CN108602842B - Heterocyclic compounds, process for their preparation and their use - Google Patents

Heterocyclic compounds, process for their preparation and their use Download PDF

Info

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

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/683Diesters of a phosphorus acid with two hydroxy compounds, e.g. phosphatidylinositols
    • A61K31/685Diesters of a phosphorus acid with two hydroxy compounds, e.g. phosphatidylinositols one of the hydroxy compounds having nitrogen atoms, e.g. phosphatidylserine, lecithin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic System
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

The present invention relates to heterocyclic compounds having antiviral activity, pharmaceutical compositions comprising the same, processes for the preparation thereof and the use thereof in the prevention or treatment of viral diseases including, but not limited to, viral hepatitis a, viral hepatitis b, viral hepatitis c, influenza, herpes and acquired immunodeficiency syndrome (AIDS).

Description

Heterocyclic compounds, process for their preparation and their use
Technical Field
The present invention relates to heterocyclic compounds having antiviral activity, pharmaceutical compositions comprising the same, processes for the preparation thereof and the use thereof in the prevention or treatment of viral diseases including, but not limited to, viral hepatitis a, viral hepatitis b, viral hepatitis c, influenza, herpes and acquired immunodeficiency syndrome (AIDS).
Background
Viruses are composed of a nucleic acid molecule (DNA or RNA) and a protein or only a protein (e.g., prions). Viruses can cause a variety of infectious diseases, common viral-caused diseases including, but not limited to, viral hepatitis a, viral hepatitis b, viral hepatitis c, influenza, herpes, and Acquired Immune Deficiency Syndrome (AIDS).
Currently clinically used antiviral drugs act by inhibiting attachment, uncoating, viral gene replication, maturation or release of viruses, or by affecting the immune system of the host, which mainly include reverse transcriptase inhibitors and capsid protein assembly inhibitors, etc.
Hepatitis B Virus (HBV) is a common hepadnatropic viral pathogen. The virus can cause diseases such as acute hepatitis, chronic hepatitis, hepatic fibrosis, liver cirrhosis, liver cancer and the like.
Viral hepatitis b can be treated with the following nucleoside analogs:
Figure BDA0001712943790000011
wherein, Tenofovir (Tenofovir) is a nucleoside analogue reverse transcriptase inhibitor which has obvious curative effect on Human Immunodeficiency Virus (HIV) and Hepatitis B Virus (HBV) infection (Antivir Ther, 2004, 9, 57-65; JViralHeapat, 2000, 7, 161 and 165). The principle of action is to inhibit reverse transcriptase activity by terminating DNA strand synthesis after competitive integration into HBV DNA with the natural substrate deoxyadenosine-5' -triphosphate. However, tenofovir is hardly absorbed by the gastrointestinal tract. And the esterified prodrug Tenofovir Disoproxil Fumarate (TDF) and the amidated prodrug Tenofovir Alafenamide Fumarate (TAF) have good water solubility (Nucleotides acids.2001, 20, 1085-.
In addition, viral hepatitis B can be treated with non-nucleoside analogs. Research by Deres et al found that heteroaryl dihydropyrimidines (Bay41-4109) could prevent HBV viral replication by inhibiting viral capsid protein assembly (Science, 2003, 299, 893-896). The specific mechanism of action is that dihydropyrimidine compounds induce core protein misassembly, thereby forming unstable capsid protein and accelerating the degradation of core protein (biochem. Pharmacol., 2003, 66, 2273-2279). The heteroaryl dihydropyrimidines HAP1(Proc. Natl. Acad. Sci., 2005, 102, 8138-8143) found by ZLotnick et al and the heteroaryl dihydropyrimidines (GLS4) reported by Guangdong-Tokyo photo pharmaceuticals, Inc. (Antimicrob. Agents Chemother., 2013, 57, 5344-5354; WO2015078391, US2016206616) also have anti-HBV activity.
Although both classes of compounds exhibit antiviral activity to varying degrees, their activity has not been satisfactory. Therefore, there is an urgent need and great significance to find more effective drugs with less toxic side effects.
Summary of The Invention
The present invention provides an antiviral heterocyclic compound which can exert an antiviral action by an inhibitory action on reverse transcriptase and/or capsid protein assembly. In particular, preferred compounds of the present invention can simultaneously exhibit an inhibitory effect on reverse transcriptase and capsid protein assembly, thereby achieving more excellent antiviral effects at a smaller dose. In addition, the compounds of the present invention have more excellent properties such as better physicochemical properties (e.g., solubility, physical and/or chemical stability), improved pharmacokinetic properties (e.g., improved bioavailability, suitable half-life and duration of action), improved safety (less toxicity and/or fewer side effects), less susceptibility to drug resistance, and the like.
One aspect of the present invention provides a compound, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, or prodrug thereof, wherein the compound has the structure of formula (I):
Figure BDA0001712943790000021
wherein:
b is selected from:
Figure BDA0001712943790000022
a is selected from optionally substituted by one or more RbSubstituted C1-6Alkylene radical, C2-6Alkenylene radical, C2-6Alkynylene, said alkylene, alkenylene or alkynylene being optionally interrupted by one or more-O-, -NR-or-S-;
or A is selected from the following groups:
Figure BDA0001712943790000023
wherein
Figure BDA0001712943790000024
Represents a single or double bond, and is linked to B at the 1-position and to the phosphorus atom (P) at the 2-position;
x, Y and Z are each independently selected from CH at each occurrence2O, S and NR;
Raand RbEach occurrence is independently selected from halogen, -OH, -CN, -NO2、-N(R)2、-N3、C1-6Alkyl and C3-6A cycloalkyl group;
R2selected from H, C1-12Alkyl radical, C3-6Cycloalkyl radical, C6-14Aryl, 5-14 membered heteroaryl, C6-20Aralkyl, -C1-6alkylene-COOH, -C1-6alkylene-C (═ O) O-C1-6Alkyl, -C1-6alkylene-OC (═ O) -C1-6Alkyl, -C1-6alkylene-OC (═ O) O-C1-6Alkyl and- ((CH)2)iO)m-(CH2)q-O-C1-6Alkyl, each of the above groups being optionally substituted by one or more groups selected from halogen, -OH, -CN, -NO2、-N(R)2、C1-6Alkyl, halo C1-6Alkyl radical, C1-6Alkylthio and C3-6Cycloalkyl substituents;
i and q are each independently at each occurrence 1, 2, 3, 4, 5, or 6;
m is any integer from 0 to 50, preferably from 0 to 20, particularly preferably from 0 to 6;
R1selected from:
Figure BDA0001712943790000025
Figure BDA0001712943790000026
represents a 3 to 14 membered azacyclic ring system optionally additionally containing a substituent independently selected from the group consisting of N, O, C ═ O, S, S ═ O and S (═ O)21, 2 or 3 ring members of (a);
Ar1and Ar2Each independently selected from C6-14Aryl and 5-14 membered heteroaryl, optionally substituted with one or more substituents selected from halogen, -OH, -CN, -NO2、-N(R)2、C1-6Alkyl, halo C1-6Alkyl radical, C1-6Alkylthio and C3-6Cycloalkyl substituents;
l is absent or selected from-O-, -S-and-NR-;
R3and R4Each independently selected from H, C1-4Alkyl and C3-6A cycloalkyl group;
R5at each occurrence, is attached to the rest of the molecule with a single or double bond;
R5and R6Independently at each occurrence, is selected from halogen, -OH, -COOH, -CN, -NO2、-N(R)2、C1-6Alkyl, halo C1-6Alkyl, -W-C1-6Alkyl, -C1-6alkylene-W-R, -W-C1-6alkylene-W' -R, -W-C2-6Alkenyl, -C2-6alkenylene-W-R, -W-C2-6alkenylene-W' -R and C3-6Cycloalkyl, wherein the alkylene and alkenylene are optionally further interrupted by one or more W;
w and W' are each independently at each occurrence selected from O, C (═ O), C (═ O) O, NR, S ═ O, and S (═ O)2
R at each occurrence is independently selected from H, C1-6Alkyl and C3-6A cycloalkyl group;
n is independently at each occurrence 0, 1, 2, 3, 4 or 5, provided that n is no greater than the number of positions on the corresponding group that may be substituted; and is
When n is greater than 1, each RaWhich may be the same or different, each RbCan be the same as orIn different, each R5Which may be the same or different, each R6May be the same or different.
Another aspect of the invention provides a pharmaceutical composition comprising a prophylactically or therapeutically effective amount of a compound of the invention, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, or prodrug thereof, and one or more pharmaceutically acceptable carriers.
Another aspect of the present invention provides a method of preparing a pharmaceutical composition, comprising combining a compound of the present invention, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, or prodrug thereof, with one or more pharmaceutically acceptable carriers.
Another aspect of the present invention provides a use of a compound of the present invention, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, or prodrug thereof, or a pharmaceutical composition of the present invention, for the preparation of a medicament for the prevention or treatment of a viral disease.
Another aspect of the present invention provides a compound of the present invention or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite or prodrug thereof, or a pharmaceutical composition of the present invention, for use in the prevention or treatment of a viral disease.
Another aspect of the present invention provides a method for preventing or treating a viral disease, the method comprising administering to a subject in need thereof an effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, or prodrug thereof, or a pharmaceutical composition of the present invention.
Such viral diseases include, but are not limited to, viral hepatitis A, viral hepatitis B, viral hepatitis C, influenza, herpes and Acquired Immune Deficiency Syndrome (AIDS).
Another aspect of the present invention provides a process for preparing a compound of the present invention, said process comprising the steps of:
Figure BDA0001712943790000031
or
Figure BDA0001712943790000032
Wherein:
R12、R13and R14Each independently selected from F, Cl, Br, I, -NHR, hydroxy, triflate, p-toluenesulfonate and boronate;
PG is selected from t-butoxycarbonyl, benzyloxycarbonyl, 9-fluorenylmethoxycarbonyl, allyloxycarbonyl, trimethylsiloxyethoxycarbonyl, methoxycarbonyl, ethoxycarbonyl, p-toluenesulfonyl, o-nitrobenzenesulfonyl, p-nitrobenzenesulfonyl, formyl, acetyl, trifluoroacetyl, propionyl, pivaloyl, phenyl, benzoyl, trityl, benzyl, 2, 4-dimethoxybenzyl and p-methoxybenzyl;
the remaining groups are as defined above;
the first step is carried out in the presence of an organic or inorganic base and/or a condensation reagent (particularly preferably DCC, DIC, EDC, BOP, PyAOP or PyBOP) in an aprotic solvent or without a solvent;
the second step is carried out under conditions suitable for removing the PG groups;
the third step is carried out in an aprotic solvent in the presence of an organic base or an inorganic base; and is
The fourth step is carried out in an aprotic solvent, preferably in the presence of an organic or inorganic base and/or a condensation reagent (particularly preferably DCC, DIC, EDC, BOP, PyAOP or PyBOP).
Another aspect of the present invention provides a process for preparing a compound of the present invention, said process comprising the steps of:
Figure BDA0001712943790000041
wherein:
R12selected from the group consisting of F, Cl, Br, I, -NHR, hydroxy, triflate, p-toluenesulfonate and borate;
LG is a leaving group, which is preferably pentafluorophenyl and p-nitrophenyl;
the remaining groups are as defined above;
the first step is carried out in an aprotic solvent in the presence of an organic or inorganic base;
the second step is carried out in an aprotic solvent in the presence of an organic base or an inorganic base; and is
The third step is carried out in an aprotic solvent in the presence of an organic or inorganic base.
Definition of
Unless defined otherwise below, all technical and scientific terms used herein are intended to have the same meaning as commonly understood by one of ordinary skill in the art. Reference to the techniques used herein is intended to refer to those techniques commonly understood in the art, including those variations of or alternatives to those techniques that would be apparent to those skilled in the art. While the following terms are believed to be well understood by those skilled in the art, the following definitions are set forth to better explain the present invention.
The terms "comprising," "including," "having," "containing," or "involving," and other variations thereof herein, are inclusive or open-ended and do not exclude additional unrecited elements or method steps.
As used herein, the term "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.
As used herein, the term "alkenylene" denotes a divalent hydrocarbon radical comprising one or more double bonds, preferably having 2, 3, 4, 5 or 6 carbon atoms, such as ethenylene, propenylene or allylene. When the compounds of the invention contain alkenylene groups, the compounds may be present in pure E (entgegen) form, in pure Z (ipsilateral (zusammen)) form or in any mixture thereof.
As used herein, the term "alkynylene" denotes a divalent hydrocarbon radical comprising one or more triple bonds, preferably having 2, 3, 4, 5 or 6 carbon atoms, e.g. ethynylene or propynyl.
As used herein, the term "alkyl" is defined as a straight or branched chain saturated aliphatic hydrocarbon group. In some embodiments, the alkyl group has 1 to 12 carbon atoms, for example 1 to 6 carbon atoms. For example, as used herein, the term "C1-6Alkyl "refers to a straight or branched chain saturated aliphatic hydrocarbon group having 1 to 6 carbon atoms (e.g., methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, or n-hexyl), which is optionally substituted with 1 or more (such as 1 to 3) suitable substituents such as halogen (in which case the group is referred to as" haloalkyl ") (e.g., CF3、C2F5、CHF2、CH2F、CH2CF3、CH2Cl or-CH2CH2CF3Etc.). The term "C1-4Alkyl "refers to a straight or branched chain saturated aliphatic hydrocarbon group having 1 to 4 carbon atoms (i.e., methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, or tert-butyl).
As used herein, the term "cycloalkyl" refers to a saturated or unsaturated non-aromatic monocyclic or polycyclic (such as bicyclic) hydrocarbon ring (e.g., monocyclic, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, cyclononyl, or bicyclic ring, 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]Nonyl, decalinyl, etc.)), optionally substituted with 1 or more (such as 1 to 3) suitable substituents. The cycloalkyl group has 3 to 15 carbon atoms. For example, the term "C3-6Cycloalkyl "refers to a saturated or unsaturated, non-aromatic, monocyclic or polycyclic (such as bicyclic) hydrocarbon ring (e.g., cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl) having 3 to 6 ring carbon atoms, optionally substituted with 1 or more (such as 1 to 3) suitable substituents, e.g., methylAnd (3) cyclopropyl.
As used herein, the term "aryl" refers to an all-carbon monocyclic or fused ring polycyclic aromatic group having a conjugated pi-electron system. For example, as used herein, the term "C6-14Aryl "means an aromatic group containing 6 to 14 carbon atoms, such as phenyl or naphthyl. Aryl is optionally substituted with 1 or more (such as 1 to 3) suitable substituents (e.g. halogen, -OH, -CN, -NO)2、C1-6Alkyl, etc.).
As used herein, the term "aralkyl" denotes an alkyl group substituted with an aryl group, wherein the aryl group and the alkyl group are as defined herein. Typically, the aryl group can have 6 to 14 carbon atoms and the alkyl group can have 1 to 6 carbon atoms. Exemplary aralkyl groups include, but are not limited to, benzyl, phenylethyl, phenylpropyl, phenylbutyl.
The term "heteroaryl" as used herein refers to a monocyclic, bicyclic or tricyclic aromatic ring system having 5, 6, 8, 9, 10, 11, 12, 13 or 14 ring atoms, in particular 1 or 2 or 3 or 4 or 5 or 6 or 9 or 10 carbon atoms, and which contains at least one heteroatom which may be the same or different (said heteroatom being, for example, oxygen, nitrogen or sulfur), and which, in addition, in each case may be benzo-fused. In particular, heteroaryl is selected from thienyl, furyl, pyrrolyl, oxazolyl, thiazolyl, imidazolyl, pyrazolyl, isoxazolyl, isothiazolyl, oxadiazolyl, triazolyl, thiadiazolyl and the like, and benzo derivatives thereof; or pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, and the like, and benzo derivatives thereof.
As used herein, the term "halo" or "halogen" group is defined to include F, Cl, Br, or I.
The term "alkylthio," as used herein, refers to an alkyl group, as defined above, appended to the parent molecular moiety through a sulfur atom. C1-6Representative examples of alkylthio include, but are not limited to, methylthio, ethylthio, tert-butylthio, and hexylthio.
As used herein, the term "azacyclic ring system" refers to a saturated or unsaturated monocyclic or bicyclic ring systemA cyclic group having 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12 or 13 carbon atoms in the ring and at least one nitrogen atom, which may also optionally contain one or more (e.g. one, two, three or four) selected from N, O, C ═ O, S, S ═ O and S (═ O)2A ring member of (a); the aza ring system is attached to the rest of the molecule through a nitrogen atom. In particular, the 3-to 14-membered nitrogen heterocycle is a cyclic group having 3-14 carbon atoms and heteroatoms (at least one of which is a nitrogen atom) in the ring, which includes, but is not limited to, aziridinyl, azetidinyl, pyrrolyl, pyrrolidinyl, pyrrolinyl, pyrrolidinonyl, imidazolyl, imidazolidinyl, imidazolinyl, pyrazolyl, pyrazolinyl, piperidinyl, morpholinyl, thiomorpholinyl, piperazinyl, indolyl, indolinyl, and the like.
The term "substituted" means that one or more (e.g., one, two, three, or four) hydrogens on the designated atom is replaced with a selection from the indicated group, provided that the designated atom's normal valency under the current circumstances is not exceeded and that the substitution results in a stable compound. Combinations of substituents and/or variables are permissible only if such combinations result in stable compounds.
If a substituent is described as "optionally substituted," the substituent may be (1) unsubstituted or (2) substituted. If a carbon of a substituent is described as being optionally substituted with one or more of the list of substituents, one or more hydrogens on the carbon (to the extent of any hydrogens present) may be replaced individually and/or together with an independently selected optional substituent. If the nitrogen of a substituent is described as being optionally substituted with one or more of the list of substituents, then one or more hydrogens on the nitrogen (to the extent any hydrogen is present) may each be replaced with an independently selected optional substituent.
If a substituent is described as being "independently selected from" a group, each substituent is selected independently of the other. Thus, each substituent may be the same as or different from another (other) substituent.
As used herein, the term "one or more" means 1 or more than 1, such as 2, 3, 4, 5 or 10, under reasonable conditions.
Unless indicated, as used herein, the point of attachment of a substituent may be from any suitable position of the substituent.
When a bond of a substituent is shown through a bond connecting two atoms in a ring, then such substituent may be bonded to any ring atom in the substitutable ring.
The invention also includes all pharmaceutically acceptable isotopically-labeled compounds, which are identical to those of the present invention, except that one or more atoms are replaced by an atom having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number prevailing in nature. Examples of isotopes suitable for inclusion in compounds of the invention include, but are not limited to, isotopes of hydrogen (e.g. hydrogen)2H、3H) (ii) a Isotopes of carbon (e.g. of11C、13C and14C) (ii) a Isotopes of chlorine (e.g. of chlorine)36Cl); isotopes of fluorine (e.g. of fluorine)18F) (ii) a Isotopes of iodine (e.g. of iodine)123I and125I) (ii) a Isotopes of nitrogen (e.g. of13N and15n); isotopes of oxygen (e.g. of15O、17O and18o); isotopes of phosphorus (e.g. of phosphorus)32P); and isotopes of sulfur (e.g. of35S). Certain isotopically-labeled compounds of the present invention (e.g., those into which a radioisotope is incorporated) are useful in drug and/or substrate tissue distribution studies (e.g., assays). Radioisotope tritium (i.e. tritium3H) And carbon-14 (i.e.14C) Are particularly useful for this purpose because of their ease of incorporation and ease of detection. Using positron-emitting isotopes (e.g. of the type11C、18F、15O and13n) can be used to examine substrate receptor occupancy in Positron Emission Tomography (PET) studies. Isotopically labeled compounds of the present invention can be prepared by processes analogous to those described in the accompanying schemes and/or in the examples and preparations by using an appropriate isotopically labeled reagent in place of the non-labeled reagent employed previously. Pharmaceutically acceptable solvates of the invention include those in which the crystallization solvent may be isotopically substituted, e.g., D2O, acetone-d6Or DMSO-d6
The term "stereoisomer" denotes an isomer formed as a result of at least one asymmetric center. In compounds having one or more (e.g., one, two, three, or four) asymmetric centers, they can give rise to racemic mixtures, single enantiomers, diastereomeric mixtures, and individual diastereomers. Certain individual molecules may also exist as geometric isomers (cis/trans). Similarly, the compounds of the invention may exist as mixtures of two or more structurally different forms (commonly referred to as tautomers) in rapid equilibrium. Representative examples of tautomers include keto-enol tautomers, phenol-keto tautomers, nitroso-oxime tautomers, imine-enamine tautomers, and the like. For example, a dihydropyrimidine radical may exist in solution in equilibrium with the following tautomeric forms:
Figure BDA0001712943790000061
it is understood that the scope of this application encompasses all such isomers or mixtures thereof in any ratio (e.g., 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99%).
Solid (-), solid wedge shapes may be used herein
Figure BDA0001712943790000062
Or virtual wedge shape
Figure BDA0001712943790000063
Carbon-carbon bonds of the compounds of the invention are depicted. The use of a solid line to depict a bond to an asymmetric carbon atom is intended to indicate that all possible stereoisomers (e.g., particular enantiomers, racemic mixtures, etc.) at that carbon atom are included. The use of solid or dashed wedges to depict bonds to asymmetric carbon atoms is intended to indicate that the stereoisomers shown are present. When present in a racemic mixture, solid and dotted wedges are used to define the relative stereochemistry, not the absolute stereochemistry. Unless otherwise indicated, the compounds of the present invention are intended to be stereoisomerizableIn the form of isomers, including cis and trans isomers, optical isomers (e.g., R and S enantiomers), diastereomers, geometric isomers, rotamers, conformers, atropisomers, and mixtures thereof. The compounds of the present invention may exhibit more than one type of isomerization and consist of mixtures thereof (e.g., racemic mixtures and diastereomeric pairs).
The present invention encompasses all possible crystalline forms or polymorphs of the compounds of the present invention, which may be single polymorphs or mixtures of more than one polymorph in any ratio.
It will also be appreciated that certain compounds of the invention may be present in free form for use in therapy or, where appropriate, in the form of a pharmaceutically acceptable derivative thereof. In the present invention, pharmaceutically acceptable derivatives include, but are not limited to, pharmaceutically acceptable salts, esters, solvates, metabolites or prodrugs, which upon administration to a patient in need thereof are capable of providing, directly or indirectly, a compound of the present invention or a metabolite or residue thereof. Thus, when reference is made herein to "a compound of the invention," it is also intended to encompass the various derivative forms of the compounds described above.
Pharmaceutically acceptable salts of the compounds of the present invention include acid addition salts and base addition salts thereof.
Suitable acid addition salts are formed from acids which form pharmaceutically acceptable salts. Examples include aspartate, bicarbonate/carbonate, fumarate, glucoheptonate, gluconate, glucuronate, hexafluorophosphate, hydrobromide/bromide, hydroiodide, naphthoate, nicotinate, nitrate, orotate, palmitate and other similar salts.
Suitable base addition salts are formed from bases which form pharmaceutically acceptable salts. Examples include aluminum salts, arginine salts, choline salts, lysine salts, magnesium salts, meglumine salts, tromethamine salts, and other similar salts.
For a review of suitable Salts see Stahl and Wermuth, "Handbook of Pharmaceutical Salts: properties, Selection, and Use "(Wiley-VCH, 2002). Methods for preparing pharmaceutically acceptable salts of the compounds of the present invention are known to those skilled in the art.
As used herein, the term "ester" means an ester derived from a compound of the respective general formula in the present application, including physiologically hydrolysable esters (which can be hydrolysed under physiological conditions to release the compound of the invention in free acid or alcohol form). The compounds of the invention may themselves also be esters.
The compounds of the invention may be present in the form of solvates, preferably hydrates, wherein the compounds of the invention comprise as structural element of the crystal lattice of the compound a polar solvent, such as in particular water, methanol or ethanol. The amount of polar solvent, particularly water, may be present in stoichiometric or non-stoichiometric proportions.
Also included within the scope of the present invention are metabolites of the compounds of the present invention, i.e., substances formed in vivo upon administration of the compounds of the present invention. Such products may result, for example, from oxidation, reduction, hydrolysis, amidation, deamidation, esterification, defatting, enzymatic hydrolysis, etc. of the administered compound. Accordingly, the present invention includes metabolites of the compounds of the present invention, including compounds made by the process of contacting the compounds of the present invention with a mammal for a time sufficient to produce a metabolite thereof.
The present invention further includes within its scope prodrugs of the compounds of the present invention which are certain derivatives of the compounds of the present invention which may themselves have little or no pharmacological activity which, when administered into or onto the body, may be converted to the compounds of the present invention having the desired activity by, for example, hydrolytic cleavage. Typically such prodrugs will be functional derivatives of the compounds which are readily convertible in vivo into the desired therapeutically active compound. Further 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," PergamonPress, 1987(E.B.Roche editions, American Pharmaceutical Association). Prodrugs of the invention may be prepared, for example, by substituting certain moieties known to those skilled in the art as "pro-moieties" (e.g., "Design of produgs", described in h. bundgaard (Elsevier, 1985)) for appropriate functional groups present in compounds of the invention.
The invention also encompasses compounds of the invention containing a protecting group. In any process for preparing the compounds of the present invention, it may be necessary and/or desirable to protect sensitive or reactive groups on any of the molecules concerned, thereby forming a chemically protected form of the compounds of the present invention. This can be achieved by conventional protecting groups, for example, as described in protective groups in Organic Chemistry, ed.j.f.w.mcomie, Plenum Press, 1973; and T.W.Greene & P.G.M.Wuts, Protective Groups in Organic Synthesis, John Wiley & Sons, 1991, which are incorporated herein by reference. The protecting group may be removed at a suitable subsequent stage using methods known in the art.
The term "about" means within. + -. 10%, preferably within. + -. 5%, more preferably within. + -. 2% of the stated value.
Detailed Description
Compound and preparation method thereof
In one embodiment, the present invention provides a compound, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, or prodrug thereof, wherein the compound has the structure of formula (I):
Figure BDA0001712943790000071
wherein:
b is selected from:
Figure BDA0001712943790000072
a is selected from optionally substituted by one or more RbSubstituted C1-6Alkylene radical, C2-6Alkenylene radical, C2-6Alkynylene, said alkylene, alkenylene or alkynylene being optionally interrupted by one or more-O-, -NR-or-S-)Breaking;
or A is selected from the following groups:
Figure BDA0001712943790000073
wherein
Figure BDA0001712943790000074
Represents a single or double bond, and is linked to B at the 1-position and to the phosphorus atom (P) at the 2-position;
x, Y and Z are each independently selected from CH at each occurrence2O, S and NR;
Raand RbEach occurrence is independently selected from halogen, -OH, -CN, -NO2、-N(R)2、-N3、C1-6Alkyl and C3-6A cycloalkyl group;
R2selected from H, C1-12Alkyl radical, C3-6Cycloalkyl radical, C6-14Aryl, 5-14 membered heteroaryl, C6-20Aralkyl, -C1-6alkylene-COOH, -C1-6alkylene-C (═ O) O-C1-6Alkyl, -C1-6alkylene-OC (═ O) -C1-6Alkyl, -C1-6alkylene-OC (═ O) O-C1-6Alkyl and- ((CH)2)iO)m-(CH2)q-O-C1-6Alkyl, each of the above groups being optionally substituted by one or more groups selected from halogen, -OH, -CN, -NO2、-N(R)2、C1-6Alkyl, halo C1-6Alkyl radical, C1-6Alkylthio and C3-6Cycloalkyl substituents;
i and q are each independently at each occurrence 1, 2, 3, 4, 5, or 6;
m is any integer from 0 to 50, preferably from 0 to 20, particularly preferably from 0 to 6;
R1selected from:
Figure BDA0001712943790000075
Figure BDA0001712943790000076
represents a 3 to 14 membered azacyclic ring system optionally additionally containing a substituent independently selected from the group consisting of N, O, C ═ O, S, S ═ O and S (═ O)21, 2 or 3 ring members of (a);
Ar1and Ar2Each independently selected from C6-14Aryl and 5-14 membered heteroaryl, optionally substituted with one or more substituents selected from halogen, -OH, -CN, -NO2、-N(R)2、C1-6Alkyl, halo C1-6Alkyl radical, C1-6Alkylthio and C3-6Cycloalkyl substituents;
l is absent or selected from-O-, -S-and-NR-;
R3and R4Each independently selected from H, C1-4Alkyl and C3-6A cycloalkyl group;
R5at each occurrence, is attached to the rest of the molecule with a single or double bond;
R5and R6Independently at each occurrence, is selected from halogen, -OH, -COOH, -CN, -NO2、-N(R)2、C1-6Alkyl, halo C1-6Alkyl, -W-C1-6Alkyl, -C1-6alkylene-W-R, -W-C1-6alkylene-W' -R, -W-C2-6Alkenyl, -C2-6alkenylene-W-R, -W-C2-6alkenylene-W' -R and C3-6Cycloalkyl, wherein the alkylene and alkenylene are optionally further interrupted by one or more W;
w and W' are each independently at each occurrence selected from O, C (═ O), C (═ O) O, NR, S ═ O, and S (═ O)2
R at each occurrence is independently selected from H, C1-6Alkyl and C3-6A cycloalkyl group;
n is independently at each occurrence 0, 1, 2, 3, 4 or 5, provided that n is not greater than the number of positions on the corresponding group which may be substituted, and
when n is greater than 1, each RaWhich may be the same or different, each RbWhich may be the same or different, each R5Which may be the same or different, each R6May be the same or different.
In a preferred embodiment, X, Y and Z are each independently selected for each occurrence from CH2O and NH.
In a particularly preferred embodiment, X, Y and Z are each independently selected at each occurrence from O and NH.
In a preferred embodiment, RaAnd RbIndependently at each occurrence, is selected from halogen, -OH, -N (R)2、-N3And C1-6An alkyl group.
In a particularly preferred embodiment, RaAnd RbEach occurrence is independently selected from the group consisting of F, -OH, amino, cyclopropylamino and methyl.
In a preferred embodiment, B is selected from:
Figure BDA0001712943790000081
in a particularly preferred embodiment, B is
Figure BDA0001712943790000082
In a preferred embodiment, a is selected from:
Figure BDA0001712943790000083
wherein the 1 position is linked to B and the 2 position is linked to the phosphorus atom (P).
In a more preferred embodiment, A is
Figure BDA0001712943790000084
Wherein the 1 position is linked to B and the 2 position is linked to the phosphorus atom (P).
In a particularly preferred embodiment, A is
Figure BDA0001712943790000085
WhereinThe 1 position is linked to B and the 2 position is linked to the phosphorus atom (P).
In a preferred embodiment of the process according to the invention,
Figure BDA0001712943790000091
represents a 3-, 4-, 5-or 6-membered azaring system.
In a particularly preferred embodiment of the process according to the invention,
Figure BDA0001712943790000092
selected from azetidinyl, pyrrolidinyl, piperidinyl, morpholinyl, and piperazinyl.
In a preferred embodiment, R1Selected from:
Figure BDA0001712943790000093
in a preferred embodiment, Ar1And Ar2Each independently selected from imidazolyl, oxazolyl, thiazolyl, pyridyl, pyrimidinyl, piperazinyl and phenyl, each optionally substituted with one or more of the same or different halogen, C1-6Alkyl, halo C1-6Alkyl and C3-6Cycloalkyl is substituted.
In a more preferred embodiment, Ar1And Ar2Each independently selected from imidazolyl, oxazolyl, thiazolyl, pyridyl, pyrimidinyl, piperazinyl and phenyl, each optionally substituted with one or more of the same or different halogen, C1-6Alkyl and C3-6Cycloalkyl is substituted.
In a preferred embodiment, Ar1Selected from:
Figure BDA0001712943790000094
wherein R iscEach occurrence is independently selected from F, Cl, Br, I, C1-6Alkyl, halo C1-6Alkyl and C3-6A cycloalkyl group;
preferably RcIndependently at each occurrenceSelected from F, Cl, Br, I, C1-6Alkyl and C3-6A cycloalkyl group;
Ar1preferably selected from:
Figure BDA0001712943790000095
Ar1more preferably from:
Figure BDA0001712943790000096
in a preferred embodiment, Ar2Selected from:
Figure BDA0001712943790000097
the above groups are optionally substituted by one or more groups selected from halogen, C1-6Alkyl, halo C1-6Alkyl and C3-6Cycloalkyl groups.
In a more preferred embodiment, Ar2Selected from:
Figure BDA0001712943790000098
the above groups are optionally substituted by one or more groups selected from halogen, C1-6Alkyl and C3-6Cycloalkyl groups.
In a preferred embodiment, L is-O-.
In a preferred embodiment, R3And R4Each independently selected from methyl, ethyl, n-propyl and isopropyl.
In a particularly preferred embodiment, R4Is H.
In a preferred embodiment, R5And R6Each occurrence is independently selected from F, Cl, Br, I, -OH, -COOH, methyl, ethyl, -CH2OH、-OCH3、-CH2COOH、-CH2CH2COOH、-CH2CH2CH2COOH、-CH=CHCOOH、-OCH2COOH、-SCH2COOH、-N(CH3)CH2COOH、-CH2OCH2COOH、-CH2SCH2COOH and-CH2N(CH3)CH2COOH。
In a preferred embodiment, R2Selected from H, C1-12Alkyl radical, C3-6Cycloalkyl radical, C6-14Aryl, 5-14 membered heteroaryl, C6-20Aralkyl, -C1-6alkylene-C (═ O) O-C1-6Alkyl, -C1-6alkylene-OC (═ O) -C1-6Alkyl, -C1-6alkylene-OC (═ O) O-C1-6Alkyl and- ((CH)2)iO)m-(CH2)q-O-C1-6Alkyl, each of the above groups being optionally substituted by one or more groups selected from halogen, -OH, -CN, -NO2、-N(R)2、C1-6Alkyl, halo C1-6Alkyl radical, C1-6Alkylthio and C3-6Cycloalkyl substituents;
in a more preferred embodiment, R2Selected from: c1-6Alkyl (- ((CH))2)iO)m-(CH2)q-O-C1-6Alkyl, aryl, heteroaryl, and heteroaryl,
Figure BDA0001712943790000101
Wherein:
i and q are each independently at each occurrence 1, 2, 3, 4, 5, or 6;
m is any integer from 0 to 50, preferably from 0 to 20, particularly preferably from 0 to 6;
R7、R8and R9Each independently selected from H, C1-10Alkyl radical, C3-6Cycloalkyl radical, C6-20Aryl and C7-20Aralkyl, said alkyl, cycloalkyl, aryl and aralkyl each optionally substituted with one or more substituents selected from the group consisting of halogen, -OH, -CN and-NO2Substituted with the substituent(s);
or R7And R8Together with the carbon atom to which they are attachedForm C3-6A cycloalkyl group;
R10and R11Each independently selected from H, halogen, -OH, -CN, -NO2、C1-10Alkyl and C3-6A cycloalkyl group.
In a more preferred embodiment, R2Selected from: c1-6Alkyl (- ((CH))2)iO)m-(CH2)q-O-C1-4Alkyl, aryl, heteroaryl, and heteroaryl,
Figure BDA0001712943790000102
And
Figure BDA0001712943790000103
wherein:
i and q are each independently at each occurrence 1, 2, 3 or 4;
m is 1, 2, 3, 4, 5 or 6;
R7and R9Each independently selected from H or C1-4Alkyl, said alkyl being optionally substituted by one or more groups selected from halogen, -OH, -CN and-NO2Substituted with the substituent(s);
R10and R11Each independently selected from H, halogen, -OH, -CN, -NO2Or C1-4An alkyl group.
In a particularly preferred embodiment, R2Selected from H, methyl, ethyl, propyl, butyl, pentyl, - ((CH)2)2O)6-(CH2)2-O-CH3
Figure BDA0001712943790000104
In a particularly preferred embodiment, R2Selected from methyl, ethyl, propyl, butyl, - ((CH)2)2O)6-(CH2)2-O-CH3
Figure BDA0001712943790000105
Figure BDA0001712943790000106
In preferred embodiments, n is 0, 1, 2 or 3.
In a preferred embodiment, the present invention provides a compound, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, or prodrug thereof, wherein the compound has the structure of formula (II) or formula (II) -1:
Figure BDA0001712943790000107
in a preferred embodiment, the present invention provides a compound, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, or prodrug thereof, wherein the compound has the structure of any of the following formulae:
Figure BDA0001712943790000108
Figure BDA0001712943790000111
the present invention encompasses compounds resulting from any combination of the various embodiments.
In a preferred embodiment, the present invention provides a compound, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite or prodrug thereof, wherein said compound is selected from the group consisting of:
Figure BDA0001712943790000112
Figure BDA0001712943790000121
one embodiment of the present invention provides a process for preparing a compound of the present invention, said process comprising the steps of:
Figure BDA0001712943790000122
or
Figure BDA0001712943790000123
Wherein:
R12、R13and R14Each independently selected from F, Cl, Br, I, -NHR, hydroxy, triflate, p-toluenesulfonate and boronate;
PG is selected from t-butoxycarbonyl, benzyloxycarbonyl, 9-fluorenylmethoxycarbonyl, allyloxycarbonyl, trimethylsiloxyethoxycarbonyl, methoxycarbonyl, ethoxycarbonyl, p-toluenesulfonyl, o-nitrobenzenesulfonyl, p-nitrobenzenesulfonyl, formyl, acetyl, trifluoroacetyl, propionyl, pivaloyl, phenyl, benzoyl, trityl, benzyl, 2, 4-dimethoxybenzyl and p-methoxybenzyl;
the remaining groups are as defined above;
the first step is carried out in the presence of an organic or inorganic base and/or a condensation agent, in an aprotic solvent or without a solvent;
the second step is carried out under conditions suitable for removing the PG groups;
the third step is carried out in an aprotic solvent in the presence of an organic base or an inorganic base; and is
The fourth step is carried out in an aprotic solvent, preferably in the presence of an organic or inorganic base and/or a condensation agent.
Another aspect of the present invention provides a process for preparing a compound of the present invention, said process comprising the steps of:
Figure BDA0001712943790000131
wherein:
R12selected from F, Cl, Br, I, -NHR, hydroxyA triflate group, a p-toluenesulfonate group and a borate group;
LG is a leaving group, which is preferably pentafluorophenyl and p-nitrophenyl;
the remaining groups are as defined above;
the first step is carried out in an aprotic solvent in the presence of an organic or inorganic base;
the second step is carried out in an aprotic solvent in the presence of an organic base or an inorganic base; and is
The third step is carried out in an aprotic solvent in the presence of an organic or inorganic base.
In a preferred embodiment, R12、R13And R14Each independently selected from F, Cl, Br, I, NH2Or a hydroxyl group.
In a preferred embodiment, PG may be t-butyloxycarbonyl and suitable conditions for removal of the t-butyloxycarbonyl group may be, for example, in a solvent such as dichloromethane at from 0 ℃ to room temperature under catalysis of an acid such as trifluoroacetic acid.
In a preferred embodiment, LG-OH may be pentafluorophenol, p-nitrophenol.
In a preferred embodiment, aprotic solvents that may be used in the methods of making the compounds of the present invention include, but are not limited to, tetrahydrofuran, dichloromethane, 1, 2-dichloroethane, acetonitrile, N-Dimethylformamide (DMF), N-methylpyrrolidone, 1, 3-dimethyl-2-imidazolidinone (DMI), dimethyl sulfoxide (DMSO), and hexamethylphosphoric triamide (HMPA).
In a preferred embodiment, organic bases that can be used in the methods of making the compounds of the present invention include, but are not limited to, sodium tert-butoxide, tert-butylmagnesium chloride, triethylamine, N-Diisopropylethylamine (DIPEA), pyridine, or 4-Dimethylaminopyridine (DMAP); inorganic bases that can be used in the process for preparing the compounds of the present invention include, but are not limited to, NaH, NaOH, Na2CO3Or K2CO3
In a preferred embodiment, the condensation reagents that may be used in the process for the preparation of the compounds of the present invention include, but are not limited to Dicyclohexylcarbodiimide (DCC), N-Diisopropylcarbodiimide (DIC), 1- (3-dimethylaminopropyl) -3-Ethylcarbodiimide (EDC), benzotriazol-1-yloxytris (dimethylamino) phosphonium hexafluorophosphate (BOP), (3H-1, 2, 3-triazolo [4, 5-b ] pyridin-3-yloxy) tris-1-pyrrolidinophosphonium hexafluorophosphate (PyAOP) and 1H-benzotriazol-1-yloxytripyrrolidinylphosphonium hexafluorophosphate (PyBOP).
Pharmaceutical compositions and methods of treatment
In another embodiment, the present invention provides 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, metabolite, or prodrug thereof, and one or more pharmaceutically acceptable carriers. In another embodiment, the pharmaceutical composition may further comprise one or more additional therapeutic agents, for example, additional therapeutic agents for preventing or treating viral diseases.
In another embodiment, the present invention provides a method of making a pharmaceutical composition comprising combining a compound of the present invention, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, or prodrug thereof, with one or more pharmaceutically acceptable carriers.
In another embodiment, the present invention provides the use of a compound of the present invention, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite, or prodrug thereof, or a pharmaceutical composition of the present invention, in the manufacture of a medicament for the prevention or treatment of a viral disease.
In another embodiment, the present invention provides a compound of the present invention or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite or prodrug thereof, or a pharmaceutical composition of the present invention, for use in the prevention or treatment of a viral disease.
In another embodiment, the present invention provides a method for preventing or treating a viral disease, comprising administering to a subject in need thereof an effective amount of a compound of the present invention, or a pharmaceutically acceptable salt, ester, stereoisomer, tautomer, polymorph, solvate, metabolite or prodrug thereof, or a pharmaceutical composition of the present invention.
Preferred compounds of the invention exert an antiviral effect through inhibition of reverse transcriptase and/or capsid protein assembly. Thus, the compounds of the present invention may be used to treat any virus involved in reverse transcriptase and/or capsid protein assembly in affecting the 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).
Thus, viral diseases that can be prevented and treated using the compounds of the present invention include, but are not limited to, viral hepatitis a, viral hepatitis b, viral hepatitis c, influenza, herpes and Acquired Immune Deficiency Syndrome (AIDS), and related symptoms or diseases caused by the above diseases (e.g., inflammation, liver fibrosis, cirrhosis, liver cancer, etc.).
By "pharmaceutically acceptable carrier" in the context of the present invention is meant a diluent, adjuvant, excipient, or vehicle that is administered together with a therapeutic agent and which is, within the scope of sound medical judgment, suitable for contact with the tissues of humans and/or other animals without excessive toxicity, irritation, allergic response, or other problem or complication commensurate with a reasonable benefit/risk ratio.
Pharmaceutically acceptable carriers that may be employed 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, mineral oil, sesame oil and the like. Water is an exemplary carrier when the pharmaceutical composition is administered intravenously. Physiological saline and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, maltose, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene glycol, water, ethanol and the like. The composition may also optionally contain minor amounts of wetting agents, emulsifying agents, or pH buffering agents. Oral formulations may contain standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate and the like. Examples of suitable pharmaceutically acceptable carriers are described in Remington's pharmaceutical sciences (1990).
The pharmaceutical compositions of the present invention may act systemically and/or locally. For this purpose, they may be administered by a suitable route, for example by injection (e.g. intravenous, intra-arterial, subcutaneous, intraperitoneal, intramuscular injection, including instillation) or transdermally; or by oral, buccal, nasal, transmucosal, topical, in the form of ophthalmic preparations or by inhalation.
For these routes of administration, the pharmaceutical compositions of the present invention may be administered in suitable dosage forms.
Such dosage forms include, but are not limited to, tablets, capsules, lozenges, hard candies, powders, sprays, creams, ointments, suppositories, gels, pastes, lotions, ointments, aqueous suspensions, injectable solutions, elixirs, syrups.
The term "effective amount" as used herein refers to an amount of a compound that, when administered, will alleviate one or more symptoms of the condition being treated to some extent.
The dosing regimen may be adjusted to provide the best desired response. For example, a single bolus may be administered, several divided doses may be administered over time, or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation. It is noted that dosage values may vary with the type and severity of the condition being alleviated, and may include single or multiple doses. It is further understood that for any particular individual, the specific dosage regimen will be adjusted over time according to the individual need and the professional judgment of the person administering the composition or supervising the administration of the composition.
The amount of a compound of the invention administered will depend on the subject being treated, the severity of the disorder or condition, the rate of administration, the disposition of the compound, and the judgment of the prescribing physician. Generally, an effective dose is from about 0.0001 to about 50mg per kg body weight per day, e.g., from about 0.01 to about 10 mg/kg/day (single or divided administration). For a 70kg human, this may amount to about 0.007 mg/day to about 3500 mg/day, e.g., about 0.7 mg/day to about 700 mg/day. In some cases, dosage levels not higher than the lower limit of the aforesaid range may be sufficient, while in other cases still larger doses may be employed without causing any harmful side effects, provided that the larger dose is first divided into several smaller doses to be administered throughout the day.
The compound of the invention may be present in the pharmaceutical composition in an amount or amount of from about 0.01mg to about 1000mg, suitably 0.1-500mg, preferably 0.5-300mg, more preferably 1-150mg, especially 1-50mg, for example 1.5mg, 2mg, 4mg, 10mg, 25mg etc.
As used herein, unless otherwise specified, the term "treating" means reversing, alleviating, inhibiting the progression of, or preventing such a disorder or condition, or one or more symptoms of such a disorder or condition, to which such term applies.
As used herein, "individual" includes a human or non-human animal. Exemplary human individuals include human individuals (referred to as patients) having a disease (e.g., a disease described herein) or normal individuals. "non-human animals" in the context of the present invention include all vertebrates, such as non-mammals (e.g., birds, amphibians, reptiles) and mammals, such as non-human primates, livestock and/or domesticated animals (e.g., sheep, dogs, cats, cows, pigs, etc.).
In another embodiment, the pharmaceutical compositions of the present invention may further comprise one or more additional therapeutic or prophylactic agents, including, but not limited to, lamivudine, telbivudine, entecavir, adefovir dipivoxil, tenofovir disoproxil fumarate, and tenofovir alafenamide fumarate.
Examples
The invention is further described below in connection with examples, which are not provided to limit the scope of the invention.
Commercial anhydrous and HPLC grade solvents were used without further purification unless otherwise noted.
Recording at ambient temperature with a Bruker instrument (400MHz)1H NMR spectrum using TMS as internal standard. Chemical shifts () are given in ppm and coupling constants (J) are given in hertz (Hz).1The split weight of H NMR peaks is abbreviated as follows: s (singlet), d (doublet), t (triplet), q (quartet), m (multiplet), br (broad).
LC-MS adopts an Aglient 1200 liquid chromatograph combined with an Aglient 6120Quadrupole type mass spectrometer to detect at 214nm and 254 nm. Preparative liquid chromatography using a SHIMADZU CBM-20A and active 1260 model preparative liquid chromatography, C18OBD 19 × 150mm5 μ M preparative column, detection wavelength 214nm, mobile phase a water, mobile phase B acetonitrile (0.5 ‰ formic acid added), linear gradient elution was performed according to the following table:
time (min) A% B%
0 90 10
15 40 60
30 10 90
EXAMPLE one Ethyl (4R) -6- ((4- (4- (((((((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) ((2-methylbenzyl) oxy) phosphonyl) oxy) phenyl) piperidin-1-yl) methyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (Compound 1)
Figure BDA0001712943790000151
The method comprises the following steps: synthesis of tert-butyl 4- (4-hydroxyphenyl) piperidine-1-carboxylate (Compound 1-2)
At room temperature, 4- (4- (benzyloxy) phenyl) -3, 6-dihydropyridine-1 (2H) -carboxylic acid tert-butyl ester (compound 1-1) (3.0g, 8.2mmol) was added to tetrahydrofuran (10mL) and methanol (20mL) and after the compound was completely dissolved, 10% Pd/C (300mg) was added under nitrogen protection, and after the addition was completed, hydrogen substitution was performed, reaction was carried out at room temperature for 18H, followed by filtration and after-treatment of the filtrate, the title compound (2.0g) was obtained. ESI-MS (m/z): 222.1[ M-55+ H]+
Step two: synthesis of tert-butyl 4- (4- (((((((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) (hydroxy) phosphonyl) oxy) phenyl) piperidine-1-carboxylate (Compound 1-3)
(R) -9- (2-methoxypropyl-phosphate) -adenine (3.1g, 10.8mmol) and compound 1-2(2.0g, 7.2mmol) were dissolved in N-methylpyrrolidone (20mL) at room temperature, and triethylamine (879mg, 8.9mmol) was added dropwise, after which the temperature was raised to 100 ℃ and a solution of dicyclohexylcarbodiimide (4.8g, 23.1mmol) in N-methylpyrrolidone (10mL) was added dropwise, and after addition, the reaction was carried out overnight at 100 ℃ to obtain the title compound (1.0 g). ESI-MS (m/z): 547.2[ M + H]+
Step three: synthesis of ((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) -4- (piperidin-4-yl) -phenyl-monophosphate (Compound 1-4)
Compound 1-3(200mg, 0.4mmol) was added to dichloromethane (5.0mL) at room temperature, after complete dissolution of the compound trifluoroacetic acid (0.2mL) was added, and after completion of addition, the reaction was allowed to proceed at room temperature for 2.5h, followed by workup to give the trifluoroacetate salt of the title compound (200 mg). ESI-MS (m/z): 447.2[ M + H]+
Step four: synthesis of ethyl (4R) -6- ((4- (4- (((((((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) (hydroxy) phosphonyl) oxy) phenyl) piperidin-1-yl) methyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (Compound 1-5)
The trifluoroacetate salt of compound 1-4 (163mg, 0.4mmol) was dissolved in 1, 2-dichloroethane (4mL) at room temperature, and ethyl (R) -6- (bromomethyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (204mg, 0.4mmol) and N, N-diisopropylethylamine (0.1mL) were added in this order, and after completion, the reaction was allowed to react overnight at room temperature, followed by workup to give the title compound (130 mg). ESI-MS (m/z): 824.2[ M + H]+
Step five: synthesis of ethyl (4R) -6- ((4- (4- (((((((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) ((2-methylbenzyl) oxy) phosphonyl) oxy) phenyl) piperidin-1-yl) methyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (Compound 1)
Compound 1-5(30mg, 0.04mmol), o-methylbenzyl alcohol (9.0mg, 0.07mmol), and 1H-benzotriazole-1-yloxytripyrrolidinophosphonium hexafluorophosphate (PyBOP) (38mg, 0.07mmol) were dissolved in N, N-dimethylformamide (3mL) at room temperature, after complete dissolution, N-diisopropylethylamine (19mg, 0.14mmol) was added dropwise, and after completion of addition, reaction was carried out at room temperature for 4 hours to obtain the title compound (9mg) after purification.
The structure is characterized as follows:
1H NMR(400MHz,DMSO-d6)9.78(s,1H),8.13(s,1H),8.05-8.03(m,2H),7.95(d,J=3.13Hz,1H),7.44-7.40(m,2H),7.30-7.14(m,9H),7.03(d,J=8.20Hz,1H),6.96(d,J=8.12Hz,1H),6.06(s,1H),5.15-5.08(m,2H),4.28-4.16(m,2H),4.11-3.87(m,7H),3.06(d,J=10.39Hz,1H),2.88(d,J=10.29Hz,1H),2.60-2.57(m,1H),2.44-2.39(m,1H),2.32-2.24(m,4H),1.88-1.67(m,4H),1.09-1.04(m,6H).ESI-MS(m/z):928.2[M+H]+
example Ethyl bis (4R) -6- ((4- (4- ((((((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) ((((S) -1-ethoxy-1-oxopropan-2-yl) oxy) phosphono) oxy) phenyl) piperidin-1-yl) methyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (Compound 2)
Figure BDA0001712943790000161
After compounds 1-5(30mg, 0.04mmol), ethyl L-lactate (8.0mg, 0.07mmol) and PyBOP (38mg, 0.07mmol) were dissolved in N, N-dimethylformamide (3mL) at room temperature and completely dissolved, N-diisopropylethylamine (19mg, 0.14mmol) was added dropwise, and after completion of the addition, the reaction was carried out at room temperature for about 4 hours to obtain the title compound (5.0mg) after purification.
The structure is characterized as follows:
1H NMR(400MHz,DMSO-d6)9.77(s,1H),8.13(s,1H),8.05(s,1H),8.03(d,J=23.26Hz,1H),7.95(d,J=3.13Hz,1H),7.44-7.40(m,2H),7.27-7.16(m,5H),7.05-7.03(m,2H),6.06(s,1H),5.00-4.92(m,1H),4.31-3.87(m,11H),3.06(d,J=9.75Hz,1H),2.88(d,J=11.02Hz,1H),2.60-2.57(m,1H),2.43-2.40(m,1H),2.33-2.26(m,1H),1.88-1.68(m,4H),1.35(dd,J=21.76Hz,6.84Hz,3H),1.10(d,J=6.22Hz,3H),1.05(t,J=7.11Hz,3H),1.17(dt,J=7.11Hz,3.05Hz,3H).ESI-MS(m/z):924.2[M+H]+
example Ethyl tris (4R) -6- ((4- (4- ((((((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) ((((R) -1-ethoxy-1-oxopropan-2-yl) oxy) phosphono) oxy) phenyl) piperidin-1-yl) methyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (Compound 3)
Figure BDA0001712943790000171
At room temperature, compound 1-5(30mg, 0.04mmol), ethyl D-lactate (8.0mg, 0.07mmol) and PyBOP (38mg, 0.07mmol) were dissolved in N, N-dimethylformamide (3mL) and after complete dissolution, N-diisopropylethylamine (19mg, 0.14mmol) was added dropwise and reacted at room temperature for 4h to obtain the title compound (6.0mg) after purification.
The structure is characterized as follows:
1H NMR(400MHz,CDCl3)9.80(brs,1H),8.34(s,1H),7.99-7.89(m,2H),7.44(s,1H),7.33(s,1H),7.19(d,J=8.15Hz,2H),7.14-7.11(m,1H),7.01-6.92(m,3H),6.22(s,1H),5.64(brs,2H),5.05-4.98(m,1H),4.39-4.34(m,1H),4.23-3.79(m,10H),3.02(d,J=46.30Hz,2H),2.54-2.38(m,3H),1.90-1.86(m,4H),1.51(dd,J=25.54Hz,6.92Hz,3H),1.29-1.21(m,6H),1.14(t,J=7.10Hz,3H).ESI-MS(m/z):924.2[M+H]+
example Ethyl tetrakis (4R) -6- ((4- (4- (((2, 5, 8, 11, 14, 17, 20-heptaoxadocosan-22-yloxy) (((((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) phosphono) oxy) phenyl) piperidin-1-yl) methyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (Compound 4)
Figure BDA0001712943790000172
After compound 1-5(30mg, 0.04mmol), heptaethyleneglycol monomethyl ether (24.0mg, 0.07mmol) and PyBOP (38mg, 0.07mmol) were dissolved in N, N-dimethylformamide (3mL) at room temperature and completely dissolved, N-diisopropylethylamine (19mg, 0.14mmol) was added dropwise, and after completion of the addition, the reaction was carried out at room temperature for 4 hours to obtain the title compound (5.0mg) after purification.
The structure is characterized as follows:
1H NMR(400MHz,CDCl3)9.78(brs,1H),8.33(d,J=2.16Hz,1H),7.97(d,J=13.35Hz,1H),7.90(dd,J=5.33Hz,3.17Hz,1H),7.44(d,J=2.98Hz,1H),7.32(dd,J=8.45Hz,6.24Hz,1H),7.22-7.16(m,2H),7.14-7.09(m,2H),7.01(d,J=7.95Hz,1H),6.91(td,J=8.36Hz,2.27Hz,1H),6.22(s,1H),5.79(brs,2H),4.36(ddd,J=14.48Hz,6.43Hz,2.82Hz,1H),4.30-4.21(m,1H),4.17-4.12(m,1H),4.06-3.98(m,4H),3.88(d,J=17.09Hz,1H),3.81-3.73(m,1H),3.64-3.53(m,28H),3.37(s,3H),3.05(d,J=10.76Hz,1H),2.92(d,J=10.87Hz,1H),2.53-2.47(m,2H),2.35(t,J=8.41 Hz,1H),1.89-1.77(m,4H),1.24(dd,J=6.02Hz,4.16Hz,3H),1.13(t,J=7.10Hz,3H).ESI-MS(m/z):573.8[M/2+H]+
example Ethyl penta (4R) -6- ((4- (4- ((((((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) (pentyloxy) phosphono) oxy) phenyl) piperidin-1-yl) methyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (Compound 5)
Figure BDA0001712943790000173
After compounds 1-5(30mg, 0.04mmol), N-pentanol (6.0mg, 0.07mmol) and PyBOP (38mg, 0.07mmol) were dissolved in N, N-dimethylformamide (3mL) at room temperature and completely dissolved, N-diisopropylethylamine (19mg, 0.14mmol) was added dropwise, and after completion of addition, the reaction was carried out at room temperature for 3.5 hours to obtain the title compound (4.0mg) after purification.
The structure is characterized as follows:
1H NMR(400MHz,DMSO-d6)9.77(s,1H),8.13(s,1H),8.04-8.03(m,2H),7.95(d,J=3.08Hz,1H),7.44-7.40(m,2H),7.27-7.16(m,5H),7.07(d,J=7.99Hz,1H),7.00(d,J=8.27Hz,1H),6.06(s,1H),4.30-4.16(m,2H),4.03-3.91(m,9H),3.06(d,J=10.38Hz,1H),2.88(d,J=11.44Hz,1H),2.60-2.58(m,1H),2.44-2.39(m,1H),2.33-2.26(m,1H),1.87-1.67(m,4H),1.53-1.43(m,2H),1.23-1.18(m,4H),1.11(t,J=6.57Hz,3H),1.05(t,J=7.11Hz,3H).ESI-MS(m/z):894.3[M+H]+
example Ethyl hexa (4R) -6- ((4- (4- ((((((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) ((pivaloyloxy) methoxy) phosphono) oxy) phenyl) piperidin-1-yl) methyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (Compound 6)
Figure BDA0001712943790000181
After compound 1-5(30mg, 0.04mmol) was dissolved in N-methylpyrrolidone (2mL) at room temperature and completely dissolved, N-diisopropylethylamine (9mg, 0.07mmol) and iodomethyl pivalate (13mg, 0.06mmol) were added in this order, and after completion of addition, reaction was carried out at room temperature for 1.5h to obtain the title compound (5.0mg) after purification.
The structure is characterized as follows:
1H NMR(400MHz,CDCl3)9.19(s,2H),8.38-8.34(m,1H),7.95-7.89(m,2H),7.44(m,1H),7.35-7.30(m,1H),7.23-7.19(m,2H),7.17-7.08(m,2H),7.03-6.90(m,3H),6.22(s,1H),5.75-5.59(m,2H),4.40-4.34(m,2H),4.11-3.98(m,5H),3.82-2.75(m,1H),3.48-3.35(m,2H),3.11-3.01(m,1H),2.94-2.84(m,1H),2.59-2.42(m,2H),1.98-1.84(m,4H),1.34-1.12(m,15H).ESI-MS(m/z):938.2[M+H]+
example Ethyl hepta (4R) -6- ((4- ((((((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) (((S) -1-ethoxy-1-oxopropan-2-yl) oxy) phosphono) oxy) -3, 3-difluoropiperidin-1-yl) methyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (Compound 27)
Figure BDA0001712943790000182
The method comprises the following steps: synthesis of 4- (((((((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) (hydroxy) phosphono) oxy) -3, 3-difluoropiperidine-1-carboxylic acid tert-butyl ester (Compound 7-2)
(R) -9- (2-methoxypropyl-phosphate) -adenine (55mg, 0.19mmol) and tert-butyl 3, 3-difluoro-4-hydroxypiperidine-1-carboxylate (compound 7-1) (30mg, 0.13mmol) were dissolved in pyridine (2mL) at room temperature, dicyclohexylcarbodiimide (80mg, 0.39mmol) was added under nitrogen, and the mixture was heated to 80 ℃ for reaction overnight. Purification gave the title compound (10 mg). ESI-MS (m/z): 507.1[ M + H]+
Step two: synthesis of ((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) - (3, 3-difluoropiperidin-4-yl) -monophosphate trifluoroacetate (Compound 7-3)
Compound 7-2(10mg, 0.02mmol) was added to methylene chloride (1.0mL) at room temperature, trifluoroacetic acid (0.2mL) was added to dissolve completely, and after completion of the addition, the reaction was carried out at room temperature. The reaction solution was evaporated to dryness under reduced pressure to give the title compound (10mg), which was used directly in the next reaction without purification. ESI-MS (m/z): 407.1[ M + H ] +.
Step three: synthesis of ethyl (4R) -6- ((4- ((((((((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) (hydroxy) phosphonyl) oxy) -3, 3-difluoropiperidin-1-yl) methyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (Compound 7-4)
Compound 7-3(10mg, 0.025mmol) was dissolved in 1, 2-dichloroethane (2mL) at room temperature, and ethyl (R) -6- (bromomethyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (14mg, 0.030mmol) and N, N-diisopropylethylamine (0.1mL) were added in this order, and after completion of the addition, the reaction was allowed to proceed at room temperature overnight. Purification gave the title compound (10 mg). ESI-MS (m/z): 784.1[ M + H]+
Step four: synthesis of ethyl (4R) -6- ((4- (((((((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) (((S) -1-ethoxy-1-oxopropan-2-yl) oxy) phosphonyl) oxy) -3, 3-difluoropiperidin-1-yl) methyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (Compound 27)
After the compounds 7-4(40mg, 0.05mmol), ethyl L-lactate (10mg, 0.1mmol) and PyBOP (52mg, 0.1mmol) were dissolved in N, N-dimethylformamide (3mL) at room temperature and dissolved completely, N-diisopropylethylamine (26mg, 0.2mmol) was added dropwise, and after completion of the addition, the reaction was allowed to proceed at room temperature overnight. Purification gave the title compound (10 mg).
The structure is characterized as follows:
1H NMR(400MHz,CDCl3)9.46(d,J=11.90Hz,1H),8.35(t,J=2.16Hz,1H),8.07-7.98(m,1H),7.85(d,J=3.24,1H),7.43(d,J=2.87Hz,1H),7.31(ddd,J=9.02,6.15,3.19Hz,1H),7.13(dd,J=8.60,2.57Hz,1H),6.93(td,J=8.32,2.42Hz,1H),6.21(t,J=2.29Hz,1H),5.91(s,2H),5.02-4.90(m,1H),4.78-4.67(m,1H),4.45-4.34(m,1H),4.27-3.90(m,9H),3.74-3.66(m,1H),3.02-2.63(m,3H),2.25-1.99(m,3H),1.55-1.44(m,3H),1.31-1.23(m,6H),1.13(t,J=7.16,1.39Hz,3H).ESI-MS(m/z):884.1[M+H]+
EXAMPLE Ethyl octa (4R) -6- ((4- ((((((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) (hydroxy) phosphono) oxy) -3, 3-difluoropyrrolidin-1-yl) methyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (Compound 28)
Figure BDA0001712943790000191
The method comprises the following steps: synthesis of tert-butyl 4- (((((((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) (hydroxy) phosphono) oxy) -3, 3-difluoropyrrolidine-1-carboxylate (Compound 8-2)
Tenofovir (Compound 8-1) (966mg, 3.36mmol) and tert-butyl 3, 3-difluoro-4-hydroxypyrrolidine-1-carboxylate (500mg, 2.24mmol) were dissolved in pyridine (6mL) at room temperature, dicyclohexylcarbodiimide (1.39g, 6.72mmol) was added under nitrogen, and the temperature was raised to 80 ℃ for reaction overnight. Purification gave the title compound (240 mg). ESI-MS (m/z): 493.1[ M + H ] +.
Step two: synthesis of ((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) -4, 4-difluoropyrrolidin-3-yl-monophosphate trifluoroacetate (Compound 8-3)
Compound 8-2(100mg, 0.2mmol) was added to dichloromethane (1.5mL) at room temperature, after completion of dissolution, trifluoroacetic acid (0.5mL) was added, and after completion of addition, the reaction was carried out at room temperature. The solvent was evaporated under reduced pressure to dryness to give the title compound (120mg), which was used in the next reaction without purification. ESI-MS (m/z): 393.1[ M + H]+
Step three: synthesis of ethyl (4R) -6- ((4- ((((((((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) (hydroxy) phosphono) oxy) -3, 3-difluoropyrrolidin-1-yl) methyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (Compound 28)
Compound 8-3(80mg, 0.2mmol) was dissolved in dichloromethane (4mL) at room temperature, and (R) -6- (bromomethyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylic acid ethyl ester (100mg, 0.22mmol) and N, N-diisopropylethylamine (0.5mL) were added in this order, and after completion, the reaction was allowed to proceed at room temperature overnight. Purification gave the title compound (80 mg).
The structure is characterized as follows:
1h NMR (400MHz, methanol-d)4)8.31(d,J=1.6Hz,1H),8.18(d,J=2.7Hz,1H),7.87(dd,J=5.3,3.2Hz,1H),7.67(dd,J=5.8,3.1Hz,1H),7.46-7.39(m,1H),7.21(dd,J=8.8,2.6Hz,1H),7.08-7.02(m,2H),6.14(d,J=2.0Hz,1H),4.87-4.75(m,1H),4.38-4.32(m,1H),4.24-4.00(m,5H),3.88-3.84(m,1H),3.77-3.68(m,1H),3.40-3.36(m,1H),3.29-2.95(m,2H),2.78-2.76(m,1H),1.13-1.07(m,6H).ESI-MS(m/z):770.1[M+H]+
Example nine Ethyl (4R) -6- ((4- ((((((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) (((S) -1-ethoxy-1-oxopropan-2-yl) oxy) phosphono) oxy) -3, 3-difluoropyrrolidin-1-yl) methyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (Compound 29)
Figure BDA0001712943790000201
The title compound (20mg) was obtained by performing a similar procedure as described in example seven step four, substituting compound 28 for compound 7-4. ESI-MS (m/z): 870.1[ M + H]+
Example Ethyl deca (4R) -6- ((4- ((((((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) (((R) -1-ethoxy-1-oxopropan-2-yl) oxy) phosphono) oxy) -3, 3-difluoropyrrolidin-1-yl) methyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (Compound 30)
Figure BDA0001712943790000202
In a similar manner to that described in example nine, ethyl L-lactate was replaced with ethyl D-lactate to give the title compound (10 mg). ESI-MS (m/z): 870.1[ M + H]+
EXAMPLE undec 2- (((((((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) ((1- (((R) -6- (2-chloro-4-fluorophenyl) -5- (ethoxycarbonyl) -2- (thiazol-2-yl) -3, 6-dihydropyrimidin-4-yl) methyl) -4, 4-difluoropyrrolidin-3-yl) oxy) phosphono) oxy) acetic acid (Compound 31)
Figure BDA0001712943790000203
Compound 28(30mg, 0.04mmol) was dissolved in N-methylpyrrolidone (2mL) at room temperature, bromoacetic acid (27mg, 0.20mmol) and potassium carbonate (16mg, 0.12mmol) were added in this order, and after the addition, the temperature was raised to 60 ℃ to react for 2 hours. The reaction solution was filtered, and the filtrate was purified to give the title compound (5.0 mg). ESI-MS (m/z): 828.0[ M + H]+
Example Ethyl dodeca (4R) -4- (2-chloro-4-fluorophenyl) -6- ((3, 3-difluoro-4- (((((2S, 3R, 5S) -3-hydroxy-5- (5-methyl-2, 4-dioxo-3, 4-dihydropyrimidin-1 (2H) -yl) tetrahydrofuran-2-yl) methoxy) (((S) -1-isopropoxy-1-oxoprop-2-yl) amino) phosphonyl) oxy) pyrrolidin-1-yl) methyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (Compound 32).
Figure BDA0001712943790000211
The method comprises the following steps: synthesis of ethyl (4R) -4- (2-chloro-4-fluorophenyl) -6- ((4- ((dichlorophosphono) oxy) -3, 3-difluoropyrrolidin-1-yl) methyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (Compound 12-2)
At room temperature, (4R) -ethyl 4- (2-chloro-4-fluorophenyl) -6- ((3, 3-difluoro-4-hydroxypyrrolidin-1-yl) methyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (compound 12-1) (50mg, 0.1mmol) was dissolved in dichloromethane (2mL), cooled to-20 ℃ under nitrogen protection, phosphorus oxychloride (0.2mL) was added dropwise, and after completion, triethylamine (0.2mL) was added dropwise and reacted at-20 ℃ for 30 min. The reaction solution is directly spin-dried, and the obtained crude product is directly used for the next reaction without purification.
Step two: synthesis of ethyl (4R) -4- (2-chloro-4-fluorophenyl) -6- ((3, 3-difluoro-4- ((((((S) -1-isopropoxy-1-oxoprop-2-yl) amino) (pentafluorophenoxy) phosphono) oxy) pyrrolidin-1-yl) methyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (Compound 12-3)
At room temperature, dissolving the compound 12-2(60mg, 0.1mmol) in dichloromethane (2mL), cooling to-20 ℃ under the protection of nitrogen, dropwise adding a dichloromethane (1.0mL) solution of L-alanine isopropyl ester hydrochloride (50mg, 0.3mmol), after the addition, dropwise adding a dichloromethane (0.5mL) solution of triethylamine (0.2mL), and moving to room temperature for reaction for 1 h. The temperature is reduced to-20 ℃ under the protection of nitrogen, a solution of pentafluorophenol (50mg, 0.3mmol) and triethylamine (0.1mL) in dichloromethane (2.0mL) is added dropwise, the reaction is carried out for 1h at-20 ℃, the reaction solution is directly dried by spinning, tetrahydrofuran (4.0mL) is used for dissolving, insoluble substances are filtered, and the filtrate is directly used for the next reaction.
Step three: synthesis of ethyl (4R) -4- (2-chloro-4-fluorophenyl) -6- ((3, 3-difluoro-4- ((((((2S, 3R, 5S) -3-hydroxy-5- (5-methyl-2, 4-dioxo-3, 4-dihydropyrimidin-1 (2H) -yl) tetrahydrofuran-2-yl) methoxy) ((((S) -1-isopropoxy-1-oxoprop-2-yl) amino) phosphonyl) oxy) pyrrolidin-1-yl) methyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylate (Compound 32)
And (2) dissolving telbivudine (50mg, 0.2mmol) in tetrahydrofuran (2mL) at room temperature, cooling to-20 ℃ under the protection of nitrogen, dropwise adding tert-butyl magnesium chloride (0.4mL, 1M), transferring to room temperature after addition, reacting for 1h, cooling to-20 ℃ under the protection of nitrogen, dropwise adding the tetrahydrofuran solution obtained in the second step, and reacting at room temperature overnight under the protection of nitrogen. Purification gave the title compound (2.0 mg). ESI-MS (m/z): 918.0[ M + H]+
EXAMPLE thirteen (4R) -6- ((4- ((((((R) -1- (6-amino-9H-purin-9-yl) propan-2-yl) oxy) methyl) (((R) -1-ethoxy-1-oxopropan-2-yl) oxy) phosphono) oxy) -3, 3-difluoropiperidin-1-yl) methyl) -4- (2-chloro-4-fluorophenyl) -2- (thiazol-2-yl) -1, 4-dihydropyrimidine-5-carboxylic acid ethyl ester (Compound 33)
Figure BDA0001712943790000212
In a similar manner to that described in seven step four of the example, ethyl L-lactate was replaced with ethyl D-lactate to give the title compound (15 mg). ESI-MS (m/z): 884.1[ M + H]+
Other compounds in the present application can be synthesized by methods similar to those in the above examples.
Biological activity assay
The compounds of the present invention were tested for their inhibitory effect on Hepatitis B Virus (HBV). The compounds of the invention were tested at the virus-cell level for cytotoxicity as well as for their effect on the level of viral (HBV) nucleic acid (DNA) replication.
Test method
HepG2.2.15 cells in the logarithmic growth phase were seeded in 96-well plates at a cell concentration of 40 cells/. mu.L. At 37 ℃ with 5% CO2Culturing in an incubator for 3 days; the medium was replaced with fresh medium (200. mu.L/well) before addition of the compound. The concentration of the mother liquor for each test compound was 200. mu.M. At 200. mu.M maximum concentration, diluted in DMSO to various concentrations, 1. mu.L of test compound was placed in the corresponding medium well and the final test concentrations of compound were 0.06, 0.24, 0.98, 3.9, 15.6, 62.5, 250, 1000nM (for half the Effective Concentration (EC) calculation50))。
The stock solutions of each test compound were diluted with DMSO at 600 μ M and 60 μ M, and 1 μ L each was added to the corresponding medium well, with the final test concentrations of compound at 3 μ M and 0.3 μ M (used to calculate percent inhibition).
Blank control: 1 μ of LDMSO was added to the corresponding medium wells as a control.
After addition of the test compound at 37 ℃ with 5% CO2The incubator was co-cultured for 10 days, the medium was changed every two days, and the compound was re-added.
At day 11, 150 μ L of cell supernatant per well was collected for qPCR detection.
Is tested forEC of the Compound50And the results of the inhibition ratios at 3. mu.M and 0.3. mu.M concentrations are shown in tables 1 and 2.
TABLE 1
Compound (I) EC50(nM)
2 59.43
3 54.65
27 52.1
29 5.18
30 8.21
32 26.95
33 53.1
As can be seen from the data in Table 1, the tested compounds showed strong inhibitory activity in the activity test for inhibiting the replication of deoxyribonucleic acid (DNA) of Hepatitis B Virus (HBV), and the EC of some compounds50< 60nM, EC of preferred compounds (e.g. compounds 29 and 30)50Less than 10nM, indicating that the compounds of the invention have very excellent inhibitionsPreparing the activity.
TABLE 2
Compound (3. mu.M) Inhibition ratio%
1 96.54
2 100.20
3 101.67
As can be seen from the above results, the tested compounds showed excellent inhibitory effects in the activity test for inhibiting deoxyribonucleic acid (DNA) replication of Hepatitis B Virus (HBV) at a concentration of 3 μ M. The tested compounds also showed excellent inhibitory effect at a concentration of 0.3 μ M, indicating that the compounds of the present invention have very excellent inhibitory effect.
Cytotoxicity assays
Diluting the test compound with DMSO to 30mM, taking 30mM as the highest concentration, diluting three times to multiple different concentrations, taking 0.2 μ L of each concentration of the test compound in a 384-well plate, adding 2000/50 μ L of HepG22.2.15 cells into each well, and obtaining the maximum final concentration of the test compound of 150 μ M; mu.L of DMSO was added to the corresponding wells as a control.
At 37 ℃ with 5% CO2The culture was carried out in an incubator for 4 days.
After 4 days, 50. mu.L of CellTiter-Glo was added to each well, and plate reading was performed to calculate half-Cytotoxic Concentration (CC)50) The value is obtained.
Preferred compounds (e.g. of example 5)Compound) CC50The value is more than 150 mu M, which indicates that the cytotoxicity is lower and the safety is higher.
Various modifications of the invention in addition to those described herein will be apparent to those skilled in the art from the foregoing description. Such modifications are also intended to fall within the scope of the appended claims. Each reference, including all patents, patent applications, journal articles, books, and any other publications, cited in this application is hereby incorporated by reference in its entirety.

Claims (20)

1. A compound, or a pharmaceutically acceptable salt, stereoisomer, or tautomer thereof, wherein the compound has the structure of formula (II):
Figure FDA0002572689930000011
wherein:
x and Y are O;
R2selected from H, C1-12Alkyl radical, C3-6Cycloalkyl radical, C6-14Aryl, 5-14 membered heteroaryl, C6-20Aralkyl, -C1-6alkylene-COOH, -C1-6alkylene-C (═ O) O-C1-6Alkyl, -C1-6alkylene-OC (═ O) -C1-6Alkyl, -C1-6alkylene-OC (═ O) O-C1-6Alkyl and- ((CH)2)iO)m-(CH2)q-O-C1-6Alkyl, each of the above groups being optionally substituted by one or more groups selected from halogen, -OH, -CN, -NO2、-N(R)2、C1-6Alkyl, halo C1-6Alkyl radical, C1-6Alkylthio and C3-6Cycloalkyl substituents;
i and q are each independently at each occurrence 1, 2, 3, 4, 5, or 6;
m is any integer selected from 0 to 6;
R1selected from:
Figure FDA0002572689930000012
Figure FDA0002572689930000013
represents a 3 to 14 membered azacyclic ring system optionally additionally containing a substituent independently selected from the group consisting of N, O, C ═ O, S, S ═ O and S (═ O)21, 2 or 3 ring members of (a);
Ar1and Ar2Each independently selected from C6-14Aryl and 5-14 membered heteroaryl, optionally substituted with one or more substituents selected from halogen, -OH, -CN, -NO2、-N(R)2、C1-6Alkyl, halo C1-6Alkyl radical, C1-6Alkylthio and C3-6Cycloalkyl substituents;
l is absent or selected from-O-, -S-and-NR-;
R3and R4Each independently selected from H, C1-4Alkyl and C3-6A cycloalkyl group;
R5at each occurrence, is attached to the rest of the molecule with a single or double bond;
R5and R6Independently at each occurrence, is selected from halogen, -OH, -COOH, -CN, -NO2、-N(R)2、C1-6Alkyl, halo C1-6Alkyl, -W-C1-6Alkyl, -C1-6alkylene-W-R, -W-C1-6alkylene-W' -R, -W-C2-6Alkenyl, -C2-6alkenylene-W-R, -W-C2-6alkenylene-W' -R and C3-6Cycloalkyl, wherein the alkylene and alkenylene are optionally further interrupted by one or more W;
w and W' are each independently at each occurrence selected from O, C (═ O), C (═ O) O, NR, S ═ O, and S (═ O)2
R at each occurrence is independently selected from H, C1-6Alkyl and C3-6A cycloalkyl group;
n is independently at each occurrence 0, 1, 2, 3, 4 or 5, provided that n is no greater than the number of positions on the corresponding group that may be substituted; and is
When n is greater than 1, each R5Which may be the same or different, each R6May be the same or different.
2. The compound of claim 1, or a pharmaceutically acceptable salt, stereoisomer, or tautomer thereof, wherein R2Selected from H, C1-12Alkyl radical, C3-6Cycloalkyl radical, C6-14Aryl, 5-14 membered heteroaryl, C6-20Aralkyl, -C1-6alkylene-C (═ O) O-C1-6Alkyl, -C1-6alkylene-OC (═ O) -C1-6Alkyl, -C1-6alkylene-OC (═ O) O-C1-6Alkyl and- ((CH)2)iO)m-(CH2)q-O-C1-6Alkyl, each of the above groups being optionally substituted by one or more groups selected from halogen, -OH, -CN, -NO2、-N(R)2、C1-6Alkyl, halo C1-6Alkyl radical, C1-6Alkylthio and C3-6Cycloalkyl substituents.
3. The compound of claim 1 or 2, or a pharmaceutically acceptable salt, stereoisomer, or tautomer thereof, wherein R1Selected from:
Figure FDA0002572689930000021
4. a compound of claim 1 or 2, or a pharmaceutically acceptable salt, stereoisomer, or tautomer thereof, wherein Ar is1Selected from:
Figure FDA0002572689930000022
wherein R iscEach occurrence is independently selected from F, Cl, Br, I, C1-6Alkyl, halo C1-6Alkyl and C3-6A cycloalkyl group.
5. The process of claim 4A compound or a pharmaceutically acceptable salt, stereoisomer or tautomer thereof, wherein RcEach occurrence is independently selected from F, Cl, Br, I, C1-6Alkyl and C3-6A cycloalkyl group.
6. The compound of claim 4, or a pharmaceutically acceptable salt, stereoisomer, or tautomer thereof, wherein Ar1Selected from:
Figure FDA0002572689930000023
7. the compound of claim 6, or a pharmaceutically acceptable salt, stereoisomer, or tautomer thereof, wherein Ar1Selected from:
Figure FDA0002572689930000024
8. a compound of claim 1 or 2, or a pharmaceutically acceptable salt, stereoisomer, or tautomer thereof, wherein Ar is2Selected from:
Figure FDA0002572689930000025
the above groups are optionally substituted by one or more groups selected from halogen, C1-6Alkyl, halo C1-6Alkyl and C3-6Cycloalkyl groups.
9. The compound of claim 8, or a pharmaceutically acceptable salt, stereoisomer, or tautomer thereof, wherein Ar is2Selected from:
Figure FDA0002572689930000026
the above groups are optionally substituted by one or more groups selected from halogen, C1-6Alkyl and C3-6Cycloalkyl groups.
10. The compound of claim 1 or 2, or a pharmaceutically acceptable salt, stereoisomer, or tautomer thereof, wherein R2Selected from: c1-6Alkyl (- ((CH))2)iO)m-(CH2)q-O-C1-6Alkyl, aryl, heteroaryl, and heteroaryl,
Figure FDA0002572689930000027
Wherein:
i and q are each independently at each occurrence 1, 2, 3, 4, 5, or 6;
m is any integer selected from 0 to 6;
R7、R8and R9Each independently selected from H, C1-10Alkyl radical, C3-6Cycloalkyl radical, C6-20Aryl and C7-20Aralkyl, said alkyl, cycloalkyl, aryl and aralkyl each optionally substituted with one or more substituents selected from the group consisting of halogen, -OH, -CN and-NO2Substituted with the substituent(s);
or R7And R8Together with the carbon atom to which they are attached form C3-6A cycloalkyl group;
R10and R11Each independently selected from H, halogen, -OH, -CN, -NO2、C1-10Alkyl and C3-6A cycloalkyl group.
11. The compound of claim 1 or 2, or a pharmaceutically acceptable salt, stereoisomer, or tautomer thereof, wherein the compound has the structure of any of the following formulae:
Figure FDA0002572689930000031
12. the compound of claim 1 or 2, or a pharmaceutically acceptable salt, stereoisomer, or tautomer thereof, wherein the compound is selected from the group consisting of:
Figure FDA0002572689930000032
Figure FDA0002572689930000041
13. a pharmaceutical composition comprising a prophylactically or therapeutically effective amount of a compound according to any one of claims 1-12, or a pharmaceutically acceptable salt, stereoisomer, or tautomer thereof, and one or more pharmaceutically acceptable carriers.
14. The pharmaceutical composition of claim 13, which is a solid formulation, a liquid formulation or a transdermal formulation.
15. A process for preparing a pharmaceutical composition comprising combining a compound of any one of claims 1-12, or a pharmaceutically acceptable salt, stereoisomer, or tautomer thereof, with one or more pharmaceutically acceptable carriers.
16. Use of a compound of any one of claims 1 to 12, or a pharmaceutically acceptable salt, stereoisomer or tautomer thereof, or a pharmaceutical composition of claim 13 or 14, for the preparation of a medicament for the prophylaxis or treatment of a viral disease.
17. The use of claim 16, wherein the medicament is a medicament for administration by oral, intravenous, intraarterial, subcutaneous, intraperitoneal, intramuscular or transdermal routes.
18. The use of claim 16, wherein the viral disease is selected from the group consisting of viral hepatitis a, viral hepatitis b, viral hepatitis c, influenza, herpes, and Acquired Immune Deficiency Syndrome (AIDS).
19. A process for the preparation of a compound according to any one of claims 1 to 12, comprising the steps of:
Figure FDA0002572689930000051
or
Figure FDA0002572689930000052
Wherein:
b is
Figure FDA0002572689930000053
A is
Figure FDA0002572689930000054
Wherein position 1 is attached to B and position 2 is attached to the phosphorus atom (P);
R12、R13and R14Each independently selected from F, Cl, Br, I, -NHR, hydroxy, triflate, p-toluenesulfonate and boronate;
PG is selected from t-butoxycarbonyl, benzyloxycarbonyl, 9-fluorenylmethoxycarbonyl, allyloxycarbonyl, trimethylsiloxyethoxycarbonyl, methoxycarbonyl, ethoxycarbonyl, p-toluenesulfonyl, o-nitrobenzenesulfonyl, p-nitrobenzenesulfonyl, formyl, acetyl, trifluoroacetyl, propionyl, pivaloyl, phenyl, benzoyl, trityl, benzyl, 2, 4-dimethoxybenzyl and p-methoxybenzyl;
the remaining groups are as defined in any one of claims 1 to 12;
the first step is carried out in the presence of an organic or inorganic base and/or a condensation agent, in an aprotic solvent or without a solvent;
the second step is carried out under conditions suitable for removing the PG groups;
the third step is carried out in an aprotic solvent in the presence of an organic base or an inorganic base; and is
The fourth step is carried out in an aprotic solvent in the presence of an organic or inorganic base and/or a condensation agent;
or the method comprises the following steps:
Figure FDA0002572689930000061
wherein:
b is
Figure FDA0002572689930000062
A is
Figure FDA0002572689930000063
Wherein position 1 is attached to B and position 2 is attached to the phosphorus atom (P);
R12selected from the group consisting of F, Cl, Br, I, -NHR, hydroxy, triflate, p-toluenesulfonate and borate;
LG is a leaving group which is pentafluorophenyl and p-nitrophenyl;
the remaining groups are as defined in any one of claims 1 to 12;
the first step is carried out in an aprotic solvent in the presence of an organic or inorganic base;
the second step is carried out in an aprotic solvent in the presence of an organic base or an inorganic base; and is
The third step is carried out in an aprotic solvent in the presence of an organic or inorganic base.
20. The process of claim 19, wherein the condensation reagent is DCC, DIC, EDC, BOP, PyAOP or PyBOP.
CN201780005344.3A 2016-11-02 2017-10-27 Heterocyclic compounds, process for their preparation and their use Active CN108602842B (en)

Applications Claiming Priority (3)

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

Publications (2)

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

Family

ID=62076620

Family Applications (1)

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

Country Status (2)

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

Families Citing this family (2)

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

Citations (4)

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

Patent Citations (4)

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

Also Published As

Publication number Publication date
CN108602842A (en) 2018-09-28
WO2018082503A1 (en) 2018-05-11

Similar Documents

Publication Publication Date Title
JP5271913B2 (en) Compound
JP6263533B2 (en) Dihydropyrimidine compounds and their application in medicine
ES2324816T3 (en) DERIVATIVES OF 2-AMINOCARBONIL-9H-PURINA.
EP2892893B1 (en) 6-amino acid heteroaryldihydropyrimidines for the treatment and prophylaxis of hepatitis b virus infection
JP6524081B2 (en) Dihydropyrimidine compounds and their application in medicine
EP3782626A1 (en) Hepatitis b core protein modulators
US7820631B2 (en) Anti-viral pyrimidine nucleoside analogues
CN109790145B (en) Dihydropyrimidine compound and preparation method and application thereof
HUE033727T2 (en) 2&#39;,4&#39;-difluoro-2&#39;-methyl substituted nucleoside derivatives as inhibitors of hcv rna replication
KR20120092153A (en) 2&#39;-fluoro-6&#39;-methylene carbocyclic nucleosides and methods of treating viral infections
CN102256991A (en) Uracyl cyclopropyl nucleotides
CN102083845A (en) Cyclopropyl polymerase inhibitors
CN108602842B (en) Heterocyclic compounds, process for their preparation and their use
KR20230091929A (en) bicyclic compound
WO2018217439A1 (en) Substituted [5,6]cyclic-4(3h)-pyrimidinones as anticancer agents
WO2020114495A1 (en) Dinucleotide compound and prodrug thereof
TW200417372A (en) Antiviral nucleoside derivatives
EP4174068A1 (en) Novel compound, and pharmaceutical composition for preventing or treating resistant cancer, comprising same
WO2003061670A1 (en) Compounds useful as a3 adenosine receptor agonists
WO2020103929A1 (en) Oligonucleotide and prodrug thereof
US11166954B2 (en) Dihydropyrimidine compound and preparation method and use thereof
CA2813993A1 (en) Method of preparation of antiviral compounds and useful intermediates thereof
EA039977B1 (en) DIHYDROPYRIMIDINE COMPOUNDS AND METHOD FOR THEIR PRODUCTION AND APPLICATION
JP2020164521A (en) Antiviral drug
US20240150351A1 (en) Bicyclic compounds

Legal Events

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