WO2023142803A1 - 抗病毒化合物及其用途 - Google Patents

抗病毒化合物及其用途 Download PDF

Info

Publication number
WO2023142803A1
WO2023142803A1 PCT/CN2022/141023 CN2022141023W WO2023142803A1 WO 2023142803 A1 WO2023142803 A1 WO 2023142803A1 CN 2022141023 W CN2022141023 W CN 2022141023W WO 2023142803 A1 WO2023142803 A1 WO 2023142803A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
compound
formula
alkoxy
pharmaceutically acceptable
Prior art date
Application number
PCT/CN2022/141023
Other languages
English (en)
French (fr)
Inventor
王伟光
陈善
Original Assignee
北京恩泰伟医药科技有限公司
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 北京恩泰伟医药科技有限公司 filed Critical 北京恩泰伟医药科技有限公司
Publication of WO2023142803A1 publication Critical patent/WO2023142803A1/zh

Links

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
    • C07H19/06Pyrimidine radicals
    • C07H19/10Pyrimidine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • 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
    • A61K31/7068Compounds 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 having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H1/00Processes for the preparation of sugar derivatives

Definitions

  • the present disclosure relates to a phosphate compound of ⁇ -DN 4 -hydroxycytidine having broad-spectrum antiviral activity, to pharmaceutical compositions comprising such compounds, and to the use of such compounds and pharmaceutical compositions comprising such compounds Use for the suppression of viruses or for the prevention and/or treatment of viral infections in a subject in need thereof.
  • ⁇ -DN 4 -hydroxycytidine (NHC, EIDD-1931) is a derivative of cytidine nucleoside (i.e. cytidine), which has the following structural formula:
  • WO 2019/113462 discloses N 4 -hydroxycytidine and its derivatives as a prodrug of EIDD-1931, wherein a representative compound is the isobutyrate EIDD2801 of EIDD-1931, which has the following structural formula:
  • EIDD2801 Compared with EIDD-1931, the oral bioavailability of EIDD2801 has improved somewhat, but there is also the problem of affecting the absorption in vivo due to the degradation by digestive enzymes (such as but not limited to hydrolytic enzymes) and gastric acid in the gastrointestinal tract in vivo. At the same time, EIDD2801 itself has no biological activity, and EIDD2801 absorbed into the body can only play a role after being hydrolyzed to expose the active structure EIDD-1931, which makes the in vivo activity of EIDD2801 greatly limited by the rate and efficiency of hydrolysis.
  • digestive enzymes such as but not limited to hydrolytic enzymes
  • the present disclosure provides a novel phosphate compound of ⁇ -DN 4 -hydroxycytidine (hereinafter referred to as the compound of the present disclosure) which has broad-spectrum virus replication inhibitory activity.
  • n is any integer from 6-20;
  • n is any integer from 1-5;
  • X is -O- or -NR 0 -;
  • R 0 is H or C 1 -C 6 alkyl
  • Z is -O- or -S-S-;
  • R is H, C 1 -C 6 alkyl, C 6 -C 12 aryl, R'-OC(O)-O-CH 2 - or R'-C(O)-O-CH 2 -;
  • R' is C 1 -C 6 alkyl, C 3 -C 8 cycloalkyl, C 6 -C 12 aryl, 3 to 12 membered containing 1 or 2 heteroatoms independently selected from N, O or S Heterocyclyl or 5 to 12 membered heteroaryl containing 1 or 2 heteroatoms independently selected from N, O or S;
  • the compound of formula I has the structure of formula I-1:
  • the compound of formula I has the structure of formula I-2:
  • the pharmaceutically acceptable salt of any of the compounds of Formula I, I-1 or I-2 is an alkali metal salt (e.g. lithium, sodium, potassium, rubidium, cesium salt), alkaline earth metal salts (such as magnesium, calcium, strontium, barium salts), aluminum salts, organic base salts (such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, diethylamine , diethylaminoethanol, ethylenediamine, imidazole, morphine, 2-hydroxyethylmorpholine, dibenzylethylenediamine, trimethylamine, piperidine, pyrrolidine, benzylamine, tetramethylammonium hydroxide salt ) or amino acid salts (eg lysine, arginine salts).
  • alkali metal salt e.g. lithium, sodium, potassium, rubidium, cesium salt
  • alkaline earth metal salts such as magnesium, calcium, strontium, barium salts
  • a pharmaceutical composition comprising a compound of any one of Formula I, I-1 or I-2 or a pharmaceutically acceptable salt, hydrate, solvate thereof Or stereoisomers and one or more pharmaceutically acceptable carriers.
  • the pharmaceutical composition comprises two, three or more pharmaceutically acceptable carriers.
  • the pharmaceutical composition is made into a pharmaceutically acceptable dosage form (for example, by making any one of formula I, I-1 or I-2 according to the conventional production method in the field of pharmacy,
  • the compound or its pharmaceutically acceptable salt, hydrate, solvate or stereoisomer is mixed with one or more pharmaceutically acceptable carriers and prepared into a pharmaceutically acceptable dosage form).
  • a compound of any one of Formula I, I-1 or I-2 or a pharmaceutically acceptable salt, hydrate, solvate or stereoisomer thereof or A compound comprising any one of Formula I, I-1 or I-2 or a pharmaceutically acceptable salt, hydrate, solvate or stereoisomer thereof and one or more pharmaceutically acceptable carriers A pharmaceutical composition for treating or preventing diseases.
  • the pharmaceutical composition of the invention is used for suppressing virus or for preventing and/or treating viral infection in a subject in need thereof.
  • a compound of any one of formula I, I-1 or I-2 or a pharmaceutically acceptable salt, hydrate, solvate or stereoisomer thereof or A compound comprising any one of Formula I, I-1 or 1-2 or a pharmaceutically acceptable salt, hydrate, solvate or stereoisomer thereof and one or more pharmaceutically acceptable carriers Use of the pharmaceutical composition of the invention in the preparation of a medicament for inhibiting viruses or for preventing and/or treating viral infections in subjects in need.
  • a method for preventing and/or treating viral infection in a patient in need thereof comprising administering an effective amount of formula I, I- A compound of any one of 1 or I-2, or a pharmaceutically acceptable salt, hydrate, solvate or stereoisomer thereof, or a compound comprising any one of formulas I, I-1 or I-2 A pharmaceutical composition of a compound or its pharmaceutically acceptable salt, hydrate, solvate or stereoisomer and one or more pharmaceutically acceptable carriers.
  • the virus is a coronavirus (e.g., a human coronavirus, such as severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus Virus (MERS-CoV) or Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2, including, for example, B.1.1.7 strain, B.1.351 strain, Delta variant strain B.1.617.2, Omic Rong variant B.1.1.529 or other SARS-CoV-2 variants), equine encephalitis viruses (such as eastern equine encephalitis virus, western equine encephalitis virus, or Venezuelan equine encephalitis virus), Chikungunya viruses, Ross River virus, Orthomyxoviridae (e.g. influenza viruses such as influenza A, B or C), Paramyxoviridae (e.g. respiratory syncytial virus), filoviruses (such as
  • the intermediate compound is a compound of Formula 2a:
  • the intermediate compound is a compound of Formula 2a-1:
  • the intermediate compound is a compound of Formula 4a:
  • the intermediate compound is a compound of formula 4a-1:
  • the intermediate compound is a compound of Formula 2-1a:
  • m, n and Z are as defined above for compounds of formula I;
  • A is chloro or nitro;
  • B is hydrogen, chloro or nitro.
  • A is chloro or nitro and B is hydrogen.
  • the intermediate compound is a compound of formula 2-1a-1:
  • the intermediate compound is a compound of Formula 4-1a:
  • m and n are as defined above for compounds of formula I; A is chloro or nitro; B is hydrogen, chloro or nitro. Preferably, A is chloro or nitro and B is hydrogen.
  • the intermediate compound is a compound of formula 4-1a-1:
  • ranges recited in this disclosure include the range endpoints and each integer subsumed within the range.
  • m is any integer from 6-20
  • m can be 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20.
  • n is any integer from 1-5" means that n can be 1, 2, 3, 4 and 5.
  • any subranges formed by these integers are intended to be within the scope of this disclosure.
  • compound of the present disclosure refers to any one or more compounds falling within the scope of any of Formula I, I-1 or I-2, or a pharmaceutically acceptable salt thereof, Hydrates, solvates or stereoisomers.
  • Cx - Cy refers to the range of the number of carbon atoms contained in the group (the endpoints of the range and each integer contained in the range and any subrange formed by these integers are intended to be included within the scope of this disclosure).
  • C 1 -C 6 alkyl means an alkyl group comprising 1 to 6 carbon atoms (eg, 1, 2, 3, 4, 5, or 6 carbon atoms).
  • alkyl refers to a linear or branched saturated monovalent hydrocarbon group having the indicated number of carbon atoms. Alkyl groups typically contain 1-6 carbon atoms (“C 1 -C 6 alkyl”), more preferably 1-5 carbon atoms (“C 1 -C 5 alkyl”), 1-4 carbon atoms (“C 1 -C 5 alkyl”), C 1 -C 4 alkyl”) or 1-2 carbon atoms (“C 1 -C 2 alkyl”).
  • Examples of preferred C 1 -C 6 alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, isopentyl, Neopentyl and n-hexyl.
  • Examples of preferred C 1 -C 4 alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl and tert-butyl.
  • alkenyl refers to a linear or branched unsaturated monovalent hydrocarbon group having the specified number of carbon atoms containing one or more double bonds. Alkenyl groups typically contain 2-6 carbon atoms (“C 2 -C 6 alkenyl”), more preferably 2-5 carbon atoms (“C 2 -C 5 alkenyl”), 2-4 carbon atoms (“C 2 -C 5 alkenyl”), C 2 -C 4 alkenyl”) or 2 carbon atoms (“vinyl”).
  • C 2 -C 6 alkenyl typically contain 2-6 carbon atoms (“C 2 -C 6 alkenyl”), more preferably 2-5 carbon atoms (“C 2 -C 5 alkenyl”), 2-4 carbon atoms (“C 2 -C 5 alkenyl”), C 2 -C 4 alkenyl”) or 2 carbon atoms (“vinyl”).
  • Preferred C 2 -C 6 alkenyl groups include vinyl, 1-propenyl, 2-propenyl, 1-butenyl, 2-butenyl, 1,3-butadien-1-enyl, 1-pentene En-3-yl, 2-penten-1-yl, 3-penten-1-yl, 3-penten-2-yl, 1,3-pentadien-1-yl, 1,4-pentene Dien-3-yl, 1-hexen-3-yl and 1,4-hexadien-1-yl.
  • alkynyl refers to a linear or branched unsaturated monovalent hydrocarbon group having the indicated number of carbon atoms containing one or more triple bonds.
  • Alkynyl groups typically contain 2-6 carbon atoms (“C 2 -C 6 alkynyl”), more preferably 2-5 carbon atoms (“C 2 -C 5 alkynyl”), 2-4 carbon atoms (“C 2 -C 5 alkynyl”), C2 - C4alkynyl ”) or 2 carbon atoms ("ethynyl”).
  • C 2 -C 6 alkynyl groups include ethynyl, propynyl, butynyl, pentynyl and hexynyl.
  • alkoxy denotes an alkyl group attached to the parent molecule through an oxygen atom (ie “-O-alkyl”), wherein “alkyl” is as previously defined.
  • Alkoxy groups generally contain 1-6 carbon atoms ("C 1 -C 6 alkoxy”), more preferably 1-4 carbon atoms ("C 1 -C 4 alkoxy”).
  • C 1 -C 4 alkoxy includes methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy and the like.
  • halogen used herein refers to fluorine, chlorine, bromine, iodine, etc., preferably fluorine atom and chlorine atom.
  • halo as used herein means that one or more hydrogen atoms in a substituent are replaced by one or more same or different halogen atoms.
  • Halogen is as defined above.
  • halogenated C 1 -C 6 alkyl refers to "C 1 -C 6 alkyl” in which one or more hydrogen atoms are replaced by one or more same or different halogen atoms, wherein “C 1 -C 6 Alkyl” is as defined above.
  • halogenated C 1 -C 6 alkoxy refers to "C 1 -C 6 alkoxy” in which one or more hydrogen atoms are replaced by one or more identical or different halogen atoms, wherein “C 1 -C 6 alkoxy” is as defined above.
  • cyano refers to a -CN group.
  • hydroxy refers to a -OH group.
  • nitro refers to the -NO2 group.
  • aryl refers to a monovalent group derived from a monocyclic or fused bicyclic or polycyclic ring system in which at least one ring contains a fully conjugated ⁇ -electron system, having the well-known aromatic character. Hydrocarbyl. Fused aryl may include an aryl ring fused to a saturated or partially unsaturated carbocyclic or heterocyclic ring or to another aryl or heteroaryl ring, provided that in such fused ring system The point of attachment to the parent molecule is an atom of the aromatic portion of the ring system. Typically, aryl groups contain 6-12 carbon atoms (“ C6 - C12 aryl"). Examples of aryl groups include, but are not limited to, phenyl, naphthyl, anthracenyl, phenanthrenyl, indanyl, indenyl, and tetrahydronaphthyl.
  • cycloalkyl refers to a monovalent hydrocarbon radical derived from a non-aromatic saturated carbocyclic ring system containing the indicated number of carbon atoms, which may be attached to the parent molecule through a carbon atom of the cycloalkyl ring Monocyclic, spiro, bridged or fused bicyclic or polycyclic ring systems.
  • the cycloalkyl groups of the present disclosure contain 3-8 carbon atoms (“C 3 -C 8 cycloalkyl”), preferably 3-7 carbon atoms (“C 3 -C 7 cycloalkyl”) or 3 - 6 carbon atoms (" C3 - C6cycloalkyl ").
  • Representative examples of cycloalkyl include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl and the like.
  • heteroatom refers to N, O or S atoms.
  • heterocyclyl refers to a non-aromatic group derived from a group containing the specified number of carbon atoms and also including at least one heteroatom, preferably one to four heteroatoms, more preferably one or two heteroatoms, as ring members.
  • Such heterocyclyl groups may be partially unsaturated.
  • Heterocyclyl includes spiro, bridged or fused rings formed with one or more other heterocycles or carbocycles, wherein such spiro, bridged or fused rings may themselves be saturated, partially unsaturated or Aromatic, provided that the point of attachment to the parent molecule is an atom of the heterocyclic portion of such a ring system.
  • a “heterocyclyl” contains 3 to 12 ring atoms (ie 3 to 12 membered heterocyclyl), preferably 4 to 7 ring atoms (ie 4 to 7 membered heterocyclyl), most preferably 5 or 6 ring atoms (ie, 5- or 6-membered heterocyclyl), wherein the ring atoms include carbon and non-carbon heteroatoms.
  • heterocyclyl groups include azetidinyl, oxetanyl, thietanyl, pyrrolidinyl, imidazolidinyl, pyrazolidinyl, oxazolidinyl , thiazolidinyl, dihydrooxazolyl, dihydroisoxazolyl, dihydropyrrolyl, dihydroimidazolyl, dihydropyrazolyl, dihydrothiazolyl, piperidinyl, dihydropyridyl, dihydro Pyrimidinyl, Piperazinyl, Dioxanyl, Oxythianyl, Azepanyl, Diazepanyl, Oxetanyl, Tetrahydrofuranyl, Tetrahydropyridine Pyranyl, tetrahydrothiophenyl, tetrahydrothiopyranyl, morpholinyl and thiomorpholinyl, etc.
  • heteroaryl refers to a group derived from an aromatic group having the specified number of carbon atoms and also including at least one heteroatom, preferably one to four heteroatoms, more preferably one or two heteroatoms, as ring members.
  • heteroaryl groups contain 5-12 ring atoms (“5-12 membered heteroaryl”), preferably 5-10 ring atoms (“5-10 membered heteroaryl”), more preferably 5 or 6 ring atom (“5- or 6-membered heteroaryl”), wherein the ring atoms include carbon and non-carbon heteroatoms.
  • a heteroaryl group is attached to the parent molecule through a ring atom of the heteroaryl ring, thereby maintaining aromaticity.
  • Heteroaryl may also be fused to another aryl or heteroaryl ring, or to a saturated or partially unsaturated carbocyclic or heterocyclic ring, provided that, on such fused ring system, the base molecule The point of attachment of is an atom of the heteroaromatic portion of the ring system.
  • heteroaryl include furyl, imidazolyl, isoxazolyl, thiazolyl, isothiazolyl, oxadiazolyl, oxazolyl, pyridyl, pyridazinyl, pyrimidine Base, pyrazinyl, pyrazolyl, pyrrolyl, tetrazolyl, thiadiazolyl, thienyl, triazolyl, triazinyl, benzimidazolyl, benzofuryl, benzothienyl, benzo Oxadiazolyl, benzothiadiazolyl, benzothiazolyl, imidazopyridyl, imidazopyrimidinyl, imidazopyridazinyl, cinnolinyl, furopyridyl, indazolyl, indolyl, Isoindolyl, isoquinolyl, naphthyridyl, purin
  • the term "pharmaceutically acceptable” means, within the scope of sound medical judgment, suitable for use in contact with the tissues of a subject, such as a human or other mammal, without undue toxicity, irritation, allergic response or other problems, Those compounds or pharmaceutical compositions that also have a commensurate and reasonable benefit/risk ratio.
  • pharmaceutically acceptable salt refers to an acid or base addition salt formed by any one of the compounds of formula I, I-1 or 1-2 with a pharmaceutically acceptable acid or base, It retains the biological effectiveness and properties of the parent compound, the compound of any of Formula I, I-1 or 1-2.
  • the pharmaceutically acceptable salts include, but are not limited to, alkali metal salts (such as lithium, sodium, potassium, rubidium, cesium salts), alkaline earth metal salts (such as magnesium, calcium, strontium, barium salts), aluminum salts, organic alkali salts (such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, diethylamine, diethylaminoethanol, ethylenediamine, imidazole, morphine, 2-hydroxyethylmorphine, Dibenzylethylenediamine, trimethylamine, piperidine, pyrrolidine, benzylamine, tetramethylammonium hydroxide salts) or amino acid salts (eg lysine, arginine salts).
  • alkali metal salts such as lithium, sodium, potassium, rubidium, cesium salts
  • alkaline earth metal salts such as magnesium, calcium, strontium, barium salts
  • aluminum salts such as
  • the pharmaceutically acceptable salt of the compound of any one of the formulas I, I-1 or 1-2 of the present disclosure can be prepared as follows: an equivalent amount of any one of the formulas I, I-1 or I-2
  • the compound and the base providing the desired counterion are stirred in a solvent at 25-70° C. for 0.5-2 h, and then the solvent is evaporated under reduced pressure to obtain the pharmaceutically acceptable salt.
  • solvent refers to a molecule comprising a compound of any of Formula I, I-1 or I-2 and one or more pharmaceutically acceptable solvent molecules (e.g., ethanol) complexes.
  • solvent e.g., water
  • hydrate is used.
  • stereoisomer refers to isomers resulting from differences in the arrangement of atoms in a molecule in space.
  • enantiomers When a compound has an asymmetric carbon atom, enantiomers will be produced; when a compound has a carbon-carbon double bond or a ring structure, cis-trans isomers will be produced. Enantiomers, diastereoisomers, racemates, cis-trans isomers, geometric Isomers, epimers and mixtures thereof.
  • pharmaceutical composition refers to a pharmaceutically active ingredient (in the context of this disclosure, specifically a compound of any one of Formula I, I-1 or I-2 or a pharmaceutically acceptable Salts, hydrates, solvates and stereoisomers) and pharmaceutically acceptable carriers are combined in a certain proportion and have specific medical uses.
  • the pharmaceutical composition can be made into a pharmaceutically acceptable dosage form, such as tablet, powder (including sterile powder for injection), capsule, granule, solution, syrup, suppository , injections, patches, etc.
  • compositions of the present disclosure can be administered to a subject (e.g., a human or non-human mammal) by any of a variety of routes of administration, including, for example, orally (e.g., as a tablet, capsule, powder, granules); mucosal (e.g., sublingual, nasal, anal, rectal, or vaginal) absorption (e.g., in the form of suppositories, creams, or foams); parenteral (e.g., intramuscular, intravenous, intraperitoneal, subcutaneous or intrathecal injection); transdermal (eg, as a patch applied to the skin); and topical administration (eg, as a cream, ointment, spray or as eye drops applied to the skin).
  • routes of administration including, for example, orally (e.g., as a tablet, capsule, powder, granules); mucosal (e.g., sublingual, nasal, anal, rectal, or vaginal
  • the pharmaceutical compositions may also be formulated for inhaled administration.
  • the pharmaceutical composition usually contains 0.1% by weight to 99.5% by weight of the active ingredient (i.e. the compound of any one of formula I, I-1 or I-2 or its pharmaceutically acceptable salt, hydrate, solvates or stereoisomers), preferably containing 0.5 wt. % by weight, 30% by weight or 50% by weight of active ingredient.
  • the active ingredient i.e. the compound of any one of formula I, I-1 or I-2 or its pharmaceutically acceptable salt, hydrate, solvates or stereoisomers
  • pharmaceutically acceptable carrier means a carrier (or excipient) that does not cause significant irritation to the organism and does not eliminate the biological activity and properties of the administered compound. Any commonly used pharmaceutically acceptable carrier can be used, the choice of which depends on factors such as the particular mode of administration, the effect of the carrier on solubility and stability, and the nature of the dosage form and is within the ordinary skill of the art.
  • materials that can be used as pharmaceutically acceptable carriers include: starches, such as corn starch and potato starch; sugars, such as lactose, glucose and sucrose; cellulose and its derivatives, such as ethyl cellulose, carboxymethyl Sodium cellulose and cellulose acetate; gelatin, acacia, guar, tragacanth; magnesium stearate, zinc stearate, talc; water, saline; oils such as peanut oil, cottonseed oil, olive oil, sesame oil , corn oil, and soybean oil; alcohols, such as ethanol, propylene glycol, glycerin, sorbitol, mannitol, and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; buffers, such as sodium chloride, phosphoric acid Saline buffer solution, etc.
  • starches such as corn starch and potato starch
  • sugars such as lactose, glucose and sucrose
  • prevention refers to preventing the appearance of a disease or the recurrence of a disease that has disappeared in a subject at risk of having the disease.
  • treating refers to controlling, alleviating or ameliorating the pathological progression of a disease and prolonging the survival of an afflicted subject.
  • the term "subject" refers to an individual animal to which a compound or pharmaceutical composition of the present disclosure is intended to be administered, including, but not limited to, humans and/or other primates (e.g., cynomolgus monkeys, rhesus monkeys, monkeys); other mammals such as horses, cows, pigs, sheep, goats, cats, dogs; and birds such as chickens, ducks, geese, quails, turkeys.
  • a preferred subject is a human.
  • effective amount refers to an amount sufficient to affect any one or more beneficial or desired symptoms of the disease, its complications, or intermediate pathological phenotypes presented during the development of the disease.
  • the "effective amount” of any one of the compounds of Formula I, I-1 or I-2 administered, or a pharmaceutically acceptable salt, hydrate, solvate or stereoisomer thereof, will depend on the amount of treatment received The species of the subject, the severity of the disease, the frequency of administration, the metabolic characteristics of the drug and other factors can be judged by the prescribing physician according to routine practice. In general, effective amounts will generally be in the range of about 0.001 to about 100 mg/kg body weight/day, preferably about 0.01 to about 50 mg/kg body weight/day (in single or divided doses).
  • the present disclosure generally relates to a phosphate compound of ⁇ -DN 4 -hydroxycytidine having broad-spectrum antiviral activity, to pharmaceutical compositions comprising such compounds, and to such compounds and pharmaceutical combinations comprising such compounds Use of a substance for inhibiting a virus or for preventing and/or treating a viral infection in a subject in need thereof.
  • the compound of formula I has the structure of formula I-1:
  • the compound of formula I has the structure of formula I-2:
  • n is any integer from 6-20.
  • m is 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20.
  • n is any integer from 1-5.
  • n 1, 2, 3, 4 or 5.
  • X is -O- or -NR 0 -.
  • X is -O-.
  • X is -NR 0 -.
  • R 0 is H or C 1 -C 6 alkyl.
  • R 0 is H.
  • R 0 is C 1 -C 6 alkyl.
  • R is methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, or tert-butyl.
  • R 0 is methyl or ethyl.
  • R 0 is methyl
  • Z is -O- or -S-S-.
  • Z is -O-.
  • Z is -S-S-.
  • R is H.
  • R is methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, or tert-butyl.
  • R is methyl or ethyl.
  • R is methyl
  • R is phenyl, wherein said phenyl is optionally replaced by 1, 2 or 3 independently selected from halogen, C 1 -C 6 alkyl, halogenated C 1 -C 6 alkyl, Substituents of C 2 -C 6 alkenyl, C 2 -C 6 alkynyl, C 1 -C 6 alkoxy, halogenated C 1 -C 6 alkoxy, cyano, hydroxy or nitro.
  • R is phenyl, wherein said phenyl is optionally replaced by 1, 2 or 3 independently selected from halogen, C 1 -C 6 alkyl, halogenated C 1 -C 6 alkyl, Substituents of C 1 -C 6 alkoxy, halogenated C 1 -C 6 alkoxy, cyano, hydroxyl or nitro.
  • R is phenyl, wherein said phenyl is optionally replaced by 1 independently selected from halogen, halogenated C 1 -C 6 alkyl, halogenated C 1 -C 6 alkoxy, cyano Substituent group or nitro substituent.
  • R is phenyl optionally substituted with 1 substituent independently selected from halo or nitro.
  • R is phenyl optionally substituted with 1 substituent independently selected from chloro or nitro.
  • R is R'-OC(O)-O- CH2- .
  • R is R'-C(O)-O- CH2- .
  • R' is methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, or tert-butyl.
  • R' is methyl, ethyl, isopropyl or t-butyl.
  • R' is methyl, isopropyl, or tert-butyl.
  • R' is cyclobutyl, cyclopentyl, cyclohexyl, or cycloheptyl.
  • R' is cyclopentyl or cyclohexyl.
  • R' is phenyl, wherein said phenyl is optionally replaced by 1, 2 or 3 independently selected from halogen, C 1 -C 6 alkyl, haloC 1 -C 6 alkyl , C 1 -C 6 alkoxy, halogenated C 1 -C 6 alkoxy, cyano, hydroxyl or nitro substituents.
  • R' is phenyl, wherein said phenyl is optionally replaced by 1 member independently selected from halogen, halogenated C 1 -C 6 alkyl, halogenated C 1 -C 6 alkoxy, Substitution with cyano or nitro substituents.
  • R' is phenyl
  • R' is pyrrolidinyl, tetrahydrofuranyl, tetrahydrothienyl, pyrazolidinyl, imidazolidinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, Dihydropyrrolyl, dihydrofuranyl, dihydrothienyl, dihydropyrazolyl, dihydroimidazolyl, dihydrooxazolyl, isodihydrooxazolyl, dihydrothiazolyl, isodihydrothiazolyl, Piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, dihydropyridyl, dihydropyrazinyl, dihydropyrimidinyl or dihydropyridazinyl, wherein said group is optionally replaced by 1 , 2 or 3 independently selected from halogen, C
  • R' is pyrrolidinyl, tetrahydrofuranyl, tetrahydrothienyl, pyrazolidinyl, imidazolidinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, Dihydropyrrolyl, dihydrofuranyl, dihydrothienyl, dihydropyrazolyl, dihydroimidazolyl, dihydrooxazolyl, isodihydrooxazolyl, dihydrothiazolyl, isodihydrothiazolyl, Piperidinyl, piperazinyl, morpholinyl, thiomorpholinyl, dihydropyridyl, dihydropyrazinyl, dihydropyrimidinyl or dihydropyridazinyl, wherein said group is optionally replaced by 1 , 2 or 3 independently selected from halogen, C
  • R' is pyrrolyl, furyl, thienyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl , tetrazolyl, triazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, benzimidazolyl, benzofuryl, benzothienyl, benzoxadiazolyl, benzo Thiadiazolyl, benzothiazolyl, imidazopyridyl, imidazopyrimidinyl, imidazopyridazinyl, cinnolinyl, furopyridyl, indazolyl, indolyl, isoindolyl, iso Quinolinyl, naphthyridinyl, purinyl, imid
  • R' is pyrrolyl, furyl, thienyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, oxadiazolyl, thiadiazolyl , tetrazolyl, triazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, benzimidazolyl, benzofuryl, benzothienyl, benzoxadiazolyl, benzo Thiadiazolyl, benzothiazolyl, imidazopyridyl, imidazopyrimidinyl, imidazopyridazinyl, cinnolinyl, furopyridyl, indazolyl, indolyl, isoindolyl, iso Quinolinyl, naphthyridinyl, purinyl, imid
  • each R a and R b is independently H or C 1 -C 6 alkyl.
  • each R a and R b is independently H.
  • each R a and R b is independently C 1 -C 6 alkyl.
  • each Ra and Rb is independently methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, or tert-butyl.
  • each R a and R b is independently methyl or ethyl.
  • each R a and R b is independently methyl.
  • n is any integer from 6-20;
  • n is any integer from 1-5;
  • X is -O- or -NR 0 -;
  • R is H, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl or tert-butyl;
  • R is H, methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, optionally substituted with 1 independently selected from halogen or nitro Substituted phenyl, R'-OC(O)-O-CH 2 - or R'-C(O)-O-CH 2 -;
  • R' is methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, or phenyl .
  • n is any integer from 6-20;
  • n is any integer from 1-5;
  • X is -O- or -NR 0 -;
  • R 0 is H, methyl or ethyl
  • R is H, methyl, ethyl, phenyl optionally substituted with 1 substituent independently selected from chloro or nitro, R'-OC(O)-O-CH 2 - or R'-C (O)-O- CH2- ;
  • R' is methyl, ethyl, isopropyl, t-butyl, cyclopentyl, cyclohexyl or phenyl.
  • n is any integer from 6-20;
  • n is any integer from 1-5;
  • X is -O- or -NR 0 -;
  • R 0 is H or methyl
  • R is H, methyl, phenyl optionally substituted with 1 substituent independently selected from chloro or nitro, R'-OC(O)-O- CH2- or R'-C(O) -O- CH2- ;
  • R' is methyl, isopropyl, t-butyl, cyclopentyl, cyclohexyl or phenyl.
  • the present disclosure provides a compound selected from Table 1, or a pharmaceutically acceptable salt, hydrate, solvate or stereoisomer thereof:
  • the pharmaceutically acceptable salt of any of the compounds of Formula I, I-1 or I-2 is an alkali metal salt (e.g. lithium, sodium, potassium, rubidium, cesium salt), alkaline earth metal salts (such as magnesium, calcium, strontium, barium salts), aluminum salts, organic base salts (such as ethanolamine, diethanolamine, triethanolamine, tromethamine, N-methylglucamine, diethylamine , diethylaminoethanol, ethylenediamine, imidazole, morphine, 2-hydroxyethylmorpholine, dibenzylethylenediamine, trimethylamine, piperidine, pyrrolidine, benzylamine, tetramethylammonium hydroxide salt ) or amino acid salts (eg lysine, arginine salts).
  • alkali metal salt e.g. lithium, sodium, potassium, rubidium, cesium salt
  • alkaline earth metal salts such as magnesium, calcium, strontium, barium salts
  • a pharmaceutical composition comprising a compound of any one of Formula I, I-1 or I-2 or a pharmaceutically acceptable salt, hydrate, solvate thereof Or stereoisomers and one or more pharmaceutically acceptable carriers.
  • the pharmaceutical composition contains 0.1% by weight to 99.5% by weight of the active ingredient (i.e. the compound of any one of Formula I, I-1 or I-2 or a pharmaceutically acceptable salts, hydrates, solvates or stereoisomers), preferably containing 0.5% by weight to 99.5% by weight, more preferably 1% by weight to 50% by weight, such as 1% by weight, 1.5% by weight, 2% by weight, 5% by weight %, 10%, 15%, 20%, 25%, 30% or 50% by weight of active ingredient.
  • the rest of the pharmaceutical composition is a pharmaceutically acceptable carrier.
  • the pharmaceutical composition comprises two, three or more pharmaceutically acceptable carriers.
  • Examples of the pharmaceutically acceptable carrier include, but are not limited to, conventional diluents, excipients, fillers, binders, wetting agents, disintegrants, lubricants, coloring agents, fragrances, absorption-promoting Agents, surfactants, adsorption carriers, etc.
  • the pharmaceutical composition is formulated into a pharmaceutically acceptable dosage form.
  • the compound of any one of Formula I, I-1 or I-2, or a pharmaceutically acceptable salt, hydrate, or solvate thereof is prepared according to conventional production methods in the pharmaceutical field.
  • stereoisomers are mixed with one or more pharmaceutically acceptable carriers and prepared into pharmaceutically acceptable dosage forms.
  • Examples of the pharmaceutically acceptable dosage forms include, but are not limited to, conventional solid dosage forms in the pharmaceutical field (such as but not limited to tablets, capsules, powders, granules, cachets and suppositories, etc.), liquid dosage forms (such as but not limited to limited to solutions, syrups, suspensions, emulsions, elixirs, etc.).
  • the pharmaceutical compositions of the present disclosure can be formulated as sustained release dosage forms to provide controlled release of the active ingredient to optimize antiviral activity.
  • Dosage forms suitable for sustained release include, but are not limited to, multilayer tablets containing layers with different disintegration rates, or capsules containing impregnated or encapsulated porous polymer matrices.
  • the amount of active ingredient in the pharmaceutical composition may range from about 1 mg to about 2000 mg. In some embodiments, the amount may be 1 mg to 1000 mg, 2 mg to 500 mg, 2.5 mg to 100 mg, or 5 mg to 75 mg.
  • a compound of any one of Formula I, I-1 or I-2 or a pharmaceutically acceptable salt, hydrate, solvate or stereoisomer thereof or A compound comprising any one of Formula I, I-1 or I-2 or a pharmaceutically acceptable salt, hydrate, solvate or stereoisomer thereof and one or more pharmaceutically acceptable carriers A pharmaceutical composition for treating or preventing diseases.
  • a compound of any one of Formula I, I-1 or I-2 or a pharmaceutically acceptable salt, hydrate, solvate or stereoisomer thereof or A compound comprising any one of Formula I, I-1 or I-2 or a pharmaceutically acceptable salt, hydrate, solvate or stereoisomer thereof and one or more pharmaceutically acceptable carriers A pharmaceutical composition for inhibiting a virus or for preventing and/or treating a viral infection in a subject in need thereof.
  • the pharmaceutical composition of the invention is used for suppressing virus or for preventing and/or treating viral infection in a subject in need thereof.
  • a compound of any one of formula I, I-1 or I-2 or a pharmaceutically acceptable salt, hydrate, solvate or stereoisomer thereof or A compound comprising any one of Formula I, I-1 or I-2 or a pharmaceutically acceptable salt, hydrate, solvate or stereoisomer thereof and one or more pharmaceutically acceptable carriers Use of the pharmaceutical composition of the invention in the preparation of a medicament for inhibiting viruses or for preventing and/or treating viral infections in subjects in need.
  • a method for preventing and/or treating viral infection in a patient in need thereof comprising administering an effective amount of formula I, I- A compound of any one of 1 or I-2, or a pharmaceutically acceptable salt, hydrate, solvate or stereoisomer thereof, or a compound comprising any one of formulas I, I-1 or I-2 A pharmaceutical composition of a compound or its pharmaceutically acceptable salt, hydrate, solvate or stereoisomer and one or more pharmaceutically acceptable carriers.
  • the virus is a coronavirus (e.g., a human coronavirus, such as severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV) or severe acute respiratory syndrome coronavirus Virus 2 (SARS-CoV-2, including, for example, B.1.1.7 strain, B.1.351 strain, delta variant strain B.1.617.2 strain, Omicron variant strain B.1.1.529 strain or other SARS- CoV-2 variants), equine encephalitis viruses (e.g.
  • SARS-CoV severe acute respiratory syndrome coronavirus
  • MERS-CoV Middle East respiratory syndrome coronavirus
  • SARS-CoV-2 severe acute respiratory syndrome coronavirus Virus 2
  • B.1.1.7 strain B.1.351 strain
  • delta variant strain B.1.617.2 strain Omicron variant strain B.1.1.529 strain or other SARS- CoV-2 variants
  • equine encephalitis viruses e.g.
  • Orthomyxoviridae viruses e.g. Influenza viruses, such as influenza A, B, or C
  • Paramyxoviridae such as respiratory syncytial virus
  • filoviruses such as Ebola, Marburg, or Cuva virus
  • Zika virus Zika virus.
  • the virus is influenza virus.
  • the virus is influenza A virus.
  • the virus is influenza A (H1N1) virus.
  • the virus is the SC09 strain.
  • the virus is influenza A(H3N2) virus.
  • the virus is the FJ09 strain.
  • the virus is influenza B virus.
  • the virus is the SC11 strain.
  • the virus is Zika virus.
  • the virus is the SZ01 strain.
  • the virus is a human coronavirus.
  • the virus is HCoV-229E strain or HCoV-OC43 strain.
  • the virus is severe acute respiratory syndrome coronavirus 2.
  • the virus is strain B.1.1.7, strain B.1.351, or strain B.1.617.2.
  • a compound or pharmaceutical composition of the present disclosure can be administered to a subject (e.g., a human or non-human mammal) by any of a variety of routes of administration, including, for example, orally (e.g., in the form of tablets, capsules, in the form of powders, granules); mucosal (e.g., sublingual, nasal, anal, rectal, or vaginal) absorption (e.g., in the form of suppositories, creams, or foams); parenteral (e.g., intramuscular, intravenous, peritoneal intrathecal injection); transdermal (eg, as a patch applied to the skin); and topical administration (eg, as a cream, ointment, spray or as eye drops applied to the skin).
  • routes of administration including, for example, orally (e.g., in the form of tablets, capsules, in the form of powders, granules); mucosal (e.g., sublingual
  • the total daily dosage may, if desired, be divided and administered in portions during the day. In some embodiments, the daily dose is administered at one time. In other embodiments, the total daily dose is administered in two, three or more divided doses over a 24 hour period.
  • the amount and frequency of administration of the disclosed compound or pharmaceutical composition can be adjusted by the attending physician taking into account factors such as the patient's age, general health, body weight and severity of the symptoms to be treated.
  • the total daily dose of the disclosed compound or pharmaceutical composition is usually about 0.1 to about 2000 mg active ingredient/day, for example about 1 to about 1500 mg/day, about 10 to about 1250 mg/day, about 50 to about 1250 mg/day, about 50 to about About 1000 mg/day, administered in a single dose or in 2, 3 or 4 divided doses.
  • the compounds of the present disclosure have broad-spectrum antiviral activity, and can efficiently inhibit viruses such as: coronaviruses (such as human coronaviruses, such as severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV) or Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2, including, for example, B.1.1.7 strain, B.1.351 strain, Delta variant strain B.1.617.2 strain, Omicron variant B.1.1.529 or other SARS-CoV-2 variants), equine encephalitis viruses (e.g.
  • coronaviruses such as human coronaviruses, such as severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV) or Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2, including, for example, B.1.1.7 strain, B.1.351 strain, Delta variant strain B.1.617.2 strain, O
  • Orthomyxoviridae e.g. influenza viruses such as influenza A, B or C
  • Paramyxoviridae e.g. respiratory syncytial virus
  • filoviruses e.g. Ebola virus, Marburg virus or Kuva virus
  • Zika virus e.g. Zika virus
  • the compounds of the present disclosure have improved oral administration stability and enhanced antiviral activity: before EIDD-2801 enters cells, it is partially converted into EIDD-2801 under the action of acid, alkali and digestive enzymes in the gastrointestinal tract. 1931, thus affecting the absorption in the body and weakening the antiviral activity.
  • the difference is that the compound of the present disclosure will not be degraded by acid, alkali and digestive enzymes in the gastrointestinal tract, and can be absorbed in the body as a compound prototype, and the cell permeability is enhanced, and it is transformed into EIDD-1931 thereby exerts antiviral effect and improves antiviral activity;
  • the compounds of the present disclosure have relatively high safety: the compounds of the present disclosure belong to lipid drugs, have improved pharmacokinetic properties (especially in vivo distribution), and can reduce the toxicity of nucleoside drugs to the kidneys, Improve the targeting of the lung and liver, thereby significantly reducing the lung damage caused by viral infection, and thus have the characteristics of drug safety.
  • Scheme 1 shows a method for the preparation of compounds of formula I. This method is especially suitable for the preparation of compounds of formula I wherein X is -O-.
  • the compound of formula 1d (can be selected from 2-chlorophenyl dichlorophosphonoester, 2-nitrophenyl dichlorophosphonoester, 4-chlorophenyldichlorophosphonoester, 4-nitrophenyl Chlorophosphonoate, 2,4-dichlorophenyldichlorophosphonoate, 2,4-dinitrophenyldichlorophosphonoate) and compounds of formula 1c (both are directly commercially available) in React in anhydrous solvent, then add the compound of formula 1a (according to Chemistry-A European Journal (2016), 22 (21), the method preparation of 7215-7224) and the compound of formula 1b (can be according to document Kyle E.Giesler, et al ., LiottaReduction Sensitive Lipid Conjugates of Tenofovir: Synthesis, Stability, and Antiviral Activity; J.Med.Chem.2016, 59, 7097-7110; US2012/0058976A1 or Susum
  • the above reaction can give the compound of formula 2a.
  • the anhydrous solvents required in the above reaction process include but are not limited to anhydrous tetrahydrofuran, 2-methyltetrahydrofuran, dichloromethane, N-methylpyrrolidone, toluene, 1,4-dioxane or their mixed solvents, etc. , preferably tetrahydrofuran and 2-methyltetrahydrofuran.
  • the reaction temperature is 0-50°C, preferably 0-35°C; the reaction time is 5-48 hours, preferably 10-20 hours.
  • the base required for the reaction can be N, N-diethylethylamine, triethylamine, pyridine (all directly commercially available) (with reference to Steven D.Dong, Chin-Chung Lin, Mel Schroeder, Synthesis and evaluation of a new phosphorylated ribavirin prodrug, Antiviral Research 99(2013) 18-26).
  • the compound of formula 2a is reacted with the compound of formula 3b (2,4,5-triisopropylbenzenesulfonyl chloride, TBSCl) to obtain formula 3c compound of.
  • the compound of formula 3c is reacted with hydroxylamine hydrochloride under basic conditions to obtain the compound of formula 4a.
  • the solvent used in this reaction is dichloromethane, chloroform, 1,2-dichloroethane, acetonitrile, tetrahydrofuran, 2-methyltetrahydrofuran, N-methylpyrrolidone, toluene, 1,4-dioxane or their Mixed solvents and the like are preferably dichloromethane, chloroform, and 1,2-dichloroethane.
  • the reaction temperature is 0-35°C.
  • the base required for the reaction is imidazole, N, N-diethylethylamine, triethylamine, pyridine (all directly commercially available), preferably imidazole (refer to WO 2019/113462).
  • the compound of formula 4a is reacted in a solvent, and the compound of formula 6a is obtained after removing the hydroxyl protecting group.
  • the solvent used for this reaction may be ammonia in methanol, ammonia in ethanol or ammonia in isopropanol.
  • the reaction temperature is 20-80°C.
  • the compound of formula 5a is reacted in a solvent, and the compound of formula I is obtained after removing the hydroxyl protecting group.
  • the solvent used for this reaction may be ammonia in methanol, ammonia in ethanol or ammonia in isopropanol.
  • the reaction temperature is 20-50°C.
  • Scheme 2 shows another method for preparing the compound of formula I, wherein compared with scheme 1, the compound of formula 1-1d is used instead of the compound of formula 1d. This method is especially suitable for the preparation of compounds of formula I wherein X is -NR 0 -.
  • the compound of formula 1-1d and the compound of formula 1c are reacted in anhydrous solvent, and then the compound of formula 1a and the compound of formula 1b are added to react.
  • the above reaction can give the compound of formula 2a-1.
  • the anhydrous solvent used in this reaction includes but not limited to anhydrous tetrahydrofuran, 2-methyltetrahydrofuran, dichloromethane, N-methylpyrrolidone, toluene, 1,4-dioxane or their mixed solvents, etc., preferably tetrahydrofuran and 2-methyltetrahydrofuran.
  • the reaction temperature is 0-50°C, preferably 0-35°C; the reaction time is 5-48 hours, preferably 10-20 hours.
  • the compound of formula 2a-1 is reacted with the compound of formula 3b (2,4,5-triisopropylbenzenesulfonyl chloride, TBSCl) to obtain A compound of formula 3c-1.
  • the compound of formula 3c-1 is reacted with hydroxylamine hydrochloride under basic conditions to obtain the compound of formula 4a-1.
  • the solvent used in this reaction is dichloromethane, chloroform, 1,2-dichloroethane, acetonitrile, tetrahydrofuran, 2-methyltetrahydrofuran, N-methylpyrrolidone, toluene, 1,4-dioxane or their Mixed solvents and the like are preferably dichloromethane, chloroform, and 1,2-dichloroethane.
  • the reaction temperature is 0-35°C.
  • the base required for the reaction can be imidazole, N, N-diethylethylamine, triethylamine, pyridine (all directly commercially available), preferably imidazole (refer to WO 2019/113462).
  • the compound of formula 4a-1 is reacted in a solvent, and the compound of formula I is obtained after removing the hydroxyl protecting group.
  • the solvent used for this reaction may be ammonia in methanol, ammonia in ethanol or ammonia in isopropanol.
  • the reaction temperature is 20-50°C.
  • variables m, n and R' are as defined above for compounds of formula I.
  • A is chlorine or nitro
  • B is hydrogen, chlorine or nitro.
  • A is chloro or nitro and B is hydrogen.
  • Step 1 Preparation of intermediate compound of formula 2-1a
  • the compound of formula 1d (can be selected from 2-chlorophenyl dichlorophosphonoester, 2-nitrophenyl dichlorophosphonoester, 4-chlorophenyldichlorophosphonoester, 4-nitrophenyl Chlorophosphonoate, 2,4-dichlorophenyl dichlorophosphonoate, 2,4-dinitrophenyl dichlorophosphonoate) and the compound of formula 1c (both can be directly commercially available) reaction , and then add the compound of formula 1-1a (prepared according to the method disclosed in WO 2019/113462 or Angewandte Chemie, International Edition, 53(19), 4862-4866; 2014) and the compound of formula 1-1b (according to US2012/0058976A1 Or Susumu Tsushima, Chew.Pharm.I3u11.30 (9) 3260-3270, the method disclosed in 1982 is prepared, specifically can be prepared as follows: use raw material CH 3 (CH 2 ) m CH 2 OH and
  • the base required for the reaction can be N, N-diethylethylamine, triethylamine, pyridine (all directly commercially available) (with reference to Steven D.Dong, Chin-Chung Lin, Mel Schroeder, Synthesis and evaluation of a new phosphorylated ribavirin prodrug, Antiviral Research 99(2013) 18-26).
  • the compound of formula 2-1a is reacted with the compound of formula 3b (2,4,5-triisopropylbenzenesulfonyl chloride, TBSCl) to obtain A compound of formula 3-1c.
  • the compound of formula 3-1c is reacted with hydroxylamine hydrochloride under basic conditions to obtain the compound of formula 4-1-a.
  • the solvent used in this reaction is dichloromethane, chloroform, 1,2-dichloroethane, acetonitrile, tetrahydrofuran, 2-methyltetrahydrofuran, N-methylpyrrolidone, toluene, 1,4-dioxane or their Mixed solvents and the like are preferably dichloromethane, chloroform, and 1,2-dichloroethane.
  • the reaction temperature is 0-35°C.
  • the base required for the reaction is imidazole, N,N-diethylethylamine, triethylamine, pyridine (all can be obtained commercially), preferably imidazole.
  • the compound of formula 4-1-a is treated with the compound of 4b (1,1,3,3-tetramethylguanidine) and the compound of formula 4c (2-pyridinecarbaldoxime) to remove the substituted phenyl moiety to obtain A compound of formula 5-1a.
  • the solvents used in this reaction are tetrahydrofuran, 2-methyltetrahydrofuran, and N-methylpyrrolidone.
  • the reaction temperature is 10-40°C.
  • the compound of formula 4-1-a is removed from isopropylidene in formic acid aqueous solution (concentration is 80%-90%, w/w) or trifluoroacetic acid aqueous solution (concentration is 80%-90%, w/w) Reaction of the methyl protecting group affords compounds of formula 6-1a.
  • the reaction temperature is 20-80°C.
  • the compound of formula 5-1a is de-isopropylmethylene in formic acid aqueous solution (concentration is 80%-90%, w/w) or trifluoroacetic acid aqueous solution (concentration is 80%-90%, w/w) Reaction of the protecting group affords the compound of formula 1-2.
  • the reaction temperature is 20-80°C.
  • a compound of formula 6-1a is treated with a compound of formula 4b (1,1,3,3-tetramethylguanidine) and a compound of formula 4c (2-pyridinecarbaldoxime) to remove the substituted phenyl moiety to obtain the formula Compounds of I-2.
  • the solvents used in this reaction are tetrahydrofuran, 2-methyltetrahydrofuran, and N-methylpyrrolidone.
  • the reaction temperature is 10-40°C (refer to WO 2019/113462).
  • Scheme 4 shows another method for preparing the compound of formula 1-2, wherein compared with scheme 3, the compound of formula 1-1d is used instead of the compound of formula 1d. This method is especially suitable for the preparation of compounds of formula I-2 wherein X is -NR 0 -.
  • Step 1 Preparation of intermediate compound of formula 2-1a-1
  • the compound of formula 2-1a-1 is reacted with the compound of formula 1c in the presence of phosphorus oxychloride to obtain the compound of formula 3-1a-1.
  • the compound of formula 3-1a-1 is then reacted with hydroxylamine hydrochloride under basic conditions to obtain the compound of formula 4-1a-1.
  • the solvent used in this reaction is acetonitrile, tetrahydrofuran, 2-methyltetrahydrofuran, N-methylpyrrolidone, toluene, 1,4-dioxane or a mixed solvent thereof, preferably acetonitrile and dioxane.
  • the reaction temperature is 0-35°C.
  • the base required for the reaction can be N, N-diethylethylamine, triethylamine, pyridine (all directly commercially available) (with reference to Steven D.Dong, Chin-Chung Lin, Mel Schroeder, Synthesis and evaluation of a new phosphorylated ribavirin prodrug, Antiviral Research 99(2013) 18-26).
  • the compound of formula 2-1a-1 is reacted with the compound of formula 3b (2,4,5-triisopropylbenzenesulfonyl chloride, TBSCl) , to obtain the compound of formula 3-1c-1.
  • the compound of formula 3-1c-1 is reacted with hydroxylamine hydrochloride under basic conditions to obtain the compound of formula 4-1a-1.
  • the solvent used in this reaction is dichloromethane, chloroform, 1,2-dichloroethane, acetonitrile, tetrahydrofuran, 2-methyltetrahydrofuran, N-methylpyrrolidone, toluene, 1,4-dioxane or their Mixed solvents and the like are preferably dichloromethane, chloroform, and 1,2-dichloroethane.
  • the reaction temperature is 0-35°C.
  • the base required for the reaction is imidazole, N,N-diethylethylamine, triethylamine, pyridine (all can be obtained commercially), preferably imidazole.
  • the compound of formula 4-1a-1 is removed from isopropylidene in formic acid aqueous solution (concentration is 80%-90%, w/w) or trifluoroacetic acid aqueous solution (concentration is 80%-90%, w/w) Reaction of the methyl protecting group affords compounds of formula 1-2.
  • the reaction temperature is 20-80°C (refer to WO 2019/113462).
  • the intermediate compound is a compound of Formula 2a:
  • the intermediate compound is a compound of Formula 2a-1:
  • the intermediate compound is a compound of Formula 4a:
  • the intermediate compound is a compound of formula 4a-1:
  • the intermediate compound is a compound of Formula 2-1a:
  • m, n and Z are as defined above for compounds of formula I;
  • A is chloro or nitro;
  • B is hydrogen, chloro or nitro.
  • A is chloro or nitro and B is hydrogen.
  • the intermediate compound is a compound of formula 2-1a-1:
  • the intermediate compound is a compound of Formula 4-1a:
  • m and n are as defined above for compounds of formula I; A is chloro or nitro; B is hydrogen, chloro or nitro. Preferably, A is chloro or nitro and B is hydrogen.
  • the intermediate compound is a compound of formula 4-1a-1:
  • step 1
  • the resulting reaction solution was stirred overnight at room temperature. After the reaction was complete, it was concentrated under reduced pressure, and the residue was dissolved in ethyl acetate (400 mL). The resulting solution was washed successively with water (200mlx2), saturated aqueous sodium bicarbonate solution (200mlx2), water (200mlx2) and saturated brine (200mlx2).
  • reaction solution was cooled to 0° C., and phosphorus oxychloride (14.5 ml, 150.0 mmol) was slowly added dropwise under a nitrogen atmosphere. After the addition was complete, the temperature of the reaction solution was raised to room temperature, and the stirring was continued for 2 hours to complete the reaction, and the reaction was monitored by TLC (100% EtOAc). After the reaction was complete, water (500 mL) and ethyl acetate (400 mL) were added to the reaction solution, and stirred at room temperature for 15 minutes to quench the reaction.
  • the concentrate was recrystallized with petroleum ether to obtain an off-white solid (((3R, 4R)-5 -(2-oxo-4-(hydroxylamino)pyrimidin-1(2H)yl)-3,4-diacetoxytetrahydrofuran-2-yl)methyl(2-(hexadecyldithio)ethyl base) Phosphate (A5) (10.35 g, 70%).
  • step 1
  • the concentrate was recrystallized from petroleum ether to give 2-chlorophenyl(((3R,4R)-5-(2-oxo-4-(hydroxyamino)pyrimidin-1(2H)yl)-3 as a white solid, 4-diacetoxytetrahydrofuran-2-yl)methyl(2-(octadecyldithio)ethyl)phosphate (1A4), yield 90%.
  • step 1
  • step 1
  • reaction was carried out with reference to the method for preparing (1A3) described in step 2 of Example 2 to obtain intermediate (A3-1). Specifically, feed according to the following table:
  • reaction was carried out with reference to the method for preparing (1A4) described in step 3 of Example 2 to obtain intermediate (A4-1). Specifically, feed according to the following table:
  • the concentrate was recrystallized from petroleum ether to give white solid N,N-dimethylamino(((3R,4R)-5-(2-oxo-4-(hydroxyamino)pyrimidin-1(2H)yl)- 7.3 g of 3,4-diacetoxytetrahydrofuran-2-yl)methyl (2-(octadecyldithio)ethyl)phosphate (A4-1), yield 91%.
  • reaction solution was lowered to room temperature, left overnight at 4°C, filtered to obtain (((2R, 3S, 4R, 5S)-3,4-di Hydroxy-5-(4-hydroxyamino)-2-oxopyrimidin-1(2H)-yl)tetrahydrofuran-2-yl)methyl(2-(hexadecyldithio)ethyl)phosphate sodium salt Alkoxy) (II-1-1) white solid (5.9 g, 92%).
  • step 1
  • the reaction mixture was cooled to 0 ⁇ 5°C, diluted slowly with water (150ml) and extracted with ethyl acetate (2x250ml). The organic phase was washed with water (200ml), dried over anhydrous Na2SO4 and concentrated to give a brown oil .
  • the crude product was dissolved in methanol (200ml) and left at 0-30°C for 12 hours. The resulting solid impurity was filtered and discarded, and the filtrate was concentrated. To the concentrate was added acetonitrile (400ml), and the mixture was left at 5-15°C for 16 hours. The solid was filtered and dried at 25-30°C to afford 3-(hexadecyloxy)propan-1-ol (1-1b-1) (31.1 g, 77%) as a white solid.
  • reaction solution was cooled to 0°C, and phosphorus oxychloride (14.5ml, 150.0mmol) was slowly added dropwise under a nitrogen atmosphere. The addition was completed, and the temperature of the reaction solution was raised to room temperature, and continued to stir for 2 hours Make the reaction complete, and monitor the reaction by TLC (100%EtOAc).After the reaction is complete, add water (500mL) and ethyl acetate (400mL), and stir at room temperature for 15 minutes to quench the reaction.Separate the organic layer, and The organic phase was washed successively with water (2x100mL), 0.5N HCl (200mL) and saturated brine (2x100mL), dried over anhydrous magnesium sulfate and filtered.
  • TLC 50%EtOAc
  • the concentrate was recrystallized from petroleum ether to give 2-chlorophenyl(3-(hexadecyloxy)propyl)(((3aR,4R,6S,6aR)-6-(4-hydroxyamino) -2-oxopyrimidin-1(2H)-yl)-2,2-dimethyltetrahydrofuro[3,4-d][1,3]dioxolan-4-yl)methyl ) Phosphate (1-1A4) (34.7 g, 90%).
  • step 1
  • the reaction mixture was cooled to 0 ⁇ 5°C, diluted slowly with water (150ml) and extracted with ethyl acetate (2x250ml). The organic phase was washed with water (200ml), dried over anhydrous Na2SO4 and concentrated to give a brown oil .
  • the crude product was dissolved in methanol (200ml) and left at 0-30°C for 12 hours. The resulting solid impurity was filtered and discarded, and the filtrate was concentrated. To the concentrate was added acetonitrile (400ml), and the mixture was left at 5-15°C for 16 hours. The solid was filtered and dried at 25-30°C to give 2-(octadecyloxy)ethanol (1-1b-2) (25.3 g, 81.6%) as a white solid.
  • the concentrate was recrystallized from petroleum ether to give 2-chlorophenyl(2-(octadecyloxy)ethyl(((3aR,4R,6S,6aR)-6-(4-hydroxyamino)- 2-oxopyrimidin-1(2H)-yl)-2,2-dimethyltetrahydrofuro[3,4-d][1,3]dioxolan-4-yl)methyl) Phosphate (1-1B4) (7.3 g, 93%).
  • reaction solution was lowered to room temperature, placed overnight at 4°C, and filtered to obtain (((2R, 3S, 4R, 5S)-3,4-dihydroxy-5-(4-hydroxylamino)-2-oxopyrimidine -1(2H)-yl)tetrahydrofuran-2-yl)methyl(3-(hexadecyloxy)alkoxy)propyl)phosphate sodium salt (II-2-1) white solid (5.9g, 92%).
  • the solution was allowed to cool below 25°C for 8-24 hours, and then the mixture was cooled to 5 ⁇ 5°C for at least 1 hour.
  • the product was filtered and washed with cold 2-propanol (5 ⁇ 5°C, 30ml).
  • the white solid product was dried at 30-35°C for 25 hours.
  • the product (((2R, 3S, 4R, 5S)-3,4-dihydroxy-5-(4-hydroxyamino)-2-oxopyrimidin-1(2H)-yl)tetrahydrofuran-2-yl)methanol (2-(octadecyloxy)alkoxy)ethyl)phosphate monoammonium salt (II-2-2) (6.0 g, 92.3%).
  • Example 1 With the compound of Example 1 (which can be replaced by the compound of any other embodiment of the present disclosure or other specific compounds disclosed in Table 1) as an active ingredient, an exemplary tablet dosage form of different specifications of the present disclosure (which for immediate-release tablets).
  • the various components of the exemplary tablet dosage forms of different specifications and their functions and dosages are shown in the following Tables 2-4, and they can be prepared by a method comprising the following steps: (1) mixing a predetermined amount of each component, Thereby preparing a blend; and (2) compressing said blend by conventional compression methods to obtain said tablet dosage form.
  • Table 2 Composition of tablets comprising 20mg of the compound of Example 1 (amount per 1000 tablets)
  • Element Function Weight Compound of Example 1 active ingredient 20 Silicified microcrystalline cellulose Diluents, Adhesives, Glidants 33 Mannitol thinner, adhesive 53 Microcrystalline Cellulose and Mannitol (3:1, w/w) Thinner 16 Crospovidone disintegrant 5.0 Magnesium stearate lubricant 1.3 .
  • Table 3 Composition of tablets comprising 50mg of the compound of Example 1 (amount per 1000 tablets)
  • Table 4 comprises the composition of the tablet of the compound of 100mg embodiment 1 (per 1000 dosage)
  • Element Function Weight Compound of Example 1 active ingredient 100 Silicified microcrystalline cellulose Diluents, Adhesives, Glidants 64 Microcrystalline Cellulose and Mannitol (3:1, w/w) thinner, adhesive 28 Mannitol Thinner 90 Crospovidone disintegrant 10 Magnesium stearate lubricant 2.0 .
  • TC 50 maximum non-toxic concentration
  • IC 50 half-inhibitory concentration
  • SI Selection index
  • the cells used in the experiment were mosquito-derived cells (C6/36), African green monkey kidney cells (VeroE6), canine kidney cells (MDCK), human liver cancer cells (Huh7 cells) and human lung cancer cells (H460 cells).
  • the basic conditions of the culture of the cells are shown in Table 5 below:
  • influenza A virus H1N1 influenza SC09 strain and H3N2 influenza FJ09 strain
  • influenza B virus SC11 strain
  • Zika virus SZ01 strain
  • human coronavirus HCoV-229E strain
  • HCoV- OC43 strain HCoV- OC43 strain
  • strains B.1.1.7, B.1.351 and B.1.617.2 belonging to the novel coronavirus (severe acute respiratory syndrome coronavirus 2, 2019-nCoV).
  • influenza A virus H1N1 influenza SC09 strain and H3N2 influenza FJ09 strain
  • influenza B virus SC11 strain
  • DMEM cell complete culture fluid containing 10% fetal bovine serum, 100U/ml penicillin, 100U/ml streptomycin
  • MDCK cell maintenance fluid containing 2% fetal bovine serum, 100U/ml penicillin, 100U/ml streptomycin
  • the virus liquid was collected by pipetting, centrifuged (4000rpm, 10min, 4°C), and the supernatant was filtered (the pore size of the filter membrane was 0.2 ⁇ m), and 200 ⁇ l/tube was distributed into cryopreservation tubes for later use.
  • the subculture operation of FJ09 strain and SC11 strain is the same as above.
  • the 96-well plate Take out the 96-well plate that has grown into a single layer of cells, pour out the supernatant, add 200 ⁇ l of virus solution to the three secondary wells of the first titer, and add 180 ⁇ l of cell maintenance solution to the other wells, and draw from the previous titer Add 20 ⁇ l to the next concentration, pipette and mix well. No virus solution was added to the last three auxiliary wells, which were used as the cell control group.
  • the 96-well plate was cultured in a cell culture incubator and observed under a microscope every day until no new cytopathic (CPE) wells appeared. As long as lesions appear in the wells, it is recorded as "+", and the wells with CPE in each dilution are counted, and the TCID 50 value of the virus is calculated by the Karber method.
  • CPE cytopathic
  • the 96-well plate that reached the end point of the experiment was taken out, the original liquid in the well was discarded, and neutral red staining solution (100 ⁇ l/well) was added, and incubated in a cell culture incubator at 37°C and 5% CO 2 . After 1 h, the 96-well plate was taken out, the neutral red staining solution was discarded, and the neutral red in the wells was washed 2-3 times with PBS solution, the PBS was discarded, and acetic acid ethanol solution was added to develop color (100 ⁇ l/well). The OD value was measured at a wavelength of 544 nm using a microplate reader. Judgment result reference: the OD value of the experimental group ⁇ 90% of the OD value of the control group, the judgment is the maximum non-toxic concentration TC 0 value.
  • the 96-well plate was cultured in a cell culture incubator at 37°C and 5% CO 2 , and observed under a microscope every day until no new cell lesions appeared as the end point of the experiment. The OD value of cells in each well was determined by neutral red staining.
  • the 96-well plate that reached the end point of the experiment was taken out, the original liquid in the well was discarded, and neutral red staining solution (100 ⁇ l/well) was added, and incubated in a cell culture incubator at 37°C and 5% CO 2 . After 1 h, the 96-well plate was taken out, the neutral red staining solution was discarded, and the neutral red in the wells was washed 2-3 times with PBS solution, the PBS was discarded, and acetic acid ethanol solution was added to develop color (100 ⁇ l/well). The OD value was measured at a wavelength of 544 nm using a microplate reader. Calculate the cell viability after measuring the OD value, the formula is as follows:
  • the median cytotoxic concentration (CC 50 value) of the compound was calculated by GraphPad Prism 5.0 software.
  • the virus control wells and the normal cell control wells were each equipped with 5 auxiliary wells, and the experimental group was equipped with 3 auxiliary wells for each concentration. Place the 96-well plate in a 37°C, 5% CO2 incubator for culture. The cytopathic conditions were observed and recorded daily under an inverted microscope. The end point of the experiment was when 4 "+" cell lesions appeared in the virus control wells. After staining with neutral red and developing color with acetic acid ethanol solution, measure the OD value of the cells in each well at a wavelength of 544 nm, and calculate the inhibition rate according to the following formula:
  • Inhibition rate (%) (OD 544 value of test wells-average OD 544 value of virus control wells/(average OD 544 value of cell control wells-average OD 544 value of virus control wells) ⁇ 100%
  • GraphPad Prism 5.0 software was used to calculate the half maximal inhibitory concentration (IC 50 value) of the compound.
  • the tested compounds have inhibitory effects on the cytopathic effects caused by influenza virus SC09 strain, FJ09 strain and SC11 strain infection, especially when combined with the positive control drug EIDD- When compared with 1931, except compound NTV-118 and EIDD-1931, the activity of other tested compounds showed a stronger inhibitory effect on the cytopathy caused by virus infection.
  • C6/36 cells were used to culture Zika virus, and Vero cells were used for drug efficacy test.
  • RPMI-1640 cell culture medium containing 10% fetal bovine serum, 100U/ml penicillin, 100U/ml streptomycin
  • RPMI-1640 cell maintenance medium containing 2% fetal bovine serum, 100U/ml penicillin, 100U/ml ml streptomycin
  • DMEM cell complete culture fluid containing 10% fetal bovine serum, 100U/ml penicillin, 100U/ml streptomycin
  • DMEM cell maintenance fluid containing 2% fetal bovine serum, 100U/ml penicillin, 100U/ml streptomycin
  • Vero cells are cultured in a cell incubator at 37°C and 5% CO 2 .
  • the virus liquid was repeatedly frozen and thawed 3 times (-80°C for 30min; 37°C for thawing; -80°C for 30min), then centrifuged in a liquid state (4000rpm, 10min, 4°C), and the supernatant was filtered (the pore size of the filter membrane was 0.2 ⁇ m), dispense 200 ⁇ l/vial into cryopreservation tubes for later use.
  • Vero cells into a 96-well plate at a concentration of 3 ⁇ 105 cells/ml, inoculate 100 ⁇ l of cell suspension per well, and culture in a cell culture incubator at 37 °C, 5% CO2 for 1-2 days to grow into a single layer. Take out the small tube containing the virus solution from the -80°C refrigerator, thaw it, and dilute it 10 times with the cell maintenance solution to obtain the first virus titer.
  • the 96-well plate Take out the 96-well plate that has grown into a single layer of cells, pour out the supernatant, add 200 ⁇ l of virus solution to the three secondary wells of the first titer, and add 180 ⁇ l of cell maintenance solution to the other wells, and draw from the previous titer Add 20 ⁇ l to the next concentration, pipette and mix well. No virus solution was added to the last three auxiliary wells, which were used as the cell control group.
  • the 96-well plate was cultured in a cell culture incubator and observed under a microscope every day until no new cytopathic (CPE) wells appeared. As long as lesions appear in the wells, it is recorded as "+", and the wells with CPE in each dilution are counted, and the TCID 50 value of the virus is calculated by the Karber method.
  • CPE cytopathic
  • virus TCID 50 value was calculated using the following formula:
  • L is the log value of the lowest titer
  • d is the d power of 10 of the titer dilution
  • s is the sum of the ratio of the cell lesion wells with different titers.
  • Cytopathic ratio recording method ⁇ 25% + 25%-50% ++ 50%-75% +++ >75% ++++ .
  • TCID 50 10 -5.83 (calculated based on 180 ⁇ l of virus liquid).
  • the 96-well plate that reached the end point of the experiment was taken out, the original liquid in the well was discarded, and neutral red staining solution (100 ⁇ l/well) was added, and incubated in a cell culture incubator at 37°C and 5% CO 2 . After 1 h, the 96-well plate was taken out, the neutral red staining solution was discarded, and the neutral red in the wells was washed 2-3 times with PBS solution, the PBS was discarded, and acetic acid ethanol solution was added to develop color (100 ⁇ l/well). The OD value was measured at a wavelength of 544 nm using a microplate reader.
  • Judgment result reference the OD value of the experimental group ⁇ 90% of the OD value of the control group, the judgment is the maximum non-toxic concentration TC 0 value.
  • the cell state will be inhibited or cytopathic phenomenon will be found, and the maximum non-toxic concentration range needs to be determined again.
  • the 96-well plate was cultured in a cell culture incubator at 37°C and 5% CO 2 , and observed under a microscope every day until no new cell lesions appeared as the end point of the experiment. The OD value of cells in each well was determined by neutral red staining.
  • the 96-well plate that reached the end point of the experiment was taken out, the original liquid in the well was discarded, and neutral red staining solution (100 ⁇ l/well) was added, and incubated in a cell culture incubator at 37°C and 5% CO 2 . After 1 h, the 96-well plate was taken out, the neutral red staining solution was discarded, and the neutral red in the wells was washed 2-3 times with PBS solution, the PBS was discarded, and acetic acid ethanol solution was added to develop color (100 ⁇ l/well). The OD value was measured at a wavelength of 544 nm using a microplate reader. Calculate the cell viability after measuring the OD value, the formula is as follows:
  • the median cytotoxic concentration (CC 50 value) of the drug was calculated by GraphPad Prism5.0 software.
  • the cytotoxicity results of the tested compounds recorded in the above table show that the compounds NTV-16, NTV-29, NTV-37 and NTV-118 are compared with the positive control drug EIDD-1931, and the tested compound cytotoxicity is less than Or the equivalent of EIDD-1931.
  • TCID 50 of Zika virus 10 ⁇ 5.83 .
  • Each of the virus control wells and the normal cell control wells had 5 auxiliary wells, and the experimental group had 5 auxiliary wells for each concentration. Place the 96-well plate in a 37°C, 5% CO2 incubator for culture. The cytopathic conditions were observed and recorded daily under an inverted microscope. The end point of the experiment was when 4 "+" cell lesions appeared in the virus control wells. After staining with neutral red and developing color with acetic acid ethanol solution, measure the OD value of the cells in each well at a wavelength of 544 nm, and calculate the inhibition rate according to the following formula:
  • Inhibition rate (%) (OD 544 value of test wells-average OD 544 value of virus control wells)/(average OD 544 value of cell control wells-average OD 544 value of virus control wells) ⁇ 100%
  • IC 50 value The half maximal inhibitory concentration (IC 50 value) of the compound was calculated by GraphPad Prism5.0 software.
  • HCoV-229E strain was passaged in Huh7 cells and stored in a -80°C refrigerator.
  • the HCoV-OC43 strain was passaged in H460 cells and stored in a -80°C refrigerator.
  • the 96-well plate was cultured in a cell culture incubator at 37°C and 5% CO 2 , and observed under a microscope every day until no new cell lesions appeared as the end point of the experiment. The OD value of cells in each well was determined by neutral red staining.
  • the 96-well plate that reached the end point of the experiment was taken out, the original liquid in the well was discarded, and neutral red staining solution (100 ⁇ l/well) was added, and incubated in a cell culture incubator at 37°C and 5% CO 2 . After 1 h, the 96-well plate was taken out, the neutral red staining solution was discarded, and the neutral red in the wells was washed 2-3 times with PBS solution, the PBS was discarded, and acetic acid ethanol solution was added to develop color (100 ⁇ l/well). The OD value was measured at a wavelength of 544 nm using a microplate reader. Calculate the cell viability after measuring the OD value, the formula is as follows:
  • the median cytotoxic concentration (CC 50 value) of the drug was calculated with GraphPad Prism 5.0 software.
  • Huh7 cells and H460 cells were seeded in 96-well plates at a concentration of 1.5x10 4 cells/well, respectively. After overnight culture, the Huh7 cells and H460 cells in the 96-well plate were infected with 100 TCID 50 of the HCoV-229E strain and the HCoV-OC43 strain virus liquid respectively.
  • A drug concentration with cumulative inhibition rate ⁇ 50%
  • C cumulative inhibition rate ⁇ 50% inhibition rate
  • D log dilution factor
  • African green monkey kidney cells (VeroE6) were cultured in DMEM medium containing 10% fetal bovine serum (FBS), glutamine and penicillin-streptomycin at 37°C in a 5% CO 2 incubator. After the cells grew to a confluent monolayer, the culture medium was sucked off, washed 2-3 times with PBS, and then the virus solution was added and incubated at 37°C for 1 hour. After aspirating the culture solution, add DMEM culture solution containing 2% FBS and cultivate until the cytopathic effect reaches >75% of the cell monolayer destruction. Virus cultures were harvested and stored at -80°C for future use.
  • FBS fetal bovine serum
  • glutamine glutamine
  • penicillin-streptomycin penicillin-streptomycin
  • VeroE6 cells (at a density of 2-3 ⁇ 10 4 cells/well) in a 96-well cell culture plate and culture overnight. After the cells are covered with a monolayer, wash with PBS for 2 to 3 times, add 100 ⁇ l of virus solution of different dilutions (10 -1 to 10 -10 ) to each well, and set 6 to 8 duplicate wells for each dilution. and cultured in a 37°C, 5% CO2 incubator. Observe and record the cytopathic situation every day until no new cytopathic wells appear.
  • the TCID 50 of virus liquid was calculated by Reed-Muench or Karber method.
  • CC 50 median cytotoxic concentration
  • the CC50 of the drug is calculated using the following formula:
  • Inhibition rate (IC): (%) (OD 540 value of test wells-average OD 540 value of virus control wells)/(average OD 540 value of cell control wells-average OD 540 value of virus control wells) ⁇ 100%
  • GraphPad Prism 5.0 software was used to calculate the half inhibitory concentration (IC 50 value) of the drug.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Virology (AREA)
  • Molecular Biology (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Oncology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Communicable Diseases (AREA)
  • Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Biotechnology (AREA)
  • Pulmonology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

本公开涉及抗病毒化合物及其用途。具体而言,本公开涉及一种具有广谱抗病毒活性的β-D-N 4-羟基胞苷的磷酸酯化合物,涉及包含这样的化合物的药物组合物,以及这样的化合物和包含这样的化合物的药物组合物用于抑制病毒或者用于在有此需要的对象中预防和/或治疗病毒感染的用途。

Description

抗病毒化合物及其用途 技术领域
本公开涉及一种具有广谱抗病毒活性的β-D-N 4-羟基胞苷的磷酸酯化合物,涉及包含这样的化合物的药物组合物,以及这样的化合物和包含这样的化合物的药物组合物用于抑制病毒或者用于在有此需要的对象中预防和/或治疗病毒感染的用途。
背景技术
病毒感染是主要的全球公共卫生问题。病毒通过多种途径侵入机体,并在易感的宿主细胞中增殖,从而导致宿主患病。β-D-N 4-羟基胞苷(NHC,EIDD-1931)是一种胞嘧啶核苷(即胞苷)的衍生物,具有如下结构式:
Figure PCTCN2022141023-appb-000001
该物质已经被发现为具有广谱的抗病毒活性(参见,例如Stuyver等人,Antimicrob Agents Chemother,2003,47(1):244-54;Purohit等人,J Med Chem,2012,55(22):9988-9997;Ivanov等人,Collection ofCzechoslovak Chem Commun,2006,71(7):1099-1106和Fox等人,JACS,1959,81:178-870)。但是EIDD-1931具有强亲水性,这导致其生物利用度差,不适合作为先导化合物用于临床药物的开发。
WO 2019/113462公开了一种作为EIDD-1931的前药的N 4-羟基胞苷及衍生物,其中代表性的化合物为EIDD-1931的异丁酸酯EIDD2801,具有如下结构式:
Figure PCTCN2022141023-appb-000002
与EIDD-1931相比,EIDD2801的口服生物利用度有一些改善,但也存在因在体内被胃肠道的消化酶(例如但不限于水解酶)和胃酸降解而影响体内吸收的问题。与此同时,EIDD2801本身没有生物活性,吸收进入体内的EIDD2801只有经水解而暴露出活性结构EIDD-1931后才能发挥作用,这使得EIDD2801的体内活性受到水解的速率和效率的很大限制。
因此,本领域仍然需要新颖的并且具有广谱抗病毒活性的基于EIDD-1931的额外的化合物。
发明内容
本公开提供了新颖的并且具有广谱抑制病毒复制活性的β-D-N 4-羟基胞苷的磷酸酯化合物(以下简称为本公开的化合物)。
因此,在本公开的第一个方面中,提供了式I的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体:
Figure PCTCN2022141023-appb-000003
其中,
m是6-20中的任一整数;
n是1-5中的任一整数;
X是-O-或-NR 0-;
R 0是H或C 1-C 6烷基;
Z是-O-或-S-S-;
R是H、C 1-C 6烷基、C 6-C 12芳基、R′-O-C(O)-O-CH 2-或R′-C(O)-O-CH 2-;
R′是C 1-C 6烷基、C 3-C 8环烷基、C 6-C 12芳基、含有1或2个独立地选自N、O或S的杂原子的3至12元杂环基或含有1或2个独立地选自N、O或S的杂原子的5至12元杂芳基;
所述R或R′基团中的烷基、环烷基、芳基、杂环基或杂芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
在该方面的一些实施方案中,所述式I的化合物具有式I-1的结构:
Figure PCTCN2022141023-appb-000004
其中变量m、n、X和R如上面关于式I的化合物所定义。
在该方面的另一些实施方案中,所述式I的化合物具有式I-2的结构:
Figure PCTCN2022141023-appb-000005
其中变量m、n、X和R如上面关于式I的化合物所定义。
在该方面的一些实施方案中,所述式I、I-1或I-2中的任一种的化合物的药学上可接受的盐是碱金属盐(例如锂、钠、钾、铷、铯盐)、碱土金属盐(例如镁、钙、锶、钡盐)、铝盐、有机碱盐(例如乙醇胺、二乙醇胺、三乙醇胺、氨丁三醇、N-甲基葡糖胺、二乙胺、二乙基氨基乙醇、乙二胺、咪唑、吗琳、2-羟基乙基吗琳、二苄基乙二胺、三甲胺、哌啶、吡咯烷、苄胺、四甲基氢氧化胺盐)或氨基酸盐(例如赖氨酸、精氨酸盐)。
在本公开的第二个方面中,提供了药物组合物,其包含式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体以及一种或多种药学上可接受的载体。
在该方面的一些实施方案中,所述药物组合物包含两种、三种或更多种药学上可接受的载体。
在该方面的一些实施方案中,所述药物组合物被制成药学上可接受的剂型(例如,通过按照药学领域的常规生产方法,使式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体与一种或多种药学上可接受的载体混合并制成药学上可接受的剂型)。
在本公开的第三个方面中,提供了式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体或包含式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体以及一种或多种药学上可接受的载体的药物组 合物,其用于治疗或者预防疾病。
在本公开的第四个方面中,提供了式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体或包含式I、I-1或1-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体以及一种或多种药学上可接受的载体的药物组合物,其用于抑制病毒或者用于在有此需要的对象中预防和/或治疗病毒感染。
在本公开的第五个方面中,提供了式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体或包含式I、I-1或1-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体以及一种或多种药学上可接受的载体的药物组合物用于抑制病毒或者用于在有此需要的对象中预防和/或治疗病毒感染的用途。
在本公开的第六个方面中,提供了式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体或包含式I、I-1或1-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体以及一种或多种药学上可接受的载体的药物组合物在制备用于抑制病毒或者用于在有此需要的对象中预防和/或治疗病毒感染的药物中的用途。
在本公开的第七个方面中,提供了用于在有此需要的患者中预防和/或治疗病毒感染的方法,所述方法包括向有此需要的对象施用有效量的式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体或包含式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体以及一种或多种药学上可接受的载体的药物组合物。
在本公开的第四、五、六、七方面的一些实施方案中,所述病毒是冠状病毒(例如人冠状病毒,诸如严重急性呼吸综合征冠状病毒(SARS-CoV)、中东呼吸综合征冠状病毒(MERS-CoV)或严重急性呼吸 综合征冠状病毒2(SARS-CoV-2,包括例如B.1.1.7株、B.1.351株、德尔塔变异株B.1.617.2株、奥密克戎变异株B.1.1.529株或其它SARS-CoV-2变异株)、马脑炎病毒(例如东部马脑炎病毒、西部马脑炎病毒或委内瑞拉马脑炎病毒)、奇昆古尼亚病毒、罗斯河病毒、正粘病毒科病毒(例如流感病毒,诸如甲型流感病毒、乙型流感病毒或丙型流感病毒)、副粘病毒科病毒(例如呼吸道合胞病毒)、丝状病毒(例如埃博拉病毒、马尔堡病毒或库瓦病毒)或寨卡病毒。
在本公开的第八个方面中,提供了用于合成本公开的式I、I-1或I-2中的任一种的化合物的中间体化合物。
在一些实施方案中,所述中间体化合物是式2a的化合物:
Figure PCTCN2022141023-appb-000006
其中m、n和Z如上面关于式I的化合物所定义;R 1是直链或直链的C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基或C 6-C 12芳基,所述烷基、烯基、炔基或芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基;A是氯或硝基;B是氢、氯或硝基。优选地,R 1是甲基、乙基或苯基,A是氯或硝基,B是氢。
在一些实施方案中,所述中间体化合物是式2a-1的化合物:
Figure PCTCN2022141023-appb-000007
其中m、n、Z、R 0和R如上面关于式I的化合物所定义;R 1是直链或直链的C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基或C 6-C 12芳基,所述烷基、烯基、炔基或芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。优选地,R 1是甲基、乙基或苯基。
在一些实施方案中,所述中间体化合物是式4a的化合物:
Figure PCTCN2022141023-appb-000008
其中m、n和Z如上面关于式I的化合物所定义;R 1是直链或直链的C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基或C 6-C 12芳基,所述烷基、烯基、炔基或芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、 氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基;A是氯或硝基;B是氢、氯或硝基。优选地,R 1是甲基、乙基或苯基,A是氯或硝基,B是氢。
在一些实施方案中,所述中间体化合物是式4a-1的化合物:
Figure PCTCN2022141023-appb-000009
其中m、n、Z、R 0和R如上面关于式I的化合物所定义;R 1是直链或直链的C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基或C 6-C 12芳基,所述烷基、烯基、炔基或芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。优选地,R 1是甲基、乙基或苯基。
在一些实施方案中,所述中间体化合物是式2-1a的化合物:
Figure PCTCN2022141023-appb-000010
其中m、n和Z如上面关于式I的化合物所定义;A是氯或硝基;B是氢、氯或硝基。优选地,A是氯或硝基,B是氢。
在一些实施方案中,所述中间体化合物是式2-1a-1的化合物:
Figure PCTCN2022141023-appb-000011
其中m、n、Z、R 0和R如上面关于式I的化合物所定义。
在一些实施方案中,所述中间体化合物是式4-1a的化合物:
Figure PCTCN2022141023-appb-000012
其中m和n如上面关于式I的化合物所定义;A是氯或硝基;B是氢、氯或硝基。优选地,A是氯或硝基,B是氢。
在一些实施方案中,所述中间体化合物是式4-1a-1的化合物:
Figure PCTCN2022141023-appb-000013
其中m、n、Z、R 0和R如上面关于式I的化合物所定义。
具体实施方式
定义
应理解,除非本文另有具体地定义,否则本文使用的术语应赋予其在本领域中的公认含义。进一步地,应理解,本文使用的术语仅意图用于描述具体的实施方案,而非意图对本发明的保护范围进行任何限制。
除非另有说明,否则当本文中描述的化合物的结构式和化学名称存在不一致时,以结构式为准。
除非另有说明,否则本文中使用的单数形式“一个”、“一种”和“所述”意图包括复数形式。
除非另有说明,否则术语“包含(或其等同术语例如“包括”、“含有”等)”或“基本上由......组成”包括了“由......组成”的情况。
除非另有说明,否则本公开中记载的范围都包括该范围的端点以及该范围中所包含的每个整数。例如,“m是6-20中的任一整数”表示m可以是6、7、8、9、10、11、12、13、14、15、16、17、18、19或20。再例如,“n是1-5中的任一整数”表示n可以是1、2、3、4和5。另外,由这些整数所构成的任意子范围都意图包括在本公开的范围内。
本文中使用的术语“本公开的化合物”是指落入式I、I-1或I-2中的任一种的范围内的任意一种或多种化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体。
基团术语的前缀“C x-C y”是指该基团所包含的碳原子数目的范围(该范围的端点以及该范围中所包含的每个整数以及由这些整数所构成的任意子范围都意图包括在本公开的范围内)。例如,“C 1-C 6烷基”表示包含1至6个碳原子(例如,1、2、3、4、5或6个碳原子)的烷基。
本文中使用的术语“烷基”是指具有指定碳原子数的直链或支链饱和一价烃基。烷基通常含有1-6个碳原子(“C 1-C 6烷基”),更优选1-5个碳原子(“C 1-C 5烷基”)、1-4个碳原子(“C 1-C 4烷基”)或1-2个 碳原子(“C 1-C 2烷基”)。优选的C 1-C 6烷基的例子包括甲基、乙基、正丙基、异丙基、正丁基、仲丁基、异丁基、叔丁基、正戊基、异戊基、新戊基和正己基。优选的C 1-C 4烷基的例子包括甲基、乙基、正丙基、异丙基、正丁基、仲丁基、异丁基和叔丁基。
本文中使用的术语“烯基”是指包含一个或多个双键的具有指定碳原子数的直链或支链不饱和一价烃基。烯基通常含有2-6个碳原子(“C 2-C 6烯基”),更优选2-5个碳原子(“C 2-C 5烯基”)、2-4个碳原子(“C 2-C 4烯基”)或2个碳原子(“乙烯基”)。优选的C 2-C 6烯基包括乙烯基、1-丙烯基、2-丙烯基、1-丁烯基、2-丁烯基、1,3-丁二-1-烯基、1-戊烯-3-基、2-戊烯-1-基、3-戊烯-1-基、3-戊烯-2-基、1,3-戊二烯-1-基、1,4-戊二烯-3-基、1-己烯-3-基和1,4-己二烯-1-基。
本文中使用的术语“炔基”是指包含一个或多个三键的具有指定碳原子数的直链或支链不饱和一价烃基。炔基通常含有2-6个碳原子(“C 2-C 6炔基”),更优选2-5个碳原子(“C 2-C 5炔基”)、2-4个碳原子(“C 2-C 4炔基”)或2个碳原子(“乙炔基”)。优选的C 2-C 6炔基包括乙炔基、丙炔基、丁炔基、戊炔基和己炔基。
本文中使用的术语“烷氧基”表示通过氧原子与母体分子连接的烷基(即“-O-烷基”),其中“烷基”如前文所定义。烷氧基通常含有1-6个碳原子(“C 1-C 6烷氧基”),更优选1-4个碳原子(“C 1-C 4烷氧基”)。例如,C 1-C 4烷氧基包括甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基和叔丁氧基等。
本文中使用的术语“卤素”是指氟、氯、溴、碘等,优选氟原子和氯原子。
本文中使用的术语“卤代”是指取代基中的一个或多个氢原子被一个或多个相同或不同的卤素原子取代。“卤素”如前文所定义。例如,“卤代C 1-C 6烷基”是指一个或多个氢原子被一个或多个相同或不同的卤素原子取代的“C 1-C 6烷基”,其中“C 1-C 6烷基”如前文所定义。再例如,“卤代C 1-C 6烷氧基”是指一个或多个氢原子被一个或多 个相同或不同的卤素原子取代的“C 1-C 6烷氧基”,其中“C 1-C 6烷氧基”如前文所定义。
本文中使用的术语“氰基”是指-CN基团。
本文中使用的术语“羟基”是指-OH基团。
本文中使用的术语“硝基”是指-NO 2基团。
本文中使用的术语“芳基”是指衍生自具有公知的芳香性特征的单环或稠合二环或多环环系(其中至少一个环含有完全共轭的π-电子系统)的一价烃基。稠合芳基可以包括与饱和的或部分不饱和的碳环或杂环稠合或与另一个芳基或杂芳基环稠合的芳基环,前提条件是,在这样的稠合环系统上与母体分子的连接点是环系统的芳族部分的原子。通常,芳基含有6-12个碳原子(“C 6-C 12芳基”)。芳基的例子包括但不限于苯基、萘基、蒽基、菲基、茚满基、茚基和四氢萘基。
本文中使用的术语“环烷基”是指衍生自含有指定数目的碳原子的非芳族饱和碳环环系的一价烃基,其可以是通过环烷基环的碳原子连接至母体分子的单环、螺环、桥连或稠合二环或多环环系。通常,本公开的环烷基含有3-8个碳原子(“C 3-C 8环烷基”),优选地3-7个碳原子(“C 3-C 7环烷基”)或3-6个碳原子(“C 3-C 6环烷基”)。环烷基的代表性例子包括环丙基、环丁基、环戊基、环己基、环庚基和环辛基等。
本文中使用的术语“杂原子”是指N、O或S原子。
本文中使用的术语“杂环基”是指衍生自含有指定数目的碳原子并且还包括至少一个杂原子,优选一至四个杂原子,更优选一个或两个杂原子作为环成员的非芳族环结构的一价基团,其中所述杂原子是指N、O或S原子且S原子任选地被一个或两个氧代基团取代(即,S(O) q,其中q是0、1或2)。这样的杂环基可以是部分不饱和的。杂环基包括与一个或多个其它杂环或碳环形成的螺环、桥连环或稠合环,其中这样的螺环、桥连环或稠合环本身可以是饱和的、部分不饱和的或芳族的,前提条件是,与母体分子的连接点是这样的环系统的杂环部分的 原子。通常,“杂环基”含有3至12个环原子(即3至12元杂环基),优选含有4至7个环原子(即4至7元杂环基),最优选含有5或6个环原子(即5或6元杂环基),其中所述环原子包括碳和非碳杂原子。在某些实施方案中,杂环基的代表性例子包括氮杂环丁基、氧杂环丁基、硫杂环丁基、吡咯烷基、咪唑烷基、吡唑烷基、噁唑烷基、噻唑烷基、二氢噁唑基、二氢异噁唑基、二氢吡咯基、二氢咪唑基、二氢吡唑基、二氢噻唑基、哌啶基、二氢吡啶基、二氢嘧啶基、哌嗪基、二氧杂环己烷基、氧硫杂环己烷基、氮杂环庚烷基、二氮杂环庚烷基、氧杂环丁基、四氢呋喃基、四氢吡喃基、四氢噻吩基、四氢噻喃基、吗啉基和硫代吗啉基等。
本文中使用的术语“杂芳基”是指衍生自含有指定数目的碳原子并且还包括至少一个杂原子,优选一至四个杂原子,更优选一个或两个杂原子作为环成员的具有芳族环结构的一价基团,其中所述杂原子是指N、O或S原子。通常,杂芳基含有5-12个环原子(“5-12元杂芳基”),优选5-10个环原子(“5-10元杂芳基”),更优选5或6个环原子(“5或6元杂芳基”),其中所述环原子包括碳和非碳杂原子。杂芳基通过杂芳环的环原子连接至母体分子,从而保持芳香性。杂芳基也可以与另一个芳基或杂芳基环稠合,或与饱和的或部分不饱和的碳环或杂环稠合,前提条件是,在这样的稠合环系统上与基础分子的连接点是环系统的杂芳族部分的原子。在某些实施方案中,杂芳基的代表性例子包括呋喃基、咪唑基、异噁唑基、噻唑基、异噻唑基、噁二唑基、噁唑基、吡啶基、哒嗪基、嘧啶基、吡嗪基、吡唑基、吡咯基、四唑基、噻二唑基、噻吩基、三唑基、三嗪基、苯并咪唑基、苯并呋喃基、苯并噻吩基、苯并噁二唑基、苯并噻二唑基、苯并噻唑基、咪唑并吡啶基、咪唑并嘧啶基、咪唑并哒嗪基、噌啉基、呋喃并吡啶基、吲唑基、吲哚基、异吲哚基、异喹啉基、萘啶基、嘌呤基、喹啉基、噻吩并吡啶基等。
本文中使用的术语“任选地”是指该术语后面所紧接着描述的情 况可以发生,但也可以不发生。例如,“所述R或R′基团中的烷基、环烷基、芳基、杂环基或杂芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代”涵盖“所述R或R′基团中的烷基、环烷基、芳基、杂环基或杂芳基被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代”或者“所述R或R′基团中的烷基、环烷基、芳基、杂环基或杂芳基不被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代”两种情况。
本文中使用的术语“药学上可接受的”是指在合理的医学判断的范围,适合用于与对象例如人或其它哺乳动物的组织接触而不产生过度毒性、刺激、过敏反应或其它问题,同时具有相称的合理受益/风险比的那些化合物或药物组合物。
本文中使用的术语“药学上可接受的盐”是指式I、I-1或1-2中的任一种的化合物与药学上可接受的酸或碱形成的酸或碱加成盐,其保留母体化合物即式I、I-1或1-2中的任一种的化合物的生物有效性和性质。所述药学上可接受的盐包括但不限于碱金属盐(例如锂、钠、钾、铷、铯盐)、碱土金属盐(例如镁、钙、锶、钡盐)、铝盐、有机碱盐(例如乙醇胺、二乙醇胺、三乙醇胺、氨丁三醇、N-甲基葡糖胺、二乙胺、二乙基氨基乙醇、乙二胺、咪唑、吗琳、2-羟基乙基吗琳、二苄基乙二胺、三甲胺、哌啶、吡咯烷、苄胺、四甲基氢氧化胺盐)或氨基酸盐(例如赖氨酸、精氨酸盐)。本公开的式I、I-1或1-2中的任一种的化合物 的药学上可接受的盐可以如下制备:将等量的式I、I-1或I-2中的任一种的化合物与提供所需抗衡离子的碱在溶剂中于25-70℃搅拌0.5-2h,然后减压蒸去溶剂,得到所述药学上可接受的盐。
本文中使用的术语‘溶剂合物’是指包含式I、I-1或I-2中的任一种的化合物和一种或多种药学上可接受的溶剂分子(例如,乙醇)的分子络合物。当溶剂是水时,采用术语“水合物”。
本文中使用的术语“立体异构体”是指由分子中的原子在空间上排列方式不同所产生的异构体。当化合物存在不对称碳原子时,会产生对映异构体;当化合物存在碳碳双键或环状结构时,会产生顺反异构体。在本公开范围中包括所有式I、I-1或I-2中的任一种的化合物的对映异构体、非对映异构体、外消旋体、顺反异构体、几何异构体、差向异构体及其混合物。
本文中使用的术语“药物组合物”是指由药物活性成分(在本公开的背景下,特指式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物和立体异构体)和药学上可接受的载体按一定比例组合而成的具有特定医学用途的物质或材料。通过本领域任何常规技术,可以将药物组合物制成药学上可接受的剂型的形式,诸如片剂、粉剂(包括注射用无菌粉末)、胶囊剂、颗粒剂、溶液剂、糖浆剂、栓剂、注射剂、贴剂等。可以通过多种施用途径中的任何一种将本公开的药物组合物施用于对象(例如人或非人哺乳动物),所述施用途径包括例如口服(例如,以片剂、胶囊剂、粉剂、颗粒剂的形式);经粘膜(例如舌下、鼻、肛门、直肠或阴道)吸收(例如,以栓剂、乳膏或泡沫的形式);肠胃外(例如,肌肉内、静脉内、腹膜内、皮下或鞘内注射);透皮(例如,作为施加到皮肤的贴片);以及局部施用(例如,作为施加于皮肤的乳膏、软膏、喷雾剂或作为滴眼剂)。该药物组合物也可以配制成用于吸入施用。一般而言,药物组合物通常含有0.1重量%-99.5重量%的活性成分(即式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体),优选含有0.5重量%-99.5 重量%,例如1重量%、1.5重量%、2重量%、5重量%、10重量%、15重量%、20重量%、25重量%、30重量%或50重量%的活性成分。
本文中使用的术语“药学上可接受的载体”表示不会对生物体造成明显刺激并且不会消除所施用的化合物的生物活性和性质的载体(或称赋形剂)。可以使用任何常用的药学上可接受的载体,其选择取决于例如特定施用模式、载体对溶解度和稳定性的影响、以及剂型的性质等因素并且在本领域技术人员的普通技能之内。一些可用作药学上可接受的载体的材料的实例包括:淀粉,诸如玉米淀粉和马铃薯淀粉;糖,诸如乳糖、葡萄糖和蔗糖;纤维素及其衍生物,诸如乙基纤维素、羧甲基纤维素钠和醋酸纤维素;明胶、阿拉伯胶、瓜尔胶、西黄蓍胶;硬脂酸镁、硬脂酸锌、滑石;水、盐水;油,诸如花生油、棉籽油、橄榄油、芝麻油、玉米油和大豆油;醇,诸如乙醇、丙二醇、甘油、山梨糖醇、甘露糖醇和聚乙二醇;酯,诸如油酸乙酯和月桂酸乙酯;缓冲剂,诸如氯化钠、磷酸盐缓冲溶液等。
本文中使用的术语“预防”是指防止有患病风险的对象中的疾病的出现或者已消失的疾病的复发。
本文中使用的术语“治疗”是指控制、减轻或缓解疾病的病理学进展和延长患病对象的存活期。
本文中使用的术语“对象”是指预期要对其施用本公开的化合物或药物组合物的动物个体,其包括但不限于人和/或其它灵长类动物(例如,食蟹猴、恒河猴);其它哺乳动物,例如马、牛、猪、绵羊、山羊、猫、狗;和禽类,如鸡、鸭、鹅、鹌鹑、火鸡。优选的对象是人。
本文中使用的术语“有效量”是指足以影响疾病、其并发症或在疾病的发展过程中呈现出的中间病理学表型的任何一种或多种有益的或期望的症状的量。所施用的式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体的“有效量”将取决于接受治疗的对象的种属、疾病的严重程度、施用频率、药物的代谢特征等因素,并可由处方医师根据常规实践来判断。一般而言, 有效量通常在约0.001至约100mg/千克体重/天的范围内,优选约0.01至约50mg/千克体重/天(呈单剂量或分份剂量)。在某些情况下,在上述范围的下限之下的剂量水平可能是绰绰有余的,而在其它情况下,可能需要使用更大的剂量而不引起任何有害的副作用,其中这样的更大的剂量通常分成数个用于在一天中施用的更小剂量。需要注意的是,本公开中提及的所有数值范围均表示包括该范围的两个端点、该范围之内的所有整数以及由这些整数形成的子范围。
本公开技术方案的详细描述
提供以下具体实施方案以使得本领域技术人员能够更清楚地理解本公开的内容。需要指出的是,这些实施方案仅仅为举例说明的目的而记载,并非是对本申请的保护范围进行限制。
本公开一般性地涉及一种具有广谱抗病毒活性的β-D-N 4-羟基胞苷的磷酸酯化合物,涉及包含这样的化合物的药物组合物,以及这样的化合物和包含这样的化合物的药物组合物用于抑制病毒或者用于在有此需要的对象中预防和/或治疗病毒感染的用途。
在本公开的第一个方面中,提供了式I的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体:
Figure PCTCN2022141023-appb-000014
其中变量m、n、X、Z和R如上面关于式I的化合物所定义。
在该方面的一些实施方案中,所述式I的化合物具有式I-1的结构:
Figure PCTCN2022141023-appb-000015
其中变量m、n、X和R如上面关于式I的化合物所定义。
在该方面的另一些实施方案中,所述式I的化合物具有式I-2的结构:
Figure PCTCN2022141023-appb-000016
其中变量m、n、X和R如上面关于式I的化合物所定义。
在一些实施方案中,m是6-20中的任一整数。
在一些实施方案中,m是6、7、8、9、10、11、12、13、14、15、16、17、18、19或20。
在一些实施方案中,n是1-5中的任一整数。
在一些实施方案中,n是1、2、3、4或5。
在一些实施方案中,X是-O-或-NR 0-。
在一些实施方案中,X是-O-。
在一些实施方案中,X是-NR 0-。
在一些实施方案中,R 0是H或C 1-C 6烷基。
在一些实施方案中,R 0是H。
在一些实施方案中,R 0是C 1-C 6烷基。
在一些实施方案中,R 0是甲基、乙基、正丙基、异丙基、正丁基、仲丁基、异丁基或叔丁基。
在一些实施方案中,R 0是甲基或乙基。
在一些实施方案中,R 0是甲基。
在一些实施方案中,Z是-O-或-S-S-。
在一些实施方案中,Z是-O-。
在一些实施方案中,Z是-S-S-。
在一些实施方案中,R是H、C 1-C 6烷基、C 6-C 12芳基、R′-O-C(O)-O-CH 2-或R′-C(O)-O-CH 2-,其中所述R基团中的烷基或芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
在一些实施方案中,R是H。
在一些实施方案中,R是C 1-C 6烷基,其中所述烷基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
在一些实施方案中,R是甲基、乙基、正丙基、异丙基、正丁基、仲丁基、异丁基或叔丁基。
在一些实施方案中,R是甲基或乙基。
在一些实施方案中,R是甲基。
在一些实施方案中,R是C 6-C 12芳基,其中所述芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是 H或C 1-C 6烷基。
在一些实施方案中,R是苯基,其中所述苯基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
在一些实施方案中,R是苯基,其中所述苯基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基或硝基的取代基取代。
在一些实施方案中,R是苯基,其中所述苯基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基或硝基的取代基取代。
在一些实施方案中,R是苯基,其中所述苯基任选地被1个独立地选自卤素、卤代C 1-C 6烷基、卤代C 1-C 6烷氧基、氰基或硝基的取代基取代。
在一些实施方案中,R是任选地被1个独立地选自卤素或硝基的取代基取代的苯基。
在一些实施方案中,R是任选地被1个独立地选自氯或硝基的取代基取代的苯基。
在一些实施方案中,R是R′-O-C(O)-O-CH 2-。
在一些实施方案中,R是R′-C(O)-O-CH 2-。
在一些实施方案中,R′是C 1-C 6烷基、C 3-C 8环烷基、C 6-C 12芳基、含有1或2个独立地选自N、O或S的杂原子的3至12元杂环基或含有1或2个独立地选自N、O或S的杂原子的5至12元杂芳基,其中所述R′基团中的烷基、环烷基、芳基、杂环基或杂芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6 炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
在一些实施方案中,R′是C 1-C 6烷基,其中所述烷基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
在一些实施方案中,R′是甲基、乙基、正丙基、异丙基、正丁基、仲丁基、异丁基或叔丁基。
在一些实施方案中,R′是甲基、乙基、异丙基或叔丁基。
在一些实施方案中,R′是甲基、异丙基或叔丁基。
在一些实施方案中,R′是C 3-C 8环烷基,其中所述环烷基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
在一些实施方案中,R′是环丁基、环戊基、环己基或环庚基。
在一些实施方案中,R′是环戊基或环己基。
在一些实施方案中,R′是C 6-C 12芳基,其中所述芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
在一些实施方案中,R′是苯基,其中所述苯基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
在一些实施方案中,R′是苯基,其中所述苯基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基或硝基的取代基取代。
在一些实施方案中,R′是苯基,其中所述苯基任选地被1个独立地选自卤素、卤代C 1-C 6烷基、卤代C 1-C 6烷氧基、氰基或硝基的取代基取代。
在一些实施方案中,R′是苯基。
在一些实施方案中,R′是含有1或2个独立地选自N、O或S的杂原子的3至12元杂环基,其中所述杂环基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
在一些实施方案中,R′是含有1或2个独立地选自N、O或S的杂原子的5或6元杂环基,其中所述杂环基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
在一些实施方案中,R′是吡咯烷基、四氢呋喃基、四氢噻吩基、吡 唑烷基、咪唑烷基、噁唑烷基、异噁唑烷基、噻唑烷基、异噻唑烷基、二氢吡咯基、二氢呋喃基、二氢噻吩基、二氢吡唑基、二氢咪唑基、二氢噁唑基、异二氢噁唑基、二氢噻唑基、异二氢噻唑基、哌啶基、哌嗪基、吗啉基、硫代吗啉基、二氢吡啶基、二氢吡嗪基、二氢嘧啶基或二氢哒嗪基,其中所述基团任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
在一些实施方案中,R′是吡咯烷基、四氢呋喃基、四氢噻吩基、吡唑烷基、咪唑烷基、噁唑烷基、异噁唑烷基、噻唑烷基、异噻唑烷基、二氢吡咯基、二氢呋喃基、二氢噻吩基、二氢吡唑基、二氢咪唑基、二氢噁唑基、异二氢噁唑基、二氢噻唑基、异二氢噻唑基、哌啶基、哌嗪基、吗啉基、硫代吗啉基、二氢吡啶基、二氢吡嗪基、二氢嘧啶基或二氢哒嗪基,其中所述基团任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NH 2或-C(=O)NH 2的取代基取代。
在一些实施方案中,R′是含有1或2个独立地选自N、O或S的杂原子的5至12元杂芳基,其中所述杂芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
在一些实施方案中,R′是吡咯基、呋喃基、噻吩基、吡唑基、咪唑基、噁唑基、异噁唑基、噻唑基、异噻唑基、噁二唑基、噻二唑基、四唑基、三唑基、吡啶基、哒嗪基、嘧啶基、吡嗪基、三嗪基、苯并咪唑基、苯并呋喃基、苯并噻吩基、苯并噁二唑基、苯并噻二唑基、 苯并噻唑基、咪唑并吡啶基、咪唑并嘧啶基、咪唑并哒嗪基、噌啉基、呋喃并吡啶基、吲唑基、吲哚基、异吲哚基、异喹啉基、萘啶基、嘌呤基、喹啉基、噻吩并吡啶基,其中所述基团任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
在一些实施方案中,R′是吡咯基、呋喃基、噻吩基、吡唑基、咪唑基、噁唑基、异噁唑基、噻唑基、异噻唑基、噁二唑基、噻二唑基、四唑基、三唑基、吡啶基、哒嗪基、嘧啶基、吡嗪基、三嗪基、苯并咪唑基、苯并呋喃基、苯并噻吩基、苯并噁二唑基、苯并噻二唑基、苯并噻唑基、咪唑并吡啶基、咪唑并嘧啶基、咪唑并哒嗪基、噌啉基、呋喃并吡啶基、吲唑基、吲哚基、异吲哚基、异喹啉基、萘啶基、嘌呤基、喹啉基、噻吩并吡啶基,其中所述基团任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NH 2或-C(=O)NH 2的取代基取代。
在一些实施方案中,每个R a和R b独立地是H或C 1-C 6烷基。
在一些实施方案中,每个R a和R b独立地是H。
在一些实施方案中,每个R a和R b独立地是C 1-C 6烷基。
在一些实施方案中,每个R a和R b独立地是甲基、乙基、正丙基、异丙基、正丁基、仲丁基、异丁基或叔丁基。
在一些实施方案中,每个R a和R b独立地是甲基或乙基。
在一些实施方案中,每个R a和R b独立地是甲基。
在一些实施方案中:
m是6-20中的任一整数;
n是1-5中的任一整数;
X是-O-或-NR 0-;
R 0是H、甲基、乙基、正丙基、异丙基、正丁基、仲丁基、异丁基或叔丁基;
R是H、甲基、乙基、正丙基、异丙基、正丁基、仲丁基、异丁基、叔丁基、任选地被1个独立地选自卤素或硝基的取代基取代的苯基、R′-O-C(O)-O-CH 2-或R′-C(O)-O-CH 2-;
R′是甲基、乙基、正丙基、异丙基、正丁基、仲丁基、异丁基、叔丁基、环丁基、环戊基、环己基、环庚基或苯基。
在一些实施方案中:
m是6-20中的任一整数;
n是1-5中的任一整数;
X是-O-或-NR 0-;
R 0是H、甲基或乙基;
R是H、甲基、乙基、任选地被1个独立地选自氯或硝基的取代基取代的苯基、R′-O-C(O)-O-CH 2-或R′-C(O)-O-CH 2-;
R′是甲基、乙基、异丙基、叔丁基、环戊基、环己基或苯基。
在一些实施方案中:
m是6-20中的任一整数;
n是1-5中的任一整数;
X是-O-或-NR 0-;
R 0是H或甲基;
R是H、甲基、任选地被1个独立地选自氯或硝基的取代基取代的苯基、R′-O-C(O)-O-CH 2-或R′-C(O)-O-CH 2-;
R′是甲基、异丙基、叔丁基、环戊基、环己基或苯基。
在该方面的一些实施方案中,本公开提供了选自表1中的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体:
表1
Figure PCTCN2022141023-appb-000017
Figure PCTCN2022141023-appb-000018
Figure PCTCN2022141023-appb-000019
Figure PCTCN2022141023-appb-000020
Figure PCTCN2022141023-appb-000021
Figure PCTCN2022141023-appb-000022
Figure PCTCN2022141023-appb-000023
Figure PCTCN2022141023-appb-000024
Figure PCTCN2022141023-appb-000025
Figure PCTCN2022141023-appb-000026
Figure PCTCN2022141023-appb-000027
Figure PCTCN2022141023-appb-000028
Figure PCTCN2022141023-appb-000029
Figure PCTCN2022141023-appb-000030
Figure PCTCN2022141023-appb-000031
Figure PCTCN2022141023-appb-000032
Figure PCTCN2022141023-appb-000033
Figure PCTCN2022141023-appb-000034
Figure PCTCN2022141023-appb-000035
Figure PCTCN2022141023-appb-000036
Figure PCTCN2022141023-appb-000037
Figure PCTCN2022141023-appb-000038
在该方面的一些实施方案中,所述式I、I-1或I-2中的任一种的化合物的药学上可接受的盐是碱金属盐(例如锂、钠、钾、铷、铯盐)、碱土金属盐(例如镁、钙、锶、钡盐)、铝盐、有机碱盐(例如乙醇胺、二乙醇胺、三乙醇胺、氨丁三醇、N-甲基葡糖胺、二乙胺、二乙基氨基乙醇、乙二胺、咪唑、吗琳、2-羟基乙基吗琳、二苄基乙二胺、三甲胺、哌啶、吡咯烷、苄胺、四甲基氢氧化胺盐)或氨基酸盐(例如赖氨酸、精氨酸盐)。
在本公开的第二个方面中,提供了药物组合物,其包含式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体以及一种或多种药学上可接受的载体。
在该方面的一些实施方案中,所述药物组合物含有0.1重量%-99.5重量%的活性成分(即式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体),优选含有0.5重量%-99.5重量%,更优选1重量%-50重量%,例如1重量%、1.5重量%、2重量%、5重量%、10重量%、15重量%、20重量%、25重量%、30重量%或50重量%的活性成分。所述药物组合物的其余部分为药学可接受的载体。在该方面的一些实施方案中,所述药物组合物包含两种、三种或更多种药学上可接受的载体。所述药学上可接受的载体的实例包括但不限于药学领域常规的稀释剂、赋形剂、填充剂、粘合剂、润湿剂、崩解剂、润滑剂、着色剂、香料、吸收促进剂、表面活性剂、吸附载体等。
在该方面的一些实施方案中,所述药物组合物被制成药学上可接受的剂型。例如,在一些实施方案中,通过按照药学领域的常规生产方法,使式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体与一种或多种药学上可接受的载体混 合并制成药学上可接受的剂型。所述药学上可接受的剂型的实例包括但不限于药学领域常规的固体剂型(例如但不限于片剂、胶囊剂、粉剂、颗粒剂、扁胶囊剂和栓剂等)、液体剂型(例如但不限于溶液剂、糖浆剂、混悬剂、乳剂、酏剂等)。除此之外,可将本公开的药物组合物制成缓释剂型以提供活性成分的控速释放,从而优化抗病毒活性。适合于缓释的剂型包括但不限于含有崩解速率不同的层的多层片剂,或含有被浸渍或被包封过的多孔聚合物基质的胶囊剂。
在该方面的一些实施方案中,所述药物组合物中活性成分的含量可以在约1mg至约2000mg的范围内变化。在一些实施方案中,所述含量可以为1mg至1000mg、2mg至500mg、2.5mg至100mg或5mg至75mg。
在本公开的第三个方面中,提供了式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体或包含式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体以及一种或多种药学上可接受的载体的药物组合物,其用于治疗或者预防疾病。
在本公开的第四个方面中,提供了式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体或包含式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体以及一种或多种药学上可接受的载体的药物组合物,其用于抑制病毒或者用于在有此需要的对象中预防和/或治疗病毒感染。
在本公开的第五个方面中,提供了式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体或包含式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体以及一种或多种药学上可接受的载体的药物组合物用于抑制病毒或者用于在有此需要的对象中预防和/或治疗病毒感染的用途。
在本公开的第六个方面中,提供了式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体或包含式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体以及一种或多种药学上可接受的载体的药物组合物在制备用于抑制病毒或者用于在有此需要的对象中预防和/或治疗病毒感染的药物中的用途。
在本公开的第七个方面中,提供了用于在有此需要的患者中预防和/或治疗病毒感染的方法,所述方法包括向有此需要的对象施用有效量的式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体或包含式I、I-1或I-2中的任一种的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体以及一种或多种药学上可接受的载体的药物组合物。
在一些实施方案中,所述病毒是冠状病毒(例如人冠状病毒,诸如严重急性呼吸综合征冠状病毒(SARS-CoV)、中东呼吸综合征冠状病毒(MERS-CoV)或严重急性呼吸综合征冠状病毒2(SARS-CoV-2,包括例如B.1.1.7株、B.1.351株、德尔塔变异株B.1.617.2株、奥密克戎变异株B.1.1.529株或其它SARS-CoV-2变异株)、马脑炎病毒(例如东部马脑炎病毒、西部马脑炎病毒或委内瑞拉马脑炎病毒)、奇昆古尼亚病毒、罗斯河病毒、正粘病毒科病毒(例如流感病毒,诸如甲型流感病毒、乙型流感病毒或丙型流感病毒)、副粘病毒科病毒(例如呼吸道合胞病毒)、丝状病毒(例如埃博拉病毒、马尔堡病毒或库瓦病毒)或寨卡病毒。
在一些实施方案中,所述病毒是流感病毒。
在一些实施方案中,所述病毒是甲型流感病毒。
在一些实施方案中,所述病毒是甲型H1N1流感病毒。
在一些实施方案中,所述病毒是SC09株。
在一些实施方案中,所述病毒是甲型H3N2流感病毒。
在一些实施方案中,所述病毒是FJ09株。
在一些实施方案中,所述病毒是乙型流感病毒。
在一些实施方案中,所述病毒是SC11株。
在一些实施方案中,所述病毒是寨卡病毒。
在一些实施方案中,所述病毒是SZ01株。
在一些实施方案中,所述病毒是人冠状病毒。
在一些实施方案中,所述病毒是HCoV-229E株或HCoV-OC43株。
在一些实施方案中,所述病毒是严重急性呼吸综合征冠状病毒2。
在一些实施方案中,所述病毒是B.1.1.7株、B.1.351株或B.1.617.2株。
可以通过多种施用途径中的任何一种将本公开的化合物或药物组合物施用于对象(例如人或非人哺乳动物),所述施用途径包括例如口服(例如,以片剂、胶囊剂、粉剂、颗粒剂的形式);经粘膜(例如舌下、鼻、肛门、直肠或阴道)吸收(例如,以栓剂、乳膏或泡沫的形式);肠胃外(例如,肌肉内、静脉内、腹膜内、皮下或鞘内注射);透皮(例如,作为施加到皮肤的贴片);以及局部施用(例如,作为施加于皮肤的乳膏、软膏、喷雾剂或作为滴眼剂)。本公开的化合物或药物组合物也可以配制成用于吸入施用。
如果需要,可以将总的每日剂量进行拆分并在一天当中分次施用。在一些实施方案中,每日剂量一次性施用。在另一些实施方案中,总的每日剂量在24小时的时间段内以两个、三个或更多个拆分的剂量施用。
主治医师在考虑到患者的年龄、一般健康状况、体重和待治疗症状的严重程度等因素的情况下,可对本公开的化合物或药物组合物的施用量和施用频率进行调整。一般而言,本公开的化合物或药物组合物的总的每日剂量通常为约0.1至约2000mg活性成分/天,例如约1至约1500mg/天、约10至约1250mg/天、约50至约1000mg/天,以单次剂量或2、3或4个分份剂量施用。
本公开的化合物的主要有益效果包括以下几个方面:
(1)本公开的化合物具有广谱抗病毒活性,可以高效地抑制诸如以 下病毒:冠状病毒(例如人冠状病毒,诸如严重急性呼吸综合征冠状病毒(SARS-CoV)、中东呼吸综合征冠状病毒(MERS-CoV)或严重急性呼吸综合征冠状病毒2(SARS-CoV-2,包括例如B.1.1.7株、B.1.351株、德尔塔变异株B.1.617.2株、奥密克戎变异株B.1.1.529株或其它SARS-CoV-2变异株)、马脑炎病毒(例如东部马脑炎病毒、西部马脑炎病毒或委内瑞拉马脑炎病毒)、奇昆古尼亚病毒、罗斯河病毒、正粘病毒科病毒(例如流感病毒,诸如甲型流感病毒、乙型流感病毒或丙型流感病毒)、副粘病毒科病毒(例如呼吸道合胞病毒)、丝状病毒(例如埃博拉病毒、马尔堡病毒或库瓦病毒)或寨卡病毒;
(2)本公开的化合物具有具有改善的口服给药稳定性和增强的抗病毒活性:EIDD-2801在进入细胞前,在胃肠道的酸、碱和消化酶作用下部分地转变为EIDD-1931,从而影响了体内吸收并减弱了抗病毒活性。与之不同的是,本公开的化合物不会被胃肠道的酸、碱和消化酶降解,能够以化合物原型在体内吸收,且细胞通透性增强,于细胞内在磷脂酶的作用下转变为EIDD-1931从而发挥抗病毒作用,提高了抗病毒活性;
(3)本公开的化合物具有较高的安全性:本公开的化合物属于脂质类药,具有改善的药代动力学性质(尤其是体内分布),能够减少核苷类药物对肾脏的毒性,提高对肺部和肝部的靶向性,从而显著降低由于病毒感染所致的肺损伤,因而更具有用药安全性的特点。
制备本公开化合物的一般方法
依照有机合成领域的技术人员公知的合成和纯化方法,可以从市售的或容易制备的起始原料制备式I的化合物。可用于制备式I的化合物的方法阐述在下面的方案和实施例中。但是,这些具体方法不对本发明构成任何限制。有机合成领域的技术人员会明白可替代的合成途径。作为合成方法的参考文献,可以提及的是Steven D.Dong,Chin-Chung Lin,Mel Schroeder,Synthesis and evaluation of a new phosphorylated ribavirin prodrug,Antiviral Research 99(2013)18-26和 WO 2019/113462。
方案1
下面的方案1显示了一种用于制备式I的化合物的方法。该方法尤其适合于制备其中X是-O-的式I的化合物。
Figure PCTCN2022141023-appb-000039
在以上方案中,变量m、n、Z、X、R和R′如上面关于式I的化合物所定义。R 1是直链或直链的C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基或C 6-C 12芳基,所述烷基、烯基、炔基或芳基任选地被1、2或3个独立 地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基;A是氯或硝基,B是氢、氯或硝基。优选地,R 1是甲基、乙基或苯基,A是氯或硝基,B是氢。
步骤1:中间体式2a的化合物的制备
将式1d的化合物(可选自2-氯苯基二氯膦酰酯、2-硝基苯基二氯膦酰酯、4-氯苯基二氯膦酰酯、4-硝基苯基二氯膦酰酯、2,4-二氯苯基二氯膦酰酯、2,4-二硝基苯基二氯膦酰酯)和式1c的化合物(二者均可直接商购获得)在无水溶剂中反应,然后加入式1a的化合物(按照Chemistry-A European Journal(2016),22(21),7215-7224的方法制备)和式1b的化合物(可按照文献Kyle E.Giesler,etal.,LiottaReduction Sensitive Lipid Conjugates of Tenofovir:Synthesis,Stability,and Antiviral Activity;J.Med.Chem.2016,59,7097-7110;US2012/0058976A1或Susumu Tsushima,Chew.Pharm.Bu11.30(9)3260-3270,1982的方法制备)。以上反应可得到式2a的化合物。以上反应过程中所需的无水溶剂包括但不限于无水四氢呋喃、2-甲基四氢呋喃、二氯甲烷、N-甲基吡咯烷酮、甲苯、1,4-二氧六环或它们的混合溶剂等,优选四氢呋喃和2-甲基四氢呋喃。反应温度是0-50℃,优选0-35℃;反应时间是5-48小时,优选10-20小时。
步骤2:中间体式4a的化合物的制备
方法1
将式2a的化合物在三氯氧磷存在下与式1c的化合物反应,得到式3a的化合物。然后将式3a的化合物在碱性条件下与盐酸羟胺反应,得到式4a的化合物。该反应所用的溶剂是乙腈、四氢呋喃、2-甲基四氢呋喃、N-甲基吡咯烷酮、甲苯、1,4-二氧六环或它们的混合溶剂等,优选乙腈和二氧六环。反应温度是0-35℃。反应所需要的碱可以是N,N- 二乙基乙胺、三乙胺、吡啶(均可直接商购获得)(参考Steven D.Dong,Chin-Chung Lin,Mel Schroeder,Synthesis and evaluation of a new phosphorylated ribavirin prodrug,Antiviral Research 99(2013)18-26)。
方法2
在碱性环境下并在4-二甲基氨基吡啶的催化下,将式2a的化合物与式3b的化合物(2,4,5-三异丙基苯磺酰氯,TBSCl)反应,得到式3c的化合物。将式3c的化合物在碱性条件下与盐酸羟胺反应,得到式4a的化合物。该反应所用的溶剂是二氯甲烷、氯仿、1,2-二氯乙烷、乙腈、四氢呋喃、2-甲基四氢呋喃、N-甲基吡咯烷酮、甲苯、1,4-二氧六环或它们的混合溶剂等,优选二氯甲烷、氯仿、1,2-二氯乙烷。反应温度是0-35℃。反应所需要的碱是咪唑、N,N-二乙基乙胺、三乙胺、吡啶(均可直接商购获得),优选咪唑(参考WO 2019/113462)。
步骤3:中间体式5a的化合物的制备
将式4a的化合物用式4b的化合物(1,1,3,3-四甲基胍)和式4c的化合物(2-吡啶甲醛肟)处理以脱去取代的苯基部分,得到式5a的化合物。该反应所用的溶剂是四氢呋喃、2-甲基四氢呋喃、N-甲基吡咯烷酮。反应温度是10-40℃(参考Steven D.Dong,Chin-Chung Lin,Mel Schroeder,Synthesis and evaluation of a new phosphorylated ribavirin prodrug,Antiviral Research 99(2013)18-26)。
步骤4:中间体式6a的化合物的制备
将式4a的化合物在溶剂中反应,脱去羟基保护基后得到式6a的化合物。该反应所用的溶剂可以是氨的甲醇溶液、氨的乙醇溶液或氨的异丙醇溶液。反应温度是20-80℃。
步骤:5式I的化合物的制备
方法1
将式5a的化合物在溶剂中反应,脱去羟基保护基后得到式I的化合物。该反应所用的溶剂可以是氨的甲醇溶液、氨的乙醇溶液或氨的异丙醇溶液。反应温度是20-50℃。
万法2
将式6a的化合物用式4b的化合物(1,1,3,3-四甲基胍)和式4c的化合物(2-吡啶甲醛肟)处理以脱去取代的苯基部分,得到式I的化合物。该反应所用的溶剂是四氢呋喃、2-甲基四氢呋喃、N-甲基吡咯烷酮。反应温度是10-40℃(参考Steven D.Dong,Chin-Chung Lin,Mel Schroeder,Synthesis and evaluation of a new phosphorylated ribavirin prodrug,Antiviral Research 99(2013)18-26)。
方案2
下面的方案2显示了另一种用于制备式I的化合物的方法,其中与方案1相比,用式1-1d的化合物代替了式1d的化合物。该方法尤其适合于制备其中X是-NR 0-的式I的化合物。
Figure PCTCN2022141023-appb-000040
在以上方案中,变量m、n、Z、R和R 0如上面关于式I的化合物所定义。R 1是直链或直链的C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基或C 6-C 12 芳基,所述烷基、烯基、炔基或芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。优选地,R 1是甲基、乙基或苯基。
步骤1:中间体式2a-1的化合物的制备
将式1-1d的化合物和式1c的化合物(二者均可直接商购获得)在无水溶剂中反应,然后加入式1a的化合物和式1b的化合物进行反应。以上反应可得到式2a-1的化合物。该反应所用的无水溶剂包括但不限于无水四氢呋喃、2-甲基四氢呋喃、二氯甲烷、N-甲基吡咯烷酮、甲苯、1,4-二氧六环或它们的混合溶剂等,优选四氢呋喃和2-甲基四氢呋喃。反应温度是0-50℃,优选0-35℃;反应时间是5-48小时,优选10-20小时。
步骤2:中间体式4a-1的化合物的制备
方法1
将式2a-1的化合物在三氯氧磷存在下与式1c的化合物反应,得到式3a-1的化合物。然后将式3a-1的化合物在碱性条件下与盐酸羟胺反应,得到式4a-1的化合物。该反应所用的溶剂是乙腈、四氢呋喃、2-甲基四氢呋喃、N-甲基吡咯烷酮、甲苯、1,4-二氧六环或它们的混合溶剂等,优选乙腈和二氧六环。反应温度是0-35℃。反应所需要的碱可以是N,N-二乙基乙胺、三乙胺或吡啶(均可直接商购获得)。
方法2
在碱性环境下并在4-二甲基氨基吡啶的催化下,将式2a-1的化合物与式3b的化合物(2,4,5-三异丙基苯磺酰氯,TBSCl)反应,得到式3c-1的化合物。将式3c-1的化合物在碱性条件下与盐酸羟胺反应,得到式4a-1的化合物。该反应所用的溶剂是二氯甲烷、氯仿、1,2-二氯乙烷、乙腈、四氢呋喃、2-甲基四氢呋喃、N-甲基吡咯烷酮、甲苯、1,4-二氧 六环或它们的混合溶剂等,优选二氯甲烷、氯仿、1,2-二氯乙烷。反应温度是0-35℃。反应所需要的碱可以是咪唑、N,N-二乙基乙胺、三乙胺、吡啶(均可直接商购获得),优选咪唑(参考WO 2019/113462)。
步骤3:式I的化合物的制备
将式4a-1的化合物在溶剂中反应,脱去羟基保护基后得到式I的化合物。该反应所用的溶剂可以是氨的甲醇溶液、氨的乙醇溶液或氨的异丙醇溶液。反应温度是20-50℃。
方案3
下面的方案3显示了一种用于制备式I-2的化合物的方法。
Figure PCTCN2022141023-appb-000041
在以上方案中,变量m、n和R′如上面关于式I的化合物所定义。A是氯或硝基,B是氢、氯或硝基。优选地,A是氯或硝基,B是氢。
步骤1:中间体式2-1a的化合物的制备
将式1d的化合物(可选自2-氯苯基二氯膦酰酯、2-硝基苯基二氯膦酰酯、4-氯苯基二氯膦酰酯、4-硝基苯基二氯膦酰酯、2,4-二氯苯基二氯膦酰酯、2,4-二硝基苯基二氯膦酰酯)和式1c的化合物(二者均可直接商购获得)反应,然后加入式1-1a的化合物(按照WO 2019/113462或Angewandte Chemie,International Edition,53(19),4862-4866;2014中公 开的方法制备)和式1-1b的化合物(按照US2012/0058976A1或Susumu Tsushima,Chew.Pharm.I3u11.30(9)3260-3270,1982中公开的方法制备,具体而言可如下制备:使用原料CH 3(CH 2) mCH 2OH与甲磺酰氯或对甲苯磺酰氯在三乙胺的存在下反应,生成取代的磺酰酯,然后再与二醇OH(CH 2) nOH(m、n如上面关于式I的化合物所定义)在氢化钠作用下反应)。以上反应可得到式2-1a的化合物(参照文献Susumu Tsushima,Chew.Pharm.Bu11.30(9)3260-3270,1982)。
步骤2:中间体式4-1-a的化合物的制备
方法1
将式2-1a的化合物在三氯氧磷存在下与式1c的化合物反应,得到式3-1a的化合物。然后将式3-1a的化合物在碱性条件下与盐酸羟胺反应,得到式4-1-a的化合物。该反应所用的溶剂是乙腈、四氢呋喃、2-甲基四氢呋喃、N-甲基吡咯烷酮、甲苯、1,4-二氧六环或它们的混合溶剂等,优选乙腈和二氧六环。反应温度是0-35℃。反应所需要的碱可以是N,N-二乙基乙胺、三乙胺、吡啶(均可直接商购获得)(参考Steven D.Dong,Chin-Chung Lin,Mel Schroeder,Synthesis and evaluation of a new phosphorylated ribavirin prodrug,Antiviral Research 99(2013)18-26)。
方法2
在碱性环境下并在4-二甲基氨基吡啶的催化下,将式2-1a的化合物与式3b的化合物(2,4,5-三异丙基苯磺酰氯,TBSCl)反应,得到式3-1c的化合物。将式3-1c的化合物在碱性条件下与盐酸羟胺反应,得到式4-1-a的化合物。该反应所用的溶剂是二氯甲烷、氯仿、1,2-二氯乙烷、乙腈、四氢呋喃、2-甲基四氢呋喃、N-甲基吡咯烷酮、甲苯、1,4-二氧六环或它们的混合溶剂等,优选二氯甲烷、氯仿、1,2-二氯乙烷。反应温度是0-35℃。反应所需要的碱是咪唑、N,N-二乙基乙胺、三乙胺、吡啶(均可直接商购获得),优选咪唑。
步骤3:中间体式5-1a的化合物的制备
将式4-1-a的化合物用4b的化合物(1,1,3,3-四甲基胍)和式4c的化 合物(2-吡啶甲醛肟)处理以脱去取代的苯基部分,得到式5-1a的化合物。该反应所用的溶剂是四氢呋喃、2-甲基四氢呋喃、N-甲基吡咯烷酮。反应温度是10-40℃。
步骤4:中间体式6-1a的化合物的制备
将式4-1-a的化合物在甲酸水溶液(浓度为80%-90%,w/w)或三氟醋酸水溶液(浓度为80%-90%,w/w)中经脱去异丙亚甲基保护基反应,得到式6-1a的化合物。反应温度是20-80℃。
步骤5:式I-2的化合物的制备
方法1
将式5-1a的化合物在甲酸水溶液(浓度为80%-90%,w/w)或三氟醋酸水溶液(浓度为80%-90%,w/w)中经脱去异丙亚甲基保护基反应,得到式I-2的化合物。反应温度是20-80℃。
方法2
将式6-1a的化合物用式4b的化合物(1,1,3,3-四甲基胍)和式4c的化合物(2-吡啶甲醛肟)处理以脱去取代的苯基部分,得到式I-2的化合物。该反应所用的溶剂是四氢呋喃、2-甲基四氢呋喃、N-甲基吡咯烷酮。反应温度是10-40℃(参考WO 2019/113462)。
方案4
下面的方案4显示了另一种用于制备式I-2的化合物的方法,其中与方案3相比,用式1-1d的化合物代替了式1d的化合物。该方法尤其适合于制备其中X是-NR 0-的式I-2的化合物。
Figure PCTCN2022141023-appb-000042
在以上方案中,变量m、n、R和R 0如上面关于式I的化合物所定义。
步骤1:中间体式2-1a-1的化合物的制备
将式1-1d的化合物和式1c的化合物(二者均可直接商购获得)反应,然后加入式1-1a的化合物(按照WO 2019/113462或Angewandte Chemie,International Edition,53(19),4862-4866;2014中公开的方法制备)和式1-1b的化合物(按照US2012/0058976A1或Susumu Tsushima,Chew.Pharm.I3u11.30(9)3260-3270,1982中公开的方法制备,具体而言可如下制备:使用原料CH 3(CH 2) mCH 2OH与甲磺酰氯或对甲苯磺酰氯在三乙胺的存在下反应,生成取代的磺酰酯,然后再与二醇OH(CH 2) nOH(m、n如上面关于式I的化合物所定义)在氢化钠作用下反应)。以上反应可得到式2-1a-1的化合物(参照文献Susumu Tsushima,Chew.Pharm.Bu11.30(9)3260-3270,1982)。
步骤2:中间体式4-1a-1的化合物的制备
方法1
将式2-1a-1的化合物在三氯氧磷存在下与式1c的化合物反应,得到式3-1a-1的化合物。然后将式3-1a-1的化合物在碱性条件下与盐酸羟胺反应,得到式4-1a-1的化合物。该反应所用的溶剂是乙腈、四氢呋喃、2-甲基四氢呋喃、N-甲基吡咯烷酮、甲苯、1,4-二氧六环或它们的混合溶剂等,优选乙腈和二氧六环。反应温度是0-35℃。反应所需要的碱可以是N,N-二乙基乙胺、三乙胺、吡啶(均可直接商购获得)(参考Steven D.Dong,Chin-Chung Lin,Mel Schroeder,Synthesis and evaluation of a new phosphorylated ribavirin prodrug,Antiviral Research 99(2013)18-26)。
方法2
在碱性环境下并在4-二甲基氨基吡啶的催化下,将式2-1a-1的化合物与式3b的化合物(2,4,5-三异丙基苯磺酰氯,TBSCl)反应,得到式3-1c-1的化合物。将式3-1c-1的化合物在碱性条件下与盐酸羟胺反应,得到式4-1a-1的化合物。该反应所用的溶剂是二氯甲烷、氯仿、1,2-二氯乙烷、乙腈、四氢呋喃、2-甲基四氢呋喃、N-甲基吡咯烷酮、甲苯、1,4-二氧六环或它们的混合溶剂等,优选二氯甲烷、氯仿、1,2-二氯乙烷。反应温度是0-35℃。反应所需要的碱是咪唑、N,N-二乙基乙胺、三乙胺、吡啶(均可直接商购获得),优选咪唑。
步骤3:式I-2的化合物的制备
将式4-1a-1的化合物在甲酸水溶液(浓度为80%-90%,w/w)或三氟醋酸水溶液(浓度为80%-90%,w/w)中经脱去异丙亚甲基保护基反应,得到式I-2的化合物。反应温度是20-80℃(参考WO 2019/113462)。
为此,在本公开的第八个方面中,提供了用于合成本公开的式I、I-1或I-2中的任一种的化合物的中间体化合物。
在一些实施方案中,所述中间体化合物是式2a的化合物:
Figure PCTCN2022141023-appb-000043
其中m、n和Z如上面关于式I的化合物所定义;R 1是直链或直链的C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基或C 6-C 12芳基,所述烷基、烯基、炔基或芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基;A是氯或硝基;B是氢、氯或硝基。优选地,R 1是甲基、乙基或苯基,A是氯或硝基,B是氢。
在一些实施方案中,所述中间体化合物是式2a-1的化合物:
Figure PCTCN2022141023-appb-000044
其中m、n、Z、R 0和R如上面关于式I的化合物所定义;R 1是直链或直链的C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基或C 6-C 12芳基,所述烷基、烯基、炔基或芳基任选地被1、2或3个独立地选自卤素、C 1-C 6 烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。优选地,R 1是甲基、乙基或苯基。
在一些实施方案中,所述中间体化合物是式4a的化合物:
Figure PCTCN2022141023-appb-000045
其中m、n和Z如上面关于式I的化合物所定义;R 1是直链或直链的C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基或C 6-C 12芳基,所述烷基、烯基、炔基或芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基;A是氯或硝基;B是氢、氯或硝基。优选地,R 1是甲基、乙基或苯基,A是氯或硝基,B是氢。
在一些实施方案中,所述中间体化合物是式4a-1的化合物:
Figure PCTCN2022141023-appb-000046
其中m、n、Z、R 0和R如上面关于式I的化合物所定义;R 1是直链或直链的C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基或C 6-C 12芳基,所述烷基、烯基、炔基或芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。优选地,R 1是甲基、乙基或苯基。
在一些实施方案中,所述中间体化合物是式2-1a的化合物:
Figure PCTCN2022141023-appb-000047
其中m、n和Z如上面关于式I的化合物所定义;A是氯或硝基;B是氢、氯或硝基。优选地,A是氯或硝基,B是氢。
在一些实施方案中,所述中间体化合物是式2-1a-1的化合物:
Figure PCTCN2022141023-appb-000048
其中m、n、Z、R 0和R如上面关于式I的化合物所定义。
在一些实施方案中,所述中间体化合物是式4-1a的化合物:
Figure PCTCN2022141023-appb-000049
其中m和n如上面关于式I的化合物所定义;A是氯或硝基;B是氢、氯或硝基。优选地,A是氯或硝基,B是氢。
在一些实施方案中,所述中间体化合物是式4-1a-1的化合物:
Figure PCTCN2022141023-appb-000050
其中m、n、Z、R 0和R如上面关于式I的化合物所定义。
实施例
下面用实施例进一步具体地描述本发明。显然,所描述的实施例仅是本发明的一部分,而不是全部。这些实施例仅用于例证本发明,不应将其理解为对本发明的保护范围的限制。基于本发明的实施例, 本领域技术人员在没有付出创造性劳动的前提下所获得的所有其它技术方案,都属于本发明保护的范围。在所述实施例中的化合物的结构均经 1HNMR或MS所确定。
另外,需要注意的是,本领域技术人员在参照“制备本公开化合物的一般方法”部分中所示的通用合成方案和以下实施例中所示的制备本公开化合物的示例性合成方法的基础上,通过选用合适的起始原料和采用适当的步骤,可以容易地制备未在实施例部分中具体例示其合成方法的表1中所列的具体化合物。
实施例1
(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)基四氢 呋喃-2-基)甲基(2-(十六烷二硫基)乙基)磷酸酯(NTV-29)
Figure PCTCN2022141023-appb-000051
步骤1:
2-氯苯基((3R,4R)-5-(2,4-二氧代-3,4-二氢嘧啶-1(2H)基)-3,4-二乙 酰氧基四氢呋喃-2-基)甲基(2-(十六烷二硫基))乙基磷酸酯(A2)
Figure PCTCN2022141023-appb-000052
(1)(3R,4R)-2-(2,4-二氧代-3,4-二氢嘧啶-1(2H)基)-5-(羟甲基)四氢呋喃-3,4-二乙酸酯(2′,3′-二-O-乙酰基脲苷)(A1)
Figure PCTCN2022141023-appb-000053
将干燥的脲苷24.4g(100.0mmol)、咪唑8.2g(120mmol)、1,4-二甲氨基吡啶0.6g(4.9mmol)一同置于300ml干燥二氯甲烷/吡啶(1:1,v/v)溶液中,搅拌并将反应液冷却至约0℃。向上述溶液中滴加叔丁基二甲基氯硅烷26g(120mmol)的二氯甲烷(50ml)溶液,并于该温度和氮气保护下反应约6h。TLC监测反应完成后,加入1ml甲醇,搅拌0.5小时,滴加醋酐22.4g(252mmol)。将反应体系温度升至室温反应6小时。反应完成后,将反应液依次用水(3x100ml)、饱和盐水(100ml)洗涤。将有机相用无水硫酸钠干燥并过滤。将滤液减压蒸干,残留物加正己烷(300ml)重结晶。将固体滤出并干燥,即得近白色固体产物2′,3′-二乙酰基5′-O-叔丁基二甲基硅烷基脲苷24.0g,收率73.0%。将2′,3′-O-二乙酰基5′-O-叔丁基二甲基硅烷基脲苷24.0g(54.2mmol)溶于干燥THF200mL中,加入醋酸3.2ml(54.2mmol),搅拌下将1M的四丁基氟化铵的THF溶液109ml(109mmol)缓慢滴入上述溶液中,滴加完毕后继续搅拌1.5-2.5h。TLC监测反应基本完成后,将溶剂减压蒸干,将得到的固体使用500mL乙酸乙酯复溶,用等体积水洗涤三次,饱和食盐水洗涤一次。将乙酸乙酯层用无水硫酸钠干燥并过滤,将滤液减压蒸干。所得固体使用异丙醚重结晶,即得近白色固体产物2′,3′-二-O-乙酰基脲苷(A1)16g,总收率65%。
(2)2-十六烷基二硫乙醇(1b-1)
C 16H 33SSCH 2CH 2OH
1b-1
向十六烷基-1-硫醇(8.74ml,28.4mmol)和巯基乙醇(2ml,28.4mmol)的MeOH/DCM(50∶50,200mL)溶液中加入吡啶(4.94ml,56.8mmol),接 着分批加入碘(7.21g,28.4mmol),直到溶液颜色维持棕色且不再褪色。将反应溶液于室温搅拌2h,将所得溶液过滤。将滤液减压浓缩,得到的固体加入水后用DCM萃取。将有机层减压浓缩,然后进行硅胶色谱法纯化(使用己烷类/EtOAc(0-8%)梯度),得到化合物2-十六烷基二硫乙醇(1-1b)(3.98g,11.89mmol,41.9%),为白色固体粉末。熔点50-51℃;MS(ESI)m/e[M+Na]+:357.225。
(3)2-氯苯基(((3R,4R)-5-(2,4-二氧代-3,4-二氢嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十六烷二硫基)乙基)磷酸酯(A2)
将1,2,4-三唑(1c)(13.8g,212mmol)和三乙胺(10.1g,200mmol)于干燥四氢呋喃(200mL)中的溶液加入到预先冷却至0℃的2-氯苯基二氯磷酸酯(1d-1)(24.5g,100mmol)于干燥四氢呋喃(150mL)中的溶液中。将所得反应液于室温反应1小时。将生成的固体滤除,并向所得滤液中加入(3R,4R)-2-(2,4-二氧代-3,4-二氢嘧啶-1(2H)基)-5-(羟甲基)四氢呋喃-3,4-二乙酸酯(2′,3′-二-O-乙酰基脲苷)(A1)(32.8g,100mmol)和1-甲基咪唑(8.4ml,100mmol)于四氢呋喃(150mL)中的溶液。加毕,于室温搅拌反应1小时。向反应液中加入2-十六烷基二硫乙醇(1b-1)(33.4g,100mmol)。将所得反应液于室温搅拌过夜,反应完毕后减压浓缩,并将残留物用乙酸乙酯(400mL)溶解。将所得溶液依次用水(200mlx2)、饱和碳酸氢钠水溶液(200mlx2)、水(200mlx2)和饱和盐水(200mlx2)洗涤。分出有机层,经无水硫酸钠干燥、过滤,将滤液减压浓缩,得到2-氯苯基(((3R,4R)-5-(2,4-二氧代-3,4-二氢嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十六烷二硫基)乙基)磷酸酯(A2)(53.0g,70%)无色油状物,其无需进一步纯化可直接用于下步反应。用于结构鉴定的样品可通过色谱柱(CH 2Cl 2+0至10%MeOH)纯化。
1HNMR(600MHz,氯仿-d4/甲醇-d4):δ7.490-7.486(m,3H);7.390-7.268(dd,J=6Hz,J=54Hz,1H);7.199-7.161(dd,J=7.8Hz,1H);6.182-6.046(dd,J=6Hz,7.2Hz,1H);5.778-5.702(dd,J=7.2Hz,1H);5.455-4.339(m,5H);3.079-2.959(m,J=6Hz,J=7.2Hz,2H);2.778-2.689(m,J= 4.2Hz,J=3Hz,2H);2.156-2.142(d,J=2.4Hz,3H);2.113-2.108(d,J=3Hz,3H);1.409-1.355(m,J=7.2Hz,2H);1.318(s,28H);0.912-0.888(t,J=7.2Hz,3H)。
13CNMR(600MHz,氯仿-d4/甲醇-d4)δ169.665-169.413(dd,J=17.4,J=59Hz);162.387;150.228(d,J=10Hz)(d,J=10Hz);146.210;139.355-139.280(d,J=45Hz);130.859-130.839(d,J=12Hz);128.199;126.500;125.422;121.509-121.494(d,J=9Hz);103.601;86.370-86.200(d,J=102Hz);80.813-80.762(d,J=30.6Hz);72.496-72.457(d,J=23.4Hz);70.192-70.171(d,J=12.6Hz);67.382;67.126;67.086;67.016;39.255;39.146;37.926-37.879(d,J=28Hz);31.929;29.699;29.660;29.607;29.520;29.363;29.241;29.144;28.542;22.690;20.539;20.368;14.121。
31P:δ-6.845,-7.188。
MS(ESI)m/e[M+H] +:835.3。
步骤2:
2-氯苯基((3R,4R)-5-(2-氧代-4-(1H-1,2,4-三唑-1-基)嘧啶-1(2H) 基)-3,4-二乙酰氧基四氢呋喃-2-基)-甲基(2-(十六烷二硫基)乙基)磷酸酯 (A3)
Figure PCTCN2022141023-appb-000054
将2-氯苯基((3R,4R)-5-(2,4-二氧代-3,4-二氢嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十六烷二硫基)乙基)磷酸酯(A2)(83.5g,100.0mmol)溶于乙腈(400.0mL)中,并加入1,2,4-三唑(48.0g,700.0mmol)和三乙胺(81g,800.0mmol),通过搅拌使固体溶解。将反应 液冷却至0℃,在氮气氛下缓慢滴加三氯氧磷(14.5ml,150.0mmol)。加毕,将反应液温度升至室温,继续搅拌2小时使反应进行完全,并通过TLC(100%EtOAc)监测反应。反应进行完全后,向反应液中加入水(500mL)和乙酸乙酯(400mL),于室温搅拌15分钟以淬灭反应。将分出的有机层依次用水(2x100mL)、0.5N HCl(200mL)和饱和盐水(2x100mL)洗涤,经无水硫酸镁干燥并过滤,将滤液浓缩,得到过渡中间体2-氯苯基(((3R,4R)-5-(2-氧代-4-(1H-1,2,4-三唑-1-基)嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十六烷二硫基)乙基)磷酸酯(A3)(80.6g,100%)(其不需要纯化直接用于下步反应)。用于结构鉴定的样品可通过色谱柱(CH 2Cl 2+0至8%MeOH)纯化。
1HNMR(600MHz,氯仿-d4/甲醇-d4):δ9.293-9.275(m,J=4.2Hz,J=6.6Hz,1H);8.317(s,1H);8.152-8.131(d,J=3.6Hz,1H);7.648-7.041(m,5H);6.315-6.191(dd,J=4.2Hz,12Hz,1H);5.486-5.310(m,2H);4.663-4.324(m,3H);3.00-2.947(m,J=6Hz,2H);2.737-2.665(m,J=7.2Hz,2H);2.137(s,3H);2.113(s,3H);1.358-1.334(m,J=7.4Hz,2H);1.260(s,30H);0.881(t,J=7.2Hz,3H)。
13CNMR(600MHz,氯仿-d4/甲醇-d4):δ169.684(dd,J=10Hz);159.554;154.541;153.768;146.675-146.595(d,J=48Hz);145.638;143.320;130.807-130.791(d,J=9Hz);128.202-128.158(d,J=26.4Hz);126.630-126.583(d,J=28.2);125.290;121.365-121.330(d,J=21Hz);95.740;93.291;89.038-88.956(d,J=49.2Hz);80.849-80.754(t,J=26.4Hz);73.752;69.433;67.149;66.776;45.961;42.305;40.261;39.231;38.919-38.898(d,J=7Hz);37.77737.696(t,J=25.8Hz);31.791;29.559;29.523;29.465;29.380;29.227;29.095;28.999;28.365;22.552;20.263-20.177(d,J=51.6Hz);13.920。
31P:δ-7.005,-7.314。
MS(ESI)m/e:885.30[M] +
步骤3:
2-氯苯基(((3R,4R)-5-(2-氧代-4-(羟氨基)嘧啶-1(2H)基)-3,4-二乙酰 氧基四氢呋喃-2-基)甲基(2-(十六烷二硫基)乙基)磷酸酯(A4)
Figure PCTCN2022141023-appb-000055
向上面所得的产物(A3)(41.8g,50mmol)中加入异丙醇(300mL)和三乙胺(7.5g,75mmol),于室温下搅拌使固体溶解。加入盐酸羟胺(5.2g,75mmol),将反应搅拌2.5小时后得到淡黄色反应液。通过TLC(EtOAc)监测反应完毕。在小于45℃的温度下减压浓缩以去除溶剂。将残留物溶于乙酸乙酯(300ml)中,将有机相依次用水(150mlx2)、饱和盐水(150mlx2)洗涤,经无水硫酸钠干燥、过滤,将滤液减压浓缩。将得到的浓缩物用石油醚重结晶,得到白色固体2-氯苯基((3R,4R)-5-(2-氧代-4-(羟氨基)嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十六烷二硫基)乙基)磷酸酯(A4)(38.3g,90%)。
1HNMR(600MHz,氯仿-d4/甲醇-d4):δ7.477-7.443(m,J=4.8Hz,J=7.8Hz,3H);7.332-7.306(t,J=7.8Hz,1H);7.272-7.160(m,J=8.4Hz,1H);6.407-6.320,5.8445.768(m,J=7.8Hz,1H);6.181-6.059(m,J=7.8Hz,1H);5.454-5.205(m,3H);4.517-4.305(m,2H);2.991-2.957(m,J=6.6Hz,2H),2.718-2.690(m,J=7.8Hz,2H);2.136(s,3H);2.109(s,3H);1.372(m,J=7.4Hz,2H);1.272(s,26H);0.898(t,J=7.2Hz,3H)。
13CNMR(600MHz,氯仿-d4/甲醇-d4):δ169.684(dd,J=10Hz);158.406;155.294;148.382;147.301;144.327;130.931;128.419;126.692;121.546;91.434;80.782;72.370;70.926;70.137;70.098;69.369;69.085;68.969;67.131;54.971;39.259;37.875;32.430;31.791;31.361;29.707;29.665;29.623;29.540;29.336;29.260;29.157;28.532;22.696;20.401;14.124。
31P:δ-6.954,-7.206,-7.326。
MS(ESI)m/e:850.30[M+1] +
步骤4:
(((3R,4R)-5-(2-氧代-4-(羟氨基)嘧啶-1(2H)基)-3,4-二乙酰氧基四氢 呋喃-2-基)甲基(2-(十六烷二硫基)乙基)磷酸酯(A5)
Figure PCTCN2022141023-appb-000056
将1,1,3,3-四甲基胍(4b)(12.9g,112.0mmol)和2-吡啶甲醛肟(4c)(13.70g,112.0mmol)于THF(120ml)中的溶液加入到2-氯苯基(((3R,4R)-5-(2-氧代-4-(羟氨基)嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十六烷二硫基)乙基)磷酸酯(A4)(17.0g,20mmol)于THF(360mL)中的溶液中。将所得反应溶液于室温搅拌过夜。通过TLC(二氯甲烷∶甲醇=20∶1,v/v)检测反应完毕后,向反应液中加入乙酸乙酯(480ml)和冷盐酸溶液(pH=1-2,200ml)。分出有机层,将水层用乙酸乙酯/四氢呋喃(1∶1)溶液100ml萃取3次。合并有机层,用2N氢氧化钠溶液(200ml×2)萃取。将水相合并,用盐酸酸化至溶液的pH为1-2,然后用乙酸乙酯(200ml×2)萃取。合并萃取液,用无水硫酸钠干燥,过滤并浓缩。将浓缩物用石油醚重结晶,得到类白色固体(((3R,4R)-5-(2-氧代-4-(羟氨基)嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十六烷二硫基)乙基)磷酸酯(A5)(10.35g,70%)。
1HNMR(600MHz,氯仿-d4/甲醇-d4):δ7.728-7.713(m,J=5.4Hz,J=3.6Hz,0.35H);7.565-7.550(m,J=5.4Hz,J=3.6Hz,0.35H);7.274-7.260,7.035-7.021,6.834-6.819(td.J=8.4Hz,0.30H);5.988-5.328(m,J=7.2Hz,0.3HJ=3Hz,2H);4.394-4.371(dd,J=8.4Hz,J=7.2Hz,2H);4.215-4.109(m,J=6.6Hz,J=7.2,3H);2.713-2.678(m,J=6.6Hz,,2H);2.532-2.505(m,J=8.4Hz,2H); 2.316-2.265(m,J=7.8Hz,2H);2.171(d,3H);2.037(d,3H);1.416(m,J=7.4Hz,2H);1.272(s,26H);0.883(t,J=7.2Hz,3H)。
13CNMR(600MHz,氯仿-d4/甲醇-d4):δ163.850;149.382;132.649;127.665;104.878;72.861;71.790;70.217;68.812;66.924;65.615;62.537;60.846;56.043;39.099;36.165;32.934;32.067;31.793;29.707;30.655;29.553;29.517;29.221;29.093;29.004;28.896;28.405;27.735;23.385;22.542;20.542;13.889。
31P:δ-6.954,-1.106,-1.413,-1.627。
MS(-ESI)m/e:738.30[M-H] +
步骤5:
(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2--氧代嘧啶-1(2H)基四 氢呋喃-2-基)甲基(2-(十六烷二硫基)乙基)磷酸酯(NTV-29)
Figure PCTCN2022141023-appb-000057
将((3R,4R)-5-(2-氧代-4-(羟氨基)嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十六烷二硫基)乙基)磷酸酯(A5)(7.4g,10mmol)溶解于甲醇(50mL)中。将反应液于室温搅拌以使物料溶解。加入氨的甲醇溶液(7mol/L,5ml),并于室温反应5h。待反应进行完毕后,将反应液减压浓缩。向得到的残留物中加入盐酸水溶液(pH1-2)30ml。将得到的溶液用乙酸乙酯(3x50ml)萃取。合并有机相,经无水硫酸钠干燥并过滤。将滤液减压浓缩,用甲醇重结晶,得到(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)基四氢呋喃-2-基)甲基(2-(十六烷二硫基)乙基)磷酸酯(NTV-29)(6.04g,90%)
1HNMR(600MHz,氯仿-d4/甲醇-d4):δ8.304-7.264(qd,J=7.8Hz,H);δ6.180-5.748(dd,J=6.0Hz,7.8Hz,2H);4.328-4.125(m,5H);3.175-2.937(dtJ=7.2Hz,J=6.0Hz,2H);2.841-2.679(m,J=7.2Hz 2H);1.772-1.617 (m,J=7.2Hz,2H);1.476-1.354(m,J=7.2Hz,J=6.0Hz,2H);1.276(s,26H);0.890(t,J=7.2Hz,3H)。
13CNMR(600MHz,氯仿-d4/甲醇-d4):δ163.649;148.928;134.091;128.979;122.135;105.786;103.191;89.422;74.500;73.341;69.152;66.924;56.036;38.716;38.527;38.092;31.552;29.281;29.253;29.208;29.141;28.964;28.865;28.732;28.088;22.259;13.338。
31P:δ-3.002,-2.964,-2.864。
MS(ESI)m/e:656.2[M+H] +
实施例2
(((2R,3S,4R,5R)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(,2H)基四 氢呋喃-2-基)甲基(2-(十八烷二硫基)乙基)磷酸酯(NTV-37)
Figure PCTCN2022141023-appb-000058
以与实施例1类似的方法制备(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)基四氢呋喃-2-基)甲基(2-(十八烷二硫基)乙基)磷酸酯(NTV-37)。
步骤1:
2-氯苯基(((3R,4R)-5-(2,4-二氧代-3,4-二氢嘧啶-1(2H)基)-3,4-二乙 酰氧基四氢呋喃-2-基)甲基(2-(十八烷二硫基)乙基)磷酸酯(1A2)
Figure PCTCN2022141023-appb-000059
参照实施例1的步骤1中所述的制备(A2)的方法,用2-(十八烷基 二硫)乙醇(1b-2)代替2-十六烷基二硫乙醇(1b-1)进行反应,得到2-氯苯基(((3R,4R)-5-(2,4-二氧代-3,4-二氢嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十八烷二硫基)乙基)磷酸酯(1A2)。
具体而言,按下表进行投料:
Figure PCTCN2022141023-appb-000060
将1,2,4-三唑(1c)、三乙胺于干燥四氢呋喃中的溶液加入到预先冷却至0℃的2-氯苯基二氯磷酸酯(1d-1)于干燥四氢呋喃中的溶液中。将所得反应液于室温反应1小时。将生成的固体滤除。向所得滤液中加入2′,3′-二-O-乙酰基脲苷(A1)和1-甲基咪唑于四氢呋喃中的溶液。加毕,于室温搅拌反应1小时。然后,向反应液中加入2-十八烷基二硫乙醇(1b-2),并将所得反应液于室温搅拌过夜。将反应液减压浓缩,并将残留物用乙酸乙酯(200mL)溶解。将所得溶液依次用水(100mlx2)、饱和碳酸氢钠水溶液(100mlx2)、水(100mlx2)和饱和盐水(100mlx2)洗涤。分出有机相,经无水硫酸钠干燥并过滤。将滤液减压浓缩,得到2-氯苯基(((3R,4R)-5-(2,4-二氧代-3,4-二氢嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十八烷二硫基)乙基)磷酸酯(1A2)(29.2g,收率71%)无色油状物,其无需进一步纯化可直接用于下步反应。用于结构鉴定的样品可通过色谱柱(CH 2Cl 2+0至10%MeOH)纯化。
1HNMR(600MHz,氯仿-d4/甲醇-d4):δ7.483-7.414(m,J=3Hz, J=8.4Hz,2H);7.308-7.266(dd,J=1.8Hz,J=7.8Hz,1H);7.199-7.161(dd,J=7.2Hz,1H);6.099-6.064(ddd,J=6Hz,3.6Hz,1.8Hz,1H);5.747-5.677(ddd,J=7.8Hz,J=1.8Hz,J=2.4Hz 1H);5.439-5.339(m,J=3.6Hz,6.6Hz,4.2H,1H);5.323-5.269(m,J=3.6Hz,2.4Hz,7.2H,1H);4.542-4.460(m,4H);2.992-2.947(t,J=6Hz,2H);2.707-2.674(m,J=4.2Hz,J=2.4J=7.6Hz,2H);2.135-2.129(t,J=1.8Hz,3H);2.104-2.096(t,J=2.4Hz,3H);1.683-1.630(m,J=7.2Hz,J=5.4Hz,J=2.4Hz,2H);1.261(s,30H);0.893-0.866(t,J=7.2Hz,3H)。
13CNMR(600MHz,氯仿-d4/甲醇-d4):δ169.766-169.743(d,J=13.8,Hz);169.580-169.564(d,J=9.6Hz);150.402;145.988-145.948(d,J=24Hz);139.674-139.585(d,J=53.4Hz);130.707-130.690(d,J=10.2Hz);128.081;126.482;125.265-125.218(d,J=26.4Hz);121.282;103.233;86.891-86.728(d,J=97.8Hz);80.418-80.312(t,J=31.8Hz);72.461;69.848-69.832(d,J=8.4.Hz);67.241-67.203(d,J=22.8Hz);67.066-67.025(d,J=24.6Hz);67.033-66.962(dd,J=24.6Hz,J=7.2Hz);39.055;38.888;37.69537.648(d,J=22.2Hz);31.752;29.516;29.476;29.424;29.341;29.180;29.056;28.953;28.326;22.502;20.211;20.075-20.067(d,J=8.4Hz);13.858。
31Pδ:-3.188,-3.445。
MS(ESI)m/e:863.3[M] +
步骤2:
2-氯苯基(((3R,4R)-5-(2-氧代-4-(1H-1,2,4-三唑-1-基)嘧啶-1(2H) 基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十八烷二硫基)乙基)磷酸酯 (1A3)
Figure PCTCN2022141023-appb-000061
参照实施例1的步骤2中所述的制备(A3)的方法,用2-氯苯基(((3R,4R)-5-(2,4-二氧代-3,4-二氢嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十八烷二硫基))乙基)磷酸酯(1A2)代替2-氯苯基((3R,4R)-5-(2,4-二氧代-3,4-二氢嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十六烷二硫基)乙基)磷酸酯(A2)进行反应,得到2-氯苯基(((3R,4R)-5-(2-氧代-4-(1H-1,2,4-三唑-1-基)嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十八烷二硫基)乙基)磷酸酯(1A3)无色油状物12.8g,收率81%。
具体而言,按下表进行投料:
Figure PCTCN2022141023-appb-000062
1HNMR(600MHz,氯仿-d4/甲醇-d4):δ9.297-9.285(t,J=3.5Hz, 0.5H);8.404(m,0.5H);8.325-8.310(dd,J=7.2Hz,J=1.8Hz,0.5H);7.465-7.439(m,J=6.6Hz,J=2.4Hz,1H);7.444-7.379(m,J=7.2Hz,J=1.8Hz,1H);7.321-7.273(m,J=8.4Hz,J=2.4Hz,J=1.8Hz,1H);7.245-7.169(m,J=3Hz,1H);7.141-7.068(dd,J=7.2Hz,J=36.6Hz,1H);6.296-5.987(dd,J=7.8Hz,J=32.4Hz,J=11.4Hz,1H),5.464-5.337(m,2H);4.664-4.309(m,4H);3.002-2.948(m,J=6.6Hz,2H);2.723-2.667(m,J=7.2Hz,2H);2.145(m,J=7.2Hz,3H);2.139(s,3H);1.366-1.362(m,2H);1.272(s,28H);0.882(t,J=7.2Hz,3H)。
13CNMR(600MHz,氯仿-d4/甲醇-d4):δ169.898(d,J=8.4Hz);169.798;159.751(d,J=11.4Hz);154.762;153.923;148.895(d,J=46.2Hz);145.459;143.477;130.981;128.354(d,J=22.4Hz);126.828(d,J=28.8Hz);121.506(t,J=29.4Hz);95.944;89.268(d,J=49.2Hz);80.981(t,J=29.4Hz);69.607;67.302(d,J=57Hz);66.961;46.244;39.096;37.913;31.981;29.745;29.705;29.650;29.564;29.409;29.280;29.181;28.547;22.731;20.476(d,J=58.2Hz);14.080。
31P:δ-6.954,-3.039,-3.342。
MS(ESI)m/e:914.50[M] +
步骤3:
2-氯苯基(((3R,4R)-5-(2-氧代-4-(羟氨基)嘧啶-1(2H)基)-3,4-二乙酰 氧基四氢呋喃-2-基)甲基(2-(十八烷二硫基)乙基)磷酸酯(1A4)
Figure PCTCN2022141023-appb-000063
参照实施例1的步骤3所述的方法合成目标产物。具体而言,按下表进行投料:
Figure PCTCN2022141023-appb-000064
向上面得到的产物(1A3)中加入异丙醇和三乙胺,于室温下搅拌以使固体溶解。向反应液中加入盐酸羟胺,搅拌1小时后得到淡黄色反应液。通过TLC(EtOAc)监测反应完毕。在小于45℃的温度下减压浓缩除去溶剂。将残留物溶解于乙酸乙酯中,将有机层依次用水、饱和盐水洗涤。将所得有机溶液用无水硫酸钠干燥、过滤并浓缩。将浓缩物用石油醚重结晶,得到白色固体2-氯苯基(((3R,4R)-5-(2-氧代-4-(羟氨基)嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十八烷二硫基)乙基)磷酸酯(1A4),收率90%。
1HNMR(600MHz,甲醇-d4):δ7.511-7.491(dd,J=7.2Hz,1H);7.480-7.466(d,J=7.2Hz,1H);7.340-7.327((dd,J=7.2Hz,1H);7.234-7.224((d,J=7.2Hz,1H),6.853-6.839(d,J=7.4Hz,1H);5.968-5.940(d,J=6.0Hz,1H);5.704-5.53(dd,J=16.8;Hz J=7.2Hz,1H);5.453-5.417(m,J=7.2Hz,1H);5.352-5.33(m,J=7.2Hz,1H);4.531-4.471(m,3H);4.315(m,2H);3.017-3.000(m,J=4.2Hz,3.6Hz,2H);2.727-2.698(dt,J=7.2Hz,J=4.2Hz,2H);2.109-2.107(d,J=1.2Hz,3H);2.071-2.068(d,J=1.8Hz,3H);1.672-1.648(t,J=7.2Hz,2H);1.434-1.243(m,30H);0.898(t,J=7.2Hz,3H)。
13CNMR(600MHz,甲醇-d4):δ169.972(d,J=15Hz),169.818(d,J=12.6Hz);149.795;146.256;144.315;130.591;130.363(d,J=38.4Hz);128.160;126.558;125.162(d,J=27Hz);121.435(d,J=24.6Hz);102.213;98 .915;88.690;87.192(d,J=69.6Hz);79.928(dd,J=16.2Hz,J=30Hz);71.840;69.886(d,J=9.6hz);69.718(m,J=24.6Hz,J=16.8Hz,J=12Hz);38.503;37.641(d,J=33Hz);31.729;29.415;29.426;29.363;19.298;29.141;28.983;28.891;28.807;28.135;22.395;19.132,(d,J=38.4Hz);13.119。
31P:δ-7.277,-7.413。
MS(ESI)m/e:878.3237[M+H] +
步骤4:
(((3R,4R)-5-(2-氧代-4-(羟氨基)嘧啶-1(2H)基)-3,4-二乙酰氧基四氢 呋喃-2-基)甲基(2-(十八烷二硫基)乙基)磷酸酯(1A5)
Figure PCTCN2022141023-appb-000065
参照实施例1的步骤4中所述的制备(A5)的方法,用2-氯苯基(((3R,4R)-5-(2-氧代-4-(羟氨基)嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十八烷二硫基)乙基)磷酸酯(1A4)代替2-氯苯基(((3R,4R)-5-(2-氧代-4-(羟氨基)嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十六烷二硫基)乙基)磷酸酯(A4)制备化合物(((3R,4R)-5-(2-氧代-4-(羟氨基)嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十八烷二硫基)乙基)磷酸酯(1A5):
具体而言,按下表进行投料:
Figure PCTCN2022141023-appb-000066
将1,1,3,3-四甲基胍(4b)和2-吡啶甲醛肟(4c)于THF中的溶液加入到2-氯苯基(((3R,4R)-5-(2-氧代-4-(羟氨基)嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十八烷二硫基)乙基)磷酸酯(1A4)于THF中的溶液中。将所得反应溶液于室温搅拌过夜。通过TLC(二氯甲烷∶甲醇=20∶1,v/v)检测反应完毕。反应完毕后,向反应液中加入乙酸乙酯和冷盐酸水溶液(pH=1-2)。分出有机层,将水层用乙酸乙酯/四氢呋喃(1∶1,v/v)溶液萃取3次。合并有机层,用2N氢氧化钠溶液萃取。将水相合并,用盐酸酸化至溶液的pH为1-2,然后用乙酸乙酯萃取。将萃取液合并,经无水硫酸钠干燥、过滤并浓缩。将浓缩物用石油醚重结晶,得到类白色固体(((3R,4R)-5-(2-氧代-4-(羟氨基)嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十八烷二硫基)乙基)磷酸酯(1A5)4.2g(收率65%)。
1HNMR(600MHz,氯仿-d4/甲醇-d4):δ7.846-7.723(m,J=7.2Hz,1H);6.165-5.413(mJ=7.8Hz,2H);4.710-4.386(m,J=4.2Hz,J=3Hz,J=14.2Hz,5H);4.189-4.176(m,J=3Hz,J=4.8Hz,2H);3.916-3.893(m,J=8.4Hz,2H);3.811-3.773(m,J=4.8Hz,,2H),2.132(d,3H);2.096(d,3H);1.408(m,J=7.4Hz,2H);1.261(s,30H);0.882(t,J=7.2Hz,3H)。
13CNMR(600MHz,氯仿-d4/甲醇-d4):δ170.158;166.927;157.177;133.281;128.768;124.491;120.504;116.385;96.866;64.274;63.160;62.814(J=27.6Hz);62.689(J=35.46Hz);52.024;33.991;31.794;29.556;29.518;29.477;29.383;29.350;29.171;29.041;24.818;22.545;16.184(m,J=13.8Hz);13.996。
31P:δ,25.554;24.651。
步骤5:
(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)基四氢 呋喃-2-基)甲基(2-(十八烷二硫基)乙基)磷酸酯(NTV-37)
Figure PCTCN2022141023-appb-000067
参照实施例1的步骤5中所述的制备化合物NTV-29的方法,使(((3R,4R)-5-(2-氧代-4-(羟氨基)嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十八烷二硫基))乙基磷酸酯(1A5)与氨的甲醇溶液反应以得到目标物(NTV-37)。
具体而言,将(((3R,4R)-5-(2-氧代-4-(羟氨基)嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十八烷二硫基))乙基磷酸酯(1A5)溶于甲醇中,并于室温搅拌以使物料溶解。加入氨的甲醇溶液(7mol/L),于室温反应5h。待反应进行完毕后,将反应液减压浓缩。向残留物中加入盐酸水溶液(pH1-2)30ml以溶解残留物。将水溶液用乙酸乙酯萃取3次,合并有机相,经无水硫酸钠干燥,过滤。将滤液减压浓缩,用甲醇重结晶,得到((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)基四氢呋喃-2-基)甲基(2-(十八烷二硫基)乙基)磷酸酯(NTV-37)(收率95%)。
1HNMR(600MHz,氯仿-d4/甲醇-d4):δ8.061(d,J=8.4Hz,0.2H);δ7.572(dd,J=7.8Hz,3.0Hz,0.8H);5.997(d,J=7.8,Hz,0.2H);5.764(d,J=8.4Hz,0.8H);5.699(d,J=8.4Hz,0.8H);5.573(d,J=8.4Hz,0.2H);4.048-3.95(m,5H);3.166-3.154(m,J=2.4Hz,2H);2.724-2.754(t,J=7.2Hz,2H);2.493-2.717(t,J=7.2Hz,2H);1.406-1.489(t,J=7.2Hz,2H);1.068-1.196(m,30H);0.688(t,J=7.2Hz,3 H)。
13CNMR(600MHz,氯仿-d4/甲醇-d4):δ156.34;154.804;147.446;128.561;125.075;119.237;113.345;92.684;89.755;74.407;69.327;64.981;39.214;38.991;31.916;29.676;29.639;29.603;29.574;29.526;29.341;29.253;29.208;28.510;22.656;13.955。
31P:δ2.800,2.667。
MS(ESI)m/e:700.29[M+H] +
实施例3
(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)基四氢 呋喃-2-基)甲基(2-(十八烷二硫基)乙基)(甲基碳酸甲基)磷酸酯 (NTV-59)
Figure PCTCN2022141023-appb-000068
按照下列路线进行合成:
Figure PCTCN2022141023-appb-000069
步骤1:
氯甲基碳酸甲酯
将氯甲酸氯甲酯(1.5g,11.72mmol)和甲醇(375mg,11.72mmol)置于反应瓶中,加入无水乙醚15ml溶解。于0℃,缓慢滴加吡啶 (0.93g,11.72mmol)。加毕,将反应溶液升至室温,搅拌反应过夜。加入水稀释,依次用1%(w/v)柠檬酸水溶液、饱和碳酸氢钠水溶液和饱和食盐水洗涤3次,分出有机层,经无水硫酸钠干燥,过滤并浓缩,得到产物1.32g,收率:90.8%。
步骤2:
(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)基四氢 呋喃-2-基)甲基(2-(十八烷二硫基)乙基)(甲基碳酸甲基)磷酸酯 (NTV-59)
向反应瓶中加入化合物NTV-37(341.8mg,0.5mmol)、氯甲基碳酸甲酯(513.8mg,4.14mmol)、碳酸钾(572.7mg,4.14mmol)、碘化钾(68.1mg,0.41mmol)和DMF(10ml)。将反应液加热至60℃反应过夜。用TLC检测反应完毕。将反应液降至室温,加入过量水稀释,用乙酸乙酯(3x30ml)萃取。合并有机相,用饱和食盐水洗涤并浓缩,用硅胶柱色谱法纯化得到154mg产物,收率:40%。
1HNMR(600MHz,甲醇-d4):δ7.961(d,J=8.4Hz,0.2H);δ7.472(dd,J=7.8Hz,3.0Hz,0.8H);6.097(d,J=7.8,Hz,0.2H);5.704(d,J=8.4Hz,0.8H);5.629(d,J=8.4Hz,0.8H);5.583(d,J=8.4Hz,0.2H);4.028-3.85(m,8H);3.164-3.152(m,J=2.4Hz,4H);2.724-2.754(t,J=7.2Hz,2H);2.493-2.717(t,J=7.2Hz,2H);1.406-1.489(t,J=7.2Hz,2H);1.068-1.196(m,30H);0.688(t,J=7.2Hz,3H)。
MS(ESI)m/e:771.32[M] +
实施例4
(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)基四氢 呋喃-2-基)甲基(2-(十八烷二硫基)乙基)(N,N-二甲氨基)磷酸酯 (NTV-84)
Figure PCTCN2022141023-appb-000070
参照实施例2的反应路线,用1d-2代替1d-1进行反应,得到化合物NTV-84。
步骤1:
N,N-二甲氨基(((3R,4R)-5-(2,4-二氧代-3,4-二氢嘧啶-1(2H)基)-3,4- 二乙酰氧基四氢呋喃-2-基)甲基(2-(十八烷二硫基)乙基)磷酸酯(A2-1)
Figure PCTCN2022141023-appb-000071
按下表进行投料:
Figure PCTCN2022141023-appb-000072
将1,2,4-三唑(1c)和三乙胺于干燥四氢呋喃中的溶液加入到预先冷 却至0℃的N,N-二甲氨基二氯磷酸酯(1d-2)于干燥四氢呋喃中的溶液中。将所得反应液于室温反应1小时。将生成的固体滤除。向所得滤液中加入2′,3′-二-O-乙酰基脲苷(A1)和1-甲基咪唑于四氢呋喃中的溶液。加毕,于室温搅拌反应1小时。然后,向反应液中加入2-十八烷基二硫乙醇(1b-2),并所得反应液于室温搅拌过夜。将反应液减压浓缩,并将残留物用乙酸乙酯(200mL)溶解。将所得溶液依次用水(100mlx2)、饱和碳酸氢钠水溶液(100mlx2)、水(100mlx2)和饱和盐水(100mlx2)洗涤。分出有机相,经无水硫酸钠干燥并过滤。将滤液减压浓缩,得到N,N-二甲氨基(((3R,4R)-5-(2,4-二氧代-3,4-二氢嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十八烷二硫基)乙基)磷酸酯(A2-1)(39.24g,收率75%)无色油状物,其无需进一步纯化可直接用于下步反应。
MS(ESI)m/e:779.36[M] +
步骤2:
N,N-二甲氨基(((3R,4R)-5-(2-氧代-4-(1H-1,2,4-三唑-1-基)嘧啶 -1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十八烷二硫基)乙基) 磷酸酯(A3-1)
Figure PCTCN2022141023-appb-000073
参照实施例2的步骤2中所述的制备(1A3)的方法进行反应,得到中间体(A3-1)。具体而言,按下表进行投料:
试剂 分子量 摩尔(mmol) 重量(g) 摩尔比
1,2,4-三唑(1c) 69.07 152.5 9 7.6
中间体(A2-1) 779.9 20 15.6 1
乙腈     100  
三乙胺 101 160 11.5 8
三氯氧磷 153.3 30 4.6 1.5
将中间体(A2-1)在搅拌下溶解于乙腈中。溶解后,加入1,2,4-三唑和三乙胺,搅拌反应液使固体溶解。将反应液冷却至0℃,在氮气氛下缓慢滴加三氯氧磷。加毕,使反应液温度升至室温,继续搅拌2小时使反应进行完全。通过TLC(100%EtOAc)监测反应。反应进行完毕后,通过加入水和乙酸乙酯并于室温搅拌15分钟以淬灭反应。分出有机层,依次用水、0.5N HCl和饱和盐水洗涤,经无水硫酸镁干燥、过滤并浓缩,得到白色固体物(A3-1)(33g,收率80%),其不需要纯化而直接用于下步反应。
MS(ESI)m/e:830.38[M] +
步骤3:
N,N-二甲氨基(((3R,4R)-5-(2-氧代-4-(羟氨基)嘧啶-1(2H)基)-3,4-二 乙酰氧基四氢呋喃-2-基)甲基(2-(十八烷二硫基)乙基)磷酸酯(A4-1)
Figure PCTCN2022141023-appb-000074
参照实施例2的步骤3中所述的制备(1A4)的方法进行反应,得到中间体(A4-1)。具体而言,按下表进行投料:
试剂 分子量 摩尔(mmol) 重量(g) 摩尔比
盐酸羟胺 69.5 15 1.04 1.5
三乙胺 101.2 15 15 1.5
中间体(A3-1) 831.04 10 8.3 1
异丙醇     45ml  
向上面得到的产物(A3-1)中加入异丙醇和三乙胺,于室温下搅拌以使固体溶解。向反应液中加入盐酸羟胺,搅拌1小时后得到淡黄色反应液。通过TLC(EtOAc)监测反应完毕。在小于45℃的温度下减压浓缩除去溶剂。将残留物溶解于乙酸乙酯中,将有机层依次用水、饱和盐水洗涤。将所得有机溶液用无水硫酸钠干燥、过滤并浓缩。将浓缩物用石油醚重结晶,得到白色固体N,N-二甲氨基(((3R,4R)-5-(2-氧代-4-(羟氨基)嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十八烷二硫基)乙基)磷酸酯(A4-1)7.3g,收率91%。
MS(ESI)m/e:794.37[M] +
步骤4:
(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)基四氢 呋喃-2-基)甲基(2-(十八烷二硫基)乙基)(N,N-二甲氨基)磷酸酯 (NTV-84)
将N,N-二甲氨基(((3R,4R)-5-(2-氧代-4-(羟氨基)嘧啶-1(2H)基)-3,4-二乙酰氧基四氢呋喃-2-基)甲基(2-(十八烷二硫基)乙基)磷酸酯(A4-1)4g溶解于甲醇30ml中。将反应液于室温搅拌以使物料溶解。加入氨的甲醇溶液10ml(7mol/L),于室温反应5h。待反应进行完毕后,减压浓缩,并将残留物加入水30ml中。将所得水溶液用乙酸乙酯萃取3次,合并有机相,经无水硫酸钠干燥并过滤。将滤液减压浓缩,用甲醇重结晶,得到化合物(NTV-84)2.85g(收率80%)。
1HNMR(600MHz,甲醇-d4):δ7.861-7.562(m J=8.4Hz,3.0Hz,1H);7.572-5.997(m,J=7.8Hz,1H)5.764-5.719(m,J=8.4Hz,1H);4.048-3.95(m,5H);3.166-3.154(m,J=2.4Hz,2H);2.724-2.754(t,J=7.2Hz,2H);2.652(s,3H);2.602(s,3H);2.487-2.713(t,J=7.2Hz,2H);1.402-1.477(t,J=7.2Hz,2H);1.068-1.196(m,30H);0.688(t,J=7.2Hz,3H)。
MS(ESI)m/e:710.35[M] +
实施例5
(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)四 氢呋喃-2-基)甲基(2-(十六烷二硫基)乙基)磷酸酯钠盐(II-1-1)
Figure PCTCN2022141023-appb-000075
向(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)基四氢呋喃-2-基)甲基(2-(十六烷二硫基)乙基)磷酸酯(NTV-29)(6.56g,10mmol)中加入无水乙醇50ml。将反应物在搅拌下加热至60℃使其溶解。在该温度下,向溶液中加入2NNaOH乙醇溶液(5ml)并搅拌10分钟。将反应液温度降至室温,在4℃放置过夜,过滤,得到(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)四氢呋喃-2-基)甲基(2-(十六烷二硫基)乙基)磷酸酯钠盐烷氧基)(II-1-1)白色固体(5.9g,92%)。
实施例6
(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)四 氢呋喃-2-基)甲基(3-(十八烷二硫基)乙基)磷酸酯铵盐(II-1-2)
Figure PCTCN2022141023-appb-000076
向烧瓶中加入(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)四氢呋喃-2-基)甲基(2-(十八烷二硫基)乙基)磷酸酯(I-2)(6.83g,0.01mol)、2-丙醇(50ml)和28-30%(w/w)氨水溶液(1ml)。搅拌反应液并使之在62-80℃回流以实现溶出(经约10分钟)。在回流下使 溶液停止搅拌,放置15分钟以上。使溶液在低于25℃的温度下冷却8-24小时,然后再将混合物冷却至5±5℃并保持至少1小时。过滤产物,并用冷却的2-丙醇(0±5℃,30ml)洗涤。将白色固体产物在30-35℃干燥25小时,得到(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)四氢呋喃-2-基)甲基(3-(十八烷二硫基)乙基)磷酸酯铵盐(II-1-2)(6.0g,92.3%)。
实施例7
((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)四 氢呋喃-2-基)甲基(3-(十六烷氧基)丙基)磷酸酯(NTV-118)
Figure PCTCN2022141023-appb-000077
步骤1:
2-氯苯基(((3aR,4R,6S,6aR)-6-(2,4-二氧代-3,4-二氢嘧啶-1(2H) 基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)甲基)(3-(十 六烷氧基)丙基)磷酸酯(1-1A2)
Figure PCTCN2022141023-appb-000078
(1)1-((3aR,4S,6R,6aR)-6-(羟基甲基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)嘧啶-2,4(1H,3H)-二酮(1-1a)
Figure PCTCN2022141023-appb-000079
将1-((3R,4S)-3,4-二羟基-5-(羟甲基)四氢呋喃-2-基]嘧啶-2,4(1H,3H)-二酮(尿苷)(48.4g,200mmol)和丙酮(2000mL)加入到反应瓶中。在室温搅拌下,向所得浆状物中加入硫酸(98%,2mL)。将反应液于室温搅拌48小时。得到澄清溶液后,用三乙胺将反应液的pH调至8。将溶液减压浓缩,得到浅黄色油状残留物。将残留物用乙酸乙酯(400mL)溶解。将所得溶液依次用水(200mlx2)、饱和碳酸氢钠水溶液(200mlx2)、水(200mlx2)和饱和盐水(200mlx2)洗涤。分出有机层,用无水硫酸钠干燥,减压浓缩,得到白色1-((3aR,4S,6R,6aR)-6-(羟基甲基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)嘧啶-2,4(1H,3H)-二酮(1-1a)(33g)。
(2)3-(十六烷氧基)丙-1-醇(1-1b-1)
HOCH 2CH 2CH 2-O-CH 2(CH 2) 14CH 3
1-1b-1
反应路线:
Figure PCTCN2022141023-appb-000080
I.对甲苯磺酸1-(十六烷基)酯
Figure PCTCN2022141023-appb-000081
在氮气保护下,向反应器中加入1-(十六烷基)醇(48.4g)、无水二氯甲烷(400mL)和三乙胺(26.3g)。将反应混合物冷却至0±5℃。在该温度下滴加对甲苯磺酰氯(41g)的二氯甲烷(200ml)溶液。加毕,将反应混合物升温至20-30℃并搅拌2小时。将反应液保持在20-30℃,并用水 (150mL)进行稀释。分离有机层,用水(50ml)洗涤并浓缩至干,得到为浅黄色固体的对甲苯磺酸1-(十六烷基)酯(48.7g,96%)。
II.3-(十六烷氧基)丙-1-醇(1-1b-1)
在氮气下,向反应器中加入1,3-丙二醇(40.7g)和N-甲基甲酰胺(NMF)(300ml)。将反应液冷却至0±5℃,分批小心地加入氢化钠(10.7g,60%)。加入完成后,将反应液在室温下搅拌另外2小时。在20-35℃向反应混合物中缓慢地加入溶解于DMF(100ml)中的对甲苯磺酸十六烷基酯(52.8g)的溶液。将该溶液在20-35℃搅拌24小时。将反应混合物冷却至0±5℃,用水(150ml)缓慢地稀释,并用乙酸乙酯(2x250ml)萃取。将有机相用水(200ml)洗涤,经无水Na 2SO 4干燥并浓缩,得到棕色的油。将粗产物溶解于甲醇(200ml)中,并在0-30℃放置12小时。过滤生成的固体杂质并弃去,浓缩滤液。向浓缩物中加入乙腈(400ml),并将混合物在5-15℃放置16小时。过滤固体并在25-30℃干燥,得到为白色固体的3-(十六烷氧基)丙-1-醇(1-1b-1)(31.1g,77%)。
(3) 2-氯苯基(((3aR,4R,6S,6aR)-6-(2,4-二氧代-3,4-二氢嘧啶-1(2H) 基)-2,2-二甲基四氢呋喃并[3,4-d]1,3]间二氧杂环戊烷-4-基)甲基)(3-(十 六烷氧基)丙基)磷酸酯(1-1A2)
Figure PCTCN2022141023-appb-000082
将1,2,4-三唑(1c)(13.8g,2002.12mmol)和三乙胺(10.1g,200mmol)于干燥四氢呋喃(200mL)中的溶液加入到2-氯苯基二氯磷酸酯(1d-1)(24.5g,100mmol)于干燥四氢呋喃(150mL)中的溶液中。将所得反应液于室温反应1小时。将生成的固体滤除,并向所得滤液中加入1-((3aR,4S,6R,6aR)-6-(羟基甲基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间 二氧杂环戊烷-4-基)嘧啶-2,4(1H,3H)-二酮(1-1a)(28.4g,100mmol)和1-甲基咪唑(8.4ml,100mmol)于四氢呋喃(150mL)中的溶液。加毕,于室温搅拌反应1小时。向反应液中加入3-(十六烷氧基)丙-1-醇(1b-3)(30g,100mmol)。将所得反应液于室温搅拌过夜,反应完毕后减压浓缩,并将残留物用乙酸乙酯(400mL)溶解。将所得溶液依次用水(200mlx2)、饱和碳酸氢钠水溶液(200mlx2)、水(200mlx2)和饱和盐水(200mlx2)洗涤。分出有机层,经无水硫酸钠干燥并减压浓缩,得到2-氯苯基(((3aR,4R,6S,6aR)-6-(2,4-二氧代-3,4-二氢嘧啶-1(2H)基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)甲基(3-(十六烷氧基)丙基)磷酸酯(1-1A2)(53g,70%)无色油状物,其无需进一步纯化可直接用于下步反应。
1HNMR(600MHz,/甲醇-d4):δ7.615(dd,J=8.4Hz,J=3.6Hz,1H);7.510(d,J=7.8Hz,1H);7.326(d,J=7.8Hz,1H);7.228(d,J=7.8Hz,1H),5.771(ddd,J=1.8,J=4.8,J=6Hz,1H);5.659(dd,J=7.8Hz,1H);5.087(ddd,J=25.8Hz,J=7.8Hz,1H);4.418-4.479(m,J=7.8Hz,J=3.6Hz,2H);4.389-4.342(m,J=6.0Hz,J=4.2Hz,2H);3.652(t,J=6.6Hz,2H);3.545-3.501(m,2H),3.454-3.401(m,2H);1.988-1.953(m,2H);1.558(s,6H);1.366-1.361(m,2x2H);1.352-1.228(m,26H);0.87(t,J=7.2Hz,3H)。
13CNMR(600MHz,/甲醇-d4):164.693(d,J=13Hz);150.454;146.399;146.357;143.445(t,J=30Hz);143.268;130,485;127.967;126.294(d,J=30Hz);121.260(t,J=7.2Hz);114.031(t,J=6Hz);101.490;94.577;94.839;85.482(d,J=30Hz);85.635(d,J=30Hz);84.271;81;008(d,J=24Hz);70.736;70.695;68.348(t,J=27Hz);67.404;66.613(d,J=25Hz);65.815(d,J=22Hz);58.787;32.320;31.702;30.163(d,J=27Hz);29.423;29.410;29.396;29.369;29.230;26.033;25.903;24.124;22.364;13.103。
31P NMR(600MHz,CD 3OD):δ-7.173;-7.185。
MS(ESI)m/e:757.35803[M+H] +
步骤2:
2-氯苯基(3-(十六烷氧基)丙基)((3aR,4R,6S,6aR)-6-(4-(1,2,4-三唑 基)-2-氧代嘧啶-1(2H)基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环 戊烷-4-基)甲基)磷酸酯(1-1A3)
Figure PCTCN2022141023-appb-000083
将2-氯苯基(((3aR,4R,6S,6aR)-6-(2,4-二氧代-3,4-二氢嘧啶-1(2H)基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)甲基(3-(十六烷氧基)丙基)磷酸酯(1-1A2)(76g,100.0mmol)在搅拌下溶解于乙腈(400.0mL)中,待溶解后加入1,2,4-三唑(1c)(48.0g,700.0mmol)和三乙胺(81g,800.0mmol),并在搅拌下使固体溶解。将反应液冷却至0℃,在氮气氛下缓慢滴加三氯氧磷(14.5ml,150.0mmol)。加毕,使反应液温度升至室温,继续搅拌2小时使反应进行完全,并通过TLC(100%EtOAc)监测反应。反应进行完毕后,加入水(500mL)和乙酸乙酯(400mL),于室温搅拌15分钟以淬灭反应。分出有机层,将有机相依次用水(2x100mL)、0.5N HCl(200mL)和饱和盐水(2x100mL)洗涤,经无水硫酸镁干燥并过滤。将滤液浓缩,得到过渡中间体2-氯苯基(3-(十六烷氧基)丙基)((3aR,4R,6S,6aR)-6-(4-(1,2,4-三唑基)-2-氧代嘧啶-1(2H)基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)甲基)磷酸酯(1-1A3)(80.6g,100%),其不需要纯化直接用于下步反应。
步骤3:
2-氯苯基(3-(十六烷氧基)丙基)(((3aR,4R,6S,6aR)-6-(4-羟氨基)-2- 氧代嘧啶-1(2H)-基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷 -4-基)甲基)磷酸酯(1-1A4)
Figure PCTCN2022141023-appb-000084
将上面所得的产物(1-1A3)(40.6g,50mmol)加入异丙醇(300mL)和三乙胺(7.5g,75mmol)的混合物中,于室温下搅拌以使固体溶解。然后,加入盐酸羟胺(5.2g,75mmol),搅拌1-2小时后得到淡黄色反应液,并通过TLC(EtOAc)监测反应完毕。在小于45℃的温度下减压浓缩以除去溶剂。将残留物溶解于乙酸乙酯(300ml)中,并将有机相依次用水(150mlx2)、饱和盐水(150mlx2)洗涤,经无水硫酸钠干燥并过滤。将滤液减压浓缩。将浓缩物用石油醚重结晶,得到白色固体2-氯苯基(3-(十六烷氧基)丙基)(((3aR,4R,6S,6aR)-6-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)甲基)磷酸酯(1-1A4)(34.7g,90%)。
1HNMR(600MHz,/甲醇-d4):δ7.517-7.504(d,J=7.8Hz,1H);7.459-7.445(d,J=7.8Hz,1H);7.347-7.321(t,J=7.8Hz,1H);7.245-7.232(d,J=7.8Hz,1H);6.851-6.826(t,J=7.8Hz,1H);5.700-5.691(t,J=3Hz,1H);5.576-5.537(dd,J=7.8Hz,1H);5.014-4.944(ddd,J=33Hz,J=1.8Hz,1H),4.443-4.415(dd,J=6Hz,J=4.2Hz,2H);4.382-4.349(dd,J=6.6Hz,2H);4.298-4.249(m,J=3.6Hz,J=2.5Hz,1H);3.582-3.509(t,J=6.6Hz,2H);3.428-3.407(t,J=6.6Hz,2H);1.996-1.965(m,J=6.6,2H);1.551(s,6H);1.398-1.327(m,2x2H);1.342-1.300(m,26H);0.87(t,J=7.2Hz,3H)。
13CNMR(600MHz,/甲醇-d4):δ149.678;148.760;146.369;144.751;132.764;130.475;127.985;126.793;121.285;114.067;98.126;93.290;84.605;83.846;83.792;80.816;80.766;79.646;70.730;68.402;66.557;65.812;65.775;31.685;30.150;30.104;29.393;29.381;29.346;29.202; 29.086;26.153;26.084;25.886;24.130;22.345;13.057。
31P NMR(600MHz,CD 3OD):δ-7.135,-7.196。
MS(ESI)m/e:772.36597[M+H] +
步骤4:
3-(十六烷氧基)丙基(((3aR,4R,6S,6aR)-6-(4-羟氨基)-2-氧代嘧啶 -1(2H)-基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)甲 基)磷酸酯(1-1A5)
Figure PCTCN2022141023-appb-000085
将1,1,3,3-四甲基胍(4b)(12.9g,112.0mmol)和2-吡啶甲醛肟(4c)(13.70g,112.0mmol)于THF(120ml)中的溶液加入到2-氯苯基(3-(十六烷氧基)丙基)(((3aR,4R,6S,6aR)-6-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)甲基)磷酸酯(1-1A4)(15.4g,20mmol)于THF(360mL)中的溶液中,并将所得反应溶液于室温搅拌过夜。通过TLC(二氯甲烷∶甲醇=20∶1,v/v)监测反应完毕后,向反应液中加入乙酸乙酯(480ml)和冷盐酸水溶液(pH=1-2,200ml)。分出有机层,并将水层用乙酸乙酯/四氢呋喃(1∶1,v/v)溶液(100ml×3)萃取。合并有机层,用2N氢氧化钠溶液(200ml×2)萃取。将水相合并,用盐酸酸化至溶液的pH为1-2,并用乙酸乙酯(200ml×2)萃取。将乙酸乙酯萃取液合并,用无水硫酸钠干燥并过滤。将滤液减压浓缩。将浓缩物用石油醚重结晶,得到类白色固体3-(十六烷氧基)丙基(((3aR,4R,6S,6aR)-6-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)甲基)磷酸酯(1-1A5)(9.2g,70%)。
1H NMR(600MHz,CD 3OD):δ7.765-7.752(d,J=7.8Hz,0.2H);δ 7.608-7.594(d,J=8.4Hz,0.8H);5.903-5.868(dd,J=2.4,Hz,J=18Hz,1H);5.854-5.726(dd,J=8.4Hz,J=68Hz,1H);4.421-4.401(m,1H);4.179-4.145(m,2H);4.127-4.067(m,2H);4.044-4.011(dd,J=6.0Hz,2H);3.535-3.532(t,J=6.6Hz,2H);3.445-3.427(t,J=6.6Hz,2H);1.934-1.886(m,J=6.6Hz,2H);1.570(s,6H);1.375(s,2H);1.308(m,26H);0.913(t,J=7.2Hz,3H)。
13C NMR(600MHz,CD 3OD):δ156.255;152.349;142.464;138.394;113.857;113.638;109.757;101.583;93.084;85.493;84.449;84.230;80.958;70.742;66.577;66.436;65.674;63.339;63.297;31.689;30.542;30.493,29.402;29.373;29.252;29.091;26.122;25.903;24.128;22.350;13.066;
31P NMR(600MHz,CD 3OD):δ-0.548,-0.547。
MS(ESI)m/e:662.37604[M+H] +;684.35162[M+Na] +
步骤5:
(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)四 氢呋喃-2-基)甲基(3-(十六烷氧基)丙基)磷酸酯(NTV-118)
将3-(十六烷氧基)丙基(((3aR,4R,6S,6aR)-6-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)甲基)磷酸酯(1-1A5)(6.6g,10mmol)溶解于TFA∶H 2O(9∶1,80mL)中,并将反应液于室温搅拌45分钟。将反应液减压浓缩,并将浓缩物用甲苯(100ml)减压浓缩2次。向残留物中加入甲醇(100ml),减压浓缩3次后用乙醇重结晶,得到(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)四氢呋喃-2-基)甲基(3-(十六烷氧基)丙基)磷酸酯(NTV-118)(5.6g,90%)。
1HNMR(600MHz,CD 3OD):δ8.013(d,J=8.4Hz,0.2H);δ7.229(d,J=8.4Hz,0.8H);5.993(d,J=4.8,Hz,0.2H);5.961(d,J=4.8Hz,0.8H);5.813(d,J=8.4Hz,0.2H);5.701(d,J=8.4Hz,0.8H);4.226(m,1H);4.068(m,1H);4.028(m 1H);3.966(dd,J=6.0Hz,2H);3.543(t,J=6.0Hz,2H);3.434(t,J=6.6Hz,2H);1.912(m,J=6.6Hz,2H);1.579(m,J=6.6Hz,2H);1.357-1.900(m,26H);0.913(t,J=7.2Hz,3H)。
13C NMR(600MHz,CD 3OD):δ150.505;144.958;141.094;130.706;101.728;98.252;87.382;83.467(d,J=59Hz);74.484;73.005;70.252;70.633;70.390;66.971;64.782;62.459;62.422;30.767;30.717;29.737;29.263;29.090;25.893;22.349;22.394;13.068。
31P NMR(600MHz,CD 3OD):δ0.328,0.293。
MS(ESI)m/e:622.34363[M+H] +
实施例8
((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)四 氢呋喃-2-基)甲基(2-(十八烷氧基)乙基)磷酸酯(NTV-125)
Figure PCTCN2022141023-appb-000086
采用与实施例7类似的合成方法制备(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)四氢呋喃-2-基)甲基(2-(十八烷氧基)乙基)磷酸酯(NTV-125)。
步骤1:
2-氯苯基(((3aR,6aR)-6-(2,4-二氧代-3,4-二氢嘧啶-1(2H)基)-2,2-二 甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷4-基)甲基)(2-(十八烷氧基) 乙基)磷酸酯(1-1B2)
Figure PCTCN2022141023-appb-000087
(1)3-(十八烷氧基)乙-1-醇(1-1b-2)
H 2CH 2COH-O-CH 2(CH 2) 16CH 3
1-1b-2
I.对甲苯磺酸十八烷基酯
在氮气保护下,向反应器中加入1-十八烷基醇(27.4g)、无水二氯甲烷(300mL)和三乙胺(30g)。将反应混合物冷却至0±5℃。在该温度下滴加对甲苯磺酰氯(22g)的二氯甲烷(200ml)溶液。加毕,将反应混合物升温至20-30℃并搅拌2小时。将反应液保持在20-30℃,并用水(150mL)进行稀释。分离有机层,用水(50ml)洗涤并浓缩至干,得到为浅黄色固体的对甲苯磺酸十八烷基酯(34.6g,90%)。
II.2-(十八烷氧基)乙醇(1-1b-2)
在氮气下,向反应器中加入1,2-乙二醇(12.2g)和NMF(300ml)。将反应液冷却至0±5℃,分批小心地加入氢化钠(10.7g,60%)。加入完成后,将反应液在室温下搅拌另外2小时。在20-55℃向反应混合物中缓慢地加入溶解于DMF(100ml)中的对甲苯磺酸十八烷基酯(42.68g)的溶液。将该溶液在20-35℃搅拌24小时。将反应混合物冷却至0±5℃,用水(150ml)缓慢地稀释,并用乙酸乙酯(2x250ml)萃取。将有机相用水(200ml)洗涤,经无水Na 2SO 4干燥并浓缩,得到棕色的油。将粗产物溶解于甲醇(200ml)中,并在0-30℃放置12小时。过滤生成的固体杂质并弃去,浓缩滤液。向浓缩物中加入乙腈(400ml),并将混合物在5-15℃放置16小时。过滤固体并在25-30℃干燥,得到为白色固体的2-(十八烷氧基)乙醇(1-1b-2)(25.3g,81.6%)。
(2)2-氯苯基(((3aR,6aR)-6-(2,4-二氧代-3,4-二氢嘧啶-1(2H)基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)甲基)(2-(十八烷氧基)乙基)磷酸酯(1-1B2)
参照实施例7的步骤1中所述的制备2-氯苯基(((3aR,4R,6S,6aR)-6-(2,4-二氧代-3,4-二氢嘧啶-1(2H)基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)甲基)(3-(十六烷氧基)丙基)磷酸酯烷氧基)(1-1A2)的方法,用2-(十八烷氧基)乙醇(1-1b-2)代替3-(十六烷氧基)丙-1-醇(1-1b-1)进行反应,得到2-氯苯基(((3aR,6aR)-6-(2,4- 二氧代-3,4-二氢嘧啶-1(2H)基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)甲基(2-(十八烷氧基)乙基)磷酸酯(1-1B2)。
具体而言,按下表进行投料:
Figure PCTCN2022141023-appb-000088
将1,2,4-三唑(1c)和三乙胺于干燥四氢呋喃中的溶液加入到2-氯苯基二氯磷酸酯(1d-1)于干燥四氢呋喃中的溶液中。将所得反应液于室温反应1小时。将生成的固体滤除,并向所得滤液中加入1-[(3aR,6aR)-6-(羟甲基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷4-基]嘧啶-2,4-二酮(1-1a)和1-甲基咪唑于四氢呋喃中的溶液。加毕,于室温搅拌反应1小时。向反应液中加入2-(十八烷氧基)乙醇(1-1b-2)。将所得反应液于室温搅拌过夜,反应完毕后减压浓缩,并将残留物用乙酸乙酯溶解。将所得溶液依次用水、饱和碳酸氢钠水溶液、水和饱和盐水洗涤。分出有机层,经无水硫酸钠干燥、过滤,将滤液减压浓缩,得2-氯苯基(((3aR,4R,6S,6aR)-6-(2,4-二氧代-3,4-二氢嘧啶-1(2H)基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)甲基(2-(十八烷氧基)乙基)磷酸酯(1-1B2)(17.5g,65%)无色油状物,其无需进一步纯化可直接用于下步反应。用于结构鉴定的样品可通过色谱柱(CH 2Cl 2+0至8%MeOH)纯化。
1HNMR(600MHz,CD 3OD):δ7.589-7.566(dd,J=7.8Hz,1H);7.473-7.438(m,J=7.8Hz,J=1.8Hz,2H);7.301-7.275(ddd,J=7.8Hz,J=1.8Hz,1H);5.767-5.759(t,J=2.4Hz,1H);5.654-5.614(dd,J=7.8,Hz,1H); 4.480-4.412(m,,5H);3.672-3.642(m,J=3Hz,2H);3.494-3.447(t,J=6.6Hz,2H);1.569-1.546(m,J=6.6Hz,J=7.2Hz 2H);1.533(s,3H);1.337(m,2H);1.337(s,3H);1.278(m,30H);0.892(t,J=7.2Hz,3H)。
13C NMR(600MHz,CD 3OD):δ164.625(d,J=12Hz);150.467;146.422(d,J=25.8Hz);143.214;143.004;130.470;127.956;126.337(d,J=26.4Hz);125.252(m,J=19.2Hz,J=9Hz);121.348(m,J=9Hz);114.053;101.594;94.530;94.264;85.628(d,J=30.6Hz);85.453(d,J=31.8Hz);80.986(d,J=33Hz);84.282(m,J=25.8Hz);71.031;69.004(m,J=27Hz);68.499(t,J=25.2Hz);68.379(t,J=25.2Hz);31.736;29.468;29.437;29.423;29.402;29.278;29.148;26.146;25.874;24.232;22.400;13.182。
31P:δ-7.1610;-7.1962。
MS(ESI)m/e:771.40[M+H] +
步骤2:
2-氯苯基(((3aR,4R,6S,6aR)-6-(4-(1H-1,2,4-三唑-1-基)-2-氧代嘧啶 -1(2H)-基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)甲 基(2-(十八烷氧基)乙基)磷酸酯(1-1B3)
Figure PCTCN2022141023-appb-000089
参照实施例7的步骤2中所述的制备2-氯苯基(3-(十六烷氧基)丙基)((3aR,4R,6S,6aR)-6-(4-(1,2,4-三唑基)-2-氧代嘧啶-1(2H)基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)甲基)磷酸酯(1-1A3)的方法,用2-氯苯基(((3aR,4R,6S,6aR)-6-(2,4-二氧代-3,4-二氢嘧啶-1(2H) 基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)甲基(2-(十八烷氧基)乙基)磷酸酯烷氧基)(1-1B2)代替2-氯苯基(((3aR,4R,6S,6aR)-6-(2,4-二氧代-3,4-二氢嘧啶-1(2H)基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烯-4-基)甲基(3-(十六烷氧基)丙基)磷酸酯烷氧基)(1-1A2)进行反应,得到2-氯苯基(((3aR,4R,6S,6aR)-6-(4-(1H-1,2,4-三唑-1-基)-2-氧代嘧啶-1(2H)-基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)甲基(2-(十八烷氧基)乙基)磷酸酯(1-1B3)。
具体而言,按下表进行投料:
Figure PCTCN2022141023-appb-000090
1HNMR(600MHz,CD 3OD):δ9.297-9.285(t,J=3.5Hz,0.5H);8.404(m,0.5H);8.325-8.310(dd,J=7.2Hz,J=1.8Hz,0.5H);7.465-7.439(m,J=6.6Hz,J=2.4Hz,1H);7.444-7.379(m,J=7.2Hz,J=1.8Hz,1H);7.321-7.273(m,J=8.4Hz,J=2.4Hz,J=1.8.Hz 1H);7.245-7.169(m,J=3Hz,1H);7.141-7.068(dd,J=7.2Hz,J=36.6Hz,1H);6.296-5.987(dd,J=7.8Hz,J=32.4Hz,J=11.4Hz1H);5.464-5.337(m,2H);4.664-4.309(m,4H);3.002-2.948(m,J=6.6Hz,2H);2.723-2.667(m,J=7.2Hz,2H);2.145(m,J=7.2Hz,3H);2.139 (s,3H);1.366-1.362(m,2H);1.272(s,28H);0.882(t,J=7.2Hz,3H)。
13C NMR(600MHz,CD 3OD):δ159.838;154.938;153.533(d,J=19.8Hz);149.705;146.299(d,J=25.2Hz);143.377(d,J=15Hz);130.482(d,J=37.8Hz);127.976(d,J=27.6Hz);126.376;126.234;125.107(tJ=30Hz);121.247(d,J=9Hz);113.842;97.068;96.734;94.845(d,J=79.8Hz);86.964(d,J=31.2Hz);86.804(d,J=31.2Hz);85.271;85.159;81.106;81.006;71.079;68.981(m,J=26.4Hz);68.600;68.500(m,J=24Hz,);31.720;29.440;29.427;29.384;29.370;29.246;29.128;26.155(d,J=10.4Hz);25.845;24.247(d,J=18Hz);22.393;13.223。
31P:δ-7.195;-7.291。
MS(ESI)m/e:822.40[M] +
步骤3:
2-氯苯[2-(十八烷氧基)乙基(((3aR,4R,6S,6aR)-6-(4-羟氨基)-2-氧代 嘧啶-1(2H)-基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基) 甲基)磷酸酯(1-1B4)
Figure PCTCN2022141023-appb-000091
按下表进行投料:
Figure PCTCN2022141023-appb-000092
将上面所得的产物(1-1B3)(8.2g,10mmol)加入异丙醇(100mL)中,于室温下搅拌以使固体溶解。然后,加入盐酸羟胺(1.1g,15mmol)和氢氧化钠(0.6g,15mmol),搅拌1小时后得到淡黄色反应液,并通过TLC(EtOAc)监测反应完毕。在小于45℃的温度下减压浓缩以除去溶剂。将残留物溶解于乙酸乙酯(100ml)中,并将有机相依次用水(50mlx2)、饱和盐水(50mlx2)洗涤,经无水硫酸钠干燥并过滤。将滤液减压浓缩。将浓缩物用石油醚重结晶,得到白色固体2-氯苯基(2-(十八烷氧基)乙基(((3aR,4R,6S,6aR)-6-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)甲基)磷酸酯(1-1B4)(7.3g,93%)。
1HNMR(600MHz,CD 3OD):δ7.586-7.573(m,J=5.4Hz,J=6Hz0.5H);8.404(m,0.5H);8.325-8.310(dd,J=7.2Hz,J=1.8Hz,0.5H);7.465-7.439(m,J=6.6Hz,J=2.4Hz,1H);7.444-7.379(m,J=7.2Hz,J=1.8Hz,1H);7.321-7.273(m,J=8.4Hz,J=2.4Hz,J=1.8.Hz,1H);7.245-7.169(mJ=3Hz,1H);7.141-7.068(dd,J=7.2Hz,J=36.6Hz,1H);6.296-5.987(dd,J=7.8Hz,J=32.4Hz,J=11.4Hz,1H);5.464-5.337(m,2H);4.664-4.309(m,4H);3.002-2.948(m,J=6.6Hz,2H);2.723-2.667(m,J=7.2Hz,2H);2.145(m,J=7.2Hz,3H);2.139(s,3H);1.366-1.362(m,2H);1.272(s,28H);0.882(t,J=7.2Hz,3H)。
13C NMR(600MHz,CD 3OD):δ150.477;149.688;146.437(m,J=7.8Hz,J=38Hz);144.757;143.254;143.042;132.875;132.686;130.466;127.970(d,J=7.2Hz);126.322(d,J=32.4Hz);125.264(t,J=28.2Hz);121.397( dd,J=9Hz,J=16.8Hz);114.046(d,J=9Hz);101.604;101.624;98.240(d,J=23.4Hz);94.589;94.318;93.424;93.132;84.665(d,J=30Hz);84.496(d,J=31.2Hz);84.219;83.817(d,J=27.6Hz);80.997(d,J=32.4Hz);80.833(d,J=35.4Hz);71.049;69.011(d,J=27Hz);68.452(m,J=7.8Hz,J=17.4Hz,J=9Hz);31.750;29.452;29.445;29.418;29.229;29.165;26.242(J=33Hz);25.889;24.319(d,J=33.6Hz);22.451;13.218。
31P:δ-7.181;-7.193。
MS(ESI)m/e:785.3801[M] +
步骤4:
2-(十八烷氧基)乙基(((3aR,4R,6S,6aR)-6-(4-羟氨基)-2-氧代嘧啶 -1(2H)-基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)甲 基)磷酸酯(1-1B5)
Figure PCTCN2022141023-appb-000093
参照实施例7的步骤4中所述的方法,用2-氯苯基(2-(十八烷氧基)乙基(((3aR,4R,6S,6aR)-6-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)甲基)磷酸酯)(1-1B4)代替2-氯苯基(3-(十六烷氧基)丙基)(((3aR,4R,6S,6aR)-6-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)甲基)磷酸酯)(1-1A4)制备1-1B5(白色固体产物3.1g,60%收率)。
具体而言,按下表进行投料:
Figure PCTCN2022141023-appb-000094
Figure PCTCN2022141023-appb-000095
1HNMR(600MHz,CD 3OD):δ7.764-7.750(d,J=8.4Hz,0.2H);7.612-7.599(d,J=8.4Hz,0.8H);5.918-5.910(d,J=4.8,Hz,0.2H);5.901-5.896(d,J=3Hz,0.2H);5.872-5.867(d,J=8.4Hz,0.8H);5.854-5.840(d,J=8.4Hz,0.8H);4.168-4.008(m,,5H);3.615-3.600(t,J=4.2Hz,2H);3.482-3.460(t,J=6.6Hz,2H);1.548(s,3H);1.353(s,3H);1.2845(m,30H);0.908-0.884(t,J=7.2Hz,3H)。
13C NMR(600MHz,CD 3OD):δ150.898;148.265;142.295;138.749;113.084;113.527;101.624;93.106;92.892;92.762;85.524;84.525;81.036;71.035;69.792;65.913;65.688;65.384;31.629;29.476;29.387;29.363;29.249;29.075;26.132;25.867;24.135;22.337;13.049。
31P NMR(600MHz,CD 3OD):δ-0.345,-0.545。
MS(ESI)m/e:676.3802[M+H] +
步骤5:
(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)四 氢呋喃-2-基)甲基(2-(十八烷氧基)烷氧基)乙基)磷酸酯(NTV-125)
Figure PCTCN2022141023-appb-000096
参照实施例7的步骤5中所述的方法用2-(十八烷氧基)乙基(((3aR,4R,6S,6aR)-6-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烷-4-基)甲基)磷酸酯(1-1B5)与三氟醋酸反应,得到目标产物(NTV-125)。
具体而言,按下表进行投料:
Figure PCTCN2022141023-appb-000097
将2-(十八烷氧基)乙基(((3aR,4R,6S,6aR)-6-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)-2,2-二甲基四氢呋喃并[3,4-d][1,3]间二氧杂环戊烯-4-基)甲基)磷酸酯(1-1B5)溶解于TFA:H2O(9∶1,80mL)中,并将反应液于室温搅拌45分钟。将反应液减压浓缩,并将浓缩物用甲苯减压浓缩2次。向残留物中加入甲醇(100ml),减压浓缩3次后用乙醇重结晶,得到类白色(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)四氢呋喃-2-基)甲基(3-(十八烷氧基)乙基)磷酸酯(NTV-125)固体(3.5g,92%)。
1HNMR(600MHz,CD 3OD):δ7.834(s,0.2H);7.683-7.670(d,J=7.8Hz,0.8H);5.960-5.952(d,J=4.8Hz,0.2H);5.928-5.921(d,J=4.2Hz 0.8H);5.877-5.864(d,J=7.8,Hz,1H);4.219-4.026(m,5H);3.624-3.615(t,J=5.4Hz,2H);3.490-3.467(t,J=7.2Hz,2H);1.583-1.537(m,J=7.2Hz,2H);1.368-1.229(m,30H);0.906-0.883(t,J=7.2Hz,3H)。
13C NMR(600MHz,CD 3OD):δ149.601;146.098;141.054;136.531;125.935;118.695;114.004;109.279;105.619;94.171;88.764;78.038;77.821;77.604,73.894;71.116;69.893;65.035;31.701;29.442;29.281;29.102;25.859,22.372;13.182。
31P NMR(600MHz,CD 3OD):δ0.757,0.231。
MS(ESI)m/e:636.35[M+H] +
实施例9
(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)四 氢呋喃-2-基)甲基)(3-(十六烷氧基)烷氧基)丙基)磷酸酯钠盐(II-2-1)
Figure PCTCN2022141023-appb-000098
向(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)四氢呋喃-2-基)甲基(3-(十六烷氧基)烷氧基)丙基)磷酸酯(NTV-118)(6.21g,10mmol)中加入无水乙醇50ml。将反应物在搅拌下加热至60℃使其溶解。在该温度下,向溶液中加入2N NaOH乙醇溶液(5ml)并搅拌10分钟。将反应液温度降至室温,在4℃放置过夜,过滤,得到(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)四氢呋喃-2-基)甲基(3-(十六烷氧基)烷氧基)丙基)磷酸酯钠盐(II-2-1)白色固体(5.9g,92%)。
实施例10
(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)四 氢呋喃-2-基)甲基(2-(十八烷氧基)烷氧基)乙基)磷酸酯单铵盐(II-2-2)
Figure PCTCN2022141023-appb-000099
向烧瓶中加入(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)四氢呋喃-2-基)甲基(2-(十八烷氧基)烷氧基)乙基)磷酸酯 (NTV-125)(6.3g,0.01mol)、2-丙醇(50ml)和28-30%(w/w)氨水溶液(1ml)。搅拌反应液并使之在62-80℃回流以实现溶出(经约10分钟)。在回流下使溶液停止搅拌,放置15分钟以上。使溶液在低于25℃的温度下冷却8-24小时,然后再将混合物冷却至5±5℃并保持至少1小时。过滤产物,并用冷却的2-丙醇(5±5℃,30ml)洗涤。将白色固体产物在30-35℃干燥25小时。得产物(((2R,3S,4R,5S)-3,4-二羟基-5-(4-羟氨基)-2-氧代嘧啶-1(2H)-基)四氢呋喃-2-基)甲基(2-(十八烷氧基)烷氧基)乙基)磷酸酯单铵盐(II-2-2)(6.0g,92.3%)。
实施例11
片剂剂型
以实施例1的化合物(其可以被替换成本公开的任何一种其它实施例的化合物或表1中公开的其它具体化合物)为活性成分,制备本公开的不同规格的示例性片剂剂型(其为速释片剂)。所述不同规格的示例性片剂剂型的各成分及其功能和用量显示在下面的表2-4中,并且它们可通过包括以下步骤的方法来制备:(1)混合预定量的各成分,从而制备掺混物;和(2)通过常规压制方法压制所述掺混物,从而得到所述片剂剂型。
表2包含20mg实施例1的化合物的片剂的组成(每1000片用量)
成分 功能 重量(g)
实施例1的化合物 活性成分 20
硅化微晶纤维素 稀释剂、粘合剂、助流剂 33
甘露醇 稀释剂、粘合剂 53
微晶纤维素和甘露醇(3∶1,w/w) 稀释剂 16
交聚维酮 崩解剂 5.0
硬脂酸镁 润滑剂 1.3
表3包含50mg实施例1的化合物的片剂的组成(每1000片用量)
成分 功能 重量(g)
实施例1的化合物 活性成分 50
硅化微晶纤维素 稀释剂、粘合剂、助流剂 32
微晶纤维素和甘露醇(3∶1,w/w) 稀释剂、粘合剂 14
甘露醇 稀释剂 45
交聚维酮 崩解剂 5
硬脂酸镁 润滑剂 1.0
表4包含100mg实施例1的化合物的片剂的组成(每1000片用量)
成分 功能 重量(g)
实施例1的化合物 活性成分 100
硅化微晶纤维素 稀释剂、粘合剂、助流剂 64
微晶纤维素和甘露醇(3∶1,w/w) 稀释剂、粘合剂 28
甘露醇 稀释剂 90
交聚维酮 崩解剂 10
硬脂酸镁 润滑剂 2.0
实施例12抗病毒细胞保护实验
为了研究本公开的化合物在体外抗病毒的药效,使用细胞病变效应(CPE)实验测定了示例性化合物对细胞的最大无毒浓度(TC 50)、对病毒的半数抑制浓度(IC 50)和选择指数(SI),其中以利巴韦林(RBV)作为阳性对照药物和β-D-N 4-羟基胞苷(NHC,EIDD-1931)作为参考药物。
实验中选用的细胞为蚊源细胞(C6/36)、非洲绿猴肾细胞(VeroE6)、犬肾细胞(MDCK)、人肝癌细胞(Huh7细胞)和人肺癌细胞(H460细胞)。所述细胞的培养基本情况如下表5所示:
表5细胞的培养基本情况
细胞 培养液 培养条件 是否胰酶消化,消化时间
C6/36细胞 RPMI-1640 28℃,5%CO 2
Vero细胞 DMEM 37℃,5%CO 2 2-3min
MDCK细胞 DMEM 37℃,5%CO 2 7-10min
H460细胞 DMEM 37℃,5%CO 2 1-2min
Huh7 DMEM 37℃,5%CO 2 2-3min
实验中选用的病毒为甲型流感病毒(H1N1流感SC09株和H3N2流感FJ09株)、乙型流感病毒(SC11株)、寨卡病毒(SZ01株)、人冠状病毒(HCoV-229E株、HCoV-OC43株;以及属于新型冠状病毒(严重急性呼吸综合征冠状病毒2,2019-nCoV)的B.1.1.7株、B.1.351株和B.1.617.2株)。
实验1体外抗流感病毒实验
本实验例测定了本公开的示例性化合物对甲型流感病毒(H1N1流感SC09株和H3N2流感FJ09株)和乙型流感病毒(SC11株)的抑制活性。
1、细胞的复苏与培养
配制DMEM细胞完全培养液(含有10%胎牛血清、100U/ml青霉素、100U/ml链霉素)和MDCK细胞维持液(含有2%胎牛血清、100U/ml青霉素、100U/ml链霉素)备用。将冻存细胞的小管从液氮中取出,迅速置于37℃水浴中快速晃动,直至完全溶解(尽量在1min内)。取1ml细胞悬液置于离心管中,加入5ml细胞完全培养液后离心(2000rpm,5min,于室温)。将上清液弃去,加入2m1完全培养液,用移液枪吹匀后转移到细胞培养瓶(25cm 2)中,补足完全培养液至3ml,放置于37℃,5%CO 2的细胞培养箱中培养,24h后更换新的完全培养液。待细胞长成单层后,弃去原培养液,用PBS荡洗2-3次后,大力吹打细胞使其掉落。反复吹吸直至细胞充分离壁且分散均匀,吸取适量细胞悬浮液置于新的细胞培养瓶中,补足完全培养液。按一定比例(一般为1∶2或1∶3)传代后,放置于37℃,5%CO 2的细胞培养箱中培养。
2、流感病毒传代
将流感病毒SC09株冻存管从-80℃冰箱取出,在冰上化开备用。将含有长成单层的MDCK细胞的培养瓶取出,倾倒培养液,用PBS荡洗1-2次。将病毒液转移到培养瓶中,铺平底壁,加入适量细胞维持液, 放置于相应条件的细胞培养箱中培养。每日取出在显微镜下观察,直至细胞全部病变(历时36h)。吹打收集病毒液,离心(4000rpm,10min,4℃),取上清液过滤(滤膜孔径为0.2μm),以200μl/支分装于冻存管中备用。FJ09株和SC11株的传代操作同上。
3、流感病毒滴度测定
将MDCK细胞以3×10 5个细胞/ml的浓度接种到96孔板中,每孔接种100μl的细胞悬浮液,并置于37℃,5%CO 2的细胞培养箱中培养1-2天以长成单层。从-80℃冰箱中取出存有病毒液的小管,在流动自来水中融化,用细胞维持液稀释10倍,为第一个病毒滴度。将长成单层细胞的96孔板取出,倾倒上清液,除第一个滴度的三个副孔加入200μl的病毒液外,其余各孔加入180μl的细胞维持液,从前一个滴度吸取20μl到下一浓度中,吹吸混合均匀。最后3个副孔不加病毒液,作为细胞对照组。将96孔板置于细胞培养箱中培养,每天于显微镜下观察,直至不再出现新的细胞病变(CPE)孔为止。只要孔中出现病变就记为“+”,计数每个稀释度出现CPE的孔,以Karber法计算病毒的TCID 50值。
4、测试化合物对细胞的最大无毒浓度(TC 0)
(1)药物溶液的配制:取适量化合物(阳性对照品和供试品),分别用DMSO配制成25mg/ml的母液,过滤除菌(滤器孔径0.2μm)后在4℃保存。
(2)中性红染液的配制:利用细胞维持液将1%的中性红溶液稀释100倍。
(3)醋酸乙醇溶液的配制:分别量取5ml水、5ml无水乙醇和100μl冰醋酸,混合均匀即得。
(4)化合物TC 0值的测定方法:采用十倍稀释的方法设定6个浓度梯度,每个浓度设3个副孔。将长成单层细胞的96孔板取出,弃去孔中的培养液。除最大浓度的3个副孔外,每孔加入180μl细胞维持液。将化合物母液用细胞维持液稀释500倍,为最大实验浓度。对于最大 浓度的3个副孔,每孔加入200μl的50μg/ml的化合物溶液。从最大浓度的孔中吸取20μl液体至第二个浓度的孔中,混合均匀;再从第二个浓度的孔中吸取20μl液体至第三个浓度的孔中,以此类推,完成浓度的十倍递减稀释,并设细胞对照孔。将96孔板置于37℃,5%CO 2的细胞培养箱中培养72h。利用中性红染色法测定各孔细胞的OD值。将达到实验终点的96孔板取出,弃去孔中原有的液体,加入中性红染液(100μl/孔),并置于37℃,5%CO 2的细胞培养箱中孵育。1h后,取出96孔板,弃去中性红染液,并用PBS溶液将孔中的中性红荡洗2-3次,弃去PBS,加入醋酸乙醇溶液显色(100μl/孔)。利用酶标仪在波长为544nm处测定OD值。判定结果参考:实验组的OD值≥90%对照组的OD值,判定是最大无毒浓度TC 0值。
5、测试化合物的半数细胞毒性浓度(CC 50)
(1)药物溶液的配制:取适量化合物(阳性对照品和供试品),分别用DMSO配制成25mg/ml的母液,过滤除菌(滤器孔径0.2μm)后在4℃保存。
(2)中性红染液的配制:利用细胞维持液将1%的中性红溶液稀释100倍。
(3)醋酸乙醇溶液的配制:分别量取5ml水、5ml无水乙醇和100μl冰醋酸,混合均匀即得。
(4)化合物的CC 50值的测定方法:采用对倍稀释的方法设定5个浓度梯度,每个浓度设3个副孔。将长成单层细胞的96孔板取出,弃去孔中的培养液。除最大浓度的3个副孔外,每孔加入100μl细胞维持液。将化合物母液用细胞维持液稀释500倍,为最大实验浓度。对于最大浓度的3个副孔,每孔加入200μl的50μg/ml的化合物溶液。从最大浓度的孔中吸取100μl液体至第二个浓度的孔中,混合均匀;再从第二个浓度的孔中吸取100μl液体至第三个浓度的孔中,以此类推,完成浓度的对倍递减稀释,并设细胞对照孔。将96孔板置于37℃,5%CO 2的细胞培养箱中培养,每天于显微镜下观察,直至不再出 现新的细胞病变时为实验终点。利用中性红染色法测定各孔细胞的OD值。将达到实验终点的96孔板取出,弃去孔中原有的液体,加入中性红染液(100μl/孔),并置于37℃,5%CO 2的细胞培养箱中孵育。1h后,取出96孔板,弃去中性红染液,并用PBS溶液将孔中的中性红荡洗2-3次,弃去PBS,加入醋酸乙醇溶液显色(100μl/孔)。利用酶标仪在波长为544nm处测定OD值。测得OD值后计算细胞存活率,公式如下:
细胞存活率(%)=(受试孔OD 544值/细胞对照孔的平均OD 544值)×100%
用GraphPad Prism 5.0软件计算化合物的半数细胞毒性浓度(CC 50值)。
6、测试化合物的半数抑制浓度(IC 50)
准备长成单层细胞的96孔板。弃去板中原有的完全培养液,用稀释成10-100TCID 50毒力的病毒液感染病毒对照孔、阳性药物对照孔和实验孔中的细胞(100μl/孔)。然后,向病毒对照孔中加入细胞维持液(100μl/孔)。向样品实验组中加入以选用的TC 0浓度为终浓度的试验化合物样品溶液对倍稀释,向下稀释四个浓度梯度,即实验组5个浓度梯度进行测试。向细胞对照孔中加入细胞维持液(200μl/孔),并设有5个空白孔作为空白对照组。病毒对照孔和正常细胞对照孔各设5个副孔,实验组每个浓度设3个副孔。将96孔板置于37℃,5%CO 2培养箱中培养。每日在倒置显微镜下观察并记录细胞病变情况。直到病毒对照孔出现4个“+”细胞病变时为实验终点。用中性红染色和醋酸乙醇溶液显色后,在波长544nm处测定各孔细胞的OD值,并根据下式计算抑制率:
抑制率(%)=(受试孔OD 544值-病毒对照孔平均OD 544值/(细胞对照孔平均OD 544值-病毒对照孔平均OD 544值)×100%
用GraphPad Prism 5.0软件计算化合物的半数抑制浓度(IC 50值)。
7、实验结果汇总
以上实验的结果汇总在下面的表6中:
表6体外抗流感病毒实验结果汇总
Figure PCTCN2022141023-appb-000100
由上表中的结果可知,在本实验的条件下,所测试的化合物对流感病毒SC09株、FJ09株和SC11株感染所致的细胞病变均有抑制作用,特别是当与阳性对照药EIDD-1931比较时,除化合物NTV-118与EIDD-1931的活性相当以外,其它测试的化合物均显示出对病毒感染所致的细胞病变具有更强的抑制作用。
实验2体外抗寨卡病毒实验
本实验例测定了本公开的示例性化合物对寨卡病毒(SZ01株)的抑制活性。
1、细胞的复苏与培养
本实验采用C6/36细胞培养寨卡病毒,并采用Vero细胞进行药效试验。
(1)C6/36细胞的复苏与培养
配制RPMI-1640细胞完全培养液(含有10%胎牛血清、100U/ml青霉素、100U/ml链霉素)和RPMI-1640细胞维持液(含有2%胎牛血清、100U/ml青霉素、100U/ml链霉素)备用。将冻存细胞的小管从液氮中取出,迅速置于37℃水浴中快速晃动,直至完全溶解(尽量在1min内)。取1ml细胞悬液于离心管中,加入5ml细胞完全培养液后离心(2000rpm,5min,于室温)。将上清液弃去,加入2m1完全培养液,用移液枪吹匀后转移到细胞培养瓶(25cm 2)中,补足完全培养液至3ml, 放置于28℃,5%CO 2的细胞培养箱中培养,24h后更换新的完全培养液。待细胞长成单层后,弃去原培养液,用PBS荡洗2-3次后,大力吹打细胞使其掉落。反复吹吸至细胞充分离壁且分散均匀,吸取适量细胞悬浮液于新的细胞培养瓶中,补足完全培养液,按一定比例(一般1∶2或1∶3)传代,放置于28℃,5%CO 2的细胞培养箱中培养。以上操作需在无菌条件下进行。
(2)Vero细胞的复苏与培养
配制DMEM细胞完全培养液(含有10%胎牛血清、100U/ml青霉素、100U/ml链霉素)和DMEM细胞维持液(含有2%胎牛血清、100U/ml青霉素、100U/ml链霉素)备用。具体操作方法同C6/36细胞的复苏与培养,不同点仅在于细胞培养条件的设定,Vero细胞在37℃,5%CO 2的细胞培养箱中培养。
2、寨卡病毒传代
将寨卡病毒SZ01株冻存管从-80℃冰箱取出,在冰上化开备用。将含有长成单层的Vero细胞的培养瓶取出,倾倒培养液,用PBS荡洗1-2次。将病毒液转移到培养瓶中,铺平底壁,加入适量细胞维持液,放置于相应条件的细胞培养箱中培养。每日取出在显微镜下观察,直至细胞全部病变(历时7天)。将病毒液反复冻融3次(-80℃保持30min;37℃融化;-80℃保持30min),然后以液体状态离心(4000rpm,10min,4℃),取上清液过滤(滤膜孔径为0.2μm),以200μl/支分装于冻存管中备用。
3、寨卡病毒滴度测定
将Vero细胞以3×10 5个细胞/ml的浓度接种到96孔板中,每孔接种100μl的细胞悬浮液,并置于37℃,5%CO 2的细胞培养箱中培养1-2天以长成单层。从-80℃冰箱中取出存有病毒液的小管,融化,用细胞维持液稀释10倍,为第一个病毒滴度。将长成单层细胞的96孔板取出,倾倒上清液,除第一个滴度的三个副孔加入200μl的病毒液外,其余各孔加入180μl的细胞维持液,从前一个滴度吸取20μl到 下一浓度中,吹吸混合均匀。最后3个副孔不加病毒液,作为细胞对照组。将96孔板置于细胞培养箱中培养,每天于显微镜下观察,直至不再出现新的细胞病变(CPE)孔为止。只要孔中出现病变就记为“+”,计数每个稀释度出现CPE的孔,以Karber法计算病毒的TCID 50值。
根据观察到的CPE结果,利用如下公式算病毒TCID 50值:
LogTCID 50=L-d(s-0.5)
其中,L是最低滴度的log值,d是滴度稀释的10的d次方,s是不同滴度细胞病变孔比例之和。
细胞病变的判定方法见下表7:
表7细胞病变判定方法(CPE法)
细胞病变比例 记录方法
<25% +
25%-50% ++
50%-75% +++
>75% ++++
利用CPE法,观察记录细胞的病变情况并记录实验结果,汇总于下表8中:
表8寨卡病毒SZ01株的TCID 50
Figure PCTCN2022141023-appb-000101
由表8中的数据计算寨卡病毒的TCID 50值:
LogTCID 50=-1-1(5.33-0.5)=-5.83,
TCID 50=10 -5.83(按180μl病毒液计算)。
4、测试化合物对细胞的最大无毒浓度(TC 0)
(1)药物溶液的配制:取适量化合物(阳性对照品和供试品),分别用DMSO配制成25mg/ml的母液,过滤除菌(滤器孔径0.2μm)后在4℃保存。
(2)中性红染液的配制:利用细胞维持液将1%的中性红溶液稀释100倍。
(3)醋酸乙醇溶液的配制:分别量取5ml水、5ml无水乙醇和100μl冰醋酸,混合均匀即得。
(4)化合物TC 0值的测定方法:采用十倍稀释的方法设定6个浓度梯度,每个浓度设3个副孔。将长成单层细胞的96孔板取出,弃去孔中的培养液。除最大浓度的3个副孔外,每孔加入180μl细胞维持液。将化合物母液用细胞维持液稀释500倍,为最大实验浓度。对于最大浓度的3个副孔,每孔加入200μl的50μg/ml的化合物溶液。从最大浓度的孔中吸取20μl液体至第二个浓度的孔中,混合均匀;再从第二个浓度的孔中吸取20μl液体至第三个浓度的孔中,以此类推,完成浓度的十倍递减稀释,并设细胞对照孔。将96孔板置于37℃,5%CO 2的细胞培养箱中培养72h。利用中性红染色法测定各孔细胞的OD值。将达到实验终点的96孔板取出,弃去孔中原有的液体,加入中性红染液(100μl/孔),并置于37℃,5%CO 2的细胞培养箱中孵育。1h后,取出96孔板,弃去中性红染液,并用PBS溶液将孔中的中性红荡洗2-3次,弃去PBS,加入醋酸乙醇溶液显色(100μl/孔)。利用酶标仪在波长为544nm处测定OD值。判定结果参考:实验组的OD值≥90%对照组的OD值,判定是最大无毒浓度TC 0值。另外,有的化合物在100μg/ml~1ng/ml的范围内会发现细胞状态受到抑制或细胞病变现象,需要再次确定最大无毒浓度范围。
5、测试化合物的半数细胞毒性浓度(CC 50)
(1)药物溶液的配制:取适量化合物(阳性对照品和供试品),分别用DMSO配制成25mg/ml的母液,过滤除菌(滤器孔径0.2μm)后在4℃ 保存。
(2)中性红染液的配制:利用细胞维持液将1%的中性红溶液稀释100倍。
(3)醋酸乙醇溶液的配制:分别量取5ml水、5ml无水乙醇和100μl冰醋酸,混合均匀即得。
(4)化合物的CC 50值的测定方法:采用对倍稀释的方法设定5个浓度梯度,每个浓度设3个副孔。将长成单层细胞的96孔板取出,弃去孔中的培养液。除最大浓度的3个副孔外,每孔加入100μl细胞维持液。将母液用细胞维持液稀释500倍,为最大实验浓度。对于最大浓度的3个副孔,每孔加入200μl的50μg/ml的化合物溶液。从最大浓度的孔中吸取100μl液体至第二个浓度的孔中,混合均匀;再从第二个浓度的孔中吸取100μl液体至第三个浓度的孔中,以此类推,完成浓度的对倍递减稀释,并设细胞对照孔。将96孔板置于37℃,5%CO 2的细胞培养箱中培养,每天于显微镜下观察,直至不再出现新的细胞病变时为实验终点。利用中性红染色法测定各孔细胞的OD值。将达到实验终点的96孔板取出,弃去孔中原有的液体,加入中性红染液(100μl/孔),并置于37℃,5%CO 2的细胞培养箱中孵育。1h后,取出96孔板,弃去中性红染液,并用PBS溶液将孔中的中性红荡洗2-3次,弃去PBS,加入醋酸乙醇溶液显色(100μl/孔)。利用酶标仪在波长为544nm处测定OD值。测得OD值后计算细胞存活率,公式如下:
细胞存活率(%)=(受试孔OD 544值/细胞对照孔的平均OD 544值)×100%
用GraphPadPrism5.0软件计算药物的半数细胞毒性浓度(CC 50值)。
(5)化合物CC 50值的测定结果:
以上实验的结果汇总在下面的表9中:
表9测试化合物的CC 50
化合物 CC 50(μM)±SD
利巴韦林 1358±13.92
EIDD-1931 11.23±0.22
NTV-16 43.23±2.23
NTV-29 13.20±0.94
NTV-37 10.51±1.12
NTV-118 18.40±2.04
上表中记载的所测试的化合物的细胞毒性结果表明,化合物NTV-16、NTV-29、NTV-37和NTV-118与阳应对照药物EIDD-1931相比,所测试的化合物细胞毒性与小于或相当于EIDD-1931。
6、测试化合物的半数抑制浓度(IC 50)
寨卡病毒的TCID 50=10 -5.83。将病毒原液用细胞维持液稀释成10-100TCID 50。用原液的10 -4浓度进行感染反应。
准备长成单层细胞的96孔板。弃去板中原有的完全培养液,用稀释的病毒液感染病毒对照孔、阳性药物对照孔和实验孔中的细胞(100μl/孔)。然后,向病毒对照孔中加入细胞维持液(100μl/孔)。向样品实验组中加入以选用的TC 0浓度为终浓度的试验化合物样品溶液对倍稀释,向下稀释四个浓度梯度(100μl/孔),即实验组5个浓度梯度进行测试。向细胞对照孔中加入细胞维持液(200μl/孔),并设有5个空白孔作为空白对照组。病毒对照孔和正常细胞对照孔各设5个副孔,实验组每个浓度设5个副孔。将96孔板置于37℃,5%CO 2培养箱中培养。每日在倒置显微镜下观察并记录细胞病变情况。直到病毒对照孔出现4个“+”细胞病变时为实验终点。用中性红染色和醋酸乙醇溶液显色后,在波长544nm处测定各孔细胞的OD值,并根据下式计算抑制率:
抑制率(%)=(受试孔OD 544值-病毒对照孔平均OD 544值)/(细胞对照孔平均OD 544值-病毒对照孔平均OD 544值)×100%
用GraphPadPrism5.0软件计算化合物的半数抑制浓度(IC 50值)。
7、实验结果汇总
以上实验的结果汇总在下面的表10中:
表10体外抗寨卡病毒(SZ01)株实验结果汇总
化合物 IC 50(μM)±SD CC 50(μM)±SD SI
利巴韦林 >400μM 1358±13.92 -
EIDD-1931 1.36±0.42 11.23±0.22 8.25
NTV-29 0.65±0.12 13.20±0.94 20.3
NTV-37 1.05±0.38 10.51±1.12 10
NTV-118 2.25±0.62 18.40±2.04 8.2
由上表中的结果可知,在本实验的条件下,所测试的化合物的体外抗寨卡病毒活性优于阳应对照药EIDD-1931或与其相当,而利巴韦林未显示出抗寨卡病毒的效果。
实验3体外抗冠状病毒活性测定
本实验例测定了本公开的示例性化合物对人冠状病毒(HCoV-229E株和HCoV-OC43株)的抑制活性。
1、实验准备
(1)细胞:人肝癌细胞Huh7细胞和人肺癌细胞H460细胞均为本实验传代保存,并在含有10%胎牛血清和1%双抗(青霉素和链霉素)的DMEM培养基中于37℃,5%CO 2培养箱中培养。每隔2-3天传代一次。
(2)毒株:HCoV-229E株于Huh7细胞中传代,保存于-80℃冰箱。HCoV-OC43株于H460细胞中传代,亦保存于-80℃冰箱。
(3)试剂:DMEM液体培养基、胎牛血清、青霉素和链霉素溶液、PBS(PH=7.4)和0.25%胰蛋白酶-EDTA均可商购获得。
(4)细胞培养:以H460细胞为例,在长满H460细胞的培养瓶内加入3m10.25%胰蛋白酶-EDTA,于7℃消化1-2分钟。弃去消化液,加入培养液吹打,以1∶3传代,每2-3天传代一次。种板时,配制成15万个细胞/ml培养液,接种于96孔细胞培养板,每孔0.1ml,于37℃,5%CO 2细胞培养箱中培养过夜。待细胞长成单层后进行实验。Huh7细胞也按照上述方法进行培养。
2、测试化合物的半数细胞毒性浓度(CC 50)
(1)药物溶液的配制:取适量化合物(阳性对照品和供试品),分别用 DMSO配制成25mg/ml的母液,过滤除菌(滤器孔径0.2μm)后在4℃保存。
(2)中性红染液的配制:利用细胞维持液将1%的中性红溶液稀释100倍。
(3)醋酸乙醇溶液的配制:分别量取5ml水、5ml无水乙醇和100μl冰醋酸,混合均匀即得。
(4)化合物的CC 50值的测定方法:采用对倍稀释的方法设定5个浓度梯度,每个浓度设3个副孔。将长成单层细胞的96孔板取出,弃去孔中的培养液。除最大浓度的3个副孔外,每孔加入100μl细胞维持液。将化合物母液用细胞维持液稀释500倍,为最大实验浓度。对于最大浓度的3个副孔,每孔加入200μl的50μg/ml的化合物溶液。从最大浓度的孔中吸取100μl液体至第二个浓度的孔中,混合均匀;再从第二个浓度的孔中吸取100μl液体至第三个浓度的孔中,以此类推,完成浓度的对倍递减稀释,并设细胞对照孔。将96孔板置于37℃,5%CO 2的细胞培养箱中培养,每天于显微镜下观察,直至不再出现新的细胞病变时为实验终点。利用中性红染色法测定各孔细胞的OD值。将达到实验终点的96孔板取出,弃去孔中原有的液体,加入中性红染液(100μl/孔),并置于37℃,5%CO 2的细胞培养箱中孵育。1h后,取出96孔板,弃去中性红染液,并用PBS溶液将孔中的中性红荡洗2-3次,弃去PBS,加入醋酸乙醇溶液显色(100μl/孔)。利用酶标仪在波长为544nm处测定OD值。测得OD值后计算细胞存活率,公式如下:
细胞存活率(%)=(受试孔OD 544值/细胞对照孔的平均OD 544值)×100%
用GraphPad Prism 5.0软件计算药物的半数细胞毒性浓度(CC 50值)。
3、测试化合物的半数抑制浓度(IC 50)
实验分别在Huh7细胞和H460细胞中进行。将Huh7细胞和H460 细胞分别以1.5x10 4个细胞/孔的浓度接种于96孔板中。过夜培养后,分别用100TCID 50的HCoV-229E株和HCoV-OC43株的病毒液感染96孔板内的Huh7细胞和H460细胞。用维持液稀释测试化合物,并于感染同时给药进行测定:将测试化合物以三倍稀释的8个剂量的样品进行实验,每个剂量设2个平行孔,待病毒对照组病变达4+(根据细胞的死亡比例,分别标记为4+(细胞死亡比例75%-100%)、3+(细胞死亡比例50%-75%)、2+(细胞死亡比例25%-50%)、1+(细胞死亡比例0-25%)、0+(细胞全部存活))时观察结果,记录数据并用Reed-Muench法根据下式计算测试化合物对病毒的半数抑制浓度,并计算选择指数SI值(SI=CC 50/IC 50):
Figure PCTCN2022141023-appb-000102
其中:A=累积抑制率<50%的药物浓度,B累积抑制率>50%的抑制率,C=累积抑制率<50%的抑制率,D=log稀释倍数。
4、实验结果汇总
以上实验的结果汇总在下面的表11中:
表11体外抗冠状病毒实验结果汇总
Figure PCTCN2022141023-appb-000103
由上表中的结果可知,在本实验的条件下,所测试的化合物的体外抗冠状病毒活性优于阳应对照药EIDD-1931或与其相当。
实验4体外抗新型冠状病毒(2019-nCoV-2)活性测定
本实验例测定了本公开的示例性化合物对新型冠状病毒(2019-nCoV-2)B.1.1.7株、B.1.351株和B.1.617.2株的抑制活性。
1、病毒培养与病毒滴度测定
将非洲绿猴肾细胞(VeroE6)用含有10%胎牛血清(FBS)、谷氨酰胺与青霉素-链霉素的DMEM培养液于37℃,5%CO 2培养箱中培养。待 细胞生长至成完全汇合的单层后,吸去培养液,用PBS洗涤2~3次,然后加入病毒液,于37℃孵育1小时。吸去培养液后,加入含有2%FBS的DMEM培养液培养,直至出现细胞病变达>75%的细胞单层破坏。收获病毒培养物,于-80℃保存备用。
在96孔细胞培养板中接种VeroE6细胞(密度为2~3×10 4个细胞/孔),培养过夜。至细胞长满单层后,用PBS洗涤2~3次,于各孔中加入100μl不同稀释度(10 -1~10 -10)的病毒液,每个稀释度设6~8个复孔,并置于37℃,5%CO 2培养箱中培养。每天观察并记录细胞病变情况,直至不再出现新的细胞病变孔。用Reed-Muench或Karber法计算病毒液的TCID 50
Figure PCTCN2022141023-appb-000104
2、测试化合物的半数细胞毒性浓度(CC 50)
如前所述,在96孔板中接种Vero E6细胞。培养至长满单层后,用PBS洗涤,然后加入200μl不同浓度的化合物的2%FBS DMEM溶液。培养5~6天后,加入3μl 1.5%中性红溶液,继续孵育24小时,然后用10%甲醛生理盐水溶液固定。用PBS洗涤2~3次以洗去残留的中性红,加入200μl酸化乙醇(由50%乙醇、49%水和1%冰醋酸组成),振荡10分钟或直至中性红完全溶出,用酶标仪测定OD 540值。用4参数逻辑斯谛回归拟合量-效曲线,计算化合物的半数细胞毒性浓度(CC 50)。
采用下述公式计算药物的CC 50
Figure PCTCN2022141023-appb-000105
最后,利用Graphpad Prism 7软件对数据进行S拟合分析,计算化合物的半数细胞毒性浓度(CC 50值)。
3、测试化合物的半数抑制浓度(IC 50)
如前所述,在96孔板中接种Vero E6细胞,培养过夜。待细胞长 满单层后,于其中6行加入100μl对倍稀释的化合物的2%FBS DMEM溶液,每个浓度设4个复孔。孵育1小时后,在该6行及另1行加入100μl病毒液(~100×TCID 50),余下1行加入200μl 2%FBS DMEM培养液。培养5~6天后,用中性红染色,测定OD 540值,计算化合物的半数抑制浓度(IC 50),并计算选择指数SI值(SI=CC 50/IC 50)。
抑制率(IC):(%)=(受试孔OD 540值-病毒对照孔平均OD 540值)/(细胞对照孔平均OD 540值-病毒对照孔平均OD 540值)×100%
用GraphPad Prism 5.0软件计算药物半数抑制浓度(IC 50值)。
4、实验结果汇总
以上实验的结果汇总在下面的表12中:
表12体外抗新型冠状病毒实验结果汇总
Figure PCTCN2022141023-appb-000106
由上表中的结果可知,在本实验的条件下,所测试的化合物的体外抗新型冠状病毒活性优于阳应对照药EIDD-1931。

Claims (40)

  1. 式I的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体:
    Figure PCTCN2022141023-appb-100001
    其中,
    m是6-20中的任一整数;
    n是1-5中的任一整数;
    X是-O-或-NR 0-;
    R 0是H或C 1-C 6烷基;
    Z是-O-或-S-S-;
    R是H、C 1-C 6烷基、C 6-C 12芳基、R′-O-C(O)-O-CH 2-或R′-C(O)-O-CH 2-;
    R′是C 1-C 6烷基、C 3-C 8环烷基、C 6-C 12芳基、含有1或2个独立地选自N、O或S的杂原子的3至12元杂环基或含有1或2个独立地选自N、O或S的杂原子的5至12元杂芳基;
    所述R或R′基团中的烷基、环烷基、芳基、杂环基或杂芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
  2. 根据权利要求1所述的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体,其中所述式I的化合物具有式I-1的结构:
    Figure PCTCN2022141023-appb-100002
  3. 根据权利要求1所述的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体,其中所述式I的化合物具有式I-2的结构:
    Figure PCTCN2022141023-appb-100003
  4. 根据权利要求1-3中的任一项所述的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体,其中X是-O-。
  5. 根据权利要求1-3中的任一项所述的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体,其中X是-NR 0-,并且其中R 0是H、甲基、乙基、正丙基、异丙基、正丁基、仲丁基、异丁基或叔丁基。
  6. 根据权利要求1-3中的任一项所述的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体,其中R是H。
  7. 根据权利要求1-3中的任一项所述的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体,其中R是C 1-C 6烷基,其中所述烷基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
  8. 根据权利要求1-3中的任一项所述的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体,其中R是C 6-C 12芳基,其中所述芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
  9. 根据权利要求1-3中的任一项所述的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体,其中R是R′-O-C(O)-O-CH 2-或R′-C(O)-O-CH 2-。
  10. 根据权利要求9所述的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体,其中R′是C 1-C 6烷基,其中所述烷基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
  11. 根据权利要求9所述的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体,其中R′是C 3-C 8环烷基,其中所述环烷基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
  12. 根据权利要求9所述的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体,其中R′是C 6-C 12芳基,其中所述芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝 基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
  13. 根据权利要求9所述的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体,其中R′是含有1或2个独立地选自N、O或S的杂原子的3至12元杂环基,其中所述杂环基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
  14. 根据权利要求9所述的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体,其中R′是含有1或2个独立地选自N、O或S的杂原子的5至12元杂芳基,其中所述杂芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
  15. 根据权利要求1-3中的任一项所述的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体,其中每个R a和R b独立地是H、甲基、乙基、正丙基、异丙基、正丁基、仲丁基、异丁基或叔丁基。
  16. 根据权利要求1-3中的任一项所述的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体,其中:
    m是6-20中的任一整数;
    n是1-5中的任一整数;
    X是-O-或-NR 0-;
    R 0是H、甲基、乙基、正丙基、异丙基、正丁基、仲丁基、异丁 基或叔丁基;
    R是H、甲基、乙基、正丙基、异丙基、正丁基、仲丁基、异丁基、叔丁基、任选地被1个独立地选自卤素或硝基的取代基取代的苯基、R′-O-C(O)-O-CH 2-或R′-C(O)-O-CH 2-;
    R′是甲基、乙基、正丙基、异丙基、正丁基、仲丁基、异丁基、叔丁基、环丁基、环戊基、环己基、环庚基或苯基。
  17. 根据权利要求1-3中的任一项所述的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体,其中:
    m是6-20中的任一整数;
    n是1-5中的任一整数;
    X是-O-或-NR 0-;
    R 0是H、甲基或乙基;
    R是H、甲基、乙基、任选地被1个独立地选自氯或硝基的取代基取代的苯基、R′-O-C(O)-O-CH 2-或R′-C(O)-O-CH 2-;
    R′是甲基、乙基、异丙基、叔丁基、环戊基、环己基或苯基。
  18. 根据权利要求1-3中的任一项所述的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体,其中:
    m是6-20中的任一整数;
    n是1-5中的任一整数;
    X是-O-或-NR 0-;
    R 0是H或甲基;
    R是H、甲基、任选地被1个独立地选自氯或硝基的取代基取代的苯基、R′-O-C(O)-O-CH 2-或R′-C(O)-O-CH 2-;
    R′是甲基、异丙基、叔丁基、环戊基、环己基或苯基。
  19. 根据权利要求1-3中的任一项所述的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体,其中所述化合物是下表中所述的化合物中的任一种:
    Figure PCTCN2022141023-appb-100004
    Figure PCTCN2022141023-appb-100005
    Figure PCTCN2022141023-appb-100006
    Figure PCTCN2022141023-appb-100007
    Figure PCTCN2022141023-appb-100008
    Figure PCTCN2022141023-appb-100009
    Figure PCTCN2022141023-appb-100010
    Figure PCTCN2022141023-appb-100011
    Figure PCTCN2022141023-appb-100012
    Figure PCTCN2022141023-appb-100013
    Figure PCTCN2022141023-appb-100014
    Figure PCTCN2022141023-appb-100015
    Figure PCTCN2022141023-appb-100016
    Figure PCTCN2022141023-appb-100017
    Figure PCTCN2022141023-appb-100018
    Figure PCTCN2022141023-appb-100019
    Figure PCTCN2022141023-appb-100020
    Figure PCTCN2022141023-appb-100021
    Figure PCTCN2022141023-appb-100022
    Figure PCTCN2022141023-appb-100023
    Figure PCTCN2022141023-appb-100024
  20. 根据权利要求1-3中的任一项所述的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体,其中所述化合物是下列化合物中的任一种:
    Figure PCTCN2022141023-appb-100025
    Figure PCTCN2022141023-appb-100026
    Figure PCTCN2022141023-appb-100027
  21. 根据权利要求1-20中的任一项所述的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体,其中:所述药学上可接受的盐是碱金属盐(例如锂、钠、钾、铷、铯盐)、碱土金属盐(例如镁、钙、锶、钡盐)、铝盐、有机碱盐(例如乙醇胺、二乙醇胺、三乙醇胺、氨丁三醇、N-甲基葡糖胺、二乙胺、二乙基氨基乙醇、乙二胺、咪唑、吗琳、2-羟基乙基吗琳、二苄基乙二胺、三甲胺、哌啶、吡咯烷、苄胺、四甲基氢氧化胺盐)或氨基酸盐(例如赖氨酸、精氨酸、精氨酸盐)。
  22. 药物组合物,其包含根据权利要求1-20中的任一项所述的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体以及一种或多种药学上可接受的载体。
  23. 根据权利要求1-20中的任一项所述的化合物或其药学上可接受的盐、水合物、溶剂合物或立体异构体或根据权利要求22所述的药物组合物在制备用于抑制病毒或者用于在有此需要的对象中预防和/或治疗病毒感染的药物中的用途。
  24. 根据权利要求23所述的用途,其中所述病毒是冠状病毒(例如人冠状病毒,诸如严重急性呼吸综合征冠状病毒(SARS-CoV)、中东呼吸综合征冠状病毒(MERS-CoV)或严重急性呼吸综合征冠状病毒2(SARS-CoV-2,包括例如B.1.1.7株、B.1.351株、德尔塔变异株B.1.617.2株、奥密克戎变异株B.1.1.529株或其它SARS-CoV-2变异株)、马脑炎病毒(例如东部马脑炎病毒、西部马脑炎病毒或委内瑞拉马脑炎病毒)、奇昆古尼亚病毒、罗斯河病毒、正粘病毒科病毒(例如流感病毒,诸如甲型流感病毒、乙型流感病毒或丙型流感病毒)、副粘病毒科病毒(例如呼吸道合胞病毒)、丝状病毒(例如埃博拉病毒、马尔堡病毒或库 瓦病毒)或寨卡病毒。
  25. 根据权利要求24所述的用途,其中所述病毒是甲型H1N1流感病毒。
  26. 根据权利要求24所述的用途,其中所述病毒是SC09株。
  27. 根据权利要求24所述的用途,其中所述病毒是甲型H3N2流感病毒。
  28. 根据权利要求24所述的用途,其中所述病毒是FJ09株。
  29. 根据权利要求24所述的用途,其中所述病毒是SC11株。
  30. 根据权利要求24所述的用途,其中所述病毒是SZ01株。
  31. 根据权利要求24所述的用途,其中所述病毒是HCoV-229E株或HCoV-OC43株。
  32. 根据权利要求24所述的用途,其中所述病毒是B.1.1.7株、B.1.351株或B.1.617.2株。
  33. 式2a的化合物:
    Figure PCTCN2022141023-appb-100028
    其中m、n和Z根据权利要求1-3中的任一项所定义;R 1是直链或直链的C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基或C 6-C 12芳基,所述烷基、烯基、炔基或芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基;A是氯或硝基;B是 氢、氯或硝基。
  34. 式2a-1的化合物:
    Figure PCTCN2022141023-appb-100029
    其中m、n、Z、R 0和R根据权利要求1-3中的任一项所定义;R 1是直链或直链的C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基或C 6-C 12芳基,所述烷基、烯基、炔基或芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
  35. 式4a的化合物:
    Figure PCTCN2022141023-appb-100030
    其中m、n和Z根据权利要求1-3中的任一项所定义;R 1是直链或直链的C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基或C 6-C 12芳基,所述烷基、烯基、炔基或芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6 烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基;A是氯或硝基;B是氢、氯或硝基。
  36. 式4a-1的化合物:
    Figure PCTCN2022141023-appb-100031
    其中m、n、Z、R 0和R根据权利要求1-3中的任一项所定义;R 1是直链或直链的C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基或C 6-C 12芳基,所述烷基、烯基、炔基或芳基任选地被1、2或3个独立地选自卤素、C 1-C 6烷基、卤代C 1-C 6烷基、C 2-C 6烯基、C 2-C 6炔基、C 1-C 6烷氧基、卤代C 1-C 6烷氧基、氰基、羟基、硝基、-NR aR b、-C(=O)R a、-C(=O)OR a、-OC(=O)R a、-C(=O)NR aR b、-SR a、-S(=O)R a、-S(=O) 2R a和-S(=O) 2NR aR b的取代基取代,其中每个R a和R b独立地是H或C 1-C 6烷基。
  37. 式2-1a的化合物:
    Figure PCTCN2022141023-appb-100032
    其中m、n和Z根据权利要求1-3中的任一项所定义;A是氯或硝 基;B是氢、氯或硝基。
  38. 式2-1a-1的化合物:
    Figure PCTCN2022141023-appb-100033
    其中m、n、Z、R 0和R根据权利要求1-3中的任一项所定义。
  39. 式4-1a的化合物:
    Figure PCTCN2022141023-appb-100034
    其中m和n根据权利要求1-3中的任一项所定义;A是氯或硝基;B是氢、氯或硝基。
  40. 式4-1a-1的化合物:
    Figure PCTCN2022141023-appb-100035
    其中m、n、Z、R 0和R根据权利要求1-3中的任一项所定义。
PCT/CN2022/141023 2022-01-28 2022-12-22 抗病毒化合物及其用途 WO2023142803A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210106177.4A CN116554249A (zh) 2022-01-28 2022-01-28 抗病毒化合物及其用途
CN202210106177.4 2022-01-28

Publications (1)

Publication Number Publication Date
WO2023142803A1 true WO2023142803A1 (zh) 2023-08-03

Family

ID=87470385

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/141023 WO2023142803A1 (zh) 2022-01-28 2022-12-22 抗病毒化合物及其用途

Country Status (2)

Country Link
CN (1) CN116554249A (zh)
WO (1) WO2023142803A1 (zh)

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015038596A1 (en) * 2013-09-11 2015-03-19 Emory University Nucleotide and nucleoside compositions and uses related thereto
CN105829333A (zh) * 2013-10-11 2016-08-03 艾丽奥斯生物制药有限公司 取代的核苷、核苷酸及其类似物
CN106573011A (zh) * 2014-06-24 2017-04-19 艾丽奥斯生物制药有限公司 取代的核苷、核苷酸和其类似物
CN106604734A (zh) * 2014-06-24 2017-04-26 艾丽奥斯生物制药有限公司 取代的核苷、核苷酸和其类似物
WO2017156380A1 (en) * 2016-03-10 2017-09-14 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
CN107250145A (zh) * 2014-12-19 2017-10-13 艾丽奥斯生物制药有限公司 被取代的核苷、核苷酸及其类似物
CN107427529A (zh) * 2014-12-26 2017-12-01 埃莫里大学 N4‑羟基胞苷和衍生物及与其相关的抗病毒用途
CN113735928A (zh) * 2021-10-21 2021-12-03 药康众拓(江苏)医药科技有限公司 一种n4-羟基胞苷衍生物及其制备方法和用途
WO2022020793A1 (en) * 2020-07-24 2022-01-27 The Regents Of The University Of California Antiviral prodrugs, pharmaceutical formulations, and methods
WO2022235874A1 (en) * 2021-05-05 2022-11-10 Emory University Nucleotide and nucleoside therapeutic compositions, combinations and uses related thereto
WO2022251663A2 (en) * 2021-05-27 2022-12-01 Emory University Novel universal anti-rna virus agents

Patent Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2015038596A1 (en) * 2013-09-11 2015-03-19 Emory University Nucleotide and nucleoside compositions and uses related thereto
CN105829333A (zh) * 2013-10-11 2016-08-03 艾丽奥斯生物制药有限公司 取代的核苷、核苷酸及其类似物
CN106573011A (zh) * 2014-06-24 2017-04-19 艾丽奥斯生物制药有限公司 取代的核苷、核苷酸和其类似物
CN106604734A (zh) * 2014-06-24 2017-04-26 艾丽奥斯生物制药有限公司 取代的核苷、核苷酸和其类似物
CN107250145A (zh) * 2014-12-19 2017-10-13 艾丽奥斯生物制药有限公司 被取代的核苷、核苷酸及其类似物
CN107427529A (zh) * 2014-12-26 2017-12-01 埃莫里大学 N4‑羟基胞苷和衍生物及与其相关的抗病毒用途
WO2017156380A1 (en) * 2016-03-10 2017-09-14 Emory University N4-hydroxycytidine and derivatives and anti-viral uses related thereto
WO2022020793A1 (en) * 2020-07-24 2022-01-27 The Regents Of The University Of California Antiviral prodrugs, pharmaceutical formulations, and methods
WO2022235874A1 (en) * 2021-05-05 2022-11-10 Emory University Nucleotide and nucleoside therapeutic compositions, combinations and uses related thereto
WO2022251663A2 (en) * 2021-05-27 2022-12-01 Emory University Novel universal anti-rna virus agents
CN113735928A (zh) * 2021-10-21 2021-12-03 药康众拓(江苏)医药科技有限公司 一种n4-羟基胞苷衍生物及其制备方法和用途

Also Published As

Publication number Publication date
CN116554249A (zh) 2023-08-08

Similar Documents

Publication Publication Date Title
CA2997051C (en) Novel pyrazolo[3,4-d]pyrimidine compound or salt thereof
ES2402791T3 (es) Forma cristalina de la sal diclorhidrato de ((1S)-1-(((2S)-2-(5-(4'-(2-((2S)-1-((2S)-2-((metoxicarbonil)amino)-3-metilbutanoil)-2-pirrolidinil)-1H-imidazol-5-il)-4-bifenilil)-1H-imidazol-2-il)-1-pirrolidinil)carbonil)-2-metilpropil)carbamato de metilo
US10562864B2 (en) Chemical modulators of immune checkpoints and therapeutic use
JP2000501694A (ja) 複素環置換シクロペンタン化合物
WO2020011246A1 (zh) 含苯环的化合物、其制备方法及应用
KR20210024574A (ko) 세포 괴사 억제제 및 이의 제조 방법과 용도
CN113527293B (zh) Kras g12c突变蛋白抑制剂及其药物组合物、制备方法和用途
CN109311891A (zh) 作为jak抑制剂的吡咯并嘧啶化合物的结晶
BR112020023861A2 (pt) Derivado de p-fenilenodiamina como regulador de canal de potássio e método de preparação e aplicação médica do mesmo
CN110461836B (zh) 一种选择性抑制激酶化合物及其用途
JP5972911B2 (ja) モリブドプテリン前駆体z誘導体を合成するための方法
CN101454309B (zh) 焦谷氨酸衍生物的合成和用途
EP3560918A1 (en) Condensed ring group azacyclobutyl triazole derivative, preparation method therefor and use thereof in medicine
WO2020238776A1 (zh) 取代的稠合双环类衍生物、其制备方法及其在医药上的应用
CA3134405A1 (en) Thienoheterocyclic derivative, preparation method therefor and medical use thereof
CN105732659A (zh) 硝基咪唑类化合物及其制备方法和在制药中的用途
WO2020151707A1 (zh) 一种新型的小分子cd73抑制剂、其制备方法及其在医药上的应用
WO2023142803A1 (zh) 抗病毒化合物及其用途
CN116375707A (zh) Menin抑制剂及其用途
WO2019228330A1 (zh) 取代的苯并[d]咪唑类化合物及其药物组合物
TW202342017A (zh) 用於治療與lpa受體活性相關的病狀的化合物及組合物
KR101872264B1 (ko) 신규한 유형의 시티딘 유도체 이량체 및 그의 용도
TWI794576B (zh) 一類含氟取代的苯并噻吩類化合物及其藥物組合物及應用
RU2768451C1 (ru) Селективный антагонист рецепторов типа A2A
WO2022116968A1 (zh) 一种新型n-杂环bet溴结构域抑制剂、其制备方法及医药用途

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 22923589

Country of ref document: EP

Kind code of ref document: A1