WO2018090862A1 - 二氢嘧啶类化合物及其制备方法和用途 - Google Patents
二氢嘧啶类化合物及其制备方法和用途 Download PDFInfo
- Publication number
- WO2018090862A1 WO2018090862A1 PCT/CN2017/110123 CN2017110123W WO2018090862A1 WO 2018090862 A1 WO2018090862 A1 WO 2018090862A1 CN 2017110123 W CN2017110123 W CN 2017110123W WO 2018090862 A1 WO2018090862 A1 WO 2018090862A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- compound
- alkyl
- group
- cooh
- pharmaceutically acceptable
- Prior art date
Links
- 0 OCC(C*C1)CC1(F)F Chemical compound OCC(C*C1)CC1(F)F 0.000 description 13
- AZKLITPVPAPTFY-UGQQDIOTSA-N OC(/C=C/[C@H]1OCCNC1)=O Chemical compound OC(/C=C/[C@H]1OCCNC1)=O AZKLITPVPAPTFY-UGQQDIOTSA-N 0.000 description 2
- YNMJEEMSDNYEBZ-SBXXRYSUSA-N CC1C=CSC(C(NCC2)=N[C@H]2c2cccc(F)c2Br)=CC1 Chemical compound CC1C=CSC(C(NCC2)=N[C@H]2c2cccc(F)c2Br)=CC1 YNMJEEMSDNYEBZ-SBXXRYSUSA-N 0.000 description 1
- SVKWSVDFYWOGGU-AWEZNQCLSA-N CCOC(C1=C(CBr)NC(c2ncc[s]2)=N[C@H]1c(ccc(F)c1)c1Cl)=O Chemical compound CCOC(C1=C(CBr)NC(c2ncc[s]2)=N[C@H]1c(ccc(F)c1)c1Cl)=O SVKWSVDFYWOGGU-AWEZNQCLSA-N 0.000 description 1
- JUWNGMAYTXVYME-INIZCTEOSA-N CCOC(C1=C(CN(C2)CC2(F)F)NC(c2ncc[s]2)=N[C@H]1c(ccc(F)c1)c1Cl)=O Chemical compound CCOC(C1=C(CN(C2)CC2(F)F)NC(c2ncc[s]2)=N[C@H]1c(ccc(F)c1)c1Cl)=O JUWNGMAYTXVYME-INIZCTEOSA-N 0.000 description 1
- NJTPXGWHZWNQIN-KTYGWHHQSA-N COC(C1=C(CN2C[C@@H](/C=C/C(O)=O)OCC2)NC(c2ncc[s]2)=N[C@H]1c(c(Cl)c1F)ccc1F)=O Chemical compound COC(C1=C(CN2C[C@@H](/C=C/C(O)=O)OCC2)NC(c2ncc[s]2)=N[C@H]1c(c(Cl)c1F)ccc1F)=O NJTPXGWHZWNQIN-KTYGWHHQSA-N 0.000 description 1
- SDWOHGBGCJVLAH-IJVJVEAYSA-N COC(C1=C(CN2C[C@@H](/C=C/C(O)=O)OCC2)NC(c2ncc[s]2)=N[C@H]1c1cccc(F)c1Br)=O Chemical compound COC(C1=C(CN2C[C@@H](/C=C/C(O)=O)OCC2)NC(c2ncc[s]2)=N[C@H]1c1cccc(F)c1Br)=O SDWOHGBGCJVLAH-IJVJVEAYSA-N 0.000 description 1
- UIYNJIYPPDNPMO-SECBINFHSA-N Fc1ccc([C@@H]2N=C(c3ncc[s]3)NCC2)c(Cl)c1F Chemical compound Fc1ccc([C@@H]2N=C(c3ncc[s]3)NCC2)c(Cl)c1F UIYNJIYPPDNPMO-SECBINFHSA-N 0.000 description 1
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D417/00—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
- C07D417/14—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/506—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/535—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
- A61K31/5355—Non-condensed oxazines and containing further heterocyclic rings
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/14—Antivirals for RNA viruses
- A61P31/16—Antivirals for RNA viruses for influenza or rhinoviruses
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/20—Antivirals for DNA viruses
- A61P31/22—Antivirals for DNA viruses for herpes viruses
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D401/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
- C07D401/14—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D403/00—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
- C07D403/14—Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D413/00—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
- C07D413/14—Heterocyclic 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
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D417/00—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
- C07D417/02—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings
- C07D417/04—Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/0012—Galenical forms characterised by the site of application
- A61K9/0014—Skin, i.e. galenical aspects of topical compositions
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/0012—Galenical forms characterised by the site of application
- A61K9/0019—Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/0012—Galenical forms characterised by the site of application
- A61K9/0053—Mouth and digestive tract, i.e. intraoral and peroral administration
-
- Y—GENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
- Y02—TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
- Y02A—TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
- Y02A50/00—TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
- Y02A50/30—Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change
Definitions
- the present invention relates to a dihydropyrimidine compound having antiviral activity, a pharmaceutical composition comprising the same, a preparation method thereof and its prevention or treatment including but not limited to hepatitis A virus, hepatitis B virus, and hepatitis C virus Use in hepatitis, influenza, herpes, and viral diseases of acquired immunodeficiency syndrome (AIDS).
- a dihydropyrimidine compound having antiviral activity a pharmaceutical composition comprising the same, a preparation method thereof and its prevention or treatment including but not limited to hepatitis A virus, hepatitis B virus, and hepatitis C virus Use in hepatitis, influenza, herpes, and viral diseases of acquired immunodeficiency syndrome (AIDS).
- AIDS acquired immunodeficiency syndrome
- a virus consists of a nucleic acid molecule (DNA or RNA) or a protein (such as a prion).
- the virus can cause a variety of infectious diseases. Common diseases caused by viruses include, but are not limited to, viral hepatitis A, hepatitis B, hepatitis C, influenza, herpes and acquired immunodeficiency syndrome. (AIDS).
- antiviral drugs play a role by inhibiting virus attachment, uncoating, viral gene duplication, maturation or release, or by affecting the host's immune system, including reverse transcriptase inhibitors and capsid protein assembly inhibitors. Wait.
- Hepatitis B virus is a common hepadnavirus-like viral pathogen. Such viruses can cause diseases such as acute hepatitis, chronic hepatitis, liver fibrosis, cirrhosis and liver cancer.
- Drugs for the treatment of hepatitis B include interferons and nucleoside analogs (such as lamivudine and adefovir dipivoxil).
- interferon inhibits the replication of hepatitis B virus by reacting with cell surface receptors to produce antiviral proteins.
- nucleoside analogs act primarily by inhibiting the replication of viral polymerase (reverse transcriptase). The disadvantage of this type of drug is that long-term use can easily cause virus mutation and produce drug resistance.
- hepatitis B can also be treated with non-nucleoside analogs.
- heteroaryldihydropyrimidines Boy 41-4109
- dihydropyrimidines induce misassembly of core proteins, resulting in unstable capsid proteins that accelerate the degradation of core proteins (Biochem. Pharmacol., 2003, 66, 2273-2279).
- the heteroaryl dihydropyrimidine compound HAP1 (Proc. Natl. Acad. Sci., 2005, 102, 8138-8143) discovered by Zlotnick et al.
- the dihydropyrimidine compounds of the present invention can inhibit the replication of HBV DNA more efficiently than the disclosed dihydropyrimidine HBV capsid protein assembly regulators (eg, cells).
- the preferred anti-viral activity of the present invention is about 10 times the activity of the preferred compound (compound of Example 9) of WO2015144093.
- the compounds of the present invention are free of the cardiotoxicity of the disclosed dihydropyrimidine compounds (such as the hERG inhibitory activity of the preferred compounds of GLS4 and WO2015144093 (compound of Example 9)); and with the disclosed dihydropyrimidine compounds (eg Compared to the compound of Example 5 in WO201403748, the compounds of the invention have a significantly reduced induction of the CYP450 enzyme 3A4 subtype. Furthermore, in the pharmacokinetic tests of rats, Beagle dogs, cynomolgus monkeys, the compounds of the invention exhibit better pharmacokinetic properties (eg better exposure, blood concentration, bioavailability) . At the same time, the preferred compounds of the present invention have good liver targeting properties, and the exposure of the drug to the liver can reach about 10 times of the exposure in plasma, indicating that the compound has liver enrichment ability, thereby facilitating improvement of liver diseases. Efficacy.
- One aspect of the invention provides a compound, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, isotopically labeled compound or prodrug thereof, wherein The compound has the structure of Formula I or Formula Ia:
- Ar 1 and Ar 2 are each independently selected from a C 6-14 aryl group and a 5-14 membered heteroaryl group, which are optionally selected from one or more selected from the group consisting of halogen, -OH, -CN, -NO 2 , -N Substituent substitution of (R) 2 , C 1-6 alkyl, halo C 1-6 alkyl, C 1-6 alkylthio and C 3-6 cycloalkyl;
- L is absent or selected from -O-, -S-, and -NR-;
- R 1 and R 2 are each independently selected from H (including 1 H, 2 H, 3 H), C 1-6 alkyl (eg, deuterated C 1-6 alkyl), and C 3-6 cycloalkyl;
- R 3 is a quaternary, five-, six- or seven-membered nitrogen heterocyclic ring selected from the following structures:
- R a , R a ' , R 4 , R 4 ' , R 5 , R 5 ' and R 6 are each independently selected from H, halogen, -OH, -COOH, -CN, -NO 2 , -N(R) 2 , C 1-6 alkyl, halo C 1-6 alkyl, -WC 1-6 alkyl, -C 1-6 alkylene-WR, -WC 1-6 alkylene -W'-R, -WC 2-6 alkenyl, -C 2-6 alkenylene-WR, -WC 2-6 alkenylene-W'-R and C 3-6 cycloalkyl, wherein The alkylene and alkenylene are optionally further separated by one or more W; or R a together with R a ' , R 4 together with R 5 and/or R 4 ' and R 5' together, each occurrence
- R 6 is attached to the ring carbon atom marked by * and / or ** in the above nitrogen heterocyclic structure
- R is each independently selected from the group consisting of H, C 1-6 alkyl and C 3-6 cycloalkyl;
- g is 1 or 2;
- t is 0, 1, 2 or 3, provided that t is not greater than the number of positions on the corresponding group that can be substituted, and when t is greater than 1, each R 6 may be the same or different.
- compositions comprising a prophylactically or therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph thereof, A solvate, metabolite, isotopically labeled compound or prodrug and one or more pharmaceutically acceptable carriers, preferably a solid formulation, a liquid formulation or a transdermal formulation.
- Another aspect of the invention provides a method of preparing a pharmaceutical composition
- a method of preparing a pharmaceutical composition comprising administering a compound of the invention, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph thereof,
- the solvate, metabolite, isotopically labeled compound or prodrug is combined with one or more pharmaceutically acceptable carriers.
- Another aspect of the invention provides a compound of the invention, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, isotopically labeled compound thereof, or Use of a prodrug or a pharmaceutical composition of the invention in the manufacture of a medicament for the prevention or treatment of a viral disease.
- Another aspect of the invention provides a compound of the invention, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, isotopically labeled compound thereof, or A prodrug or a pharmaceutical composition of the invention for use in preventing or treating a viral disease.
- Another aspect of the invention provides a method of preventing or treating a viral disease, the method comprising administering to an individual in need thereof an effective amount of a compound of the invention, or a pharmaceutically acceptable salt, ester, stereoisomer thereof, Tautomers, polymorphs, solvates, metabolites, isotopically labeled compounds or prodrugs or pharmaceutical compositions of the invention.
- the above viral diseases include, but are not limited to, viral hepatitis A, hepatitis B virus, hepatitis C virus, influenza, herpes, and acquired immunodeficiency syndrome (AIDS).
- viral hepatitis A hepatitis A
- hepatitis B virus hepatitis B virus
- hepatitis C virus influenza
- herpes and acquired immunodeficiency syndrome (AIDS).
- AIDS acquired immunodeficiency syndrome
- Another aspect of the invention provides a method of preparing a compound of the invention, the method comprising the steps of:
- Hal is selected from the group consisting of F, Cl, Br, and I;
- the halogenating agent is selected from the group consisting of Cl 2 , Br 2 , I 2 , N-chlorosuccinimide, N-bromosuccinimide, and N-iodosuccinimide;
- Step one is carried out in a protic solvent in the presence of an alkali metal salt
- Step two is carried out in an aprotic solvent
- Step 3 is carried out in an aprotic solvent in the presence of an organic base or an inorganic base;
- R 2 in the compound of the present invention is a C 1-6 alkyl group
- it can also be synthesized by a method comprising the following steps:
- R 2 ' is H or C 1-5 alkyl
- Hal is selected from the group consisting of F, Cl, Br, and I;
- the halogenating agent is selected from the group consisting of Cl 2 , Br 2 , I 2 , N-chlorosuccinimide, N-bromosuccinimide, and N-iodosuccinimide;
- Step I is carried out in the presence of a Lewis acid in a non-polar solvent
- Step II is carried out in an aprotic solvent in the presence of an organic or inorganic base
- Step III is carried out in an aprotic solvent
- Step IV is carried out in an aprotic solvent in the presence of an organic or inorganic base.
- Figure 1 Induction of CYP450 enzyme 3A4 subtype by compound 10-227, control compound 3 and rifampicin.
- alkylene denotes a saturated divalent hydrocarbon group, preferably a saturated divalent hydrocarbon group having 1, 2, 3, 4, 5 or 6 carbon atoms, such as methylene, ethylene, Propylene or butylene.
- alkenylene denotes a divalent hydrocarbon radical containing one or more double bonds, preferably having 2, 3, 4, 5 or 6 carbon atoms, such as ethenylene, propenylene or Allylene.
- alkenylene groups the compounds may exist in pure E (ent ought) form, pure Z (zusammen) form, or any mixture thereof.
- alkyl is defined as a straight or branched chain saturated aliphatic hydrocarbon.
- an alkyl group has from 1 to 12, such as from 1 to 6 carbon atoms.
- C1-6 alkyl refers to a linear or branched group of 1 to 6 carbon atoms (eg, methyl, ethyl, n-propyl, isopropyl, n-butyl) Base, isobutyl, sec-butyl, tert-butyl, n-pentyl or n-hexyl), which is optionally substituted by one or more (such as 1 to 3) suitable substituents such as halogen (in this case the group)
- the group is referred to as "haloalkyl” (for example, CF 3 , C 2 F 5 , CHF 2 , CH 2 F, CH 2 CF 3 , CH 2 Cl or -CH 2 CH 2 CF 3
- C 1-4 alkyl refers to a linear or branched aliphatic hydrocarbon chain of 1 to 4 carbon atoms (ie methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, Sec-butyl or tert-butyl).
- cycloalkyl refers to a saturated or unsaturated, non-aromatic monocyclic or polycyclic (such as bicyclic) hydrocarbon ring (eg, a monocyclic ring such as cyclopropyl, cyclobutyl, cyclopentyl, Cyclohexyl, cycloheptyl, cyclooctyl, cyclodecyl, or bicyclic, including spiro, fused or bridged systems (such as bicyclo [1.1.1] pentyl, bicyclo [2.2.1] heptyl, bicyclo [ 3.2.1] Octyl or bicyclo [5.2.0] anthracenyl, decalinyl, etc.), which is optionally substituted by one or more (such as 1 to 3) suitable substituents.
- bicyclic hydrocarbon ring eg, a monocyclic ring such as cyclopropyl, cyclobutyl, cyclopentyl, Cyclohex
- C 3-6 cycloalkyl refers to a saturated or unsaturated, non-aromatic monocyclic or polycyclic (such as bicyclic) hydrocarbon ring of 3 to 6 ring-forming carbon atoms ( For example, cyclopropyl, cyclobutyl, cyclopentyl or cyclohexyl), which is optionally substituted by one or more (such as 1 to 3) suitable substituents, such as methyl substituted cyclopropyl.
- aryl refers to an all-carbon monocyclic or fused-ring polycyclic aromatic group having a conjugated pi-electron system.
- C6-14 aryl means an aromatic group containing from 6 to 14 carbon atoms, such as phenyl or naphthyl.
- the aryl group is optionally substituted with one or more (such as 1 to 3) suitable substituents (e.g., halogen, -OH, -CN, -NO 2 , C 1-6 alkyl, etc.).
- aralkyl preferably denotes an aryl-substituted alkyl group, wherein the aryl group and the alkyl group are as defined herein.
- the aryl group can have from 6 to 14 carbon atoms and the alkyl group can have from 1 to 6 carbon atoms.
- Exemplary aralkyl groups include, but are not limited to, benzyl, phenylethyl, phenylpropyl, phenylbutyl.
- heteroaryl refers to a monovalent monocyclic, bicyclic or tricyclic aromatic ring system having 5, 6, 7, 8, 9, 10, 11, 12, 13 or 14 rings.
- the heteroaryl is selected from the group consisting of thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thia A oxazolyl group or the like, and a benzo derivative thereof; or a pyridyl group, a pyridazinyl group, a pyrimidinyl group, a pyrazinyl group, a triazinyl group or the like, and a benzo derivative thereof.
- halo or halogen group, as used herein, is defined to include F, Cl, Br or I.
- alkylthio refers to an alkyl group, as defined above, appended to the parent molecular moiety through a sulfur atom.
- Representative examples of C1-6 alkylthio include, but are not limited to, methylthio, ethylthio, tert-butylthio, and hexylthio.
- the 3 to 14 membered nitrogen heterocycle is a group having 3 to 14 carbon atoms and a hetero atom (at least one of which is a nitrogen atom) in the ring, including but not limited to a ternary nitrogen heterocyclic ring (eg, Aziridine), a quaternary nitrogen heterocyclic ring (such as azetidinyl), a five-membered nitrogen heterocyclic ring (such as pyrrolyl, tetrahydropyrrolyl, pyrrolinyl, pyrrolidinone, imidazolyl, imidazolidine) a group, an imidazolinyl group, a pyrazolyl group, a pyrazolinyl group, a six-membered nitrogen heterocyclic ring (such as piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl), a seven-membered nitrogen heterocyclic ring, and the like.
- substituted means that one or more (eg, 1, 2, 3 or 4) hydrogens on the designated atom are replaced by a selected group, provided that the specified atom is not exceeded in the current situation. Normal valence and the substitution forms a stable compound. The number of alternative groups selected is permissible when such combinations form stable compounds.
- substituent may be unsubstituted or (2) substituted. If a carbon of a substituent is described as being optionally substituted by one or more of the list of substituents, one or more hydrogens on the carbon may be replaced individually and/or together by an optional substituent that is independently selected. If the nitrogen of the substituent is described as being optionally substituted by one or more of the list of substituents, then one or more hydrogens on the nitrogen may each be replaced by an independently selected optional substituent.
- each substituent may be the same or different than another (other) substituent.
- one or more means 1 or more than 1, such as 2, 3, 4, 5 or 10 under reasonable conditions.
- a point of attachment of a substituent may come from any suitable position of the substituent.
- the invention also includes all pharmaceutically acceptable isotopically-labeled compounds which are identical to the compounds of the invention, except that one or more atoms are of the same atomic number but the atomic mass or mass number differs from the atomic mass prevailing in nature. Or atomic substitution of mass.
- isotopes suitable for inclusion in the compounds of the invention include, but are not limited to, isotopes of hydrogen (e.g., hydrazine (D, 2 H), hydrazine (T, 3 H)); isotopes of carbon (e.g., 11 C, 13 C) And 14 C); isotope of chlorine (eg 36 Cl); isotope of fluorine (eg 18 F); isotope of iodine (eg 123 I and 125 I); isotopes of nitrogen (eg 13 N and 15 N); isotopes of oxygen (eg 15 O, 17 O and 18 O); phosphorus isotopes (eg 32 P); and sulfur isotopes (eg 35 S).
- isotopes of hydrogen e.g., hydrazine (D, 2 H), hydrazine (T, 3 H)
- isotopes of carbon e.g., 11 C, 13 C) And 14 C
- Certain isotopically-labeled compounds of the invention are useful in drug and/or substrate tissue distribution studies (e.g., assays).
- the pharmaceutically acceptable solvates of the present invention include those in which the crystallization solvent can be substituted with an isotope, for example, D 2 O, acetone-d 6 or DMSO-d 6 .
- stereoisomer denotes an isomer formed by at least one asymmetric center.
- asymmetric center which can produce a racemic mixture, a single enantiomer, a mixture of diastereomers, and Separate diastereomers.
- Specific individual molecules can also exist as geometric isomers (cis/trans).
- the compounds of the invention may exist as mixtures (often referred to as tautomers) of two or more different forms in a rapidly balanced structure.
- tautomers include keto-enol tautomers, phenol-keto tautomers, nitroso-oxime tautomers, imine-enamine tautomers Wait.
- a dihydropyrimidinyl group can exist in equilibrium in the following tautomeric forms: It is to be understood that the scope of the present application covers all such ratios in any ratio (eg, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99). %) isomer or a mixture thereof.
- Solid line (-), solid wedge can be used in this paper.
- Virtual wedge Or wavy line The carbon-carbon bonds of the compounds of the invention are depicted.
- a solid line is used to delineate a bond design that is bonded to an asymmetric carbon atom, including all possible stereoisomers at that carbon atom (eg, specific enantiomers, racemic mixtures, etc.).
- a wavy line is used to delineate a bond representation linked to an alkenylene group, including all possible stereoisomers at the chemical bond (eg, a particular cis-trans isomer, a mixture of cis-trans isomers in any ratio, or Racemic mixture, etc.).
- the use of a solid or virtual wedge to characterize the bond to an asymmetric carbon atom indicates the presence of the stereoisomers shown.
- solid and virtual wedges are used to define relative stereochemistry rather than absolute stereochemistry.
- the compounds of the invention may be stereoisomers (including cis and trans isomers, optical isomers (eg, R and S enantiomers), diastereomers, Geometric isomers, rotamers, conformers, atropisomers, and mixtures thereof exist.
- the compounds of the invention may exhibit more than one type of isomerism and consist of a mixture thereof (e.g., a racemic mixture and a diastereomeric pair).
- the invention encompasses all possible crystalline forms or polymorphs of the compounds of the invention, which may be a single polymorph or a mixture of more than one polymorph in any ratio.
- the compounds of the invention may exist in free form for treatment or, where appropriate, in the form of their pharmaceutically acceptable derivatives.
- pharmaceutically acceptable derivatives include, but are not limited to, pharmaceutically acceptable salts, esters, solvates, metabolites, isotopically labeled compounds or prodrugs, which are administered to a patient in need thereof
- the compound of the invention, or a metabolite or residue thereof, can be provided directly or indirectly after the drug.
- a “compound of the invention” it is also intended to encompass the various derivative forms described above for the compound.
- the pharmaceutically acceptable salts of the compounds of the present invention include the acid addition salts and base addition salts thereof.
- Suitable acid addition salts are formed from acids which form pharmaceutically acceptable salts, including suitable inorganic and organic acids. Examples include aspartate, benzoate, bicarbonate/carbonate, hydrogen sulfate/sulfate, fumarate, glucoheptonate, gluconate, glucuronate, hexafluorophosphate Phosphate, hydrobromide/bromide, hydroiodide/iodide, maleate, malonate, methyl sulfate, naphthylate, nicotinate, nitrate , orotate, oxalate, palmitate and other similar salts.
- Suitable base addition salts are formed from bases which form pharmaceutically acceptable salts, including suitable inorganic bases and organic bases. Examples include aluminum salts, arginine salts, choline salts, diethylamine salts, lysine salts, magnesium salts, meglumine salts, potassium salts, and other similar salts.
- esters means an ester derived from a compound of the formulae herein, which includes a physiologically hydrolyzable ester, which is hydrolyzable under physiological conditions to release the free acid or alcohol form of the invention.
- Compound The compounds of the invention may also be esters per se.
- the compound of the present invention may exist in the form of a solvate (e.g., hydrate) wherein the compound of the present invention contains a polar solvent as a structural element of the crystal lattice of the compound, particularly such as water, methanol or ethanol.
- a polar solvent as a structural element of the crystal lattice of the compound, particularly such as water, methanol or ethanol.
- the amount of polar solvent, particularly water may be present in stoichiometric or non-stoichiometric ratios.
- metabolites of the compounds of the invention i.e., substances formed in vivo upon administration of a compound of the invention. Such products may be produced, for example, by oxidation, reduction, hydrolysis, amidation, deamidation, esterification, enzymatic hydrolysis, and the like of the administered compound. Accordingly, the invention includes metabolites of the compounds of the invention, including compounds prepared by contacting a compound of the invention with a mammal for a time sufficient to produce a metabolic product thereof.
- the invention further includes within its scope prodrugs of the compounds of the invention which are certain derivatives of the compounds of the invention which may or may not be pharmacologically active, which are administered to or into the body It can be converted to a compound of the invention having the desired activity by, for example, hydrolytic cleavage.
- prodrugs will be functional group derivatives of the compounds which are readily converted in vivo to the desired therapeutically active compound. Additional information on the use of prodrugs can be found in "Pro-drugs as Novel Delivery Systems", Volume 14, ACS Symposium Series (T. Higuchi and V. Stella) and "Bioreversible Carriers in Drug Design," Pergamon Press, 1987 ( Edited by EBRoche, American Pharmaceutical Association).
- Prodrugs of the invention may, for example, be known by those skilled in the art as “pro-moiety” (e.g., “Design of Prodrugs", H. Bundgaard (Elsevier, 1985))" It is prepared in place of the appropriate functional groups present in the compounds of the invention.
- the invention also encompasses compounds of the invention containing a protecting group.
- a protecting group In any process for preparing a compound of the invention, it may be necessary and/or desirable to protect a sensitive group or reactive group on any of the molecules of interest, thereby forming a chemically protected form of the compound of the invention. This can be achieved by conventional protecting groups, for example, as described in Protective Groups in Organic Chemistry, ed. JFW McOmie, Plenum Press, 1973; and TW Greene & PGM Wuts, Protective Groups in Organic Synthesis, John Wiley & Sons, 1991. Those protecting groups, these references are incorporated herein by reference. Use known in the art The method can remove the protecting group at an appropriate subsequent stage.
- the term "about” refers to within ⁇ 10% of the stated value, preferably within ⁇ 5%, more preferably within ⁇ 2%.
- the invention provides a compound or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, isotopically labeled compound thereof or a pharmaceutically acceptable salt thereof Medicament wherein the compound has the structure of Formula I or Formula Ia:
- Ar 1 and Ar 2 are each independently selected from a C 6-14 aryl group and a 5-14 membered heteroaryl group, which are optionally selected from one or more selected from the group consisting of halogen, -OH, -CN, -NO 2 , -N Substituent substitution of (R) 2 , C 1-6 alkyl, halo C 1-6 alkyl, C 1-6 alkylthio and C 3-6 cycloalkyl;
- L is absent or selected from -O-, -S-, and -NR-;
- R 1 and R 2 are each independently selected from H (including 1 H, 2 H, 3 H), C 1-6 alkyl (eg, deuterated C 1-6 alkyl), and C 3-6 cycloalkyl;
- R 3 is a quaternary, five-, six- or seven-membered nitrogen heterocyclic ring selected from the following structures:
- R a , R a ' , R 4 , R 4 ' , R 5 , R 5 ' and R 6 are each independently selected from H, halogen, -OH, -COOH, -CN, -NO 2 , -N(R) 2 , C 1-6 alkyl, halo C 1-6 alkyl, -WC 1-6 alkyl, -C 1-6 alkylene-WR, -WC 1-6 alkylene -W'-R, -WC 2-6 alkenyl, -C 2-6 alkenylene-WR, -WC 2-6 alkenylene-W'-R and C 3-6 cycloalkyl, wherein The alkylene and alkenylene are optionally further separated by one or more W; or R a together with R a ' , R 4 together with R 5 and/or R 4 ' and R 5' together, each occurrence
- R 6 is attached to the ring carbon atom marked by * and / or ** in the above nitrogen heterocyclic structure
- R is each independently selected from the group consisting of H, C 1-6 alkyl and C 3-6 cycloalkyl;
- g is 1 or 2;
- t is 0, 1, 2 or 3, provided that t is not greater than the number of positions on the corresponding group that can be substituted, and when t is greater than 1, each R 6 may be the same or different.
- the invention provides a compound, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, isotopically labeled compound thereof, or a prodrug wherein the compound has the structure of Formula II or Formula IIa:
- the invention provides a compound, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, isotopically labeled compound thereof, or Prodrug, wherein Ar 1 is selected from:
- R c is each independently selected from the group consisting of F, Cl, Br, I, halo C 1-6 alkyl, C 1-6 alkyl, and C 3-6 cycloalkyl;
- R c is each independently selected from the group consisting of F, Cl, Br, I, C 1-6 alkyl and C 3-6 cycloalkyl;
- Ar 1 is more preferably from:
- Ar 1 is particularly preferred from:
- the invention provides a compound, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, isotopically labeled compound thereof, or Prodrug, wherein Ar 2 is selected from:
- R b is each independently selected from H, halogen, halo C 1-6 alkyl, C 1-6 alkyl and C 3-6 cycloalkyl at each occurrence; preferably, R b is present each time Each is independently selected from the group consisting of H, halogen, C 1-6 alkyl, and C 3-6 cycloalkyl;
- i 0, 1 or 2;
- Ar 2 is preferably selected from:
- L is -O-.
- R 1 and R 2 are each independently selected from H (including 1 H, 2 H, 3 H), methyl, ethyl, n-propyl and isopropyl.
- R 3 is a quaternary, penta, hexa or seven-membered nitrogen heterocycle selected from the group consisting of:
- R 6 is bonded to the ring carbon atom marked by * and / or ** in the above nitrogen heterocyclic structure.
- R 7 , R 7 ' , R 7a , R 7a′ are each independently selected from H, C 1-4 alkyl and C 3-6 cycloalkyl at each occurrence;
- R is selected from the group consisting of H, methyl, ethyl, propyl and cyclopropyl;
- n 0, 1, 2, 3 or 4;
- n 1, 2, 3 or 4;
- j 0, 1, or 2.
- R 3 is selected from the group consisting of
- R 3 is selected from the group consisting of
- the invention provides a compound, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, isotopically labeled compound thereof, or a pharmaceutically acceptable salt thereof Medicament wherein the compound has the structure:
- the invention provides a compound, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, isotopically labeled compound thereof, or a pharmaceutically acceptable salt thereof Medicament wherein the compound has the structure:
- the invention provides a compound, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, isotopically labeled compound thereof, or a pharmaceutically acceptable salt thereof Medicine wherein the compound has the following knot Structure:
- R 1 and R 2 are each independently selected from H (including 1 H, 2 H, 3 H), C 1-6 alkyl (eg, deuterated C 1-6 alkyl), and C 3-6 cycloalkyl, And R 1 is preferably methyl, ethyl, n-propyl, isopropyl or cyclopropyl;
- Q is -(CR a R a' ) g - or -O-;
- R a , R a ' , R 4 , R 5 and R 6 are each independently selected from H, halogen (eg F), -OH, -COOH, -CN, -NO 2 , -N(R) at each occurrence. 2 , C 1-6 alkyl, halo C 1-6 alkyl, -WC 1-6 alkyl, -C 1-6 alkylene-WR, -WC 1-6 alkylene-W'- R, -WC 2-6 alkenyl, -C 2-6 alkenylene-WR, -WC 2-6 alkenylene-W'-R and C 3-6 cycloalkyl, wherein the alkylene group The alkenylene group is optionally further separated by one or more W;
- R b is each independently selected from the group consisting of H, halogen, halogenated C 1-6 alkyl, C 1-6 alkyl, and C 3-6 cycloalkyl;
- R c is each independently selected from the group consisting of F, Cl, Br, I, halo C 1-6 alkyl, C 1-6 alkyl and C 3-6 cycloalkyl, and R c is preferably Cl Or Br;
- R 6 is attached to a ring carbon atom marked by * and / or ** in the general structure
- R is each independently selected from the group consisting of H, C 1-6 alkyl and C 3-6 cycloalkyl;
- g is 1 or 2;
- i 0, 1 or 2;
- n 0, 1, 2, 3 or 4;
- each R 6 may be the same or different.
- R b is each independently selected from the group consisting of H, halo, C 1-6 alkyl, and C 3-6 cycloalkyl;
- R c is each independently selected from the group consisting of F, Cl, Br, I, C 1-6 alkyl, and C 3-6 cycloalkyl, and R c is preferably Cl or Br;
- the invention provides a compound, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, isotopically labeled compound thereof, or a pharmaceutically acceptable salt thereof Medicament wherein the compound has the structure:
- the invention encompasses compounds obtained by any suitable combination of the various embodiments.
- the invention provides a compound, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, isotopically labeled compound thereof, or a prodrug wherein the compound is selected from the group consisting of
- Embodiments of the invention provide methods of making the compounds of the invention, the methods comprising the steps of:
- Hal is selected from the group consisting of F, Cl, Br, and I;
- the halogenating agent is selected from the group consisting of Cl 2 , Br 2 , I 2 , N-chlorosuccinimide, N-bromosuccinimide, and N-iodosuccinimide;
- Step 1 in the presence of an alkali metal salt (such as potassium acetate, sodium acetate, potassium carbonate, sodium carbonate, cesium carbonate, sodium hydrogencarbonate, etc.) in a protic solvent (eg 2,2,2-trifluoroethanol, 2,2- In difluoroethanol, 2-fluoroethanol, ethanol, fluoromethanol, hexafluoroisopropanol, etc.);
- an alkali metal salt such as potassium acetate, sodium acetate, potassium carbonate, sodium carbonate, cesium carbonate, sodium hydrogencarbonate, etc.
- a protic solvent eg 2,2,2-trifluoroethanol, 2,2- In difluoroethanol, 2-fluoroethanol, ethanol, fluoromethanol, hexafluoroisopropanol, etc.
- Step two is carried out in an aprotic solvent (for example, carbon tetrachloride, dichloromethane, 1,2-dichloroethane, etc.);
- an aprotic solvent for example, carbon tetrachloride, dichloromethane, 1,2-dichloroethane, etc.
- Step 3 in an aprotic solvent eg dichloromethane, 1,2-dichloroethane, acetonitrile, tetrahydrofuran, 2-methyltetrahydrofuran, diethyl ether, tert-butyl methyl ether, N,N-dimethylformamide, N , N-dimethylacetamide, N-methylpyrrolidone, etc.
- an organic base eg N,N-diisopropylethylamine, triethylamine, 1,8-diazabicyclo[5.4.0 ]-7-undecene, 4-dimethylaminopyridine, N-methylmorpholine, N-methylpiperidine, N-methyltetrahydropyrrole, etc.
- inorganic bases eg potassium acetate, sodium acetate, carbonic acid
- R 2 in the compound of the formula I or formula Ia of the present invention is a C 1-6 alkyl group, it can also be synthesized by a method comprising the following steps:
- R 2 ' is H or C 1-5 alkyl
- Hal is selected from the group consisting of F, Cl, Br, and I;
- the halogenating agent is selected from the group consisting of Cl 2 , Br 2 , I 2 , N-chlorosuccinimide, N-bromosuccinimide, and N-iodosuccinimide;
- Step I is in a Lewis acid (for example, a triflate (such as indium triflate, ytterbium triflate, etc.), a triflate (such as trimethylsilyl trifluoromethanesulfonate) , boron trifluoride, aluminum trichloride, etc.) in the presence of a non-polar solvent (such as o-xylene, toluene, anisole, etc.);
- a Lewis acid for example, a triflate (such as indium triflate, ytterbium triflate, etc.), a triflate (such as trimethylsilyl trifluoromethanesulfonate) , boron trifluoride, aluminum trichloride, etc.) in the presence of a non-polar solvent (such as o-xylene, toluene, anisole, etc.);
- a non-polar solvent such as o-xylene, toluene, anisole
- Step II is in an organic base (for example, N,N-diisopropylethylamine, triethylamine, 1,8-diazabicyclo[5.4.0]-7-undecene, 4-dimethylaminopyridine, N -methylmorpholine, N-methylpiperidine, N-methyltetrahydropyrrole, etc.) or inorganic bases (eg potassium acetate, sodium acetate, potassium carbonate, sodium carbonate, cesium carbonate, sodium hydrogencarbonate, sodium hydride, uncle In the presence of potassium butoxide, etc., in an aprotic solvent (eg dichloromethane, 1,2-dichloroethane, acetonitrile, tetrahydrofuran, 2-methyltetrahydrofuran, diethyl ether, tert-butyl methyl ether, N, N- In dimethylformamide, N,N-dimethylacetamide, N-methylpyrrolidone, etc.);
- Step III is carried out in an aprotic solvent such as carbon tetrachloride, dichloromethane, 1,2-dichloroethane, etc.;
- Step IV is in an aprotic solvent (eg dichloromethane, 1,2-dichloroethane, acetonitrile, tetrahydrofuran, 2-methyltetrahydrofuran, diethyl ether, tert-butyl methyl ether, N,N-dimethylformamide, N , N-dimethylacetamide, N-methylpyrrolidone, etc.) in an organic base (eg N,N-diisopropylethylamine, triethylamine, 1,8-diazabicyclo[5.4.0 ]-7-undecene, 4-dimethylaminopyridine, N-methylmorpholine, N-methylpiperidine, N-methyltetrahydropyrrole, etc.) or inorganic bases (eg potassium acetate, sodium acetate, carbonic acid) It is carried out in the presence of potassium, sodium carbonate, cesium carbonate, sodium hydrogencarbonate, sodium hydride, potassium t-
- R 2 in the compound of Formula I or Formula Ia of the present invention is a methyl group
- it can also be synthesized by a method comprising the following steps:
- compositions and methods of treatment are provided.
- the present invention provides a pharmaceutical composition
- a pharmaceutical composition comprising a prophylactically or therapeutically effective amount of a compound of the present invention or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate thereof, A metabolite, an isotopically labeled compound or prodrug, and one or more pharmaceutically acceptable carriers or excipients.
- the pharmaceutical composition may further comprise one or more Other therapeutic agents, such as other therapeutic agents for the prevention or treatment of viral diseases.
- the invention also provides a process for the preparation of a pharmaceutical composition
- a pharmaceutical composition comprising the compound of the invention or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate thereof
- a metabolite, isotopically labeled compound or prodrug is combined with one or more pharmaceutically acceptable carriers or excipients.
- the present invention provides a compound of the present invention or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, isotopically labeled compound or prodrug thereof or the present invention
- a pharmaceutical composition of the invention in the manufacture of a medicament for the prevention or treatment of a viral disease.
- the present invention provides a compound of the present invention or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, isotopically labeled compound or prodrug thereof or the present invention
- a pharmaceutical composition of the invention for use in the prevention or treatment of a viral disease.
- the invention provides a method of preventing or treating a viral disease, the method comprising administering to an individual in need thereof an effective amount of a compound of the invention, or a pharmaceutically acceptable salt, ester, or stereoisod thereof
- a construct, tautomer, polymorph, solvate, metabolite, isotopically labeled compound or prodrug or a pharmaceutical composition of the invention comprising administering to an individual in need thereof an effective amount of a compound of the invention, or a pharmaceutically acceptable salt, ester, or stereoisod thereof.
- the compounds of the present invention exert an antiviral effect by inhibiting the assembly of capsid proteins.
- the compounds of the invention are useful in the treatment of any virus involved in the assembly of capsid proteins in the process of affecting a host, including but not limited to hepatitis A virus (HAV), hepatitis B virus (HBV), hepatitis C virus (HCV), influenza virus, herpes virus (HSV) and human immunodeficiency virus (HIV).
- HAV hepatitis A virus
- HBV hepatitis B virus
- HCV hepatitis C virus
- influenza virus influenza virus
- HSV herpes virus
- HSV human immunodeficiency virus
- viral diseases which can be prevented and treated using the compounds of the present invention include, but are not limited to, viral hepatitis A, hepatitis B virus, hepatitis C virus, influenza, herpes, and acquired immunodeficiency syndrome. (AIDS), and related symptoms or diseases caused by the above diseases (for example, inflammation, liver fibrosis, cirrhosis, liver cancer, etc.).
- “Pharmaceutically acceptable carrier” in the context of the present invention means a diluent, adjuvant, excipient or vehicle with which the therapeutic agent is administered, and which is suitable for contacting humans and/or within the scope of sound medical judgment. Tissues of other animals without excessive toxicity, irritation, allergic reactions, or other problems or complications corresponding to a reasonable benefit/risk ratio.
- Pharmaceutically acceptable carriers that can be used in the pharmaceutical compositions of the present invention include, but are not limited to, sterile liquids such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, minerals. Oil, sesame oil, etc. Water is an exemplary carrier when the pharmaceutical composition is administered intravenously. It is also possible to use physiological saline and an aqueous solution of glucose and glycerin as a liquid carrier, particularly for injection.
- Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, maltose, chalk, silica gel, sodium stearate, glyceryl monostearate, talc, sodium chloride, skimmed milk powder, glycerin, propylene glycol, water, Ethanol and the like.
- the composition may also contain minor amounts of wetting agents, emulsifying agents or pH buffering agents as needed.
- Oral formulations may contain standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharin, cellulose, magnesium carbonate, and the like. Examples of suitable pharmaceutically acceptable carriers are as described in Remington's Pharmaceutical Sciences (1990).
- compositions of the invention may act systemically and/or locally.
- they may be administered in a suitable route, for example by injection (for example intravenous, intraarterial, subcutaneous, intraperitoneal, intramuscular, including instillation) or transdermal administration; or by oral, buccal, or oral administration.
- routes of administration it can be administered in a suitable dosage form.
- the dosage forms include, but are not limited to, tablets, capsules, troches, hard candy, powders, sprays, creams, ointments, suppositories, gels, pastes, lotions, ointments, aqueous suspensions. Injectable solutions, elixirs, syrups.
- an effective amount refers to an amount of a compound that, to a certain extent, relieves one or more symptoms of the condition being treated after administration.
- the dosing regimen can be adjusted to provide the optimal desired response. For example, a single administration may be administered, several divided doses may be administered over time, or the dose may be proportionally reduced or increased depending on the condition being treated. It is noted that the dose value can vary with the type and severity of the condition and can include single or multiple doses. It is to be further understood that for any particular individual, the particular dosage regimen should be adjusted over time based on individual needs.
- an effective dose will be from about 0.0001 to about 50 mg per kg body weight per day, for example from about 0.01 to about 10 mg/kg/day (single or divided doses). For a 70 kg person, this would add up to about 0.007 mg/day to about 3500 mg/day, such as from about 0.7 mg/day to about 700 mg/day.
- a dose level that is not higher than the lower limit of the aforementioned range may be sufficient, while in other cases, a larger dose may still be employed without causing any harmful side effects, provided that the larger The dose is divided into several smaller doses to be administered throughout the day.
- the amount or amount of the compound of the present invention in the pharmaceutical composition may be from about 0.01 mg to about 1000 mg, suitably from 0.1 to 500 mg, preferably from 0.5 to 300 mg, more preferably from 1 to 150 mg, particularly preferably from 1 to 50 mg, for example, 1.5 mg, 2mg, 4mg, 10mg, 25mg Wait.
- treating means reversing, alleviating, inhibiting the progression of a condition or condition to which such a term applies or one or more symptoms of such a condition or condition, or Prevention of such a condition or condition or one or more symptoms of such condition or condition.
- “Individual” as used herein includes human or non-human animals.
- Exemplary human individuals include a human individual (referred to as a patient) or a normal individual having a disease, such as the disease described herein.
- “Non-human animals” in the present invention include all vertebrates, such as non-mammals (eg, birds, amphibians, reptiles) and mammals, such as non-human primates, domestic animals, and/or domesticated animals (eg, sheep, dogs). , cats, cows, pigs, etc.).
- the pharmaceutical composition of the present invention may further comprise one or more additional therapeutic or prophylactic agents, which are anti-hepatitis B drugs including, but not limited to, Lamiv Ding, telbivudine, entecavir, adefovir dipivoxil, tenofovir, tenofovir disoproxil fumarate and tenofovir alafenamide fumarate.
- additional therapeutic or prophylactic agents which are anti-hepatitis B drugs including, but not limited to, Lamiv Ding, telbivudine, entecavir, adefovir dipivoxil, tenofovir, tenofovir disoproxil fumarate and tenofovir alafenamide fumarate.
- LC-MS was detected on an Aglient 1200 liquid chromatograph using an Aglient 6120 Quadrupole mass spectrometer at 214 nm and 254 nm.
- Preparative liquid chromatography using SHIMADZU CBM-20A and Aglient 1260 to prepare a liquid phase meter, C18 OBD 19 ⁇ 150mm 5 ⁇ M preparative column, detection wavelength 214nm, mobile phase A is water, mobile phase B is acetonitrile (add 0.5 ⁇ formic acid), Linear gradient elution as follows:
- Step 2 6-(Bromomethyl)-4-(2-chloro-4-fluorophenyl)-2-(thiazol-2-yl)-1,4-dihydropyrimidine-5-carboxylic acid ethyl ester (1 -2) synthesis
- Step 3 4-(2-Chloro-4-fluorophenyl)-6-((3,3-difluoro-4-hydroxypiperidin-1-yl)methyl)-2-(thiazol-2-yl) Synthesis of ethyl 4-1,4-dihydropyrimidine-5-carboxylate (10-88)
- Step 1 4-(2-Chloro-4-fluorophenyl)-6-methyl-2-(thiazol-2-yl)-1,4-dihydropyrimidine-5-carboxylic acid ethyl ester (3-1) Separation
- Step 1 Synthesis of benzyl 3,3-difluoro-4-hydroxypiperidine-1-carboxylate (5-2)
- 3,3-Difluoropiperidin-4-ol hydrochloride (100 mg, 0.73 mmol) was dissolved in dichloromethane (2 mL) and triethylamine (147 mg, 1.46 mmol) A solution of N-(benzyloxycarbonyloxy) succinimide (12 mg, 0.48 mmol) in dichloromethane (2 mL) was evaporated. The title compound was obtained after workup of 100 mg. ESI-MS (m/z): 272.2 [M+H] + .
- Step 2 Synthesis of 4-(2-ethoxy-2-oxoethoxy)-3,3-difluoropiperidine-1-carboxylic acid benzyl ester (5-3)
- Step 5 2-((1-((6-(2-Chloro-4-fluorophenyl)-5-(ethoxycarbonyl)-2-(thiazol-2-yl)-3,6-dihydropyrimidine) Synthesis of 4-yl)methyl)-3,3-difluoropiperidin-4-yl)oxy)acetic acid (10-95)
- the title compound was obtained in a manner similar to that described in the third step of Example 1 except that the compound (5-5) was used instead of 3,3-difluoropiperidin-4-ol hydrochloride.
- the title compound 4 mg was obtained by a method similar to that described in the first step of Example 1 (3,3-difluoroazetidine hydrochloride instead of 3,3-difluoropiperidin-4-ol hydrochloride). .
- Step 2 (R)-1-((6-(2-chloro-4-fluorophenyl)-5-(ethoxycarbonyl)-2-(thiazol-2-yl)-3,6-dihydropyrimidine Synthesis of 4-yl)methyl)-3-fluoroazetidin-3-carboxylic acid (10-11)
- Step 1 Synthesis of tert-butyl 3,3-difluoro-4-methoxypiperidine-1-carboxylate (8-2)
- the title compound 50 mg was obtained by a method similar to that described in the first step of Example 1 (substituting compound (8-3) for 3,3-difluoropiperidin-4-ol hydrochloride).
- Step 2 (R)-4-(2-Chloro-4-fluorophenyl)-6-((3R,4R)-3-fluoro-4-hydroxytetrahydropyrrol-1-yl)methyl)- Synthesis of ethyl 2-(thiazol-2-yl)-1,4-dihydropyrimidine-5-carboxylate (10-34)
- Step 1 Synthesis of (3R,4R)-3-(2-ethoxy-2-oxoethoxy)-4-fluorotetrahydropyrrole-1-carboxylic acid tert-butyl ester (10-b)
- Step 4 2-(((3R,4R)-1-(((R)-6-(2-chloro-4-fluorophenyl)-5-(ethoxycarbonyl)-2-(thiazole-2-) Synthesis of 3-(6-6-dihydropyrimidin-4-yl)methyl)-4-fluorotetrahydropyrrol-3-yl)oxy)acetic acid (10-36)
- the title compound 33 mg was obtained by a method similar to the one described in the first step of Example 1 (substituting compound (11-5) for 3,3-difluoropiperidin-4-ol hydrochloride).
- Step 4 2-((1-((R)-6-(2-Chloro-4-fluorophenyl)-5-(ethoxycarbonyl)-2-(thiazol-2-yl)-3,6 Synthesis of dihydropyrimidin-4-yl)methyl)-4,4-difluorotetrahydropyrrol-3-yl)oxy)acetic acid
- the title compound 25 mg was obtained by a method similar to that described in the first step of Example 1 (substituting compound (12-4) for 3,3-difluoropiperidin-4-ol hydrochloride).
- Step 2 Synthesis of 3-formylmorpholine-4-carboxylic acid tert-butyl ester (13-3)
- Step 3 Synthesis of (E)-3-(3-ethoxy-3-oxoprop-1-en-1-yl)morpholine-4-carboxylic acid tert-butyl ester (13-4)
- Step 6 (E)-3-(4-((R)-6-(2-chloro-4-fluorophenyl)-5-(ethoxycarbonyl)-2-(thiazol-2-yl)- Synthesis of 3,6-dihydropyrimidin-4-yl)methyl)morpholin-3-yl)acrylic acid (10-182)
- Step 1 Synthesis of 3-((2-(tert-butoxy)-2-oxoethoxy)methyl)morpholine-4-carboxylic acid tert-butyl ester (15-2)
- tert-butyl 3-(hydroxymethyl)morpholine-4-carboxylate 200 mg, 0.99 mmol
- tetrahydrofuran 6 mL
- sodium hydride 47.3 mg, 1.2 mmol
- tert-butyl bromoacetate 192 mg, 0.99 mmol
- the reaction mixture was poured into 10 mL of water, and the pH was adjusted to 2 to 3 with 1N aqueous hydrochloric acid, and extracted with ethyl acetate.
- Step 3 2-((4-((R)-6-(2-chloro-4-fluorophenyl)-5-(ethoxycarbonyl)-2-(thiazol-2-yl)-3,6 Synthesis of dihydropyrimidin-4-yl)methyl)morpholin-3-yl)methoxy)acetic acid (10-162)
- the title compound 73 mg was obtained by a method similar to that described in the first step of Example 1 (substituting compound (15-3) for 3,3-difluoropiperidin-4-ol hydrochloride).
- Step 3 Synthesis of tert-butyl 3,3-difluoro-5-(hydroxymethyl)piperidine-1-carboxylate (16-4)
- Step 4 5-((2-(tert-Butoxy)-2-oxoethoxy)methyl)-3,3-difluoropiperidine-1-carboxylic acid tert-butyl ester (16-5) synthesis
- Step 6 2-((1-((R)-6-(2-chloro-4-fluorophenyl)-5-(ethoxycarbonyl)-2-(thiazol-2-yl)-3,6 Synthesis of dihydropyrimidin-4-yl)methyl)-5,5-difluoropiperidin-3-yl)methoxy)acetic acid (10-136)
- Step 1 Synthesis of tert-butyl 3,3-difluoro-5-formylpiperidine-1-carboxylate (17-2)
- tert-Butyl 3,3-difluoro-5-(hydroxymethyl)piperidine-1-carboxylate (17-1) (50 mg, 0.2 mmol) was dissolved in dichloromethane (2.0 mL). After completely dissolved, the reaction was cooled to 0 ° C, and Dess-Martin reagent (102 mg, 0.24 mmol) was added with stirring. After the addition, the reaction was carried out for 3 h at room temperature. A large amount of a white solid was precipitated from the reaction mixture, and the filtrate was evaporated.
- Step 5 (E)-3-(1-(((R)-6-(2-chloro-4-fluorophenyl)-5-(ethoxycarbonyl)-2-(thiazol-2-yl)-) Synthesis of 3,6-dihydropyrimidin-4-yl)methyl)-5,5-difluoropiperidin-3-yl)acrylic acid (10-116)
- the title compound 10 mg was obtained by a method similar to the one described in the first step of Example 1 (substituting compound (17-5) for 3,3-difluoropiperidin-4-ol hydrochloride).
- Step 1 Synthesis of 2-((2-ethoxy-2-oxoethoxy)methyl)-4,4-difluorotetrahydropyrrole-1-carboxylic acid tert-butyl ester (18-2)
- Step 4 2-((1-((R)-6-(2-Chloro-4-fluorophenyl)-5-(ethoxycarbonyl)-2-(thiazol-2-yl)-3,6 Synthesis of dihydropyrimidin-4-yl)methyl)-4,4-difluorotetrahydropyrrole-2-yl)methoxy)acetic acid (10-56)
- the title compound 25 mg was obtained by a method similar to the one described in the first step of Example 1 (substituting compound (18-4) for 3,3-difluoropiperidin-4-ol hydrochloride).
- Step 1 Synthesis of 2-(hydroxymethyl)morpholine-4-carboxylic acid tert-butyl ester (19-2)
- the target compound 70 mg was obtained from the compound (19-2) by the operation similar to the procedure of the procedure of the step of the above.
- Step 1 Synthesis of 2-(((2-methoxy-2-oxoethyl)(methyl)amino)methyl)morpholine-4-carboxylic acid tert-butyl ester (20-2)
- Step 4 N-((4-((R)-6-(2-chloro-4-fluorophenyl)-5-ethoxycarbonyl-2-(thiazol-2-yl)-3,6-di Synthesis of Hydropyrimidin-4-yl)methyl)morpholin-2-yl)methyl)-N-methylglycine (10-168)
- Step 1 Synthesis of (E)-2-acetyl-3-(4-fluorophenyl)but-2-enoic acid ethyl ester (21-1)
- Step 2 Synthesis of ethyl 4-(4-fluorophenyl)-4,6-dimethyl-2-(thiazol-2-yl)-1,4-dihydropyrimidine-5-carboxylate
- Step 3 to Step 4 2-((1-(((S)-5-(ethoxycarbonyl))-6-(4-fluorophenyl)-6-methyl-2-(thiazol-2-yl)) -3,6-dihydropyrimidin-4-yl)methyl)-3,3-difluoropiperidin-4-yl)oxy)acetic acid (10-224)
- the target compound 15 mg was obtained from the compound (21-2') of Example 21, using a procedure similar to that described in Steps 21 to 4 of Example.
- Example 21 The compound (21-2') of Example 21 was reacted with 2-((4,4-difluorotetrahydropyrrole) after the bromination reaction using a procedure similar to that described in the first step and the third step of Example 1.
- -3-yl)oxy)acetic acid Compound (12-4) in Example 12 (29 mg, 0.16 mmol)
- Step 1 Synthesis of (S)-2-formylmorpholine-4-carboxylic acid tert-butyl ester (25-2)
- Step 2 (R,E)-2-(3-(tert-Butoxy)-3-oxoprop-1-en-1-yl)morpholine-4-carboxylic acid tert-butyl ester (25-3) synthesis
- Step 4 (E)-3-((R)-4-(((R)-6-(2-chloro-4-fluorophenyl)-5-(ethoxycarbonyl)-2-(thiazole-2) Synthesis of 3-yl-3,6-dihydropyrimidin-4-yl)methyl)morpholin-2-yl)acrylic acid (10-226)
- Step 2 Synthesis of (S)-2-(methoxy(methyl)carbamoyl)morpholine-4-carboxylic acid tert-butyl ester (39-3)
- Step 3 Synthesis of (S)-2-acetylmorpholine-4-carboxylic acid tert-butyl ester (39-4)
- Step 4 (R,E)-2-(4-(tert-Butoxy)-4-oxobut-2-en-2-yl)morpholine-4-carboxylic acid tert-butyl ester (39-5) synthesis
- Step 6 (E)-3-((R)-4-(((R)-6-(2-chloro-4-fluorophenyl)-5-(methoxycarbonyl)-2-(thiazole-2) Synthesis of 3-yl-3,6-dihydropyrimidin-4-yl)methyl)morpholin-2-yl)but-2-enoic acid (10-246)
- control compound 1 The structure of the control compound 1 is The structure of the control compound 2 is The structure of the control compound 3 is
- HBV hepatitis B virus
- HepG2.2.15 cells in logarithmic growth phase were seeded in 96-well plates at a cell concentration of 40/ ⁇ L. Incubate for 3 days at 37 ° C in a 5% CO 2 incubator; replace with fresh medium (200 ⁇ L/well) before adding the compound.
- the mother liquor concentration of each of the examples was 200 ⁇ M.
- the highest concentration was 200 ⁇ M, diluted to a number of different concentrations in DMSO, and 1 ⁇ L of the test compound was placed in the corresponding medium well.
- the final test concentrations of the compounds were 0.06, 0.24, 0.98, 3.9, 15.6, 62.5, 250, 1000 nM (using Calculate the half effective concentration (EC 50 )).
- the test results are shown in Table 1-1 and Table 1-2.
- the tested compounds have strong inhibitory activity against hepatitis B virus (HBV).
- the anti-HBV virus activity of the compound of the present invention having a single configuration was about 10 times that of the control compound 2.
- the compound of the present invention has strong inhibitory activity against hepatitis B virus (HBV).
- the remaining compounds of the invention have inhibitory activities similar to those described above.
- the test compound was diluted to 30 mM with DMSO, diluted to a maximum concentration of 30 mM, and diluted to a plurality of different concentrations.
- 0.2 ⁇ L of each compound was added to a 384-well plate, and 2000/50 ⁇ L of HepG2.2.15 was added to each well.
- the highest concentration of the test compound was 150 ⁇ M; 1 ⁇ L of DMSO was added to the corresponding wells as a control.
- the cells were co-cultured for 4 days at 37 ° C in a 5% CO 2 incubator. After 4 days, 50 ⁇ L of CellTiter-Glo was added to each well to perform a plate reading test, and a half-cytotoxic concentration (CC 50 ) value was calculated.
- Table 2 The test results are shown in Table 2.
- hERG human Ether-a-go-go Related Gene
- IKr inhibition is the most important mechanism for drug-induced QT interval prolongation.
- the criterion was that if the compound IC 50 > 10 ⁇ M, it was judged that the compound had no inhibitory effect on hERG.
- test compounds on the hERG potassium channel were tested for the role Predictor TM hERG Fluorescence Polarization Assay. The test results are shown in Table 3 below:
- Control compound 1 2.85 Control compound 2 1 to 10 10-36 of the tenth embodiment >10 10-40 of the eleventh embodiment >10 10-42 of the twelfth embodiment >10 10-88 of the first embodiment >10 Isomer B of Example 4 >10 Tetraisomer A of the Examples >10 10-93 of the second embodiment >10 10-95 of the fifth embodiment >10 Example twenty-fifth 10-226 >10
- Control Compound 1 and Control Compound 2 have different degrees of cardiotoxicity (significant inhibition of hERG potassium channel in cardiomyocytes) and thus have a potential risk of inducing arrhythmia; and the present invention tested The compound has no hERG potassium channel inhibition, no obvious cardiotoxicity, and thus has higher safety. Other compounds of the invention have similar safety.
- Test compounds were administered to male SD rats by intravenous (iv) and gavage (po), respectively.
- the doses administered by iv and po were 1 mg/kg and 2 mg/kg, respectively, and the iv system was 5% DMSO. : 5% solutol: 90% saline, the vehicle system was 0.5% MC when po was administered.
- Blood was collected at various time points after iv administration and po administration for PK studies. Plasma samples and liver tissue samples were subjected to protein precipitation and subjected to LC-MS/MS analysis.
- the mass spectrometer is API 5500, the liquid chromatograph is Waters ACQUITY I CLASS system; the column is Agela ASB C 18 column (2.1mm ⁇ 50mm, 1.9 ⁇ m); the mobile phase A phase is water +0.1% formic acid, and the B phase is acetonitrile.
- the flow rate was 0.4 mL/min and the column temperature was 40 °C.
- the ion source is used as the ESI source positive ion mode, and the scanning mode is multiple reaction monitoring (MRM).
- MRM multiple reaction monitoring
- the compound of the present invention has a better exposure in the blood of the body at an intravenous dose of 1.00 mg/kg as compared with the control compound 2.
- AUC INF a higher plasma concentrations
- C max a higher plasma concentrations
- the compound of the present invention has a better blood in the body at a dose of 2.00 mg/kg compared to the control compound 2. Exposure (AUC INF ) and higher blood concentration (C max ), thus having better absorption.
- the compound of the present invention has better exposure in the liver at an intragastric administration dose of 2.00 mg/kg as compared with the control compound 2.
- the amount (AUC INF ) and the higher blood concentration (C max ) further illustrate that the compounds of the invention (e.g., 10-226 of Example 25) have better absorbency.
- the compound of the present invention has a bioavailability (F) of 47.6% at an intragastric administration dose of 2.00 mg/kg, which is significantly higher than the bioavailability of the control compound 2.
- the compound of the present invention (for example, 10-230 of Example 26) is exposed to about 10 times in the liver in vivo, and the blood concentration in the liver is in plasma. About 10 times; while the amount of the control compound 2 exposed in the liver is about 1.5 times that in plasma, and the blood concentration in the liver is about 0.5 times that in plasma.
- This indicates that the compound of the present invention has excellent exposure (AUC INF ) and blood concentration (C max ) in the liver in vivo, and thus has liver targeting properties.
- Test compounds were administered to male Beagle dogs by intravenous (iv) and gavage (po), respectively.
- the doses administered by iv and po were 0.5 mg/kg and 2.5 mg/kg, respectively, and the solvent system was 5% when administered iv.
- Blood was collected at various time points after iv administration and po administration for PK studies. Plasma samples were subjected to protein precipitation and subjected to LC-MS/MS analysis.
- the mass spectrometer is API 5500, the liquid chromatograph is Waters ACQUITY I CLASS system; the column is Agela ASB C 18 column (2.1mm ⁇ 50mm, 1.9 ⁇ m); the mobile phase A phase is water +0.1% formic acid, and the B phase is acetonitrile.
- the flow rate was 0.4 mL/min and the column temperature was 40 °C.
- the ion source is used as the ESI source positive ion mode, and the scanning mode is multiple reaction monitoring (MRM).
- MRM multiple reaction monitoring
- the compound of the present invention has a better exposure in the blood of the body at an intravenous dose of 0.50 mg/kg as compared with the control compound 3. (AUC INF ) and higher plasma concentrations (C max ), thus having better pharmacokinetic parameters.
- the compound of the present invention (for example, 10-227 of Example 27) was exposed to plasma in an amount of about 14 times that of the control compound 3 at an intragastric administration dose of 2.50 mg/kg.
- the blood drug concentration was about 6 times that of the control compound 3.
- the compounds of the invention e.g., 10-227 of Example 27
- the compound of the present invention has a bioavailability (F) of 75.9% at an intragastric administration dose of 2.50 mg/kg, which is significantly superior to the bioavailability of the control compound 3.
- Degree (F) 41.7% is significantly superior to the bioavailability of the control compound 3.
- Test compounds were administered to male cynomolgus monkeys by intravenous (iv) and gavage (po), respectively.
- the doses administered by iv and po were 0.5 mg/kg and 2.5 mg/kg, respectively, and the solvent system was 5 when iv was administered.
- Blood was collected at various time points after iv administration and po administration for PK studies. Plasma samples were subjected to protein precipitation and subjected to LC-MS/MS analysis.
- the mass spectrometer is API 5500, the liquid chromatograph is Waters ACQUITY I CLASS system; the column is Agela ASB C 18 column (2.1mm ⁇ 50mm, 1.9 ⁇ m); the mobile phase A phase is water +0.1% formic acid, and the B phase is acetonitrile.
- the flow rate was 0.4 mL/min and the column temperature was 40 °C.
- the ion source is used as the ESI source positive ion mode, and the scanning mode is multiple reaction monitoring (MRM).
- MRM multiple reaction monitoring
- the compound of the present invention (for example, 10-227 of Example 27) was compared with the control compound 3 in the cynomolgus PK study at an intravenous dose of 0.50 mg/kg in vivo.
- AUC INF blood concentration
- C max blood concentration
- the compound of the present invention (for example, 10-227 of Example 27) in the cynomolgus PK study, the exposure amount in plasma at the dose of 2.50 mg/kg is About 21 times the control compound 3, the blood concentration was about 33 times that of the control compound 3. It is further illustrated that the compounds of the invention (e.g., 10-227 of Example 27) have better absorbency. Moreover, the compound of the present invention (for example, 10-227 of Example 27) has a bioavailability (F) of 37.2% at an intragastric administration dose of 2.50 mg/kg, which is significantly superior to the bioavailability of the control compound 3. Degree (F) 6.77%.
- HepG2C3A/pCYP3A4-Luc one C8 cell, was removed from the liquid nitrogen tank, and the cells were resuscitated in a 37 ° C sterile water bath and gently shaken until the ice cubes all melted.
- the resuscitated cells were transferred to a 15 mL sterile centrifuge tube and 5-10 mL of pre-warmed basal cell culture medium at 37 °C was added. The cells were allowed to settle naturally for 2 minutes and then centrifuged (1000 rpm) for 8 minutes. The supernatant was removed and the cells were resuspended in 10 mL of prewarmed cell culture medium.
- the cell suspension was transferred to a 10 cm cell culture dish and cultured in a 37 ° C 5% CO 2 incubator. After 24 hours, the original cell culture medium was replaced with the selected cell culture medium.
- the cells were grown to 80% to 90% culture dishes, the cells were digested and transferred to a 15 mL sterile centrifuge tube. The cells were collected by centrifugation (1000 rpm) for 8 minutes. The supernatant was removed and the cells were resuspended in 3 mL of prewarmed complete medium. The cell suspension was subcultured at a ratio of 1:3 or 1:5.
- the cell suspension was inoculated into 384-well white cell plates at 25 ⁇ L per well.
- the cell plates were centrifuged (300 rpm) for 1 minute and placed in a 37 ° C 5% CO 2 incubator for 24 hours.
- 300 nL of 100X compound was transferred from the compound plate into the cell plate.
- the cell plates were centrifuged (300 rpm) for one minute and placed in a 37 ° C 5% CO 2 incubator for 72 hours.
- the cell plate and Bright-Glo luciferase reagent were taken out and equilibrated to room temperature.
- Bright-Glo luciferase reagent was added to the cell plates (30 ⁇ L per well).
- the cell plates were centrifuged at 1000 rpm for 1 minute and incubated for 2 minutes at room temperature. The fluorescence signal is measured on a plate reader.
- the concentration curve of the test compound was prepared using the mapping software Prism 5, and the EC 50 value was calculated.
- the compound of the present invention (for example, 10-227 of Example 27) has a weaker induction effect on the CYP450 enzyme 3A4 subtype than Comparative Compound 3 and rifampicin, and thus has better safety. Sex.
Landscapes
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Virology (AREA)
- Medicinal Chemistry (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- General Health & Medical Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Oncology (AREA)
- Communicable Diseases (AREA)
- Epidemiology (AREA)
- Molecular Biology (AREA)
- Biotechnology (AREA)
- Engineering & Computer Science (AREA)
- Pulmonology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Plural Heterocyclic Compounds (AREA)
Abstract
Description
时间(min) | A% | B% |
0 | 90 | 10 |
15 | 40 | 60 |
30 | 10 | 90 |
缩写 | 含义 | 缩写 | 含义 |
DAST | 二乙胺基三氟化硫 | HOAc | 乙酸 |
DEA | 二乙胺 | IPA | 异丙醇 |
EA | 乙酸乙酯 | MeOH | 甲醇 |
EtOH | 乙醇 | PE | 石油醚 |
HEX | 己烷 | TLC | 薄层色谱法 |
化合物编号 | EC50(nM) |
实施例六的10-7 | 102.3 |
实施例九的10-34 | 13.2 |
实施例十的10-36 | 98.1 |
实施例十一的10-40 | 6.2 |
实施例十二的10-42 | 164.5 |
实施例一的10-88 | 24.7 |
实施例四的异构体B | 34.7 |
实施例四的异构体A | 35.1 |
实施例五的10-95 | 314.4 |
实施例十六的10-136 | 110.4 |
实施例十五的10-162 | 213.1 |
实施例十四的10-180 | 23.5 |
实施例十三的10-182 | 61.8 |
化合物编号 | EC50(nM) |
对照化合物2 | 89 |
实施例二十五的10-226 | 9.2 |
实施例二十七的10-227 | 10.0 |
实施例二十八的10-229 | 8.3 |
实施例二十六的10-230 | 11.2 |
实施例三十的10-237 | 10.9 |
实施例三十一的10-238 | 10.2 |
实施例三十二的10-239 | 28.9 |
实施例三十三的10-240 | 21.1 |
化合物编号 | CC50(μM) |
实施例六的10-7 | 129.5 |
实施例九的10-34 | 79.6 |
实施例十的10-36 | >150.0 |
实施例十一的10-40 | 73.5 |
实施例十二的10-42 | >150.0 |
实施例一的10-88 | 61.1 |
实施例四的异构体B | 51.3 |
实施例四的异构体A | 43.9 |
实施例五的10-95 | 131.3 |
实施例十六的10-136 | 122.3 |
实施例十五的10-162 | >150.0 |
实施例十四的10-180 | >150.0 |
实施例十三的10-182 | >150.0 |
实施例二十五的10-226 | >150.0 |
实施例二十六的10-230 | 137.0 |
实施例二十七的10-227 | >150.0 |
实施例二十八的10-229 | >150.0 |
实施例三十的10-237 | >150.0 |
实施例三十一的10-238 | >150.0 |
实施例三十二的10-239 | >150.0 |
实施例三十三的10-240 | >150.0 |
化合物编号 | IC50值(μM) |
对照化合物1 | 2.85 |
对照化合物2 | 1~10 |
实施例十的10-36 | >10 |
实施例十一的10-40 | >10 |
实施例十二的10-42 | >10 |
实施例一的10-88 | >10 |
实施例四的异构体B | >10 |
实施例的四异构体A | >10 |
实施例二的10-93 | >10 |
实施例五的10-95 | >10 |
实施例二十五的10-226 | >10 |
实施例二十七的10-227 | >10 |
实施例二十六的10-230 | >10 |
化合物编号 | 实施例二十七的10-227 | 对照化合物3 | 利福平 |
EC50(μM) | 122.8 | 5.641 | 1.828 |
Claims (20)
- 化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂合物、代谢物、同位素标记的化合物或前药,其中所述化合物具有式I或式Ia的结构:其中:Ar1和Ar2各自独立地选自C6-14芳基和5-14元杂芳基,其任选地被一个或多个选自卤素、-OH、-CN、-NO2、-N(R)2、C1-6烷基、卤代C1-6烷基、C1-6烷基硫基和C3-6环烷基的取代基取代;L不存在或者选自-O-、-S-和-NR-;R1和R2各自独立地选自H(包括1H、2H、3H)、C1-6烷基(例如:氘代C1-6烷基)和C3-6环烷基;R3为选自下列结构的四元、五元、六元或七元氮杂环系:Q选自-(CRaRa’)g-、-NRa-、-O-、-S-、-S(=O)-和-S(=O)2-;Ra、Ra’、R4、R4’、R5、R5’和R6在每次出现时各自独立地选自H、卤素、-OH、-COOH、-CN、-NO2、-N(R)2、C1-6烷基、卤代C1-6烷基、-W-C1-6烷基、-C1-6亚烷基-W-R、-W-C1-6亚烷基-W’-R、-W-C2-6烯基、-C2-6亚烯基-W-R、-W-C2-6亚烯基-W’-R和C3-6环烷基,其中所述亚烷基和亚烯基任选地进一步被一个或多个W间隔;或者Ra与Ra’一起、R4与R5一起和/或R4’与R5’一起,在每次出现时各自独立地形成基团=CH-W-R;条件是当R3不是四元氮杂环系时,Ra、Ra’、R4、R4’、R5、R5’和R6不同时为氢,并且不为选自-COOH、-C1-6亚烷基-OH和-C1-6亚烷基-C(=O)OH的基团;且当R3为四元氮杂环时,R4、R5和R6不同时为氢;R6连接在上述氮杂环系结构中*和/或**标记的环碳原子上;W和W’在每次出现时各自独立地选自O、C(=O)、C(=O)O、NR、NC(=O)、N(S=O)、NS(=O)2、S、S=O和S(=O)2;R在每次出现时各自独立地选自H、C1-6烷基和C3-6环烷基;g为1或2;并且t为0、1、2或3,条件是t不大于对应基团上可被取代的位置的数目,并且当t大于1时,每个R6可以相同或不同。
- 权利要求3的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂合物、代谢物、同位素标记的化合物或前药,其中Rc在每次出现时各自独立地选自F、Cl、Br、I、C1-6烷基和C3-6环烷基。
- 权利要求5的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂合物、代谢物、同位素标记的化合物或前药,其中Rb在每次出现时各自独立地选自H、卤素、C1-6烷基和C3-6环烷基。
- 权利要求1-6中任一项的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂合物、代谢物、同位素标记的化合物或前药,其中:Ra、Ra’、R4、R4’、R5、R5’和R6在每次出现时各自独立地选自H、F、Cl、Br、-(CR7R7a)mOH、-O-C1-6烷基、-(CR7R7a)mCOOH、-C(R7’)=C(R7a’)(CR7R7a)mCOOH和-(CR7R7a)m-W-(CR7’R7a’)nCOOH,所述-(CR7R7a)m-W-(CR7’R7a’)nCOOH优选为-(CR7R7a)mO(CR7’R7a’)nCOOH、-(CR7R7a)mNR(CR7’R7a’)nCOOH或-(CR7R7a)mS(=O)j(CR7’R7a’)nCOOH;或者Ra与Ra’一起、R4与R5一起和/或R4’与R5’一起,在每次出现时各自独立地形成基团=CH-W-R;优选地,Ra、Ra’、R4、R4’、R5、R5’和R6在每次出现时各自独立地选自H、F、-OH、-CH2OH、-OCH3、-COOH、-CH2COOH、-(CH2)2COOH、-(CH2)3COOH、-CH=CHCOOH、-OCH2COOH、-SCH2COOH、-N(CH3)CH2COOH、-CH2OCH2COOH、-CH2SCH2COOH、-CH2N(CH3)CH2COOH、-C(CH3)=CHCOOH和-CH=C(CH3)COOH;R7、R7’、R7a、R7a’在每次出现时各自独立地选自H、C1-4烷基和C3-6环烷基;R选自H、甲基、乙基、丙基和环丙基;m为0、1、2、3或4;n为1、2、3或4;并且j为0、1或2。
- 化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂合物、代谢物、同位素标记的化合物或前药,其中所述化合物具有以下结构:其中:R1和R2各自独立地选自H(包括1H、2H、3H)、C1-6烷基(例如:氘代C1-6烷基)和C3-6环烷基;Q为-(CRaRa’)g-或-O-;Ra、Ra’、R4、R5和R6在每次出现时各自独立地选自H、卤素、-OH、-COOH、-CN、-NO2、-N(R)2、C1-6烷基、卤代C1-6烷基、-W-C1-6烷基、-C1-6亚烷基-W-R、-W-C1-6亚烷基-W’-R、-W-C2-6烯基、-C2-6亚烯基-W-R、-W-C2-6亚烯基-W’-R和C3-6环烷基,其中所述亚烷基和亚烯基任选地进一步被一个或多个W间隔;Rb在每次出现时各自独立地选自H、卤素、卤代C1-6烷基、C1-6烷基和C3-6环烷基;Rc在每次出现时各自独立地选自F、Cl、Br、I、卤代C1-6烷基、C1-6烷基和C3-6环烷基;R6连接在通式结构中*和/或**标记的环碳原子上;W和W’在每次出现时各自独立地选自O、C(=O)、C(=O)O、NR、NC(=O)、N(S=O)、NS(=O)2、S、S=O和S(=O)2;R在每次出现时各自独立地选自H、C1-6烷基和C3-6环烷基;g为1或2;i为0、1或2;m为0、1、2、3或4;并且t为0、1或2,条件是当t大于1时,每个R6可以相同或不同。
- 权利要求11的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂合物、代谢物、同位素标记的化合物或前药,其中Rb在每次出现时各自独立地选自H、卤素、C1-6烷基和C3-6环烷基;Rc在每次出现时各自独立地选自F、Cl、Br、I、C1-6烷基和C3-6环烷基。
- 药物组合物,其包含预防或治疗有效量的权利要求1-14中任一项的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂合物、代谢物、同位素标记的化合物或前药以及一种或多种药学上可接受的载体,所述药物组合物优选是固体制剂、液体制剂或透皮制剂。
- 制备药物组合物的方法,所述方法包括将权利要求1-14中任一项的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂合物、代谢物、同位素标记的化合物或前药与一种或多种药学上可接受的载体组合。
- 权利要求1-14中任一项的化合物或其药学上可接受的盐、酯、立体异构体、互变异构体、多晶型物、溶剂合物、代谢物、同位素标记的化合物或前药或者权利要求15的药物组合物在制备用于预防或治疗病毒性疾病的药物中的用途。
- 权利要求17的用途,其中所述药物为通过口服、静脉内、动脉内、皮下、腹膜内、肌内或经皮途径给药的药物。
- 权利要求17的用途,其中所述病毒性疾病选自甲型病毒性肝炎、乙型病毒性肝炎、丙型病毒性肝炎、流行性感冒、疱疹和获得性免疫缺陷综合征(AIDS)。
- 制备权利要求1-14中任一项的化合物的方法,所述方法包括以下步骤:或者其中:Hal选自F、Cl、Br和I;卤化试剂选自Cl2、Br2、I2、N-氯代琥珀酰亚胺、N-溴代丁二酰亚胺和N-碘代丁二酰亚胺;其余各基团如权利要求1-14中任一项所定义;步骤一在碱金属盐存在下于质子性溶剂中进行;步骤二在非质子性溶剂中进行;并且步骤三在非质子性溶剂中,在有机碱或无机碱的存在下进行;当权利要求1-14中任一项的化合物中的R2为C1-6烷基时,其也可通过包括以下步骤的方法合成:或者其中:R2’为H或C1-5烷基;Hal选自F、Cl、Br和I;卤化试剂选自Cl2、Br2、I2、N-氯代琥珀酰亚胺、N-溴代丁二酰亚胺和N-碘代丁二酰亚胺;其余各基团如上述所定义;步骤I在路易斯酸存在下于非极性溶剂中进行;步骤II在有机碱或无机碱的存在下,在非质子性溶剂中进行;步骤III在非质子性溶剂中进行;并且步骤IV在非质子性溶剂中,在有机碱或无机碱的存在下进行。
Priority Applications (16)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP17870982.0A EP3508483B1 (en) | 2016-11-18 | 2017-11-09 | Dihydropyrimidine compound and preparation method and use thereof |
MX2019005119A MX2019005119A (es) | 2016-11-18 | 2017-11-09 | Compuesto de dihidropirimidina y metodo de preparacion y uso del mismo. |
US16/334,237 US10696669B2 (en) | 2016-11-18 | 2017-11-09 | Dihydropyrimidine compound and preparation method and use thereof |
ES17870982T ES2901401T3 (es) | 2016-11-18 | 2017-11-09 | Compuesto de dihidropirimidina y método de preparación y uso del mismo |
CA3037218A CA3037218A1 (en) | 2016-11-18 | 2017-11-09 | Dihydropyrimidine compound and preparation method and use thereof |
JP2019513814A JP7139568B2 (ja) | 2016-11-18 | 2017-11-09 | ジヒドロピリミジン化合物並びにその調製方法及び使用 |
KR1020197007824A KR102496508B1 (ko) | 2016-11-18 | 2017-11-09 | 디하이드로피리미딘 화합물 및 이러한 화합물의 제조 방법 및 용도 |
EA201990528A EA201990528A1 (ru) | 2017-05-11 | 2017-11-09 | Дигидропиримидиновые соединения и способ их получения и применения |
AU2017359773A AU2017359773B2 (en) | 2016-11-18 | 2017-11-09 | Dihydropyrimidine compound and preparation method and use thereof |
BR112019005205A BR112019005205A2 (pt) | 2016-11-18 | 2017-11-09 | composto de di-hidropirimidina e método de preparação e uso do mesmo |
MYPI2019001442A MY194471A (en) | 2016-11-18 | 2017-11-09 | Dihydropyrimidine compound and preparation method and use thereof |
CN201780057446.XA CN109790145B (zh) | 2016-11-18 | 2017-11-09 | 二氢嘧啶类化合物及其制备方法和用途 |
PL17870982T PL3508483T3 (pl) | 2016-11-18 | 2017-11-09 | Związek dihydropirymidynowy oraz sposób jego wytwarzania i jego zastosowanie |
PH12019550039A PH12019550039A1 (en) | 2016-11-18 | 2019-03-18 | Dihydropyrimidine compound and preparation method and use thereof |
IL266345A IL266345A (en) | 2016-11-18 | 2019-04-30 | Dihydropyrimidine compound and method of preparation and use thereof |
US16/859,910 US11166954B2 (en) | 2016-11-18 | 2020-04-27 | Dihydropyrimidine compound and preparation method and use thereof |
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
CN201611015150 | 2016-11-18 | ||
CN201611015150.5 | 2016-11-18 | ||
CN201710328659.3 | 2017-05-11 | ||
CN201710328659 | 2017-05-11 |
Related Child Applications (2)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
US16/334,237 A-371-Of-International US10696669B2 (en) | 2016-11-18 | 2017-11-09 | Dihydropyrimidine compound and preparation method and use thereof |
US16/859,910 Continuation-In-Part US11166954B2 (en) | 2016-11-18 | 2020-04-27 | Dihydropyrimidine compound and preparation method and use thereof |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2018090862A1 true WO2018090862A1 (zh) | 2018-05-24 |
Family
ID=62145967
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/CN2017/110123 WO2018090862A1 (zh) | 2016-11-18 | 2017-11-09 | 二氢嘧啶类化合物及其制备方法和用途 |
Country Status (16)
Country | Link |
---|---|
US (1) | US10696669B2 (zh) |
EP (1) | EP3508483B1 (zh) |
JP (1) | JP7139568B2 (zh) |
KR (1) | KR102496508B1 (zh) |
CN (1) | CN109790145B (zh) |
AU (1) | AU2017359773B2 (zh) |
BR (1) | BR112019005205A2 (zh) |
CA (1) | CA3037218A1 (zh) |
ES (1) | ES2901401T3 (zh) |
IL (1) | IL266345A (zh) |
MX (1) | MX2019005119A (zh) |
MY (1) | MY194471A (zh) |
PH (1) | PH12019550039A1 (zh) |
PL (1) | PL3508483T3 (zh) |
SG (1) | SG10202010180YA (zh) |
WO (1) | WO2018090862A1 (zh) |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN108329308A (zh) * | 2018-05-16 | 2018-07-27 | 四川科伦博泰生物医药股份有限公司 | 一种二氢嘧啶类化合物的固体形式及其制备方法 |
WO2020255015A1 (en) | 2019-06-18 | 2020-12-24 | Janssen Sciences Ireland Unlimited Company | Combination of hepatitis b virus (hbv) vaccines and dihydropyrimidine derivatives as capsid assembly modulators |
CN114853761A (zh) * | 2021-02-05 | 2022-08-05 | 刘沛 | 一种含有二氢嘧啶的双功能衍生物及其用途 |
Families Citing this family (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN112575389B (zh) * | 2019-09-30 | 2023-09-19 | 成都先导药物开发股份有限公司 | 一种dna编码化合物库的液相色谱纯化方法 |
EP4289842A1 (en) | 2021-02-05 | 2023-12-13 | Hepagene Therapeutics (HK) Limited | Phenyldihydropyrimidine compound and use thereof |
CN113512035B (zh) * | 2021-04-26 | 2023-11-24 | 山东大学 | 二氢嘧啶-泊马度胺缀合物及其制备方法与应用 |
Citations (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2014003748A1 (en) | 2012-06-28 | 2014-01-03 | Nuance Communications, Inc. | Meta-data inputs to front end processing for automatic speech recognition |
CN104144924A (zh) * | 2012-03-31 | 2014-11-12 | 弗·哈夫曼-拉罗切有限公司 | 用于治疗和预防乙型肝炎病毒感染的新的4-甲基-二氢嘧啶类 |
CN104650068A (zh) * | 2013-11-19 | 2015-05-27 | 广东东阳光药业有限公司 | 二氢嘧啶类化合物及其在药物中的应用 |
WO2015078391A1 (en) | 2013-11-27 | 2015-06-04 | Sunshine Lake Pharma Co., Ltd. | Processes for preparing dihydropyrimidine derivatives and intermediates thereof |
WO2015132276A1 (en) * | 2014-03-07 | 2015-09-11 | F. Hoffmann-La Roche Ag | Novel 6-fused heteroaryldihydropyrimidines for the treatment and prophylaxis of hepatitis b virus infection |
WO2015144093A1 (en) | 2014-03-28 | 2015-10-01 | Sunshine Lake Pharma Co., Ltd. | Dihydropyrimidine compounds and their application in pharmaceuticals |
CN105209470A (zh) * | 2013-05-17 | 2015-12-30 | 豪夫迈·罗氏有限公司 | 用于治疗和预防乙型肝炎病毒感染的新的6-桥连的杂芳基二氢嘧啶 |
Family Cites Families (14)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
DE10013126A1 (de) * | 2000-03-17 | 2001-09-20 | Bayer Ag | Arzneimittel gegen virale Erkrankungen |
CN101225084A (zh) | 2007-01-16 | 2008-07-23 | 北京摩力克科技有限公司 | 二氢嘧啶类化合物及其用于制备治疗和预防病毒性疾病的药物的用途 |
WO2010069147A1 (zh) * | 2008-12-17 | 2010-06-24 | 张中能 | 二氢嘧啶类化合物、其组合物及其应用 |
CN101575318B (zh) * | 2009-06-25 | 2012-02-08 | 中国人民解放军军事医学科学院毒物药物研究所 | 二氢嘧啶类化合物及其用于制备治疗和/或预防病毒性疾病的药物的用途 |
AU2013305390C1 (en) * | 2012-08-24 | 2015-12-24 | Ruyuan Wei Xiang Technology Co., Ltd. | Dihydropyrimidine compounds and their application in pharmaceuticals |
CN103664925B (zh) * | 2012-09-07 | 2018-01-23 | 广东东阳光药业有限公司 | 杂芳基取代的二氢嘧啶类化合物及其在药物中的应用 |
MA37942B1 (fr) | 2012-09-10 | 2020-01-31 | Hoffmann La Roche | Hétéroaryldihydropyrimidines d'acide 6-aminé pour le traitement et la prophylaxie d'une infection par le virus de l'hépatite b |
CN103664899B (zh) * | 2012-09-11 | 2017-06-16 | 广东东阳光药业有限公司 | 杂芳基取代的二氢嘧啶类化合物及其在药物中的应用 |
WO2014165128A2 (en) | 2013-03-12 | 2014-10-09 | Novira Therapeutics, Inc. | Hepatitis b antiviral agents |
CN105188703A (zh) * | 2013-03-20 | 2015-12-23 | 美国印第安纳大学研究和技术公司 | 荧光-hap:用于细胞中的hbv核的诊断性染色剂 |
ES2640063T3 (es) | 2013-04-03 | 2017-10-31 | Janssen Sciences Ireland Uc | Derivados de n-fenil-carboxamida y su uso como medicamentos para el tratamiento de la hepatitis B |
CN104650069B (zh) * | 2013-11-19 | 2019-04-19 | 广东东阳光药业有限公司 | 4-甲基二氢嘧啶类化合物及其在药物中的应用 |
CN104650070B (zh) * | 2013-11-25 | 2018-09-14 | 广东东阳光药业有限公司 | 二氢嘧啶类化合物及其在药物中的应用 |
CN108329308B (zh) * | 2018-05-16 | 2022-11-01 | 四川科伦博泰生物医药股份有限公司 | 一种二氢嘧啶类化合物的固体形式及其制备方法 |
-
2017
- 2017-11-09 BR BR112019005205A patent/BR112019005205A2/pt not_active Application Discontinuation
- 2017-11-09 ES ES17870982T patent/ES2901401T3/es active Active
- 2017-11-09 MX MX2019005119A patent/MX2019005119A/es unknown
- 2017-11-09 US US16/334,237 patent/US10696669B2/en active Active
- 2017-11-09 WO PCT/CN2017/110123 patent/WO2018090862A1/zh active Application Filing
- 2017-11-09 JP JP2019513814A patent/JP7139568B2/ja active Active
- 2017-11-09 EP EP17870982.0A patent/EP3508483B1/en active Active
- 2017-11-09 PL PL17870982T patent/PL3508483T3/pl unknown
- 2017-11-09 MY MYPI2019001442A patent/MY194471A/en unknown
- 2017-11-09 CA CA3037218A patent/CA3037218A1/en active Pending
- 2017-11-09 SG SG10202010180YA patent/SG10202010180YA/en unknown
- 2017-11-09 CN CN201780057446.XA patent/CN109790145B/zh active Active
- 2017-11-09 AU AU2017359773A patent/AU2017359773B2/en active Active
- 2017-11-09 KR KR1020197007824A patent/KR102496508B1/ko active IP Right Grant
-
2019
- 2019-03-18 PH PH12019550039A patent/PH12019550039A1/en unknown
- 2019-04-30 IL IL266345A patent/IL266345A/en unknown
Patent Citations (8)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN104144924A (zh) * | 2012-03-31 | 2014-11-12 | 弗·哈夫曼-拉罗切有限公司 | 用于治疗和预防乙型肝炎病毒感染的新的4-甲基-二氢嘧啶类 |
WO2014003748A1 (en) | 2012-06-28 | 2014-01-03 | Nuance Communications, Inc. | Meta-data inputs to front end processing for automatic speech recognition |
CN105209470A (zh) * | 2013-05-17 | 2015-12-30 | 豪夫迈·罗氏有限公司 | 用于治疗和预防乙型肝炎病毒感染的新的6-桥连的杂芳基二氢嘧啶 |
CN104650068A (zh) * | 2013-11-19 | 2015-05-27 | 广东东阳光药业有限公司 | 二氢嘧啶类化合物及其在药物中的应用 |
US20160206616A1 (en) | 2013-11-19 | 2016-07-21 | Sunshine Lake Pharma Co., Ltd. | Dihydropyrimidine compounds and their application in pharmaceuticals |
WO2015078391A1 (en) | 2013-11-27 | 2015-06-04 | Sunshine Lake Pharma Co., Ltd. | Processes for preparing dihydropyrimidine derivatives and intermediates thereof |
WO2015132276A1 (en) * | 2014-03-07 | 2015-09-11 | F. Hoffmann-La Roche Ag | Novel 6-fused heteroaryldihydropyrimidines for the treatment and prophylaxis of hepatitis b virus infection |
WO2015144093A1 (en) | 2014-03-28 | 2015-10-01 | Sunshine Lake Pharma Co., Ltd. | Dihydropyrimidine compounds and their application in pharmaceuticals |
Non-Patent Citations (12)
Title |
---|
"Bioreversible Carriers in Drug Design", 1987, PERGAMON PRESS |
"Protective Groups in Organic Chemistry", 1973, PLENUM PRESS |
"Remington's Pharmaceutical Sciences", 1990 |
ANTIMICROB. AGENTS CHEMOTHER., vol. 57, 2013, pages 5344 - 5354 |
BIOCHEM. PHARMACOL., vol. 66, 2003, pages 2273 - 2279 |
H. BUNDGAARD: "Design of Prodrugs", 1985, ELSEVIER |
SCIENCE, vol. 299, 2003, pages 893 - 896 |
See also references of EP3508483A4 |
STAHL; WERMUTH: "Hand book of Pharmaceutical Salts: Properties, Selection, and Use", 2002, WILEY-VCH |
T. HIGUCHI; V. STELLA: "Pro-drugs as Novel Delivery Systems", ACS SYMPOSIUM SERIES, vol. 14 |
T.W. GREENE; P.G.M. WUTS: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY & SONS |
ZLOTNICK ET AL., PROC. NATL. ACAD. SCI., vol. 102, 2005, pages 8138 - 8143 |
Cited By (8)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
CN108329308A (zh) * | 2018-05-16 | 2018-07-27 | 四川科伦博泰生物医药股份有限公司 | 一种二氢嘧啶类化合物的固体形式及其制备方法 |
WO2019218883A1 (zh) * | 2018-05-16 | 2019-11-21 | 四川科伦博泰生物医药股份有限公司 | 一种二氢嘧啶类化合物的固体形式及其制备方法和用途 |
CN111868057A (zh) * | 2018-05-16 | 2020-10-30 | 四川科伦博泰生物医药股份有限公司 | 一种二氢嘧啶类化合物的固体形式及其制备方法和用途 |
US11434235B2 (en) | 2018-05-16 | 2022-09-06 | Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd. | Solid form of dihydropyrimidine compound and preparation method therefor and use thereof |
CN108329308B (zh) * | 2018-05-16 | 2022-11-01 | 四川科伦博泰生物医药股份有限公司 | 一种二氢嘧啶类化合物的固体形式及其制备方法 |
CN111868057B (zh) * | 2018-05-16 | 2024-03-05 | 四川科伦博泰生物医药股份有限公司 | 一种二氢嘧啶类化合物的固体形式及其制备方法和用途 |
WO2020255015A1 (en) | 2019-06-18 | 2020-12-24 | Janssen Sciences Ireland Unlimited Company | Combination of hepatitis b virus (hbv) vaccines and dihydropyrimidine derivatives as capsid assembly modulators |
CN114853761A (zh) * | 2021-02-05 | 2022-08-05 | 刘沛 | 一种含有二氢嘧啶的双功能衍生物及其用途 |
Also Published As
Publication number | Publication date |
---|---|
SG10202010180YA (en) | 2020-11-27 |
US10696669B2 (en) | 2020-06-30 |
MY194471A (en) | 2022-11-30 |
CA3037218A1 (en) | 2018-05-24 |
ES2901401T3 (es) | 2022-03-22 |
CN109790145A (zh) | 2019-05-21 |
PH12019550039A1 (en) | 2019-11-25 |
CN109790145B (zh) | 2020-09-22 |
MX2019005119A (es) | 2019-06-20 |
BR112019005205A2 (pt) | 2019-06-11 |
AU2017359773A1 (en) | 2019-04-11 |
IL266345A (en) | 2019-06-30 |
PL3508483T3 (pl) | 2022-02-07 |
JP2019535644A (ja) | 2019-12-12 |
JP7139568B2 (ja) | 2022-09-21 |
US20190225603A1 (en) | 2019-07-25 |
KR102496508B1 (ko) | 2023-02-03 |
AU2017359773B2 (en) | 2021-05-20 |
EP3508483A4 (en) | 2020-04-15 |
EP3508483A1 (en) | 2019-07-10 |
EP3508483B1 (en) | 2021-10-06 |
KR20190077312A (ko) | 2019-07-03 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
WO2018090862A1 (zh) | 二氢嘧啶类化合物及其制备方法和用途 | |
EP2892893B1 (en) | 6-amino acid heteroaryldihydropyrimidines for the treatment and prophylaxis of hepatitis b virus infection | |
TWI668217B (zh) | 二氫嘧啶類化合物及其在藥物中的應用 | |
WO2015124063A1 (zh) | 丙肝病毒抑制剂及其制药用途 | |
JP2020537677A (ja) | ジヒドロピリミジン化合物、及び医薬におけるその使用 | |
EP3441389B1 (en) | Pyrazole-oxazolidinone compound for anti-hepatitis b virus | |
CA2935811A1 (en) | 6-fused heteroaryldihydropyrimidines for the treatment and prophylaxis of hepatitis b virus infection | |
WO2022100623A1 (zh) | 氮取代杂环噻吩类化合物及其用途 | |
CN112543755A (zh) | 一类细胞坏死抑制剂及其制备方法和用途 | |
CN112771027A (zh) | 溶血磷脂酸受体拮抗剂及其制备方法 | |
WO2013151707A1 (en) | Kynurenine-3-monooxygenase inhibitors, pharmaceutical compositions, and methods of use thereof | |
AU2017326356A1 (en) | Hepatitis B core protein modulators | |
US20180263991A1 (en) | Complexes and salts of dihydropyrimidine derivatives and their application in pharmaceuticals | |
TW202416959A (zh) | 經取代四氫環戊[c]吡咯、經取代二氫吡咯𠯤,其類似物及使用其之方法 | |
WO2022199599A1 (zh) | 丙烯酰基取代的化合物、包含其的药物组合物及其用途 | |
WO2018082503A1 (zh) | 杂环化合物及其制备方法和用途 | |
US11166954B2 (en) | Dihydropyrimidine compound and preparation method and use thereof | |
CN115698001A (zh) | 具有作为usp30抑制剂活性的n-氰基吡咯烷类化合物 | |
CN114901649A (zh) | Ssao抑制剂及其用途 | |
EA039977B1 (ru) | Дигидропиримидиновые соединения и способ их получения и применения | |
WO2022052923A1 (zh) | 二氢嘧啶类化合物、其应用 | |
OA19422A (en) | Dihydropyrimidine compound and preparation method and use thereof. | |
WO2024088408A1 (zh) | 一种含氮杂环化合物、其药学上可接受的盐及其制备方法与应用 | |
TW201542521A (zh) | 丙肝病毒抑制劑及其製藥用途 |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 17870982 Country of ref document: EP Kind code of ref document: A1 |
|
ENP | Entry into the national phase |
Ref document number: 2019513814 Country of ref document: JP Kind code of ref document: A |
|
ENP | Entry into the national phase |
Ref document number: 3037218 Country of ref document: CA Ref document number: 20197007824 Country of ref document: KR Kind code of ref document: A |
|
REG | Reference to national code |
Ref country code: BR Ref legal event code: B01A Ref document number: 112019005205 Country of ref document: BR |
|
ENP | Entry into the national phase |
Ref document number: 2017870982 Country of ref document: EP Effective date: 20190401 |
|
ENP | Entry into the national phase |
Ref document number: 2017359773 Country of ref document: AU Date of ref document: 20171109 Kind code of ref document: A |
|
WWE | Wipo information: entry into national phase |
Ref document number: 266345 Country of ref document: IL |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
ENP | Entry into the national phase |
Ref document number: 112019005205 Country of ref document: BR Kind code of ref document: A2 Effective date: 20190318 |