CN107793409B - Dihydropyrimidine compound and application thereof in medicine - Google Patents

Dihydropyrimidine compound and application thereof in medicine Download PDF

Info

Publication number
CN107793409B
CN107793409B CN201710783447.4A CN201710783447A CN107793409B CN 107793409 B CN107793409 B CN 107793409B CN 201710783447 A CN201710783447 A CN 201710783447A CN 107793409 B CN107793409 B CN 107793409B
Authority
CN
China
Prior art keywords
compound
ethyl
hbv
methyl
group
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201710783447.4A
Other languages
Chinese (zh)
Other versions
CN107793409A (en
Inventor
任青云
刘辛昌
张英俊
S·戈尔德曼
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.)
Guangdong HEC Pharmaceutical
Original Assignee
Sunshine Lake Pharma 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 Sunshine Lake Pharma Co Ltd filed Critical Sunshine Lake Pharma Co Ltd
Publication of CN107793409A publication Critical patent/CN107793409A/en
Application granted granted Critical
Publication of CN107793409B publication Critical patent/CN107793409B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention relates to a dihydropyrimidine compound and application thereof as a medicament, in particular to application of the dihydropyrimidine compound as a medicament for treating and preventing hepatitis B. Specifically, the invention relates to a compound shown in a general formula (I) or (Ia) or an enantiomer, a diastereoisomer, a tautomer, a hydrate, a solvate or a pharmaceutically acceptable salt thereof, wherein each variable is defined in the specification. The invention also relates to the use of the compounds of general formula (I) or (Ia) or enantiomers, diastereomers, tautomers, hydrates, solvates or pharmaceutically acceptable salts thereof as medicaments, in particular for the treatment and prophylaxis of hepatitis B.
Figure DDA0001397442510000011

Description

Dihydropyrimidine compound and application thereof in medicines
Technical Field
The invention relates to a dihydropyrimidine compound and application thereof as a medicament, in particular to application thereof as a medicament for treating and preventing hepatitis B. The invention also relates to a composition consisting of the dihydropyrimidine compounds and other antiviral agents and application of the dihydropyrimidine compounds and other antiviral agents in treating and preventing Hepatitis B (HBV) infection.
Background
Hepatitis b virus belongs to the hepadnaviridae family. It can cause acute and/or persistent progressive chronic disease. Hepatitis b virus also causes many other clinical manifestations in pathological morphology-in particular chronic inflammation of the liver, cirrhosis and canceration of hepatocytes. In addition, co-infection with hepatitis delta can have adverse effects on the progression of the disease.
The conventional drugs licensed for the treatment of chronic hepatitis are interferon and lamivudine (lamivudine). However, interferons have only moderate activity and high toxic side effects; while lamivudine (lamivudine) has good activity, its resistance increases rapidly during treatment and often produces a rebound effect after cessation of treatment, the IC of lamivudine (3-TC) 50 The value was 300nM (Science 299(2003), 893-896).
Deres et al reported heteroaromatic substituted dihydropyrimidine (HAP) compounds represented by Bay41-4109, Bay39-5493, which act to inhibit HBV replication by preventing normal nucleocapsid formation. Bay41-4109 shows better drug metabolism property in clinical research (Science,299(2003),893-896), and through the research of action mechanism, the aromatic ring substituted dihydropyrimidine compound changes the included angle between the dimers forming nucleocapsid through the action with 113-143 amino acid residues of core protein, leading to the formation of unstable swollen nucleocapsid, accelerating the degradation of core protein (biochem. Pharmacol.66(2003), 2273-2279).
There is still a need for new compounds which are effective as antiviral agents, in particular for use as agents for the treatment and/or prevention of hepatitis b.
The novel dihydropyrimidine compound provided by the invention has the advantages of good inhibitory activity, pharmacokinetic property, solubility, stability, low toxicity and the like, and has a good application prospect in the aspect of resisting HBV viruses.
Summary of the invention
The invention relates to a novel dihydropyrimidine compound and application thereof in preparing a medicament for treating and preventing HBV infection. In particular, the compounds of the present invention, and the pharmaceutically acceptable compositions thereof, are effective in inhibiting HBV infection.
In one aspect, the invention relates to a compound of formula (I) or (Ia),
Figure BDA0001397442500000011
or an enantiomer, diastereomer, tautomer, solvate, or pharmaceutically acceptable salt thereof, wherein each R, R is 1 、R 2 、R 3 And f have the meaning according to the invention.
In some embodiments, R is-X-Z;
x is- (CR) 7 R 7a ) t -or-C (═ O) -;
z is a substructure represented by formula (II):
Figure BDA0001397442500000012
w is CR 7 Or N;
y is- (CR) 7 R 7a ) t -、-O-、-S(=O) q -or-NR 6 -;
Each R 1 Independently hydrogen, F, Cl, Br, I, cyano, methyl, ethyl, n-propyl, isopropyl, methoxy, ethoxy, methylamino, ethylamino, nitro, 4-trifluoromethylphenyl, 3, 5-bis (trifluoromethyl) phenyl, or trifluoromethyl;
R 2 is hydrogen, alkyl, alkenyl, alkynyl, arylalkyl, heteroarylalkyl, cycloalkyl, cycloalkylalkyl or heterocyclylalkyl;
R 3 is C 6-10 Aryl or heteroaryl of 5 to 6 ring atoms, wherein C is 6-10 Aryl and heteroaryl of 5 to 6 ring atoms may be independently optionally substituted by 1,2,3,4 or 5 substituents selected from fluoro, chloro, bromo, iodo, (═ O), (═ S), alkyl, alkoxy, cyano, hydroxy, nitro, alkylaminoSubstituted with a substituent of yl, amino, trifluoromethyl, trifluoromethoxy, haloalkyl substituted aryl, halogen substituted aryl, or trifluoromethanesulfonyl;
each R 4 Independently hydrogen, deuterium, F, Cl, Br or C 1-4 An alkyl group;
R 5 is- (CR) 9 R 9a ) m -R 8
R 6 Is alkyl, alkenyl or alkynyl;
each R 7a And R 7 Independently hydrogen, deuterium, F, Cl, Br, alkyl, haloalkyl, - (CH) 2 ) m -OH or- (CH) 2 ) m -C(=O)O-R 8
R 8 Is heteroaryl or heterocyclyl, wherein said aryl and heteroaryl may be independently optionally substituted by 1,2,3,4 or 5 substituents selected from fluoro, chloro, bromo, iodo, (═ O), (═ S), alkyl, alkoxy, cyano, hydroxy, nitro, alkylamino, amino, trifluoromethyl, trifluoromethoxy, haloalkyl substituted aryl, halo substituted aryl or trifluoromethanesulfonyl;
each R 9a And R 9 Independently hydrogen, deuterium, F, Cl, Br, C 1-4 Alkyl or C 1-4 A haloalkyl group;
n is 0, 1,2,3,4 or 5;
each t is independently 0, 1 or 2;
each m is independently 0, 1,2,3 or 4;
f is 0, 1,2,3 or 4;
q is 0, 1 or 2.
In some embodiments, Z is a substructure represented by formula (III):
Figure BDA0001397442500000021
each R 4 Independently hydrogen, deuterium, F, Cl, Br, methyl, ethyl, n-propyl or isopropyl;
R 5 is- (CR) 9 R 9a ) m -R 8
Y is- (CR) 7 R 7a ) t -、-O-、-S(=O) q -or-NR 6 -;
R 6 Is C 1-4 An alkyl group;
each R 7a And R 7 Independently hydrogen, deuterium, F, Cl, Br, C 1-4 Alkyl radical, C 1-4 Haloalkyl or- (CH) 2 ) m -C(=O)O-R 8
R 8 Is heteroaryl consisting of 5-6 ring atoms or heterocyclic group consisting of 5-6 ring atoms, wherein, the heteroaryl consisting of 5-6 ring atoms and the heterocyclic group consisting of 5-6 ring atoms can be independently and optionally substituted by 1,2,3,4 or 5 groups selected from (═ O), (═ S), C 1-4 Alkyl or C 1-4 Substituted by a substituent of alkoxy;
each R 9a And R 9 Independently hydrogen, deuterium, F, Cl, methyl, ethyl or trifluoromethyl.
In some other embodiments, Z is a substructural formula as shown below:
Figure BDA0001397442500000022
each R 4 Independently hydrogen, deuterium, F, Cl, Br, methyl, ethyl, n-propyl or isopropyl;
each R 5 Independently is- (CR) 9 R 9a ) m -R 8
R 6 Is C 1-4 An alkyl group;
each R 9a And R 9 Independently hydrogen, deuterium, F, Cl, methyl or ethyl;
each R 8 Independently is a heteroaryl group consisting of 5-6 ring atoms or a heterocyclic group consisting of 5-6 ring atoms, wherein the heteroaryl group consisting of 5-6 ring atoms and the heterocyclic group consisting of 5-6 ring atoms can be independently and optionally substituted by 1,2,3,4 or 5 groups selected from (═ O), (═ S) or C 1-4 Alkyl substituents.
In some other embodiments, the R is 8 Is the following subformula:
Figure BDA0001397442500000031
wherein each R is 10 Independently hydrogen, methyl, ethyl, n-propyl or isopropyl.
In some embodiments, the R is 2 Is methyl, ethyl, n-propyl or isopropyl;
R 3 is phenyl, thiazolyl, thienyl, pyridyl, pyrazolyl, oxazolyl, furyl or imidazolyl, wherein said phenyl, thiazolyl, thienyl, pyridyl, pyrazolyl, oxazolyl, furyl and imidazolyl may be independently optionally substituted with 1,2,3,4 or 5 substituents selected from fluoro, chloro, bromo, iodo, methyl, ethyl, n-propyl or isopropyl.
In other embodiments, the compounds of the invention have a structure as shown in formula (IV) or (IVa),
Figure BDA0001397442500000032
or an enantiomer, diastereomer, tautomer, solvate, or pharmaceutically acceptable salt thereof, wherein,
z is a sub-structural formula shown in formula (III):
Figure BDA0001397442500000033
y is- (CR) 7 R 7a ) t -or-O-;
each R 4 Independently hydrogen, deuterium, F, Cl, Br, methyl, ethyl, n-propyl or isopropyl;
each R 1 Independently hydrogen, F, Cl, Br, I, methyl, ethyl, n-propyl, isopropyl, cyano, trifluoromethyl or methoxyA group;
R 2 is methyl, ethyl, n-propyl or isopropyl;
R 3 is phenyl, thiazolyl, thienyl, pyridyl, pyrazolyl, oxazolyl, furyl or imidazolyl, wherein said phenyl, thiazolyl, thienyl, pyridyl, pyrazolyl, oxazolyl, furyl and imidazolyl may be independently optionally substituted with 1,2,3,4 or 5 substituents selected from fluoro, chloro, bromo, iodo, methyl, ethyl, n-propyl or isopropyl;
R 5 is- (CR) 9 R 9a ) m -R 8
Each R 7a And R 7 Independently hydrogen, deuterium, F, Cl, Br, methyl, ethyl, n-propyl or isopropyl;
each R 9a And R 9 Independently hydrogen, deuterium, F, Cl, methyl or ethyl;
R 8 is composed of
Figure BDA0001397442500000034
Figure BDA0001397442500000041
Each R 10 Independently hydrogen, methyl, ethyl, n-propyl or isopropyl;
each n is independently 0, 1,2,3,4 or 5;
t is 0, 1 or 2;
m is 0, 1,2,3 or 4;
f is 0, 1,2 or 3.
In some further embodiments, Z is selected from the following substructures:
Figure BDA0001397442500000042
Figure BDA0001397442500000051
Figure BDA0001397442500000061
Figure BDA0001397442500000071
in another aspect, the present invention also provides a pharmaceutical composition comprising a compound of the present invention, further comprising a pharmaceutically acceptable carrier or a combination thereof.
In some embodiments, the pharmaceutical composition of the present invention further comprises an additional anti-HBV agent.
In some embodiments, the pharmaceutical composition of the invention, wherein the other anti-HBV agent is an HBV polymerase inhibitor, an immunomodulator, or an interferon.
In some embodiments, the pharmaceutical composition of the invention, wherein the other anti-HBV agent is lamivudine, telbivudine, tenofovir disoproxil, entecavir, adefovir dipivoxil, alfafenone, Alloferon, simon interleukin, cladribine, emtricitabine, faplovir, interferon, calamine CP, intefine, interferon alpha-1 b, interferon alpha-2 a, interferon beta-1 a, interferon alpha-2, interleukin-2, mevoxil, nitazoxanide, peginterferon alpha-2 a, ribavirin, roscovitine-a, cizopyran, Euforavac, azapril, Phosphazid, heplisv, interferon alpha-2 b, levamisole or propafege.
In another aspect, the invention also provides the use of the compound or the pharmaceutical composition in the preparation of a medicament for preventing, treating or alleviating a viral disease in a patient.
In some embodiments, the use of the invention, wherein the viral disease is hepatitis b infection or a disease caused by hepatitis b infection.
In still other embodiments, the use of the present invention, wherein the disease caused by hepatitis B infection is liver cirrhosis or hepatocellular carcinoma.
In another aspect, the invention relates to the use of a compound or pharmaceutical composition as described herein for the manufacture of a medicament for the prevention, treatment or alleviation of hepatitis b disease in a patient, comprising administering to the patient a therapeutically effective amount of a compound or a pharmaceutical composition as described herein.
Another aspect of the invention relates to a method of preventing, treating or ameliorating HBV disorders in a patient, comprising administering to the patient a pharmaceutically acceptable effective amount of a compound of the invention.
Another aspect of the invention relates to a method of preventing, treating or ameliorating HBV disorders in a patient, comprising administering to the patient a pharmaceutically acceptable effective amount of a pharmaceutical composition comprising a compound of the invention.
Another aspect of the invention relates to the use of a compound of the invention for the manufacture of a medicament for the prevention, treatment or treatment of HBV disorders in a patient and for lessening the severity thereof.
Another aspect of the present invention relates to the use of a pharmaceutical composition comprising a compound of the present invention for the manufacture of a medicament for the prevention, treatment or treatment of HBV disorders in a patient and for lessening the severity thereof.
Another aspect of the invention relates to a method of inhibiting HBV infection comprising contacting a cell with a compound or composition of the invention effective to inhibit HBV. In other embodiments, the method further comprises contacting the cell with another HBV therapeutic agent.
Another aspect of the present invention relates to the treatment of HBV disease in a patient, comprising administering to the patient a compound of the present invention or a composition thereof in a dosage effective to treat the disease. In other embodiments, the method further comprises administering an additional HBV therapeutic agent.
Another aspect of the present invention relates to a method of inhibiting HBV infection in a patient, comprising administering to the patient a compound of the present invention or a composition thereof in a dosage effective to treat the patient in need thereof. In other embodiments, the method further comprises administering a dose of an additional HBV therapeutic agent.
Another aspect of the invention relates to methods for the preparation, isolation and purification of compounds encompassed by formula (I) or formula (Ia).
The foregoing merely summarizes certain aspects of the invention and is not intended to be limiting. These and other aspects will be more fully described below.
Detailed description of the invention
Definitions and general terms
The invention will be described in detail in the literature corresponding to the identified embodiments, and the examples are accompanied by the graphic illustrations of structural formulae and chemical formulae. The present invention is intended to cover all alternatives, modifications and equivalents that may be included within the scope of the present invention as defined by the appended claims. Those skilled in the art will recognize many methods and materials similar or equivalent to those described herein which can be used in the practice of the present invention. The present invention is in no way limited to the description of methods and materials. There are many documents and similar materials that may be used to distinguish or contradict the present application, including, but in no way limited to, the definition of terms, their usage, the techniques described, or the scope as controlled by the present application.
The following definitions will apply to the invention unless otherwise indicated. For the purposes of the present invention, the chemical elements are described in the periodic table of elements, CAS version and handbook of chemicals, 75, th ed, 1994. In addition, the general principles of Organic Chemistry are described in "Organic Chemistry," Thomas Sorrell, University Science Books, Sausalito:1999, and "March's Advanced Organic Chemistry," by Michael B.Smith and Jerry March, John Wiley&Sons, New York:2007, all of which are hereby incorporated by reference.
As described herein, the compounds of the present invention may be optionally substituted with one or more substituents, such as those of the above general formula, or as specified in the examples, subclasses, and groups encompassed by the present invention. In general, the term "substituted" indicates that one or more hydrogen atoms in a given structure are replaced with a particular substituent. Unless otherwise indicated, an optional substituent group may have one substituent substituted at each substitutable position of the group. When more than one position in a given formula can be substituted with one or more substituents selected from a particular group, the substituents may be substituted at each position, identically or differently. Wherein the substituent may be, but is not limited to, fluorine, chlorine, bromine, iodine, (═ O), (═ S), alkyl, alkoxy, cyano, hydroxy, nitro, alkylamino, amino, trifluoromethyl, trifluoromethoxy, haloalkyl-substituted aryl, halo-substituted aryl, or trifluoromethanesulfonyl.
The term "alkyl" as used herein includes saturated straight or branched chain monovalent hydrocarbon radicals of 1 to 20 carbon atoms, wherein the alkyl radical may independently be optionally substituted with one or more substituents described herein. In some embodiments, the alkyl group contains 1 to 10 carbon atoms, in other embodiments, the alkyl group contains 1 to 8 carbon atoms, in other embodiments, the alkyl group contains 1 to 6 carbon atoms, in other embodiments, the alkyl group contains 1 to 4 carbon atoms, and in other embodiments, the alkyl group contains 1 to 3 carbon atoms. Further examples of alkyl groups include, but are not limited to, methyl (Me, -CH) 3 ) Ethyl (Et, -CH) 2 CH 3 ) N-propyl (n-Pr, -CH) 2 CH 2 CH 3 ) Isopropyl group (i-Pr, -CH (CH) 3 ) 2 ) N-butyl (n-Bu, -CH) 2 CH 2 CH 2 CH 3 ) 2-methylpropyl or isobutyl (i-Bu, -CH) 2 CH(CH 3 ) 2 ) 1-methylpropyl or sec-butyl (s-Bu, -CH (CH) 3 )CH 2 CH 3 ) T-butyl (t-Bu, -C (CH) 3 ) 3 ) N-pentyl (-CH) 2 CH 2 CH 2 CH 2 CH 3 ) 2-pentyl (-CH (CH) 3 )CH 2 CH 2 CH 3 ) 3-pentyl (-CH (CH) 2 CH 3 ) 2 ) 2-methyl-2-butyl (-C (CH) 3 ) 2 CH 2 CH 3 ) 3-methyl-2-butyl (-CH (CH) 3 )CH(CH 3 ) 2 ) 3-methyl-1-butyl (-CH) 2 CH 2 CH(CH 3 ) 2 ) 2-methyl-1-butyl (-CH) 2 CH(CH 3 )CH 2 CH 3 ) N-hexyl (-CH) 2 CH 2 CH 2 CH 2 CH 2 CH 3 ) 2-hexyl (-CH (CH) 3 )CH 2 CH 2 CH 2 CH 3 ) 3-hexyl (-CH (CH) 2 CH 3 )(CH 2 CH 2 CH 3 ) 2-methyl-2-pentyl (-C (CH)) 3 ) 2 CH 2 CH 2 CH 3 ) 3-methyl-2-pentyl (-CH (CH) 3 )CH(CH 3 )CH 2 CH 3 ) 4-methyl-2-pentyl (-CH (CH) 3 )CH 2 CH(CH 3 ) 2 ) 3-methyl-3-pentyl (-C (CH) 3 )(CH 2 CH 3 ) 2 ) 2-methyl-3-pentyl (-CH (CH) 2 CH 3 )CH(CH 3 ) 2 ) 2, 3-dimethyl-2-butyl (-C (CH) 3 ) 2 CH(CH 3 ) 2 ) 3, 3-dimethyl-2-butyl (-CH (CH) 3 )C(CH 3 ) 3 ) N-heptyl, n-octyl, and the like. The term "alkyl" and its prefix "alkane" as used herein, both include straight and branched saturated carbon chains. The term "alkylene" is used herein to denote a saturated divalent hydrocarbon radical resulting from the elimination of two hydrogen atoms from a straight or branched chain saturated hydrocarbon, examples of which include, but are not limited to, methylene, ethylidene, and the like.
As used herein, the term "haloaliphatic" or "haloalkyl" means that an aliphatic group or alkyl group is substituted with one or more of the same or different halogen atoms, wherein the aliphatic group or alkyl group has the meaning described herein, i.e., fluorine, chlorine, bromine, or iodine, examples of which include, but are not limited to, trifluoromethyl, trifluoroethyl, and the like.
The term "alkenyl" denotes a straight or branched chain monovalent hydrocarbon group of 2 to 12 carbon atoms, at least one of which C-C is sp 2 Double bonds in which the alkenyl group may be independently optionally substituted with one or more substituents described herein, including the positioning of the groups as "trans", "cis" or "E" or "Z", withExamples of entities include, but are not limited to, vinyl (-CH ═ CH) 2 ) Allyl (-CH) 2 CH=CH 2 ) And so on.
The term "alkynyl" denotes a straight or branched chain monovalent hydrocarbon radical of 2 to 12 carbon atoms wherein at least one C-C is an sp triple bond, wherein the alkynyl radical may independently be optionally substituted with one or more substituents as described herein, specific examples include, but are not limited to, ethynyl (-C.ident.CH), propargyl (-CH) 2 C ≡ CH), and so forth.
The terms "cyclic aliphatic", "carbocyclic", "carbocyclyl" refer to mono-or polyvalent, non-aromatic, saturated or partially unsaturated rings, including monocyclic 3 to 12 carbon atoms or bicyclic 7 to 12 carbon atoms. The carbocycle having 7 to 12 atoms may be a bicyclo [4,5], [5,5], [5,6] or [6,6] system, while the carbocycle having 9 or 10 atoms may be a bicyclo [5,6] or [6,6] system. Suitable cycloaliphatic groups include, but are not limited to, cycloalkyl, cycloalkenyl and cycloalkynyl. Examples of cycloaliphatic radicals further include, but are in no way limited to, cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopentyl-1-alkenyl, 1-cyclopentyl-2-alkenyl, 1-cyclopentyl-3-alkenyl, cyclohexyl, 1-cyclohexyl-1-alkenyl, 1-cyclohexyl-2-alkenyl, 1-cyclohexyl-3-alkenyl, cyclohexadienyl, cycloheptyl, cyclooctyl, cyclononyl, cyclodecyl, cycloundecyl, cyclododecyl, and the like.
The term "cycloalkyl" denotes a monovalent or polyvalent saturated monocyclic, bicyclic or tricyclic ring system containing from 3 to 12 carbon atoms. In one embodiment, the cycloalkyl group contains 3 to 12 carbon atoms; in another embodiment, the cycloalkyl group contains 3 to 8 carbon atoms; in yet another embodiment, the cycloalkyl group contains 3 to 6 carbon atoms. The cycloalkyl groups may be independently unsubstituted or substituted with one or more substituents described herein.
The term "h ring atoms", where h is an integer, typically describes the number of ring-forming atoms in the molecule, which is h. For example, piperidinyl is a heterocyclic group consisting of 6 atoms.
The term "heteroBy "cyclyl" is meant a non-aromatic, saturated or partially unsaturated, monocyclic, bicyclic or tricyclic ring system containing from 3 to 12 ring atoms, wherein at least one ring atom is selected from nitrogen, sulfur or oxygen. Wherein said heterocyclyl group may be optionally substituted with one or more substituents as described herein. Unless otherwise specified, a heterocyclic group may be carbon-or nitrogen-based, and-CH 2 -a group may optionally be replaced by-C (═ O) -or-C (═ S) -. The sulfur atom of the ring may optionally be oxidized to the S-oxide. The nitrogen atoms of the ring may optionally be oxidized to the N-oxide. In some embodiments, heterocyclyl is a 3-7 atom heterocyclyl and refers to a monovalent or polyvalent, saturated or partially unsaturated, non-aromatic, monocyclic or bicyclic ring containing 3-7 ring atoms, wherein at least one ring atom is selected from the group consisting of nitrogen, sulfur, and oxygen atoms. In other embodiments, heterocyclyl is a 5-atom heterocyclyl and refers to a monovalent or multivalent, saturated or partially unsaturated, non-aromatic monocyclic ring comprising 5 ring atoms, wherein at least one ring atom is selected from the group consisting of nitrogen, sulfur, and oxygen atoms. In other embodiments, heterocyclyl is a 6-atom heterocyclyl and refers to a monovalent or multivalent, saturated or partially unsaturated, non-aromatic monocyclic ring comprising 6 ring atoms, wherein at least one ring atom is selected from the group consisting of nitrogen, sulfur, and oxygen atoms. The heterocyclic group of 6 atoms includes a saturated heterocyclic group and a partially unsaturated heterocyclic group of 6 atoms.
Examples of heterocyclic groups include, but are not limited to: the heterocyclic group may be a carbon-based or heteroatom group. "Heterocyclyl" likewise includes heterocyclic groups fused to saturated or partially unsaturated rings or heterocycles. Examples of heterocycles include, but are not limited to, pyrrolidinyl, tetrahydrofuryl, dihydrofuranyl, tetrahydrothienyl, tetrahydropyranyl, dihydropyranyl, tetrahydrothiopyranyl, piperidinyl, morpholinyl, thiomorpholinyl, thiaxanyl, piperazinyl, homopiperazinyl, azetidinyl, oxetanyl, thietanyl, homopiperidinyl, epoxypropyl, azepinyl, oxepinyl, thietanyl, oxazepinyl, diazepinyl, thiazepinyl, 2-pyrrolinyl, 3-pyrrolinyl, indolinyl, 2H-pyranyl, 4H-pyranyl, dioxacyclohexyl, 1, 3-dioxolyl, pyrazolinyl, dithianyl, dithiazolyl, dithienyl, dihydrothienyl, pyrazolylimidazolinyl, imidazolidinyl, 1,2,3, 4-tetrahydroisoquinolinyl, dihydrofuranyl, tetrahydroquinolinyl, oxathiofuranyl, oxacycloheptanyl, thianyl, oxanyl, oxacycloheptanyl, oxanyl, thianyl, thiafuranyl, thianyl, and thianyl, 3-azabicyclo [3.1.0] hexyl, 3-azabicyclo [4.1.0] heptyl, azabicyclo [2.2.2] hexyl, 3H-indolylquinazinyl, and N-pyridylurea. Examples of heterocyclic groups also include, 1, 1-dioxothiomorpholinyl. Examples of the group in which the carbon atom on the ring is substituted with oxo (═ O) include, but are not limited to, pyrimidinedione, 1,2, 4-thiadiazol-5 (4H) -keto, 1,2, 4-oxadiazol-5 (4H) -keto, 1,3, 4-oxadiazol-2 (3H) -keto, 1H-1,2, 4-triazol-5 (4H) -keto, and the like, and examples in which the carbon atom on the ring is substituted with ═ S include, but are not limited to, 1,2, 4-oxadiazol-5 (4H) -thioketo, 1,3, 4-oxadiazol-2 (3H) -thioketo, and the like. And the heterocyclic group may be substituted or unsubstituted, wherein the substituent may be, but is not limited to, fluorine, chlorine, bromine, iodine, (═ O), (═ S), alkyl, alkoxy, cyano, hydroxy, nitro, alkylamino, amino, trifluoromethyl, trifluoromethoxy, haloalkyl substituted aryl, halo substituted aryl, or trifluoromethanesulfonyl.
The term "heterocyclylalkyl" includes heterocyclyl-substituted alkyl groups, examples of which include, but are not limited to, pyrrole-2-methyl and morpholine-4-methyl, and the like.
The term "heterocyclylalkoxy" includes heterocyclyl-substituted-alkoxy groups in which an oxygen atom is attached to the rest of the molecule, examples of which include, but are not limited to, pyrrole-2-methoxy and piperidine-2-ethoxy, and the like.
The term "heterocyclylalkylamino" includes heterocyclyl-substituted alkylamino groups in which the nitrogen atom is attached to the remainder of the molecule; wherein the heterocyclyl and alkylamino groups have the meaning as described herein, examples of which include, but are not limited to, piperazine-2-ethylamino, morpholine-4-propoxy, morpholine-4-ethylamino, and the like.
The term "heteroatom" means one or more of O, S, N, P and Si, including any oxidation state form of N, S and P; primary, secondary, tertiary amines and quaternary ammonium salt forms; or a form in which a hydrogen on a nitrogen atom in the heterocycle is substituted, for example, N (like N in 3, 4-dihydro-2H-pyrrolyl), NH (like NH in pyrrolidinyl) or NR (like NR in N-substituted pyrrolidinyl).
The term "halogen" or "halogen atom" refers to F, Cl, Br or I.
The term "unsaturated" as used herein means that the moiety contains one or more degrees of unsaturation.
The term "alkoxy", as used herein, relates to an alkyl group, as defined herein, attached to the main carbon chain through an oxygen atom ("alkoxy").
The terms "haloalkyl", "haloalkenyl" or "haloalkoxy" denote alkyl, alkenyl or alkoxy groups which may be substituted by one or more halogen atoms which may be the same or different. Wherein alkyl, alkenyl and alkoxy groups have the meaning as described herein, examples of which include, but are not limited to, trifluoromethyl, trifluoromethoxy, 2-fluorovinyl, and the like.
The term "aryl" used alone or as a majority of "aralkyl", "aralkoxy", or "aryloxyalkyl" refers to monocyclic, bicyclic, and tricyclic carbon ring systems of 6-14 ring atoms in combination, wherein at least one ring system is aromatic, wherein each ring system contains 3-7 ring atoms with only one attachment point attached to the rest of the molecule. The term "aryl" may be used interchangeably with the term "aromatic ring", e.g., aromatic rings may include phenyl, naphthyl and anthracenyl. And the aryl group may be substituted or unsubstituted, wherein the substituent may be, but is not limited to, fluorine, chlorine, bromine, iodine, (═ O), (═ S), alkyl, alkoxy, cyano, hydroxyl, nitro, alkylamino, amino, trifluoromethyl, trifluoromethoxy, haloalkyl-substituted aryl, halogen-substituted aryl, or trifluoromethanesulfonyl.
The term "heteroaryl" denotes monocyclic, bicyclic and tricyclic ring systems containing 5 to 12 ring atoms, or 5 to 10 ring atoms, or 5 to 6 ring atoms, wherein at least one ring is aromatic and at least one aromatic ring contains one or more heteroatoms, wherein each ring system contains a ring of 5 to 7 ring atoms with one or more attachment points to the rest of the molecule. The term "heteroaryl" may be used interchangeably with the terms "heteroaromatic", "heteroaromatic ring" or "heteroaromatic compound". In some embodiments, heteroaryl is a heteroaryl consisting of 5 to 12 ring atoms containing 1,2,3, or 4 heteroatoms independently selected from nitrogen, sulfur, and oxygen. In other embodiments, heteroaryl is a heteroaryl consisting of 5 to 10 ring atoms containing 1,2,3, or 4 heteroatoms independently selected from nitrogen, sulfur, and oxygen. In other embodiments, heteroaryl is a heteroaryl consisting of 5 to 6 ring atoms containing 1,2,3, or 4 heteroatoms independently selected from nitrogen, sulfur, and oxygen. And the heteroaryl group may be substituted or unsubstituted, wherein the substituent may be, but is not limited to, fluorine, chlorine, bromine, iodine, (═ O), (═ S), alkyl, alkoxy, cyano, hydroxyl, nitro, alkylamino, amino, trifluoromethyl, trifluoromethoxy, haloalkyl-substituted aryl, halogen-substituted aryl, or trifluoromethanesulfonyl.
Examples of heteroaromatic rings include, but are not limited to, the following monocyclic rings: 1,2, 4-oxadiazol-5 (4H) -thioketo group, 1,2, 4-thiadiazol-5 (4H) -keto group, 1,2, 4-oxadiazol-5 (4H) -keto group, 1,3, 4-oxadiazol-2 (3H) -thioketo group, 1H-1,2, 4-triazol-5 (4H) -keto group, 2-furyl group, 3-furyl group, N-imidazolyl group, 2-imidazolyl group, 4-imidazolyl group, 5-imidazolyl group, 3-isoxazolyl group, 4-isoxazolyl group, 5-isoxazolyl group, 2-oxazolyl group, 4-oxazolyl group, 5-oxazolyl group, N-pyrrolyl group, 2-pyrrolyl group, 3-pyrrolyl group, 2-pyridyl group, azoxy group, thiodiazolyl group, 2-pyridyl group, 2-oxazolyl group, 2-pyridyl group, and the like, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, pyridazinyl (e.g. 3-pyridazinyl), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (e.g. 5-tetrazolyl), triazolyl (e.g. 2-triazolyl and 5-triazolyl), 2-thienyl, 3-thienyl, pyranyl, pyrazolyl (e.g. 2-pyrazolyl), isothiazolyl, 1,2, 3-oxadiazolyl, 1,2, 5-oxadiazolyl, 1,2, 4-oxadiazolyl, 1,2, 3-triazolyl, 1,2, 3-thiadiazolyl, 1,3, 4-thiadiazolyl, 1,2, 5-thiadiazolyl, pyrazinyl, 1,3, 5-triazinyl, Diazolyl, thiadiazolyl, triazinyl, and the like; the following bicyclic rings are also included, but in no way limited to these: benzothiazolyl, benzimidazolyl, benzofuranyl, benzothienyl, indolyl (e.g., 2-indolyl), purinyl, quinolyl (e.g., 2-quinolyl, 3-quinolyl, 4-quinolyl), isoquinolyl (e.g., 1-isoquinolyl, 3-isoquinolyl, or 4-isoquinolyl), and the like.
The term "heteroarylalkyl" means that an alkyl group is substituted with one or more identical or different heteroaryl groups, wherein alkyl and heteroaryl groups have the meaning as described herein, examples of which include, but are not limited to, pyridine-2-ethyl, thiazole-2-methyl, imidazole-2-ethyl, pyrimidine-2-propyl, and the like.
The term "sulfonyl", whether used alone or in combination with other terms such as "alkylsulfonyl", denotes the divalent group-SO 2 -. The term "alkylsulfonyl" refers to an alkyl-substituted sulfonyl group that forms an alkylsulfonyl group (e.g., -SO) 2 CH 3 )。
The term "alkylthio" includes C 1-10 A linear or branched alkyl group is attached to a divalent sulfur atom, wherein the alkyl group has the meaning as described herein. In some of these embodiments, the alkylthio group is a lower C 1-3 Alkylthio groups, and such examples include, but are not limited to, methylthio (CH) 3 S-), ethylthio, and the like.
The terms "aralkyl", "arylalkyl" and "arylalkyl" include aryl-substituted alkyl groups in which the aryl and alkyl groups have the meaning as set forth herein. In some of these examples, an aralkyl group or arylalkyl group refers to a "lower aralkyl" group, i.e., the aryl group is attached to C 1-6 On the alkyl group. In other embodiments, the aralkyl or arylalkyl group is C-containing 1-3 "phenylalkylene" of an alkyl group. Specific examples thereof include phenylmethyl (i.e., benzyl), diphenylmethyl, phenylethyl and the like. And the aryl group on the aralkyl group may be optionally further substituted with a substituent of fluorine, chlorine, bromine, iodine, (═ O), (═ S), alkyl, alkoxy, cyano, hydroxy, nitro, alkylamino, amino, trifluoromethyl, trifluoromethoxy, haloalkyl-substituted aryl, halogen-substituted aryl, or trifluoromethanesulfonyl.
The terms "alkylamino", "alkylamino" include "N-alkylamino" and "N, N-dialkylamino" in which the hydrogen atoms in the amino groups are each independently substituted by one or two identical or different alkyl groups, wherein the alkyl groups have the meaning as described herein. In some of these embodiments, the alkylamino group is one or two C 1-6 Lower alkylamino groups in which the alkyl group is attached to the nitrogen atom. In other embodiments, the alkylamino group is C 1-3 Lower alkylamino groups of (a). Suitable alkylamino groups can be monoalkylamino or dialkylamino, and such examples include, but are not limited to, N-methylamino, N-ethylamino, N-dimethylamino, N-diethylamino, and the like.
The term "haloalkyl-substituted aryl" includes aryl groups which may be substituted with one or more of the same or different haloalkyl groups, wherein the haloalkyl and aryl groups have the meaning as described herein. Examples include, but are not limited to, 2-trifluoromethylphenyl, 3, 5-bis (trifluoromethyl) phenyl, 3-trifluoromethylphenyl, 4-trifluoromethylphenyl, 2, 6-bis (trifluoromethyl) phenyl, and the like.
The term "halogen-substituted aryl" includes aryl groups which may be substituted by one or more of the same or different halogen atoms, wherein the halogen atom (halogen) and the aryl group have the meaning as described herein. Examples include, but are not limited to, fluorophenyl, difluorophenyl, trifluorophenyl, chlorophenyl, dichlorophenyl, trichlorophenyl, bromophenyl, tribromophenyl, dibromophenyl, fluorophenylphenyl, fluorobromophenyl, chlorobromophenyl, and the like.
The term "cycloalkylalkyl" denotes that an alkyl group may be substituted by one or more identical or different cycloalkyl groups, wherein cycloalkyl and alkyl groups have the meaning as described herein. Examples include, but are not limited to, cyclohexylmethyl, cyclopropylethyl, and the like.
As described herein, the ring system formed by a substituent that is attached to the central ring through a bond (as shown in formula a) represents that the substituent may be substituted at any substitutable position on the ring, and may be an enantiomer-containing substitution, as shown in formulae b, c, d, e, f, g, and h.
Figure BDA0001397442500000111
In addition, unless otherwise expressly indicated, the descriptions "… and … are each independently," "… and … are each independently," and "… and … are each independently" used throughout this document are interchangeable and should be broadly construed to mean that particular items expressed between the same symbols in different groups do not affect each other, or that particular items expressed between the same symbols in the same groups do not affect each other. For example, as in formula p, a plurality of R 4 Are not affected by each other.
Figure BDA0001397442500000121
As described herein, there are two linking sites in the system that are attached to the rest of the molecule, for example, as shown by formula q, which means that either the E or E' terminus is attached to the rest of the molecule, i.e., the linking modes at the two ends can be interchanged under the circumstances of reasonable molecular structure.
Figure BDA0001397442500000122
Unless otherwise indicated, the structural formulae depicted herein include all isomeric forms (e.g., enantiomers, diastereomers, and geometric (or conformational isomers) such as the R, S configuration containing an asymmetric center, (Z), (E) isomers of double bonds, and conformational isomers of (Z), (E). accordingly, a single stereochemical isomer of a compound of the invention or an enantiomer, diastereomer, or mixture of geometric isomers (or conformational isomers) thereof is within the scope of the invention.
The invention isThe term "prodrug" as used herein, represents a compound that is converted in vivo to a compound of formula (I) or (Ia). Such conversion is effected by hydrolysis of the prodrug in the blood or by enzymatic conversion to the parent structure in the blood or tissue. The prodrug compound of the invention can be ester, and in the prior invention, the ester can be used as the prodrug and comprises phenyl ester and aliphatic (C) 1-24 ) Esters, acyloxymethyl esters, carbonates, carbamates and amino acid esters. For example, a compound of the present invention contains a hydroxy group, i.e., it can be acylated to provide the compound in prodrug form. Other prodrug forms include phosphate esters, such as those obtained by phosphorylation of a hydroxyl group on the parent. For a complete discussion of prodrugs, reference may be made to the following: T.Higuchi and V.Stella, Pro-drugs as Novel Delivery Systems, Vol.14of the A.C.S.Symphosis Series, Edward B.Roche, ed., Bioreversible Carriers in Drug designs, American Pharmaceutical Association and Pergamon Press,1987, J.Rautio et al, Prodrugs: Design and Clinical Applications, Nature Review Drug Discovery,2008,7, 255-.
Unless otherwise indicated, all tautomeric forms of the compounds of the invention are included within the scope of the invention. In addition, unless otherwise indicated, the structural formulae of the compounds described herein include isotopically enriched concentrations of one or more different atoms.
"metabolite" refers to the product of a particular compound or salt thereof obtained by metabolism in vivo. Metabolites of a compound can be identified by techniques well known in the art, and its activity can be characterized by assays as described herein. Such products may be obtained by administering the compound by oxidation, reduction, hydrolysis, amidation, deamidation, esterification, defatting, enzymatic cleavage, and the like. Accordingly, the present invention includes metabolites of compounds, including metabolites produced by contacting a compound of the present invention with a mammal for a sufficient period of time.
The definition and convention of stereochemistry in the present invention is generally used with reference to the following documents: S.P. Parker, Ed., McGraw-Hill Dictionary of Chemical Terms (1984) McGraw-Hill Book Company, Ne York; and Eliel, E.and Wilen, S., "stereoschemistry of Organic Compounds", John Wiley & Sons, Inc., New York,1994. All stereoisomeric forms of the compounds of the present invention, including, but in no way limited to, diastereomers, enantiomers, atropisomers, and mixtures thereof, such as racemic mixtures, form part of the present invention. Many organic compounds exist in optically active form, i.e., they have the ability to rotate the plane of plane polarized light. In describing optically active compounds, the prefix D, L or R, S is used to indicate the absolute configuration of the chiral center of the molecule. The prefix d, l or (+), (-) is used to designate the sign of the rotation of the plane polarized light of the compound, with (-) or l indicating that the compound is left-handed and the prefix (+) or d indicating that the compound is right-handed. The chemical structures of these stereoisomers are identical, but their stereo structures are different. A particular stereoisomer may be an enantiomer, and a mixture of isomers is commonly referred to as a mixture of enantiomers. 50: 50 is called a racemic mixture or racemate, which may result in no stereoselectivity or stereospecificity during the chemical reaction. The terms "racemic mixture" and "racemate" refer to an equimolar mixture of two enantiomers, lacking optical activity.
The term "tautomer" or "tautomeric form" means that isomers of structures of different energies may be interconverted through a low energy barrier. For example, proton tautomers (i.e., prototropic tautomers) include tautomers that move through protons, such as keto-enol and imine-enamine isomerizations. Valence (valence) tautomers include tautomers that recombine to form bonded electrons.
As used herein, "pharmaceutically acceptable salts" refers to both organic and inorganic salts of the compounds of the present invention. Pharmaceutically acceptable saltsAre well known in the art, such as in the literature: berge et al, descriptive pharmacologically acceptable salts in detail in J. pharmaceutical Sciences,66:1-19,1977. Pharmaceutically acceptable non-toxic acid salts include, but are not limited to, salts of inorganic acids such as hydrochlorides, hydrobromides, phosphates, sulfates, perchlorates, and salts of organic acids such as acetates, oxalates, maleates, tartrates, citrates, succinates, malonates, which are formed by reaction with amino groups, or which are obtained by other methods described in the literature, such as ion exchange. Other pharmaceutically acceptable salts include adipate, malate, 2-hydroxypropionate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, cyclopentylpropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumerate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodiate, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, palmitate, pamoate, pectate, persulfate, 3-phenylpropionate, alginate, pectin, and mixtures thereof, Picrates, pivalates, propionates, stearates, thiocyanates, p-toluenesulfonates, undecanoates, pentanoates, and the like. Salts obtained with appropriate bases include alkali metals, alkaline earth metals, ammonium and N + (C 1-4 Alkyl radical) 4 A salt. The present invention also contemplates quaternary ammonium salts formed from compounds containing groups of N. Water-soluble or oil-soluble or dispersion products can be obtained by quaternization. Alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Pharmaceutically acceptable salts further include suitable, non-toxic ammonium, quaternary ammonium salts and amine cations resistant to formation of counterions, such as halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, C 1-8 Sulfonates and aromatic sulfonates.
"solvate" of the present invention refers to an association of one or more solvent molecules with a compound of the present invention. Solvents that form solvates include, but are not limited to, water, isopropanol, ethanol, methanol, dimethyl sulfoxide, ethyl acetate, acetic acid, and aminoethanol. The term "hydrate" refers to an association of solvent molecules with water.
The term "protecting group" or "Pg" refers to a substituent that when reacted with another functional group, is typically used to block or protect a particular functionality. For example, "amino protecting group" refers to a substituent attached to an amino group to block or protect the functionality of the amino group in a compound, and suitable amino protecting groups include acetyl, trifluoroacetyl, tert-Butoxycarbonyl (BOC), benzyloxycarbonyl (CBZ), and 9-fluorenylmethyleneoxycarbonyl (Fmoc). Similarly, "hydroxyl protecting group" refers to the functionality of a substituent of a hydroxyl group to block or protect the hydroxyl group, and suitable protecting groups include acetyl and silyl groups. "carboxyl protecting group" refers to a substituent of a carboxyl group used to block or protect the functionality of the carboxyl group, and typical carboxyl protecting groups include-CH 2 CH 2 SO 2 Ph, cyanoethyl, 2- (trimethylsilyl) ethyl, 2- (trimethylsilyl) ethoxymethyl, 2- (p-toluenesulfonyl) ethyl, 2- (p-nitrobenzenesulfonyl) ethyl, 2- (diphenylphosphino) ethyl, nitroethyl, and the like. General descriptions of protecting groups can be found in the literature: greene, Protective Groups in Organic Synthesis, John Wiley&Sons,New York,1991;and P.J.Kocienski,Protecting Groups,Thieme,Stuttgart,2005。
Description of the Compounds of the invention
The compound and the pharmaceutically acceptable composition thereof can effectively inhibit HBV infection.
In one aspect, the invention relates to a compound of formula (I) or (Ia),
Figure BDA0001397442500000141
or itAn enantiomer, diastereomer, tautomer, solvate, or pharmaceutically acceptable salt, wherein each R, R is 1 、R 2 、R 3 And f have the meaning described in the present invention.
In some embodiments, R is-X-Z;
x is- (CR) 7 R 7a ) t -or-C (═ O) -;
z is a substructure represented by formula (II):
Figure BDA0001397442500000142
w is CR 7 Or N;
y is- (CR) 7 R 7a ) t -、-O-、-S(=O) q -or-NR 6 -;
Each R 1 Independently hydrogen, F, Cl, Br, I, cyano, methyl, ethyl, n-propyl, isopropyl, methoxy, ethoxy, methylamino, ethylamino, nitro, 4-trifluoromethylphenyl, 3, 5-bis (trifluoromethyl) phenyl, or trifluoromethyl;
R 2 is hydrogen, alkyl, alkenyl, alkynyl, arylalkyl, heteroarylalkyl, cycloalkyl, cycloalkylalkyl or heterocyclylalkyl;
R 3 is C 6-10 Aryl or heteroaryl of 5 to 6 ring atoms, wherein C is 6-10 Aryl and heteroaryl consisting of 5 to 6 ring atoms may be independently optionally substituted by 1,2,3,4 or 5 substituents selected from fluoro, chloro, bromo, iodo, (═ O), (═ S), alkyl, alkoxy, cyano, hydroxy, nitro, alkylamino, amino, trifluoromethyl, trifluoromethoxy, haloalkyl substituted aryl, halo substituted aryl or trifluoromethanesulfonyl;
each R 4 Independently hydrogen, deuterium, F, Cl, Br or C 1-4 An alkyl group;
R 5 is- (CR) 9 R 9a ) m -R 8
R 6 Is alkyl, alkenyl or alkynyl;
each R 7a And R 7 Independently hydrogen, deuterium, F, Cl, Br, alkyl, haloalkyl, - (CH) 2 ) m -OH or- (CH) 2 ) m -C(=O)O-R 8
R 8 Is heteroaryl or heterocyclyl, wherein said aryl and heteroaryl may be independently optionally substituted by 1,2,3,4 or 5 substituents selected from fluoro, chloro, bromo, iodo, (═ O), (═ S), alkyl, alkoxy, cyano, hydroxy, nitro, alkylamino, amino, trifluoromethyl, trifluoromethoxy, haloalkyl substituted aryl, halo substituted aryl or trifluoromethanesulfonyl;
each R 9a And R 9 Independently hydrogen, deuterium, F, Cl, Br, C 1-4 Alkyl or C 1-4 A haloalkyl group;
n is 0, 1,2,3,4 or 5;
each t is independently 0, 1 or 2;
each m is independently 0, 1,2,3 or 4;
f is 0, 1,2,3 or 4;
q is 0, 1 or 2.
In some embodiments, Z is a substructure represented by formula (III):
Figure BDA0001397442500000143
each R 4 Independently hydrogen, deuterium, F, Cl, Br, methyl, ethyl, n-propyl or isopropyl;
R 5 is- (CR) 9 R 9a ) m -R 8
Y is- (CR) 7 R 7a ) t -、-O-、-S(=O) q -or-NR 6 -;
R 6 Is C 1-4 An alkyl group;
each R 7a And R 7 Independently hydrogen, deuterium, F, Cl, Br, C 1-4 Alkyl radical, C 1-4 Haloalkyl or- (CH) 2 ) m -C(=O)O-R 8
R 8 Is heteroaryl consisting of 5-6 ring atoms or heterocyclic group consisting of 5-6 ring atoms, wherein, the heteroaryl consisting of 5-6 ring atoms and the heterocyclic group consisting of 5-6 ring atoms can be independently and optionally substituted by 1,2,3,4 or 5 groups selected from (═ O), (═ S), C 1-4 Alkyl or C 1-4 Substituted with a substituent of alkoxy;
each R 9a And R 9 Independently hydrogen, deuterium, F, Cl, methyl, ethyl or trifluoromethyl.
In some other embodiments, Z is a substructural formula as shown below:
Figure BDA0001397442500000151
each R 4 Independently hydrogen, deuterium, F, Cl, Br, methyl, ethyl, n-propyl or isopropyl;
each R 5 Independently is- (CR) 9 R 9a ) m -R 8
R 6 Is C 1-4 An alkyl group; each R 9a And R 9 Independently hydrogen, deuterium, F, Cl, methyl or ethyl;
each R 8 Independently is a heteroaryl group consisting of 5-6 ring atoms or a heterocyclic group consisting of 5-6 ring atoms, wherein the heteroaryl group consisting of 5-6 ring atoms and the heterocyclic group consisting of 5-6 ring atoms can be independently optionally substituted by 1,2,3,4 or 5 substituents selected from (═ O), (═ S) or C 1-4 Alkyl substituents.
In some other embodiments, the R is 8 Is the following subformula:
Figure BDA0001397442500000152
wherein each R is 10 Independently hydrogen, methyl, ethyl, n-propyl or isopropyl.
In some embodiments, the R is 2 Is a firstAlkyl, ethyl, n-propyl or isopropyl;
R 3 is phenyl, thiazolyl, thienyl, pyridyl, pyrazolyl, oxazolyl, furyl or imidazolyl, wherein said phenyl, thiazolyl, thienyl, pyridyl, pyrazolyl, oxazolyl, furyl and imidazolyl may be independently optionally substituted with 1,2,3,4 or 5 substituents selected from fluoro, chloro, bromo, iodo, methyl, ethyl, n-propyl or isopropyl.
In other embodiments, the compounds of the invention have a structure as shown in formula (IV) or (IVa),
Figure BDA0001397442500000153
or an enantiomer, diastereomer, tautomer, solvate, or pharmaceutically acceptable salt thereof, wherein,
z is a sub-structural formula shown in formula (III):
Figure BDA0001397442500000161
y is- (CR) 7 R 7a ) t -or-O-;
each R 4 Independently hydrogen, deuterium, F, Cl, Br, methyl, ethyl, n-propyl or isopropyl;
each R 1 Independently hydrogen, F, Cl, Br, I, methyl, ethyl, n-propyl, isopropyl, cyano, trifluoromethyl or methoxy;
R 2 is methyl, ethyl, n-propyl or isopropyl;
R 3 is phenyl, thiazolyl, thienyl, pyridyl, pyrazolyl, oxazolyl, furyl or imidazolyl, wherein said phenyl, thiazolyl, thienyl, pyridyl, pyrazolyl, oxazolyl, furyl and imidazolyl may be independently optionally substituted with 1,2,3,4 or 5 substituents selected from fluoro, chloro, bromo, iodo, methyl, ethyl, n-propyl or isopropyl;
R 5 is- (CR) 9 R 9a ) m -R 8
Each R 7a And R 7 Independently hydrogen, deuterium, F, Cl, Br, methyl, ethyl, n-propyl or isopropyl;
each R 9a And R 9 Independently hydrogen, deuterium, F, Cl, methyl or ethyl;
R 8 is composed of
Figure BDA0001397442500000162
Figure BDA0001397442500000163
Each R 10 Independently hydrogen, methyl, ethyl, n-propyl or isopropyl;
each n is independently 0, 1,2,3,4 or 5;
t is 0, 1 or 2;
m is 0, 1,2,3 or 4;
f is 0, 1,2 or 3.
In some further embodiments, Z is selected from the following subformulas:
Figure BDA0001397442500000164
Figure BDA0001397442500000171
Figure BDA0001397442500000181
Figure BDA0001397442500000191
in other embodiments, the compounds of the present invention comprise a compound of one of the following, or an enantiomer, diastereomer, tautomer, solvate, or pharmaceutically acceptable salt thereof, but are in no way limited to such compounds:
Figure BDA0001397442500000192
Figure BDA0001397442500000201
Figure BDA0001397442500000211
Figure BDA0001397442500000221
Figure BDA0001397442500000231
Figure BDA0001397442500000241
Figure BDA0001397442500000251
Figure BDA0001397442500000261
Figure BDA0001397442500000271
in another aspect, the invention also provides a pharmaceutical composition containing the compound of the invention, and a pharmaceutically acceptable carrier or a combination thereof.
In some embodiments, the pharmaceutical composition of the present invention further comprises other anti-HBV agents.
In some embodiments, the pharmaceutical composition of the invention, wherein the other anti-HBV agent is an HBV polymerase inhibitor, an immunomodulator, or an interferon.
In some embodiments, the pharmaceutical composition of the invention, wherein the other anti-HBV agent is lamivudine, telbivudine, tenofovir disoproxil, entecavir, adefovir dipivoxil, alfafenone, Alloferon, simon interleukin, clevudine, emtricitabine, faplovir, interferon, calamine CP, intefene, interferon alpha-1 b, interferon alpha-2 a, interferon beta-1 a, interferon alpha-2, interleukin-2, mevotil, nitazoxanide, peginterferon alpha-2 a, ribavirin, roscovitine-a, cizopyran, Euforavac, azapril, phposphazid, heissav, interferon alpha-2 b, levamisole, or propagum.
In another aspect, the invention also provides the use of the compound or the pharmaceutical composition in the preparation of a medicament for preventing, treating or alleviating a viral disease in a patient.
In some embodiments, the use of the invention, wherein the viral disease is hepatitis b infection or a disease caused by hepatitis b infection.
In still other embodiments, the use of the present invention, wherein the disease caused by hepatitis B infection is liver cirrhosis or hepatocellular carcinoma.
In another aspect, the compounds of the present invention or the pharmaceutical compositions are used in the manufacture of a medicament for preventing, treating or ameliorating a viral disease in a patient.
In some embodiments, the compound of the present invention or the pharmaceutical composition is used, wherein the viral disease is hepatitis b infection or a disease caused by hepatitis b infection.
In still other embodiments, the use of the compound of the present invention or the pharmaceutical composition, wherein the disease caused by hepatitis b infection is liver cirrhosis or hepatocellular carcinoma.
In another aspect, the invention relates to a method of preventing, treating or ameliorating a viral disease in a patient, wherein the method comprises administering to the patient an effective amount of a compound or pharmaceutical composition of the invention that is pharmaceutically acceptable.
In some embodiments, the method of the invention, wherein the viral disease is hepatitis b infection or a disease caused by hepatitis b infection.
In still other embodiments, the method of the present invention, wherein the disease caused by hepatitis B infection is liver cirrhosis or hepatocellular carcinoma.
In another aspect, the invention relates to the use of the compound or the pharmaceutical composition for the preparation of a medicament for preventing, treating or alleviating hepatitis b disease in a patient.
Another aspect of the invention relates to a method of preventing, treating or ameliorating HBV disorders in a patient, comprising administering to the patient a pharmaceutically acceptable effective amount of a compound of the invention.
Another aspect of the invention relates to a method of preventing, treating or ameliorating HBV disorders in a patient, comprising administering to the patient a pharmaceutically acceptable effective amount of a pharmaceutical composition comprising a compound of the invention.
Another aspect of the invention relates to the use of a compound of the invention for the manufacture of a medicament for the prevention, treatment or treatment of HBV disorders in a patient and for lessening the severity thereof.
Another aspect of the present invention relates to the use of a pharmaceutical composition comprising a compound of the present invention for the manufacture of a medicament for the prevention, treatment or amelioration of HBV disorders in a patient and the severity thereof.
In some embodiments, the organism is a mammal, and in other embodiments, the organism is a human. In other embodiments, the method further comprises contacting the kinase with an HBV therapeutic agent.
Another aspect of the invention relates to a method of inhibiting HBV infection comprising contacting a cell with a compound or composition of the invention in an amount effective to inhibit HBV. In other embodiments, the method further comprises contacting the cell with another HBV therapeutic agent.
Another aspect of the present invention relates to the treatment of HBV disease in a patient, comprising administering to the patient a compound of the present invention or a composition thereof in a dosage effective to treat the disease. In other embodiments, the method further comprises administering an additional HBV therapeutic agent.
Another aspect of the present invention relates to a method of inhibiting HBV infection in a patient, comprising administering to the patient a compound of the present invention or a composition thereof in an amount effective to treat the patient in need thereof. In other embodiments, the method further comprises administering a dose of an additional HBV therapeutic agent.
Another aspect of the invention relates to methods for the preparation, isolation and purification of compounds encompassed by formula (I) or formula (Ia).
The invention also encompasses the use of the compounds of the invention and pharmaceutically acceptable salts thereof for the manufacture of a pharmaceutical product effective in inhibiting HBV infection, including those described herein. The application of the compound of the invention in the production of the drugs for effectively inhibiting HBV infection. The compounds of the invention are also useful in the manufacture of a medicament for alleviating, preventing, controlling or treating a condition of hepatitis b in a patient. The present invention encompasses pharmaceutical compositions comprising a therapeutically effective amount of a compound represented by formula (I) or (Ia) in association with at least one pharmaceutically acceptable carrier, adjuvant or diluent.
The invention also encompasses a method of effectively inhibiting HBV infection, or a disease susceptible to such a condition, comprising treating a patient with a therapeutically effective amount of a compound represented by formula (I) or (Ia).
Unless otherwise indicated, all stereoisomers, geometric isomers, tautomers, nitroxides, hydrates, solvates, metabolites, salts and pharmaceutically acceptable prodrugs of the compounds of the invention are within the scope of the present invention.
In particular, the salts are pharmaceutically acceptable salts. The term "pharmaceutically acceptable" includes materials or compositions which must be compatible chemically or toxicologically, with the other components comprising the formulation, and with the mammal being treated.
Salts of the compounds of the present invention also include, but are not necessarily pharmaceutically acceptable salts of intermediates used in the preparation or purification of the compounds of formula (I) or (Ia) or isolated enantiomers of the compounds of formula (I) or (Ia).
If the compounds of the invention are basic, the desired salts may be prepared by any suitable method provided in the literature, for example, using inorganic acids such as hydrochloric, hydrobromic, sulfuric, nitric, and phosphoric acids, and the like. Or using organic acids such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, malic acid, 2-hydroxypropionic acid, citric acid, oxalic acid, glycolic acid, and salicylic acid; pyranonic acids, such as glucuronic acid and galacturonic acid; alpha-hydroxy acids such as citric acid and tartaric acid; amino acids such as aspartic acid and glutamic acid; aromatic acids such as benzoic acid and cinnamic acid; sulfonic acids such as p-toluenesulfonic acid, benzenesulfonic acid, methanesulfonic acid, ethanesulfonic acid, trifluoromethanesulfonic acid, and the like, or combinations thereof.
If the compounds of the invention are acidic, the desired salts can be prepared by suitable methods, e.g., using inorganic or organic bases, such as ammonia (primary, secondary, tertiary), alkali metal hydroxides, ammonium, N + (R 14 ) 4 Salts and alkaline earth metal hydroxides, and the like. Suitable salts include, but are not limited to, organic salts derived from amino acids, such as glycine and arginine, ammonia, such as primary, secondary and tertiary amines, N + (R 14 ) 4 Salts, e.g. R 14 Is H, C 1-4 Alkyl radical, C 6-10 Aryl radical, C 6-10 Aryl radical C 1-4 Alkyl, etc., and cyclic amines such as piperidine, morpholine, piperazine, etc., and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum, and lithium. Also included are suitable, non-toxic ammonium, quaternary ammonium salts and amine cations resistant to formation of counterions, e.g., halides, hydroxides, carboxylates, sulfates, phosphates, nitrates, sulfates,C 1-8 Sulfonates and aromatic sulfonates.
Compositions, formulations, administration of the compounds of the invention and uses of the compounds and compositions
According to another aspect, the pharmaceutical compositions of the invention may be characterized by comprising a compound of formula (I) or (Ia), a compound of the invention as set forth in the examples, or a compound of the examples, and a pharmaceutically acceptable carrier, adjuvant, or vehicle. The compounds in the composition can effectively inhibit hepatitis B virus, and are suitable for treating diseases caused by virus, particularly acute and chronic persistent HBV virus infection, HBV-caused chronic viral diseases can cause serious morbidity, and chronic hepatitis B virus infection can cause cirrhosis and/or hepatocellular carcinoma in many cases.
For the compounds of the invention, mention may be made of the indicator regions, for example: treatment of acute and chronic viral infections, which may lead to infectious hepatitis, e.g., hepatitis B virus infection. The compounds of the invention are particularly suitable for the treatment of chronic hepatitis B infections and acute and chronic hepatitis B virus infections.
The invention encompasses pharmaceutical preparations which, in addition to a non-toxic, inert pharmaceutically suitable carrier, also contain one or more compounds (I) or (Ia) or compositions according to the invention or one or more active ingredients (I) or (Ia) or compositions according to the invention.
The above pharmaceutical preparation may also contain other active pharmaceutical ingredients than compound (I) or (Ia).
The compounds of the invention exist in free form or, where appropriate, as pharmaceutically acceptable derivatives. According to the present invention, pharmaceutically acceptable derivatives include, but are not limited to, pharmaceutically acceptable prodrugs, salts, esters, salts of esters, or any other adduct or derivative capable of being administered directly or indirectly to a patient in need thereof, compounds described in other aspects of the invention, metabolites thereof, or residues thereof.
As described herein, the pharmaceutical composition of the present invention comprises any one of the compounds of formula (I) or (Ia) of the present invention, and further comprises pharmaceutically acceptable excipients, such as any solvent, solid excipient, diluent, binder, disintegrant, or other liquid excipient, dispersant, flavoring agent or suspending agent, surfactant, isotonizing agent, thickening agent, emulsifier, preservative, solid binder or lubricant, and the like, as used herein, suitable for the specific intended dosage form. As described in the following documents: in Remington, The Science and Practice of Pharmacy,21st edition,2005, ed.D.B.Troy, Lippincott Williams & Wilkins, Philadelphia, and Encyclopedia of Pharmaceutical Technology, eds.J.Swarbrick and J.C.Boylan, 1988. Annu 1999, Marcel Dekker, New York, taken together with The disclosure of this document, indicates that different adjuvants can be used In The preparation of pharmaceutically acceptable compositions and their well-known methods of preparation. Except insofar as any conventional adjuvant is incompatible with the compounds of the invention, e.g., any adverse biological effect produced or interaction in a deleterious manner with any other component of a pharmaceutically acceptable composition, their use is contemplated by the present invention.
Substances that may serve as pharmaceutically acceptable excipients include, but are not limited to, ion exchangers; aluminum; aluminum stearate; lecithin; serum proteins, such as human serum albumin; buffer substances such as phosphates; glycine; sorbic acid; potassium sorbate; partial glyceride mixtures of saturated vegetable fatty acids; water; salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts; colloidal silica; magnesium trisilicate; polyvinylpyrrolidone; polyacrylate esters; a wax; polyethylene-polyoxypropylene-blocking polymers; lanolin; sugars, such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; gum powder; malt; gelatin; talc powder; adjuvants such as cocoa butter and suppository wax; oils such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol and polyethylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic salt; ringer's solution; ethanol; phosphoric acid buffer solution; and other non-toxic suitable lubricants such as sodium lauryl sulfate and magnesium stearate; a colorant; a release agent; coating the coating material; a sweetener; a flavoring agent; a fragrance; preservatives and antioxidants.
Pharmaceutical compositions of the compounds of the invention may be administered in any of the following ways: oral administration, inhalation by spray, topical administration, rectal administration, nasal administration, topical administration, vaginal administration, parenteral administration such as subcutaneous, intravenous, intramuscular, intraperitoneal, intrathecal, intraventricular, intrasternal, or intracranial injection or infusion, or by means of a reservoir of external value. Preferred modes of administration are oral, intramuscular, intraperitoneal or intravenous.
The compounds of the present invention or compositions containing them which are pharmaceutically acceptable may be administered in unit dosage form. The administration dosage form can be liquid dosage form or solid dosage form. The liquid dosage forms can be true solutions, colloids, microparticles, and suspensions. Other dosage forms such as tablet, capsule, dripping pill, aerosol, pill, powder, solution, suspension, emulsion, granule, suppository, lyophilized powder for injection, clathrate, implant, patch, liniment, etc.
Oral tablets and capsules may contain excipients such as binding agents, for example syrup, acacia, sorbitol, tragacanth, or polyvinylpyrrolidone; fillers, such as lactose, sucrose, corn starch, calcium phosphate, sorbitol, glycine; lubricants, such as magnesium stearate, talc, polyethylene glycol, silica; disintegrants, such as potato starch; or acceptable humectants such as sodium lauryl sulfate. The tablets may be coated by methods known in the art of pharmacy.
Oral liquids may be formulated as suspensions in aqueous oil, solutions, emulsions, syrups or elixirs, or may be formulated as a dry product for constitution with water or other suitable vehicle before use. Such liquid preparations may contain conventional additives such as suspending agents, sorbitol, cellulose methyl ether, glucose syrup, gelatin, hydroxyethyl cellulose, carboxymethyl cellulose, aluminum stearate gelatin, hydrogenated edible fats and oils, emulsifying agents such as lecithin, sorbitan monooleate, gum arabic; or a non-aqueous carrier (which may comprise an edible oil), such as almond oil, an oil such as glycerol, ethylene glycol, or ethanol; preservatives, e.g. methyl or propyl p-hydroxybenzoates, sorbic acid. Flavoring or coloring agents may be added if desired.
Suppositories may contain conventional suppository bases such as cocoa butter or other glycerides.
For parenteral administration, the liquid dosage forms are generally prepared from the compound and a sterile vehicle. The carrier is preferably water. The compound can be dissolved in the carrier or made into suspension solution according to the different carrier and drug concentration, when making injection solution, the compound is dissolved in water, filtered and sterilized, and then filled into sealed bottle or ampoule.
When applied topically to the skin, the compounds of the present invention may be formulated in the form of a suitable ointment, lotion, or cream in which the active ingredient is suspended or dissolved in one or more carriers, which may be used in ointment formulations including, but not limited to: mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyethylene oxide, polypropylene oxide, emulsifying wax and water; lotions and creams may employ carriers including, but not limited to: mineral oil, sorbitan monostearate, tween 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
In general, it has proven advantageous, both in human medicine and in veterinary medicine, to administer the active compounds according to the invention in a total amount of from about 0.5 to 500mg, preferably from 1 to 100mg, per kg of body weight per 24 hours, if appropriate in divided single doses, in order to achieve the desired effect. The amount of active compound contained in a single dose is preferably about 1 to 80mg, more preferably 1 to 50mg per kg body weight, but may be varied from the dosages described above, i.e. depending on the type and body weight of the subject to be treated, the nature and severity of the disease, the type of preparation and the mode of administration of the drug, and the period or interval of administration.
The pharmaceutical composition provided by the invention also comprises an anti-HBV medicament. Wherein the anti-HBV drug is an HBV polymerase inhibitor, an immunomodulator or an interferon.
The HBV drugs include lamivudine, telbivudine, tenofovir disoproxil, entecavir, adefovir dipivoxil, alfafenone, Alloferon, simon interleukin, cladribine, emtricitabine, faprolivir, interferon, calamine CP, intefine, interferon alpha-1 b, interferon alpha-2 a, interferon beta-1 a, interferon alpha-2, interleukin-2, mequitylate, nitazoxanide, peginterferon alpha-2 a, ribavirin, roscovarin-A, Sizopyran, Euforavac, ritolimod, Phosphazid, Heplinav, interferon alpha-2 b, levamisole or propafegermanium.
Another aspect of the present invention relates to the use of a compound or pharmaceutical composition of the present invention for the manufacture of a medicament for the prevention, treatment or amelioration of hepatitis B disease in a patient, which comprises administering to the patient a pharmaceutically acceptable effective amount. Hepatitis B disease refers to liver diseases caused by hepatitis B virus infection or hepatitis B infection, including acute hepatitis, chronic hepatitis, liver cirrhosis and stem cell carcinoma. Acute hepatitis b virus infection may be asymptomatic or manifest as acute hepatitis symptoms. Patients with chronic viral infections have active disease that can progress to cirrhosis and liver cancer.
The anti-HBV agent may be administered separately from a composition comprising a compound of the invention as part of a multiple dosing regimen. Alternatively, those agents may be part of a single dosage form, mixed together with the compounds of the present invention to form a single composition. If administered as part of a multiple dosing regimen, the two active agents can be delivered to each other simultaneously, sequentially or over a period of time, to achieve the desired agent activity.
The amount of compound and composition that can be combined with a carrier material to produce a single dosage form (those containing a composition like that described herein) will vary depending upon the indication and the particular mode of administration. Normally, the amount of the composition of the invention will not exceed the amount of the composition normally administered containing as the only active agent. In another aspect, the amount of the presently disclosed compositions ranges from about 50% to 100% of the normal amount of the presently disclosed compositions, including the agent as the sole active therapeutic agent. In those compositions that are included, the compositions will act synergistically with the compounds of the present invention.
The compound of the invention shows stronger antiviral effect. The compounds have unexpected antiviral activity on HBV, and are suitable for treating various diseases caused by viruses, especially diseases caused by acute and chronic persistent HBV virus infection. Chronic viral diseases caused by HBV can lead to a variety of syndromes of varying severity, and chronic hepatitis b virus infection is known to cause cirrhosis and/or hepatocellular carcinoma.
Examples of indications that can be treated with the compounds of the invention are: treating acute and chronic viral infections that can lead to infectious hepatitis, such as heterohepatitis virus infection. Particularly preferred are the treatment of chronic hepatitis B infection and the treatment of acute hepatitis B virus infection.
The invention also relates to the use of the compounds and compositions of the invention for the preparation of medicaments for the treatment and prophylaxis of viral diseases, in particular hepatitis b.
General synthetic methods
In general, the compounds of the invention may be prepared by the methods described herein, wherein the substituents are as defined for formula (I) or (Ia), unless otherwise indicated. The following reaction schemes and examples serve to further illustrate the context of the invention.
Those skilled in the art will recognize that: the chemical reactions described herein may be used to suitably prepare a number of other compounds of the invention, and other methods for preparing the compounds of the invention are considered to be within the scope of the invention. For example, the synthesis of those non-exemplified compounds according to the present invention can be successfully accomplished by those skilled in the art by modification, such as appropriate protection of interfering groups, by the use of other known reagents in addition to those described herein, or by some routine modification of reaction conditions. In addition, the reactions disclosed herein or known reaction conditions are also recognized as being applicable to the preparation of other compounds of the present invention.
In the examples described below, all temperatures are given in degrees Celsius (. degree. C.) unless otherwise indicated. Reagents were purchased from commercial suppliers such as Aldrich Chemical Company, Arco Chemical Company and Alfa Chemical Company and were used without further purification unless otherwise indicated. General reagents were purchased from Shantou Wen Long chemical reagent factory, Guangdong Guanghua chemical reagent factory, Guangzhou chemical reagent factory, Tianjin HaoLiyu Chemicals Co., Ltd, Qingdao Tenglong chemical reagent Co., Ltd, and Qingdao Kaseiki chemical plant.
The column used silica gel column, silica gel (200-300 mesh) purchased from Qingdao oceanic plant. Nuclear magnetic resonance spectroscopy with CDC1 3 ,d 6 -DMSO,CD 3 OD or d 6 Acetone as solvent (reported in ppm) with TMS (0ppm) or chloroform (7.25ppm) as reference standard. When multiple peaks occur, the following abbreviations will be used: s (singleton), d (doublet), t (triplet ), m (multiplet, multiplet), br (broadpede, broad), dd (doublet of doublets, quartet), dt (doublet of triplets), br. Coupling constant J, in Hertz (Hz).
Low resolution Mass Spectral (MS) data were measured by an Agilent6320 series LC-MS spectrometer equipped with a G1312A binary pump and a G1316A TCC (column temperature maintained at 30 ℃), a G1329A autosampler and a G1315B DAD detector were applied for analysis, and an ESI source was applied to the LC-MS spectrometer.
Low resolution Mass Spectral (MS) data were determined by Agilent 6120 series LC-MS spectrometer equipped with a G1311A quaternary pump and a G1316A TCC (column temperature maintained at 30 ℃), a G1329A autosampler and a G1315D DAD detector were used for analysis, and an ESI source was used for the LC-MS spectrometer.
Both spectrometers were equipped with an Agilent Zorbax SB-C18 column, 2.1X 30mm, 5 μm. The injection volume is determined by the sample concentration; the flow rate is 0.6 mL/min; peaks of HPLC were recorded by UV-Vis wavelength at 210nm and 254 nm. The mobile phases were 0.1% formic acid in acetonitrile (phase a) and 0.1% formic acid in ultrapure water (phase B). Gradient elution conditions are shown in table 1:
table 1: gradient elution conditions
Time (min) A(CH 3 CN,0.1%HCOOH) B(H 2 O,0.1%HCOOH)
0-3 5-100 95-0
3-6 100 0
6-6.1 100-5 0-95
6.1-8 5 95
The compound purification was assessed by Agilent 1100 series High Performance Liquid Chromatography (HPLC) with UV detection at 210nm and 254nm on a Zorbax SB-C18 column, 2.1X 30mm, 4 μm, 10min, flow rate 0.6mL/min, 5-95% (0.1% formic acid in acetonitrile) (0.1% formic acid in water), the column temperature was maintained at 40 ℃.
The following acronyms are used throughout the invention:
MeCN,CH 3 CN acetonitrile
DCM,CH 2 Cl 2 Methylene dichloride
CHCl 3 Chloroform, trichloroMethane
CDC1 3 Deuterated chloroform
CCl 4 Carbon tetrachloride
Boc-t-butyloxycarbonyl group
PE Petroleum Ether
EtOAc, EA ethyl acetate
EtOH ethanol
TCDI thiocarbonyldiimidazole
CDI carbonyl imidazole
DBU 1, 5-diazabicyclo [5.4.0] undec-5-ene
HCl hydrogen chloride
K 2 CO 3 Potassium carbonate
NaHCO 3 Sodium bicarbonate
NaOH sodium hydroxide
NaCl sodium chloride
Na 2 SO 4 Sodium sulfate
Et 3 N, TEA Triethylamine
NBS N-bromosuccinimide
D 2 Heavy O water
H 2 O water
mL of
RT, RT Room temperature
Rt Retention time
H 2 Hydrogen gas
HCl/EA hydrogen chloride in ethyl acetate
HOAt 1-hydroxy-7-azobenzotriazol
DIPEA N, N-diisopropylethylamine
DCC N, N' -dicyclohexylcarbodiimide
DMF dimethyl formamide
THF tetrahydrofuran
DMSO dimethyl sulfoxide
CuCN cuprous cyanide
CH 3 OH methanol
N 2 Nitrogen gas
NH 4 Cl ammonium chloride
Ac 2 O acetic anhydride
t 1/2 Half life period
AUC area under the time curve
Vss steady state apparent distribution volume
Clearance of CL, clearance
Bioavailability of F, absolute bioavailalability
Dose of Dose
T max Time to peak
C max Maximum concentration of
hr * ng/mL blood concentration time
Synthesis method
The following synthetic schemes set forth the experimental procedures for preparing the compounds disclosed in the present invention. Wherein each R is 1 、R 2 、R 4 、R 5 N, m and f have the meanings as described in the invention.
Synthesis scheme 1
Figure BDA0001397442500000331
Compound 6a can be prepared by scheme 1 by first reacting 1a with Boc anhydride to give compound 2a, reacting compound 2a with ethyl chloroformate to give 3a under the action of base (e.g., TEA, etc.), reacting compound 3a with aqueous ammonia to give compound 4a, reacting compound 4a with oxalyl chloride to give 5a, and finally reacting compound 5a with sodium azide and ammonium chloride to give intermediate compound 6 a.
Synthesis scheme 2
Figure BDA0001397442500000341
Compound 11a can be prepared according to scheme 2 by first reacting 8a with hydrazine hydrate to give compound 9a, and then reacting compound 9a with triphosgene under basic conditions (e.g., DIPEA) to give intermediate compound 10 a.
Synthesis scheme 3
Figure BDA0001397442500000342
Compound 13a can be prepared by scheme 3 by first reacting compound 5a with hydroxylamine hydrochloride in the presence of sodium bicarbonate to give compound 12a, and then reacting compound 12a with thiocarbonyldiimidazole in the presence of a catalyst (e.g., DBU, etc.) to give intermediate compound 13 a.
Synthesis scheme 4
Figure BDA0001397442500000343
Compound 14a can be prepared by synthesis scheme 4 by first reacting compound 14a with thiocarbonyldiimidazole to form intermediate compound b, and then reacting compound b with boron trifluoride in diethyl etherate to form intermediate compound 14 a.
Synthesis scheme 5
Figure BDA0001397442500000344
Compound 15a can be prepared by synthesis scheme 5, and compound 12a is reacted with carbonyldiimidazole to form intermediate compound 15 a.
Synthesis scheme 6
Figure BDA0001397442500000345
Compound 19a can be prepared by scheme 6, intermediate 16a is deprotected from the Boc protecting group by the action of an acid (e.g. hydrogen chloride in ethyl acetate) to give compound 17a, and compound 17a is reacted with compound 18a (compound 18a can be prepared by reference to method 2 and embodiments of CN104650068 or scheme 7 and examples in WO 2015144093) under basic conditions (e.g. TEA, potassium carbonate, etc.) to give anti-HBV active compound 19 a.
Examples
Example 1: (R) -6- (((R) -3- (1H-tetrazol-5-yl) morpholine) methyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester
Figure BDA0001397442500000351
The synthetic route is as follows:
Figure BDA0001397442500000352
step 1) (3S) -3-carbamoylmorpholine-4-carboxylic acid tert-butyl ester
(S) -4- (di-tert-butyl dicarbonate) morpholine-3-carboxylic acid (2.0g,8.6mmol) and THF (20mL) were added in this order to a 250mL three-necked flask, and after the mixture was stirred at room temperature, triethylamine (1.3g,13mmol) was added, and the reaction system was cooled to 0 ℃ and then ethyl chloroformate (1.2g,11mmol) was added. The reaction mixture was stirred for 20 minutes with warming and then stirred for 2 hours at room temperature. After the reaction, the reaction solution was cooled to 0 ℃, and then ammonia (8mL) was added thereto, stirred for 10 minutes under heat preservation, and then heated to room temperature and stirred for 3 hours. The solvent was evaporated under reduced pressure, water (50mL) was added to the obtained residue, followed by extraction with ethyl acetate (50mL × 2), and the combined organic phases were washed with saturated brine (50mL × 2), and then concentrated under reduced pressure to give the title compound as a colorless oil (1.75g, 89%).
MS-ESI:(ESI,pos.ion)m/z:131.3[M+H-100] +
Step 2) (R) -N-Boc-3-cyanomorpholine
To a 50mL reaction flask were added DMF (0.2g,2.74mmol) and acetonitrile (8mL) in that order, the mixture was cooled to 0 deg.C, and then a solution of oxalyl chloride (0.34g,2.7mmol) in dichloromethane (2mL) was added dropwise, after which the mixture was stirred for an additional 20 minutes while maintaining the temperature. Subsequently, a solution of tert-butyl (3S) -3-carbamoylmorpholine-4-carboxylate (0.3g,1.3mmol) and pyridine (0.12g,1.52mmol) in acetonitrile (3mL) was added, and after the addition was complete, the resulting mixture was stirred at room temperature for 1 hour. After completion of the reaction, the reaction was quenched with water, extracted with ethyl acetate (25 mL. times.2), and the combined organic phases were washed with saturated brine (30 mL. times.2), dried over anhydrous sodium sulfate, and the solvent was evaporated under reduced pressure to give the title compound as a white solid (0.21g, 75.9%).
MS-ESI:(ESI,pos.ion)m/z:213.2[M+H] + ,235.2[M+Na] +
Step 3) (R) -3- (1H-tetrazol-5-yl) morpholine-4-carboxylic acid tert-butyl ester
(R) -N-Boc-3-cyanomorpholine (0.2g,0.94mmol) and DMF (2mL) were added sequentially to a 50mL single-neck flask, and after the mixture was completely dissolved by stirring at room temperature, sodium azide (0.1g,1.54mmol) and NH were added 4 Cl (74mg,1.4mmol), under nitrogen, the resulting reaction mixture was heated to 100 ℃ and stirred for 6 hours, then cooled to room temperature and stirred overnight. After completion of the reaction, the reaction was quenched by addition of water (20mL), adjusted to pH 3 with dilute hydrochloric acid, extracted with ethyl acetate (25 mL. times.3), and the combined organic phases were washed with saturated brine (30 mL. times.3), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated by evaporation under reduced pressure to give the title compound as a colorless oil (0.15g, 62.4%).
MS-ESI:(ESI,neg.ion)m/z:254.2[M-H]
Step 4) (R) -3- (1H-tetrazole-5-yl) morpholine hydrochloride
To a 100mL single-neck flask were added (R) -3- (1H-tetrazol-5-yl) morpholine-4-carboxylic acid tert-butyl ester (0.15g,0.59mmol) and HCl in ethyl acetate (4mol/L,15mL) in that order, and the reaction mixture was stirred at room temperature for 2 hours. After the reaction was complete, filtration was carried out and the filter cake was washed with a small amount of ethyl acetate and then dried under vacuum at 50 ℃ to give the title compound as a white solid (0.11g, 82.1%).
Step 5 methyl (R) -6- (((R) -3- (1H-tetrazol-5-yl) morpholine) methyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate
To a 50mL single-necked flask were added in sequence (R) -methyl 6- (bromomethyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (0.21g,0.48mmol), K 2 CO 3 (0.17g,1.2mmol), (R) -3- (1H-tetrazol-5-yl) morpholine hydrochloride (0.11g,0.48mmol) andaqueous ethanol (8mL) and the reaction mixture was stirred under nitrogen at room temperature overnight. The filtrate was filtered, concentrated under reduced pressure, diluted with water (25mL), the pH of the resulting solution was adjusted to about 3 with dilute hydrochloric acid, and extracted with ethyl acetate (30 mL. times.2). The combined organic phases were dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated by evaporation under reduced pressure, and the resulting residue was purified by silica gel column chromatography (PE/EA (V/V) ═ 3/1) to afford the title compound as a yellow solid (0.18g, 71.91%).
MS-ESI:(ESI,pos.ion)m/z:519.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ(ppm):9.70(s,1H),8.06(d,,1H),7.96(d,1H),7.43–7.38(m,2H),7.17(td,1H),5.97(s,1H),4.38(s,1H),4.04-3.95(m,3H),3.83–3.77(m,2H),3.47(s,3H),3.37–3.27(m,1H),3.03–2.93(m,1H),2.63–2.57(m,1H)。
Example 2: (R) -4- (2-chloro-4-fluorophenyl) -6- (((S) -3- (5-oxo-4, 5-dihydro-1, 3, 4-oxadiazol-2-yl) morpholine) methyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester
Figure BDA0001397442500000361
The synthetic route is as follows:
Figure BDA0001397442500000371
step 1) (S) -3- (formylhydrazine) morpholine-4-carboxylic acid tert-butyl ester
To a 100mL single-neck flask were added (S) -N-Boc-3-morpholinocarboxylic acid methyl ester (2.0g,8.2mmol), methanol (20mL) and hydrazine hydrate (1.2g,24mmol) in that order, and the mixture was heated to 80 ℃ and stirred overnight. After completion of the reaction, the reaction mixture was concentrated under reduced pressure, 50mL each of water and ethyl acetate was added to the residue, the mixture was allowed to stand, the layers were separated, the aqueous layer was extracted with EA (50mL × 3), the combined organic layers were washed with saturated brine, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (PE/EA (V/V) ═ 15/1) to give the title compound as a white solid (1.4g, 70%).
MS-ESI:(ESI,pos.ion)m/z:146.2[M+H-100] +
Step 2) (S) -3- (5-oxo-4, 5-dihydro-1, 3, 4-oxadiazol-2-yl) morpholine-4-carboxylic acid tert-butyl ester
To a 100mL one-neck flask were added (S) -N-Boc-3-formylhydrazine-morpholine (0.9g,3.6693mmol) and THF (40mL) in that order, and the mixture was stirred at room temperature, diisopropylethylamine (1.2g,9.3mmol) was added and, after complete dissolution of the solids, triphosgene (0.55g,1.853mmol) in THF (6mL) was added slowly. After the addition was complete, the reaction mixture was warmed to 80 ℃ and stirred for 4 hours. After completion of the reaction, the temperature was lowered to room temperature, and the reaction was quenched with saturated aqueous sodium bicarbonate solution (200mL), and the resulting mixture was extracted with ethyl acetate (100 mL. times.3). The combined organic phases were washed with saturated brine (100mL × 2), dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (pure EA) to give the title compound as a colorless oil (0.78g, 78.36%).
MS-ESI:(ESI,pos.ion)m/z:172.2[M+H-100] + ,294.2[M+Na] +
Step 3) (S) -5- (morpholin-3-yl) -1,3, 4-oxadiazol-2 (3H) -one hydrochloride
To a 50mL single neck flask were added tert-butyl (S) -3- (5-oxo-4, 5-dihydro-1, 3, 4-oxadiazol-2-yl) morpholine-4-carboxylate (0.25g,0.92mmol) and ethyl acetate (5mL) in that order, and after the mixture was completely dissolved by stirring at room temperature, a solution of HCl in ethyl acetate (4mol/L,10mL) was added and after the addition, a white solid gradually precipitated and stirring was continued for 1 hour, followed by filtration. The filter cake was washed with ethyl acetate and dried in vacuo to afford the title compound as a white solid (0.11g, 57.5%).
MS-ESI:(ESI,pos.ion)m/z:172.1[M+H] +
Step 4) (R) -4- (2-chloro-4-fluorophenyl) -6- (((S) -3- (5-oxo-4, 5-dihydro-1, 3, 4-oxadiazol-2-yl) morpholine) methyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester
To a 50mL single-necked flask were added in sequence (R) -methyl 6- (bromomethyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (0.64g,1.439mmol), K 2 CO 3 (0.5g,3.62mmol)、(S)-5-(Morpholin-3-yl) -1,3, 4-oxadiazol-2 (3H) -one hydrochloride (0.3g,1.44mmol) and absolute ethanol (20mL), the reaction mixture was stirred overnight at room temperature under nitrogen. After completion of the reaction, the reaction mixture was filtered, and the filtrate was concentrated under reduced pressure. The resulting residue was diluted with 80mL of each of water and ethyl acetate, and the resulting solution was adjusted to pH 7 with dilute hydrochloric acid, allowed to stand, separated into layers, and the aqueous phase was extracted with ethyl acetate (50mL × 2). The combined organic phases were dried over anhydrous sodium sulfate, filtered and the filtrate was concentrated under reduced pressure. The residue was chromatographed by preparative thin layer chromatography (DCM/CH) 3 OH (V/V) ═ 15/1) purified to give the title compound as a yellow solid (0.18g, 23.28%).
MS-ESI:(ESI,pos.ion)m/z:535.1[M+H] +
1 HNMR(400MHz,DMSO-d 6 )δ(ppm):12.35(s,1H),9.62(s,1H),8.05(d,1H),7.95(d,1H),7.43–7.38(m,2H),7.17(td,1H),6.02(s,1H),4.26(d,1H),4.03(d,1H),3.95-3.82(m,3H),3.77–3.73(m,2H),3.50(s,3H),3.03–3.00(m,1H),2.56–2.53(m,1H)。
Example 3: (R) -6- (((R) -2- (2- (1H-tetrazol-5-yl) ethyl) morpholine) methyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester
Figure BDA0001397442500000381
The synthetic route is as follows:
Figure BDA0001397442500000382
step 1: (R) -N-Boc-2- (3-amino-3-oxopropyl) morpholine
A500 mL three-necked flask was charged with (R) -N-Boc-3-morpholinopropionic acid (12.0g,46.32mmol) and tetrahydrofuran (300mL), the starting materials were dissolved and stirred well, then cooled to 0 ℃ and, under nitrogen, triethylamine (7.0g,69.18mmol) and ethyl chloroformate (5.7g,60.32mmol) were added in that order. The reaction mixture was stirred for 20 minutes at the same temperature, then warmed to 25 ℃ and stirred for 2 hours, then cooled to 0 ℃, ammonia (40mL) was added, stirred for 10 minutes at the same temperature, warmed to 25 ℃ again and stirred for 3 hours. After completion of the reaction, the reaction solution was concentrated, and water (250mL) was added to the residue to dilute, the aqueous layer was extracted with ethyl acetate (250mL × 3), and the combined organic phases were washed with saturated brine (250mL × 2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to obtain the title compound as a white solid (10g, 83.6%).
MS-ESI:(ESI,pos.ion)m/z:159.10[M+H-100] +
Step 2: (R) -N-Boc- (2-cyanoethyl) morpholine
DMF (170mg,2.33mmol) and acetonitrile (8mL) were added sequentially to a 50mL reaction flask and, after stirring, the temperature was reduced to 0 ℃ and a solution of oxalyl chloride (0.30g,2.42mmol) in dichloromethane (2mL) was added dropwise, after which stirring was maintained for 20min, followed by addition of a solution of (R) -N-Boc-3-morpholinopropionamide (300mg,1.16mmol) and pyridine (110mg,1.39mmol) in acetonitrile (3mL) and, after addition, heating to 25 ℃ and stirring continued for 1 h. After completion of the reaction, quenched with water (20mL), the aqueous layer extracted with ethyl acetate (25 mL. times.2), and the combined organic layers were washed successively with aqueous hydrochloric acid (1%) and saturated brine (30 mL. times.2), dried over anhydrous sodium sulfate, national standard and concentrated under reduced pressure to give the title compound as a pale yellow oil (250mg, 89.6%).
MS-ESI:(ESI,pos.ion)m/z:263.10[M+Na] +
And step 3: (R) -N-Boc-2- (2- (1H-tetrazol-5-yl) ethyl) morpholine
(R) -N-Boc- (2-cyanoethyl) morpholine (400mg,1.66mmol) and DMF (6mL) were added to a 50mL single-neck flask in this order, and after complete dissolution with stirring, sodium azide (860mg,13.23mmol) and NH were added in this order 4 Cl (705mg,13.30mmol), the reaction mixture was warmed to 100 ℃ under nitrogen and stirred for 20 h with incubation. After completion of the reaction, the reaction system was cooled to 25 ℃, and then pH was adjusted to 3 with dilute hydrochloric acid (2M), water and ethyl acetate were added to dilute each (50mL), the layers were separated by extraction, the aqueous phase was extracted with ethyl acetate (25mL × 2), the combined organic phases were washed with saturated brine (50mL × 3), dried over anhydrous sodium sulfate, and concentrated, and the resulting residue was purified by silica gel column chromatography (PE/EA (V/V) ═ 1/1) to give the title compound as a colorless oil (137mg, 29%).
MS-ESI:(ESI,pos.ion)m/z:284.30[M+H] +
And 4, step 4: (R) -2- (2- (1H-tetrazol-5-yl) ethyl) morpholine hydrochloride
(R) -N-Boc-2- (2- (1H-tetrazol-5-yl) ethyl) morpholine (130mg,0.46mmol) and ethyl acetate (5mL) were added to a 50mL single vial, stirred and dissolved, and HCl in ethyl acetate (4mol/L,10mL) was added and, after addition, the reaction mixture was stirred for 3 hours at 25 ℃. After completion of the reaction, the reaction mixture was concentrated under reduced pressure to give the title compound as a brown oil (100mg, 99.2%).
And 5: (R) -6- (((R) -2- (2- (1H-tetrazol-5-yl) ethyl) morpholine) methyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester
To a 50mL single-necked flask were added (R) -methyl 6- (bromomethyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (250mg,0.56mmol) and potassium carbonate (183mg,1.32 mmol), (R) -2- (2- (1H-tetrazol-5-yl) ethyl) morpholine hydrochloride (100mg,0.45mmol) followed by ethanol (8 mL). The reaction mixture was stirred under nitrogen overnight at 25 ℃, then filtered, the filtrate was concentrated under reduced pressure, the residue was diluted with water (50mL) and ethyl acetate (50mL), the pH was adjusted to 4 with dilute hydrochloric acid (2M), the solution was separated, the aqueous phase was extracted with ethyl acetate (50mL × 2), the combined organic phases were dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated by evaporation under reduced pressure, and the resulting residue was purified by silica gel column chromatography (PE/EA (V/V) ═ 2/1) to give the title compound as a yellow solid (150mg, 60.24%).
MS-ESI:(ESI,pos.ion)m/z:547.1[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ15.93(s,1H),9.68(s,1H),7.95(d,J=3.4Hz,2H),7.49–7.32(m,2H),7.19–7.17(m,,1H),6.04(s,1H),3.95–3.84(m,3H),3.63–3.48(m,5H),3.00–2.91(m,2H),2.83–2.72(m,2H),2.38(s,1H),2.07(s,1H),1.88–1.78(m,2H)。
Example 4: (R) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -6- (((R) -2- (2- (5-thio-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) ethyl) morpholine) methyl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester
Figure BDA0001397442500000391
The synthetic route is as follows:
Figure BDA0001397442500000401
step 1: (R, Z) -N-Boc-2- (3-amino-3- (hydroxyimino) propyl) morpholine
To a 100mL single-neck flask were added (R) -N-Boc- (2-cyanoethyl) morpholine (2.3g,9.6mmol), hydroxylamine hydrochloride (1.0g,14.39mmol), NaHCO in that order 3 (1.3g,15.47mmol) and methanol (50mL), the reaction mixture was reacted at 60 ℃ for 12 h, then cooled to 25 ℃ and filtered, and the crude product was purified by silica gel column chromatography (EA) to afford the title compound as a white solid (2.1g, 80%).
MS-ESI:(ESI,pos.ion)m/z:274.10[M+H] +
Step 2: (R) -N-Boc-2- (2- (5-thio-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) ethyl) morpholine
To a 50mL one-necked flask were added (R, Z) -N-Boc-2- (3-amino-3- (hydroxyimino) propyl) morpholine (200mg,0.73mmol), thiocarbonyldiimidazole (260mg,1.46mmol), DBU (445mg,2.93mmol, 100% mass) and acetonitrile (10mL) in that order. The reaction mixture was blanketed with nitrogen and stirred at 25 ℃ for 12 hours. After completion of the reaction, the reaction system was concentrated under reduced pressure, and the residue was diluted with water (50mL), then adjusted to pH 3 with dilute hydrochloric acid (2M), and extracted with ethyl acetate (50 mL. times.3). The combined organic phases were washed with dilute hydrochloric acid (2M,50mL × 2), dried over anhydrous sodium sulfate, and concentrated to afford the title compound as a beige solid (200mg, 86.66%).
MS-ESI:(ESI,pos.ion)m/z:216.0[M+H-100] + ;338.1[M+Na] +
Step 3 (R) -3- (2- (morpholin-2-yl) ethyl) -1,2, 4-oxadiazole-5 (4H) -thione hydrochloride
To a 50mL single-necked flask were added (R) -N-Boc-2- (2- (5-thio-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) ethyl) morpholine (200mg,0.63mmol), ethyl acetate (5mL) in that order,the mixture was stirred to dissolve, and a solution of HCl in ethyl acetate (4mol/L,15mL) was added and, after addition, the reaction mixture was stirred for an additional 2 hours. After completion of the reaction, the reaction was concentrated under reduced pressure to give the title compound as a pale yellow oil (150mg, 93.97%). MS-ESI (ESI, pos.ion) m/z: 216.2[ M + H] +
And 4, step 4: (R) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -6- (((R) -2- (2- (5-thio-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) ethyl) morpholine) methyl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester
To a 50mL single-necked flask were added in this order (R) -6- (bromomethyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester (270mg,0.61mmol), potassium carbonate (200mg,1.45mmol), (R) -3- (2- (morpholin-2-yl) ethyl) -1,2, 4-oxadiazole-5 (4H) -thione hydrochloride (150mg,0.59mmol), and ethanol (10 mL). The reaction mixture was stirred under nitrogen at 25 ℃ for 12 hours, then filtered, the filtrate was concentrated under reduced pressure, the residue was diluted with water (30mL) and ethyl acetate (50mL), the pH of the system was adjusted to 4 with dilute hydrochloric acid (2M), the aqueous phase was extracted with ethyl acetate (50mL × 3), the combined organic phases were dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (PE/EA (V/V) ═ 1/1) to give the title compound as a yellow solid (180mg, 52.16%).
MS-ESI:(ESI,pos.ion)m/z:579.1[M+H] +
Example 5: (R) -4- (2-chloro-4-fluorophenyl) -6- (((R) -2- (2- (5-oxo-4, 5-dihydro-1, 3, 4-oxadiazol-2-yl) ethyl) morpholine) methyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester
Figure BDA0001397442500000411
The synthetic route is as follows:
Figure BDA0001397442500000412
step 1: (R) -N-Boc-2- (3-hydrazino-3-oxopropyl) morpholine
To a 100mL one-neck flask were added (R) -N-Boc-2- (3-methoxy-3-oxopropyl) morpholine (3.0g,11.02mmol) and methanol (20mL), and after stirring, hydrazine hydrate (4mL) was added. The reaction mixture was stirred at 60 ℃ for 12 hours, then cooled to 25 ℃ and concentrated, the residue was diluted with water (100mL), the aqueous layer was extracted with EA (100mL × 3), the combined organic phases were concentrated with drying, and the resulting residue was purified by silica gel column chromatography (PE/EA (V/V) ═ 1/1) to give the title compound as a colorless oil (2.3g, 77%).
MS-ESI:(ESI,pos.ion)m/z:174.2[M+H-100] + ;296.3[M+Na] +
Step 2: (R) -N-Boc-2- (2- (5-oxo-4, 5-dihydro-1, 3, 4-oxadiazol-2-yl) ethyl) morpholine
To a 100mL one-neck flask were added (R) -N-Boc-2- (3-hydrazino-3-oxopropyl) morpholine (600mg,2.20mmol) and THF (20mL) in that order, and after stirring to homogeneity, DIPEA (700mg,5.42mmol) was added and triphosgene (325mg,1.09mmol) in tetrahydrofuran (6mL) was added slowly. After the addition, the reaction mixture was reacted at 80 ℃ for 1 hour, then cooled to 25 ℃ and quenched with saturated sodium bicarbonate solution (30mL), followed by extraction with ethyl acetate (30 mL. times.3). The combined organic phases were washed with saturated brine (30 mL. times.2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure. The resulting residue was purified by preparative thin layer chromatography (PE/EA (V/V) ═ 1/2) to give the title compound as a colorless oil (430mg, 65.45%).
MS-ESI:(ESI,pos.ion)m/z:322.2[M+Na] +
And step 3: (R) -5- (2- (morpholin-2-yl) ethyl) -1,3, 4-oxadiazol-2 (3H) -one hydrochloride
To a 50mL single vial was added (R) -N-Boc-2- (2- (5-oxo-4, 5-dihydro-1, 3, 4-oxadiazol-2-yl) ethyl) morpholine (450mg,1.50mmol), dissolved in 5mL of ethyl acetate, and HCl in ethyl acetate (4mol/L,10mL) was slowly added. After the addition was complete, the reaction mixture was stirred at 25 ℃ for 1 hour, then concentrated under reduced pressure to give the title compound as a slightly brown oil (250mg, 83.47%).
MS-ESI:(ESI,pos.ion)m/z:200.2[M+H] +
And 4, step 4: (R) -4- (2-chloro-4-fluorophenyl) -6- (((R) -2- (2- (5-oxo-4, 5-dihydro-1, 3, 4-oxadiazol-2-yl) ethyl) morpholine) methyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester
To a 50mL single-necked flask were added in this order (R) -6- (bromomethyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester (470mg,1.06mmol), potassium carbonate (366mg,2.65mmol), (R) -5- (2- (morpholin-2-yl) ethyl) -1,3, 4-oxadiazol-2 (3H) -one hydrochloride (250mg,1.06mmol), and ethanol (10 mL). The reaction mixture was stirred for 12 hours at 25 ℃ under nitrogen, then filtered, the filtrate was concentrated under reduced pressure, the residue was diluted with water (50mL) and EA (50mL), the resulting mixture was adjusted to neutral pH with dilute hydrochloric acid (2M) under stirring, the aqueous layer was extracted with ethyl acetate (50mL × 3), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (PE/EA (V/V) ═ 1/2) to give the title compound as a colorless oil (240mg, 40.19%).
MS-ESI:(ESI,pos.ion)m/z:563.12[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ12.02(br,1H),9.69(s,1H),8.01–7.92(m,2H),7.44–7.37(m,2H),7.19–7.15(m,1H),6.04(s,1H),3.89(d,J=8.3Hz,3H),3.63–3.47(m,5H),2.78(t,J=10.5Hz,2H),2.61–2.58(m,2H),2.40–2.35(m,1H),2.06(t,J=10.5Hz,1H),1.75–1.65(m,2H)。
Example 6: (R) -methyl-4- (2-chloro-4-fluorophenyl) -6- (((R) -2- (2- (5-oxo-4, 5-dihydro-1, 2, 4-thiadiazol-3-yl) ethyl) morpholine) methyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester
Figure BDA0001397442500000421
The synthesis route is as follows:
Figure BDA0001397442500000422
step 1: (R) -N-Boc-2- (2- (5-oxo-4, 5-dihydro-1, 2, 4-thiadiazol-3-yl) ethyl) morpholine
To a 50mL one-neck flask were added (R, Z) -N-Boc-2- (3-amino-3- (hydroxyimino) propyl) morpholine (510mg,1.86mmol), thiocarbonyldiimidazole (665mg,3.73mmol), and THF (20mL) in that order. The reaction mixture was stirred for 1 hour at 25 ℃ under nitrogen and then concentrated under reduced pressure, the resulting residue was diluted with water (50mL), the aqueous layer was extracted with EA (100 mL. times.4), the organic phases were combined and then dried and concentrated.
To the above concentrated residue was added THF (20mL), and after stirring uniformly, boron trifluoride ether (794mg,5.59mmol) was added thereto at 25 ℃ and after the addition was completed, the resulting mixture was stirred for 3 hours, then sodium bicarbonate solution (50mL) was added to quench the reaction, the aqueous layer was extracted with ethyl acetate (40 mL. times.3), the organic phases were combined, the combined organic phases were washed with saturated brine (50 mL. times.2), dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the title compound as a pale brown oil (350mg, 59.5%).
MS-ESI:(ESI,pos.ion)m/z:338.1[M+Na] +
Step 2: (R) -3- (2- (morpholin-2-yl) ethyl) -1,2, 4-thiadiazol-5 (4H) -one hydrochloride
To a 50mL single vial were added (R) -N-Boc-2- (2- (5-oxo-4, 5-dihydro-1, 2, 4-thiadiazol-3-yl) ethyl) morpholine (350mg,1.11mmol) and ethyl acetate (5mL) in that order, and after complete dissolution with stirring, a solution of HCl in ethyl acetate (4mol/L,15mL) was added. The reaction mixture was stirred at 25 ℃ for 2h, then concentrated under reduced pressure to give the title compound as a light brown oil (280mg, 100%).
MS-ESI:(ESI,pos.ion)m/z:216.0[M+H] +
And 3, step 3: (R) -methyl-4- (2-chloro-4-fluorophenyl) -6- (((R) -2- (2- (5-oxo-4, 5-dihydro-1, 2, 4-thiadiazol-3-yl) ethyl) morpholine) methyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester
To a 50mL single-necked flask were added in this order (R) -6- (bromomethyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester (530mg,1.19mmol), potassium carbonate (410mg,2.97mmol), (R) -3- (2- (morpholin-2-yl) ethyl) -1,2, 4-thiadiazol-5 (4H) -one hydrochloride (300mg,1.19mmol), and ethanol (10 mL). The reaction mixture was stirred under nitrogen at 25 ℃ for 12 h, then filtered, the filtrate was concentrated under reduced pressure, the residue was diluted with water (50mL) and EA (50mL), then the pH was adjusted to neutral with dilute hydrochloric acid (2M), the aqueous layer was extracted with ethyl acetate (50mL × 3), the organic phases were combined, the combined organic phases were concentrated with drying, and the resulting residue was purified by silica gel column chromatography (PE/EA (V/V) ═ 1/1) to afford the title compound as a yellow solid (180mg, 26.1%).
MS-ESI:(ESI,pos.ion)m/z:578.05[M+H] +
Example 7: (R) -methyl-4- (2-chloro-4-fluorophenyl) -6- (((R) -2- (2- (5-oxo-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) ethyl) morpholine) methyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester
Figure BDA0001397442500000431
The synthetic route is as follows:
Figure BDA0001397442500000432
step 1: (R) -N-Boc-2- (2- (5-oxo-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) ethyl) morpholine
To a 50mL one-neck flask were added (R, Z) -N-Boc-2- (3-amino-3- (hydroxyimino) propyl) morpholine (200mg,0.73mmol), carbonylimidazole (240mg,1.48mmol), and dioxane (10mL) in that order. The reaction mixture was stirred under nitrogen at 110 ℃ for 12 hours, then concentrated under reduced pressure, the resulting residue was diluted with water (100mL) and EA (100mL), the resulting mixture was adjusted to pH 4 with dilute hydrochloric acid (2M), the aqueous layer was extracted with ethyl acetate (50 mL. times.3), the organic phases were combined, the combined organic phases were washed with dilute hydrochloric acid (50 mL. times.2) and saturated brine (50 mL. times.2), respectively, the organic layer was dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the title compound as a brown oily product (150mg, 68.5%). MS-ESI (ESI, pos.ion) m/z: 200.20[ M + H-100 ]] + ;322.20[M+Na] +
Step 2: (R) -3- (2- (morpholin-2-yl) ethyl) -1,2, 4-oxadiazol-5 (4H) -one hydrochloride
In a 50mL single vial were added (R) -N-Boc-2- (2- (5-oxo-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) ethyl) morpholine (130mg,0.43mmol) and ethyl acetate (5mL) in that order, and after complete dissolution with stirring, a solution of HCl in ethyl acetate (4mol/L,10mL) was added and the reaction mixture was stirred for 3 hours at 25 ℃ and then concentrated under reduced pressure to give the title compound as a brown oil (100mg, 97.7%).
MS-ESI:(ESI,pos.ion)m/z:200.1[M+H] +
And step 3: (R) -4- (2-chloro-4-fluorophenyl) -6- (((R) -2- (2- (5-oxo-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) ethyl) morpholino) methyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester
To a 50mL single-necked flask were added in this order (R) -6- (bromomethyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester (188mg,0.42mmol), potassium carbonate (150mg,1.08mmol), (R) -3- (2- (morpholin-2-yl) ethyl) -1,2, 4-oxadiazol-5 (4H) -one hydrochloride (100mg,0.42mmol), and ethanol (10 mL). The reaction mixture was stirred under nitrogen at 25 ℃ for 12 h, then filtered, the filtrate was concentrated under reduced pressure, the residue was diluted with water (50mL) and EA (50mL), then the pH was adjusted to 4 with dilute hydrochloric acid (2M), the aqueous layer was extracted with ethyl acetate (50mL × 3), the organic phases were combined, the combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure, and the resulting residue was purified by silica gel column chromatography (PE/EA (V/V) ═ 1/2) to give the title compound as a yellow solid (150mg, 62.8%).
1 HNMR(400MHz,CDCl 3 )δ9.57(s,1H),7.86(d,J=3.1Hz,1H),7.48(d,J=3.1Hz,1H),7.27(s,1H),7.18–7.10(m,1H),6.95–6.88(m,1H),6.21(s,1H),4.12–4.01(m,2H),3.96–3.85(m,2H),3.82–3.75(m,1H),3.62(s,3H),2.91–2.79(m,2H),2.78–2.64(m,2H),2.63–2.55(m,1H),2.24–2.15(m,1H),1.95–1.84(m,1H),1.82–1.73(m,1H)。
Example 8 methyl (R) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -6- (((R) -3- (5-thioxo-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) morpholino) methyl) -1, 4-dihydropyrimidine-5-carboxylate
Figure BDA0001397442500000441
The synthetic route is as follows:
Figure BDA0001397442500000442
step 1: (R, Z) -N-Boc-3- (N' -hydroxyformamidine) morpholine
To a 100mL single-necked flask were added (R) -N-Boc-3-cyanomorpholine (500mg,2.36mmol), hydroxylamine hydrochloride (196mg,2.82mmol), NaHCO in that order 3 (277mg,3.30mmol) and methanol (10 mL). The reaction mixture was reacted at 60 ℃ for 3 hours, then concentrated under reduced pressure, and the concentrated residue was slurried with ethyl acetate (15mL), filtered, and the filtrate was concentrated to give the title compound as a white solid (520mg, 90.00%). MS-ESI (ESI, pos.ion) m/z: 246.2[ M + H] +
And 2, step: (R) -N-Boc-3- (5-thio-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) morpholine
To a 50mL single-necked flask were added (R, Z) -N-Boc-3- (N' -hydroxyformamidine) morpholine (200mg,0.82mmol), thiocarbonyldiimidazole (300mg,1.68mmol), DBU (500mg,3.29mmol), and acetonitrile (10mL) in that order. The reaction mixture was stirred at 25 ℃ for 12 hours, then concentrated under reduced pressure, the residue was diluted with water (50mL) and ethyl acetate (50mL), the pH of the system was adjusted to 3 with dilute hydrochloric acid (2M), the layers were separated, the aqueous layer was extracted with ethyl acetate (50 mL. times.2), the combined organic layers were washed with dilute hydrochloric acid (2M) (50 mL. times.2) and saturated brine, respectively, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated by reduced pressure evaporation to give the title compound as a pale brown solid (200mg, 85.4%). MS-ESI (ESI, pos.ion) m/z: 188.1[ M + H] +
And step 3: (R) -3- (morpholin-3-yl) -1,2, 4-oxadiazole-5 (4H) -thione hydrochloride
To a 50mL single vial were added (R) -N-Boc-3- (5-thio-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) morpholine (200mg,0.70mmol) and ethyl acetate (5mL) in that order, and after complete dissolution with stirring, a solution of HCl in ethyl acetate (4mol/L,10mL) was added. The reaction mixture was stirred at 25 ℃ for 2h, then concentrated under reduced pressure to give the title compound as a white solid (150mg, 96.33%).
MS-ESI:(ESI,pos.ion)m/z:188.1[M+H] +
And 4, step 4: (R) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -6- (((R) -3- (5-thio-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) morpholino) methyl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester
To a 50mL single-necked flask were added in this order (R) -6- (bromomethyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester (300mg,0.67mmol), potassium carbonate (231mg,1.67mmol), (R) -3- (morpholin-3-yl) -1,2, 4-oxadiazole-5 (4H) -thione hydrochloride (150mg,0.67mmol), and ethanol (10 mL). The reaction mixture was stirred at 25 ℃ for 12 h, then filtered, the filtrate was concentrated under reduced pressure, the residue was diluted with water (50mL) and EA (50mL), then the pH was adjusted to 3 with dilute hydrochloric acid (2M), the aqueous layer was extracted with ethyl acetate (50mL × 3), the organic phases were combined, the combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated, and the resulting residue was purified by silica gel column chromatography (PE/EA (V/V) ═ 1/5) to afford the title compound as a yellow solid (153mg, 41.4%).
MS-ESI:(ESI,pos.ion)m/z:552.0[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ10.15–9.75(m,1H),8.08–7.86(m,2H),7.50–7.28(m,2H),7.26–7.08(m,1H),6.04–5.91(m,1H),4.90–4.70(m,2H),4.07–3.78(m,2H),3.72–3.63(m,1H),3.62–3.43(m,5H),2.93–2.69(m,2H)。
Example 9 methyl (R) -4- (2-chloro-4-fluorophenyl) -6- (((R) -3- (5-oxo-4, 5-dihydro-1, 2, 4-thiadiazol-3-yl) morpholine) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate
Figure BDA0001397442500000451
The synthesis route is as follows:
Figure BDA0001397442500000461
step 1: (R) -N-Boc-3- (5-oxo-4, 5-dihydro-1, 2, 4-thiadiazol-3-yl) morpholine
To a 50mL single-neck vial were added (R, Z) -N-Boc-3- (N' -hydroxyformamidine) morpholine (300mg,1.22mmol), thiocarbonyldiimidazole (430mg,2.41mmol), and THF (10mL) in that order. The reaction mixture was stirred at 25 ℃ for 1 hour under nitrogen, then diluted with water (30mL) and ethyl acetate (20mL), extracted by liquid separation, the aqueous phase was extracted with ethyl acetate (20 mL. times.2), and the organic phases were combined. The organic phase was washed with brine (50 mL. times.2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give a light brown oil.
The above oil was dissolved in THF (10mL), to which boron trifluoride diethyl etherate (520mg,3.66mmol) was slowly added at 25 ℃. After the addition was completed, the reaction mixture was stirred at 25 ℃ for 2 hours, followed by addition of water (30mL) to quench the reaction, followed by extraction with ethyl acetate (30 mL. times.3), and the organic phases were combined, washed with saturated brine (30 mL. times.2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the title compound as a light brown oily product (200mg, 56.92%).
MS-ESI:(ESI,pos.ion)m/z:188.2[M+H-100] + ;310.2[M+Na] +
Step 2: (R) -3- (morpholin-3-yl) -1,2, 4-thiadiazol-5 (4H) -one hydrochloride
To a 50mL single vial were added (R) -N-Boc-3- (5-oxo-4, 5-dihydro-1, 2, 4-thiadiazol-3-yl) morpholine (200mg,0.70mmol) and ethyl acetate (5mL) in that order, and after complete dissolution of the original, a solution of HCl in ethyl acetate (4mol/L,10mL) was added slowly and the reaction mixture stirred at 25 ℃ for 2 h. After completion of the reaction, the reaction mixture was concentrated under reduced pressure to give the title compound as a white solid (150mg, 96.33%).
MS-ESI:(ESI,pos.ion)m/z:188.1[M+H] +
And 3, step 3: (R) -4- (2-chloro-4-fluorophenyl) -6- (((R) -3- (5-oxo-4, 5-dihydro-1, 2, 4-thiadiazol-3-yl) morpholine) methyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester
To a 50mL single-necked flask were added in this order (R) -6- (bromomethyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester (300mg,0.67mmol), potassium carbonate (231mg,1.67mmol), (R) -3- (morpholin-3-yl) -1,2, 4-thiadiazol-5 (4H) -one hydrochloride (150mg,0.67mmol), and ethanol (10 mL). The reaction mixture was stirred under nitrogen at 25 ℃ for 12 h, then filtered, the filtrate was concentrated under reduced pressure, the residue was diluted with water (50mL) and ethyl acetate (50mL), the pH was adjusted to 3 with dilute hydrochloric acid (2M), the solution was separated, the aqueous phase was extracted with ethyl acetate (50mL × 2), the combined organic phases were dried over anhydrous sodium sulfate, filtered, the filtrate was concentrated under reduced pressure by evaporation, and the resulting residue was purified by silica gel column chromatography (PE/EA (V/V) ═ 1/1) to give the title compound as a yellow solid (180mg, 48.71%).
MS-ESI:(ESI,pos.ion)m/z:552.2[M+H] +
Example 10: (R) -4- (2-chloro-4-fluorophenyl) -6- (((R) -3- (5-oxo-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) morpholine) methyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester
Figure BDA0001397442500000471
The synthetic route is as follows:
Figure BDA0001397442500000472
step 1: (R) -N-Boc-3- (5-oxo-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) morpholine
To a 50mL single-neck flask were added (R, Z) -N-Boc-3- (N' -hydroxyformamidine) morpholine (500mg,2.04mmol), carbonyldiimidazole (1.0g,6.20mmol), and dioxane (15mL) in that order. The reaction mixture was stirred at 110 ℃ for 12 hours, then filtered, the filtrate was concentrated under reduced pressure, the residue was diluted with water (50mL) and EA (50mL), then the pH was adjusted to 3 with dilute hydrochloric acid (2M), the layers were separated, the aqueous layer was extracted with ethyl acetate (50 mL. times.3), the organic phases were combined, the organic phase was washed successively with dilute hydrochloric acid (50 mL. times.2) and saturated brine (50 mL. times.2), dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure to give the title compound as a brown oily product (320mg, 57.87%).
MS-ESI:(ESI,pos.ion)m/z:172.2[M+H-100] + ;294.2[M+23] +
Step 2: (R) -3- (morpholin-3-yl) -1,2, 4-oxadiazol-5 (4H) -one hydrochloride
To a 50mL single vial were added (R) -N-Boc-3- (5-oxo-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) morpholine (450mg,1.66mmol) and ethyl acetate (7mL) in that order, and after complete dissolution with stirring, a solution of HCl in ethyl acetate (4mol/L,20mL) was added. After the addition was complete, the reaction mixture was stirred at 25 ℃ for 2 hours, then concentrated under reduced pressure to give the title compound as a white solid (320mg, 92.9%).
MS-ESI:(ESI,pos.ion)m/z:172.1[M+H] +
And 3, step 3: (R) -4- (2-chloro-4-fluorophenyl) -6- (((R) -3- (5-oxo-4, 5-dihydro-1, 2, 4-oxadiazol-3-yl) morpholine) methyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylic acid methyl ester
To a 50mL single-necked flask were added in sequence (R) -methyl 6- (bromomethyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (685mg,1.54mmol), potassium carbonate (530mg,3.83mmol), (R) -3- (morpholin-3-yl) -1,2, 4-oxadiazol-5 (4H) -one hydrochloride (320mg,1.54mmol), and ethanol (10 mL). The reaction mixture was stirred under nitrogen at 25 ℃ for 12 h, then filtered, the filtrate was concentrated under reduced pressure, the residue was diluted with water (50mL) and EA (50mL), then pH was adjusted to 6-7 with dilute hydrochloric acid (2M), the layers were separated, the aqueous layer was extracted with ethyl acetate (50mL × 3), the organic phases were combined, the combined organic phases were dried over anhydrous sodium sulfate, filtered and concentrated under reduced pressure, and the residue was purified by silica gel column chromatography (PE/EA (V /) ═ 1/1) to give the title compound as a yellow solid (240mg, 29.11%).
MS-ESI:(ESI,pos.ion)m/z:536.2[M+H] +
1 H NMR(400MHz,DMSO-d 6 )δ12.40(br,1H),9.58(s,1H),8.13–7.83(m,2H),7.52–7.23(m,2H),7.27–7.04(m,1H),6.03(s,1H),4.21–4.01(m,2H),3.91(d,J=2.9Hz,3H),3.79–3.69(m,2H),3.50(s,3H),3.08–2.96(m,1H)。
Biological activity assay
Test 1: anti-HBV EC 50 Test method
In vitro anti-HBV pharmacodynamic activity determination experiment using HBV HepG2.2.15 cell strain
The experimental method comprises the following steps:
qPCR was performed to determine the viral DNA content of the cell culture fluid and calculate the concentration of the compound at which half of the viral inhibition (EC) occurred 50 ) The specific experimental method is as follows:
HepG2.2.15 cells were seeded into 96-well cell culture plates (40,000 cells/well) and the cells were treated the next day by adding cell culture medium containing different concentrations of test compound (compound maximum final concentration 16.4. mu.M, 3-fold gradient dilution, 9 dilution spots, duplicate wells). And replacing the culture solution containing the drug to be detected on the fifth day, and collecting the culture supernatant and extracting DNA in the supernatant on the eighth day.
And (3) virus DNA extraction: reference is made to the QIAamp 96DNA Blood Kit (QIAGEN 51161).
Quantitative PCR: preparing a reaction mixed solution according to a PCR system, and adding the mixed solution into a 96-hole PCR reaction plate (special for quantification); adding the standard template diluted in proportion (the highest concentration of the standard template is 1 multiplied by 10) 7 copies/ul, 9-fold dilution for 7 points, and minimum concentration of 10 copies/ul); adding a sample template; sealing the 96-well plate by using a plate sealing film; the quantitative PCR instrument was run according to the set program.
Percentage inhibition of HBV replication by compounds was calculated: % inh. ═ 1-amount of HBV DNA treated with compound/amount of HBV DNA treated with DMSO control × 100.
Calculating the EC of Compounds on HBV replication 50 The value: EC was calculated using GraphPad Prism 5 analysis software using the "four parameter s i equation 50 The value is obtained.
TABLE 2: EC of compounds on HBV replication 50 Value of
Examples EC 50
Example 1 365.65nmol
Example 3 250.45nmol
And (4) conclusion: experimental data show that the compound has good inhibitory activity on HBV and has good application prospect in the aspect of resisting HBV.
And (3) testing 2: cytotoxic and Selectivity indices
Test compound cytotoxicity and selection index experiments
The experimental method comprises the following steps:
HepDE cells were plated in 96-well plates (5 × 103 cells per well) and treated with compound for determination of EC 50. After 5 days of treatment, 20uL of CCK-8 reagent was added to test cell viability, and after incubation at 37 ℃ for 2 hours, the absorbance at wavelengths of 450nm and 630nm was measured using a microplate reader to determine the concentration of each compound that caused 50% lethality in the host cells (CC 50).
The relative effectiveness of a compound to induce cell death and inhibit viral replication was defined as the selection index (CC50 value/EC 50 value). Based on the CC50 and EC50 data, a selection index was determined.
TABLE 3: compound cytotoxic CC 50 Value of
Examples CC 50
Example 1 >150μmol
Example 3 >150μmol
And (4) conclusion: experimental data of cytotoxicity and selectivity indexes show that the compound has low toxicity.
And (3) testing: pharmacokinetic experiments of the compounds of the invention in beagle dogs, mice and rats
(1) Beagle PK test experiment
Experimental methods for PK determination of compounds in beagle dogs:
beagle dogs were administered 2.5mg/kg or 5mg/kg by oral gavage or 1mg/kg or 2mg/kg by intravenous injection of the test compound.
Blood was collected intravenously at time points (0.083, 0.25, 0.5, 1,2,4, 6, 8 and 24 hours) after administration and collected at the time of EDTA-K addition 2 In the anticoagulation tube. Plasma samples were subjected to liquid-liquid extraction and then quantitatively analyzed by multiplex reaction ion monitoring (MRM) on a triple quadrupole tandem mass spectrometer. Pharmacokinetic parameters were calculated using a non-compartmental model using WinNonlin 6.3 software.
And (4) conclusion: the pharmaceutical experiment data show that the compound has better pharmacokinetic property and has good application prospect in the aspect of anti-HBV.
(2) Mouse PK test experiments:
experimental methods for PK determination of compounds in mice:
ICR mice were orally gavaged with 10mg/kg or injected via the tail vein with 2mg/kg or 10mg/kg of the test compound. Blood was collected at time points (0.083, 0.25, 0.5, 1,2,4, 6, 8 and 24 hours) from the orbital vein after administration and collected by adding EDTA-K 2 In the anticoagulation tube. Plasma samples were subjected to liquid-liquid extraction and then quantitatively analyzed by multiplex reaction ion monitoring (MRM) on a triple quadrupole tandem mass spectrometer. Pharmacokinetic parameters were calculated using a non-compartmental model method using WinNonlin 6.1 software.
And (4) conclusion: the data of the pharmaceutical experiment show that the compound has better pharmacokinetic property and has good application prospect in the aspect of anti-HBV virus.
(3) SD rat PK test experiment:
experimental methods for PK assay of compounds in SD rats:
rats were dosed either 2.5mg/kg or 5mg/kg per oral gavage or 1mg/kg per intravenous injection of the test compound.
Blood was collected intravenously at time points (0.083, 0.25, 0.5, 1,2,5, 7 and 24 hours) after administration and collected by adding EDTA-K 2 In the anticoagulation tube. Plasma samples were subjected to liquid-liquid extraction and then quantitatively analyzed by multiplex reaction ion monitoring (MRM) on a triple quadrupole tandem mass spectrometer. Pharmacokinetic parameters were calculated using a non-compartmental model using WinNonlin 6.3 software.
And (4) conclusion: the data of the pharmaceutical experiment show that the compound has better pharmacokinetic property and has good application prospect in the aspect of anti-HBV virus.
And (4) testing: stability test of liver microsomes of different species
Experimental methods for stability of liver microsomes of compounds in different species:
30. mu.L of a mixed solution of the blank solution and the liver microsomes was added to a 96-well plate, and 15. mu.L of a buffer containing the compound to be detected was added to each well, and two samples were prepared in parallel. After preincubation at 37 ℃ for 10min, 15. mu.L of NADPH solution (8mM) was added at time points, with a final concentration of test compound of 1. mu.M, liver microsome concentration of 0.1mg/mL, and NADPH concentration of 2 mM. Incubate for 0, 15, 30, 60min, respectively, after incubation, add 150. mu.L acetonitrile (containing internal standard) into the mixed system. The acetonitrile diluted sample was centrifuged at 4000rpm for 5min and 150. mu.L of the supernatant was taken to LC-MS/MS for analysis.
And (4) conclusion: the stability experimental data of the liver microsome show that the compound has better stability.
And (5) testing: solubility test method
Experimental test method for compound solubility:
except for other provisions, a test sample ground into fine powder is weighed or a liquid test sample is weighed, the test sample is placed in a solvent with a certain volume at 25 +/-2 ℃, strong shaking is carried out for 30s every 5min, and the dissolution condition within 30min is observed, if no visible solute particles or liquid drops exist, the test sample is regarded as complete dissolution. Very soluble means that 1g (mL) of solute can be dissolved in less than 1mL of solvent;
soluble means that 1g (mL) of solute can be dissolved in 1-less than 10mL of solvent;
the dissolving means that 1g (mL) of solute can be dissolved in 10-less than 30mL of solvent;
slightly soluble means that 1g (mL) of solute can be dissolved in 30-less than 100mL of solvent;
slightly soluble means that solute lg (mL) can be dissolved in 100-less than 1000mL of solvent;
the minimal dissolution means that 1g (mL) of solute can be dissolved in 1000-less than 10000mL of solvent;
by hardly soluble or insoluble is meant that 1g (mL) of solute is not completely dissolved in 10000mL of solvent.
And (4) conclusion: solubility experimental data show that the compound has better solubility.
And 6, testing: hERG test method
Method for testing heart experiment by compound
In 384 well plates were added compound/positive control/negative control, membrane fragments containing the hERG channel, tracer with high affinity for the hERG channel in sequence and incubated for 4 hours at 25 ℃ and 250 rpm. The fluorescence polarization value of each well is measured by a multifunctional microplate reader, and the relative inhibition rate and 50% Inhibition Concentration (IC) of the compound on the hERG channel are calculated 50 )。
And (4) conclusion: the hERG test data show that the compound has low toxicity to the heart.
Although the invention has been described in detail hereinabove by way of general description, specific embodiments and experiments, it will be apparent to those skilled in the art that many modifications and improvements can be made thereto based on the invention. Accordingly, it is intended that all such modifications and alterations be included within the scope of this invention as defined in the appended claims.

Claims (8)

1. A compound having a structure as shown in formula (IV) or (IVa), or a tautomer or a pharmaceutically acceptable salt thereof,
Figure FDA0003716182200000011
wherein Z is a sub-structural formula shown as follows:
Figure FDA0003716182200000012
each R 1 Independently hydrogen, F, Cl, Br, I, methyl or ethyl;
R 2 is methyl, ethyl, n-propyl or isopropyl;
R 3 is thiazolyl;
f is 0, 1,2 or 3.
2. The compound of claim 1, selected from the structures of one of:
Figure FDA0003716182200000013
or a tautomer or pharmaceutically acceptable salt thereof.
3. A pharmaceutical composition comprising a compound of claim 1 or 2, and a pharmaceutically acceptable carrier therefor.
4. The pharmaceutical composition according to claim 3, further comprising other anti-HBV drugs.
5. The pharmaceutical composition according to claim 4, wherein the other anti-HBV drug is an HBV polymerase inhibitor, an immunomodulator, or an interferon.
6. The pharmaceutical composition according to claim 4, wherein the other anti-HBV drug is lamivudine, telbivudine, tenofovir disoproxil, entecavir, adefovir dipivoxil, Alfaferone, Alloferon, simon interleukin, clevudine, emtricitabine, faprolivir, interferon, calamin CP, intefene, interferon alpha-1 b, interferon alpha-2 a, interferon beta-1 a, interferon alpha-2, interleukin-2, mevotil, nitazoxanide, peginterferon alpha-2 a, ribavirin, roscovitine-A, cizopyran, Euforavac, azapril, Phosphazid, Heplissav, interferon alpha-2 b, levamisole, or propafenib.
7. Use of a compound of claim 1 or 2 or a pharmaceutical composition of any one of claims 3-6 in the manufacture of a medicament for preventing, treating or ameliorating a viral disease in a patient, wherein the viral disease is hepatitis b infection or a disease caused by hepatitis b infection.
8. The use according to claim 7, wherein the disease caused by hepatitis B infection is liver cirrhosis or hepatocellular carcinoma.
CN201710783447.4A 2016-09-05 2017-09-04 Dihydropyrimidine compound and application thereof in medicine Active CN107793409B (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201610802768 2016-09-05
CN201610802768X 2016-09-05

Publications (2)

Publication Number Publication Date
CN107793409A CN107793409A (en) 2018-03-13
CN107793409B true CN107793409B (en) 2022-09-27

Family

ID=61532243

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201710783447.4A Active CN107793409B (en) 2016-09-05 2017-09-04 Dihydropyrimidine compound and application thereof in medicine

Country Status (1)

Country Link
CN (1) CN107793409B (en)

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020205867A1 (en) 2019-04-02 2020-10-08 Aligos Therapeutics, Inc. Compounds targeting prmt5
CN110615797A (en) * 2019-10-11 2019-12-27 李丽丽 Compound for treating hepatitis B and application thereof
CN114853761A (en) * 2021-02-05 2022-08-05 刘沛 Bifunctional derivative containing dihydropyrimidine and application thereof

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010069147A1 (en) * 2008-12-17 2010-06-24 张中能 Dihydropyrimidine derivatives, compositions thereof and their use
CN103626752A (en) * 2012-08-24 2014-03-12 广东东阳光药业有限公司 Dihydropyrimidine compounds and application of same in drugs

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010069147A1 (en) * 2008-12-17 2010-06-24 张中能 Dihydropyrimidine derivatives, compositions thereof and their use
CN103626752A (en) * 2012-08-24 2014-03-12 广东东阳光药业有限公司 Dihydropyrimidine compounds and application of same in drugs
CN104926808A (en) * 2012-08-24 2015-09-23 广东东阳光药业有限公司 Dihydro-pyrimidine compound and application thereof in medicine

Also Published As

Publication number Publication date
CN107793409A (en) 2018-03-13

Similar Documents

Publication Publication Date Title
CN110066278B (en) Fused tricyclic compound and application thereof in medicines
AU2018351400B2 (en) Dihydropyrimidine compounds and uses thereof in medicine
CN110036005B (en) Amide derivatives and their use in medicine
CN108976223B (en) Fused tricyclic compound and application thereof in medicines
CN113166154B (en) Deuterated dihydropyrimidine compound and application thereof in medicines
CN110950860B (en) Fused tricyclic compound and application thereof in medicines
CN110862390B (en) Fused tricyclic compounds and application thereof in medicines
CN111848627B (en) Dihydropyrimidine compound and application thereof in medicine
CN107793409B (en) Dihydropyrimidine compound and application thereof in medicine
CN111825676B (en) Dihydropyrimidine compounds and application thereof in medicines
CN111116577A (en) Fused tetracyclic compounds and application thereof in medicines
CN110964023B (en) Fused tetracyclic compounds and application thereof in medicines
CN111116588B (en) Fused tetracyclic compound and application thereof in medicines
CN111217811B (en) Fused tricyclic compounds and application thereof in medicines
WO2021197486A1 (en) Novel spiro compound and application thereof in drug
CN113831359A (en) Dihydropyrimidine compound and application thereof in medicine
CN109485676B (en) Novel nucleoside reverse transcriptase inhibitors for antiviral use
CN110903284A (en) Fused tricyclic compound and application thereof in medicines
CN113493441B (en) Novel spiro compound and application thereof in medicines
WO2022143610A1 (en) Novel amide pyrrole compounds and use thereof in medicaments
CN113135937A (en) Fused tetracyclic compounds and application thereof in medicines
CN117143109A (en) Fused tetracyclic compound and application thereof in medicines
CN114075206A (en) Novel fused ring compound and application thereof in medicines
WO2022156758A1 (en) Novel amide pyrrole compound and use thereof in drugs
CN113444102A (en) Fused tetracyclic compounds and application thereof in medicines

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
CP03 Change of name, title or address

Address after: 523808 No.1, Gongye North Road, Songshanhu Park, Dongguan City, Guangdong Province

Patentee after: Guangdong Dongyangguang Pharmaceutical Co.,Ltd.

Address before: 523808 No. 1 Industrial North Road, Songshan Industrial Park, Songshan, Guangdong, Dongguan, Hubei

Patentee before: SUNSHINE LAKE PHARMA Co.,Ltd.

CP03 Change of name, title or address