CN111434671A - Liver specificity AMPK agonist and preparation method and application thereof - Google Patents

Liver specificity AMPK agonist and preparation method and application thereof Download PDF

Info

Publication number
CN111434671A
CN111434671A CN201910027560.9A CN201910027560A CN111434671A CN 111434671 A CN111434671 A CN 111434671A CN 201910027560 A CN201910027560 A CN 201910027560A CN 111434671 A CN111434671 A CN 111434671A
Authority
CN
China
Prior art keywords
group
amino
compound
purin
substituted
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CN201910027560.9A
Other languages
Chinese (zh)
Other versions
CN111434671B (en
Inventor
徐华强
栾林波
张振伟
孙锋
杨生生
高善云
戴金威
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.)
Kaisi Kaidi Shanghai Pharmaceutical Technology Co ltd
Original Assignee
Kaisi Kaidi Shanghai Pharmaceutical Technology 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 Kaisi Kaidi Shanghai Pharmaceutical Technology Co ltd filed Critical Kaisi Kaidi Shanghai Pharmaceutical Technology Co ltd
Priority to CN201910027560.9A priority Critical patent/CN111434671B/en
Priority to PCT/CN2020/071525 priority patent/WO2020143800A1/en
Publication of CN111434671A publication Critical patent/CN111434671A/en
Application granted granted Critical
Publication of CN111434671B publication Critical patent/CN111434671B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals
    • C07H19/20Purine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • C07H19/213Purine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids containing cyclic phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7032Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a polyol, i.e. compounds having two or more free or esterified hydroxy groups, including the hydroxy group involved in the glycosidic linkage, e.g. monoglucosyldiacylglycerides, lactobionic acid, gangliosides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/04Anorexiants; Antiobesity agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/06Antihyperlipidemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H1/00Processes for the preparation of sugar derivatives

Abstract

The invention provides a liver specificity AMPK agonist, a preparation method and application thereof, in particular to a compound shown as a formula I, and application thereof in treating nonalcoholic fatty liver disease (NAF L D) including nonalcoholic fatty liver disease (NAF L), nonalcoholic steatohepatitis (NASH) and related liver cirrhosis and liver cancer, or obesity, diabetes, hypertriglyceridemia, hypercholesterolemia, atherosclerosis, cardiovascular diseases, metabolic diseases and other diseases.

Description

Liver specificity AMPK agonist and preparation method and application thereof
Technical Field
For example, the compounds of the present invention are useful in the treatment of nonalcoholic fatty liver disease (NAF L D), including nonalcoholic fatty liver disease (NAF L), nonalcoholic steatohepatitis (NASH) and its associated cirrhosis, liver cancer, as well as metabolic diseases such as obesity, diabetes, hypertriglyceridemia, hypercholesterolemia, atherosclerosis, cardiovascular disease, and the like.
Background
AMPK is known as AMP Activated Protein Kinase, namely AMP-dependent Protein Kinase, and is a heterotrimeric Protein Kinase which is composed of a catalytic subunit α, a regulatory subunit β and gamma, wherein the Protein Kinase is an energy Kinase which can sense in vivo AMP changes, and when the concentration of AMP in vivo rises to a certain extent, AMPK is Activated, which inhibits anabolism and promotes catabolism, and the net effect of activation is the inhibition of ATP consuming processes and the activation of ATP producing pathways, thereby regenerating ATP stores.
AMPK can control the invasion and metastasis of tumors by regulating the activity of cancer suppressor genes and improving energy metabolism disorder. When the energy metabolism of cells is unbalanced, AMPK can be activated by phosphorylating serine residue of downstream signal P53, and further negatively regulates the m TOR pathway to interrupt the proliferation of tumors and reduce the invasion and metastasis of the tumors. Protein Kinase B (PKB) is associated with metastasis of tumor, while AMPK may affect metastasis of tumor a through modulation of PKB. In liver cancer patients taking chemotherapeutic drugs for a long time, Fas Receptor (FasR) promoting tumor cell apoptosis can be activated in drug-resistant tumor cells, tumor invasiveness is increased by inducing activation of transcription factor NF-k B (NF-k B), and SNARK in AMPK family can affect invasive metastasis by regulating CD95-NF-k B pathway. Also, AMPK can reduce the invasiveness of tumor cells by affecting MMP-2 and MMP-9 proteins in the Matrix Metalloproteinase (MMP) family.
Due to different combination forms of various subunits of AMPK, the AMPK theoretically has a 12-protein combination form, and α 1 subunit and gamma 2 subunit have different variable splice bodies (alternative splice variants), so that the complexity of the AMPK trimer is further increased.
Disclosure of Invention
The invention synthesizes prodrug molecules of thio nucleoside monophosphate (AMPS) and analogues thereof, and the drugs are metabolized in liver tissues to obtain parent molecules after being taken orally.
In a first aspect of the present invention, there is provided a compound represented by the following formula I, or a stereoisomer or a tautomer thereof, or a pharmaceutically acceptable salt, hydrate, or solvate thereof,
Figure BDA0001943047350000021
wherein:
R1selected from the group consisting of: substituted or unsubstituted C6-C18 aryl, substituted or unsubstituted 5-12 membered heteroaryl;
R2selected from the group consisting of: hydrogen, substituted or unsubstituted C1-C18 alkyl, substituted or unsubstituted C3-C8 cycloalkyl, substituted or unsubstituted C2-C18 alkanoyl, substituted or unsubstituted C2-C18 alkoxycarbonyl, mono-or di-C2-C18 alkylaminocarbonyl: halogen, haloalkyl, nitro, hydroxy, amino and cyano;
R3and R4Each independently selected from the group consisting of: hydrogen, fluorine, C1-C6 alkyl, C1-C6 alkoxy; or R3And R4Together form a group selected from: a C3-C8 carbocyclic ring, or a 5-12 membered heterocyclic ring;
x is O, S, NH, substituted or unsubstituted C1-C4 alkylene;
wherein said substitution means that the hydrogen atoms on the group are substituted by one or more (e.g. 2,3,4, etc.) substituents selected from the group consisting of: halogen, deutero, C1-C6 alkoxy, halogenated C1-C6 alkyl, halogenated C1-C6 alkoxy, halogenated C3-C8 cycloalkyl, methylsulfonyl, -S (═ O)2NH2Oxo (═ O), -CN, hydroxy, -NH2Carboxy, C1-C6 amido (-C (═ O) -N (Rc)2or-NH-C (═ O) (Rc), Rc being H or C1-C5 alkyl), C1-C6 alkyl- (C1-C6 carboxamido)Or a substituted or unsubstituted group selected from: C1-C6 alkyl, C3-C8 cycloalkyl, C1-C6 amino, C6-C10 aryl, 5-10 membered heteroaryl having 1-3 heteroatoms selected from N, S and O, 5-12 membered heterocyclyl having 1-3 heteroatoms selected from N, S and O, - (CH) 6 alkyl2) -C6-C10 aryl, - (CH)2) - (5-to 10-membered heteroaryl having 1 to 3 heteroatoms selected from N, S and O), and the substituents are selected from the group consisting of: halogen, C1-C6 alkyl, C1-C6 alkoxy, oxo, -CN, -NH2OH, -C6-C10 aryl, -C1-C6 amino, -C1-C6 amido, -5-10 membered heteroaryl having 1-3 heteroatoms selected from N, S and O.
In another preferred embodiment, the compound of formula I has a structure selected from the group consisting of:
Figure BDA0001943047350000022
in another preferred embodiment, said X is selected from the group consisting of: o, S, NH, CH2、CF2Or CD2
In another preferred embodiment, R is1Has a structure as shown in formula II, III, IV, V, VI or VII:
Figure BDA0001943047350000031
wherein:
the dotted line is a bond or nothing;
each A is1、A2、A3、A5、A6、A7、A8Each independently is O, S, N, NH, CH or CH2;A4And A9Each independently is C or N;
each B1、B2、B3、B4、B6、B7、B8、B9Each independently is O, S, N, NH, CH or CH2;B5And B10Each independently is C or N;
each R is5、R6、R7And R8Each independently selected from the group consisting of: halogen, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C1-C6 alkoxy;
i is 0, 1,2, 3,4 or 5;
Y1o, S or NH;
Y2and Y3Each independently selected from O, N or CH;
j is 0, 1,2, 3 or 4;
m is 0, 1,2 or 3;
n is 0, 1,2, 3 or 4;
in another preferred embodiment, R is2Selected from the group consisting of: acetyl and butyryl.
In another preferred embodiment, R is3,R4Each independently a hydrogen atom.
In another preferred embodiment, the compound of formula (I) is a prodrug of a direct AMPK agonist.
In another preferred embodiment, the compounds of formula (I) may have one or more chiral centers and thus exist in a variety of stereoisomeric forms, including tautomers, cis-trans isomers, conformers, meso compounds, optical isomers having enantiomeric or diastereomeric relationships, and mixtures of the various isomers which may occur.
In another preferred embodiment, the compound has a structure selected from the group consisting of:
Figure BDA0001943047350000032
in another preferred embodiment, the compound is selected from the structures shown in the following group:
Figure BDA0001943047350000041
in another preferred embodiment, the compound has the structure shown in table 1:
in a second aspect of the invention, there is provided a pharmaceutical composition comprising (a) a therapeutically effective amount of a compound as described in the first aspect of the invention, or a pharmaceutically acceptable salt, hydrate or solvate thereof; and (b) a pharmaceutically acceptable carrier.
In another preferred embodiment, the disease or condition is selected from the group consisting of nonalcoholic fatty liver disease (NAF L), nonalcoholic steatohepatitis (NASH) and its associated cirrhosis, liver cancer, nonalcoholic fatty liver disease (NAF L D), obesity, diabetes, hypertriglyceridemia, hypercholesterolemia, atherosclerosis, cardiovascular disease, metabolic disease.
In a third aspect of the invention, there is provided the use of a compound of formula I as described in the first aspect of the invention for the preparation of a pharmaceutical composition for the treatment or prevention of a disease or condition associated with AMPK activation.
In another preferred embodiment, the disease or condition is selected from the group consisting of nonalcoholic fatty liver disease (NAF L), nonalcoholic steatohepatitis (NASH) and its associated cirrhosis, liver cancer, nonalcoholic fatty liver disease (NAF L D), obesity, diabetes, hypertriglyceridemia, hypercholesterolemia, atherosclerosis, cardiovascular disease, metabolic disease.
It is to be understood that within the scope of the present invention, the above-described features of the present invention and those specifically described below (e.g., in the examples) may be combined with each other to form new or preferred embodiments. Not to be reiterated herein, but to the extent of space.
Drawings
FIG. 1 shows the concentration-time profile of the metabolically released active molecule AMPS in the liver in vivo after gavage administration of 20. mu. mol/kg of a liver-targeted AMPS prodrug in rats.
FIG. 2 shows the concentration-time profile of AMPS, the metabolically released active molecule in vivo, in the liver, following gavage administration to rats of 20. mu. mol/kg of a prodrug of the CS0002 series.
Figure 3 shows the effect of compounds on AMPK phosphorylation levels in mouse primary hepatocytes.
Detailed Description
Unless explicitly indicated otherwise, the terms used according to the invention and herein have the following meanings:
as used herein, the term "C1-C6 alkyl" refers to a straight or branched chain alkyl group having 1 to 6 carbon atoms, such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, and the like, or the like. Preferably, the alkyl group is a straight or branched saturated chain having 1 to 3 carbon atoms, such as methyl, ethyl, n-propyl or isopropyl.
As used herein, the term "C1-C18 alkyl" refers to a straight or branched chain alkyl group having 1 to 18 carbon atoms, such as methyl, ethyl, propyl, isopropyl, butyl, isobutyl, sec-butyl, tert-butyl, and the like, or the like.
As used herein, the term "C3-C8 cycloalkyl" refers to cyclic alkyl groups having 1 to 8 carbon atoms, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl.
The term "C1-C6 alkoxy" as used herein refers to a C1-C6 alkyl group, as defined above, which is attached to the remainder of the molecule through an oxygen atom. Preferably, the C1-C6 alkoxy group may include methoxy, ethoxy, and isopropoxy.
The term "C1-C6 alkylamino" as used herein refers to a C1-C6 alkyl group, as defined above, which is attached to the rest of the molecule through a nitrogen atom. Preferably, the alkylamino group may include dimethylamino group and diethylamino group.
As used herein, the term "C1-C6 carboxy" refers to a substituent such as a "straight or branched alkyl-carboxy" structure having 1-5 carbon atoms, which is attached to the remainder of the molecule through an alkyl carbon atom. Such as a formate group, an acetate group, a propionate group, a butyrate group, or the like.
As used herein, the term "C1-C6 ester group" refers to a substituent such as a "straight or branched alkyl-ester group having 1-5 carbon atoms" structure that is attached to the remainder of the molecule through an alkyl carbon atom; wherein the alkyl part of the ester group is C1-C6 alkyl. Such as a methyl formate, ethyl formate, methyl acetate, or the like.
As used herein, the term "C2-C6 alkanoyl" refers to a substituent such as a "straight or branched alkyl-carbonyl having 1-5 carbon atoms" structure which is attached to the remainder of the molecule through a carbonyl group. Such as acetyl, propionyl, butyryl, or the like.
As used herein, the term "C2-C18 alkanoyl" refers to a substituent such as a "straight or branched chain alkyl-carbonyl group having 1-17 carbon atoms" structure, such as acetyl, propionyl, butyryl, or the like.
As used herein, the term "C2-C6 alkanoylamino" refers to a substituent as "having the structure C2-C6 alkanoyl-amino" which is attached to the remainder of the molecule through a nitrogen atom; such as acetamido, propionamido, butyramido, or the like.
As used herein, the term "C2-C18 alkoxycarbonyl" refers to, for example, a straight or branched alkyl-oxy-carbonyl group having 1 to 17 carbon atoms, which is attached to the remainder of the molecule through a carbonyl group.
As used herein, the term "C2-C18 alkylaminocarbonyl" refers to, for example, a straight or branched alkyl-nitrogen-carbonyl group having 1-17 carbon atoms, which is attached to the remainder of the molecule through a carbonyl group.
The term "halogen" refers to F, Cl, Br and I.
The term "haloalkyl" refers to a C1-C3 alkyl group substituted with a halogen. Preferably, haloalkyl is trifluoromethyl, difluoromethyl, trifluoromethoxy. Here, "C1-C3 alkyl" means a straight or branched chain alkyl group having 1 to 3 carbon atoms, such as methyl, ethyl, n-propyl, isopropyl.
The term "aryl" refers to a C6-C18 aromatic radical, such as phenyl or naphthyl, an aryl radical that is unsubstituted, substituted with one or more (e.g., 2,3,4, or 5) atoms or groups selected from: halogen, nitro, hydroxyl, amino, cyano, haloalkyl, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C3-C8 cycloalkyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted C1-C6 alkylamino, substituted or unsubstituted C1-C6 carboxyl, substituted or unsubstituted C1-C6 ester, substituted or unsubstituted C2-C6 alkanoyl, and substituted or unsubstituted C2-C6 alkylamide.
The term "heteroaryl" refers to a 5-12 membered aromatic group containing one or more heteroatoms selected from nitrogen, oxygen and sulfur. Heteroaryl groups may include pyridine, pyrazine, pyrimidine, thiophene, furan, isoxazole, isothiazole, pyrazole, imidazole. Such groups may be unsubstituted, heteroaryl substituted with one or more (e.g. 2,3,4 or 5) atoms or groups selected from: halogen, nitro, hydroxyl, amino, cyano, haloalkyl, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C3-C8 cycloalkyl, substituted or unsubstituted C1-C6 alkoxy, substituted or unsubstituted C1-C6 alkylamino, substituted or unsubstituted C1-C6 carboxyl, substituted or unsubstituted C1-C6 ester, substituted or unsubstituted C2-C6 alkanoyl, and substituted or unsubstituted C2-C6 alkylamide.
The term "heterocycle" or "heterocyclyl" refers to a 5-12 membered non-aromatic group (including saturated, partially saturated, or unsaturated groups) containing one or more heteroatoms selected from nitrogen, oxygen, and sulfur, having a single ring or fused rings (including bridged ring systems and spiro ring systems, where one or more rings may be cycloalkyl, aryl, or heteroaryl in fused ring systems, hi one embodiment, the nitrogen and/or sulfur atoms of the heterocyclic group are optionally oxidized to provide N-oxide, sulfinyl, and sulfonyl moieties examples of "heterocyclyl" and its condensed analogs include pyrrolidinyl, piperidinyl, piperazinyl, imidazolidinyl, 2, 3-dihydrofuran (2,3-b) pyridinyl, benzoxazinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, indolinyl, and the like, the term also includes non-aromatic partially unsaturated monocyclic rings, such as 2-or 4-pyridones or N-substituted- (1H, 3H) -pyrimidine-2, 4-diones (N-substituted uracils) attached via the nitrogen atom.
As used herein, the terms "comprising," "including," or "including" mean that the various ingredients may be used together in a mixture or composition of the invention. Thus, the terms "consisting essentially of and" consisting of are encompassed by the term "comprising.
In the present invention, the term "pharmaceutically acceptable" ingredient refers to a substance that is suitable for use in humans and/or animals without undue adverse side effects (such as toxicity, irritation, and allergic response), i.e., at a reasonable benefit/risk ratio.
In the present invention, the term "effective amount" refers to an amount of a therapeutic agent that treats, alleviates, or prevents a target disease or condition, or an amount that exhibits a detectable therapeutic or prophylactic effect. The precise effective amount for a subject will depend upon the size and health of the subject, the nature and extent of the disorder, and the therapeutic agent and/or combination of therapeutic agents selected for administration. Therefore, it is not useful to specify an exact effective amount in advance. However, for a given condition, the effective amount can be determined by routine experimentation and can be determined by a clinician.
As used herein, the term "pharmaceutically acceptable salt" refers to a salt of a compound of the present invention with an acid or base that is suitable for use as a pharmaceutical. Pharmaceutically acceptable salts include inorganic and organic salts. One preferred class of salts is that formed by reacting a compound of the present invention with an acid. Suitable acids for forming the salts include, but are not limited to: inorganic acids such as hydrochloric acid, hydrobromic acid, hydrofluoric acid, sulfuric acid, nitric acid, phosphoric acid, etc., organic acids such as formic acid, acetic acid, propionic acid, oxalic acid, malonic acid, succinic acid, fumaric acid, maleic acid, lactic acid, malic acid, tartaric acid, citric acid, picric acid, methanesulfonic acid, phenylmethanesulfonic acid, benzenesulfonic acid, etc.; and acidic amino acids such as aspartic acid and glutamic acid.
Some of the compounds of the present invention may be crystallized or recrystallized using water or various organic solvents, in which case various solvates may be formed. Solvates of the invention include stoichiometric solvates such as hydrates and the like, as well as compounds containing variable amounts of water formed when prepared by the low pressure sublimation drying method.
The term "prodrug" as used herein refers to any compound that produces a "drug" substance (biologically active compound) when administered to a biological system as a result of one or more spontaneous chemical reactions, one or more enzymatic chemical reactions, and/or one or more metabolic chemical reactions. It also includes biodegradable polymer derivatives of the compounds of the invention, for example as described by int.j.pharm.115,61-67 (1995).
The invention also comprisesAll suitable isotopic variations of the compounds of the present invention. Isotopic variations of the compounds of the present invention are defined as those wherein at least one atom is replaced by an atom having the same atomic number but an atomic mass different from the atomic mass usually found in nature. Examples of isotopes that can be incorporated into compounds of the invention include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine, such as2H、3H、11C、13C、14C、15N、17O、18O、35S、18F and36and (4) Cl. Some isotopic variations of the present invention, for example, in which a radioactive isotope is incorporated (e.g.3H or14C) Are used in drug and/or substrate tissue distribution studies. Tritiated, i.e.,3h, and carbon-14, i.e.,14c, isotopes are particularly preferred because of their ease of preparation and detection. In addition, the compounds are prepared with isotopes such as deuterium (i.e.,2H) may provide some therapeutic advantages resulting from increased metabolic stability, e.g., increased in vivo half-life or reduced dosage requirements and may therefore be preferred in some circumstances. Isotopic variations of the compounds of the present invention can generally be prepared by conventional procedures, for example by employing appropriate reagents for the appropriate isotopic variation, by any of the procedures exemplified or otherwise described in the experimental section below.
The invention relates to a preparation method of a formula (I):
the compounds of the present invention may be prepared by a number of methods well known to those skilled in the art, including but not limited to those described below, or by modifying these methods using standard techniques known to those skilled in the art of organic synthesis. All methods disclosed in connection with the present invention are performed on any scale, including milligram, gram, grams (multigram), kilogram (multikilogram) or commercial industrial scale. In the following equations and below, unless otherwise indicated, R1To R4As defined in the first aspect. These methods form further aspects of the invention.
Throughout the specification, the general formula is represented by roman numerals (I), (II), (III), (IV), and the like. A subset of these formulae is defined as (Ia), (Ib), (Ic), etc. …, (IVa), (IVb), (IVc), etc.; or … (I-a), (I-b), (I-c), etc., (IV-a), (IV-b), (IV-c), etc.
The general preparation of the compounds of the invention is shown below:
Figure BDA0001943047350000071
the following description will be made of the production method by taking the case where X is O in the general formula (I):
reaction formula one
Figure BDA0001943047350000081
The compounds of formula (I') can be prepared by: according to reaction scheme one, triethylamine trihydrofluoride salt is reacted with general formula (Ia/b). Typical reaction conditions include reaction of triethylamine trihydrofluoride salt and the compound of formula (Ia/Ib) in dry tetrahydrofuran at room temperature for about 12 hours. After the reaction is finished, the reaction solution is directly subjected to column chromatography (mobile phase is water and acetonitrile) by a C18 column through a rapid column chromatography instrument, and the separated fraction is subjected to freeze drying by a freeze dryer to obtain the compound of the general formula (I').
Reaction formula II
Figure BDA0001943047350000082
The compounds of general formula (Ib) can be prepared by: according to the reaction formula two, R2The substituent and the general formula (Ia) are subjected to condensation reaction to obtain the general formula (Ib).
Reaction formula III
Figure BDA0001943047350000083
The compounds of general formula (Ia) can be prepared by: according to the third reaction formula, trichloro-sulfur phosphorus, triethylamine and the general formula (Ic) are reacted. Then, the reaction mixture was reacted with the diol (Id), or the reaction mixture was concentrated and subjected to silica gel column chromatography (mobile phase was petroleum ether and ethyl acetate) and then reacted with the diol (Id). Typical reaction conditions include reaction of trichlorothiophosphoryl, triethylamine and the compound of formula (Ic) in an inert solvent such as DCM at room temperature for about 0.5 hour, followed by addition of the diol (ld) and reaction at room temperature for about 12 hours. After the reaction solution is concentrated, column chromatography purification is carried out by a C18 column through a flash column chromatography (mobile phase is water and acetonitrile), and the fraction obtained by separation is freeze-dried by a freeze dryer to obtain the compound of the general formula (Ia).
Reaction type IV
Figure BDA0001943047350000084
Compounds of general formula (Ic) can be prepared by: according to the fourth reaction formula, tert-butyldimethylsilyl chloride (TBSCl), the general formula (Ie) and imidazole are reacted. Typical reaction conditions include reacting t-butyldimethylsilyl chloride, formula (Ie) and imidazole in a solvent such as DMF at room temperature for about 12 hours. After the reaction, the reaction mixture was concentrated and subjected to silica gel column chromatography (mobile phase was petroleum ether and ethyl acetate), and the fraction obtained by separation was concentrated to obtain the general formula (If).
ii removal of the 5' -terminal tert-butyldimethylsilane protecting group from the general formula (If) under the action of trifluoroacetic acid and water gives the general formula Ic). Typical reaction conditions include reaction of formula (If), trifluoroacetic acid and water in a solvent such as THF at 0 deg.C for about 5 hours. Adjusting the pH value of the reaction solution to be neutral, filtering and collecting a filter cake to obtain the compound of the general formula (Ic).
Reaction formula five
Figure BDA0001943047350000091
Compounds of general formula (Ih) may be prepared by: general formula (Ig), thionyl chloride and catalytic amount of DMF are reacted. Typical reaction conditions include reacting a compound of formula (Ig), thionyl chloride and a catalytic amount of DMF in a solvent such as DCM at room temperature for about 2 hours. Then the reaction solution is concentrated, after adding ethanol and reacting for about one hour at room temperature, after the reaction solution is concentrated, the reaction solution is subjected to silica gel column chromatography (the mobile phase is petroleum ether and ethyl acetate), and the fraction obtained by separation is concentrated to obtain the general formula (Ih).
ii, reacting the intermediate of the general formula (Ih) with an ester under the action of lithium hexamethyldisilazide at a low temperature (about-60 ℃). Typical reaction conditions include: the reaction of general formula (Ih), lithium hexamethyldisilazide and ethyl acetate was carried out at a temperature of about-60 c for about 20min at this low temperature. The reaction mixture was adjusted to pH to neutrality with acetic acid, concentrated, purified by silica gel column chromatography (mobile phase was petroleum ether and ethyl acetate), and the fraction obtained by separation was concentrated to obtain the general formula (Ii).
iii in a protic solvent (e.g., MeOH) with a reducing agent (e.g., NaBH)4) With general formula (Ii) to obtain the compound of general formula (Id).
Reaction type six
In a particular embodiment, the chiral 1, 3-propanediol derivative (ll/lm) of the compound of formula (ld) is prepared by:
the method comprises the following steps:
Figure BDA0001943047350000092
hexamethyldisilazane and trimethylsilyl trifluoromethanesulfonate in an inert solvent such as DCM in the presence of an acid such as TMSOTf, washing with water, and concentrating to obtain a crude product, which is reacted with (2S,5R) -2-isopropyl-5-methylcyclohexanone under the action of an acid such as TMSOTf to obtain intermediate compounds of general formulae (Ij) and (Ik).
ii, respectively reacting concentrated hydrochloric acid with general formulas (Ij) and (Ik) at room temperature, concentrating the reaction solution, performing silica gel column chromatography (mobile phase is petroleum ether and ethyl acetate), and concentrating the separated fraction to obtain general formula (Il/Im).
The second method comprises the following steps:
Figure BDA0001943047350000101
i.e. at 40 ℃ to 80 ℃, in an inert solvent (such as DMF), formic acid, triethylamine, (S, S) -N- (p-toluenesulfonyl) -1, 2-diphenylethylenediamine (dichloro) (p-cymene) ruthenium (II) and intermediate Ij are reacted to obtain the intermediate of the general formula (Ip).
ii by reduction with a reducing agent (e.g., NaBH) in a protic solvent (e.g., MeOH)4) With general formula (Ip) to obtain a compound of general formula (In).
The preparation of the general formulae (VI) and (VII) can be referred to from reaction scheme one to reaction scheme five, where it is necessary to replace the achiral diol of reaction scheme five with the chiral diol obtained in reaction scheme six. It is understood that within the scope of the present invention, the above-mentioned technical features of the present invention and the technical features specifically described below (e.g. in the examples) may be combined with each other to constitute new or preferred embodiments.
Pharmaceutical compositions and methods of administration
The compound of the present invention and its various crystal forms, pharmaceutically acceptable inorganic or organic salts, hydrates or solvates, and pharmaceutical compositions containing the compound as a main active ingredient are useful for the treatment, prevention and alleviation of diseases caused by hepatitis B virus, according to the prior art, the compound of the present invention can be used for the treatment of nonalcoholic fatty liver disease (NAF L), nonalcoholic steatohepatitis (NASH) and associated cirrhosis, nonalcoholic fatty liver disease (NAF L D) including liver cancer, and also for metabolic diseases such as obesity, diabetes, hypertriglyceridemia, hypercholesterolemia, atherosclerosis, cardiovascular diseases, etc.
The pharmaceutical composition of the present invention comprises the compound of the present invention or a pharmacologically acceptable salt thereof in a safe and effective amount range and a pharmacologically acceptable excipient or carrier. Wherein "safe and effective amount" means: the amount of the compound is sufficient to significantly improve the condition without causing serious side effects. Typically, the pharmaceutical composition contains 0.1-1000mg of a compound of the invention per dose, more preferably, 0.5-500mg of a compound of the invention per dose. Preferably, said "dose" is a capsule or tablet.
"pharmaceutically acceptable carrier" refers to: one or more compatible solid or liquid fillers or gel substances which are suitable for human use and must be of sufficient purity and sufficiently low toxicity. By "compatible" is meant herein that the components of the composition are capable of intermixing with and with the compounds of the present invention without significantly diminishing the efficacy of the compounds. Examples of pharmaceutically acceptable carrier moieties are cellulose and its derivatives (e.g., sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, etc.), gelatin, talc, solid lubricants (e.g., stearic acid, magnesium stearate), calcium sulfate, vegetable oils (e.g., soybean oil, sesame oil, peanut oil, olive oil, etc.), polyols (e.g., propylene glycol, glycerin, mannitol, sorbitol, etc.), emulsifiers (e.g., tween), wetting agents (e.g., sodium lauryl sulfate), colorants, flavors, stabilizers, antioxidants, preservatives, pyrogen-free water, etc.
The mode of administration of the compounds or pharmaceutical compositions of the present invention is not particularly limited, and representative modes of administration include (but are not limited to): oral, rectal, parenteral (intravenous, intramuscular, or subcutaneous), and topical administration. A particularly preferred mode of administration is oral.
Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules. In these solid dosage forms, the active compound is mixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with the following ingredients: (a) fillers or extenders, for example, starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) binders, for example, hydroxymethylcellulose, alginates, gelatin, polyvinylpyrrolidone, sucrose and acacia; (c) humectants, for example, glycerol; (d) disintegrating agents, for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) slow solvents, such as paraffin; (f) absorption accelerators, e.g., quaternary ammonium compounds; (g) wetting agents, such as cetyl alcohol and glycerol monostearate; (h) adsorbents, for example, kaolin; and (i) lubricants, for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, or mixtures thereof. In capsules, tablets and pills, the dosage forms may also comprise buffering agents.
Solid dosage forms such as tablets, dragees, capsules, pills, and granules can be prepared using coatings and shells such as enteric coatings and other materials well known in the art. They may contain opacifying agents and the release of the active compound or compounds in such compositions may be delayed in release in a certain part of the digestive tract. Examples of embedding components which can be used are polymeric substances and wax-like substances. If desired, the active compound may also be in microencapsulated form with one or more of the above-mentioned excipients.
Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly employed in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1, 3-butylene glycol, dimethylformamide and oils, in particular, cottonseed, groundnut, corn germ, olive, castor and sesame oils or mixtures of such materials and the like.
In addition to these inert diluents, the compositions can also contain adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar, or mixtures of these substances, and the like.
Compositions for parenteral injection may comprise physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols and suitable mixtures thereof.
Dosage forms for topical administration of the compounds of the present invention include ointments, powders, patches, sprays, and inhalants. The active ingredient is mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants which may be required if necessary.
The compounds of the present invention may be administered alone or in combination with other pharmaceutically acceptable compounds.
When the pharmaceutical composition is used, a safe and effective amount of the compound of the present invention is suitable for mammals (such as human beings) to be treated, wherein the administration dose is a pharmaceutically-considered effective administration dose, and for a human body with a weight of 60kg, the daily administration dose is usually 0.2 to 1000mg, preferably 0.5 to 500 mg. Of course, the particular dosage will depend upon such factors as the route of administration, the health of the patient, and the like, and is within the skill of the skilled practitioner.
The main advantages of the invention include:
the invention prepares thio nucleoside monophosphate into a cyclic phosphorothioate prodrug with liver tissue specificity, drug molecules have better stability to gastrointestinal tracts and blood plasma and are not easy to be hydrolyzed by ester hydrolase in vivo, the drug is oxidized by CYP3A in cytochrome P450 isozyme family in tissue cells after entering the liver, the 4-position ring opening of the phosphorothioate ring of the drug molecule generates an intermediate with negative charge of the monophosphate, and the intermediate is catalyzed and hydrolyzed by phosphatase and subjected to β -elimination reaction to release the thio nucleoside monophosphate of parent drug, the thio nucleoside monophosphate is in a protonation state in cells and is not easy to be retained in the cells through cell membranes, so that the drug concentration in the liver cells is higher than that of normal tissues, the aryl ketene serving as a byproduct after the prodrug is oxidized by CYP3A can be rapidly combined with glutathione which is rich in oxidation resistance and free radicals in the liver cells to be eliminated, and no side effect report about the thio nucleoside monophosphate is found at present.
(1) The prodrug of the invention, namely the compound of the general formula (I), is metabolized in liver cells after being orally taken to generate a parent drug; the drug molecule has high negative charge, and is not easy to be discharged out of liver, so that the concentration in liver is higher, and the liver targeting effect is achieved.
(2) The invention belongs to liver targeting drugs, and has good tissue distribution of active drug molecules, more drug molecules exist in liver cells, and the drug effect is greatly improved. Since the drug molecules are mainly concentrated in liver cells and are metabolized extrahepatic to active molecules in small amounts, side effects on the kidney, heart, etc. are greatly reduced.
The invention will be further illustrated with reference to the following specific examples. It should be understood that these examples are for illustrative purposes only and are not intended to limit the scope of the present invention. The experimental procedures, in which specific conditions are not noted in the following examples, are generally carried out under conventional conditions or conditions recommended by the manufacturers. Unless otherwise indicated, percentages and parts are by weight.
Intermediate preparation example 1: ((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methanol
The reaction steps are as follows:
step 1: preparation of 9- ((2R,3R,4R,5R) -3, 4-bis ((tert-butyldimethylsilyl) oxy) -5- (((tert-butyldimethylsilyl) oxy) methyl) tetrahydrofuran-2-yl) -9H-purin-6-amino
(2R,3R,4S,5R) -2- (6-amino-9H-purin-9-yl) -5- (hydroxymethyl) tetrahydrofuran-3, 4-diol (16.0g,60.0mmol) was dissolved in N, N-dimethylformamide (120m L), the temperature was reduced to 0 ℃ and imidazole (20.4g,300.0mmol) and tert-butyldimethylchlorosilane (36.2g,240.0mmol) were added, respectively, the reaction solution was stirred at the reaction solution temperature, after completion of the reaction, the reaction solution was distilled under reduced pressure to remove the organic solvent, the crude product was dissolved in ethyl acetate (100m L) and saturated ammonium chloride (100m L) was washed twice, and the organic phase was dried over anhydrous sodium sulfate, concentrated, and subjected to silica gel column chromatography (petroleum ether: ethyl acetate ═ 2:1) to give 34g of a white solid, yield 93%. MS (ES)+)m/z 610(M+H+).
Step 2: preparation of ((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-di ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methanol
9- ((2R,3R,4R,5R) -3, 4-bis ((tert-butyldimethylsilyl) oxy) -5- (((tert-butyldimethylsilyl) oxy) methyl) tetrahydrofuran-2-yl) -9H-purin-6-amino (3.9g,6.4mmol) was dissolved in tetrahydrofuran (30m L), trifluoroacetic acid/water (12m L/12 m L) was added at 0 ℃ the reaction mixture was reacted at 0 ℃ for 16 hours, after completion of the reaction mixture was poured into a saturated sodium bicarbonate solution to adjust pH 8, ethyl acetate was extracted (150m L x 2), ethyl acetate phases were combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate and dried, the crude product was slurried with ethyl acetate (15m L) to give 2.2g of a white solid in 73% yield.1H NMR(400MHz,DMSO-d6):8.41(s,1H),8.14(s,1H),7.40(s,2H),5.91(d,J=4.0Hz,1H),5.76-5.74(m,1H),4.91-4.90(m,1H),4.31(d,J=4.0Hz,1H),4.00(s,1H),3.77-3.76(m,1H),3.58-3.35(m,1H),0.93(s,9H),0.70(s,9H),0.13-0.12(m,6H),-0.14(s,3H),-0.46(s,3H).
Intermediate preparation example 2:1- (3-chloro-2-fluorophenyl) propane-1, 3-diol
The reaction steps are as follows:
step 1: preparation of ethyl 3-chloro-2-fluorobenzoate
3-chloro-2-fluorobenzoic acid (4.9g,28.5mmol) and DMF (10 drops) were dissolved in dichloromethane (40m L), oxalyl chloride (4.3g,34.2mmol) was added at 0 deg.C, the reaction was stirred for 1 hour and then concentrated to dryness, the crude product was added with ethanol (40m L) at 0 deg.C, stirred for 1 hour at room temperature, then concentrated and washed with ethyl acetate (50m L), sodium bicarbonate and saturated sodium chloride, dried over anhydrous sodium sulfate and concentrated to give 5.8g of an anhydrous oil with 100% yield.1H NMR(400MHz,CDCl3):7.84-7.81(m,1H),7.59-7.56(m,1H),7.17-7.13(m,1H),4.41(q,J=4.0Hz,H),1.40(t,J=4.0Hz,3H).
Step 2: preparation of ethyl 3- (3-chloro-2-fluorobenzene) -3-oxopropanoate
Dissolving ethyl 3-chloro-2-fluorobenzoate (5.8g,28.5mmol) and ethyl acetate (17.6g,199.5mmol) in tetrahydrofuran (60m L), cooling to-60 deg.C, adding L iHMDS (85.5m L, 85.5mmol), stirring the reaction solution at-60 deg.C for 30min, adding acetic acid (9m L) to quench, adding water (50m L), extracting the reaction solution with ethyl acetate (40m L x 2), mixing the organic phase, drying with anhydrous sodium sulfateConcentration gave 8.2g of crude brown oil.1H NMR(400MHz,CDCl3):12.68(s,0.4H),7.84-7.81(m,1H),7.64-7.61(m,1H),7.23-7.20(m,1H),5.83(s,0.4H),4.29-4.22(m,2H),4.05-3.99(m,2H),1.36-1.29(m,3H).
And step 3: preparation of 1- (3-chloro-2-fluorophenyl) propane-1, 3-diol
Ethyl 3- (3-chloro-2-fluorobenzene) -3-oxopropanoate (8.2g,28.5mmol) was dissolved in methanol (70m L), NaBH4(5.4g,14.2mmol) was added at 0 ℃, the reaction stirred at room temperature for 3 hours, ethyl acetate (20m L) was added, the reaction was concentrated, ethyl acetate/water (50m L/50 m L) was added, the ethyl acetate phase was concentrated, and the crude product was purified by silica gel column chromatography (dichloromethane: methanol 10:1) to give 3.2g of a colorless oil, 55% yield in steps 2 and 3.1H NMR(400MHz,CDCl3):7.51-7.41(m,1H),7.36-7.32(m,1H),7.16-7.12(m,1H),5.34-5.30(m,1H),3.96-3.91(m,2H),3.27(d,J=4.0Hz,1H),2.19-2.16(m,1H),2.07-2.00(m,2H).
Intermediate preparation example 3: 1-phenyl propane-1, 3-diol
Referring to the preparation method of intermediate preparation example 2, except that benzoic acid was used as the starting material, yield was 74%,1HNMR(400MHz,CDCl3):7.45-7.35(m,5H),5.17-5.12(m,1H),4.19-4.05(m,2H),2.23-2.16(m,1H),2.07-2.00(m,1H).
intermediate preparation example 4:1- (pyridin-4-yl) propane-1, 3-diol
Referring to the preparation method of intermediate preparation example 2, except that isonicotinic acid is used as the starting material, the yield is 50%,1HNMR(400MHz,CDCl3):8.56-8.53(m,2H),7.55-7.54(m,2H),5.13-5.11(m,1H),4.11-4.10(m,1H),3.97-3.95(m,2H),2.38(brs,1H),2.01-1.99(m,2H).
intermediate preparation example 5:1- (3-chlorophenyl) propane-1, 3-diol
Referring to the preparation method of intermediate preparation example 2, except that 3-chlorobenzoic acid is used as the starting material, the yield is 25%,1H NMR(400MHz,CDCl3)7.39(t,J=1.8Hz,1H),7.34-7.25(m,3H),4.95(dd,J=8.4,4.2Hz,1H),3.87(t,J=5.4Hz,2H),1.99-1.88(m,2H).
intermediate preparation example 6: 1- (3- (trifluoromethyl) phenyl) propane-1, 3-diol
Reference was made to the preparation of intermediate preparation example 2 except that 3- (trifluoromethyl) benzoic acid was used as starting material in 48% yield and MS (ES)+)m/z 221(M+H+).
Intermediate preparation example 7: 1- (3-methylphenyl) propane-1, 3-diol
Referring to the preparation method of intermediate preparation example 2, except that 3-methylbenzoic acid was used as a starting material,1H NMR(400MHz,CDCl3)7.27-7.09(m,4H),4.95-4.92(m,1H),3.88-3.85(m,2H),2.36(s,3H),2.04-1.91(m,2H).
intermediate preparation example 8: 1- (3-methoxyphenyl) propane-1, 3-diol
Referring to the preparation method of intermediate preparation example 2, except that 3-methoxy chlorobenzoic acid is used as a starting material,1HNMR(400MHz,CDCl3)7.29-7.25(m,1H),6.95-6.93(m,2H),6.83-6.81(m,1H),4.97-4.94(m,1H),3.89-3.86(m,2H),3.82(s,3H),1.99-1.92(m,2H).
intermediate preparation example 91- (3-fluorophenyl) propane-1, 3-diol
Reference was made to the preparation of intermediate preparation example 2 except that 3-fluorobenzoic acid was used as the starting material in 50% yield and MS (ES)+)m/z 171(M+H+).
Intermediate preparation example 10: 1- (5-chloro-2-fluorophenyl) propane-1, 3-diol
Referring to the preparation method of intermediate preparation example 2, except that 5-chloro-2-fluorobenzoic acid was used as the starting material, the yield was 55%,1H NMR(400MHz,CDCl3):7.51(t,J=8.0Hz,1H),7.18(d,J=8.0Hz,1H),7.18-7.05(m,1H),5.27-5.24(m,1H),3.93-3.89(m,2H),3.37(brs,1H),2.38(brs,1H),2.00-1.98(m,2H).
intermediate preparation example 11: 1- (4-chloro-2-fluorophenyl) propane-1, 3-diol
Referring to the preparation method of intermediate preparation example 2, except that 4-chloro-2-fluorobenzoic acid was used as the starting material, the yield was 64%,1H NMR(400MHz,CDCl3):7.51(t,J=8.0Hz,1H),7.18(d,J=8.0Hz,1H),7.18-7.05(m,1H),5.27-5.24(m,1H),3.93-3.89(m,2H),3.37(brs,1H),2.38(brs,1H),2.00-1.98(m,2H).
intermediate preparation example 12 1- (2, 5-difluorophenyl) propane-1, 3-diol
Referring to the preparation method of intermediate preparation example 2, except using 2, 5-difluorobenzoic acid as the starting material, the yield was 50%,1H NMR(400MHz,CDCl3)7.38-7.27(m,1H),7.10-6.88(m,2H),5.48-5.13(m,1H),4.26-4.00(m,1H),3.90-3.94(m,1H),3.34(d,J=3.6Hz,1H),2.15-1.81(m,2H).
intermediate preparation example 13: 1- (2, 5-dichlorophenyl) propane-1, 3-diol
Reference was made to the preparation of intermediate preparation example 2 except that 2, 5-dichlorobenzoic acid was used as starting material in 54% yield MS (ES)+)m/z 221(M+H+).
Intermediate preparation example 14 1- (2-chloro-4-fluorophenyl) propane-1, 3-diol
Referring to the preparation method of intermediate preparation example 2, except using 2-chloro-4-fluorobenzoic acid as the starting material, the yield was 64%,1H NMR(400MHz,CDCl3)7.70-7.45(m,1H),7.09-7.07(m,2H),5.49-5.28(m,1H),4.27-4.01(m,2H),2.34-2.31(m,1H),1.97-1.71(m,1H).
intermediate preparation example 15 1- (2,4, 5-trifluorophenyl) propane-1, 3-diol
Referring to the preparation method of intermediate preparation example 2, except using 2,4, 5-trifluorobenzoic acid as the starting material, the yield was 71%,1H NMR(400MHz,MeOD)7.41-7.36(m,1H),7.14-7.07(m,1H),5.09-5.06(m,1H),3.68-3.62(m,2H),1.90-1.85(m,2H).
intermediate preparation example 16: 1- (2-chloro-4, 5-difluorophenyl) propane-1, 3-diol
Referring to the preparation method of intermediate preparation example 2, except using 2-chloro-4, 5-difluorobenzoic acid as the starting material, the yield was 75%,1HNMR:(400MHz,CDCl3)7.50(dd,J=11.2,8.6Hz,1H),7.17(dd,J=9.6,7.0Hz,1H),5.26(d,J=7.4Hz,1H),3.96-3.91(m,2H),2.02(dd,J=5.2,2.8Hz,1H),1.89-1.78(m,1H).MS(ES+)m/z 245.1(M+Na+)
intermediate preparation example 17: 1- (5-chloro-2, 4-difluorophenyl) propane-1, 3-diol
Preparation of reference intermediate preparation example 2Except that 5-chloro-2, 4-difluorobenzoic acid is used as the initial raw material, the yield is 74 percent,1HNMR:(400MHz,CDCl3)7.62(t,J=7.8Hz,1H),6.87(dd,J=9.6,9.0Hz,1H),5.22(dd,J=8.0,3.4Hz,1H),3.93-3.90(m,2H),1.97-1.93(m,2H).MS(ES+)m/z245.1(M+Na+)
intermediate preparation example 18: 1- (2, 4-dichloro-5-fluorophenyl) propane-1, 3-diol
Referring to the preparation method of intermediate preparation example 2, except using 2, 4-dichloro-5-fluorobenzoic acid as a starting material, the yield was 91%,1H-NMR:(400MHz CDCl3)7.48(d,J=9.8Hz,1H),7.38(d,J=6.6Hz,1H),5.25(dd,J=8.8,2.2Hz,1H),3.99-3.85(m,2H),2.04-2.00(m,1H),1.86-1.79(m,1H).
intermediate preparation example 19: 1- (2,3,4, 5-tetrafluorophenyl) propane-1, 3-diol
Referring to the preparation method of intermediate preparation example 2, except that 2,3,4, 5-tetrafluorobenzoic acid was used as a starting material, yield was 40%,1HNMR:(400MHz,CDCl3)7.26-7.18(m,1H),5.26(dd,J=8.4,2.5Hz,1H),4.11-3.88(m,2H),1.97-1.90(m,2H).
intermediate preparation example 20: 1- (3-chloro-2, 4, 5-trifluorophenyl) propane-1, 3-diol
Referring to the preparation method of intermediate preparation example 2, except that 3-chloro-2, 4, 5-trifluorobenzoic acid was used as the starting material, the yield was 78%,1HNMR:(400MHz,CDCl3)7.38(ddd,J=10.4,8.4,6.6Hz,1H),5.26(dd,J=8.2,2.4Hz,1H),4.01-3.83(m,2H),1.98-1.93(m,2H).
intermediate preparation example 21: 1- (pentafluorophenyl) propane-1, 3-diol
Referring to the preparation method of intermediate preparation example 2, except using pentafluorobenzoic acid as the starting material, the yield was 88%,1HNMR:(400MHz,CD3OD)5.25(dd,J=8.6,5.8Hz,1H),3.75-3.66(m,1H),3.66-3.58(m,1H),2.302.16(m,1H),2.04-1.90(m,1H).
intermediate preparation example 22: chiral 1- (4-chloro-2-fluorophenyl) propane-1, 3-diol
Step 1: preparation of (2R,6S,7S,10R) -2- (4-chloro-2-fluorophenyl) -7-isopropyl-10-methyl-1, 5-dioxaspiro [5.5] undecane
Dissolving 1- (4-chloro-2-fluorophenyl) propane-1, 3-diol (3.0g,14.7mmol) and hexamethyldisilazane (6.73m L, 32.3mmol) in tetrahydrofuran (12m L), adding multiple drops of trimethylsilyl trifluoromethanesulfonate at room temperature, stirring at room temperature for 2 hours, adding ethyl acetate (50m L), washing the organic phase with saturated sodium chloride, drying over anhydrous sodium sulfate, concentrating, dissolving the crude product in dry dichloromethane (30m L), adding (2S,5R) -2-isopropyl-5-methylcyclohexanone (2.72g,17.64mmol), cooling to-40 deg.C, adding trimethylsilyl trifluoromethanesulfonate (391mg,1.764mmol), stirring the reaction solution overnight at-40 deg.C, after the reaction is complete, adding pyridine (2m L) to quench, adding dichloromethane (30m L) to the reaction solution, washing twice with saturated sodium bicarbonate solution (30m L), concentrating the crude product, column chromatography (petroleum ether ═ 1: 1.5 g,5 g of colorless isopropyl-5-2 m 3583), obtaining colorless oily product [ 5R-5-methyl-5-propyl ] -2-dioxanone, 1.7-5-methyl ether (1.10 g, 7-5-methyl-1-5-methyl-1-5-methyl-10-2-7-methyl-7-methyl-2]Undecane, yield 38%,1H NMR(400MHz,CDCl3)7.54(t, J ═ 8.0Hz,1H),7.20(dd, J ═ 8.0,2.0Hz,1H),7.07(dd, J ═ 10.0,2.0Hz,1H),5.41-5.37(m,1H),4.10-4.03(m,1H),3.89-3.85(m,1H),2.91-2.86(m,1H),2.63-2.59(m,1H),1.87-1.24(m,8H),1.10-0.71(m, 12H); and oil (2S,6S,7S,10R) -2- (4-chloro-2-fluorophenyl) -7-isopropyl-10-methyl-1, 5-dioxaspiro [5.5]]Undecane, yield 30%,1H NMR(400MHz,CDCl3)7.52(t,J=8.0Hz,1H),7.18(dd,J=8.0,2.0Hz,1H),7.07(dd,J=10.0,2.0Hz,1H),5.19-5.16(m,1H),4.32-4.26(m,1H),3.94-3.89(m,1H),2.92-2.88(m,1H),2.55-2.51(m,1H),1.91-1.22(m,8H),1.11-0.65(m,12H).
step 2:
preparation of (R) -1- (4-chloro-2-fluorophenyl) propane-1, 3-diol
Reacting (2R,6S,7S,10R) -2- (4-chloro-2-fluorophenyl) -7-isopropyl-10-methyl-1, 5-dioxaspiro [5.5]]Undecane (1.9g,5.59mmol) was dissolved in methanol (10m L), concentrated hydrochloric acid (1.5m L) was added at room temperature, the reaction solution was stirred overnight at room temperature, concentrated, water (30m L) was added, ethyl acetate was extracted (30m L x 2), the organic phases were combined and concentrated, the crude product was subjected to silica gel column chromatography (dichloromethane: methanol ═ 10:1) to give 0.9g of (R) -1- (4-chloro-2-fluorophenyl) propane-1, 3-diol as a colorless oil in 80% yield,1H NMR(400MHz,CDCl3):7.51(t,J=8.0Hz,1H),7.18(d,J=8.0Hz,1H),7.09-7.05(m,1H),5.27-5.26(m,1H),3.93-3.90(m,2H),3.30(brs,1H),2.28-2.26(m,1H),2.06-1.98(m,2H).
and step 3:
preparation of (S) -1- (4-chloro-2-fluorophenyl) propane-1, 3-diol
Referring to the preparation method of (R) -1- (4-chloro-2-fluorophenyl) propane-1, 3-diol in step 2, the yield is 78%,1HNMR(400MHz,CDCl3):7.51(t,J=8.0Hz,1H),7.18(d,J=8.0Hz,1H),7.09-7.05(m,1H),5.27-5.26(m,1H),3.93-3.90(m,2H),3.31(brs,1H),2.27-2.25(m,1H),2.06-1.98(m,2H).
intermediate preparation example 23 (R) -1- (3-chlorophenyl) propane-1, 3-diol
Reference intermediate preparation 22, step 1 and step 2, except that 1- (3-chloro-phenyl) propane-1, 3-diol was substituted for 1- (4-chloro-2-fluorophenyl) propane-1, 3-diol in reference intermediate preparation 22. The yield thereof is 9%,1H NMR(400MHz,CDCl3)7.38-7.07(m,4H),4.84-4.80(m,1H),4.22-4.20(m,1H),3.75-3.72(m,2H),3.54-3.53(m,1H),1.88-1.82(m,2H).
intermediate preparation example 24: (S) -1- (3-chlorophenyl) propane-1, 3-diol
Reference intermediate preparation 22, step 1 and step 3, except that 1- (3-chloro-phenyl) propane-1, 3-diol was substituted for 1- (4-chloro-2-fluorophenyl) propane-1, 3-diol in reference intermediate preparation 22. The yield is 17 percent,1H NMR(400MHz,CDCl3)7.37-7.07(m,4H),4.84-4.80(m,1H),4.23-4.20(m,1H),3.75-3.72(m,2H),3.55-3.53(m,1H),1.88-1.82(m,2H).
intermediate preparation example 25: (R) -1- (2, 5-dichlorophenyl) propane-1, 3-diol
Reference intermediate preparation 22, step 1 and step 2, except that 1- (2, 5-dichloro-phenyl) propane-1, 3-diol was substituted for 1- (4-chloro-2-fluorophenyl) propane-1, 3-diol in reference intermediate preparation 22. The yield is 12 percent,1H NMR(400MHz,CDCl3):7.66-7.65(m,1H),7.25-7.18(m,2H),5.32-5.29(m,1H),3.95-3.92(m,2H),3.65(brs,1H),2.53(brs,1H),2.09-2.03(m,1H),1.92-1.78(m,1H).
intermediate preparation 26: (S) -1- (2, 5-dichlorophenyl) propane-1, 3-diol
Reference intermediate preparation 22, step 1 and step 3, except that 1- (2, 5-dichloro-phenyl) propane-1, 3-diol was substituted for 1- (4-chloro-2-fluorophenyl) propane-1, 3-diol in reference intermediate preparation 22. The yield is 17 percent,1H NMR(400MHz,CDCl3):7.66-7.65(m,1H),7.25-7.18(m,2H),5.32-5.29(m,1H),3.95-3.92(m,2H),3.64(brs,1H),2.52(brs,1H),2.09-2.03(m,1H),1.91-1.71(m,1H).
intermediate preparation 27- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (3-chloro-2-fluorobenzene) -1,3, 2-dioxaphosphorinane 2-sulfide
((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methanol (396mg,0.8mmol) was dissolved in anhydrous pyridine (4m L) and PSCl was added at 0 deg.C3(189mg,1.1 mmol.) after reaction at 0 ℃ for 30 minutes, a solution of 1- (3-chloro-2-fluorophenyl) propane-1, 3-diol (277mg,1.4mmol) in DCM (0.5m L) is added, the reaction stirred overnight, concentrated, and the crude product is purified by C18 preparative column to give 220mg of a white solid, yield 36%. MS (ES)+)m/z 760(M+H+).
Intermediate preparation example 28: 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4-phenyl-1, 3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of intermediate preparation example 27 except that 1-phenylpropane-1, 3-diol was used as a raw material, yield was 25%,1H NMR(400MHz,CDCl3)8.37(d,J=1.4Hz,1H),8.21(d,J=2.2Hz,1H),7.39-7.36(m,5H),5.75-5.72(m,3H),4.88-4.68(m,3H),4.45-4.31(m,5H),2.49-2.27(m,1H),2.27-2.01(m,1H),0.95(d,J=2.8Hz,9H),0.80(d,J=2.4Hz,9H),0.14-0.11(m,6H),-0.02(d,J=5.0Hz,3H),-0.23(d,J=1.6Hz,3H).
intermediate preparation example 29: 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (pyridin-4-yl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of intermediate preparation example 27 except that 1- (pyridin-4-yl) propane-1, 3-diol was used as a starting material, the yield was 40%,1H NMR(400MHz,CDCl3):8.66-8.61(m,2H),8.36(s,1H),8.12(s,1H),7.30-7.24(m,2H),6.02-6.00(m,1H),5.76-5.72(m,1H),5.62(s,2H),4.91-4.90(m,1H),4.81-4.72(m,2H),4.46-4.13(m,4H),2.35-2.21(m,1H),2.10-2.07(m,1H),0.96(s,9H),0.83-0.81(s,9H),0.17-0.13(m,6H),0.02(s,3H),-0.18(s,3H).
intermediate preparation example 30: 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of intermediate preparation example 27, except that 1- (3-chlorophenyl) propane-1, 3-diol was used as a starting material, the yield was 59%,1H NMR(400MHz,CDCl3)8.37(d,J=3.6Hz,1H),8.18-7.99(m,1H),7.58-7.32(m,4H),6.13-5.98(m,1H),5.91(d,J=5.0Hz,1H),5.59(s,2H),5.18-5.08(m,1H),4.96-4.66(m,2H),4.53-4.29(m,2H),4.09(dd,J=11.8,6.8Hz,1H),3.77(dd,J=11.8,4.4Hz,1H),2.33-2.00(m,2H),0.96(d,J=2.8Hz,9H),0.86-0.80(m,9H),0.19-0.07(m,6H),-0.01(d,J=1.4Hz,3H),-0.21(d,J=7.4Hz,3H).
intermediate preparation example 31: 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (3- (trifluoromethyl) phenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of intermediate preparation example 27 except for using 1- (3- (trifluoromethyl) phenyl) propane-1, 3-diol as a raw material, yield 41%,1H NMR(400MHz,CDCl3)8.36(d,J=2.6Hz,1H),8.14(d,J=0.8Hz,1H),7.67-7.58(m,2H),7.53(dd,J=14.8,8.6Hz,2H),6.01(dd,J=5.2,3.4Hz,1H),4.94-4.67(m,3H),4.59-4.35(m,4H),4.33(s,1H),2.43-2.20(m,2H),0.95(d,J=2.6Hz,9H),0.81(d,J=4.8Hz,9H),0.20-0.05(m,6H),0.00--0.06(m,3H),-0.16--0.25(m,3H).
intermediate preparation example 32: 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (3-methylphenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of intermediate preparation example 27 except for using 1- (3-methylphenyl) propane-1, 3-diol as a starting material, yield 54%,1H NMR(400MHz,DMSO-d6)8.55-8.33(m,1H),8.25-8.04(m,1H),7.47-7.03(m,6H),5.97(d,J=6.8Hz,1H),5.66(d,J=11.2Hz,1H),5.04-4.88(m,1H),4.65-4.59(m,2H),4.54-4.37(m,3H),4.23-4.17(m,1H),2.31-2.09(m,5H),0.99-0.82(m,9H),0.78-0.60(m,9H),0.20-0.05(m,6H),-0.06--0.12(m,3H),-0.34--0.39(m,3H).
intermediate preparation 33: 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (3-trimethoxyphenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of intermediate preparation example 27 except for using 1- (3-trimethoxyphenyl) propane-1, 3-diol as a raw material, yield 47%,1H NMR(400MHz,DMSO-d6)8.48-8.39(m,1H),8.18-8.14(m,1H),7.61-7.15(m,3H),7.09-6.78(m,3H),5.98-5.96(m,1H),5.68(d,J=11.0Hz,1H),5.50-4.91(m,1H),4.72-4.00(m,6H),3.76-3.74(m,3H),2.39-2.09(m,2H),1.03-0.80(m,9H),0.80-0.51(m,9H),0.22-0.05(m,6H),-0.04--0.16(m,3H),-0.39(d,J=4.4Hz,3H).
intermediate preparation example 34: 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (3-fluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of intermediate preparation example 27 except for using 1- (3-fluorophenyl) propane-1, 3-diol as a starting material, yield 87%,1H NMR(400MHz,MeOD)8.39(d,J=1.4Hz,1H),8.24(s,1H),7.46-7.24(m,1H),7.24-6.97(m,3H),6.14-6.10(m,1H),5.76-5.67(m,1H),4.73-4.70(m,2H),4.65-4.50(m,2H),4.50-4.31(m,3H),2.35-2.16(m,1H),2.15-2.00(m,1H),0.96(d,J=1.4Hz,9H),0.82(d,J=3.2Hz,9H),0.16-0.13(m,6H),0.02-0.01(m,3H),-0.19(d,J=6.2Hz,3H).
intermediate preparation example 35: 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (5-chloro-2-fluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of intermediate preparation example 27 except for using 1- (5-chloro-2-fluorophenyl) propane-1, 3-diol as a starting material in a yield of 31%,1H NMR(400MHz,CDCl3)8.37(s,1H),8.16-8.15(m,1H),7.54-7.41(m,2H),7.05-7.01(m,1H),6.01-5.91(m,2H),5.71(s,2H),4.91-4.78(m,3H),4.56-4.33(m,4H),2.32-2.22(m,1H),2.13-2.03(m,1H),0.98-0.79(m,18H),0.17-0.13(m,6H),0.02-0.01(m,3H),-0.07(s,3H).
intermediate preparation 36: 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (4-chloro-2-fluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of intermediate preparation example 27 except for using 1- (4-chloro-2-fluorophenyl) propane-1, 3-diol as a starting material in a yield of 45%,1H NMR(400MHz,MeOD):8.36(s,1H),8.17(s,1H),7.41-7.28(m,1H),7.15-7.13(m,2H),6.02-5.93(m,2H),5.64(brs,2H),4.89-4.80(m,3H),4.44-4.32(m,4H),2.31-2.11(m,2H),0.98-0.78(m,18H),0.16-0.12(m,6H),-0.01--0.19(m,3H),-0.21(s,3H).
intermediate preparation example 37: 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (2, 5-difluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of intermediate preparation example 27 except that 1- (2, 5-difluorophenyl) propane-1, 3-diol was used as a raw material, the yield was 68%,1H NMR(400MHz,CDCl3)8.37(s,1H),8.16(d,J=3.6Hz,1H),7.11-6.96(m,3H),6.04-5.93(m,2H),5.56(s,2H),4.94-4.87(m,1H),4.87-4.70(m,2H),4.56-4.41(m,3H),4.34(s,1H),2.32-1.99(m,2H),0.96(t,J=2.0Hz,9H),0.83(d,J=3.6Hz,9H),0.19-0.12(m,6H),0.09(s,3H),-0.16(d,J=4.8Hz,3H).
intermediate preparation example 38: 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (2, 5-dichlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of intermediate preparation example 27 except that 1- (2, 5-dichlorophenyl) propane-1, 3-diol was used as a raw material, the yield was 31%,1H NMR(400MHz,CDCl3)8.37(s,1H),8.15(s,1H),7.51(dd,J=23.8,2.5Hz,1H),7.36–7.28(m,3H),6.02(t,J=5.0Hz,1H),5.67(s,2H),5.00–4.72(m,3H),4.62–4.30(m,5H),2.19–2.01(m,2H),0.97(s,9H),0.83(d,J=9.5Hz,9H),0.21–0.11(m,6H),0.02(s,3H),-0.16(d,J=23.6Hz,3H).
intermediate preparation example 39: 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (2-chloro-4-fluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of intermediate preparation example 27 except for using 1- (2-chloro-4-fluorophenyl) propane-1, 3-diol as a starting material in a yield of 33%,1H NMR(400MHz,CDCl3)8.36(d,J=2.4Hz,1H),8.14(d,J=1.8Hz,1H),7.49-7.45(m,1H),7.19-7.12(m,1H),7.12-6.94(m,1H),6.02(t,J=5.6Hz,1H),5.70(s,2H),4.96-4.89(m,1H),4.89-4.69(m,2H),4.59-4.30(m,5H),2.28-1.91(m,2H),0.96(d,J=1.6Hz,9H),0.82(d,J=3.2Hz,9H),0.16-0.14(m,6H),0.01(s,3H),-0.19(s,3H).
intermediate preparation example 40: 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (2,4, 5-trifluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Reference was made to the preparation of intermediate preparation 27 except that 1- (2,4, 5-trifluorophenyl) propane-1, 3-diol was used as a starting material in 50% yield, MS (ES)+)m/z 762(M+H+)。
Intermediate preparation example 41: 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (2-chloro-4, 5-difluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of intermediate preparation example 27 except for using 1- (2-chloro-4, 5-trifluorophenyl) propane-1, 3-diol as a raw material, yield 50%,1H NMR(400MHz,CDCl3)8.37(d,J=2.0Hz,1H),8.15(d,J=4.4Hz,1H),7.56-7.33(m,1H),7.28-7.22(m,1H),6.06-5.91(m,2H),5.65(s,2H),4.97-4.71(m,3H),4.63-4.25(m,4H),2.34-2.07(m,2H),1.08-0.59(m,18H),0.20-0.06(m,6H),0.02(d,J=1.5Hz,3H),-0.08--0.29(m,3H).
intermediate preparation example 42: 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (5-chloro-2, 4-difluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of intermediate preparation example 27 except for using 1- (5-chloro-2, 4-difluorophenyl) propane-1, 3-diol as a raw material, yield 56%,1H NMR(400MHz,CDCl3)8.37(s,1H),8.14(s,1H),7.59-7.47(m,1H),6.99-6.92(m,1H),6.03–6.02(m,1H),5.96-5.92(m,1H),5.72(s,2H),4.90-4.71(m,3H),4.55-4.33(m,4H),2.33-2.19(m,2H),0.98-0.78(m,18H),0.16-0.12(m,6H),0.02(s,3H),-0.08--0.39(m,3H).
intermediate preparation 43: 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (2, 4-dichloro-5-fluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of intermediate preparation example 27 except for using 1- (2, 4-dichloro-5-fluorophenylphenyl) propane-1, 3-diol as a starting material in a yield of 50%,1H NMR(400MHz,DMSO-d6)8.49-8.30(m,1H),8.24-8.11(m,1H),8.05-7.87(m,1H),7.73-7.53(m,1H),7.42-7.38(m,2H),6.03-5.78(m,2H),4.98-4.86(m,1H),4.72-4.33(m,5H),4.17-4.14(m,1H),2.39-2.25(m,1H),2.18-2.08(m,1H),1.01-0.79(m,9H),0.79-0.53(m,9H),0.20-0.04(m,6H),-0.05--0.18(m,3H),-0.33--0.53(m,3H).
intermediate preparation example 44: 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (2,3,4, 5-tetrafluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of intermediate preparation example 27 except for using 1- (2,3,4, 5-tetrafluorophenyl) propane-1, 3-diol as a starting material, yield 51%,1H NMR(400MHz,CDCl3)8.36(d,J=1.8Hz,1H),8.24-7.92(m,1H),7.27-7.06(m,1H),6.05-5.86(m,2H),5.66-5.59(m,2H),4.97-4.69(m,3H),4.58-4.32(m,4H),2.25-2.18(m,1H),2.11-2.08(m,1H),0.98-0.78(m,18H),0.23-0.07(m,6H),0.03-0.01(m,3H),-0.08--0.29(m,3H).
intermediate preparation example 45: 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (3-chloro-2, 4, 5-trifluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of intermediate preparation example 27 except for using 1- (3-chloro-2, 4, 5-trifluorophenyl) propane-1, 3-diol as a raw material, yield 86%,1H NMR(400MHz,CDCl3)8.37-8.36(m,1H),8.14-8.13(m,1H),7.45-7.20(m,1H),6.01(t,J=4.0Hz,1H),5.95(t,J=12.0Hz,1H),5.61(brs,2H),4.90(q,J=9.4Hz,1H),4.84-4.72(m,2H),4.55-4.30(m,4H),2.28-2.15(m,1H),2.15-2.05(m,1H),0.96-0.92(m,9H),0.88-0.80(m,9H),0.18-0.10(m,6H),0.02(s,3H),-0.02(s,3H),-0.12--0.16(m,3H).
intermediate preparation example 46: 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (pentafluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of intermediate preparation example 27 except for using 1- (pentafluorophenyl) propane-1, 3-diol as a starting material, yield 55%,1H NMR(400MHz,CDCl3)8.37(d,J=1.4Hz,1H),8.11(d,J=5.0Hz,1H),6.12-5.97(m,2H),5.63(s,2H),4.88-4.65(m,3H),4.53-4.30(m,4H),2.79-2.71(m,1H),1.96-1.90(m,1H),1.03-0.72(m,18H),0.21-0.06(m,6H),-0.01--0.04(m,3H),-0.19--0.21(m,3H).
intermediate preparation 47: n- (9- (3, 4-bis ((tert-butyldimethylsilyl) oxy) -5- (((4- (3-chlorophenyl) -2-thio-1, 3, 2-dioxaphosphorinan-2-yl) oxy) methyl) tetrahydrofuran-2-yl) -9H-purin-6-yl) acetamide
2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide (1.1g,1.6mmol) is dissolved in anhydrousAcetyl chloride (187mg,2.39mmol) was added to pyridine (5m L) in water at 0 deg.C, the reaction stirred at room temperature for 1 hour, concentrated, and crude silica gel column chromatography (petroleum ether: ethyl acetate 1:1) gave 380mg of a white solid in 30% yield,1H NMR(400MHz,DMSO-d6)10.73-10.74(m,1H),8.69-8.70(m,1H),8.64-8.65(m,1H),7.38-7.50(m,4H),6.08-6.09(m,1H),5.72-5.74(m,1H),4.94-4.98(m,1H),4.40-4.69(m,6H),2.24-2.28(m,3H),1.96–2.04(m,1H),0.92-0.93(m,9H),0.83-0.88(m,1H),0.68-0.69(m,9H),0.09-0.15(m,6H),(-0.11)-(-0.08)(m,3H),(-0.40)-(-0.38)(m,3H).
intermediate preparation example 48: n- (9- (3, 4-bis ((tert-butyldimethylsilyl) oxy) -5- (((4- (3-chlorophenyl) -2-sulfanyl-1, 3, 2-dioxaphosphorinan-2-yl) oxy) methyl) tetrahydrofuran-2-yl) -9H-purin-6-yl) butanamide
Reference is made to the procedure for the preparation of intermediate preparation 47 except that 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide is reacted with butyryl chloride in 33% yield,1H NMR(CDCl3,400MHz)8.34(s,1H),8.23(s,1H),7.18-7.42(m,4H),6.07-6.10(m,1H),5.65-5.75(m,1H),4.68-4.85(m,4H),4.33-4.53(m,3H),2.85-2.88(m,2H),2.25-2.41(m,2H),1.57-1.963(m,2H),1.04-1.08(m,3H),0.95(s,9H),0.80(s,9H),0.11-0.14(m,6H),(-0.2)-0.02(m,3H),(-0.22)–(-0.20)(m,3H).
intermediate preparation example 49: (4R) -2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (4-chloro-2-fluorobenzene) -1,3, 2-dioxaphosphorinane 2-sulfide
Reference is made to the procedure for the preparation of intermediate preparation 27 except that (R) -1- (4-chloro-2-fluorophenyl) propane-1, 3-diol is used as starting material in 39% yield. MS (ES)+)m/z 760(M+H+).
Intermediate preparation example 50: (4S) -2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (4-chloro-2-fluorobenzene) -1,3, 2-dioxaphosphorinane 2-sulfide
Reference was made to the preparation of intermediate preparation 27 except that (S) -1- (4-chloro-2-fluorophenyl) propane-1, 3-diol was used as a starting material in 21.8% yield,1H NMR(400MHz,MeOD)8.37(s,1H),8.13(s,1H),7.41(t,J=8.0Hz,1H),7.27-7.12(m,2H),6.03-5.94(m,2H),5.67(m,2H),4.91-4.72(m,3H),4.52-4.33(m,4H),2.33-2.24(m,1H),2.10-2.07(m,1H),0.98-0.95(m,9H),0.83-0.78(m,9H),0.17-0.12(m,6H),-0.02(s,3H),-0.20(s,3H).
intermediate preparation example 51: (4R) -2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (3-chlorobenzene) -1,3, 2-dioxaphosphorinane 2-sulfide
Reference was made to the preparation of intermediate preparation 27 except that (R) -1- (3-chloro-phenyl) propane-1, 3-diol was used as a starting material in a yield of 42%,1H NMR(400MHz,CDCl3)8.39-8.38(m,1H),8.18(s,1H),7.41-7.29(m,3H),7.23-7.20(m,1H),6.02(d,J=5.0Hz,1H),5.84(s,2H),5.70(d,J=9.4Hz,1H),4.93-4.66(m,3H),4.55-4.24(m,4H),2.43-2.21(m,1H),2.05-2.01(m,1H),0.96-0.94(m,9H),0.83-0.80(m,9H),0.21-0.10(m,6H),0.02(s,1H),-0.02(s,2H),-0.20(s,3H).
intermediate preparation example 52: (4S) -2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (3-chlorobenzene) -1,3, 2-dioxaphosphorinane 2-sulfide
Reference was made to the preparation of intermediate preparation 27 except that (S) -1- (3-chloro-phenyl) propane-1, 3-diol was used as a starting material in 52.7% yield,1H NMR(400MHz,CDCl3)8.40(s,1H),8.25(s,1H),7.41-7.29(m,3H),7.28-7.26(m,1H),6.20(s,2H),6.03-6.01(m,1H),5.73-5.69(m,1H),4.86-4.31(m,7H),2.41-2.34(m,1H),2.05-2.01(m,1H),0.99-0.75(m,18H),0.20-0.07(m,6H),-0.01(s,3H),-0.20(d,J=16.0Hz,3H).
intermediate preparation 53: (4R) -2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (2, 5-dichlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Preparation of reference intermediate preparation 27Except that the raw material of (R) -1- (2, 5-dichloro-phenyl) propane-1, 3-diol is used, the yield is 42 percent,1H NMR(400MHz,DMSO-d6)8.43-8.41(m,1H),8.16-8.15(m,1H),7.58-7.29(m,5H),6.04-5.76(m,2H),5.50(d,J=4.8Hz,1H),4.97-4.94(m,1H),4.66-4.45(m,4H),4.35-4.18(m,1H),2.33-2.16(m,2H),0.94-0.91(m,6H),0.70(s,9H),0.19-0.08(m,6H),-0.09(d,J=5.4Hz,3H),-0.39(d,J=11.0Hz,3H).
intermediate preparation 54: (4S) -2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (2, 5-dichlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Reference was made to the preparation of intermediate preparation 27 except that the starting material was (S) -1- (2, 5-dichloro-phenyl) propane-1, 3-diol, in a yield of 41%,1H NMR(400MHz,DMSO-d6)8.39-8.34(m,1H),8.16-8.02(m,1H),7.63-7.31(m,5H),5.98-5.89(m,2H),5.01-4.98(m,1H),4.73-4.38(m,5H),4.40-4.18(m,1H),2.38-2.14(m,2H),0.94-0.90(m,9H),0.71-0.68(m,9H),0.22-0.03(m,6H),-0.12(s,3H),-0.43(s,3H).
intermediate preparation example 55: 9- ((2R,3R,4R,5R) -5- (aminomethyl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) -9H-purin-6-amino
The reaction steps are as follows:
step 1: preparation of N- (9- ((2R,3R,4R,5R) -3, 4-bis ((tert-butyldimethylsilyl) oxy) -5- (((tert-butyldimethylsilyl) oxy) methyl) tetrahydrofuran-2-yl) -9H-purin-6-yl) benzamide
N- (9- ((2R,3R,4S,5R) -3, 4-dihydroxy-5- (hydroxymethyl) tetrahydrofuran-2-yl) -9H-purin-6-yl) benzamide (1.0g,2.7mmol) was dissolved in N, N-dimethylformamide (20m L), the temperature was reduced to 0 ℃, imidazole (1.8g,26.9mmol) and tert-butyldimethylchlorosilane (3.2g,21.5mmol) were added, respectively, the reaction solution was stirred overnight at room temperature, after completion of the reaction, the reaction solution was distilled under reduced pressure to remove the organic solvent, the crude product was dissolved in ethyl acetate (50m L), saturated ammonium chloride (20m L) was washed twice, the organic phase was dried over anhydrous sodium sulfate, concentrated, and column chromatography was performed on silica gel (petroleum ether: ethyl acetate ═ 3:1), yielding 1.6g of a white solid in 84%.1H NMR(400MHz,CDCl3)8.83(s,1H),8.45(s,1H),8.08-8.06(m,2H),7.61(d,J=7.4Hz,1H),7.55-7.52(m,2H),6.13(d,J=5.0Hz,1H),4.67(t,J=4.6Hz,1H),4.31(t,J=3.8Hz,1H),4.17(d,J=2.8Hz,1H),4.07-4.00(m,1H),3.81(dd,J=11.4,2.6Hz,1H),0.96(s,9H),0.95(s,9H),0.80(s,9H),0.15(s,3H),0.14(s,3H),0.11(s,3H),0.10(s,3H),-0.02(s,3H),-0.23(s,3H)。
Step 2: preparation of N- (9- ((2R,3R,4R,5R) -3, 4-bis ((tert-butyldimethylsilyl) oxy) -5- (hydroxymethyl) tetrahydrofuran-2-yl) -9H-purin-6-yl) benzamide
N- (9- ((2R,3R,4R,5R) -3, 4-bis ((tert-butyldimethylsilyl) oxy) -5- (((tert-butyldimethylsilyl) oxy) methyl) tetrahydrofuran-2-yl) -9H-purin-6-yl) benzamide (400.0mg,0.6mmol) was dissolved in tetrahydrofuran (3m L), trifluoroacetic acid/water (1.2m L/1.2 m L) was added at 0 ℃, the reaction mixture was reacted at 0 ℃ for 6 hours, after completion of the reaction, the reaction mixture was poured into a saturated sodium bicarbonate solution to adjust pH to 8, ethyl acetate was extracted (50m L × 2), the ethyl acetate phases were combined, washed with saturated sodium chloride, dried over anhydrous sodium sulfate and slurried, the crude product was slurried with ethyl acetate (15m L) to obtain 300.0mg of a white solid with a yield of 88%,1H NMR(400MHz,CDCl3)8.83(s,1H),8.08(s,1H),8.06-8.01(m,2H),7.66-7.59(m,1H),7.54(t,J=7.6Hz,2H),5.87(d,J=7.8Hz,1H),5.04(dd,J=7.8,4.4Hz,1H),4.36(d,J=4.4Hz,1H),4.20(s,1H),3.98(dd,J=13.2,1.8Hz,1H),3.74(dd,J=13.2,1.8Hz,1H),0.94(s,9H),0.75(s,9H),0.14(s,3H),0.13(s,3H),-0.12(s,3H),-0.63(s,3H)。
and step 3: preparation of methyl ((2R,3R,4R,5R) -5- (6-benzoyl-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methanesulfonate
Methanesulfonyl chloride (152.0mg,1.3mmol) was added to a solution of N- (9- ((2R,3R,4R,5R) -3, 4-bis ((t-butyldimethylsilyl) oxy) -5- (hydroxymethyl) tetrahydrofuran-2-yl) -9H-purin-6-yl) benzamide (200.0mg,0.3mmol) and triethylamine (202.0mg,2.0mmol) in dichloromethane (5m L) under ice-cooling, after stirring at room temperature for 12 hours, water (20m L) was added for quenching, dichloromethane was extracted (20m L x 2), the organic phase was combined and concentrated, and the crude product was chromatographed on a silica gel column (petroleum ether: ethyl acetate ═ 2:1) to give 140mgA pale yellow oil, yield 87%,1H NMR(400MHz,CDCl3)8.81(s,1H),8.23(s,1H),8.05-8.03(m,2H),7.66-7.58(m,1H),7.53(t,J=7.4Hz,2H),6.00(d,J=4.8Hz,1H),4.97(t,J=4.2Hz,1H),4.61(dd,J=11.0,3.8Hz,1H),4.49(dd,J=11.2,4.4Hz,1H),4.41-4.32(m,2H),3.03(s,3H),0.94(s,9H),0.82(s,9H),0.14(s,3H),0.13(s,3H),-0.00(s,3H),-0.21(s,3H).MS(ES+)m/z 678(M+H+)。
and 4, step 4: preparation of N- (9- ((2R,3R,4R,5R) -5- (azidomethyl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) -9H-purin-6-yl) benzamide
Sodium azide (56.0mg,0.9mmol) was added to a solution of methyl ((2R,3R,4R,5R) -5- (6-benzoyl-9H-purin-9-yl) -3, 4-bis ((t-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methanesulfonate (140.0mg,0.2mmol) in DMF (2m L) at room temperature, the reaction mixture was stirred at room temperature for 12 hours, water (30m L) was added and quenched, ethyl acetate (30.0m L x 2) was extracted, the organic phase was combined and concentrated, the crude product was subjected to silica gel column chromatography (petroleum ether: ethyl acetate 2:1) to give 80mg of a pale yellow oil with a yield of 54%,1H NMR(400MHz,CDCl3)8.82(s,1H),8.39(s,1H),8.07-8.04(m,2H),7.62(d,J=7.4Hz,1H),7.54(t,J=7.4Hz,2H),6.01(d,J=3.8Hz,1H),4.83(t,J=3.8Hz,1H),4.33-4.21(m,2H),3.80(d,J=4.0Hz,1H),3.76-3.67(m,1H),0.94(s,9H),0.84(s,9H),0.13(s,3H),0.12(s,3H),0.03(s,3H),-0.10(s,3H)。
and 5: preparation of N- (9- ((2R,3R,4R,5R) -5- (aminomethyl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) -9H-purin-6-yl) benzamide
N- (9- ((2R,3R,4R,5R) -5- (azidomethyl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) -9H-purin-6-yl) benzamide (80.0mg,0.1mmol.) was dissolved in methanol (3.0m L), Pd (OH) was added2C (30.0mg), stirring at room temperature under hydrogen atmosphere (1atm) for 2 hours, filtering the reaction solution, concentrating the filtrate to obtain 50mg of a pale yellow solid with a yield of 65%,1H NMR(400MHz,CDCl3)8.83(s,1H),8.21(s,1H),8.04(d,J=7.2Hz,2H),7.65-7.58(m,1H),7.53(t,J=7.4Hz,2H),5.92(d,J=6.2Hz,1H),4.95(dd,J=6.0,4.6Hz,1H),4.40(d,J=3.2Hz,1H),4.22(s,1H),3.20-3.19(m,2H),0.94(s,9H),0.78(s,9H),0.11(s,6H),-0.03(s,3H),-0.41(s,3H).MS(ES+)m/z 599(M+H+)。
step 6: preparation of 9- ((2R,3R,4R,5R) -5- (aminomethyl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) -9H-purin-6-amino
N- (9- ((2R,3R,4R,5R) -5- (aminomethyl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) -9H-purin-6-yl) benzamide (1.0g,1.7mmol) was dissolved in NH3In MeOH (30.0m L, 7.0N), stirred at room temperature for 12 hours, concentrated, and the crude product was subjected to silica gel column chromatography (dichloromethane: methanol 10:1) to give 500mg of a pale yellow solid with a yield of 60%,1H NMR(400MHz,CD3OD)8.30(s,1H),8.20(s,1H),5.96(d,J=7.0Hz,1H),5.06(dd,J=6.8,4.6Hz,1H),4.32(d,J=4.6Hz,1H),4.15-4.11(m,1H),3.15-3.11(m,1H),3.00(dd,J=13.4,3.6Hz,1H),0.98(s,9H),0.76(s,9H),0.17(d,J=3.6Hz,6H),-0.05(s,3H),-0.45(s,3H).MS(ES+)m/z 495(M+H+)。
intermediate preparation 56: 2- (((((2R, 3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methyl) amino) -4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
The reaction steps are as follows:
step 1: preparation of 2-chloro-4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Triethylamine (19.5mg,0.2mmol) and 1- (3-chlorophenyl) propyl-1, 3-diol (30.0mg,0.16mmol) were dissolved in tetrahydrofuran (3m L), trichlorosulfur phosphate (27.0mg,0.2mmol) was added under ice bath, the reaction was stirred at room temperature for 12 hours and concentrated, the crude product was chromatographed on silica gel column (petroleum ether: ethyl acetate ═ 3:1) to give 20.0mg of oil in 47% yield,1HNMR(400MHz,CDCl3)7.41-7.33(m,1H),7.29-7.26(m,3H),5.00-4.87(m,1H),4.62-4.59(m,1H),4.45-4.41(m,1H),2.18-2.16(m,1H),1.29-1.24(m,1H)。
step 2: preparation of 2- (((((2R, 3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methyl) amino) -4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
9- ((2R,3R,4R,5R) -5- (aminomethyl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) -9H-purin-6-amino (216.0mg,0.4mmol) and triethylamine (64.0mg,0.6mmol) were dissolved in dichloromethane (6m L), 2-chloro-4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide (120mg,0.42mmol) was added under ice-cooling, the reaction solution was stirred at room temperature for 12 hours, concentrated, the crude product was chromatographed on silica gel column (dichloromethane: methanol ═ 10:1) to give 160mg of a pale yellow solid in 51% yield,1H NMR(400MHz,MeOD)8.41-8.17(m,2H),7.58-7.20(m,4H),5.92-5.86(m,1H),5.81-5.54(m,1H),5.16-4.99(m,1H),4.78-4.74(m,1H),4.60-4.26(m,3H),3.67-3.42(m,1H),3.40-3.32(m,1H),2.78-2.43(m,1H),2.19-2.08(m,1H),1.04-0.84(m,9H),0.78-0.58(m,9H),0.24-0.10(m,4H),0.07-0.03(m,1H),-0.02(d,J=1.6Hz,1H),-0.07--0.25(m,3H),-0.51--0.75(m,3H).MS(ES+)m/z 741(M+H+)。
example 1: 2- (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (3-chloro-2-fluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (3-chloro-2-fluorobenzene) -1,3, 2-dioxaphosphorinane 2-sulfide (200mg,0.3mmol) is dissolved in THF (3m L), triethylamine trihydrofluoride (838mg,5.2mmol) is added at room temperature, the reaction is stirred at room temperature overnight after completion of the reaction, concentrated and the crude product is passed through a C18 preparative column (CH 18)3CN/H2O2-50%) to yield 75mg of a white solid in 54% yield.1H NMR(400MHz,MeOD):8.36-8.35(m,1H),8.22(s,1H),7.38-7.35(m,2H),7.19-7.04(m,1H),6.12-6.11(m,1H),5.96-5.91(m,1H),4.89-4.33(m,7H),2.37-2.27(m,1H),2.14-2.09(m,1H).MS(ES+)m/z 532(M+H+).
Example 2: 2- (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4-phenyl-1, 3, 2-dioxaphosphorinane 2-sulfide
Preparation method of reference example 1Except that 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4-phenyl-1, 3, 2-dioxaphosphorinane 2-sulfide is used as a starting material in 87% yield,1H NMR(400MHz,DMSO-d6)8.32(d,J=3.2Hz,1H),8.15(s,1H),7.43-7.31(m,5H),5.97-5.95(m,1H),5.68-5.62(m,2H),5.45(t,J=4.8Hz,1H),4.65-4.41(m,7H),2.29-2.17(m,1H),2.16-2.06(m,1H).MS(ES+)m/z480(M+H+).
example 3: 2- (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (pyridin-4-yl) -1,3, 2-dioxaphosphorinane 2-sulfide
With reference to the preparation process of example 1, except that 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (pyridin-4-yl) -1,3, 2-dioxaphosphorinane 2-sulfide is used as a starting material, yield 67%,1H NMR(400MHz,DMSO-d6):8.58-8.54(m,2H),8.33(d,J=4.0Hz,1H),8.15(s,1H),7.38-7.25(m,4H),5.97(d,J=4.0Hz,1H),5.74-5.65(m,2H),5.64-5.46(m,1H),4.68-4.31(m,6H),4.27-4.16(m,1H),2.21-2.08(m,2H).MS(ES+)m/z 481(M+H+).
example 4: 2- (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of example 1, except that 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide is used as a starting material, yield is 59%,1H NMR(400MHz,MeOD)8.37(d,J=2.0Hz,1H),8.22(s,1H),7.42(d,J=2.2Hz,1H),7.39-7.23(m,3H),6.11(t,J=4.6Hz,1H),4.80-4.67(m,2H),4.67-4.51(m,2H),4.51-4.30(m,4H),2.38-2.15(m,1H),2.11(dt,J=14.6,2.4Hz,1H).MS(ES+)m/z 514(M+H+).
example 5: 2- (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (3- (trifluoromethyl) phenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Reference is made to the preparation of example 1, except that 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (3- (trifluoromethyl) phenyl) -1,3, 2-dioxaphosphorinane 2-sulfide is used as starting material in 31% yield,1H NMR(400MHz,MeOD)8.37(d,J=1.2Hz,1H),8.22(s,1H),7.75-7.48(m,4H),6.16-6.08(m,1H),5.36(t,J=4.6Hz,1H),4.73-4.45(m,5H),3.28-3.19(m,2H),2.05(d,J=5.4Hz,2H).MS(ES+)m/z 545(M+H+)。
example 6: 2- (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (3-methylphenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Reference is made to the preparation of example 1, except that 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (3-methylphenyl) -1,3, 2-dioxaphosphorinane 2-sulfide is used as starting material in a yield of 72%,1H NMR(400MHz,DMSO-d6)8.56-8.29(m,1H),8.25-8.07(m,1H),7.52–6.79(m,6H),6.03–5.88(m,1H),5.73–5.57(m,1H),5.53–5.39(m,1H),5.39–5.21(m,1H),4.68–3.87(m,7H),2.39–1.98(m,4H),1.89–1.68(m,1H).MS(ES+)m/z 494(M+H+)。
example 7:2- (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (3-methoxyphenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Reference was made to the preparation of example 1 except that 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (3-methoxyphenyl) -1,3, 2-dioxaphosphorinane 2-sulfide was used as a starting material in 62% yield,1H NMR(400MHz,DMSO-d6)8.41–8.24(m,1H),8.24–8.10(m,1H),7.41–7.23(m,2H),7.03–6.87(m,2H),5.95(d,J=5.3Hz,1H),5.64(d,J=11.0Hz,1H),4.70–4.12(m,7H),3.74(d,J=8.0Hz,3H),2.30–2.02(m,2H).MS(ES+)m/z 510(M+H+)。
example 8: 2- (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (3-fluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of example 1, except that 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (3-fluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide is used as a raw material, yield is 41%,1H NMR(400MHz,MeOD)8.39(d,J=1.4Hz,1H),8.24(s,1H),7.44-7.29(m,1H),7.24-6.97(m,3H),6.14-6.11(m,1H),5.76-5.67(m,1H),4.74-4.31(m,7H),2.36-2.10(m,2H).MS(ES+)m/z 498(M+H+)。
example 9: 2- (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (5-chloro-2-fluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of example 1, except that 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (5-chloro-2-fluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide is used as a raw material, yield 51%,1H NMR(400MHz,DMSO-d6):8.31-8.29(m,1H),8.15-8.09(m,1H),7.57-7.49(m,2H),7.36-7.30(m,3H),5.97-5.95(m,1H),5.85-5.82(m,1H),5.63-5.62(m,1H),5.46-5.44(m,1H),4.62-4.14(m,7H),2.37-2.27(m,1H),2.10-2.09(m,1H).MS(ES+)m/z 532(M+H+)。
example 10: 2- (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (4-chloro-2-fluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of example 1, except that 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (4-chloro-2-fluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide is used as a raw material, yield 53%,1H NMR(400MHz,MeOD):8.36-8.34(m,1H),8.22(s,1H),7.46-7.40(m,1H),7.27-7.07(m,2H),6.12-6.10(m,1H),5.90-5.89(m,1H),4.89-4.33(m,7H),2.38-2.26(m,1H),2.11-2.06(m,1H).MS(ES+)m/z 532(M+H+)。
example 11: 2- (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (2, 5-difluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of example 1, except that 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((t-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (2, 5-difluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide is used as a raw material, yield 41%,1H NMR(400MHz,MeOD)8.36(d,J=4.4Hz,1H),8.23(d,J=1.6Hz,1H),7.27-7.02(m,3H),6.12(t,J=5.0Hz,1H),5.94-5.84(m,1H),4.68-4.54(m,3H),4.53-4.30(m,4H),2.40-2.19(m,1H),2.19-1.98(m,1H).MS(ES+)m/z 516(M+H+).
example 12: 2- (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (2, 5-dichlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Reference is made to the preparation of example 1, except that 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (2, 5-dichlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide is used as starting material in 29% yield,1H NMR(400MHz,MeOD)8.37(d,J=11.0Hz,1H),8.23(d,J=1.6Hz,1H),7.51-7.26(m,3H),6.13(dd,J=10.8,5.0Hz,1H),4.77-4.57(m,4H),4.57-4.34(m,4H),2.26-2.02(m,2H).MS(ES+)m/z 548(M+H+).
example 13: 2- (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (2-chloro-4-fluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Reference was made to the preparation of example 1 except that 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (2-chloro-4-fluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide was used as a starting material, and the yield was 56%,1H NMR(400MHz,CDCl3)8.30-8.27(m,1H),8.09-8.07(m,1H),7.37-6.84(m,3H),6.12-5.74(m,3H),4.92-4.13(m,6H),2.17-1.98(m,2H).MS(ES+)m/z 532(M+H+).
Example 14: 2- (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (2,4, 5-trifluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of example 1, except that 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((t-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (2,4, 5-trifluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide is used as a raw material, yield is 65%,1H NMR(400MHz,DMSO-d6)8.29(d,J=6.4Hz,1H),8.14-8.05(m,1H),7.77-7.49(m,2H),7.30-7.25(m,2H),5.95(d,J=5.4Hz,1H),5.88-5.74(m,1H),5.63(d,J=4.8Hz,1H),5.44(t,J=5.0Hz,1H),4.69-4.09(m,6H),2.47-2.30(m,1H),2.06-2.04(m,1H).MS(ES+)m/z 534(M+H+).
example 15: 2- (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (2-chloro-4, 5-difluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of example 1, except that 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (2-chloro-4, 5-difluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide is used as a raw material, yield is 65%,1H NMR(400MHz,DMSO-d6)8.35-8.25(m,1H),8.14-8.05(m,1H),7.92-7.73(m,1H),7.72-7.57(m,1H),7.31-7.23(m,2H),5.96(d,J=5.4Hz,1H),5.88-5.76(m,1H),5.65-5.63(m,1H),5.48-5.45(m,1H),4.65-4.15(m,7H),2.38-2.21(m,1H),2.20-2.03(m,1H).MS(ES+)m/z 550(M+H+).
example 16: 2- (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (5-chloro-2, 4-difluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Reference example 1 was made to the preparation method except that 2- (((1)2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (5-chloro-2, 4-difluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide as starting material in 61% yield,1H NMR(400MHz,DMSO-d6)8.39-8.23(m,1H),8.21-8.03(m,1H),7.84-7.71(m,1H),7.66-7.59(m,1H),7.38-7.21(m,2H),5.96-5.95(d,J=5.2Hz,1H),5.84-5.81(m,1H),5.64-5.62(m,1H),5.45(t,J=5.2Hz,1H),4.63-4.20(m,6H),4.16-4.13(m,1H),2.47-2.32(m,1H),2.11-2.05(m,1H).MS(ES+)m/z550(M+H+).
example 17: 2- (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (2, 4-dichloro-5-fluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of example 1, except that 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((t-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (2, 4-dichloro-5-fluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide is used as a starting material,1H NMR(400MHz,DMSO-d6)8.33-8.21(m,1H),8.14(s,1H),8.01–7.86(m,1H),7.60(dd,J=16.2,9.8Hz,1H),7.30(d,J=8.4Hz,2H),5.96(d,J=5.4Hz,1H),5.82(s,1H),5.75–5.40(m,2H),4.73–4.09(m,7H),2.31–2.10(m,2H).MS(ES+)m/z 566(M+H+).
example 18: 2- (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (2,3,4, 5-tetrafluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Reference was made to the preparation of example 1 except that 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (2,3,4, 5-tetrafluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide was used as a starting material in 49% yield,1H NMR(400MHz,DMSO-d6)8.28(d,J=11.0Hz,1H),8.14(s,1H),7.58-7.48(m,1H),7.29-7.22(m,2H),5.95(d,J=5.4Hz,1H),5.85(t,J=9.4Hz,1H),5.67-5.62(m,1H),5.46-5.38(m,1H),4.64-4.36(m,4H),4.33-3.93(m,3H),2.45-2.32(m,1H),2.13-2.08(m,1H).MS(ES+)m/z 552(M+H+).
example 19: 2- (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (3-chloro-2, 4, 5-trifluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of example 1, except that 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((t-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (3-chloro-2, 4, 5-trifluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide is used as a raw material, yield 86%,1H NMR(400MHz,DMSO-d6)8.30-8.22(m,1H),8.14(s,1H),7.77-7.60(m,1H),7.30-7.22(m,2H),5.95(d,J=4.0Hz,1H),5.85(t,J=10.0Hz,1H),5.66-5.60(m,1H),5.46-5.40(m,1H),4.67-4.56(m,2H),4.55-4.35(m,3H),4.27-4.22(m,1H),4.16-4.10(m,1H),2.45-2.32(m,1H),2.15-2.05(m,1H).MS(ES+)m/z 568(M+H+).
example 20: 2- (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (pentafluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Referring to the preparation method of example 1, except that 2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((t-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (pentafluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide is used as a starting material in a yield of 59%,1H NMR(400MHz,DMSO-d6)8.34-8.25(m,1H),8.14-8.07(m,1H),7.29-7.22(m,2H),5.99-5.92(m,2H),5.63-5.61(m,1H),5.44-5.43(m,1H),4.66-4.09(m,7H),2.66-2.55(m,1H),2.22-2.10(m,1H).MS(ES+)m/z 570(M+H+).
example 21: n- (9- ((2R,3R,4S,5R) -5- (((4- (3-chlorophenyl) -2-thio-1, 3, 2-dioxaphosphorinan-2-yl) oxy) methyl) -3, 4-dihydroxytetrahydrofuran-2-yl) -9H-purin-6-yl) acetamide
Referring to the preparation method of example 1, except that N- (9- (3, 4-bis ((t-butyldimethylsilyl) oxy) -5- (((4- (3-chlorophenyl) -2-thio-1, 3, 2-dioxaphosphorinan-2-yl) oxy) methyl) tetrahydrofuran-2-yl) -9H-purin-6-yl) acetamide is used as a starting material, yield 11%,1H NMR(400MHz,MeOD)8.64(s,1H),8.58(d,J=4.8Hz,1H),7.23-7.39(m,4H),6.20(t,J=5.2Hz,1H),5.61-5.70(m,1H),4.75-4.80(m,1H),4.66-4.73(m,1H),4.53-4.65(m,2H),4.49-4.51(m,1H),4.31-4.48(m,2H),2.38(d,J=4Hz,3H),2.17–2.27(m,1H),2.05-2.13(m,1H).MS(ES+)m/z556(M+H+).
example 22: n- (9- ((2R,3R,4S,5R) -5- (((4- (3-chlorophenyl) -2-thio-1, 3, 2-dioxaphosphorinan-2-yl) oxy) methyl) -3, 4-dihydroxytetrahydrofuran-2-yl) -9H-purin-6-yl) butanamide
Referring to the preparation method of example 1, except that N- (9- (3, 4-bis ((t-butyldimethylsilyl) oxy) -5- (((4- (3-chlorophenyl) -2-thio-1, 3, 2-dioxaphosphorinan-2-yl) oxy) methyl) tetrahydrofuran-2-yl) -9H-purin-6-yl) butanamide was used as a starting material, yield was 26%,1H NMR(400MHz,CDCl3)8.59-8.63(m,1H),8.32-8.33(m,1H),7.09-7.31(m,4H),6.01-6.13(m,1H),5.48-5.66(m,1H),4.46-4.90(m,6H),4.17-4.40(m,2H),2.79-2.83(m,2H),2.12-2.26(m,2H),1.95-1.99(m,2H),1.04-1.09(m,3H).MS(ES+)m/z 580(M+H+).
example 23: (4R) -2- (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (4-chloro-2-fluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Reference is made to the procedure for the preparation of example 1 except that (4R) -2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (4-chloro-2-fluorobenzene) -1,3, 2-dioxaphosphorinane 2-sulfide is used as a starting material in the form of a white solid in 61% yield.1H NMR(400MHz,MeOD)8.34-8.31(m,1H),8.22-8.21(m,1H),7.37(t,J=8.0Hz,1H),7.24-7.20(m,1H),7.09-7.06(m,1H),6.12-6.11(m,1H),5.90-5.86(m,1H),4.80-4.23(m,7H),2.42-2.15(m,1H),2.14-2.05(m,1H).31P NMR(MeOD):=64.86,61.84ppm.MS(ES+)m/z 532(M+H+).
Example 24: (4S) -2- (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (4-chloro-2-fluorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
With reference to the preparation process of example 1, except that (4S) -2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (4-chloro-2-fluorobenzene) -1,3, 2-dioxaphosphorinane 2-sulfide is used as a starting material in a yield of 62%,1H NMR(400MHz,MeOD)8.36-8.34(m,1H),8.22(s,1H),7.44(t,J=8.0Hz,1H),7.27-7.19(m,2H),6.12-6.10(m,1H),5.92-5.89(m,1H),4.88-4.32(m,7H),2.36-2.33(m,1H),2.10-2.07(m,1H).31P NMR(MeOD):=64.99,61.92ppm.MS(ES+)m/z 532(M+H+).
example 25: (4R) - (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
With reference to the preparation of example 1 except that (4R) -2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (3-chlorobenzene) -1,3, 2-dioxaphosphorinane 2-sulfide was used as a starting material in 67% yield,1H NMR(400MHz,DMSO-d6)8.39-8.32(m,1H),8.15(s,1H),7.48-7.21(m,6H),5.97(d,J=4.0Hz,1H),5.72-5.63(m,2H),5.45-5.34(m,1H),4.74-4.15(m,7H),2.24-1.98(m,2H).31P NMR(MeOD):=63.84,61.21ppm.MS(ES+)m/z 514(M+H+).
example 26: (4S) - (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
With reference to the preparation of example 1 except that (4S) -2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (3-chlorobenzene) -1,3, 2-dioxaphosphorinane 2-sulfide is used as a starting material in a yield of 72%,1H NMR(400MHz,DMSO-d6)8.35-8.32(m,1H),8.14-8.13(m,1H),7.49-7.21(m,6H),5.97(d,J=4.0Hz,1H),5.71-5.62(m,2H),5.45-5.34(m,1H),4.65-4.15(m,7H),2.23-2.08(m,2H).31P NMR(MeOD):=63.62,61.25ppm.MS(ES+)m/z 514(M+H+).
example 27: (4R) - (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (2, 5-dichlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Reference is made to the preparation of example 1, except that (4R) -2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (2, 5-dichlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide is used as starting material in 73% yield,1H NMR(400MHz,MeOD)8.35-8.29(m,1H),8.15-8.14(m,1H),7.58-7.46(m,3H),7.28(s,2H),5.98-5.83(m,2H),5.63-5.61(m,1H),5.46-5.45(m,1H),4.74-4.16(m,7H),2.33-2.28(m,1H),2.18-2.14(m,1H).31PNMR(MeOD):=64.00,60.98ppm.MS(ES+)m/z 548(M+H+).
example 28: (4S) - (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (2, 5-dichlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
Reference is made to the preparation of example 1, except that (4S) -2- (((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methoxy) -4- (2, 5-dichlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide is used as starting material in 73% yield,1H NMR(400MHz,DMSO-d6)8.31-8.25(m,1H),8.14-8.06(m,1H),7.59-7.22(m,5H),5.97-5.85(m,2H),5.64-5.57(m,1H),5.45-5.43(m,1H),4.65-4.08(m,7H),2.33-2.02(m,2H).31P NMR(MeOD):=63.87,61.08ppm.MS(ES+)m/z 548(M+H+).
example 29 and example 30:
example 29(4S) - (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide isomer 1
Example 30(4S) - (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide isomer 2
Mixing (4S) - (((2R,3S, 4R)5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide (200mg,0.39mmol) HP L C (acetonitrile/water ═ 1/10-1/1) is prepared chirally to obtain (4S) - (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide isomer 1, 105mg white solid in 53% yield,1H NMR(400MHz,DMSO-d6)8.32(s,1H),8.13(s,1H),7.49-7.31(m,6H),5.97(d,J=4.0Hz,1H),5.72-5.60(m,2H),5.44(d,J=4.0Hz,1H),4.65-4.63(m,5H),4.28-4.27(m,1H),4.16-4.14(m,1H),2.20-2.10(m,2H).31P NMR(DMSO-d6):=63.64ppm.MS(ES+)m/z 514(M+H+) And (4S) - (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide isomer 2, 8mg white solid in 4% yield,1H NMR(400MHz,Methanol-d4)8.25(s,1H),8.15(s,1H),7.22-7.31(m,3H),7.04-7.06(m,1H),6.05(d,J=4.6Hz,1H),5.49(d,J=11.6Hz,1H),4.78(t,J=5.0Hz,1H),4.29-4.60(m,6H),2.88-3.01(m,1H),210-2.21(m,1H),1.90-1.92(m,1H)。31P NMR(Methanol-d4):=62.07ppm.MS(ES+)m/z 514(M+H+).
example 31 and example 32:
EXAMPLE 31(4R) - (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide isomer 1
EXAMPLE 32(4R) - (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide isomer 2
(4R) - (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide (200mg,0.39mmol) was chirally prepared HP L C (acetonitrile/water ═ 1/10-1/1) to obtain (4R) - ((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxyPhenyl) -4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide isomer 1, 75mg of a white solid, yield 38%,1H NMR(400MHz,DMSO-d6)8.31(s,1H),8.15(s,1H),7.48(s,1H),7.45-7.23(m,5H),5.96(d,J=5.2Hz,1H),5.76-5.62(m,2H),5.46(d,J=5.2Hz,1H),4.69-4.35(m,5H),4.27-4.15(m,2H),2.32-2.08(m,2H).31P NMR(DMSO-d6):=63.86ppm.MS(ES+)m/z 514(M+H+) (ii) a And (4R) - (((2R,3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl) methoxy) -4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide isomer 2, 6mg of a white solid in 3% yield,1H NMR(400MHz,DMSO-d6)8.38(s,1H),8.15(s,1H),7.41–7.42(m,2H),7.31(s,2H),7.21-7.23(m,1H),5.96(d,J=4.5Hz,1H),5.63(d,J=5.6Hz,1H),5.34-5.42(m,2H),4.74(d,J=5.1Hz,1H),4.39-4.49(m,2H),4.14-4.38(m,5H),2.28–2.12(m,1H),1.83-1.85(m,1H).31P NMR(DMSO-d6):=61.20ppm.MS(ES+)m/z 514(M+H+)。
example 33: 2- (((((2R, 3S,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-dihydroxytetrahydrofuran-2-yl)) methyl) amino) -4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide
With reference to the preparation of example 1, except that 2- ((((2R,3R,4R,5R) -5- (6-amino-9H-purin-9-yl) -3, 4-bis ((tert-butyldimethylsilyl) oxy) tetrahydrofuran-2-yl) methyl) amino) -4- (3-chlorophenyl) -1,3, 2-dioxaphosphorinane 2-sulfide is used as a starting material in a yield of 35%,1H NMR(400MHz,MeOD)8.38-8.29(m,1H),8.22-8.13(m,1H),7.46-7.29(m,4H),5.95-5.83(m,1H),5.71-5.55(m,1H),5.05-4.93(m,1H),4.82-4.69(m,1H),4.67-4.51(m,1H),4.42-4.26(m,2H),3.68-3.52(m,1H),3.43-3.34(m,1H),2.46-2.28(m,1H),2.21-2.04(m,1H).MS(ES+)m/z 513.0(M+H+)。
the compounds prepared in each example of table 1 are as follows:
Figure BDA0001943047350000321
Figure BDA0001943047350000331
Figure BDA0001943047350000341
Figure BDA0001943047350000351
example 33 in vitro metabolic stability experiments
1. Method of producing a composite material
In order to understand the possible metabolism of the compounds in vivo, the metabolic stability of human liver microsomes in vitro (X008070, RI L D) and CYP3A4 enzyme (C3A4R046B, CYPEX) was examined for various compounds by adding each compound (including CS0002, CS0005, CS0008, CS0009, CS0012, CS0013, CS0018) to human liver microsomes or CYP3A4 enzyme reaction system containing 200nM compound, 5mM MgCl2, 1mM NADH, 0.33mg/ml human liver microsomes (or CYP3A4) in 0.1M Tris-HCl buffer (pH 7.4) with a total volume of 500ul, shaking in a 37 ℃ water pan, taking 50 bath reaction solutions in 0min, 5min, 15min and 30min, adding 200 mg/ml CYP3A4), incubating the solution in the total volume of 500ul, vortexing the solution, measuring the concentration of each compound in vitro (XXX) in a chromatographic analysis (MRX) at 100 rpm, measuring the concentration of each compound in the sample (ATMS) after centrifugation, taking the concentration of the sample (ATX) and measuring the concentration of each of the sample (ATX) in the sample (ATMS) according to the concentration of the expected concentration of the concentration of each of the sample (ATX 3A 3) in 0.3) of the sample (ATMS) in 0.1M) and measuring the sample (ATMS) in the sample (ATMS) under the concentration of1/2× protein mass) and the formula (2) the rate of formation of metabolites (V)AMPS) The calculation is made as concentration of metabolite/reaction time.
2. Results
As shown in table 2, each compound had a faster clearance in human liver microsomes, with CS0005 and CS0009 being the fastest clearance. And the compounds are not all converted to the desired product AMPS, with CS0008 and CS0009 producing AMPS at the fastest rate. The results show that the compounds with the structures can be effectively converted into AMPS under the action of human liver microsomes and CYP3A4 enzyme, and can be converted into active substance AMPS to play a role after being taken by the liver in vivo as a medicament.
TABLE 2 intrinsic clearance and metabolite production of compounds in human liver microsome and CYP3A4 enzyme reaction System
Rate of formation
Figure BDA0001943047350000361
N.d. compound was rapidly cleared, not detected at 5 min.
Example 34 liver delivery of Compounds experiment
1. Method of producing a composite material
1.1. Animal experiments
Male SD rats weighing 180-300 g, supplied by Shanghai Sphere-Bikeka laboratory animals Co., Ltd, the male animals were acclimatized for more than 3 days, fasted for 12 hours in the evening before the experiment, and water was not prohibited.A solution of AMPS prodrug (Cremophor E L: ethanol: PEG 400: physiological saline 1: 1: 4: 4) including CS0002, CS0005, CS0008, CS0009, CS0012, CS0013, CS0015, CS0018, CS0028, CS0029, CS0033, CS0034, CS0035, CS0036, CS0037, CS0038, CS0045, CS0046, CS0053 and CS0054 was prepared before the administration by checking whether the body weight of the animals conforms to the experimental requirements or not, 14 rats were selected for grouping, 2 rats per group were filled with 20. mu. mol/kg of the drug solution, 0.167, 0.5, 1,3, 6, 12 and 24 hours, the rat was collected with a gas, and stored in a sterile refrigerated blood collection tube at 355 ℃ after the centrifugation, the centrifugation of the rat, the sample was collected in a sterile heparinized, and stored in a sterile refrigerated tube at 355 ℃ for a temperature of 0.80 mm, and weighed.
Determination of the content of AMPS prodrug active metabolites in biological samples
Sample pretreatment
Plasma sample 40 μ L was placed in a centrifuge tube, 200 μ L containing 0.5 μ g/m L PMPA in methanol was added, vortexed for 1min, centrifuged at 4 deg.C (15000rpm) for 5min, the supernatant was mixed with water 1:1 and injected for analysis.
Quantitatively weighing tissue sample, adding 5 times volume of methanol solution containing 0.5 μ g/m L PMPA into a homogenizing tube, homogenizing at low temperature, performing low temperature ultrasonic treatment for 15min, centrifuging at 4 deg.C (15000rpm) for 5min, mixing supernatant with water at a ratio of 1:1, and analyzing by sample injection.
Chromatographic mass spectrometry conditions
L C-MS/MS-AJ (Triple Quad 5500, AB SCIEX) for the analysis of the samples column AcquistyUP L C HSS T3(2.1 × mm, 1.8 μm), column temperature 40 ℃, flow rate 0.5m L/min, mobile phase A0.1% aqueous formic acid, mobile phase B acetonitrile/methanol/formic acid (900/100/1, V/V), sample separation using gradient elution, procedure as in Table 3, and mass spectrometry conditions for the corresponding internal standards electrospray ionization (ESI) positive ion mode, monitored ion pair m/z 364/136(AMPS) for Multiple Reaction Monitoring (MRM), 288/176(PMPA), capillary voltage 16.0kV, temperature 500 ℃, desolvation gas flow 1000L/h, scan time 0.025 seconds, collision energy 25V.
TABLE 3AMPS liquid phase elution gradient conditions
Figure BDA0001943047350000371
1.3 data analysis
Concentration-time profiles of AMPS released from each prodrug were prepared in plasma, liver and heart. The area under the AMPS tissue concentration-time curve (AUC0-T) and the time to peak (T) were obtained by fitting calculation using the log-linear trapezoidal method in a non-compartmental model of WinNonlin8.0(Pharsight, CA)max) And peak concentration in AMPS tissue (C)max)。
2. Results
Results of liver tissue distribution after gavage of 20 μmol/kg in rats showed that the exposure and peak concentration of the active molecule AMPS released from CS0002, CS0009 and CS0013 was more than twice as high as CS0005, CS0008, CS0012, CS0015 and CS0018 (table 4 and figure 1), indicating that 3 chloro substitution, 2 fluoro-4 chloro substitution and 2,5 dichloro substitution on the phenyl ring all contribute to the release and enrichment of the prodrug at the liver site. CS0002, substituted with 3 chloride, showed a longer peak time than CS0009 and CS0013 (table 4 and figure 1), indicating that CS0002 is more likely to maintain AMPS in the liver above effective therapeutic concentrations for a longer period of time.
As shown in Table 4 and FIG. 1, the compounds CS0002, CS0009, CS0013, CS0029, CS0033 and CS0037 all showed higher liver exposure after the administration of 20. mu. mol/kg of the drug solution to the gavage of the rats. The result of liver tissue AMPS distribution shows that CS0034 is 1.5 times that of CS0035, and chiral separation is carried out on CS0034, so that AMPS liver exposure of CS0053 is 275 times higher than that of corresponding CS0054, 3.7 times and 2.8 times higher than that of CS0002 and CS0034, and peak concentration of CS0053 is 4.5 times and 2.2 times higher than that of CS0002 and CS 0034. In addition to the higher AMPS liver exposure and peak concentration, CS0053 also retained a 3 hour peak-reaching time similar to CS 0002. Chiral resolution of CS0035 resulted in 44-fold higher AMPS liver exposure of CS0045 than the corresponding CS0046, 94.6% and 112.3% of CS0002 and CS 0035.
Candidate prodrugs include CS0053, CS0045, CS0034, CS0035 and CS0002, the concentration of AMPS in rat plasma was near or below the limit of detection for the LC-MS, and the AMPS exposure and peak concentration in rat heart tissue could not be determined (table 4). AMPK agonists are reported to be exposed to sufficient amounts in the heart as a major cause of toxic side effects that cause myocardial hypertrophy (Science, Myers et al 2017), and therefore the present subject is concerned with liver-specific delivery properties through cyclic phosphate (4-aryl-2-oxo-1, 3, 2-dioxaphosphorinane) precursor structures, i.e. 1) the release of AMPS is specifically catalyzed by CYP3A in cytochrome P450 isozyme family in hepatocytes, 2) while highly polar AMPS cannot efficiently enter the circulation system from hepatocytes and reach the heart, thus hopefully reducing cardiotoxicity caused by AMPK agonist AMPS. The above results show that the compounds of formula CS0045 and formula CS0053 according to the present invention have higher hepatic deliverability, which results in lower dosage required for treatment, and thus higher safety or lower toxic and side effects, thus greatly improving the clinical therapeutic index of AMPS (fig. 2).
Table 4. exposure of the metabolite AMPS in liver, heart and plasma (h x nmol/m L, concentration/tissue volume), peak concentration (nmol/m L) and peak time (h) within 24 hours after gavage administration of each AMPS precursor compound in rats.
Figure BDA0001943047350000381
N.D. indicates that the metabolite specific tissue concentrations were all below the detection lower limit of the L C-MS/MS method, 5ng/m L, at the time of the experimental design detection.
Example 35 mouse liver Primary cell level Compound AMPK activation assay
1. Method of producing a composite material
1.1 isolation and culture of mouse liver Primary cells
Male C57B L/6 mice weighing 18-20 g, supplied by Shanghai Sepul-Bikai laboratory animals Co., Ltd. for primary mouse liver cell isolation by the two-step method of in situ liver perfusion, the method comprises preheating D-Hank's perfusate (with addition of 0.5mM EGTA, 25mM HEPES in 1 × HBSS) and low sugar DMEM digestive juice (with addition of 100CDU/ml collagenase IV, 15mM HEPES, 100ug/ml Streptomyces and 100IU/ml Penicilin), anaesthetizing the mice with Suuta 50 (Zoletail 50), shaving, sterilizing with 75% ethanol, fixing on a super clean bench, opening the abdominal cavity, exposing the inferior vena cava and portal vein, cannulating the inferior vena using a trocar, perfusing with D-Hank's fluid, after the mice are slightly swollen, cutting the portal vein to perfuse with 10ml/min perfuse flow rate about 80ml, replacing the perfusate with 10 min, removing the perfusate, continuously inoculating 10 min with a low sugar DMEM-containing cell suspension, centrifuging the perfusate, collecting the supernatant, inoculating the supernatant with 10ml perfusate, centrifuging the supernatant, and collecting the supernatant, and the supernatant, wherein the supernatant is obtained by the centrifugation are obtained by the method comprises the steps of 10ml DMEM 20ml DMEM 2, and the steps of 10ml DMEM 4 min, the steps of 10ml perfusate, and the centrifugation2) Culturing in medium.
1.2 HTRF phosphorylation assay of AMPK activating Activity of Compounds
Separating the obtained mouse liver primary cells to obtain 2 × 105The cells were seeded at a cell density of 96-well plates and placed in an incubator (37 ℃ C., 5% CO)2) Medium culture for 12h, then adding compounds (including CS0002, CS0005, CS0008, CS0009, CS0015, CS0018, CS0034, CS0035, CS0045, CS0046, CS0053 and CS0054) at a final concentration of 200uM for treatment. After 1h of compound action, HTRF (Homogeneous Time Resolved fluorescence) assay was carried out according to the Phospho-AMPK (Thr172) cellular assay Kit (Cat:64MPKPEG) specification from Cisbio. The specific method comprises the following steps: throwing away the culture solution, sucking on absorbent paper, immediately adding 50ul of lysis solution into each hole, and oscillating at room temperature for 30 min; taking out 16ul of lysate, transferring the lysate to a 384-well plate, adding 4ul of antibody detection solution, and carrying out oscillation reaction for 2 hours at room temperature; in Perkinelmer
Figure BDA0001943047350000391
Detection was performed on a Multilabel Plate Reader, and signal values were recorded. The activation efficiency was used as an index for evaluating AMPK activation activity of the compound.
2. Results
After compound treatment of mouse primary hepatocytes, HTRF assay results showed that CS0002, CS0008, CS0009, CS0015 and CS0018 had strong AMPK activating activity, with CS0005 being inactive only, with CS0015 and CS0018 activating activity being most pronounced (fig. 3). The results show that the structural compound can enter cells at the in vitro cell level to exert AMPK activation effect.
Compounds CS0034, CS0035, CS0045, CS0046, CS0053 and CS0054 are structural split bodies of compound CS 0002. The results show that the activation effect of compound CS0034 is about 2 times that of compound CS0035, and that the activation effect of CS0053 in the chiral resolving body of CS0034 is about three times that of CS0054, with a better AMPK activation effect.
All documents referred to herein are incorporated by reference into this application as if each were individually incorporated by reference. Furthermore, it should be understood that various changes and modifications of the present invention can be made by those skilled in the art after reading the above teachings of the present invention, and these equivalents also fall within the scope of the present invention as defined by the appended claims.

Claims (10)

1. A compound represented by the following formula I, or a stereoisomer or tautomer thereof, or a pharmaceutically acceptable salt, hydrate or solvate thereof,
Figure FDA0001943047340000011
wherein:
R1selected from the group consisting of: substituted or unsubstituted C6-C18 aryl, substituted or unsubstituted 5-12 membered heteroaryl;
R2selected from the group consisting of: hydrogen, substituted or unsubstituted C1-C18 alkyl, substituted or unsubstituted C3-C8 cycloalkyl, substituted or unsubstituted C2-C18 alkanoyl, substituted or unsubstituted C2-C18 alkoxycarbonyl, mono-or di-C2-C18 alkylaminocarbonyl: halogen, haloalkyl, nitro, hydroxy, amino and cyano;
R3and R4Each independently selected from the group consisting of: hydrogen, fluorine, C1-C6 alkyl, C1-C6 alkoxy; or R3And R4Together form a group selected from: a C3-C8 carbocyclic ring, or a 5-12 membered heterocyclic ring;
x is O, S, NH, substituted or unsubstituted C1-C4 alkylene;
wherein said substitution means that the hydrogen atoms on the group are substituted by one or more (e.g. 2,3,4, etc.) substituents selected from the group consisting of: halogen, deutero, C1-C6 alkoxy, halogenated C1-C6 alkyl, halogenated C1-C6 alkoxy, halogenated C3-C8 cycloalkyl, methylsulfonyl, -S (═ O)2NH2Oxo (═ O), -CN, hydroxy, -NH2Carboxy, C1-C6 amido (-C (═ O) -N (Rc)2or-NH-C (═ O) (Rc), Rc being H or C1-C5 alkyl), C1-C6 alkyl- (C1-C6 amido), or a substituted or unsubstituted group selected from: C1-C6 alkyl, C3-C8 cycloalkyl, C1-C6 amino, C6-C10 aryl, 5-10 membered heteroaryl having 1-3 heteroatoms selected from N, S and O, having 1-3 heteroatoms selected from N, S and5-12 membered heterocyclic group of hetero atom of O, - (CH)2) -C6-C10 aryl, - (CH)2) - (5-to 10-membered heteroaryl having 1 to 3 heteroatoms selected from N, S and O), and the substituents are selected from the group consisting of: halogen, C1-C6 alkyl, C1-C6 alkoxy, oxo, -CN, -NH2OH, -C6-C10 aryl, -C1-C6 amino, -C1-C6 amido, -5-10 membered heteroaryl having 1-3 heteroatoms selected from N, S and O.
2. The compound of claim 1, or a stereoisomer or tautomer thereof, or a pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein the compound of formula I has a structure selected from the group consisting of:
Figure FDA0001943047340000012
3. the compound of claim 1, or a stereoisomer or tautomer thereof, or pharmaceutically acceptable salt, hydrate or solvate thereof, wherein R is1Has a structure as shown in formula II, III, IV, V, VI or VII:
Figure FDA0001943047340000021
wherein:
the dotted line is a bond or nothing;
each A is1、A2、A3、A5、A6、A7、A8Each independently is O, S, N, NH, CH or CH2;A4And A9Each independently is C or N;
each B1、B2、B3、B4、B6、B7、B8、B9Each independently is O, S, N, NH, CH or CH2;B5And B10Each independently is C or N;
each R is5、R6、R7And R8Each independently selected from the group consisting of: halogen, substituted or unsubstituted C1-C6 alkyl, substituted or unsubstituted C1-C6 alkoxy;
i is 0, 1,2, 3,4 or 5;
Y1o, S or NH;
Y2and Y3Each independently selected from O, N or CH;
j is 0, 1,2, 3 or 4;
m is 0, 1,2 or 3;
n is 0, 1,2, 3 or 4.
4. The compound of claim 1, or a stereoisomer or tautomer thereof, or pharmaceutically acceptable salt, hydrate or solvate thereof, wherein said compound has a structure selected from the group consisting of:
Figure FDA0001943047340000022
5. the compound of claim 1, or a stereoisomer or tautomer thereof, or pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein said compound is selected from the group consisting of the structures shown in seq id no:
Figure FDA0001943047340000031
6. the compound of claim 1, or a stereoisomer or tautomer thereof, or pharmaceutically acceptable salt, hydrate, or solvate thereof, wherein said compound is selected from the group consisting of the structures shown in seq id no:
Figure FDA0001943047340000032
Figure FDA0001943047340000041
Figure FDA0001943047340000051
Figure FDA0001943047340000061
Figure FDA0001943047340000071
7. a pharmaceutical composition comprising (a) a therapeutically effective amount of a compound of claim 1, or a pharmaceutically acceptable salt, hydrate, or solvate thereof; and (b) a pharmaceutically acceptable carrier.
8. The pharmaceutical composition of claim 7, wherein the disease or condition is selected from the group consisting of nonalcoholic fatty liver disease (NAF L), nonalcoholic steatohepatitis (NASH) and associated cirrhosis, liver cancer, nonalcoholic fatty liver disease (NAF L D), obesity, diabetes, hypertriglyceridemia, hypercholesterolemia, atherosclerosis, cardiovascular disease, metabolic disease.
9. Use of a compound of formula I according to claim 1 for the preparation of a pharmaceutical composition for the treatment or prevention of diseases or disorders associated with AMPK activation.
10. The use according to claim 9, wherein the disease or condition is selected from the group consisting of nonalcoholic fatty liver disease (NAF L), nonalcoholic steatohepatitis (NASH) and associated cirrhosis, liver cancer, nonalcoholic fatty liver disease (NAF L D), obesity, diabetes, hypertriglyceridemia, hypercholesterolemia, atherosclerosis, cardiovascular disease, metabolic disease.
CN201910027560.9A 2019-01-11 2019-01-11 Liver-specific AMPK agonist and preparation method and application thereof Active CN111434671B (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CN201910027560.9A CN111434671B (en) 2019-01-11 2019-01-11 Liver-specific AMPK agonist and preparation method and application thereof
PCT/CN2020/071525 WO2020143800A1 (en) 2019-01-11 2020-01-10 Liver-specific ampk agonist, preparation method therefor and uses thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910027560.9A CN111434671B (en) 2019-01-11 2019-01-11 Liver-specific AMPK agonist and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN111434671A true CN111434671A (en) 2020-07-21
CN111434671B CN111434671B (en) 2023-07-11

Family

ID=71520433

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910027560.9A Active CN111434671B (en) 2019-01-11 2019-01-11 Liver-specific AMPK agonist and preparation method and application thereof

Country Status (2)

Country Link
CN (1) CN111434671B (en)
WO (1) WO2020143800A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999045016A2 (en) * 1998-03-06 1999-09-10 Metabasis Therapeutics, Inc. Novel prodrugs for phosphorus-containing compounds
WO2000052015A2 (en) * 1999-03-05 2000-09-08 Metabasis Therapeutics, Inc. Novel phosphorus-containing prodrugs
US20090118223A1 (en) * 2005-08-12 2009-05-07 Erion Mark D Novel 2'-c-methyl and 4'c-methyl nucleoside derivatives
CN102164939A (en) * 2007-11-29 2011-08-24 配体制药公司 Antiviral nucleoside compounds
WO2015120237A2 (en) * 2014-02-06 2015-08-13 Riboscience Llc 4'-difluoromethyl substituted nucleoside derivatives as inhibitors of influenza rna replication

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2011504489A (en) * 2007-11-23 2011-02-10 バーイラン ユニバーシティー Non-hydrolyzable nucleoside di- or triphosphate derivatives and uses thereof
CN101659676B (en) * 2009-09-07 2012-06-20 徐奎 Sulfo-Adefovir and Tenofovir liver targeting ester prodrug

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999045016A2 (en) * 1998-03-06 1999-09-10 Metabasis Therapeutics, Inc. Novel prodrugs for phosphorus-containing compounds
WO2000052015A2 (en) * 1999-03-05 2000-09-08 Metabasis Therapeutics, Inc. Novel phosphorus-containing prodrugs
CN1353719A (en) * 1999-03-05 2002-06-12 症变治疗公司 Novel phosphorus-containing prodrugs
US20090118223A1 (en) * 2005-08-12 2009-05-07 Erion Mark D Novel 2'-c-methyl and 4'c-methyl nucleoside derivatives
CN102164939A (en) * 2007-11-29 2011-08-24 配体制药公司 Antiviral nucleoside compounds
WO2015120237A2 (en) * 2014-02-06 2015-08-13 Riboscience Llc 4'-difluoromethyl substituted nucleoside derivatives as inhibitors of influenza rna replication

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
JERZYBE?TOWSKI等: "Nucleoside monophosphorothioates as the new hydrogen sulfide precursors with unique properties", 《PHARMACOLOGICAL RESEARCH》 *

Also Published As

Publication number Publication date
CN111434671B (en) 2023-07-11
WO2020143800A1 (en) 2020-07-16

Similar Documents

Publication Publication Date Title
EP4092026A1 (en) Aryl or heteroaryl pyridone or pyrimidine derivative, preparation method and use thereof
US20050026853A1 (en) Anti-viral 7-deaza D-nucleosides and uses thereof
BR112021016751B1 (en) COMPOUNDS, PHARMACEUTICAL COMPOSITIONS AND USES OF COMPOUNDS
JP2021519325A (en) Inhibitors for assembly of N-heterocyclic 5-membered ring-containing capsid proteins, their pharmaceutical compositions and uses
EP3386992B1 (en) Inhibitors of bruton's tyrosine kinase and methods of their use
EP3816163A1 (en) Cell necrosis inhibitor, preparation method therefor and use thereof
KR20010112354A (en) Inhibitors of impdh enzyme
TW201317215A (en) Therapeutically active compositions and their methods of use
EP3719022A1 (en) Polycyclic compounds as inhibitors of bruton's tyrosine kinase
EP4364804A2 (en) Inhibitors of keap1-nrf2 protein-protein interaction
CA3036195A1 (en) Di-substituted pyrazole compounds for the treatment of diseases
EP4289823A1 (en) Substituted pyridazine phenol derivatives
CN113121509B (en) JAK inhibitor compounds and uses thereof
CN114591315A (en) Cathepsin C small-molecule inhibitor
TWI789479B (en) Phosphor(n)amidatacetal and phosph(on)atalcetal compounds
WO2020227421A1 (en) Modified cyclic dinucleoside compounds as sting modulators
KR102502749B1 (en) Liver Delivery Entecavir Prodrug Nucleotide Cyclophosphate Compounds and Applications
CN111718332B (en) 2-substituted pyrazol amino-4-substituted amino-5-pyrimidine formamide compound, composition and application thereof
EP3818065B1 (en) Phosphate and phosphonate derivatives of 7-amino-5-thio-thiazolo[4,5-d]pyrimidines and their use in treating conditions associated with elevated levels of cx3cr1 and/or cx3cl1
WO2020206308A1 (en) Selective inhibitors of protein arginine methyltransferase 5
CN111434671B (en) Liver-specific AMPK agonist and preparation method and application thereof
US10899787B2 (en) Cytarabine prodrug nucleoside cyclic phosphate compound based on liverspecific delivery and use
JP4310191B2 (en) Taxane derivative crystal and method for producing the same
WO2022245584A1 (en) Unnatural configuration nucleotide prodrug compounds
KR102437095B1 (en) Prodrug of caspase inhibitor

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