WO2025195347A1 - Polycyclic pyridone derivative and use thereof - Google Patents
Polycyclic pyridone derivative and use thereofInfo
- Publication number
- WO2025195347A1 WO2025195347A1 PCT/CN2025/083049 CN2025083049W WO2025195347A1 WO 2025195347 A1 WO2025195347 A1 WO 2025195347A1 CN 2025083049 W CN2025083049 W CN 2025083049W WO 2025195347 A1 WO2025195347 A1 WO 2025195347A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- compound
- pharmaceutically acceptable
- added
- hydrogen
- tautomer
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Pending
Links
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/53—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/535—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
- A61K31/5375—1,4-Oxazines, e.g. morpholine
- A61K31/5383—1,4-Oxazines, e.g. morpholine ortho- or peri-condensed with heterocyclic ring systems
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P31/00—Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
- A61P31/12—Antivirals
- A61P31/14—Antivirals for RNA viruses
- A61P31/16—Antivirals for RNA viruses for influenza or rhinoviruses
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D471/00—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
- C07D471/12—Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains three hetero rings
- C07D471/14—Ortho-condensed systems
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07D—HETEROCYCLIC COMPOUNDS
- C07D498/00—Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
- C07D498/12—Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains three hetero rings
- C07D498/14—Ortho-condensed systems
Definitions
- the present invention relates to the field of medical technology, and in particular to a class of polycyclic pyridone derivatives, compositions and uses thereof.
- Influenza is a highly contagious respiratory disease caused by the influenza virus.
- influenza A virus has the widest host range, infecting both birds and mammals, making it highly likely to cause a worldwide pandemic.
- vaccination is generally effective in preventing influenza infection, but the vaccine is only partially effective, with an overall efficacy of approximately 60%.
- the virus may mutate during this period, thereby reducing the effectiveness of the vaccine.
- influenza neuraminidase protein inhibitors oseltamivir, zanamivir, peramivir, and nanamivir
- M2 proton channel blockers the amantadine class of drugs (amantadine and rimantadine).
- amantadine and rimantadine the amantadine class of drugs
- Cap-dependent endonucleases enzymes derived from influenza viruses, are considered suitable targets for anti-influenza drugs because they are essential for viral proliferation and possess virus-specific enzymatic activity not possessed by the host.
- the influenza virus cap-dependent endonuclease is an endonuclease activity that generates a fragment containing 9 to 13 bases of the cap structure (the bases of the cap structure are not included in the above base count) with the host mRNA precursor as a substrate. This fragment functions as a primer for the viral RNA polymerase, used for the synthesis of mRNA encoding viral proteins.
- cap-dependent endonucleases are believed to inhibit viral protein synthesis by inhibiting viral mRNA synthesis, thereby inhibiting viral proliferation.
- Mabaloxavir a drug already marketed in multiple countries, is an innovative cap-dependent endonuclease inhibitor. With a single oral dose, it can halt viral shedding within about 24 hours, shorten the infectious period, and quickly relieve flu symptoms such as fever and body aches. Based on the fact that cap-dependent nuclease drugs have been proven to have strong anti-influenza virus effects, it is still necessary to develop new drugs with richer types and more outstanding effects.
- the present invention provides a novel polycyclic pyridone derivative, a pharmaceutical composition thereof and use thereof in resisting influenza virus.
- the present invention provides a compound as shown in Formula I, its tautomers, stereoisomers or pharmaceutically acceptable salts thereof:
- R 1 , R 2 , R 3 and R 4 are all hydrogen or all deuterium, R 5 is absent or is an oxo group, and R 6 is hydrogen or
- R 1 , R 2 , R 3 and R 4 are deuterium, R 5 is absent or is oxo, and R 6 is hydrogen or
- R 1 , R 2 , R 3 , and R 4 are hydrogen, R 5 is oxo, and R 6 is hydrogen or
- R 1 , R 2 , R 3 , and R 4 are deuterium, R 5 is an oxo group, the configuration of the sulfur atom is S or R, and R 6 is hydrogen or
- R 1 , R 2 , R 3 , and R 4 are hydrogen
- R 5 is an oxo group
- the configuration of the sulfur atom is S or R
- R 6 is hydrogen or
- the compound of the present invention is selected from:
- the present invention provides a pharmaceutical composition
- a pharmaceutical composition comprising a compound of Formula I, its tautomers, stereoisomers or pharmaceutically acceptable salts thereof, and optionally, a pharmaceutically acceptable carrier.
- the present invention provides the use of a compound represented by Formula I, its tautomers, stereoisomers or pharmaceutically acceptable salts thereof, or the pharmaceutical composition described in the second aspect of the present invention in the preparation of a drug for treating and/or preventing diseases or conditions mediated by cap-dependent nucleases.
- the present invention also provides the use of the compound represented by Formula I, its tautomers, stereoisomers or pharmaceutically acceptable salts thereof, or the pharmaceutical composition described in the second aspect of the present invention in the preparation of drugs for treating and/or preventing diseases or conditions caused by viral infections.
- influenza virus is selected from type A, type B, type C, or avian influenza (H5N1, H7N9); further, the disease of the present invention is selected from cold-like symptoms accompanied by fever, chills, headache, muscle pain, general fatigue, etc., or respiratory inflammation with sore throat, runny nose, nasal congestion, cough, and sputum; gastrointestinal symptoms of abdominal pain, vomiting, and diarrhea, and then accompanied by complications of acute encephalopathy, secondary pneumonia infection, or a combination thereof.
- the present invention also provides the use of the compound represented by Formula I, its tautomers, stereoisomers or pharmaceutically acceptable salts thereof, or the pharmaceutical composition described in the second aspect of the present invention as a drug or in the preparation of a drug.
- the drug is a drug for treating and/or preventing a disease or condition mediated by a cap-dependent endonuclease.
- the disease or condition is an influenza infectious disease.
- influenza infectious disease is caused by an influenza virus selected from influenza A, B, C or avian influenza (H5N1, H7N9).
- influenza infectious disease is accompanied by one or more of the following symptoms: cold-like symptoms such as fever, chills, headache, muscle pain, general fatigue, or respiratory inflammation such as sore throat, runny nose, nasal congestion, cough, and sputum; gastrointestinal symptoms such as abdominal pain, vomiting, and diarrhea.
- it is further accompanied by complications of acute encephalopathy, secondary pneumonia infection, or a combination thereof.
- D in the present invention represents deuterium ( 2 H).
- oxo refers to a group in which two hydrogen atoms at the same substitution position are replaced by the same oxygen atom to form a double bond.
- the term "pharmaceutically acceptable salt” or “pharmaceutically acceptable salt” refers to a salt that is suitable for use in contact with mammalian tissues, particularly human tissues, without excessive toxicity, irritation, allergic response, etc., and is commensurate with a reasonable benefit/risk ratio, within the scope of sound medical judgment.
- pharmaceutically acceptable salts of amines, carboxylic acids, and other types of compounds are well known in the art.
- the salts can be prepared in situ during the final isolation and purification of the compounds of the present invention, or separately by reacting the free base or free acid with a suitable reagent.
- pharmaceutically acceptable salts of the compounds of the present invention also include “solvates” thereof.
- solvate and “solvate” refer to the physical association of a salt of a compound of the present invention with one or more solvent molecules (whether organic or inorganic). This physical association includes hydrogen bonding. In certain cases, for example, when one or more solvent molecules are incorporated into the crystal lattice of a crystalline solid, the solvate will be able to be separated.
- the solvent molecules in the solvate may exist in a regular and/or disordered arrangement.
- the solvate may contain stoichiometric or non-stoichiometric amounts of solvent molecules.
- “Solvate” encompasses solution phases and separable solvates. Exemplary solvates include, but are not limited to, hydrates, ethanolates, methanolates, and isopropanolates. Solvation methods are well known in the art.
- pharmaceutically acceptable salts of the compounds of the present invention also include “hydrates” thereof.
- hydrate refers to a substance formed by water molecules binding to cations or anions in the compound by coordinate bonds or covalent bonds, or refers to a substance formed by water ions not directly binding to cations or anions but existing in a certain proportion at a certain position in the solid crystal lattice.
- the compounds of the present invention also include their "prodrugs,” which refers to drugs that are converted into the parent drug in vivo.
- Prodrugs are generally useful because they can improve certain, undesirable physical or biological properties. Physical properties are often related to solubility (excessive or insufficient lipid or water solubility) or stability, while problematic biological properties include rapid metabolism or poor bioavailability, which may themselves be related to physicochemical properties. For example, they may be bioavailable by oral administration, whereas the parent drug is not. Prodrugs also have improved solubility in pharmaceutical compositions compared to the parent drug.
- prodrug any compound of the present invention administered as an ester ("prodrug") to facilitate transport across cell membranes, where water solubility is detrimental to mobility, but once inside the cell, water solubility is beneficial, which is then metabolically hydrolyzed to the carboxylic acid, the active entity.
- prodrug is a short peptide (polyamino acid) conjugated to an acid group, where the peptide is metabolized to reveal the active moiety.
- stereoisomer refers to compounds that have the same chemical constitution but differ in the way the atoms or groups are arranged in space.
- Stereoisomers include enantiomers, diastereomers, conformers (rotamers), geometric isomers (cis/trans) isomers, atropisomers, etc. Any resulting mixture of stereoisomers can be separated into pure or substantially pure geometric isomers, enantiomers, and diastereomers based on the differences in the physicochemical properties of the components, for example, by chromatography and/or fractional crystallization.
- the term “geometric (cis/trans) isomers” may contain carbon-carbon double bonds or carbon-nitrogen double bonds in the E or Z configuration, wherein the term “E” represents higher-order substituents on opposite sides of the carbon-carbon or carbon-nitrogen double bond and the term “Z” represents higher-order substituents on the same side of the carbon-carbon or carbon-nitrogen double bond (determined using the Cahn-Ingold Prelog priority rules).
- the compounds of the present invention may also exist as mixtures of "E” and "Z” isomers.
- tautomer refers to structural isomers of different energies that are interconvertible through a low energy barrier. If tautomerism is possible (e.g., in solution), a chemical equilibrium of the tautomers can be achieved.
- proton tautomers also known as prototropic tautomers
- Valence tautomers include interconversions that occur via reorganization of some of the bonding electrons.
- the structural formulas described herein include all isomeric forms (e.g., enantiomers, diastereomers, and geometric isomers (or conformers)): for example, R and S configurations containing asymmetric centers, (Z) and (E) isomers of double bonds, and (Z) and (E) conformers. Therefore, single stereochemical isomers of the compounds of the present invention or mixtures of their enantiomers, diastereomers, or geometric isomers (or conformers) are within the scope of the present invention.
- an “effective amount,” also referred to as a “therapeutically effective amount,” refers to an amount of a compound or pharmaceutical composition described herein sufficient to achieve the intended application, including but not limited to treatment of a disease or alleviation of symptoms.
- the amount is a dose that can induce a specific response in cells, or a dose that can exert a therapeutic effect on a disease in a model animal.
- the specific amount will vary depending on, for example, the specific compound selected, the type of subject and their age/existing health condition or risk of health condition, the dosing regimen followed, the severity of the disease, whether it is administered in combination with other agents, the timing of administration, the tissue to which it is administered, and the physical delivery system used to carry it.
- Each carrier must be “acceptable” in the sense of being compatible with the other components of the formulation and harmless to the patient.
- the preparation of the pharmaceutical compositions described herein includes, but is not limited to, for example, mixing the compound of Formula I or a pharmaceutically acceptable salt thereof described in the first aspect with a pharmaceutically acceptable carrier.
- This invention designs a class of novel compounds, offering a new direction for the development of drugs known as cap-dependent endonuclease inhibitors.
- Experimental studies have shown that these compounds exhibit potent inhibitory effects against influenza virus, good PK properties, and/or oral absorption, making them promising compounds for the treatment of cap-dependent endonuclease-mediated diseases.
- the structures of the compounds of the present invention are determined by nuclear magnetic resonance (NMR) and/or liquid chromatography-mass spectrometry (LC-MS) and/or liquid chromatography (HPLC).
- NMR nuclear magnetic resonance
- LC-MS liquid chromatography-mass spectrometry
- HPLC liquid chromatography
- the starting materials in the examples of the present invention are known and can be purchased commercially, or can be synthesized using or according to methods known in the art.
- Polyphosphoric acid (59 g) was added to the reaction flask, stirred and heated to 80°C, compound 1-4 (12 g) was added, the temperature was raised to 120°C and stirred for reaction for 3 hours, the temperature was lowered to 80°C, water (30 mL) was slowly added, the temperature was lowered to 30°C, water (120 mL) and ethyl acetate (120 mL) were added to extract the phases, the organic phase was washed with 10% sodium bicarbonate aqueous solution (35 mL), and the organic phase was concentrated under reduced pressure.
- Polyphosphoric acid 45 g was added to the reaction flask, stirred and heated to 80°C, compound 2-2 (9.0 g) was added, the temperature was raised to 120°C and stirred for 3 hours, the temperature was lowered to 80°C, water (25 mL) was slowly added, the temperature was lowered to 30°C, water (90 mL) and ethyl acetate (90 mL) were added for phase separation, the organic phase was washed with 10% aqueous sodium bicarbonate solution (30 mL), and the organic phase was concentrated under reduced pressure.
- Aluminum chloride (30.0 g) and toluene (150 mL) were added to a reaction flask, stirred and cooled to 0°C.
- a solution of 1,1,3,3-tetramethyldisiloxane (30.0 g) in toluene (60 mL) was added dropwise. After the addition was completed, the temperature was raised to 25°C.
- a solution of compound 2-1 (22.8 g) in toluene (90 mL) was slowly added. The mixture was stirred at 25°C for 2.5 hours. 15% aqueous sulfuric acid solution (210 mL) was added for extraction and phase separation. The organic phase was washed twice with water (120 mL) and concentrated under reduced pressure.
- N-methylpyrrolidone (5 mL), intermediate compound 1-8 (1 g), and lithium chloride (0.57 g) were added to a reaction flask. The temperature was raised to 75°C, and methanesulfonic acid (70 mg) was added. The reaction was maintained at 75°C for 20 h. The temperature was lowered to 45°C, and acetonitrile (1.5 mL) and water 1 (10 mL) were added. The mixture was stirred at 45°C for 1 hour, then lowered to 15°C, stirred at this temperature for 1 hour, and filtered.
- Example 2-5 The following compounds of Example 2-5 were prepared by referring to the preparation method of Example 1
- Tetrahydrofuran (5 mL), N,N-dimethylacetamide (0.5 mL), compound 6 (0.5 g), anhydrous potassium carbonate (0.19 g), potassium iodide (70 mg), isopropyl alcohol (30 mL), and purified water (15 mL) were added to the reaction flask and heated to 60°C with stirring. Chloromethyl methyl carbonate (0.22 g) was then added. The mixture was reacted at 60°C for 10 hours. The mixture was cooled to room temperature, glacial acetic acid (0.1 g) was added, and the mixture was stirred for 15 minutes. Water (2.5 mL) was added and stirred for 30 minutes. The mixture was extracted and separated.
- Example 7-35 The following compound of Example 7-35 was prepared by referring to the preparation method of Example 6
- DMSO dimethyl sulfoxide
- PEG400 polyethylene glycol 400
- MC methylcellulose
- control compound mabaloxavir
- compounds 1-5 and 11-35 of the present invention were prepared respectively using the following formulations.
- Intravenous administration Dosage: 1 mg/kg, volume: 5 ml/kg, concentration: 0.2 mg/ml; 5% DMSO + 10% PEG400 + 85% aqueous solution;
- Oral administration Dosage: 5 mg/kg, Dosage volume: 10 ml/kg, Dosage concentration: 0.5 mg/ml; 0.5% methylcellulose (MC) suspension.
- iv 1 mg/kg or oral gavage 0.2 mL of blood was collected from the jugular vein in EDTA-K2 anticoagulant tubes at 5 min, 15 min, 0.5, 1, 2, 4, 8, and 24 h after administration.
- the blood samples were placed on wet ice and centrifuged within 1 h (4°C, 3500 g, 5 min) to separate the plasma.
- the plasma was placed in a labeled 1.5 mL EP tube and stored in a -80°C ultra-low temperature freezer for testing. In the po.
- 0.2 mL of blood was collected from the jugular vein before administration and 0.25, 0.5, 1, 2, 4, 6, 8, and 24 h after administration.
- the treatment method was the same as that of the iv administration group.
- the concentration of the unchanged drug in plasma was determined by LC/MS/MS, and the blood concentration-time curve was drawn.
- the main pharmacokinetic parameters were calculated using WinNonlin 7.2 software.
- Test Example 2 Detection of virus inhibition activity (EC50) and cytotoxicity (CC50) of compounds 1
- MDCK cells were seeded at a certain density in a 96-well plate, and then the cells were cultured in a 37°C, 5% CO2 incubator overnight.
- Test compounds (control compound baloxavir and compounds 6-10 of the present invention) were serially diluted with DMSO to a final DMSO concentration of 0.5% and added to the cell culture plates. CC50 determinations were performed using a 3-fold dilution from 50 ⁇ M for 10 concentrations. EC50 determinations were performed using a 3-fold dilution from 100 nM for 10 concentrations.
- Virus Inoculation Add diluted influenza virus (A/WSN/33 (H1N1) or A/PR/8/34 (H1N1)) to the antiviral activity test wells; add only the compound, without virus, to the cytotoxicity test wells. Incubate the cells in a 37°C, 5% CO2 incubator for 3-5 days until the cytopathic effect (CPE) rate in the virus control wells (no compound) reaches 80-95%.
- CPE cytopathic effect
- Cell viability assay reagent (CCK-8 assay reagent) was added to each well. After culturing the cells in a 37°C, 5% CO2 incubator for 3-4 hours, the signal value was detected by a microplate reader. The raw data was used to calculate the anti-influenza virus activity and cytotoxicity of the compound.
- the experimental results show that the compound of the present invention has good influenza virus inhibitory activity and low cytotoxicity.
- Test Example 3 Detection of Virus Inhibitory Activity (EC50) and Cytotoxicity (CC50) of Compounds 2
- the in vitro anti-influenza virus activity of the compounds of the present invention was investigated using an indirect immunofluorescence virus quantitative assay (FFA) method.
- FFA indirect immunofluorescence virus quantitative assay
- Influenza Anti-HA Serum strain-specific serum
- Type A H1N1 A/Victoria/2570/2019 & A/Victoria/4897/2022
- Type A H3N2A/Cambodia/E0826360/2020&A/Darwin/9/2021
- Cell preparation Observe the cell status and ensure the confluence is 85% to 95% and the viability is ⁇ 90%. Digest the cells and inoculate them into 96-well cell culture plates at a concentration of 2.5 to 3.0 ⁇ 10 5 cells/mL, 100 ⁇ L/well, and culture in a 37°C CO 2 incubator for approximately 24 hours.
- Virus dilution Take the virus solution and dilute it with MEM medium according to the virus titer of each strain to 8000 PFU/mL as the virus working solution. Place it on ice or at 2-8°C until ready for use.
- Test compounds (control compound 1: baloxavir, control compound 2: Example compound 1A in CN108440564A, and compounds of the present invention) were serially diluted with DMSO to a final DMSO concentration of 0.5% and added to the cell culture plate.
- EC50 determination dilution factor 20 concentrations, starting from 100 nM, with 3-fold dilutions.
- Compound Dosing and Virus Inoculation Remove the pre-seeded cell plate from the 37°C CO2 incubator, spin dry the culture supernatant, add 100 ⁇ L/well MEM medium, and wash twice. Spin dry any remaining liquid. According to the designed plate layout, pipette the compound serial dilutions and compound blank dilutions at a rate of 50 ⁇ L/well onto the antiviral assay plate using a multichannel pipette. Except for wells CC (cell control), add 50 ⁇ L of the corresponding virus working solution to each well. Gently shake the plate to mix thoroughly. Incubate the plate in a 37°C, 5% CO2 incubator for 18–24 hours.
- the experimental results show that the compound of the present invention is significantly superior to the control compound and has good influenza virus inhibitory activity.
- the distribution levels of the compounds of the present invention in the lungs and plasma were investigated after oral administration to mice.
- MC methylcellulose
- mice Balb/c mice, male.
- Oral administration The dosage is 10 mg/kg, the administration volume is 10 ml/kg, and 0.5% methylcellulose (MC) is used to prepare a uniform suspension with a dosage concentration of 1.0 mg/ml.
- MC methylcellulose
- mice 6-8 week old Balb/c mice (3 animals at each time point) were fasted overnight (with free access to water) and then orally administered with 10 mg/kg of compound (such as compound 1 of the present invention); blood samples were collected at 0.5, 1, 2, 4, and 8 hours after the drug administration for each compound group, and the mice were immediately killed and lung tissue was collected.
- the blood samples were collected into test tubes with stabilizers added in advance, mixed by inversion, and centrifuged within 30 minutes (4°C, 3000g, 10 minutes).
- the plasma was separated and frozen for testing. After the lung tissue was collected, it was immediately placed in an ice water bath, weighed within 20 minutes, and glacial acetonitrile was quickly added according to 1g:3mL. The homogenate was completed within 1 hour for testing.
- the metabolite compounds (such as compound 6 of the present invention) in the plasma were determined by LC/MS/MS method, and the results are as follows:
- DMSO dimethyl sulfoxide
- PEG400 polyethylene glycol 400
- MC methylcellulose
- Intravenous administration Dosage: 1 mg/kg, dosing volume: 2 ml/kg, dosing concentration: 0.5 mg/ml; 5% DMSO + 30% PEG400 + 65% aqueous solution;
- Oral administration Dosage: 10 mg/kg, administration volume: 2 ml/kg, administration concentration: 0.5 mg/ml; 0.5% methylcellulose (MC) suspension.
- Blood samples (0.3 mL) were collected from all four limb veins in the iv group at 2, 5, 0.25, 0.5, 1, 2, 4, 8, 24, and 48 h after administration. The samples were placed in EDTA-K2 anticoagulant tubes (pre-added with 5 ⁇ L PMSF solution (300 mM in methanol)). The tubes were gently shaken several times, placed in an ice-water bath, and centrifuged within 30 min (3000 g, 4°C, 10 min).
- Plasma was separated and frozen at -70°C as soon as possible for analysis. Blood samples were collected from the po group before administration and at 0.25, 0.5, 1, 2, 4, 8, 24, and 48 h after administration and processed as described for the intravenous group.
- the metabolites in plasma (such as compound 6, the metabolite of the invention compound) were determined by LC/MS/MS, and the blood drug concentration-time curve was drawn. The main pharmacokinetic parameters were calculated using WinNonlin 8.0 software.
- the experimental results show that the compound of the present invention has good PK properties and oral absorption properties after oral administration.
- Test Example 6 Stability test of compound in human liver S9 in vitro
- Test samples 1 ⁇ M baloxavir and compound 1 of the present invention
- Reagents Phosphate buffer: 200 mM, 200 ⁇ L, final concentration 100 mM; Ultrapure water: 138 ⁇ L, MgCl2 solution: 50 mM, 40 ⁇ L, final concentration 5 mM; S9 component: 20 mg/mL, 20 ⁇ L, final concentration 1 mg/mL
- the experimental results show that the compound of the present invention is metabolized faster in human liver S9 than Yangshen, and is more likely to release the original drug molecule.
- Test Example 7 Stability test of compound in human intestinal S9 in vitro
- Test samples 1 ⁇ M baloxavir and compound 1 of the present invention
- Reagents Phosphate buffer: 200 mM, 200 ⁇ L, final concentration 100 mM; Ultrapure water: 138 ⁇ L, MgCl2 solution: 50 mM, 40 ⁇ L, final concentration 5 mM; S9 component: 20 mg/mL, 20 ⁇ L, final concentration 1 mg/mL
- Test Example 8 Stability test of compound in human plasma
- Test samples Yangshenmabaloxavir and compound 1 of the present invention
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Organic Chemistry (AREA)
- Life Sciences & Earth Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Virology (AREA)
- Pharmacology & Pharmacy (AREA)
- Medicinal Chemistry (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Oncology (AREA)
- Communicable Diseases (AREA)
- Epidemiology (AREA)
- Pulmonology (AREA)
- Molecular Biology (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Description
本发明涉及医药技术领域,具体而言,涉及一类多环吡啶酮衍生物、其组合物及用途。The present invention relates to the field of medical technology, and in particular to a class of polycyclic pyridone derivatives, compositions and uses thereof.
流行性感冒是流感病毒引起的具有高度传染性的呼吸道疾病,其中甲型流感病毒宿主范围最广,既可以感染禽类也可以感染哺乳类,极易造成世界范围内大流行。目前,疫苗接种在预防流感感染方面一般是有效的,但是,疫苗只是部分有效,总体有效率约为60%。此外,由于在抗原鉴定到疫苗生产之间一般需要6个月左右的时间,病毒可能会在此期间发生变异,从而导致疫苗的有效性降低。在抗流感病毒的药物方面,经FDA批准的抗流感感染药物有两类:流感神经氨酸酶蛋白抑制剂(奥司他韦、扎那米韦、帕那米韦和那尼纳米韦等)和M2质子通道阻滞剂金刚烷胺类药物(金刚烷胺和金刚乙胺等)。然而,有耐药性菌株的出现、副作用的问题,又有病原性或致死性较高的新型流行性感冒病毒世界大流行等顾虑,因此期望开发出新机理的抗流行性感冒药。Influenza is a highly contagious respiratory disease caused by the influenza virus. Among them, influenza A virus has the widest host range, infecting both birds and mammals, making it highly likely to cause a worldwide pandemic. Currently, vaccination is generally effective in preventing influenza infection, but the vaccine is only partially effective, with an overall efficacy of approximately 60%. Furthermore, since it typically takes about six months between antigen identification and vaccine production, the virus may mutate during this period, thereby reducing the effectiveness of the vaccine. Regarding anti-influenza drugs, there are two types of FDA-approved anti-influenza drugs: influenza neuraminidase protein inhibitors (oseltamivir, zanamivir, peramivir, and nanamivir) and M2 proton channel blockers, the amantadine class of drugs (amantadine and rimantadine). However, there are concerns about the emergence of drug-resistant strains, side effects, and the global prevalence of novel influenza viruses with high pathogenicity or lethality, leading to the development of anti-influenza drugs with novel mechanisms.
源自流行性感冒病毒的酶的帽依赖性核酸内切酶,由于其对于病毒增殖而言是必须的,且具有宿主所不具有的病毒特异性酶活性,故而认为该帽依赖性核酸内切酶适于抗流行性感冒药的目标物。流行性感冒病毒的帽依赖性内切酶是生成以宿主mRNA前体为底物且包含帽结构的9~13个碱基(帽结构的碱基并不包含于上述碱基数量中)的片段的核酸内切酶活性。该片段作为病毒RNA聚合酶的引子发挥功能,用于编码病毒蛋白质的mRNA的合成。即,认为抑制帽依赖性核酸内切酶的物质通过抑制病毒mRNA的合成而抑制病毒蛋白质的合成,结果抑制病毒增殖。已经在多国上市的药物玛巴洛沙韦即是一款创新的帽状结构依赖性核酸内切酶抑制剂类药物,全程只需一次口服,就能在24小时左右停止病毒排毒,缩短传染期并快速缓解发热及全身酸痛等流感症状。在已经证实了帽依赖性核酸内切酶类药物具有很强的抗流感病毒作用的基础上,研发出种类更加丰富、效果更加突出的新型药物仍十分必要。Cap-dependent endonucleases, enzymes derived from influenza viruses, are considered suitable targets for anti-influenza drugs because they are essential for viral proliferation and possess virus-specific enzymatic activity not possessed by the host. The influenza virus cap-dependent endonuclease is an endonuclease activity that generates a fragment containing 9 to 13 bases of the cap structure (the bases of the cap structure are not included in the above base count) with the host mRNA precursor as a substrate. This fragment functions as a primer for the viral RNA polymerase, used for the synthesis of mRNA encoding viral proteins. In other words, substances that inhibit cap-dependent endonucleases are believed to inhibit viral protein synthesis by inhibiting viral mRNA synthesis, thereby inhibiting viral proliferation. Mabaloxavir, a drug already marketed in multiple countries, is an innovative cap-dependent endonuclease inhibitor. With a single oral dose, it can halt viral shedding within about 24 hours, shorten the infectious period, and quickly relieve flu symptoms such as fever and body aches. Based on the fact that cap-dependent nuclease drugs have been proven to have strong anti-influenza virus effects, it is still necessary to develop new drugs with richer types and more outstanding effects.
本发明提供了一种新型的多环吡啶酮衍生物,其药物组合物及在抗流感病毒方面的用途。The present invention provides a novel polycyclic pyridone derivative, a pharmaceutical composition thereof and use thereof in resisting influenza virus.
第一方面,本发明提供了一种如式I所示的化合物、其互变异构体、立体异构体或其药学上可接受的盐:
In a first aspect, the present invention provides a compound as shown in Formula I, its tautomers, stereoisomers or pharmaceutically acceptable salts thereof:
其中,R1、R2、R3、R4全部为氢或全部为氘,R5不存在或者为氧代基,R6为氢或 wherein R 1 , R 2 , R 3 and R 4 are all hydrogen or all deuterium, R 5 is absent or is an oxo group, and R 6 is hydrogen or
在本发明的一个优选实施方案中,R1、R2、R3、R4为氘,R5不存在或者为氧代基,R6为氢或 In a preferred embodiment of the present invention, R 1 , R 2 , R 3 and R 4 are deuterium, R 5 is absent or is oxo, and R 6 is hydrogen or
在本发明的一个优选实施方案中,R1、R2、R3、R4为氢,R5为氧代基,R6为氢或 In a preferred embodiment of the present invention, R 1 , R 2 , R 3 , and R 4 are hydrogen, R 5 is oxo, and R 6 is hydrogen or
在本发明的一个优选实施方案中,R1、R2、R3、R4为氘,R5为氧代基,硫原子的构型为S或R,R6为氢或 In a preferred embodiment of the present invention, R 1 , R 2 , R 3 , and R 4 are deuterium, R 5 is an oxo group, the configuration of the sulfur atom is S or R, and R 6 is hydrogen or
在本发明的一个优选实施方案中,R1、R2、R3、R4为氢,R5为氧代基,硫原子的构型为S或R,R6为氢或 In a preferred embodiment of the present invention, R 1 , R 2 , R 3 , and R 4 are hydrogen, R 5 is an oxo group, the configuration of the sulfur atom is S or R, and R 6 is hydrogen or
本发明所述化合物,选自:
The compound of the present invention is selected from:
及其互变异构体、立体异构体或其药学上可接受的盐。and its tautomers, stereoisomers or pharmaceutically acceptable salts.
本发明第二方面,提供了一种药物组合物,其包含式I所示的化合物、其互变异构体、立体异构体或其药学上可接受的盐,任选地,还包括药学上可接受的载体。In a second aspect, the present invention provides a pharmaceutical composition comprising a compound of Formula I, its tautomers, stereoisomers or pharmaceutically acceptable salts thereof, and optionally, a pharmaceutically acceptable carrier.
本发明第三方面,提供了式I所示的化合物、其互变异构体、立体异构体或其药学上可接受的盐或本发明第二方面所述的药物组合物在制备治疗和/或预防帽依赖性核酸内切酶介导的疾病或病症的药物中的用途。In a third aspect, the present invention provides the use of a compound represented by Formula I, its tautomers, stereoisomers or pharmaceutically acceptable salts thereof, or the pharmaceutical composition described in the second aspect of the present invention in the preparation of a drug for treating and/or preventing diseases or conditions mediated by cap-dependent nucleases.
本发明还提供了式I所示的化合物、其互变异构体、立体异构体或其药学上可接受的盐或本发明第二方面所述的药物组合物在制备治疗和/或预防病毒感染的疾病或病症的药物中的用途。The present invention also provides the use of the compound represented by Formula I, its tautomers, stereoisomers or pharmaceutically acceptable salts thereof, or the pharmaceutical composition described in the second aspect of the present invention in the preparation of drugs for treating and/or preventing diseases or conditions caused by viral infections.
本发明第四方面,提供了一种治疗和/或预防帽依赖性核酸内切酶介导的疾病或病症的方法,包括向患者施用治疗有效剂量的式I所示的化合物、其互变异构体、立体异构体或其药学上可接受的盐或本发明第二方面所述的药物组合物。In a fourth aspect, the present invention provides a method for treating and/or preventing diseases or conditions mediated by cap-dependent nucleases, comprising administering to a patient a therapeutically effective dose of a compound of formula I, its tautomers, stereoisomers or pharmaceutically acceptable salts thereof, or the pharmaceutical composition described in the second aspect of the present invention.
本发明还提供了一种治疗和/或预防病毒感染的疾病或病症的方法,包括向患者施用治疗有效剂量的式I所示的化合物、其互变异构体、立体异构体或其药学上可接受的盐或本发明第二方面所述的药物组合物。优选的,本发明所述病毒感染为流感病毒感染。进一步的,所述流感病毒选自甲型、乙型、丙型或禽流感(H5N1,H7N9);进一步的,本发明所述病症选自伴有发热、发冷、头痛、肌肉痛、全身感到倦怠等的类感冒症状,或咽痛、流鼻涕、鼻塞、咳嗽、痰的呼吸道炎症;腹痛、呕吐、腹泻的胃肠症状,进而伴有急性脑病、肺炎二次感染的并发症或其组合。The present invention also provides a method for treating and/or preventing a disease or condition caused by a viral infection, comprising administering to a patient a therapeutically effective dose of a compound shown in Formula I, a tautomer, a stereoisomer, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition described in the second aspect of the present invention. Preferably, the viral infection of the present invention is an influenza virus infection. Further, the influenza virus is selected from type A, type B, type C, or avian influenza (H5N1, H7N9); further, the disease of the present invention is selected from cold-like symptoms accompanied by fever, chills, headache, muscle pain, general fatigue, etc., or respiratory inflammation with sore throat, runny nose, nasal congestion, cough, and sputum; gastrointestinal symptoms of abdominal pain, vomiting, and diarrhea, and then accompanied by complications of acute encephalopathy, secondary pneumonia infection, or a combination thereof.
本发明还提供了式I所示的化合物、其互变异构体、立体异构体或其药学上可接受的盐或本发明第二方面所述的药物组合物在作为药物或制备药物中的用途。The present invention also provides the use of the compound represented by Formula I, its tautomers, stereoisomers or pharmaceutically acceptable salts thereof, or the pharmaceutical composition described in the second aspect of the present invention as a drug or in the preparation of a drug.
优选地,所述药物为用于治疗和/或预防帽依赖性核酸内切酶介导的疾病或病症的药物。优选地,所述疾病或病症为流感传染病。优选地,所述流感传染病由流感病毒引起,所述流感病毒选自甲型、乙型、丙型或禽流感(H5N1,H7N9)。优选地,所述流感传染病伴有以下症状中的一种或多种:发热、发冷、头痛、肌肉痛、全身感到倦怠等的类感冒症状,或咽痛、流鼻涕、鼻塞、咳嗽、痰的呼吸道炎症;腹痛、呕吐、腹泻的胃肠症状。优选地,进一步伴有急性脑病、肺炎二次感染的并发症或其组合。Preferably, the drug is a drug for treating and/or preventing a disease or condition mediated by a cap-dependent endonuclease. Preferably, the disease or condition is an influenza infectious disease. Preferably, the influenza infectious disease is caused by an influenza virus selected from influenza A, B, C or avian influenza (H5N1, H7N9). Preferably, the influenza infectious disease is accompanied by one or more of the following symptoms: cold-like symptoms such as fever, chills, headache, muscle pain, general fatigue, or respiratory inflammation such as sore throat, runny nose, nasal congestion, cough, and sputum; gastrointestinal symptoms such as abdominal pain, vomiting, and diarrhea. Preferably, it is further accompanied by complications of acute encephalopathy, secondary pneumonia infection, or a combination thereof.
定义definition
除另有规定外,本发明所述D代表氘(2H)。Unless otherwise specified, D in the present invention represents deuterium ( 2 H).
除另有规定外,术语“氧代基”是指相同取代位的两个氢原子被同一个氧原子替代形成双键。Unless otherwise specified, the term "oxo" refers to a group in which two hydrogen atoms at the same substitution position are replaced by the same oxygen atom to form a double bond.
除另有规定外,术语“药学上可接受的盐”或“可药用盐”是指在合理医学判断范围内适用于与哺乳动物特别是人的组织接触而无过度毒性、刺激、过敏反应等并与合理的效益/风险比相称的盐,比如胺、羧酸和其他类型化合物的医学上可接受的盐在所属领域中是被熟知的。可以在本发明化合物的最终分离和纯化期间原位制备所述盐,或单独通过将游离碱或游离酸与合适的试剂反应制备所述盐。Unless otherwise specified, the term "pharmaceutically acceptable salt" or "pharmaceutically acceptable salt" refers to a salt that is suitable for use in contact with mammalian tissues, particularly human tissues, without excessive toxicity, irritation, allergic response, etc., and is commensurate with a reasonable benefit/risk ratio, within the scope of sound medical judgment. For example, pharmaceutically acceptable salts of amines, carboxylic acids, and other types of compounds are well known in the art. The salts can be prepared in situ during the final isolation and purification of the compounds of the present invention, or separately by reacting the free base or free acid with a suitable reagent.
除另有规定外,本发明化合物的药学上可接受的盐还包括其“溶剂化物”,术语“溶剂化物”、“溶剂合物”意指本发明化合物的盐与一个或多个溶剂分子(无论有机的还是无机的)的物理缔合。该物理缔合包括氢键。在某些情形中,例如当一个或多个溶剂分子纳入结晶固体的晶格中时,溶剂化物将能够被分离。溶剂化物中的溶剂分子可按规则排列和/或无序排列存在。溶剂化物可包含化学计量或非化学计量的溶剂分子。“溶剂化物”涵盖溶液相和可分离的溶剂化物。示例性溶剂化物包括但不限于水合物、乙醇合物、甲醇合物和异丙醇合物。溶剂化方法是本领域公知的。Unless otherwise specified, pharmaceutically acceptable salts of the compounds of the present invention also include "solvates" thereof. The terms "solvate" and "solvate" refer to the physical association of a salt of a compound of the present invention with one or more solvent molecules (whether organic or inorganic). This physical association includes hydrogen bonding. In certain cases, for example, when one or more solvent molecules are incorporated into the crystal lattice of a crystalline solid, the solvate will be able to be separated. The solvent molecules in the solvate may exist in a regular and/or disordered arrangement. The solvate may contain stoichiometric or non-stoichiometric amounts of solvent molecules. "Solvate" encompasses solution phases and separable solvates. Exemplary solvates include, but are not limited to, hydrates, ethanolates, methanolates, and isopropanolates. Solvation methods are well known in the art.
除另有规定外,本发明化合物的药学上可接受的盐还包括其“水合物”,术语“水合物”是指水分子以配位键或共价键与化合物中的阳离子或阴离子结合,或指水离子不直接与阳离子或阴离子结合而是以一定比例存在于固体晶格的确定位置而形成的物质。Unless otherwise specified, pharmaceutically acceptable salts of the compounds of the present invention also include "hydrates" thereof. The term "hydrate" refers to a substance formed by water molecules binding to cations or anions in the compound by coordinate bonds or covalent bonds, or refers to a substance formed by water ions not directly binding to cations or anions but existing in a certain proportion at a certain position in the solid crystal lattice.
除另有规定外,本发明化合物还包括其“前药”,“前药”是指在体内转化为母体药物的药物。前药通常是有用的,其可以改善一些确定的、不合需要的物理或生物学性质。物理性能通常是相关的溶解度(过高或不足的脂质或水溶性)或稳定性,而有问题的生物学特性包括代谢太快或生物利用率差,这本身可能与物理化学性质相关。例如,它们可以通过口服而被生物利用,而母体则不能。与母体药物相比,前药在药物组合物中的溶解度也有所提高。前药的一个例子,但不限于此,可以是任何本发明的化合物,其作为酯(“前药”)给药,以促进穿过细胞膜的传递,其中水溶性对迁移性有害,但一旦进入细胞内水溶性是有益的,其随后被代谢水解成羧酸,即活性实体。前药的另一个例子可以是与酸基团结合的短肽(聚氨基酸),其中肽被代谢以显示活性部分。Unless otherwise specified, the compounds of the present invention also include their "prodrugs," which refers to drugs that are converted into the parent drug in vivo. Prodrugs are generally useful because they can improve certain, undesirable physical or biological properties. Physical properties are often related to solubility (excessive or insufficient lipid or water solubility) or stability, while problematic biological properties include rapid metabolism or poor bioavailability, which may themselves be related to physicochemical properties. For example, they may be bioavailable by oral administration, whereas the parent drug is not. Prodrugs also have improved solubility in pharmaceutical compositions compared to the parent drug. An example, but not limited to, of a prodrug is any compound of the present invention administered as an ester ("prodrug") to facilitate transport across cell membranes, where water solubility is detrimental to mobility, but once inside the cell, water solubility is beneficial, which is then metabolically hydrolyzed to the carboxylic acid, the active entity. Another example of a prodrug is a short peptide (polyamino acid) conjugated to an acid group, where the peptide is metabolized to reveal the active moiety.
除另有规定外,术语“立体异构体”是指具有相同化学构造,但原子或基团在空间上排列方式不同的化合物。立体异构体包括对映异构体、非对映异构体、构象异构体(旋转异构体)、几何异构体(顺/反)异构体、阻转异构体等。所得的任何立体异构体的混合物可以依据组分物理化学性质上的差异被分离成纯的或基本纯的几何异构体,对映异构体,非对映异构体,例如,通过色谱法和/或分步结晶法。Unless otherwise specified, the term "stereoisomer" refers to compounds that have the same chemical constitution but differ in the way the atoms or groups are arranged in space. Stereoisomers include enantiomers, diastereomers, conformers (rotamers), geometric isomers (cis/trans) isomers, atropisomers, etc. Any resulting mixture of stereoisomers can be separated into pure or substantially pure geometric isomers, enantiomers, and diastereomers based on the differences in the physicochemical properties of the components, for example, by chromatography and/or fractional crystallization.
除另有规定外,术语“几何异构体(顺/反)异构体”可以含有E或Z构型的碳-碳双键或碳-氮双键,其中术语“E”代表碳-碳或碳-氮双键的对侧的更高顺序取代基,术语“Z”代表碳-碳或碳-氮双键的同侧上的更高顺序取代基(利用Cahn-Ingold Prelog优先规则确定)。本发明的化合物还可以以“E”和“Z”异构体的混合物形态存在。Unless otherwise specified, the term "geometric (cis/trans) isomers" may contain carbon-carbon double bonds or carbon-nitrogen double bonds in the E or Z configuration, wherein the term "E" represents higher-order substituents on opposite sides of the carbon-carbon or carbon-nitrogen double bond and the term "Z" represents higher-order substituents on the same side of the carbon-carbon or carbon-nitrogen double bond (determined using the Cahn-Ingold Prelog priority rules). The compounds of the present invention may also exist as mixtures of "E" and "Z" isomers.
除另有规定外,术语“互变异构体”是指具有不同能量的可通过低能垒互相转化的结构异构体。若互变异构是可能的(如在溶液中),则可以达到互变异构体的化学平衡。例如,质子互变异构体(也称为质子转移互变异构体)包括通过质子迁移来进行的互相转化,如酮-烯醇异构化和亚胺-烯胺异构化。价键互变异构体包括通过一些成键电子的重组来进行的互相转化。Unless otherwise specified, the term "tautomer" refers to structural isomers of different energies that are interconvertible through a low energy barrier. If tautomerism is possible (e.g., in solution), a chemical equilibrium of the tautomers can be achieved. For example, proton tautomers (also known as prototropic tautomers) include interconversions that occur via proton migration, such as keto-enol isomerization and imine-enamine isomerization. Valence tautomers include interconversions that occur via reorganization of some of the bonding electrons.
除非其他方面表明,本发明所描述的结构式包括所有的同分异构形式(如对映异构,非对映异构,和几何异构(或构象异构)):例如含有不对称中心的R、S构型,双键的(Z)、(E)异构体,和(Z)、(E)的构象异构体。因此,本发明的化合物的单个立体化学异构体或其对映异构体,非对映异构体,或几何异构体(或构象异构体)的混合物都属于本发明的范围。Unless otherwise indicated, the structural formulas described herein include all isomeric forms (e.g., enantiomers, diastereomers, and geometric isomers (or conformers)): for example, R and S configurations containing asymmetric centers, (Z) and (E) isomers of double bonds, and (Z) and (E) conformers. Therefore, single stereochemical isomers of the compounds of the present invention or mixtures of their enantiomers, diastereomers, or geometric isomers (or conformers) are within the scope of the present invention.
“任选”或“任选地”意味着随后所描述的事件或情况可以但不必发生,该说明包括该事件或情况发生或不发生的场合。"Optional" or "optionally" means that the subsequently described event or circumstance may but need not occur, and that the description includes instances where the event or circumstance occurs and instances where it does not.
“有效量”也称“治疗有效量”是指足以实现预期的应用的本文所述的化合物或药物组合物的量,包括但不限于疾病的治疗或症状的减轻。在一些实施方案中,例如,所述量是可以诱导细胞产生特定响应的剂量,也可以是在模型动物体内发挥疾病的治疗作用的剂量。具体将将取决于例如所选的具体化合物、受试者的种类和他们的年龄/现有的健康状况或健康状况的风险、所遵循的给药方案、疾病的严重性、是否与其它药剂组合给药、给药的时间、给药的组织和携带其的物理递送系统而变化。An "effective amount," also referred to as a "therapeutically effective amount," refers to an amount of a compound or pharmaceutical composition described herein sufficient to achieve the intended application, including but not limited to treatment of a disease or alleviation of symptoms. In some embodiments, for example, the amount is a dose that can induce a specific response in cells, or a dose that can exert a therapeutic effect on a disease in a model animal. The specific amount will vary depending on, for example, the specific compound selected, the type of subject and their age/existing health condition or risk of health condition, the dosing regimen followed, the severity of the disease, whether it is administered in combination with other agents, the timing of administration, the tissue to which it is administered, and the physical delivery system used to carry it.
“药学上可接受的载体”也称为“药学上可接受的辅料”或“药学上可接受的赋形剂”,是指涉及将目标活性成分从一个器官或身体的一部分输送或输送至另一个器官或身体的一部分的药学上可接受的材料、组合物或媒介物,例如液体或固体的赋形剂、溶剂或包封材料。各载体与制剂的其它组分相容并且对患者无害的意义上必须是“可接受的”。本文中所述的药物组合物的制备,包括但不限于,例如,第一方面所述的式I所示化合物或其药学上可接受的盐与药学上可接受的载体相混合。A "pharmaceutically acceptable carrier," also known as a "pharmaceutically acceptable excipient" or "pharmaceutically acceptable vehicle," refers to a pharmaceutically acceptable material, composition, or vehicle, such as a liquid or solid excipient, solvent, or encapsulating material, that is used to transport the active ingredient from one organ or part of the body to another. Each carrier must be "acceptable" in the sense of being compatible with the other components of the formulation and harmless to the patient. The preparation of the pharmaceutical compositions described herein includes, but is not limited to, for example, mixing the compound of Formula I or a pharmaceutically acceptable salt thereof described in the first aspect with a pharmaceutically acceptable carrier.
应理解,在本发明的范围内,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征中间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅不再一一赘述。It should be understood that within the scope of the present invention, the above-mentioned technical features of the present invention and the technical features described in detail below (such as in the embodiments) can be combined with each other to form new or preferred technical solutions. Due to space limitations, they will not be described in detail one by one.
本发明的有益效果为:The beneficial effects of the present invention are:
本发明设计了一类结构新颖的化合物,为帽依赖性核酸内切酶抑制剂类的药物的发展提供了一个新的方向。试验研究显示,这些化合物对流感病毒具有较强的抑制作用,有较好的PK性质和/或口服吸收性质,可作为治疗帽依赖性核酸内切酶介导的疾病的前景化合物。This invention designs a class of novel compounds, offering a new direction for the development of drugs known as cap-dependent endonuclease inhibitors. Experimental studies have shown that these compounds exhibit potent inhibitory effects against influenza virus, good PK properties, and/or oral absorption, making them promising compounds for the treatment of cap-dependent endonuclease-mediated diseases.
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件或者按照制造厂商所建议的条件。除非另行定义,文中所使用的所有专业与科学用语与本领域专业人员所熟悉的意义相同。此外,任何与所记载内容相似或均等的方法及材料皆可应用于本发明方法之中。文中所示的较佳实施方法与材料仅做示范之用。The present invention will be further described below in conjunction with specific examples. It should be understood that these examples are intended to illustrate the present invention only and are not intended to limit the scope of the present invention. Experimental methods in the following examples where specific conditions are not specified are generally performed under conventional conditions or according to the conditions recommended by the manufacturer. Unless otherwise defined, all professional and scientific terms used herein have the same meanings as those familiar to professionals in the field. In addition, any methods and materials similar or equivalent to those described herein can be applied to the present invention. The preferred embodiments and materials shown herein are for demonstration purposes only.
本发明的化合物结构是通过核磁共振(NMR)或/和液质联用色谱(LC-MS)或/和液相色谱(HPLC)来确定的。NMR的测定使用的仪器是核磁共振谱仪Bruker Advance 600,LC-MS使用的仪器是液相色谱-质谱联用仪Waters ACQUITY UPLC I-Class Xevo TQ-S;HPLC使用的仪器是高效液相色谱仪Waters e2695。The structures of the compounds of the present invention are determined by nuclear magnetic resonance (NMR) and/or liquid chromatography-mass spectrometry (LC-MS) and/or liquid chromatography (HPLC). The NMR spectrometer used is a Bruker Advance 600, the LC-MS spectrometer is a Waters ACQUITY UPLC I-Class Xevo TQ-S, and the HPLC spectrometer is a Waters e2695.
本发明实施例中的起始原料是已知的并且可以在市场上买到,或者可以采用或按照本领域已知的方法来合成。The starting materials in the examples of the present invention are known and can be purchased commercially, or can be synthesized using or according to methods known in the art.
制备例1:中间体1-8的制备
Preparation Example 1: Preparation of Intermediate 1-8
方法一:将甲苯(36mL)、化合物1-1(5.5g,29.6mmo1)、氘代苯硫酚(8.2g,35.4mmo1)和D-樟脑磺酸(2.32g,10.0mmol)加入到圆底烧瓶中,60℃搅拌反应4小时,然后降温至5℃。加入2mol/L氢氧化钠溶液(20mL),搅拌恢复至室温,加入甲苯(20mL)萃取分相,有机相用2mol/L氢氧化钠(20mL)和水(20mL)各洗绦一次,减压浓缩后得到化合物1-3(9.2g)。MS m/z:284.1[M+H]。Method 1: Toluene (36 mL), compound 1-1 (5.5 g, 29.6 mmol), deuterated thiophenol (8.2 g, 35.4 mmol), and D-camphorsulfonic acid (2.32 g, 10.0 mmol) were added to a round-bottom flask and stirred at 60°C for 4 hours, then cooled to 5°C. 2 mol/L sodium hydroxide solution (20 mL) was added and stirred until the mixture returned to room temperature. Toluene (20 mL) was added for extraction and phase separation. The organic phase was washed once with 2 mol/L sodium hydroxide (20 mL) and once with water (20 mL), and then concentrated under reduced pressure to obtain compound 1-3 (9.2 g). MS m/z: 284.1 [M+H].
将氯化铝(11.1g,82.9mmol)和甲苯(50mL)加入到反应瓶中,搅拌并降温至0℃,滴加1,1,3,3-四甲基二硅氧烷(11.1g,82.8mmol)的甲苯(20mL)溶液,加毕后升温至25℃,缓慢加入化合物1-3的甲苯溶液,25℃搅拌反应2.5小时。加入15%硫酸水溶液(70mL)萃取分相,有机相用水(40mL)洗涤两次,减压浓缩。加入正庚烷(80mL)0℃打浆,过滤,滤饼用正庚烷淋洗,干燥,得到化合物1-4(14.5g)。MS m/z:286.1[M+H]。Aluminum chloride (11.1 g, 82.9 mmol) and toluene (50 mL) were added to a reaction flask, stirred and cooled to 0°C. A solution of 1,1,3,3-tetramethyldisiloxane (11.1 g, 82.8 mmol) in toluene (20 mL) was added dropwise. After the addition was completed, the temperature was raised to 25°C and the toluene solution of compound 1-3 was slowly added. The mixture was stirred at 25°C for 2.5 hours. 15% aqueous sulfuric acid solution (70 mL) was added for extraction and phase separation. The organic phase was washed twice with water (40 mL) and concentrated under reduced pressure. n-heptane (80 mL) was added and the mixture was slurried at 0°C. The mixture was filtered, the filter cake was rinsed with n-heptane, and dried to obtain compound 1-4 (14.5 g). MS m/z: 286.1 [M+H].
反应瓶中加入多聚磷酸(59g),搅拌升温至80℃,加入化合物1-4(12g),升温至120℃搅拌反应3小时,降温至80℃,缓慢加水(30mL),降温至30℃,加水(120mL)和乙酸乙酯(120mL)萃取分相,有机相用10%碳酸氢钠水溶液(35mL)洗涤,有机相减压浓缩。加入正庚烷(60mL)室温打浆,过滤,滤饼用正庚烷淋洗,干燥,得到化合物1-5(10.2g)。MS m/z:267.1[M+H]。Polyphosphoric acid (59 g) was added to the reaction flask, stirred and heated to 80°C, compound 1-4 (12 g) was added, the temperature was raised to 120°C and stirred for reaction for 3 hours, the temperature was lowered to 80°C, water (30 mL) was slowly added, the temperature was lowered to 30°C, water (120 mL) and ethyl acetate (120 mL) were added to extract the phases, the organic phase was washed with 10% sodium bicarbonate aqueous solution (35 mL), and the organic phase was concentrated under reduced pressure. n-heptane (60 mL) was added and the mixture was slurried at room temperature, filtered, the filter cake was rinsed with n-heptane, and dried to obtain compound 1-5 (10.2 g). MS m/z: 267.1 [M+H].
在反应瓶中加入异丙醇(40mL)、水(4.5mL)和化合物1-5(9.2g,17.2mmol),搅拌升温至40℃,缓慢加入硼氢化钠(0.47g,12.4mmol)至0.5%氢氧化钠水溶液中(3.6mL)制备为悬浮液,将悬浮液缓慢加入至反应体系中,40℃搅拌反应1.5小时,降至室温,加入水(65mL),然后加入4%硫酸水溶液(15mL),降温至5℃,过滤,滤饼用水(20mL)淋洗,干燥,得到化合物1-6(8.7g)。MS m/z:269.1[M+H]。Isopropanol (40 mL), water (4.5 mL), and compound 1-5 (9.2 g, 17.2 mmol) were added to a reaction flask, stirred, and heated to 40°C. Sodium borohydride (0.47 g, 12.4 mmol) was slowly added to a 0.5% aqueous sodium hydroxide solution (3.6 mL) to prepare a suspension. The suspension was slowly added to the reaction system, stirred at 40°C for 1.5 hours, cooled to room temperature, and water (65 mL) was added, followed by a 4% aqueous sulfuric acid solution (15 mL). The temperature was lowered to 5°C, filtered, and the filter cake was rinsed with water (20 mL) and dried to obtain compound 1-6 (8.7 g). MS m/z: 269.1 [M+H].
反应瓶中加入乙酸乙酯(16mL)、环己烷(4mL)、化合物1-6(3.0g,11.2mmol)和化合物1-7(4.5g,13.4mmol),室温搅拌,加入丙基磷酸酐50%乙酸乙酯溶液(7.8g,12.3mmol),然后加入甲磺酸(1.5g,16.1mmol),加热至60℃搅拌反应24小时。降至室温后加入四氢呋喃(20mL)和水(12mL)然后缓慢加入24%氢氧化钠水溶液(12mL)萃取分相,有机相浓缩,用乙酸乙酯和正己烷混合溶剂柱层析分离得到化合物1-8(5.2g)。MS m/z:572.2[M+H]。Ethyl acetate (16 mL), cyclohexane (4 mL), compound 1-6 (3.0 g, 11.2 mmol) and compound 1-7 (4.5 g, 13.4 mmol) were added to the reaction flask and stirred at room temperature. A 50% ethyl acetate solution of propylphosphonic anhydride (7.8 g, 12.3 mmol) was added, followed by methanesulfonic acid (1.5 g, 16.1 mmol). The mixture was heated to 60°C and stirred for 24 hours. After cooling to room temperature, tetrahydrofuran (20 mL) and water (12 mL) were added, followed by slow addition of 24% aqueous sodium hydroxide solution (12 mL) for extraction and separation. The organic phase was concentrated and separated by column chromatography using a mixed solvent of ethyl acetate and n-hexane to give compound 1-8 (5.2 g). MS m/z: 572.2 [M+H].
方法二:将甲苯(150mL)、化合物1-1(15g)、氘代苯硫酚(10g)和D-樟脑磺酸(2.8g)加入到圆底烧瓶中,60℃搅拌反应4小时,然后降温至5℃。加入2mol/L氢氧化钠水溶液(30mL),搅拌恢复至室温,加入甲苯(30mL)萃取分相,有机相用2mol/L氢氧化钠水溶液(30mL)和水(30mL)各洗涤一次,收集有机相,减压浓缩后,用乙酸乙酯和正己烷混合溶剂柱层析分离得到化合物1-3(19.7g)。MS m/z:284.1[M+H]。Method 2: Toluene (150 mL), compound 1-1 (15 g), deuterated thiophenol (10 g), and D-camphorsulfonic acid (2.8 g) were added to a round-bottom flask, stirred at 60°C for 4 hours, and then cooled to 5°C. 2 mol/L sodium hydroxide aqueous solution (30 mL) was added, stirred and returned to room temperature, and toluene (30 mL) was added for phase separation. The organic phase was washed once with 2 mol/L sodium hydroxide aqueous solution (30 mL) and once with water (30 mL). The organic phase was collected, concentrated under reduced pressure, and separated by column chromatography using a mixed solvent of ethyl acetate and n-hexane to obtain compound 1-3 (19.7 g). MS m/z: 284.1 [M+H].
将氯化铝(23.7g)和甲苯(100mL)加入到反应瓶中,搅拌并降温至0℃,滴加1,1,3,3-四甲基二硅氧烷(23.9g)的甲苯(40mL)溶液,加毕后升温至25℃缓慢加入化合物1-3(18g)的甲苯(60mL)溶液,25℃搅拌反应2.5小时。加入15%硫酸水溶液(140mL)萃取分相,有机相用水(80mL)洗涤两次,减压浓缩。加入正庚烷(80mL)0℃打浆,过滤,滤饼用正庚烷淋洗,干燥,得到化合物1-4(14g)。MS m/z:286.1[M+H]。Aluminum chloride (23.7 g) and toluene (100 mL) were added to a reaction flask, stirred and cooled to 0°C. A solution of 1,1,3,3-tetramethyldisiloxane (23.9 g) in toluene (40 mL) was added dropwise. After the addition was completed, the temperature was raised to 25°C and a solution of compound 1-3 (18 g) in toluene (60 mL) was slowly added. The mixture was stirred at 25°C for 2.5 hours. 15% aqueous sulfuric acid solution (140 mL) was added for extraction and phase separation. The organic phase was washed twice with water (80 mL) and concentrated under reduced pressure. n-heptane (80 mL) was added and the mixture was slurried at 0°C. The mixture was filtered, the filter cake was rinsed with n-heptane, and dried to obtain compound 1-4 (14 g). MS m/z: 286.1 [M+H].
反应瓶中加入多聚磷酸(60g)和环丁砜(120g),搅拌升温至80℃,加入化合物1-4(12g),升温至120℃搅拌反应3小时,降温至80℃,缓慢加水(30mL),降温至30℃,加水(120mL)和乙酸乙酯(120mL)萃取分相,有机相用10%碳酸氢钠水溶液(35mL)洗涤,有机相减压浓缩。加入正庚烷(60mL)室温打浆,过滤,滤饼用正庚烷淋洗,干燥,得到化合物1-5(10.6g)。MS m/z:267.1[M+H]。Polyphosphoric acid (60 g) and sulfolane (120 g) were added to the reaction flask, stirred and heated to 80°C, compound 1-4 (12 g) was added, the temperature was raised to 120°C and stirred for reaction for 3 hours, the temperature was lowered to 80°C, water (30 mL) was slowly added, the temperature was lowered to 30°C, water (120 mL) and ethyl acetate (120 mL) were added for phase separation, the organic phase was washed with 10% aqueous sodium bicarbonate solution (35 mL), and the organic phase was concentrated under reduced pressure. n-heptane (60 mL) was added and the mixture was slurried at room temperature, filtered, the filter cake was rinsed with n-heptane, and dried to obtain compound 1-5 (10.6 g). MS m/z: 267.1 [M+H].
在反应瓶中加入异丙醇(40mL)、水(4.5mL)和化合物1-5(9.2g),搅拌升温至40℃,缓慢加入硼氢化钠(0.47g)至0.5%氢氧化钠水溶液中(3.6mL)制备为悬浮液,将悬浮液缓慢加入至反应体系中,40℃搅拌反应1.5小时,降至室温,加入水(65mL),然后加入4%硫酸水溶液(15mL),降温至5℃,过滤,滤饼用水(20mL)淋洗,干燥,得到化合物1-6(8.7g)。MS m/z:269.1[M+H]。Isopropanol (40 mL), water (4.5 mL), and compound 1-5 (9.2 g) were added to a reaction flask, stirred and heated to 40°C. Sodium borohydride (0.47 g) was slowly added to a 0.5% aqueous sodium hydroxide solution (3.6 mL) to prepare a suspension. The suspension was slowly added to the reaction system, stirred at 40°C for 1.5 hours, cooled to room temperature, added with water (65 mL), and then added with a 4% aqueous sulfuric acid solution (15 mL). The temperature was cooled to 5°C, filtered, and the filter cake was rinsed with water (20 mL) and dried to obtain compound 1-6 (8.7 g). MS m/z: 269.1 [M+H].
反应瓶中加入乙酸乙酯(16mL)、环己烷(4mL)、化合物1-6(4.0g)和化合物1-7(7.35g),室温搅拌,加入丙基磷酸酐50%乙酸乙酯溶液(19g),然后加入甲磺酸(2.9g),加热至60℃搅拌反应24小时。降至室温后加入四氢呋喃(20mL)和水(12mL)然后缓慢加入24%氢氧化钠水溶液(12mL)萃取分相,有机相浓缩,用乙酸乙酯和正己烷混合溶剂柱层析分离得到化合物1-8(7.2g)。MS m/z:572.2[M+H]。Ethyl acetate (16 mL), cyclohexane (4 mL), compound 1-6 (4.0 g) and compound 1-7 (7.35 g) were added to the reaction flask and stirred at room temperature. A 50% ethyl acetate solution (19 g) of propylphosphoric anhydride was added, followed by methanesulfonic acid (2.9 g). The mixture was heated to 60°C and stirred for 24 hours. After cooling to room temperature, tetrahydrofuran (20 mL) and water (12 mL) were added, followed by slow addition of 24% aqueous sodium hydroxide solution (12 mL) for extraction and separation. The organic phase was concentrated and separated by column chromatography using a mixed solvent of ethyl acetate and n-hexane to give compound 1-8 (7.2 g). MS m/z: 572.2 [M+H].
制备例2:中间体1-9的制备
Preparation Example 2: Preparation of Intermediate 1-9
反应瓶中加入二氯甲烷(20mL)、化合物1-8(2.0g)、S-1,1'-联-2-萘酚(0.2g)、钛酸四异丙酯(0.2g)、水(0.14g),室温搅拌2小时。控温不超过40℃滴加过氧化叔丁醇70%水溶液(0.6g),滴毕室温反应2h。反应液浓缩,用乙酸乙酯和正己烷混合溶剂柱层析分离得到化合物1-9(1.6g)。MS m/z:588.2[M+H]。To the reaction flask, dichloromethane (20 mL), compound 1-8 (2.0 g), S-1,1'-bi-2-naphthol (0.2 g), tetraisopropyl titanate (0.2 g), and water (0.14 g) were added and stirred at room temperature for 2 hours. A 70% aqueous solution of tert-butyl peroxide (0.6 g) was added dropwise, maintaining the temperature at 40°C. The mixture was allowed to react at room temperature for 2 hours. The reaction solution was concentrated and separated by column chromatography using a mixed solvent of ethyl acetate and n-hexane to afford compound 1-9 (1.6 g). MS m/z: 588.2 [M+H].
制备例3:中间体1-10的制备
Preparation Example 3: Preparation of Intermediate 1-10
反应瓶中加入二氯甲烷(20mL)、化合物1-8(2.0g)、R-1,1'-联-2-萘酚(0.2g)、钛酸四异丙酯(0.2g)、水(0.14g),室温搅拌2小时。控温不超过40℃滴加过氧化叔丁醇70%水溶液(0.6g),滴毕室温反应2h。反应液浓缩,用乙酸乙酯和正己烷混合溶剂柱层析分离得到化合物1-10(1.6g)。MS m/z:588.2[M+H]。To the reaction flask, dichloromethane (20 mL), compound 1-8 (2.0 g), R-1,1'-bi-2-naphthol (0.2 g), tetraisopropyl titanate (0.2 g), and water (0.14 g) were added and stirred at room temperature for 2 hours. A 70% aqueous solution of tert-butyl peroxide (0.6 g) was added dropwise, maintaining the temperature at 40°C. The mixture was allowed to react at room temperature for 2 hours. The reaction solution was concentrated and separated by column chromatography using a mixed solvent of ethyl acetate and n-hexane to afford compound 1-10 (1.6 g). MS m/z: 588.2 [M+H].
制备例4:中间体2-6的制备
Preparation Example 4: Preparation of Intermediate 2-6
方法一:将甲苯(36mL)、化合物1-1(5.0g)、苯硫酚(7.2g)和D-樟脑磺酸(2.1g)加入到圆底烧瓶中,60℃搅拌反应4小时,然后降温至5℃。加入2mol/L氢氧化钠溶液(18mL),搅拌恢复至室温,加入甲苯(20mL)萃取分相,有机相用2mol/L氢氧化钠(20mL)和水(20mL)各洗绦一次,减压浓缩后得到化合物2-1(7.8g)。MS m/z:279.0[M+H]。Method 1: Toluene (36 mL), compound 1-1 (5.0 g), thiophenol (7.2 g), and D-camphorsulfonic acid (2.1 g) were added to a round-bottom flask and stirred at 60°C for 4 hours. The temperature was then lowered to 5°C. 2 mol/L sodium hydroxide solution (18 mL) was added and the mixture was stirred and returned to room temperature. Toluene (20 mL) was added for extraction and phase separation. The organic phase was washed once with 2 mol/L sodium hydroxide (20 mL) and once with water (20 mL). The mixture was concentrated under reduced pressure to obtain compound 2-1 (7.8 g). MS m/z: 279.0 [M+H].
将氯化铝(10.0g)和甲苯(50mL)加入到反应瓶中,搅拌并降温至0℃,滴加1,1,3,3-四甲基二硅氧烷(10.0g)的甲苯(20mL)溶液,加毕后升温至25℃,缓慢加入化合物2-1的甲苯溶液,25℃搅拌反应2.5小时。加入15%硫酸水溶液(70mL)萃取分相,有机相用水(40mL)洗涤两次,减压浓缩。加入正庚烷(80mL)0℃打浆,过滤,滤饼用正庚烷淋洗,干燥,得到化合物2-2(13.0g)。MS m/z:281.0[M+H]。Aluminum chloride (10.0 g) and toluene (50 mL) were added to a reaction flask, stirred and cooled to 0°C. A solution of 1,1,3,3-tetramethyldisiloxane (10.0 g) in toluene (20 mL) was added dropwise. After the addition was completed, the temperature was raised to 25°C and the toluene solution of compound 2-1 was slowly added. The mixture was stirred at 25°C for 2.5 hours. 15% aqueous sulfuric acid solution (70 mL) was added to extract the phases. The organic phase was washed twice with water (40 mL) and concentrated under reduced pressure. n-heptane (80 mL) was added and the mixture was slurried at 0°C. The mixture was filtered, the filter cake was rinsed with n-heptane, and dried to obtain compound 2-2 (13.0 g). MS m/z: 281.0 [M+H].
反应瓶中加入多聚磷酸(45g),搅拌升温至80℃,加入化合物2-2(9.0g),升温至120℃搅拌反应3小时,降温至80℃,缓慢加水(25mL),降温至30℃,加水(90mL)和乙酸乙酯(90mL)萃取分相,有机相用10%碳酸氢钠水溶液(30mL)洗涤,有机相减压浓缩。加入正庚烷(50mL)室温打浆,过滤,滤饼用正庚烷淋洗,干燥,得到化合物2-3(7.5g)。MS m/z:263.0[M+H]。Polyphosphoric acid (45 g) was added to the reaction flask, stirred and heated to 80°C, compound 2-2 (9.0 g) was added, the temperature was raised to 120°C and stirred for 3 hours, the temperature was lowered to 80°C, water (25 mL) was slowly added, the temperature was lowered to 30°C, water (90 mL) and ethyl acetate (90 mL) were added for phase separation, the organic phase was washed with 10% aqueous sodium bicarbonate solution (30 mL), and the organic phase was concentrated under reduced pressure. n-heptane (50 mL) was added and the mixture was slurried at room temperature, filtered, the filter cake was rinsed with n-heptane, and dried to obtain compound 2-3 (7.5 g). MS m/z: 263.0 [M+H].
在反应瓶中加入异丙醇(25mL)、水(3mL)和化合物2-3(6.0g),搅拌升温至40℃,缓慢加入硼氢化钠(0.3g)至0.5%氢氧化钠水溶液中(2.0mL)制备为悬浮液,将悬浮液缓慢加入至反应体系中,40℃搅拌反应1.5小时,降至室温,加入水(45mL),然后加入4%硫酸水溶液(10mL),降温至5℃,过滤,滤饼用水(15mL)淋洗,干燥,得到化合物2-4(10.0g)。MS m/z:265.0[M+H]。Isopropanol (25 mL), water (3 mL), and compound 2-3 (6.0 g) were added to a reaction flask, stirred, and heated to 40°C. Sodium borohydride (0.3 g) was slowly added to a 0.5% aqueous sodium hydroxide solution (2.0 mL) to prepare a suspension. The suspension was slowly added to the reaction system, stirred at 40°C for 1.5 hours, cooled to room temperature, and water (45 mL) was added, followed by a 4% aqueous sulfuric acid solution (10 mL). The temperature was lowered to 5°C, filtered, and the filter cake was rinsed with water (15 mL) and dried to obtain compound 2-4 (10.0 g). MS m/z: 265.0 [M+H].
反应瓶中加入乙酸乙酯(10mL)、环己烷(2mL)、化合物2-4(2.0g)和化合物1-7(3.0g),室温搅拌,加入丙基磷酸酐50%乙酸乙酯溶液(5.1g),然后加入甲磺酸(1.0g),加热至60℃搅拌反应24小时。降至室温后加入四氢呋喃(15mL)和水(10mL)然后缓慢加入24%氢氧化钠水溶液(10mL)萃取分相,有机相浓缩,用乙酸乙酯和正己烷混合溶剂柱层析分离得到化合物2-5(3.5g)。MS m/z:568.2[M+H]。Ethyl acetate (10 mL), cyclohexane (2 mL), compound 2-4 (2.0 g) and compound 1-7 (3.0 g) were added to the reaction flask and stirred at room temperature. A 50% ethyl acetate solution of propylphosphoric anhydride (5.1 g) was added, followed by methanesulfonic acid (1.0 g). The mixture was heated to 60°C and stirred for 24 hours. After cooling to room temperature, tetrahydrofuran (15 mL) and water (10 mL) were added, followed by slow addition of 24% aqueous sodium hydroxide solution (10 mL) for extraction and separation. The organic phase was concentrated and separated by column chromatography using a mixed solvent of ethyl acetate and n-hexane to give compound 2-5 (3.5 g). MS m/z: 568.2 [M+H].
反应瓶中加入二氯甲烷(20mL)、化合物2-5(2.0g)、S-1,1'-联-2-萘酚(0.2g)、钛酸四异丙酯(0.2g)、水(0.14g),室温搅拌2小时。控温不超过40℃滴加过氧化叔丁醇70%水溶液(0.6g),滴毕室温反应2h。反应液浓缩,用乙酸乙酯和正己烷混合溶剂柱层析分离得到化合物2-6(1.6g)。MS m/z:584.2[M+H]。To the reaction flask, dichloromethane (20 mL), compound 2-5 (2.0 g), S-1,1'-bi-2-naphthol (0.2 g), tetraisopropyl titanate (0.2 g), and water (0.14 g) were added and stirred at room temperature for 2 hours. A 70% aqueous solution of tert-butyl peroxide (0.6 g) was added dropwise, maintaining the temperature at 40°C. The mixture was allowed to react at room temperature for 2 hours. The reaction solution was concentrated and separated by column chromatography using a mixed solvent of ethyl acetate and n-hexane to afford compound 2-6 (1.6 g). MS m/z: 584.2 [M+H].
方法二:将甲苯(100mL)、化合物1-1(15.0g)、苯硫酚(21.6g)和D-樟脑磺酸(6.3g)加入到圆底烧瓶中,60℃搅拌反应4小时,然后降温至5℃。加入2mol/L氢氧化钠溶液(54mL),搅拌恢复至室温,加入甲苯(60mL)萃取分相,有机相用2mol/L氢氧化钠(60mL)和水(60mL)各洗涤一次,减压浓缩后得到化合物2-1(20.4g)。MS m/z:279.0[M+H]。Method 2: Toluene (100 mL), compound 1-1 (15.0 g), thiophenol (21.6 g), and D-camphorsulfonic acid (6.3 g) were added to a round-bottom flask and stirred at 60°C for 4 hours, then cooled to 5°C. 2 mol/L sodium hydroxide solution (54 mL) was added, and the mixture was stirred and returned to room temperature. Toluene (60 mL) was added for extraction and phase separation. The organic phase was washed once with 2 mol/L sodium hydroxide (60 mL) and once with water (60 mL). After concentration under reduced pressure, compound 2-1 (20.4 g) was obtained. MS m/z: 279.0 [M+H].
将氯化铝(30.0g)和甲苯(150mL)加入到反应瓶中,搅拌并降温至0℃,滴加1,1,3,3-四甲基二硅氧烷(30.0g)的甲苯(60mL)溶液,加毕后升温至25℃,缓慢加入化合物2-1(22.8g)的甲苯(90mL)溶液,25℃搅拌反应2.5小时。加入15%硫酸水溶液(210mL)萃取分相,有机相用水(120mL)洗涤两次,减压浓缩。加入正庚烷(240mL)0℃打浆,过滤,滤饼用正庚烷淋洗,干燥,得到化合物2-2(18.0g)。MS m/z:281.0[M+H]。Aluminum chloride (30.0 g) and toluene (150 mL) were added to a reaction flask, stirred and cooled to 0°C. A solution of 1,1,3,3-tetramethyldisiloxane (30.0 g) in toluene (60 mL) was added dropwise. After the addition was completed, the temperature was raised to 25°C. A solution of compound 2-1 (22.8 g) in toluene (90 mL) was slowly added. The mixture was stirred at 25°C for 2.5 hours. 15% aqueous sulfuric acid solution (210 mL) was added for extraction and phase separation. The organic phase was washed twice with water (120 mL) and concentrated under reduced pressure. n-heptane (240 mL) was added and the mixture was slurried at 0°C. The mixture was filtered, the filter cake was rinsed with n-heptane, and dried to obtain compound 2-2 (18.0 g). MS m/z: 281.0 [M+H].
反应瓶中加入多聚磷酸(45g)和环丁砜(70mL),搅拌升温至80℃,加入化合物2-2(9.0g),升温至120℃搅拌反应3小时,降温至80℃,缓慢加水(25mL),降温至30℃,加水(90mL)和乙酸乙酯(90mL)萃取分相,有机相用10%碳酸氢钠水溶液(30mL)洗涤,有机相减压浓缩。加入正庚烷(50mL)室温打浆,过滤,滤饼用正庚烷淋洗,干燥,得到化合物2-3(7.5g)。MS m/z:263.0[M+H]。Polyphosphoric acid (45 g) and sulfolane (70 mL) were added to the reaction flask, stirred and heated to 80°C, compound 2-2 (9.0 g) was added, the temperature was raised to 120°C and stirred for 3 hours, the temperature was lowered to 80°C, water (25 mL) was slowly added, the temperature was lowered to 30°C, water (90 mL) and ethyl acetate (90 mL) were added for phase separation, the organic phase was washed with 10% aqueous sodium bicarbonate solution (30 mL), and the organic phase was concentrated under reduced pressure. n-heptane (50 mL) was added and the mixture was slurried at room temperature, filtered, the filter cake was rinsed with n-heptane, and dried to obtain compound 2-3 (7.5 g). MS m/z: 263.0 [M+H].
在反应瓶中加入异丙醇(25mL)、水(3mL)和化合物2-3(6.0g),搅拌升温至40℃,缓慢加入硼氢化钠(0.3g)至0.5%氢氧化钠水溶液中(2.0mL)制备为悬浮液,将悬浮液缓慢加入至反应体系中,40℃搅拌反应1.5小时,降至室温,加入水(45mL),然后加入4%硫酸水溶液(10mL),降温至5℃,过滤,滤饼用水(15mL)淋洗,干燥,得到化合物2-4(4.0g)。MS m/z:265.0[M+H]。Isopropanol (25 mL), water (3 mL), and compound 2-3 (6.0 g) were added to a reaction flask, stirred, and heated to 40°C. Sodium borohydride (0.3 g) was slowly added to a 0.5% aqueous sodium hydroxide solution (2.0 mL) to prepare a suspension. The suspension was slowly added to the reaction system, stirred at 40°C for 1.5 hours, cooled to room temperature, and water (45 mL) was added, followed by a 4% aqueous sulfuric acid solution (10 mL). The temperature was cooled to 5°C, filtered, and the filter cake was rinsed with water (15 mL) and dried to obtain compound 2-4 (4.0 g). MS m/z: 265.0 [M+H].
反应瓶中加入乙酸乙酯(10mL)、环己烷(2mL)、化合物2-4(2.4g)和化合物1-7(3.0g),室温搅拌,加入丙基磷酸酐50%乙酸乙酯溶液(7.7g),然后加入甲磺酸(1.0g),加热至60℃搅拌反应24小时。降至室温后加入四氢呋喃(15mL)和水(10mL)然后缓慢加入24%氢氧化钠水溶液(10mL)萃取分相,有机相浓缩,用乙酸乙酯和正己烷混合溶剂柱层析分离得到化合物2-5(4.1g)。MS m/z:568.2[M+H]。Ethyl acetate (10 mL), cyclohexane (2 mL), compound 2-4 (2.4 g) and compound 1-7 (3.0 g) were added to the reaction flask and stirred at room temperature. A 50% ethyl acetate solution of propylphosphonic anhydride (7.7 g) was added, followed by methanesulfonic acid (1.0 g). The mixture was heated to 60°C and stirred for 24 hours. After cooling to room temperature, tetrahydrofuran (15 mL) and water (10 mL) were added, followed by slow addition of 24% aqueous sodium hydroxide solution (10 mL) for extraction and separation. The organic phase was concentrated and separated by column chromatography using a mixed solvent of ethyl acetate and n-hexane to give compound 2-5 (4.1 g). MS m/z: 568.2 [M+H].
反应瓶中加入二氯甲烷(20mL)、化合物2-5(2.0g)、S-1,1'-联-2-萘酚(0.2g)、钛酸四异丙酯(0.2g)、水(0.14g),室温搅拌2小时。控温不超过40℃滴加过氧化叔丁醇70%水溶液(0.6g),滴毕室温反应2h。反应液浓缩,用乙酸乙酯和正己烷混合溶剂柱层析分离得到化合物2-6(1.6g)。MS m/z:584.2[M+H]。To the reaction flask, dichloromethane (20 mL), compound 2-5 (2.0 g), S-1,1'-bi-2-naphthol (0.2 g), tetraisopropyl titanate (0.2 g), and water (0.14 g) were added and stirred at room temperature for 2 hours. A 70% aqueous solution of tert-butyl peroxide (0.6 g) was added dropwise, maintaining the temperature at 40°C. The mixture was allowed to react at room temperature for 2 hours. The reaction solution was concentrated and separated by column chromatography using a mixed solvent of ethyl acetate and n-hexane to afford compound 2-6 (1.6 g). MS m/z: 584.2 [M+H].
制备例5、中间体2-7的制备
Preparation Example 5, Preparation of Intermediate 2-7
反应瓶中加入二氯甲烷(20mL)、化合物2-5(2.0g)、R-1,1'-联-2-萘酚(0.2g)、钛酸四异丙酯(0.2g)、水(0.14g),室温搅拌2小时。控温不超过40℃滴加过氧化叔丁醇70%水溶液(0.6g),滴毕室温反应2h。反应液浓缩,用乙酸乙酯和正己烷混合溶剂柱层析分离得到化合物2-7(1.8g)。MS m/z:584.2[M+H]。To the reaction flask, dichloromethane (20 mL), compound 2-5 (2.0 g), R-1,1'-bi-2-naphthol (0.2 g), tetraisopropyl titanate (0.2 g), and water (0.14 g) were added and stirred at room temperature for 2 hours. A 70% aqueous solution of tert-butyl peroxide (0.6 g) was added dropwise, maintaining the temperature at 40°C. The mixture was allowed to react at room temperature for 2 hours. The reaction solution was concentrated and separated by column chromatography using a mixed solvent of ethyl acetate and n-hexane to afford compound 2-7 (1.8 g). MS m/z: 584.2 [M+H].
实施例1:化合物6的制备Example 1: Preparation of Compound 6
反应瓶中加入N-甲基吡咯烷酮(5mL)、中间体化合物1-8(1g),氯化锂(0.57g),升温至75℃,加入甲磺酸(70mg),75℃保温反应20h。降温至45℃,加入乙腈(1.5mL)和水1(10mL),45℃保温搅拌1小时,降温至15℃,保温搅拌1小时,过滤。滤饼用异丙醇(10mL)室温打浆2小时,过滤,滤饼用异丙醇(5mL)淋洗,干燥,得到化合物6(0.7g)。MS m/z:488.1[M+H]。1H NMR(600MHz,CDCl3)δ11.52(s,1H),7.10(dd,J=16.8,8.5Hz,1H),7.06–6.97(m,2H),5.77(d,J=7.7Hz,1H),5.29 -2.27(m,2H),4.67(d,J=13.5Hz,1H),4.58(dd,J=10.0,2.8Hz,1H),4.07(d,J=13.9Hz,1H),3.96(dd,J=11.1,2.8Hz,1H),3.81(dd,J=12.0,3.2Hz,1H),3.62(t,J=10.6Hz,1H),3.47(td,J=11.8,2.1Hz,1H),3.06–2.95(m,1H).。N-methylpyrrolidone (5 mL), intermediate compound 1-8 (1 g), and lithium chloride (0.57 g) were added to a reaction flask. The temperature was raised to 75°C, and methanesulfonic acid (70 mg) was added. The reaction was maintained at 75°C for 20 h. The temperature was lowered to 45°C, and acetonitrile (1.5 mL) and water 1 (10 mL) were added. The mixture was stirred at 45°C for 1 hour, then lowered to 15°C, stirred at this temperature for 1 hour, and filtered. The filter cake was slurried with isopropanol (10 mL) at room temperature for 2 hours, filtered, rinsed with isopropanol (5 mL), and dried to yield compound 6 (0.7 g). MS m/z: 488.1 [M+H]. 1 H NMR (600MHz, CDCl 3 ) δ11.52(s,1H),7.10(dd,J=16.8,8.5Hz,1H),7.06–6.97(m,2H),5.77(d,J=7.7Hz,1H),5.29 -2.27(m,2H),4.67(d,J=13.5Hz,1H),4.58(dd,J=10.0,2.8Hz,1H),4.07(d,J=13.9Hz,1H),3.96(dd,J=11.1,2. 8Hz,1H),3.81(dd,J=12.0,3.2Hz,1H),3.62(t,J=10.6Hz,1H),3.47(td,J=11.8,2.1Hz,1H),3.06–2.95(m,1H).
实施例2-5:参照实施例1的制备方法制备获得实施例2-5的以下化合物
Example 2-5: The following compounds of Example 2-5 were prepared by referring to the preparation method of Example 1
实施例6:化合物1的制备Example 6: Preparation of Compound 1
反应瓶中加入四氢呋喃(5mL)、N,N-二甲基乙酰胺(0.5mL)、化合物6(0.5g),无水碳酸钾(0.19g),碘化钾(70mg),异丙醇(30mL),纯化水(15mL),搅拌升温至60℃,随后加入氯甲基甲基碳酸酯(0.22g)。60℃下反应10小时。降至室温,加入冰乙酸(0.1g)搅拌15分钟。加入水(2.5mL)搅拌30分钟,萃取分相,有机相浓缩,用乙酸乙酯和正己烷混合溶剂柱层析分离得到化合物1(0.47g)。MS m/z:576.2[M+H]。1H NMR(600MHz,CDCl3)δ7.14(d,J=7.6Hz,1H),7.10(dd,J=16.8,8.4Hz,1H),7.01(dd,J=8.2,4.3Hz,1H),5.93(d,J=7.7Hz,1H),5.90(s,2H),5.33(s,1H),5.27(d,J=14.3Hz,1H),4.64(d,J=13.2Hz,1H),4.52(dd,J=9.9,2.6Hz,1H),4.07(d,J=13.9Hz,1H),3.94(dd,J=10.9,2.4Hz,1H),3.86(s,3H),3.76(dd,J=11.8,2.9Hz,1H),3.56(t,J=10.5Hz,1H),3.44(dd,J=11.7,10.0Hz,1H),2.98–2.90(m,1H)。Tetrahydrofuran (5 mL), N,N-dimethylacetamide (0.5 mL), compound 6 (0.5 g), anhydrous potassium carbonate (0.19 g), potassium iodide (70 mg), isopropyl alcohol (30 mL), and purified water (15 mL) were added to the reaction flask and heated to 60°C with stirring. Chloromethyl methyl carbonate (0.22 g) was then added. The mixture was reacted at 60°C for 10 hours. The mixture was cooled to room temperature, glacial acetic acid (0.1 g) was added, and the mixture was stirred for 15 minutes. Water (2.5 mL) was added and stirred for 30 minutes. The mixture was extracted and separated. The organic phase was concentrated and separated by column chromatography using a mixed solvent of ethyl acetate and n-hexane to obtain compound 1 (0.47 g). MS m/z: 576.2 [M+H]. 1 H NMR (600 MHz, CDCl 3 )δ7.14(d,J=7.6Hz,1H),7.10(dd,J=16.8,8.4Hz,1H),7.01(dd,J=8.2,4.3Hz,1H),5.93(d,J= 7.7Hz,1H),5.90(s,2H),5.33(s,1H),5.27(d,J=14.3Hz,1H),4.64(d,J=13.2Hz,1H),4.52(dd, J=9.9,2.6Hz,1H),4.07(d,J=13.9Hz,1H),3.94(dd,J=10.9,2.4Hz,1H),3.86(s,3H),3.76(dd ,J=11.8,2.9Hz,1H),3.56(t,J=10.5Hz,1H),3.44(dd,J=11.7,10.0Hz,1H),2.98–2.90(m,1H).
实施例7-35:参照实施例6的制备方法制备获得实施例7-35的以下化合物
Example 7-35: The following compound of Example 7-35 was prepared by referring to the preparation method of Example 6
生物测试Biological testing
以下试验例进一步描述解释本发明,但这些实施例并非意味着限制本发明的范围。The following test examples further illustrate the present invention, but these examples are not intended to limit the scope of the present invention.
试验例1:大鼠体内药代动力学实验Test Example 1: Pharmacokinetics experiment in rats
1、目的:1. Purpose:
考察本发明化合物经静脉注射和口服给药后的体内药代动力学的特征。The pharmacokinetic characteristics of the compounds of the present invention after intravenous and oral administration were investigated.
2、试剂及试验动物:2. Reagents and experimental animals:
试剂:二甲基亚砜(DMSO)、聚乙二醇400(PEG400)、甲基纤维素(MC)。Reagents: dimethyl sulfoxide (DMSO), polyethylene glycol 400 (PEG400), methylcellulose (MC).
试验动物:SD大鼠,雄性。Experimental animals: SD rats, male.
3、大鼠体内药代动力学对比实验方法3. Comparative Pharmacokinetic Experimental Methods in Rats
(1)药物配制(1) Drug preparation
采用以下配制方式,分别配制对照化合物(玛巴洛沙韦)及本发明化合物1-5和化合物11-35。The control compound (mabaloxavir) and the compounds 1-5 and 11-35 of the present invention were prepared respectively using the following formulations.
静脉给药:给药剂量1mg/kg,给药体积5ml/kg,给药浓度0.2mg/ml;5%DMSO+10%PEG400+85%水溶液;Intravenous administration: Dosage: 1 mg/kg, volume: 5 ml/kg, concentration: 0.2 mg/ml; 5% DMSO + 10% PEG400 + 85% aqueous solution;
口服给药:给药剂量5mg/kg,给药容积10ml/kg,给药浓度0.5mg/ml;0.5%甲基纤维素(MC)的混悬液。Oral administration: Dosage: 5 mg/kg, Dosage volume: 10 ml/kg, Dosage concentration: 0.5 mg/ml; 0.5% methylcellulose (MC) suspension.
(2)给药方案(2) Dosage regimen
健康成年SD大鼠(每组3只动物),禁食过夜(自由饮水)后,分别尾静脉注射(iv 1mg/kg)及灌胃(po 5mg/kg)给药;iv组分别于给药后5min、15min、0.5、1、2、4、8、24h由颈静脉采血0.2mL于EDTA-K2抗凝管中,血样采集后置于湿冰中,1h内(4℃,3500g,离心5min)分离血浆。将血浆装在贴有标记的1.5mL EP管中,保存于-80℃超低温冰箱待测。po.组于给药前及给药后0.25、0.5、1、2、4、6、8、24h由颈静脉采血0.2mL,处理方法同静脉注射给药组。LC/MS/MS法测定血浆中的原形药物浓度,绘制血药浓度-时间曲线,采用WinNonlin 7.2软件计算主要药动学参数。Healthy adult SD rats (3 animals per group) were fasted overnight (with free access to water) and then administered with the drug via tail vein injection (iv 1 mg/kg) or oral gavage (po 5 mg/kg). In the iv group, 0.2 mL of blood was collected from the jugular vein in EDTA-K2 anticoagulant tubes at 5 min, 15 min, 0.5, 1, 2, 4, 8, and 24 h after administration. The blood samples were placed on wet ice and centrifuged within 1 h (4°C, 3500 g, 5 min) to separate the plasma. The plasma was placed in a labeled 1.5 mL EP tube and stored in a -80°C ultra-low temperature freezer for testing. In the po. group, 0.2 mL of blood was collected from the jugular vein before administration and 0.25, 0.5, 1, 2, 4, 6, 8, and 24 h after administration. The treatment method was the same as that of the iv administration group. The concentration of the unchanged drug in plasma was determined by LC/MS/MS, and the blood concentration-time curve was drawn. The main pharmacokinetic parameters were calculated using WinNonlin 7.2 software.
示例性试验结果见下表:Exemplary test results are shown in the table below:
表1大鼠体内药代动力学结果
Table 1 Pharmacokinetic results in rats
实验结果表明,本发明化合物有较好的PK性质和/或口服吸收性质。The experimental results show that the compounds of the present invention have good PK properties and/or oral absorption properties.
试验例2:化合物抑制病毒活性(EC50)和化合物的细胞毒性(CC50)检测1Test Example 2: Detection of virus inhibition activity (EC50) and cytotoxicity (CC50) of compounds 1
细胞铺板:将MDCK细胞以一定密度接种于96孔板内,随后细胞置于37℃,5%CO2培养箱中培养过夜。Cell plating: MDCK cells were seeded at a certain density in a 96-well plate, and then the cells were cultured in a 37°C, 5% CO2 incubator overnight.
化合物处理:待测化合物(对照化合物巴洛沙韦及本发明的化合物6-10)用DMSO梯度稀释后加入细胞培养板中,DMSO终浓度为0.5%。CC50测定稀释倍数:50μM起,3倍稀释,10个浓度;EC50测定稀释倍数:100nM起,3倍稀释,10个浓度。Compound Treatment: Test compounds (control compound baloxavir and compounds 6-10 of the present invention) were serially diluted with DMSO to a final DMSO concentration of 0.5% and added to the cell culture plates. CC50 determinations were performed using a 3-fold dilution from 50 μM for 10 concentrations. EC50 determinations were performed using a 3-fold dilution from 100 nM for 10 concentrations.
病毒接种:在抗病毒活性检测孔内加入稀释好的流感病毒液(A/WSN/33(H1N1)或A/PR/8/34(H1N1));细胞毒性测试孔中仅加化合物,不加病毒。将细胞置于37℃,5%CO2培养箱中培养3~5天,直到病毒对照孔(无化合物)中的细胞病变(CPE)率达到80-95%。Virus Inoculation: Add diluted influenza virus (A/WSN/33 (H1N1) or A/PR/8/34 (H1N1)) to the antiviral activity test wells; add only the compound, without virus, to the cytotoxicity test wells. Incubate the cells in a 37°C, 5% CO2 incubator for 3-5 days until the cytopathic effect (CPE) rate in the virus control wells (no compound) reaches 80-95%.
细胞活性检测:每孔加入细胞活性检测试剂(CCK-8检测试剂),37℃,5%CO2培养箱中培养细胞3-4小时后,酶标仪检测信号值,原始数据用于化合物抗流感病毒活性和细胞毒性计算。Cell viability assay: Cell viability assay reagent (CCK-8 assay reagent) was added to each well. After culturing the cells in a 37°C, 5% CO2 incubator for 3-4 hours, the signal value was detected by a microplate reader. The raw data was used to calculate the anti-influenza virus activity and cytotoxicity of the compound.
GraphPad Prism软件分析,得到化合物剂量反应曲线及其EC50和CC50值,并计算治疗指数(治疗指数=CC50/EC50)。GraphPad Prism software was used for analysis to obtain the compound dose-response curve and its EC50 and CC50 values, and the therapeutic index was calculated (therapeutic index = CC50/EC50).
实验结果表明,本发明化合物有较好的流感病毒抑制活性和较低的细胞毒性。The experimental results show that the compound of the present invention has good influenza virus inhibitory activity and low cytotoxicity.
试验例3:化合物抑制病毒活性(EC50)和化合物的细胞毒性(CC50)检测2Test Example 3: Detection of Virus Inhibitory Activity (EC50) and Cytotoxicity (CC50) of Compounds 2
1、目的:1. Purpose:
基于间接免疫荧光的病毒定量测定(FFA)法,考察本发明化合物体外抗流感病毒活性测定。The in vitro anti-influenza virus activity of the compounds of the present invention was investigated using an indirect immunofluorescence virus quantitative assay (FFA) method.
2、关键试剂,毒株和细胞:2. Key reagents, strains and cells:
关键试剂:Influenza Anti-HA Serum(毒株特异性血清),Alexa Fluor 488驴抗羊IgGKey reagents: Influenza Anti-HA Serum (strain-specific serum), Alexa Fluor 488 donkey anti-sheep IgG
毒株:A型(H1N1)A/Victoria/2570/2019&A/Victoria/4897/2022Strain: Type A (H1N1) A/Victoria/2570/2019 & A/Victoria/4897/2022
A型(H3N2)A/Cambodia/E0826360/2020&A/Darwin/9/2021Type A (H3N2)A/Cambodia/E0826360/2020&A/Darwin/9/2021
细胞铺板:MDCK细胞Cell plating: MDCK cells
3、检测方法:3. Detection method:
细胞准备:观察细胞状态,确保汇合度:85%~95%,活率≥90%,细胞消化接种至96孔细胞培养板中,2.5~3.0×105cells/mL,100μL/孔,置于37℃二氧化碳培养箱培养24h左右;Cell preparation: Observe the cell status and ensure the confluence is 85% to 95% and the viability is ≥90%. Digest the cells and inoculate them into 96-well cell culture plates at a concentration of 2.5 to 3.0 × 10 5 cells/mL, 100 μL/well, and culture in a 37°C CO 2 incubator for approximately 24 hours.
病毒稀释:取病毒液,依据各毒株病毒滴度,以MEM培养基稀释,稀释至8000PFU/mL,作为病毒工作液,置于冰上或2~8℃备用;Virus dilution: Take the virus solution and dilute it with MEM medium according to the virus titer of each strain to 8000 PFU/mL as the virus working solution. Place it on ice or at 2-8°C until ready for use.
化合物稀释:待测化合物(对照化合物1为巴洛沙韦、对照化合物2为CN108440564A中的实施例化合物1A、本发明的化合物)用DMSO梯度稀释后加入细胞培养板中,DMSO终浓度为0.5%。EC50测定稀释倍数:100nM起,3倍稀释,20个浓度;Compound Dilution: Test compounds (control compound 1: baloxavir, control compound 2: Example compound 1A in CN108440564A, and compounds of the present invention) were serially diluted with DMSO to a final DMSO concentration of 0.5% and added to the cell culture plate. EC50 determination dilution factor: 20 concentrations, starting from 100 nM, with 3-fold dilutions.
化合物加药和病毒接种:从37℃二氧化碳培养箱取出提前接种的细胞板,甩干培养上清,加入100μL/孔MEM培养基重复洗2遍,甩干残留的液体。按设计的板布局取化合物梯度稀释液和化合物空白稀释液用多通道移液器按50μL/孔移取待检测液至抗病毒实验板;除CC孔(细胞对照),其余孔每孔加入50μL对应毒株病毒工作液,轻摇混匀实验板,将试验板放入37℃、5%CO2培养箱培养18~24h。。Compound Dosing and Virus Inoculation: Remove the pre-seeded cell plate from the 37°C CO2 incubator, spin dry the culture supernatant, add 100 μL/well MEM medium, and wash twice. Spin dry any remaining liquid. According to the designed plate layout, pipette the compound serial dilutions and compound blank dilutions at a rate of 50 μL/well onto the antiviral assay plate using a multichannel pipette. Except for wells CC (cell control), add 50 μL of the corresponding virus working solution to each well. Gently shake the plate to mix thoroughly. Incubate the plate in a 37°C, 5% CO2 incubator for 18–24 hours.
GraphPad Prism软件分析,得到化合物剂量反应曲线及其EC50值。GraphPad Prism software was used to analyze the compound dose-response curve and its EC50 value.
表2化合物体外抗流感病毒活性
Table 2 In vitro anti-influenza virus activity of compounds
实验结果表明,本发明化合物明显优于对照化合物,有很好的流感病毒抑制活性。The experimental results show that the compound of the present invention is significantly superior to the control compound and has good influenza virus inhibitory activity.
试验例4:小鼠体内药代动力学实验Test Example 4: Pharmacokinetics experiment in mice
1、目的:1. Purpose:
考察本发明化合物经口服给予小鼠后,在肺脏和血浆中的分布水平。The distribution levels of the compounds of the present invention in the lungs and plasma were investigated after oral administration to mice.
2、试剂及试验动物:2. Reagents and experimental animals:
试剂:甲基纤维素(MC);Reagents: methylcellulose (MC);
试验动物:Balb/c小鼠,雄性。Experimental animals: Balb/c mice, male.
3、小鼠体内药代动力学对比实验方法3. Comparative Pharmacokinetic Experimental Methods in Mice
(1)药物配制(1) Drug preparation
口服给药:给药剂量10mg/kg,给药容积为10ml/kg,采用0.5%甲基纤维素(MC)配制成给药浓度1.0mg/ml的均一混悬液。Oral administration: The dosage is 10 mg/kg, the administration volume is 10 ml/kg, and 0.5% methylcellulose (MC) is used to prepare a uniform suspension with a dosage concentration of 1.0 mg/ml.
(2)给药方案(2) Dosage regimen
6-8周Balb/c小鼠(每时间点各3只动物),禁食过夜(自由饮水)后,分别口服给予10mg/kg化合物(如本发明化合物1);各化合物组分别于药后0.5、1、2、4、8h采集血样并立即处死采集肺组织。血样采集至提前加入稳定剂的试管中颠倒混匀,并于30min内离心处理(4℃,3000g,10min),分离血浆后动冻存待测,肺组织采集后立即置于冰水浴中,在20min内称取重量后按照1g:3mL迅速加入冰乙腈,在1h内完成匀浆待测。LC/MS/MS法分别测定血浆中的代谢物化合物(如本发明化合物6),结果如下:6-8 week old Balb/c mice (3 animals at each time point) were fasted overnight (with free access to water) and then orally administered with 10 mg/kg of compound (such as compound 1 of the present invention); blood samples were collected at 0.5, 1, 2, 4, and 8 hours after the drug administration for each compound group, and the mice were immediately killed and lung tissue was collected. The blood samples were collected into test tubes with stabilizers added in advance, mixed by inversion, and centrifuged within 30 minutes (4°C, 3000g, 10 minutes). The plasma was separated and frozen for testing. After the lung tissue was collected, it was immediately placed in an ice water bath, weighed within 20 minutes, and glacial acetonitrile was quickly added according to 1g:3mL. The homogenate was completed within 1 hour for testing. The metabolite compounds (such as compound 6 of the present invention) in the plasma were determined by LC/MS/MS method, and the results are as follows:
实验结果表明,本发明化合物在小鼠血浆和肺脏中的暴露水平以及肺血暴露量的比值均较好。The experimental results show that the exposure levels of the compound of the present invention in mouse plasma and lungs, as well as the ratio of lung blood exposure, are both good.
试验例5:食蟹猴体内药代动力学实验Experimental Example 5: Pharmacokinetic Experiment in Cynomolgus Monkeys
1、目的:1. Purpose:
考察本发明化合物经静脉注射和口服给予食蟹猴后的体内药代动力学的特征。The pharmacokinetic characteristics of the compounds of the present invention were investigated after intravenous and oral administration to cynomolgus monkeys.
2、试剂及试验动物:2. Reagents and experimental animals:
试剂:二甲基亚砜(DMSO)、聚乙二醇400(PEG400)、甲基纤维素(MC)。Reagents: dimethyl sulfoxide (DMSO), polyethylene glycol 400 (PEG400), methylcellulose (MC).
试验动物:食蟹猴,雄性。Experimental animals: Cynomolgus monkeys, male.
3、食蟹猴体内药代动力学对比实验方法3. Comparative Pharmacokinetic Experimental Methods in Cynomolgus Monkeys
(1)药物配制(1) Drug preparation
静脉给药:给药剂量1mg/kg,给药容为2ml/kg,给药浓度0.5mg/ml;5%DMSO+30%PEG400+65%水溶液;Intravenous administration: Dosage: 1 mg/kg, dosing volume: 2 ml/kg, dosing concentration: 0.5 mg/ml; 5% DMSO + 30% PEG400 + 65% aqueous solution;
口服给药:给药剂量10mg/kg,给药容积为2ml/kg,给药浓度0.5mg/ml;0.5%甲基纤维素(MC)的混悬液。Oral administration: Dosage: 10 mg/kg, administration volume: 2 ml/kg, administration concentration: 0.5 mg/ml; 0.5% methylcellulose (MC) suspension.
(2)给药方案(2) Dosage regimen
健康洗脱食蟹猴(口服组每组4只,静脉组每组2只),禁食过夜(自由饮水)后,分别尾静脉注射(iv 1mg/kg)及口服(po 1mg/kg)给药;iv组分别于给药后2min、5min、0.25、0.5、1、2、4、8、24、48h由四肢静脉采血0.3mL于EDTA-K2抗凝管中(提前加入5μLPMSF溶液,PMSF溶液浓度为300mM,甲醇配制),轻轻震荡混匀多次,置于冰水浴中,于30min内离心处理(4℃,3000g,10min),分离血浆,尽快于-70℃冰箱中冷冻,待测。po.组于给药前及给药后0.25、0.5、1、2、4、8、24、48h采集血样,处理方法同静脉注射给药组。LC/MS/MS法分别测定血浆中的代谢物(如发明化合物的代谢物化合物6),绘制血药浓度-时间曲线,采用WinNonlin 8.0软件计算主要药动学参数。Healthy eluted cynomolgus monkeys (n=4 per oral group and n=2 per intravenous group) were fasted overnight (with free access to water) and then administered with either tail vein injection (1 mg/kg) or oral administration (1 mg/kg). Blood samples (0.3 mL) were collected from all four limb veins in the iv group at 2, 5, 0.25, 0.5, 1, 2, 4, 8, 24, and 48 h after administration. The samples were placed in EDTA-K2 anticoagulant tubes (pre-added with 5 μL PMSF solution (300 mM in methanol)). The tubes were gently shaken several times, placed in an ice-water bath, and centrifuged within 30 min (3000 g, 4°C, 10 min). Plasma was separated and frozen at -70°C as soon as possible for analysis. Blood samples were collected from the po group before administration and at 0.25, 0.5, 1, 2, 4, 8, 24, and 48 h after administration and processed as described for the intravenous group. The metabolites in plasma (such as compound 6, the metabolite of the invention compound) were determined by LC/MS/MS, and the blood drug concentration-time curve was drawn. The main pharmacokinetic parameters were calculated using WinNonlin 8.0 software.
实验结果表明,本发明化合物口服给药后具有较好的PK性质/口服吸收性质。The experimental results show that the compound of the present invention has good PK properties and oral absorption properties after oral administration.
试验例6:化合物在人的体外肝S9的稳定性试验Test Example 6: Stability test of compound in human liver S9 in vitro
供试品:1μM的阳参玛巴洛沙韦及本发明化合物1Test samples: 1 μM baloxavir and compound 1 of the present invention
试剂:磷酸盐缓冲液:200mM,200μL,最终浓度100mM;超纯水:138μL,MgCl2溶液:50mM,40μL,最终浓度5mM;S9组分:20mg/mL,20μL,最终浓度1mg/mLReagents: Phosphate buffer: 200 mM, 200 μL, final concentration 100 mM; Ultrapure water: 138 μL, MgCl2 solution: 50 mM, 40 μL, final concentration 5 mM; S9 component: 20 mg/mL, 20 μL, final concentration 1 mg/mL
操作步骤:在上述试剂系统中加入2μL的200μM测试化合物或对照化合物溶液。维拉帕米作为阳性对照,测试化合物和对照化合物的最终浓度为1μM;其后在0、5、15、30、45和60分钟后,将反应溶液在旋涡混合器上混合10秒,然后转移50μL到含有400μL乙腈中,密封后置于冰上,继续混合10分钟后,于4℃、3,220g离心50分钟后,采用LC-MS/MS分析玛巴洛沙韦和化合物1的浓度,所测得浓度使用Microsoft Excel计算出各时间点的剩余百分比,并根据斜率计算出半衰期T1/2和体外固有清除率CLint。Procedure: Add 2 μL of a 200 μM test compound or control compound solution to the above reagent system. Verapamil serves as a positive control, and the final concentration of the test and control compounds is 1 μM. After 0, 5, 15, 30, 45, and 60 minutes, the reaction solution is mixed on a vortex mixer for 10 seconds. Then, 50 μL is transferred to 400 μL of acetonitrile, sealed, and placed on ice. Mixing is continued for 10 minutes. After centrifugation at 3,220 g for 50 minutes at 4°C, mabaloxavir and compound 1 concentrations are analyzed by LC-MS/MS. The measured concentrations are used to calculate the percentage remaining at each time point using Microsoft Excel. The slopes are then used to calculate the half-life (T 1/2) and in vitro intrinsic clearance (CL int ) .
表3化合物人肝S9的体外稳定性结果
Table 3 In vitro stability results of compound human liver S9
实验结果表明,本发明化合物在人肝S9中的代谢快于阳参,更容易释放出原药分子。The experimental results show that the compound of the present invention is metabolized faster in human liver S9 than Yangshen, and is more likely to release the original drug molecule.
试验例7:化合物在人的体外肠S9的稳定性试验Test Example 7: Stability test of compound in human intestinal S9 in vitro
供试品:1μM的阳参玛巴洛沙韦及本发明化合物1Test samples: 1 μM baloxavir and compound 1 of the present invention
试剂:磷酸盐缓冲液:200mM,200μL,最终浓度100mM;超纯水:138μL,MgCl2溶液:50mM,40μL,最终浓度5mM;S9组分:20mg/mL,20μL,最终浓度1mg/mLReagents: Phosphate buffer: 200 mM, 200 μL, final concentration 100 mM; Ultrapure water: 138 μL, MgCl2 solution: 50 mM, 40 μL, final concentration 5 mM; S9 component: 20 mg/mL, 20 μL, final concentration 1 mg/mL
操作步骤:在上述试剂系统中加入2μL的200μM测试化合物或对照化合物溶液。维拉帕米作为阳性对照,测试化合物和对照化合物的最终浓度为1μM;其后在0、5、15、30、45和60分钟后,将反应溶液在旋涡混合器上混合10秒,然后转移50μL到含有400μL乙腈中,密封后置于冰上,继续混合10分钟后,于4℃、3,220g离心50分钟后,采用LC-MS/MS分析玛巴洛沙韦和化合物1的浓度,所测得浓度使用Microsoft Excel计算出各时间点的剩余百分比,并根据斜率计算出半衰期T1/2和体外固有清除率CLint。Procedure: Add 2 μL of a 200 μM test compound or control compound solution to the above reagent system. Verapamil serves as a positive control, and the final concentration of the test and control compounds is 1 μM. After 0, 5, 15, 30, 45, and 60 minutes, the reaction solution is mixed on a vortex mixer for 10 seconds. Then, 50 μL is transferred to 400 μL of acetonitrile, sealed, and placed on ice. Mixing is continued for 10 minutes. After centrifugation at 3,220 g for 50 minutes at 4°C, mabaloxavir and compound 1 concentrations are analyzed by LC-MS/MS. The measured concentrations are used to calculate the percentage remaining at each time point using Microsoft Excel. The slopes are then used to calculate the half-life (T 1/2) and in vitro intrinsic clearance (CL int ) .
表4化合物人肠S9的体外稳定性结果
Table 4 In vitro stability results of compound human intestinal S9
实验结果表明,本发明化合物在人肠S9中的代谢快于阳参化合物,更容易释放出原药分子。The experimental results show that the compounds of the present invention are metabolized faster in human intestinal S9 than the Yangshen compounds and are more likely to release the original drug molecules.
试验例8:化合物在人的血浆稳定性试验Test Example 8: Stability test of compound in human plasma
供试品:阳参玛巴洛沙韦及本发明化合物1Test samples: Yangshenmabaloxavir and compound 1 of the present invention
操作步骤:在人血浆中加入50μL的400ng/mL测试化合物或对照化合物溶液。测试化合物和对照化合物的最终浓度为20ng/mL;其后在37℃于0、5、15、30和60分钟后,将稳定性血浆混合后,然后转移30μL到含有270μL乙腈中,混合后迅速于4℃、10000g离心5分钟后,采用LC-MS/MS分析玛巴洛沙韦和化合物1的浓度,所测得浓度使用Microsoft Excel计算出各时间点的剩余百分比。Procedure: 50 μL of a 400 ng/mL solution of test compound or control compound was added to human plasma. The final concentration of the test compound and control compound was 20 ng/mL. After incubation at 37°C for 0, 5, 15, 30, and 60 minutes, the stabilized plasma was pooled and 30 μL was transferred to 270 μL of acetonitrile. After mixing, the mixture was rapidly centrifuged at 10,000 g for 5 minutes at 4°C. The concentrations of mabaloxavir and compound 1 were analyzed by LC-MS/MS. The measured concentrations were used to calculate the percentage remaining at each time point using Microsoft Excel.
表5化合物人血浆中的体外稳定性结果
Table 5 In vitro stability results of compounds in human plasma
实验结果表明,本发明化合物在人血浆中的代谢快于阳参化合物,更容易释放出原药分子。Experimental results show that the compound of the present invention is metabolized faster in human plasma than the Yangshen compound and is more likely to release the original drug molecules.
Claims (10)
The compound represented by formula I, its tautomer, stereoisomer or pharmaceutically acceptable salt thereof:
A compound selected from the group consisting of:
Priority Applications (1)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| CN202580000334.5A CN121127476A (en) | 2024-03-18 | 2025-03-18 | Polycyclic pyridone derivatives and use thereof |
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| CN202410304994 | 2024-03-18 | ||
| CN202410304994.X | 2024-03-18 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025195347A1 true WO2025195347A1 (en) | 2025-09-25 |
Family
ID=97138357
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/CN2025/083049 Pending WO2025195347A1 (en) | 2024-03-18 | 2025-03-18 | Polycyclic pyridone derivative and use thereof |
Country Status (2)
| Country | Link |
|---|---|
| CN (1) | CN121127476A (en) |
| WO (1) | WO2025195347A1 (en) |
-
2025
- 2025-03-18 WO PCT/CN2025/083049 patent/WO2025195347A1/en active Pending
- 2025-03-18 CN CN202580000334.5A patent/CN121127476A/en active Pending
Also Published As
| Publication number | Publication date |
|---|---|
| CN121127476A (en) | 2025-12-12 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| CN114426568B (en) | 2-oxo-3-pyrrolidinyl propionitrile compounds, pharmaceutical composition and application thereof | |
| JP7050751B2 (en) | Compounds and compositions for treating conditions associated with NLRP activity | |
| JP2021191796A (en) | Preparation of azaindole compound | |
| WO2020020267A1 (en) | Substituted polycyclic pyridone compound and prodrug thereof | |
| CN105849100B (en) | Influenza virus replication inhibitor | |
| WO2019196891A1 (en) | Polycyclic carbamoylpyridone derivatives, pharmaceutical compositions and use thereof | |
| TW202333743A (en) | Crystalline forms of (s)-2-ethylbutyl 2-(((s)-(((2r,3s,4r,5r)-5-(4-aminopyrrolo[2,1-f] [1,2,4]triazin-7-yl)-5-cyano-3,4-dihydroxytetrahydrofuran-2-yl)methoxy)(phenoxy) phosphoryl)amino)propanoate | |
| JP2017128605A (en) | Solid forms of antiviral compound | |
| CN113272298B (en) | Compounds and compositions for treating respiratory diseases | |
| CN112062800B (en) | Phosphoramidate derivatives of nucleoside compounds and uses thereof | |
| WO2020241759A1 (en) | Prophylactic and/or therapeutic agent for influenza virus infection or corona virus infection | |
| TWI844564B (en) | Inhibitors of influenza virus replication | |
| CN111620815B (en) | Chiral chloroquine, hydroxychloroquine and derivatives thereof and their preparation method and use | |
| JP7158808B2 (en) | Treatment of influenza with substituted polycyclic pyridone derivatives and their prodrugs in subjects with influenza and complication risk factors | |
| CN116514902A (en) | Deuterated peptidomimetic compounds and application thereof | |
| WO2025195347A1 (en) | Polycyclic pyridone derivative and use thereof | |
| CN111494380A (en) | Application of DB-1 in preparation of medicine for preventing and treating N L RP3 inflammasome-related diseases and pharmaceutical composition thereof | |
| CN115260100B (en) | A substituted acylguanidine compound for preparing coronavirus therapeutic drugs and its use | |
| CN102532114A (en) | Niacin derivative, preparation method thereof and medicine composition thereof | |
| JP7711088B2 (en) | Compounds for the Treatment of SARS | |
| EP4471017A1 (en) | Cyclic(alkyl)(amino)alkoxy-substituted aryl-pyrone compound and use thereof | |
| CN112638477A (en) | Pyrrolo [2,3-B ] pyridine derivatives as inhibitors of influenza virus replication | |
| CN119019400B (en) | GS-441524 prodrug and its pharmaceutical uses | |
| HK40070765A (en) | 2-oxo-3-pyrrolidinylpropionitrile compounds and their pharmaceutical compositions and uses | |
| CN118786127A (en) | Compounds and methods for treating viral infections |
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: 25773046 Country of ref document: EP Kind code of ref document: A1 |