independently selected from methyl, ethyl, n-propyl and isopropyl.
In particularly preferred embodiments, R4For H.
In preferred embodiments, R5And R6It is each independently selected from F, Cl, Br, I ,-OH ,-COOH, methyl, ethyl ,-CH at each occurrence2OH、-OCH3、-CH2COOH、-CH2CH2COOH、-CH2CH2CH2COOH ,-CH=CHCOOH ,-OCH2COOH、-SCH2COOH、-N(CH3)CH2COOH、-CH2OCH2COOH、-CH2SCH2COOH and -CH2N(CH3)CH2COOH。
In preferred embodiments, R2Selected from H, C1-12Alkyl, C3-6Naphthenic base, C6-14Aryl, 5-14 unit's heteroaryl, C6-20Aralkyl ,-C1-6Alkylidene-C (=O) O-C1-6Alkyl ,-C1-6Alkylidene-OC (=O)-C1-6Alkyl ,-C1-6Alkylidene-OC (=O) O-C1-6Alkyl and-((CH2)iO)m-(CH2)q-O-C1-6Alkyl, above-mentioned group are respectively optionally selected from halogen ,-OH ,-CN ,-NO by one or more2、-N(R)2、C1-6Alkyl, halogenated C1-6Alkyl, C1-6Alkyl sulfenyl and C3-6The substituent group of naphthenic base replaces;
In a more preferred embodiment, R2It is selected from: C1-6Alkyl ,-((CH2)iO)m-(CH2)q-O-C1-6Alkyl,
Wherein:
I and q is each independently 1,2,3,4,5 or 6 at each occurrence;
M is the arbitrary integer in 0-50, preferably 0-20, particularly preferred 0-6;
R7、R8And R9It is each independently selected from H, C1-10Alkyl, C3-6Naphthenic base, C6-20Aryl and C7-20Aralkyl, the alkyl, naphthenic base, aryl and aralkyl are respectively optionally selected from halogen ,-OH ,-CN and-NO by one or more2Substituent group replace;
Or R7And R8C is collectively formed in the carbon atom connected together with it3-6Naphthenic base;
R10And R11It is each independently selected from H, halogen ,-OH ,-CN ,-NO2、C1-10Alkyl and C3-6Naphthenic base.
In a more preferred embodiment, R2It is selected from: C1-6Alkyl ,-((CH2)iO)m-(CH2)q-O-C1-4Alkyl,
Wherein:
I and q is each independently 1,2,3 or 4 at each occurrence;
M is 1,2,3,4,5 or 6;
R7And R9It is each independently selected from H or C1-4Alkyl, the alkyl are optionally selected from halogen ,-OH ,-CN and-NO by one or more2Substituent group replace;
R10And R11It is each independently selected from H, halogen ,-OH ,-CN ,-NO2Or C1-4Alkyl.
In particularly preferred embodiments, R2Selected from H, methyl, ethyl, propyl, butyl, amyl ,-((CH2)2O)6-(CH2)2-O-CH3
In particularly preferred embodiments, R2Selected from methyl, ethyl, propyl, butyl ,-((CH2)2O)6-(CH2)2-O-CH3
In preferred embodiments, 0,1,2 or 3 n.
In preferred embodiments, the present invention provides compound or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug, wherein the compound has the structure of formula (II) or formula (II) -1:
In preferred embodiments, the present invention provides compound or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug, wherein the compound has the structure of any following formula:
The present invention, which covers, carries out the resulting compound of any combination to each embodiment.
In preferred embodiments, the present invention provides compound or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug, wherein the compound is selected from:
One embodiment of the invention provides the method for preparation the compound of the present invention, the described method comprises the following steps:
Or
Wherein:
R12、R13And R14It is each independently selected from F, Cl, Br, I ,-NHR, hydroxyl, trifluoromethanesulfonic acid ester group, p-methyl benzenesulfonic acid ester group and boric acid ester group;
PG is selected from tertbutyloxycarbonyl, benzyloxycarbonyl group, 9-fluorenylmethyloxycarbonyl, allyloxycarbonyl, trimethylsilyl ethoxycarbonyl, methoxycarbonyl group, carbethoxyl group, p-toluenesulfonyl, ortho-nitrophenyl sulfonyl, p-nitrophenyl sulfonyl, formoxyl, acetyl group, trifluoroacetyl group, propiono, pivaloyl group, phenyl, benzoyl, trityl, benzyl, 2,4- dimethoxy-benzyl and to methoxy-benzyl;
Remaining each group is as defined above;
The first step carries out in non-protonic solvent or under condition of no solvent in the presence of organic base or inorganic base and/or condensation reagent;
Second step carries out under conditions of being suitable for removing PG group;
Third step carries out in the presence of organic base or inorganic base in non-protonic solvent;And
4th step preferably carries out in the presence of organic base or inorganic base and/or condensation reagent in non-protonic solvent.
Another aspect of the present invention provides the method for preparation the compound of the present invention, the described method comprises the following steps:
Wherein:
R12Selected from F, Cl, Br, I ,-NHR, hydroxyl, trifluoromethanesulfonic acid ester group, p-methyl benzenesulfonic acid ester group and boric acid ester group;
LG is leaving group, is preferably pentafluorophenyl group and p-nitrophenyl;
Remaining each group is as defined above;
The first step carries out in non-protonic solvent in the presence of organic base or inorganic base;
Second step carries out in non-protonic solvent in the presence of organic base or inorganic base;And
Third step carries out in non-protonic solvent in the presence of organic base or inorganic base.
In preferred embodiments, R12、R13And R14It is each independently selected from F, Cl, Br, I, NH2Or hydroxyl.
In preferred embodiments, PG can be tertbutyloxycarbonyl, suitable for remove tertbutyloxycarbonyl condition can for for example under sour (such as trifluoroacetic acid) catalysis in solvent (such as methylene chloride), at 0 DEG C to carrying out at room temperature.
In preferred embodiments, LG-OH can be Pentafluorophenol, p-nitrophenol.
In preferred embodiments, can the non-protonic solvent used in the compound of the present invention preparation method include but is not limited to tetrahydrofuran, methylene chloride, 1,2- dichloroethanes, acetonitrile, N, dinethylformamide (DMF), N-Methyl pyrrolidone, 1,3-Dimethyl-2-imidazolidinone (DMI), dimethyl sulfoxide (DMSO) and hexamethylphosphoramide (HMPA).
In preferred embodiments, can the organic base used in the compound of the present invention preparation method include but is not limited to sodium tert-butoxide, tert-butyl magnesium chloride, triethylamine, n,N-diisopropylethylamine (DIPEA), pyridine or 4-dimethylaminopyridine (DMAP);Can the inorganic base used in the compound of the present invention preparation method include but is not limited to NaH, NaOH, Na2CO3Or K2CO3
In preferred embodiments, can the condensation reagent used in the compound of the present invention preparation method include but is not limited to dicyclohexylcarbodiimide (DCC), N, N- diisopropylcarbodiimide (DIC), 1- (3- dimethylamino-propyl) -3- ethyl carbodiimide (EDC), three (dimethylamino) phosphorus hexafluorophosphate (BOP) of benzotriazole -1- base oxygroup, (3H-1, 2, 3- triazol [4, 5-b] pyridine -3- oxygroup) three -1- pyrrolidinyl phosphorus hexafluorophosphates (PyAOP) and 1H- benzotriazole -1- base oxygen tripyrrole alkyl phosphorus hexafluorophosphate (PyBOP).
Pharmaceutical composition and treatment method
In another kind of embodiment, the present invention provides pharmaceutical composition, and it includes prevention or the compound of the present invention or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug and one or more pharmaceutically acceptable carriers of therapeutically effective amount.In another kind of embodiment, described pharmaceutical composition also may include one or more other therapeutic agents, such as other therapeutic agents for preventing or treating viral disease.
In another kind of embodiment, the present invention provides the method for preparation pharmaceutical composition, and the method includes combining the compound of the present invention or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug with one or more pharmaceutically acceptable carriers.
In another kind of embodiment, the present invention provides the compound of the present invention or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug or pharmaceutical composition of the invention and is preparing the purposes in the drug for preventing or treating viral disease.
In another kind of embodiment, the present invention provides the compound of the present invention or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug or pharmaceutical composition of the invention, and it is used to prevent or treat viral diseases.
In another kind of embodiment, the method that the present invention provides prevention or treatment viral disease, the method includes to needing its individual that a effective amount of the compound of the present invention or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug or pharmaceutical composition of the invention is administered.
Preferred compounds of the invention plays antivirus action by the inhibiting effect to reverse transcriptase and/or capsid protein assembly.Therefore, the compound of the present invention can be used for treating is related to any virus of reverse transcriptase and/or capsid protein assembly during influencing host comprising but it is not limited to hepatitis A virus (HAV), hepatitis type B virus (HBV), Hepatitis C Virus (HCV), influenza virus, herpesviral (HSV) and human immunodeficiency virus (HIV).
Therefore, the viral disease that usable the compound of the present invention prevents and treats includes but is not limited to viral hepatitis type A, virus B hepatitis, viral hepatitis type C, influenza, bleb and acquired immunodeficiency syndrome (AIDS), and the related symptoms or disease (such as inflammation, liver fibrosis, cirrhosis and liver cancer etc.) that are caused by above-mentioned disease.
" pharmaceutically acceptable carrier " refers to the diluent being administered together with therapeutic agent, adjuvant, excipient or medium in the present invention, and its tissue that the mankind and/or other animals are adapted for contact in the range of reasonable medical judgment without excessive toxicity, stimulation, allergic reaction or with reasonable benefit/risk than corresponding other problems or complication.
Workable pharmaceutically acceptable carrier includes but is not limited to sterile liquid, such as water and oil, oil including those petroleum, animal, plant or synthesis source, such as peanut oil, soybean oil, mineral oil, sesame oil etc. in pharmaceutical composition of the invention.When described pharmaceutical composition is administered intravenously (IV, water is exemplary carrier.Physiological saline and glucose and glycerine water solution can also be used as liquid-carrier, be especially used for injection.Suitable drug excipient includes starch, glucose, lactose, sucrose, gelatin, maltose, chalk, silica gel, odium stearate, glycerin monostearate, talcum, sodium chloride, skimmed milk power, glycerol, propylene glycol, water, ethyl alcohol etc..The composition can also optionally include a small amount of wetting agent, emulsifier or pH buffer.Oral preparation may include standard vector, such as mannitol, lactose, starch, magnesium stearate, saccharin sodium, cellulose, the magnesium carbonate of pharmaceutical grade.The example of suitable pharmaceutically acceptable carrier is as described in Remington ' s Pharmaceutical Sciences (1990).
Pharmaceutical composition of the invention can be acted on systematically and/or locally be acted on.For this purpose, the approach administration that they can be suitble to, for example, by injection (as in intravenous, intra-arterial, subcutaneous, peritonaeum, intramuscular injection, including instil) or percutaneous dosing;Or oral, buccal, intranasal, it is transmucosal, local, in the form of eye-drops preparations or pass through inhalation.
For these administration routes, pharmaceutical composition of the invention is administered in the dosage form that can be suitble to.
The dosage form includes but is not limited to tablet, capsule, pastille, hard candy agent, powder, spray, cream, ointment, suppository, gelling agent, paste, lotion, ointment, aqueous suspension, injectable solutions, elixir, syrup.
" effective quantity " refers to as used herein, the term be administered after can alleviate to a certain extent treated illness one or more symptoms compound amount.
Dosage regimen be can adjust to provide optimal required response.For example, single bolus can be administered, several divided doses can be administered at any time, or dosage can be proportionally reduced or increased as indicated in the urgent need for the treatment of condition.It it should be noted that dose value can change with the type and seriousness for the patient's condition to be mitigated, and may include single or multiple dosage.It further understands, for any particular individual, specific dosage regimen should adjust at any time according to the professional judgement of individual need and administration composition or the personnel for the administration for supervising composition.
The amount of the compound of the present invention being administered can depend on individual treated, the seriousness of illness or the patient's condition, the rate of administration, the disposition of compound and the judgement of prescriber.In general, effective dose is in per kg body weight per day about 0.0001 to about 50mg, for example, about 0.01 to about 10mg/kg/ days (single or divided doses).For the people of 70kg, this can be added up to about 0.007mg/ to about 3500mg/, for example, about 0.7mg/ to about 700mg/.In some cases, it can be not higher than the dosage level of the lower limit of aforementioned range enough, and in other cases, larger dose can be still used in the case where not causing any harmful side effect, condition is that the larger dose is divided into several smaller doses to be administered throughout the day first.
Content or dosage of the compound of the present invention in pharmaceutical composition can be about 0.01mg to about 1000mg, be compatibly 0.1-500mg, preferably 0.5-300mg, more preferable 1-150mg, particularly preferred 1-50mg, such as 1.5mg, 2mg, 4mg, 10mg, 25mg etc..
Unless otherwise stated, otherwise as used herein, term " treating (treating) " means to reverse, mitigates, inhibits illness applied by such term or the patient's condition perhaps one or more symptoms of the progress of one or more symptoms of such illness or the patient's condition or the such illness of prevention or the patient's condition or such illness or the patient's condition.
" individual " includes people or non-human animal as used herein.Exemplary individual human includes individual human (referred to as patient) or normal individual with disease (such as disease as described herein)." non-human animal " includes all vertebrates in the present invention, such as nonmammalian (such as birds, amphibian, reptile) and mammal, such as non-human primates, domestic animal and/or domesticated animal (such as sheep, dog, cat, milk cow, pig etc.).
In another kind of embodiment, pharmaceutical composition of the invention can also include one or more other therapeutic agents or prophylactic comprising but be not limited to Lamivudine, Sebivo, Entecavir, Aldoforwe ester, tenofovir, tenofovir dipivoxil fumarate and tenofovir Chinese mugwort and draw phenol amine fumarate.
Embodiment
The present invention is further described with reference to embodiments, but these embodiments are provided and are not intended to limit the scope of the present invention.
Unless otherwise stated, using commercially available anhydrous solvent and HPLC grades of solvents without further purifying.
It is recorded at ambient temperature with Bruker instrument (400MHz)1H NMR spectra is internal standard using TMS.Chemical shift (δ) is provided as unit of ppm, and coupling constant (J) is provided with hertz (Hz) for unit.1H NMR spectra peak to split the abbreviation of point tuple as follows: s (unimodal), d (bimodal), t (triplet), q (quartet), m (multiplet), br (broad peak).
LC-MS is combined Aglient 6120Quadrupole type mass spectrograph using 1200 liquid chromatograph of Aglient, detects at 214nm and 254nm.Preparative liquid chromatography prepares liquid phase instrument using 1260 type of SHIMADZU CBM-20A and Aglient, 5 μM of 19 × 150mm of C18OBD prepare column, Detection wavelength 214nm, and mobile phase A is water, Mobile phase B is acetonitrile (0.5 ‰ formic acid of addition), and according to the form below carries out linear gradient elution:
Time (min) A% B%
0 90 10
15 40 60
30 10 90
Embodiment one (4R) -6- ((4- (4- ((((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl) ((2- methylbenzyl) oxygroup) phosphono) oxygroup) phenyl) piperidin-1-yl) methyl) -4- (the chloro- 4- fluorophenyl of 2-) -2- (thiazol-2-yl) -1,4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (compound 1)
Step 1: the synthesis of 4- (4- hydroxy phenyl) piperidines -1- carboxylic acid tert-butyl ester (compound 1-2)
At room temperature; by 4- (4- (benzyloxy) phenyl) -3; 6- dihydropyridine -1 (2H)-carboxylic acid tert-butyl ester (compound 1-1) (3.0g; it 8.2mmol) is added in tetrahydrofuran (10mL) and methanol (20mL); after compound dissolution completely; 10%Pd/C (300mg) is added under nitrogen protection; it finishes; hydrogen displacement; 18h is reacted at room temperature; filtering, post-processes to obtain title compound (2.0g) for filtrate.ESI-MS(m/z):222.1[M-55+H]+
Step 2: the synthesis of 4- (4- ((((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl) (hydroxyl) phosphono) oxygroup) phenyl) piperidines -1- carboxylic acid tert-butyl ester (compound 1-3)
At room temperature, by (R) -9- (2- phosphate methoxy propyl)-adenine (3.1g, 10.8mmol), compound 1-2 (2.0g, it 7.2mmol) is dissolved in N-Methyl pyrrolidone (20mL), it is warming up to 85 DEG C, it is added dropwise triethylamine (879mg, 8.9mmol), is continuously heating to after finishing 100 DEG C, N-Methyl pyrrolidone (10mL) solution of dicyclohexylcarbodiimide (4.8g, 23.1mmol) is added dropwise, drop finishes, and reacts overnight at 100 DEG C, post-processes to obtain title compound (1.0g).ESI-MS(m/z):547.2[M+H]+
Step 3: ((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl) -4- (piperidin-4-yl)-phenyl-phosplate (compound 1-4) synthesis
At room temperature, by compound 1-3 (200mg, it 0.4mmol) is added in methylene chloride (5.0mL), trifluoroacetic acid (0.2mL) is added after compound dissolution completely, it finishes, so that it is reacted 2.5h at room temperature, post-process title compound trifluoroacetate (200mg).ESI-MS(m/z):447.2[M+H]+
Step 4: the synthesis of (4R) -6- ((4- (4- ((((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl) (hydroxyl) phosphono) oxygroup) phenyl) piperidin-1-yl) methyl) -4- (the chloro- 4- fluorophenyl of 2-) -2- (thiazol-2-yl) -1,4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (compound 1-5)
At room temperature, by the trifluoroacetate (163mg of compound 1-4,0.4mmol) it is dissolved in 1, in 2- dichloroethanes (4mL), sequentially add (R) -6- (bromomethyl) -4- (the chloro- 4- fluorophenyl of 2-) -2- (thiazol-2-yl) -1,4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (204mg, 0.4mmol) and N, N- diisopropylethylamine (0.1mL), it finishes, reaction overnight, post-processes to obtain title compound (130mg) at room temperature.ESI-MS(m/z):824.2[M+H]+
Step 5: the synthesis of (4R) -6- ((4- (4- ((((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl) ((2- methylbenzyl) oxygroup) phosphono) oxygroup) phenyl) piperidin-1-yl) methyl) -4- (the chloro- 4- fluorophenyl of 2-) -2- (thiazol-2-yl) -1,4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (compound 1)
At room temperature, by compound 1-5 (30mg, 0.04mmol), o-methyl-benzene methanol (9.0mg, 0.07mmol), 1H- benzotriazole -1- base oxygen tripyrrole alkyl phosphorus hexafluorophosphate (PyBOP) (38mg, 0.07mmol) it is dissolved in N, in dinethylformamide (3mL), after dissolution completely, N is added dropwise, N- diisopropylethylamine (19mg, 0.14mmol), finishes, 4h is reacted at room temperature, obtains title compound (9mg) after purification.
Its structural characterization is as follows:
1H NMR(400MHz,DMSO-d6) δ 9.78 (s, 1H), 8.13 (s, 1H), 8.05-8.03 (m, 2H), 7.95 (d, J=3.13Hz, 1H), 7.44-7.40 (m, 2H), 7.30-7.14 (m, 9H), 7.03 (d, J=8.20Hz, 1H), 6.96 (d, J=8.12Hz, 1H), 6.06 (s, 1H), 5.15-5.08 (m, 2H), 4.28-4.16 (m, 2H), 4.11-3.87 (m, 7H), (3.06 d, J=10.39Hz, 1H), (2.88 d, J=10.29Hz, 1H), (2.60-2.57 m, 1H), 2.44-2. 39 (m, 1H), 2.32-2.24 (m, 4H), 1.88-1.67 (m, 4H), 1.09-1.04 (m, 6H) .ESI-MS (m/z): 928.2 [M+H]+
Embodiment two (4R) -6- ((4- (4- ((((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl) (((S) -1- ethyoxyl -1- oxo propyl- 2- yl) oxygroup) phosphono) oxygroup) phenyl) piperidin-1-yl) methyl) -4- (the chloro- 4- fluorophenyl of 2-) -2- (thiazol-2-yl) -1,4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (compound 2)
At room temperature, by compound 1-5 (30mg, 0.04mmol), Pfansteihl ethyl ester (8.0mg, 0.07mmol) and PyBOP (38mg, it 0.07mmol) is dissolved in n,N-Dimethylformamide (3mL), after dissolution completely, N is added dropwise, N- diisopropylethylamine (19mg, 0.14mmol), finishes, about 4h is reacted at room temperature, obtains title compound (5.0mg) after purification.
Its structural characterization is as follows:
1H NMR(400MHz,DMSO-d6) δ 9.77 (s, 1H), 8.13 (s, 1H), 8.05 (s, 1H), 8.03 (d, J=23.26Hz, 1H), 7.95 (d, J=3.13Hz, 1H), 7.44-7.40 (m, 2H), 7.27-7.16 (m, 5H), 7.05-7.03 (m, 2H), 6.06 (s, 1H), 5.00-4.92 (m, 1H), 4.31-3.87 (m, 11H), (3.06 d, J=9.75Hz, 1H), (2.88 d, J=11.02Hz, 1H), 2.60-2.57 (m, 1H), 2.43-2.40 (m, 1H), 2.33-2.26 (m, 1H) , 1.88-1.68 (m, 4H), 1.35 (dd, J=21.76Hz, 6.84Hz, 3H), 1.10 (d, J=6.22Hz, 3H), 1.05 (t, J=7.11Hz, 3H), 1.17 (dt, J=7.11Hz, 3.05Hz, 3H) .ESI-MS (m/z): 924.2 [M+H]+
Embodiment three (4R) -6- ((4- (4- ((((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl) (((R) -1- ethyoxyl -1- oxo propyl- 2- yl) oxygroup) phosphono) oxygroup) phenyl) piperidin-1-yl) methyl) -4- (the chloro- 4- fluorophenyl of 2-) -2- (thiazol-2-yl) -1,4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (compound 3)
At room temperature, by compound 1-5 (30mg, 0.04mmol), D-ALPHA-Hydroxypropionic acid ethyl ester (8.0mg, 0.07mmol) and PyBOP (38mg, it 0.07mmol) is dissolved in n,N-Dimethylformamide (3mL), after dissolution completely, N is added dropwise, N- diisopropylethylamine (19mg, 0.14mmol), finishes, 4h is reacted at room temperature, obtains title compound (6.0mg) after purification.
Its structural characterization is as follows:
1H NMR(400MHz,CDCl3) δ 9.80 (brs, 1H), 8.34 (s, 1H), 7.99-7.89 (m, 2H), 7.44 (s, 1H), 7.33 (s, 1H), 7.19 (d, J=8.15Hz, 2H), 7.14-7.11 (m, 1H), 7.01-6.92 (m, 3H), 6.22 (s, 1H), 5.64 (brs, 2H), 5.05-4.98 (m, 1H), 4.39-4.34 (m, 1H), 4.23-3.79 (m, 10H), (3.02 d, J=46.30Hz, 2H), 2.54-2.38 (m, 3H), 1.90-1.86 (m, 4H), 1.51 (dd, J=25.5 4Hz, 6.92Hz, 3H), 1.29-1.21 (m, 6H), 1.14 (t, J=7.10Hz, 3H) .ESI-MS (m/z): 924.2 [M+H]+
Example IV (4R) -6- ((4- (4- (((2; 5; 8; 11; 14; 17; seven oxa- docosane -22- base oxygroup of 20-) ((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl) phosphono) oxygroup) phenyl) piperidin-1-yl) methyl) -4- (the chloro- 4- fluorophenyl of 2-) -2- (thiazol-2-yl) -1,4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (compound 4)
At room temperature, by compound 1-5 (30mg, 0.04mmol), seven glycol monomethyl ether (24.0mg, 0.07mmol) and PyBOP (38mg, it 0.07mmol) is dissolved in n,N-Dimethylformamide (3mL), after dissolution completely, N is added dropwise, N- diisopropylethylamine (19mg, 0.14mmol), finishes, 4h is reacted at room temperature, obtains title compound (5.0mg) after purification.
Its structural characterization is as follows:
1H NMR(400MHz,CDCl3) δ 9.78 (brs, 1H), 8.33 (d, J=2.16Hz, 1H), 7.97 (d, J=13.35Hz, 1H), 7.90 (dd, J=5.33Hz, 3.17Hz, 1H), 7.44 (d, J=2.98Hz, 1H), 7.32 (dd, J=8.45Hz, 6.24Hz, 1H), 7.22-7.16 (m, 2H), 7.14-7.09 (m, 2H), 7.01 (d, J=7.95Hz, 1H), 6.91 (td, J=8.36Hz, 2.27Hz, 1H), 6.22 (s, 1H), 5.79 (brs, 2H), 4.36 (ddd, J=14.48Hz, 6 .43Hz, 2.82Hz, 1H), 4.30-4.21 (m, 1H), 4.17-4.12 (m, 1H), 4.06-3.98 (m, 4H), 3.88 (d, J=17.09Hz, 1H), 3.81-3.73 (m, 1H), 3.64-3.53 (m, 28H), 3.37 (s, 3H), 3.05 (d, J=10.76Hz, 1H), 2.92 (d, J=10.87Hz, 1H), 2.53-2.47 (m, 2H), 2.35 (t, J=8.41Hz, 1H), 1.89-1.77 (m, 4H), 1.24 (dd, J=6.02Hz, 4.16Hz, 3H), 1.13 (t, J=7.10Hz, 3H) .ESI-MS (m/z): 573.8 [M/2+H]+
Embodiment five (4R) -6- ((4- (4- ((((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl) (amoxy) phosphono) oxygroup) phenyl) piperidin-1-yl) methyl) -4- (the chloro- 4- fluorophenyl of 2-) -2- (thiazol-2-yl) -1,4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (compound 5)
At room temperature, by compound 1-5 (30mg, 0.04mmol), n-amyl alcohol (6.0mg, 0.07mmol) and PyBOP (38mg, it 0.07mmol) is dissolved in n,N-Dimethylformamide (3mL), after dissolution completely, N is added dropwise, N- diisopropylethylamine (19mg, 0.14mmol), finishes, 3.5h is reacted at room temperature, manages to obtain title compound (4.0mg) after purification.
Its structural characterization is as follows:
1H NMR(400MHz,DMSO-d6) δ 9.77 (s, 1H), 8.13 (s, 1H), 8.04-8.03 (m, 2H), 7.95 (d, J=3.08Hz, 1H), 7.44-7.40 (m, 2H), 7.27-7.16 (m, 5H), 7.07 (d, J=7.99Hz, 1H), 7.00 (d, J=8.27Hz, 1H), 6.06 (s, 1H), 4.30-4.16 (m, 2H), 4.03-3.91 (m, 9H), 3.06 (d, J=10.38Hz, 1H), 2.88 (d, J=11.44Hz, 1H), 2.60-2.58 (m, 1H), (2.44-2.39 m, 1H), 2.33-2. 26 (m, 1H), 1.87-1.67 (m, 4H), 1.53-1.43 (m, 2H), 1.23-1.18 (m, 4H), 1.11 (t, J=6.57Hz, 3H), 1.05 (t, J=7.11Hz, 3H) .ESI-MS (m/z): 894.3 [M+H]+
Embodiment six (4R) -6- ((4- (4- ((((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl) ((pivaloyl oxygroup) methoxyl group) phosphono) oxygroup) phenyl) piperidin-1-yl) methyl) -4- (the chloro- 4- fluorophenyl of 2-) -2- (thiazol-2-yl) -1,4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (compound 6)
At room temperature, by compound 1-5 (30mg, it 0.04mmol) is dissolved in N-Methyl pyrrolidone (2mL), after dissolution completely, sequentially adds N, N- diisopropylethylamine (9mg, 0.07mmol) with iodometyl pivalate (13mg, 0.06mmol), finish, 1.5h is reacted at room temperature, obtains title compound (5.0mg) after purification.
Its structural characterization is as follows:
1H NMR(400MHz,CDCl3)δ9.19(s,2H),8.38-8.34(m,1H),7.95-7.89(m,2H),7.44(m,1H),7.35-7.30(m,1H),7.23-7.19(m,2H),7.17-7.08(m,2H),7.03-6.90(m,3H),6.22(s,1H),5.75-5.59(m,2H),4.40-4.34(m,2H),4.11-3.98(m,5H),3.82-2.75(m,1H),3.48-3.35(m,2H),3.11-3.01(m,1H),2.94-2.84(m,1H),2.59-2.42(m,2H),1.98-1.84(m,4H),1.34-1.12(m,15H).ESI-MS(m/z):938.2[M+H]+
Embodiment seven (4R) -6- ((4- ((((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl) (((S) -1- ethyoxyl -1- oxo propyl- 2- yl) oxygroup) phosphono) oxygroup) -3; 3- difluoropiperdin -1- base) methyl) -4- (the chloro- 4- fluorophenyl of 2-) -2- (thiazol-2-yl) -1,4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (compound 27)
Step 1: the synthesis of 4- ((((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl) (hydroxyl) phosphono) oxygroup) -3,3- difluoropiperdin -1- carboxylic acid tert-butyl ester (compound 7-2)
At room temperature; by (R) -9- (2- phosphate methoxy propyl)-adenine (55mg; 0.19mmol) and 3; fluoro- 4- hydroxy piperidine -1- carboxylic acid tert-butyl ester (compound the 7-1) (30mg of 3- bis-; 0.13mmol) the dissolution in pyridine (2mL); dicyclohexylcarbodiimide (80mg, 0.39mmol) is added under nitrogen protection, is warming up to 80 DEG C of reactions overnight.Obtain title compound (10mg) after purification.ESI-MS(m/z):507.1[M+H]+
Step 2: the synthesis of ((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl)-(3,3- difluoropiperdin -4- base)-phosplate trifluoroacetate (compound 7-3)
At room temperature, compound 7-2 (10mg, 0.02mmol) is added in methylene chloride (1.0mL), dissolution is added completely into trifluoro second Sour (0.2mL), after addition, reacts at room temperature.Reaction solution evaporated under reduced pressure solvent is obtained into title compound (10mg), it is directly used in the case where not purifying and is reacted in next step.ESI-MS(m/z):407.1[M+H]+
Step 3: (4R) -6- ((4- ((((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl) (hydroxyl) phosphono) oxygroup) -3; 3- difluoropiperdin -1- base) methyl) -4- (the chloro- 4- fluorophenyl of 2-) -2- (thiazol-2-yl) -1,4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (compound 7-4) synthesis
At room temperature, by compound 7-3 (10mg, 0.025mmol) it is dissolved in 1, in 2- dichloroethanes (2mL), (R) -6- (bromomethyl) -4- (the chloro- 4- fluorophenyl of 2-) -2- (thiazol-2-yl)-Isosorbide-5-Nitrae-dihydro-pyrimidin -5- carboxylic acid, ethyl ester (14mg is sequentially added, 0.030mmol) and N, N- diisopropylethylamine (0.1mL), after addition, reaction is stayed overnight at room temperature.Obtain title compound (10mg) after purification.ESI-MS(m/z):784.1[M+H]+
Step 4: (4R) -6- ((4- ((((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl) (((S) -1- ethyoxyl -1- oxo propyl- 2- yl) oxygroup) phosphono) oxygroup) -3; 3- difluoropiperdin -1- base) methyl) -4- (the chloro- 4- fluorophenyl of 2-) -2- (thiazol-2-yl) -1,4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (compound 27) synthesis
At room temperature, by compound 7-4 (40mg, 0.05mmol), Pfansteihl ethyl ester (10mg, 0.1mmol) and PyBOP (52mg, 0.1mmol) it is dissolved in N, in dinethylformamide (3mL), after dissolution completely, N is added dropwise, N- diisopropylethylamine (26mg, 0.2mmol), after addition, reaction is stayed overnight at room temperature.Obtain title compound (10mg) after purification.
Its structural characterization is as follows:
1H NMR(400MHz,CDCl3) δ 9.46 (d, J=11.90Hz, 1H), 8.35 (t, J=2.16Hz, 1H), 8.07-7.98 (m, 1H), 7.85 (d, J=3.24,1H), 7.43 (d, J=2.87Hz, 1H), 7.31 (ddd, J=9.02,6.15,3.19Hz 1H), 7.13 (dd, J=8.60,2.57Hz, 1H), 6.93 (td, J=8.32,2.42Hz, 1H), 6.21 (t, J=2.29Hz, 1H), 5.91 (s, 2H), (5.02-4.90 m, 1H), 4.78-4.67 (m, 1H), 4.45-4.34 (m 1H), 4.27-3.90 (m, 9H), 3.74-3.66 (m, 1H), 3.02-2.63 (m, 3H), 2.25-1.99 (m, 3H), 1.55-1.44 (m, 3H), 1.31-1.23 (m, 6H), 1.13 (t, J=7.16,1.39Hz, 3H) .ESI-MS (m/z): 884.1 [M+H]+
Embodiment eight (4R) -6- ((4- ((((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl) (hydroxyl) phosphono) oxygroup) -3; 3- difluoropyrrolidin -1- base) methyl) -4- (the chloro- 4- fluorophenyl of 2-) -2- (thiazol-2-yl) -1,4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (compound 28)
Step 1: the synthesis of 4- ((((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl) (hydroxyl) phosphono) oxygroup) -3,3- difluoropyrrolidin -1- carboxylic acid tert-butyl ester (compound 8-2)
At room temperature; by tenofovir (compound 8-1) (966mg; 3.36mmol) and 3; the fluoro- 4- hydroxyl pyrrolidine -1- carboxylic acid tert-butyl ester (500mg of 3- bis-; it 2.24mmol) is dissolved in pyridine (6mL), under nitrogen protection, dicyclohexylcarbodiimide (1.39g is added; 6.72mmol), 80 DEG C of reactions are warming up to overnight.Obtain title compound (240mg) after purification.ESI-MS(m/z):493.1[M+H]+
Step 2: ((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl) -4,4- difluoropyrrolidin -3- base-phosplate trifluoroacetate (compound 8-3) synthesis
At room temperature, compound 8-2 (100mg, 0.2mmol) is added in methylene chloride (1.5mL), trifluoroacetic acid (0.5mL) is added after dissolving completely, finishes, reacts at room temperature.Evaporated under reduced pressure solvent obtains title compound (120mg), it is not purified to be directly used in and is reacted in next step.ESI-MS(m/z):393.1[M+H]+
Step 3: (4R) -6- ((4- ((((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl) (hydroxyl) phosphono) oxygroup) -3; 3- difluoropyrrolidin -1- base) methyl) -4- (the chloro- 4- fluorophenyl of 2-) -2- (thiazol-2-yl) -1,4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (compound 28) synthesis
At room temperature, by compound 8-3 (80mg, it 0.2mmol) is dissolved in methylene chloride (4mL), sequentially add (R) -6- (bromomethyl) -4- (the chloro- 4- fluorophenyl of 2-) -2- (thiazol-2-yl) -1,4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (100mg, 0.22mmol) and n,N-diisopropylethylamine (0.5mL), it finishes, room temperature reaction is overnight.Obtain title compound (80mg) after purification.
Its structural characterization is as follows:
1H NMR (400MHz, methanol-d4) δ 8.31 (d, J=1.6Hz, 1H), 8.18 (d, J=2.7Hz, 1H), 7.87 (dd, J=5.3,3.2Hz, 1H), 7.67 (dd, J=5.8,3.1Hz, 1H), 7.46-7.39 (m, 1H), 7.21 (dd, J=8.8,2.6Hz, 1H), 7.08-7.02 (m, 2H), 6.14 (d, J=2.0Hz, 1H), 4.87-4.75 (m, 1H), 4.38-4.32 (m, 1H), 4.24-4.00 (m, 5H), 3.88-3.84 (m, 1H), 3.77-3.68 (m, 1H), 3.40-3.36 (m, 1 ), H 3.29-2.95 (m, 2H), 2.78-2.76 (m, 1H), 1.13-1.07 (m, 6H) .ESI-MS (m/z): 770.1 [M+H]+
Embodiment nine (4R) -6- ((4- ((((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl) (((S) -1- ethyoxyl -1- oxo propyl- 2- yl) oxygroup) phosphono) oxygroup) -3; 3- difluoropyrrolidin -1- base) methyl) -4- (the chloro- 4- fluorophenyl of 2-) -2- (thiazol-2-yl) -1,4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (compound 29)
Title compound (20mg) is obtained with compound 28 instead of compound 7-4 using with similar operations described in seven step 4 of embodiment.ESI-MS(m/z):870.1[M+H]+
Embodiment ten (4R) -6- ((4- ((((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl) (((R) -1- ethyoxyl -1- oxo propyl- 2- yl) oxygroup) phosphono) oxygroup) -3; 3- difluoropyrrolidin -1- base) methyl) -4- (the chloro- 4- fluorophenyl of 2-) -2- (thiazol-2-yl) -1,4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (compound 30)
Title compound (10mg) is obtained with D-ALPHA-Hydroxypropionic acid ethyl ester instead of Pfansteihl ethyl ester using with similar operations described in embodiment nine.ESI-MS(m/z):870.1[M+H]+
11 2- of embodiment ((((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl) ((1- (((R) -6- (the chloro- 4- fluorophenyl of 2-) -5- (carbethoxyl group) -2- (thiazol-2-yl) -3; 6- dihydro-pyrimidin -4- base) methyl) -4,4- difluoropyrrolidin -3- base) oxygroup) phosphono) oxygroup) acetic acid (compound 31)
At room temperature, compound 28 (30mg, 0.04mmol) is dissolved in N-Methyl pyrrolidone (2mL), sequentially add bromoacetic acid (27mg, 0.20mmol) and potassium carbonate (16mg, 0.12mmol), it finishes, is warming up to 60 DEG C of reaction 2h.Filtrate is obtained title compound (5.0mg) after purification by reaction solution filtering.ESI-MS(m/z):828.0[M+H]+
Embodiment 12 (4R) -4- (the chloro- 4- fluorophenyl of 2-) -6- ((3; the fluoro- 4- (((((2S of 3- bis-; 3R; 5S) -3- hydroxyl -5- (5- methyl -2; 4- dioxo -3; 4- dihydro-pyrimidin -1 (2H)-yl) tetrahydrofuran -2- base) methoxyl group) (((S) -1- isopropyl oxygen -1- oxo propyl- 2- yl) amino) phosphono) oxygroup) pyrrolidin-1-yl) methyl) -2- (thiazol-2-yl) -1,4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (compound 32)
Step 1: (4R) -4- (the chloro- 4- fluorophenyl of 2-) -6- ((4- ((dichloro phosphono) oxygroup) -3; 3- difluoropyrrolidin -1- base) methyl) -2- (thiazol-2-yl) -1,4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (compound 12-2) synthesis
At room temperature; by (4R) -4- (the chloro- 4- fluorophenyl of 2-) -6- ((3; the fluoro- 4- hydroxyl pyrrolidine -1- base of 3- bis-) methyl) -2- (thiazol-2-yl) -1; 4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (compound 12-1) (50mg; it 0.1mmol) is dissolved in methylene chloride (2mL); -20 DEG C are cooled under nitrogen protection; it is added dropwise phosphorus oxychloride (0.2mL); it finishes; it is added dropwise triethylamine (0.2mL), reacts 30min at -20 DEG C.Reaction solution is directly spin-dried for, gained crude product does not purify to be directly used in react in next step.
Step 2: (4R) -4- (the chloro- 4- fluorophenyl of 2-) -6- ((3; the fluoro- 4- of 3- bis- (((((S) -1- isopropyl oxygen -1- oxo propyl- 2- yl) amino) (phenyl-pentafluoride oxygroup) phosphono) oxygroup) pyrrolidin-1-yl) methyl) -2- (thiazol-2-yl) -1,4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (compound 12-3) synthesis
At room temperature; by compound 12-2 (60mg; it 0.1mmol) is dissolved in methylene chloride (2mL); -20 DEG C are cooled under nitrogen protection; methylene chloride (1.0mL) solution of l-Alanine isopropyl ester hydrochloride (50mg, 0.3mmol) is added dropwise, finishes; methylene chloride (0.5mL) solution of triethylamine (0.2mL) is added dropwise, finishes and moves to room temperature reaction 1h.- 20 DEG C are cooled under nitrogen protection again; Pentafluorophenol (50mg is added dropwise; 0.3mmol) and methylene chloride (2.0mL) solution of triethylamine (0.1mL); it finishes; 1h is reacted at -20 DEG C, reaction solution is directly spin-dried for, is dissolved with tetrahydrofuran (4.0mL); insoluble matter is filtered off, filtrate is directly used in and is reacted in next step.
Step 3: (4R) -4- (the chloro- 4- fluorophenyl of 2-) -6- ((3; the fluoro- 4- (((((2S of 3- bis-; 3R; 5S) -3- hydroxyl -5- (5- methyl -2; 4- dioxo -3; 4- dihydro-pyrimidin -1 (2H)-yl) tetrahydrofuran -2- base) methoxyl group) (((S) -1- isopropyl oxygen -1- oxo propyl- 2- yl) amino) phosphono) oxygroup) pyrrolidin-1-yl) methyl) and -2- (thiazol-2-yl) -1,4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (compound 32) synthesis
At room temperature; by Sebivo (50mg; it 0.2mmol) is dissolved in tetrahydrofuran (2mL), -20 DEG C is cooled under nitrogen protection, tert-butyl magnesium chloride (0.4mL is added dropwise; 1M); it finishes and moves to room temperature reaction 1h, be cooled to -20 DEG C again under nitrogen protection, the resulting tetrahydrofuran solution of a dropping step two; it finishes, is reacted at room temperature overnight under nitrogen protection.Obtain title compound (2.0mg) after purification.ESI-MS(m/z):918.0[M+H]+
Embodiment 13 (4R) -6- ((4- ((((((R) -1- (6- amino -9H- purine -9- base) propyl- 2- yl) oxygroup) methyl) (((R) -1- ethyoxyl -1- oxo propyl- 2- yl) oxygroup) phosphono) oxygroup) -3; 3- difluoropiperdin -1- base) methyl) -4- (the chloro- 4- fluorophenyl of 2-) -2- (thiazol-2-yl) -1,4- dihydro-pyrimidin -5- carboxylic acid, ethyl ester (compound 33)
Title compound (15mg) is obtained with D-ALPHA-Hydroxypropionic acid ethyl ester instead of Pfansteihl ethyl ester using with similar operations described in seven step 4 of embodiment.ESI-MS(m/z):884.1[M+H]+
Other compounds in the application can refer to method synthesis similar in above-described embodiment.
Bioactivity detection
The compound of the present invention is tested to the inhibiting effect of hepatitis type B virus (HBV).In the cytotoxicity of virus-cell horizontal checkout the compound of the present invention and influence to viral (HBV) nucleic acid (DNA) levels of replication.
Test method
By the HepG2 2.2.15 cell inoculation of logarithmic growth phase in 96 orifice plates, cell concentration is 40/μ L.In 37 DEG C, 5%CO2It is cultivated 3 days in incubator;It is replaced before compound is added with new culture medium (200 hole μ L/).The mother liquid concentration of each untested compound is 200 μM.With 200 μM for maximum concentration, multiple and different concentration are diluted to DMSO, take 1 μ L untested compound to be placed in corresponding culture datum hole, the final test concentration of compound is 0.06,0.24,0.98,3.9,15.6,62.5,250,1000nM is (for calculating medium effective concentration (EC50))。
The mother liquor of each untested compound is diluted to 600 μM and 60 μM with DMSO, respectively takes 1 μ L to be added in corresponding culture datum hole, the final test concentration of compound is 3 μM and 0.3 μM (for calculating percent inhibition).
Blank control: 1 μ L DMSO is taken to be added in corresponding culture datum hole as control.
It is added after untested compound in 37 DEG C, 5%CO2It is co-cultured 10 days in incubator, replaces culture medium every three days, and add compound again.
In the 11st day, every hole was collected 150 μ L cell supernatants and is detected for qPCR.
The EC of tested compound50, and under 3 μM and 0.3 μM of concentration inhibiting rate result as shown in table 1, table 2.
Table 1
Compound EC50(nM)
2 59.43
3 54.65
27 52.1
29 5.18
30 8.21
32 26.95
33 53.1
From the data in table 1, it can be seen that tested compound shows stronger inhibitory activity, the EC of some compounds in the active testing for DNA (DNA) duplication for inhibiting hepatitis type B virus (HBV)50< 60nM, the EC of preferred compound (such as compound 29 and 30)50< 10nM shows that the compound of the present invention has very excellent inhibitory activity.
Table 2
Compound (3 μM) Inhibiting rate %
1 96.54
2 100.20
3 101.67
From the above results, it can be seen that tested compound under 3 μM of concentration, shows excellent inhibitory effect in the active testing for DNA (DNA) duplication for inhibiting hepatitis type B virus (HBV).The compound tested equally shows excellent inhibitory effect under 0.3 μM of concentration, shows that the compound of the present invention has very excellent inhibitory effect.
Cytotoxicity detection
Untested compound is diluted to 30mM with DMSO, using 30mM as maximum concentration, three times are diluted to multiple and different concentration, take the compound of 0.2 each concentration of μ L in 384 orifice plates, the HepG2 2.2.15 cell of 2000/50 μ L of every hole addition, final concentration of 150 μM of untested compound highest;1 μ L DMSO is added in corresponding aperture as control.
In 37 DEG C, 5%CO2It is co-cultured 4 days in incubator.
50 μ L CellTiter-Glo are added in every hole after 4 days, carry out read plate detection, are calculated and cause half cytotoxic concentration (CC50) value.
The CC of preferred compound (such as compound of embodiment 5)50Value is greater than 150 μM, this illustrates that its cytotoxicity is lower, and safety is higher.
In addition to those described herein, according to foregoing description, a variety of modifications of the invention can be obvious to those skilled in the art.Such modification is also intended to fall within the scope of the appended claims.Cited each bibliography (including all patents, patent application, journal of writings, books and any other disclosure) quotes addition herein with its entirety in the application.

Claims (17)

  1. Compound or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug, wherein the compound has the structure of formula (I):
    Wherein:
    B is selected from:
    A is selected from optionally by one or more RbSubstituted C1-6Alkylidene, C2-6Alkenylene, C2-6Alkynylene, the alkylidene, alkenylene or alkynylene are optionally interrupted by one or more-O- ,-NR- or-S-;
    Or A is selected from following groups:
    WhereinIt indicates singly-bound or double bond, and is connect at 1 position with B, is connect at 2 positions with phosphorus atoms (P);
    X, Y and Z are each independently selected from CH at each occurrence2, O, S and NR;
    RaAnd RbIt is each independently selected from halogen ,-OH ,-CN ,-NO at each occurrence2、-N(R)2、-N3、C1-6Alkyl and C3-6Naphthenic base;
    R2Selected from H, C1-12Alkyl, C3-6Naphthenic base, C6-14Aryl, 5-14 unit's heteroaryl, C6-20Aralkyl ,-C1-6Alkylidene-COOH ,-C1-6Alkylidene-C (=O) O-C1-6Alkyl ,-C1-6Alkylidene-OC (=O)-C1-6Alkyl ,-C1-6Alkylidene-OC (=O) O-C1-6Alkyl and-((CH2)iO)m-(CH2)q-O-C1-6Alkyl, above-mentioned group are respectively optionally selected from halogen ,-OH ,-CN ,-NO by one or more2、-N(R)2、C1-6Alkyl, halogenated C1-6Alkyl, C1-6Alkyl sulfenyl and C3-6The substituent group of naphthenic base replaces;
    I and q is each independently 1,2,3,4,5 or 6 at each occurrence;
    M is the arbitrary integer in 0-50, preferably 0-20, particularly preferred 0-6;
    R1It is selected from:
    3 to 14 yuan of azepine ring systems are indicated, optionally in addition containing independently selected from N, O, C=O, S, S=O and S (=O)21,2 or 3 ring members;
    Ar1And Ar2It is each independently selected from C6-14Aryl and 5-14 unit's heteroaryl are optionally selected from halogen ,-OH ,-CN ,-NO by one or more2、-N(R)2、C1-6Alkyl, halogenated C1-6Alkyl, C1-6Alkyl sulfenyl and C3-6The substituent group of naphthenic base replaces;
    L is not present or selected from-O- ,-S- and-NR-;
    R3And R4It is each independently selected from H, C1-4Alkyl and C3-6Naphthenic base;
    R5At each occurrence with the connection of the rest part of singly-bound or double bond and molecule;
    R5And R6It is each independently selected from halogen ,-OH ,-COOH ,-CN ,-NO at each occurrence2、-N(R)2、C1-6Alkyl, halogenated C1-6Alkyl ,-W-C1-6Alkyl ,-C1-6Alkylidene-W-R ,-W-C1-6Alkylidene-W '-R ,-W-C2-6Alkenyl ,-C2-6Alkenylene-W-R ,-W-C2-6Alkenylene-W '-R and C3-6Naphthenic base, wherein the alkylidene and alkenylene are optionally further by one or more intervals W;
    W and W ' is each independently selected from O, C (=O), C (=O) O, NR, S, S=O and S (=O) at each occurrence2
    R is each independently selected from H, C at each occurrence1-6Alkyl and C3-6Naphthenic base;
    N is each independently 0,1,2,3,4 or 5 at each occurrence, condition be n be not more than on corresponding group can substituted position number;And
    When n is greater than 1, each RaCan be identical or different, each RbCan be identical or different, each R5Can be identical or different, each R6It can be identical or different.
  2. The compound of claim 1 or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug, wherein R2Selected from H, C1-12Alkyl, C3-6Naphthenic base, C6-14Aryl, 5-14 unit's heteroaryl, C6-20Aralkyl ,-C1-6Alkylidene-C (=O) O-C1-6Alkyl ,-C1-6Alkylidene-OC (=O)-C1-6Alkyl ,-C1-6Alkylidene-OC (=O) O-C1-6Alkyl and-((CH2)iO)m-(CH2)q-O-C1-6Alkyl, above-mentioned group are respectively optionally selected from halogen ,-OH ,-CN ,-NO by one or more2、-N(R)2、C1-6Alkyl, halogenated C1-6Alkyl, C1-6Alkyl sulfenyl and C3-6The substituent group of naphthenic base replaces.
  3. The compound of claims 1 or 2 or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug, wherein B is selected from:
  4. The compound of any one of claim 1-3 or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug, wherein A is selected from:
    It wherein connect at 1 position with B, is connect at 2 positions with phosphorus atoms (P).
  5. The compound of any one of claim 1-4 or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug, wherein R1It is selected from:
  6. The compound of any one of claim 1-5 or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug, wherein Ar1It is selected from:
    Wherein RcIt is each independently selected from F, Cl, Br, I, C at each occurrence1-6Alkyl, halogenated C1-6Alkyl and C3-6Naphthenic base;
    Preferably, RcIt is each independently selected from F, Cl, Br, I, C at each occurrence1-6Alkyl and C3-6Naphthenic base;
    Ar1It preferably is selected from:
    Ar1Particularly preferably certainly:
  7. The compound of any one of claim 1-6 or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug, wherein Ar2It is selected from:
    The above group is optionally selected from halogen, C by one or more1-6Alkyl, halogenated C1-6Alkyl and C3-6The group of naphthenic base replaces;
    Preferably, Ar2It is selected from:
    The above group is optionally selected from halogen, C by one or more1-6Alkyl and C3-6The group of naphthenic base replaces.
  8. The compound of any one of claim 1-7 or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug, wherein R2It is selected from: C1-6Alkyl ,-((CH2)iO)m-(CH2)q-O-C1-6Alkyl,
    Wherein:
    I and q is each independently 1,2,3,4,5 or 6 at each occurrence;
    M is the arbitrary integer in 0-50, preferably 0-20, particularly preferred 0-6;
    R7、R8And R9It is each independently selected from H, C1-10Alkyl, C3-6Naphthenic base, C6-20Aryl and C7-20Aralkyl, the alkyl, naphthenic base, aryl and aralkyl are respectively optionally selected from halogen ,-OH ,-CN and-NO by one or more2Substituent group replace;
    Or R7And R8C is collectively formed in the carbon atom connected together with it3-6Naphthenic base;
    R10And R11It is each independently selected from H, halogen ,-OH ,-CN ,-NO2、C1-10Alkyl and C3-6Naphthenic base.
  9. The compound of any one of claim 1-8 or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug, wherein the compound has the structure of formula (II) or formula (II) -1:
  10. The compound of any one of claim 1-9 or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug, wherein the compound has the structure of any following formula:
  11. The compound of any one of claim 1-10 or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug, wherein the compound is selected from:
  12. Pharmaceutical composition, it includes the compounds of any one of the claim 1-11 of prevention or therapeutically effective amount or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug and one or more pharmaceutically acceptable carriers, described pharmaceutical composition to be preferably solid pharmaceutical preparation, liquid preparation or preparation capable of permeating skin.
  13. The method for preparing pharmaceutical composition, the method includes combining the compound of any one of claim 1-11 or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug with one or more pharmaceutically acceptable carriers.
  14. The pharmaceutical composition of the compound of any one of claim 1-11 or its pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolin or prodrug or claim 12 is preparing the purposes in the drug for preventing or treating viral disease.
  15. The purposes of claim 14, wherein the drug be by oral, intravenous, intra-arterial, subcutaneous, peritonaeum, intramuscular or transdermal routes of administration drug.
  16. The purposes of claim 14, wherein the viral disease is selected from viral hepatitis type A, virus B hepatitis, viral hepatitis type C, influenza, bleb and acquired immunodeficiency syndrome (AIDS).
  17. The method for preparing the compound of any one of claim 1-11, the described method comprises the following steps:
    Or
    Wherein:
    R12、R13And R14It is each independently selected from F, Cl, Br, I ,-NHR, hydroxyl, trifluoromethanesulfonic acid ester group, p-methyl benzenesulfonic acid ester group and boric acid ester group;
    PG is selected from tertbutyloxycarbonyl, benzyloxycarbonyl group, 9-fluorenylmethyloxycarbonyl, allyloxycarbonyl, trimethylsilyl ethoxycarbonyl, methoxycarbonyl group, carbethoxyl group, p-toluenesulfonyl, ortho-nitrophenyl sulfonyl, p-nitrophenyl sulfonyl, formoxyl, acetyl group, trifluoroacetyl group, propiono, pivaloyl group, phenyl, benzoyl, trityl, benzyl, 2,4- dimethoxy-benzyl and to methoxy-benzyl;
    Any one of remaining each group such as claim 1-11 is defined;
    The first step carries out in non-protonic solvent or under condition of no solvent in the presence of organic base or inorganic base and/or condensation reagent (particularly preferred DCC, DIC, EDC, BOP, PyAOP or PyBOP);
    Second step carries out under conditions of being suitable for removing PG group;
    Third step carries out in the presence of organic base or inorganic base in non-protonic solvent;And
    4th step preferably carries out in the presence of organic base or inorganic base and/or condensation reagent (particularly preferred DCC, DIC, EDC, BOP, PyAOP or PyBOP) in non-protonic solvent;
    Or it the described method comprises the following steps:
    Wherein:
    R12Selected from F, Cl, Br, I ,-NHR, hydroxyl, trifluoromethanesulfonic acid ester group, p-methyl benzenesulfonic acid ester group and boric acid ester group;
    LG is leaving group, is preferably pentafluorophenyl group and p-nitrophenyl;
    Any one of remaining each group such as claim 1-11 is defined;
    The first step carries out in non-protonic solvent in the presence of organic base or inorganic base;
    Second step carries out in non-protonic solvent in the presence of organic base or inorganic base;And
    Third step carries out in non-protonic solvent in the presence of organic base or inorganic base.
CN201780005344.3A 2016-11-02 2017-10-27 Heterocyclic compounds, process for their preparation and their use Active CN108602842B (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
CN201610945433 2016-11-02
CN2016109454333 2016-11-02
PCT/CN2017/108016 WO2018082503A1 (en) 2016-11-02 2017-10-27 Heterocyclic compound and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN108602842A true CN108602842A (en) 2018-09-28
CN108602842B CN108602842B (en) 2020-10-09

Family

ID=62076620

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201780005344.3A Active CN108602842B (en) 2016-11-02 2017-10-27 Heterocyclic compounds, process for their preparation and their use

Country Status (2)

Country Link
CN (1) CN108602842B (en)
WO (1) WO2018082503A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020113094A1 (en) 2018-11-30 2020-06-04 Nuvation Bio Inc. Pyrrole and pyrazole compounds and methods of use thereof
CN113248539B (en) * 2021-06-10 2021-09-07 中以海德人工智能药物研发股份有限公司 Imidazopyrimidines compounds

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5733890A (en) * 1996-10-09 1998-03-31 National Science Council Adenylate analogs as potent anti-herpes virus agents
CN103665043A (en) * 2012-08-30 2014-03-26 上海源力生物技术有限公司 Tenofovir prodrug and medical application thereof
CN104031104A (en) * 2013-03-08 2014-09-10 南京圣和药业有限公司 Novel nucleoside phosphoramidite chemical compounds and applications thereof
CN105315319A (en) * 2014-07-30 2016-02-10 南京圣和药业股份有限公司 Hepatitis C virus inhibitor and application thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5733890A (en) * 1996-10-09 1998-03-31 National Science Council Adenylate analogs as potent anti-herpes virus agents
CN103665043A (en) * 2012-08-30 2014-03-26 上海源力生物技术有限公司 Tenofovir prodrug and medical application thereof
CN104031104A (en) * 2013-03-08 2014-09-10 南京圣和药业有限公司 Novel nucleoside phosphoramidite chemical compounds and applications thereof
CN105315319A (en) * 2014-07-30 2016-02-10 南京圣和药业股份有限公司 Hepatitis C virus inhibitor and application thereof

Also Published As

Publication number Publication date
CN108602842B (en) 2020-10-09
WO2018082503A1 (en) 2018-05-11

Similar Documents

Publication Publication Date Title
JP6524081B2 (en) Dihydropyrimidine compounds and their application in medicine
ES2345102T3 (en) ANTIVIRAL ACTIVITY AND RESOLUTION OF 2-HYDROXIMETHYL-5- (5-FLUOROCITOSIN-1-IL) -1,3-OXATIOLANE.
JP2019516714A (en) Novel pyrazine compounds with oxygen, sulfur and nitrogen linkers for the treatment of infections
WO2016161268A1 (en) Hepatitis b antviral agents
ES2619937T3 (en) New 5-fluorouracil derivative
CA2913194C (en) Bisulfate of janus kinase (jak) inhibitor and preparation method therefor
CZ2003122A3 (en) 3 -Substituted-beta-L-nucleosides
JP7328254B2 (en) Tetrazoles containing apoptosis signal-regulating kinase 1 inhibitors and methods of use thereof
CN109790145B (en) Dihydropyrimidine compound and preparation method and application thereof
CN107250137A (en) It is used as the hexa-atomic saturation heterolipid ring class of the substituted amino of long-acting DPP IV inhibitor
CA3230060A1 (en) Benzamides of pyrazolyl-amino-pyrimidinyl derivatives, and compositions and methods thereof
KR20230091929A (en) bicyclic compound
WO2019213239A1 (en) Apoptosis signal-regulating kinase 1 inhibitors and methods of use thereof
WO2015197006A1 (en) Substituted amino acid thioester compound, and composition and application thereof
CN108602842A (en) Heterocyclic compound and its preparation method and application
CN111801100A (en) 7-substituted sulfoxy-purinone compounds and derivatives for the treatment and prevention of liver cancer
WO2018217439A1 (en) Substituted [5,6]cyclic-4(3h)-pyrimidinones as anticancer agents
CN113710276A (en) Benzodiazepine derivatives as RSV inhibitors
WO2020114495A1 (en) Dinucleotide compound and prodrug thereof
CN110938104A (en) Cyclic dinucleotide analogs, pharmaceutical compositions and uses thereof
WO2019091492A1 (en) Smac mimetics used as iap inhibitors and use thereof
WO2014000541A1 (en) Acyclic nucleotide analogue, preparation method therefor, and application thereof
CN111484541B (en) Dinucleotide prodrugs and methods of making same
TWI616453B (en) Substituted amino acid thioester compounds, materials and uses thereof
CN106132972B (en) Phosphoramidate for treating HCV infection

Legal Events

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