WO2017148290A1 - Composé d'adénine substituée et composition pharmaceutique correspondante - Google Patents

Composé d'adénine substituée et composition pharmaceutique correspondante Download PDF

Info

Publication number
WO2017148290A1
WO2017148290A1 PCT/CN2017/074135 CN2017074135W WO2017148290A1 WO 2017148290 A1 WO2017148290 A1 WO 2017148290A1 CN 2017074135 W CN2017074135 W CN 2017074135W WO 2017148290 A1 WO2017148290 A1 WO 2017148290A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
mmol
added
reverse transcriptase
nucleoside reverse
Prior art date
Application number
PCT/CN2017/074135
Other languages
English (en)
Chinese (zh)
Inventor
王义汉
赵九洋
Original Assignee
深圳市塔吉瑞生物医药有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 深圳市塔吉瑞生物医药有限公司 filed Critical 深圳市塔吉瑞生物医药有限公司
Priority to CN201780003912.6A priority Critical patent/CN108350007B/zh
Publication of WO2017148290A1 publication Critical patent/WO2017148290A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings

Definitions

  • the invention belongs to the technical field of medicine, and in particular relates to a substituted adenine compound and a pharmaceutical composition thereof, which can be used for treating diseases related to viral infection.
  • Nucleoside reverse transcriptase inhibitors analogs of DNA reverse transcriptase substrate deoxynucleotides that synthesize HIV, are converted in vivo to active nucleoside triphosphate derivatives, and natural deoxynucleotides The glycosidic competition binds to HIV reverse transcriptase (RT), inhibits the action of RT, and blocks the synthesis of provirus. Similar to nucleosides, NRTIs are dideoxynucleoside derivatives that competitively bind to reverse transcriptase with cellular nucleosides, thereby terminating reverse transcription.
  • Nucleotide HIV reverse transcriptase inhibitors act on the active site of the reverse transcriptase binding to its natural substrate nucleoside.
  • These drugs are natural nucleoside drugs that enter the body and undergo multiple steps of phosphorylation to be metabolized into true active molecule triphosphorylated nucleosides (NRTI-ppp), which compete with endogenous dNTPs.
  • NRTI-ppp true active molecule triphosphorylated nucleosides
  • the active site of the substrate of the enzyme Since the structure of NRTI-ppp is very similar to the dNTP substrate, the enzyme mistakes these drugs for substrates and embeds them in the extended DNA strand. Once these drugs enter the DNA strand, there is no structure in the drug molecule. 3'-hydroxyl group attached to the next dNTP 3'-5'. This blocks the prolongation of the viral DNA strand and inhibits HIV replication.
  • Anti-HIV drugs mainly include four categories: nucleoside reverse transcriptase inhibitors (NRTIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs), protease inhibitors (PIs) and HIV integrase inhibitors, of which NRTIs are applications.
  • NRTIs nucleoside reverse transcriptase inhibitors
  • NRTIs non-nucleoside reverse transcriptase inhibitors
  • PIs protease inhibitors
  • HIV integrase inhibitors of which NRTIs are applications.
  • the earliest and most diverse categories include zidovudine (AZT), lamivudine, dehydroxyanhydride, stanfordin, abacavir and tenofovir.
  • AIDS is a serious disease caused by HIV infection. Since the first AIDS case was reported in 1981, nearly 70 million people worldwide have been infected with HIV and more than 20 million have died of AIDS. In the past 20 years, although effective drug treatment has reduced AIDS mortality, millions of people are still infected with HIV every year, and the number of AIDS patients worldwide has been on the rise.
  • HIV is currently resistant to almost all clinically used anti-HIV drugs, and the emergence of drug-resistant HIV is considered to be the main cause of failure of anti-HIV drug treatment.
  • chronic hepatitis is one of the most serious infectious diseases that threaten global human health. About 2 billion people worldwide have been infected with Hepatitis B Virus (HBV), and the number of deaths due to HBV infection is 1 million per year. HBV infection is not only an important biological factor causing chronic hepatitis B, but also causing primary liver cancer. China, Southeast Asia and Africa are high-risk areas of HBV infection, and the incidence of primary liver cancer is significantly higher than that of low-incidence areas of HBV infection in central and southern America.
  • the current treatment of chronic hepatitis B mainly includes interferon, nucleoside drugs, and thymosin, but these drugs may have serious side effects or drug resistance and are expensive. Therefore, finding new and effective anti-HBV drugs is an urgent problem to be solved.
  • the present invention discloses a nucleoside reverse transcriptase inhibitor, a pharmaceutical composition and use thereof, which have better nucleoside reverse transcriptase inhibitory activity and/or have better pharmacodynamics/ Pharmacokinetic properties.
  • a nucleoside reverse transcriptase inhibitor such as an adenine compound substituted by the formula (I), or a crystalline form, a pharmaceutically acceptable salt, a prodrug, a stereoisomer, a hydrate or a solvent compound,
  • R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 18 , R 19 , R 20 , R 21 , R 22 , R 23 , R 24 , R 25 , R 26 are each independently hydrogen, deuterium, halogen or trifluoromethyl;
  • Additional conditions are R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 , R 8 , R 9 , R 10 , R 11 , R 12 , R 13 , R 14 , R 15 , R 16 And at least one of R 17 , R 18 , R 19 , R 20 , R 21 , R 22 , R 23 , R 24 , R 25 and R 26 is deuterated or deuterated.
  • R 1 and R 2 are each independently hydrazine or hydrogen.
  • R 3 , R 4 , R 5 , R 6 , R 7 and R 8 are each independently hydrazine or hydrogen.
  • R 9 and R 10 are each independently hydrazine or hydrogen.
  • R 11 , R 12 , R 13 , R 14 , R 15 , R 16 , R 17 , R 18 , R 19 , R 20 and R 21 are each independently hydrazine or hydrogen.
  • R 23 , R 24 , R 25 and R 26 are each independently hydrazine or hydrogen.
  • the compound may be selected from the following compounds or a pharmaceutically acceptable salt thereof, but is not limited to the following compounds:
  • the shape and volume of the ruthenium in the drug molecule are substantially the same as those of the hydrogen. If the hydrogen in the drug molecule is selectively replaced with hydrazine, the deuterated drug generally retains the original biological activity and selectivity. At the same time, the inventors have confirmed through experiments that the binding of carbon-germanium bonds is more stable than the combination of carbon-hydrogen bonds, which can directly affect the absorption, distribution, metabolism and excretion of some drugs, thereby improving the efficacy, safety and tolerability of the drugs.
  • the strontium isotope content of the cerium in the deuterated position is at least greater than the natural strontium isotope content (0.015%), preferably greater than 30%, more preferably greater than 50%, more preferably greater than 75%, and even more preferably greater than 95. %, more preferably greater than 99%.
  • the osmium isotope content of each of the R 5 , R 16 , R 17 , R 18 , R 19 , R 20 , R 21 , R 22 , R 23 , R 24 , R 25 and R 26 is at least 5%, preferably More than 10%, more preferably more than 15%, more preferably more than 20%, more preferably more than 25%, more preferably more than 30%, more preferably more than 35%, more preferably more than 40%, more preferably More than 45%, more preferably more than 50%, more preferably more than 55%, more preferably more than 60%, more preferably more than 65%, more preferably more than 70%, more preferably more than 75%, more preferably more than 80%, more preferably more than 85%, more preferably more than 90%, more
  • the two Rs contain ruthenium, more preferably three R ⁇ , more preferably four R ⁇ , more preferably five R ⁇ , more preferably six R ⁇ , more preferably seven R ⁇ , preferably eight R ⁇ , more preferably nine R ⁇ , more preferably ten R ⁇ , more preferably eleven R ⁇ , more preferably twelve R ⁇ , More preferably, thirteen R ⁇ , more preferably fourteen R ⁇ , more preferably fifteen R ⁇ , more preferably sixteen R ⁇ , more preferably seventeen R
  • the compound does not include a non-deuterated compound.
  • the present invention also discloses a pharmaceutical composition
  • a pharmaceutical composition comprising a pharmaceutically acceptable carrier and the nucleoside reverse transcriptase inhibitor as described above, or a crystalline form thereof, a pharmaceutically acceptable salt, a hydrate thereof Or a pharmaceutical composition of a solvate, stereoisomer, prodrug or isotopic variation.
  • the pharmaceutically acceptable carrier includes a glidant, a sweetener, a diluent, a preservative, a dye/colorant, a flavor enhancer, a surfactant, a wetting agent, a dispersant At least one of a disintegrant, a suspending agent, a stabilizer, an isotonic agent, a solvent or an emulsifier.
  • the pharmaceutical composition is a tablet, a pill, a capsule, a powder, a granule, an ointment, an emulsion, a suspension, a solution, a suppository, an injection, an inhalant, a gel, a microsphere or Aerosol.
  • Typical routes of administration of the pharmaceutical compositions of the invention include, but are not limited to, oral, rectal, transmucosal, enteral, or topical, transdermal, inhalation, parenteral, sublingual, intravaginal, intranasal, intraocular, intraperitoneal , intramuscular, subcutaneous, intravenous administration. Oral administration or injection administration is preferred.
  • the pharmaceutical composition of the present invention can be produced by a method known in the art, such as a conventional mixing method, a dissolution method, a granulation method, a sugar-coating method, a pulverization method, an emulsification method, a freeze-drying method, and the like.
  • the present invention also provides a method of preparing a pharmaceutical composition comprising the steps of: administering a pharmaceutically acceptable carrier to a nucleoside reverse transcriptase inhibitor as described above, or a crystalline form thereof, a pharmaceutically acceptable salt, The hydrate or solvate is mixed to form a pharmaceutical composition.
  • NRTIs non-nucleoside reverse transcriptase inhibitors
  • the active ingredients of the invention may also be used in combination with other active ingredients.
  • the choice of such combination is based on the condition of the treatment, the cross-reactivity of the ingredients, and the combined pharmaceutical properties. It is also possible to administer any of the compounds of the invention in combination with one or more other active ingredients in a single dosage form for simultaneous or sequential administration to a patient.
  • Combination therapies can be administered simultaneously or sequentially. When administered continuously, the combination can be administered in two or more administrations.
  • Combination therapy can provide "synergistic effect” or “synergistic effect”, in other words, when the active ingredients are together The effect obtained using the effect is greater than the sum of the effects obtained by using the compound separately.
  • the active ingredient (1) is co-formulated and administered or delivered simultaneously in a combined formulation; (2) administered as a separate formulation or administered in parallel; or (3) obtained by some other dosage regimen Synergy.
  • synergistic effects can be obtained when the compounds are administered or released sequentially, for example, as separate tablets, pills or capsules, or by separate injections of separate syringes.
  • the effective dose of each active ingredient is administered sequentially, i.e., continuously, while in combination therapy, the effective dose of two or more active ingredients is administered together.
  • the present invention also discloses the use of a substituted adenosine nucleoside reverse transcriptase inhibitor as described above, i.e., the compound of the present invention can be advantageously used as a therapeutic agent for treating conditions such as AIDS and hepatitis B.
  • halogen means F, Cl, Br, and I unless otherwise specified. More preferably, the halogen atom is selected from the group consisting of F, Cl and Br.
  • deuterated means that one or more hydrogens in the compound or group are replaced by deuterium; deuteration may be monosubstituted, disubstituted, polysubstituted or fully substituted.
  • deuteration may be monosubstituted, disubstituted, polysubstituted or fully substituted.
  • deuterated is used interchangeably with “one or more deuterated”.
  • non-deuterated compound means a compound containing a proportion of germanium atoms not higher than the natural helium isotope content (0.015%).
  • compositions of the present invention optionally comprise a salt of a compound herein, particularly a pharmaceutically acceptable non-toxic salt, containing, for example, Na + , Li + , K + , Ca + 2 and Mg +2 .
  • a pharmaceutically acceptable non-toxic salt containing, for example, Na + , Li + , K + , Ca + 2 and Mg +2 .
  • These salts may include salts derived by the combination of suitable cations such as alkali and alkaline earth metal ions or ammonium and tetravalent amino ions and acid anion moieties, typically carboxylic acids. If a water-soluble salt is desired, a monovalent salt is preferred.
  • Metal salts are typically prepared by reacting a metal hydroxide with a compound of the invention. An example of a metal salt prepared in this manner is a salt containing Li + , Na + and K + .
  • the more insoluble metal salt can be precipitated from the more soluble salt solution by the addition of a suitable metal compound.
  • the salt may be formed by the addition of certain organic and inorganic acids, for example, HCl, HBr, H 2 SO 4 , H 3 PO 4 or an organic sulfonic acid, to a basic center, typically an amine, or to an acidic group. And formed.
  • the compositions herein comprise the compounds of the invention in their unionized, and zwitterionic form, and in combination with stoichiometric amounts of water, such as in hydrates.
  • salts of the parent compound with one or more amino acids are also included within the scope of the invention.
  • amino acids are suitable, particularly as naturally occurring amino acids found as protein components, although the amino acid is typically an amino acid with a side chain having a basic or acidic group, for example, lysine Acid, arginine or glutamic acid, or an amino acid having a side chain having a neutral group, for example, glycine, serine, threonine, alanine, isoleucine or leucine.
  • amino acid is typically an amino acid with a side chain having a basic or acidic group, for example, lysine Acid, arginine or glutamic acid, or an amino acid having a side chain having a neutral group, for example, glycine, serine, threonine, alanine, isoleucine or leucine.
  • the compounds of the invention may have a chiral center, for example, a chiral carbon or a phosphorus atom.
  • the compounds of the invention thus include racemic mixtures of all stereoisomers, including enantiomers, diastereomers and atropisomers. Additionally, the compounds of the invention include enriched or resolved optical isomers on any or all of the asymmetric chiral atoms. In other words, the chiral centers apparent from the description are provided as chiral isomers or racemic mixtures. Racemic mixtures and diastereomeric mixtures, as well as enantiomerically or diastereomeric partners which are substantially free of them, isolated or synthesized individual optical isomers, are all within the scope of the invention.
  • the compounds of the invention may also exist in the form of tautomers. Although only one type of non-localized resonant structure may be described, it is contemplated that all such forms fall within the scope of the present invention.
  • olefin-amine tautomers may be present, and all their possible tautomeric forms fall within the scope of the invention.
  • solvate refers to a complex of a compound of the invention that is coordinated to a solvent molecule to form a specific ratio.
  • Hydrophilate means a complex formed by the coordination of a compound of the invention with water.
  • the beneficial effects of the present invention are: the compound of the present invention has excellent inhibition to nucleoside reverse transcriptase; the technology of sputum is used to change the metabolism of the compound in the organism, so that the compound has more Good pharmacokinetic parameter characteristics.
  • the dosage can be changed and a long-acting preparation can be formed to improve the applicability; the substitution of a hydrogen atom in the compound with hydrazine increases the drug concentration of the compound in the animal due to its strontium isotope effect, and improves the therapeutic effect of the drug; Substituting a hydrogen atom in a compound inhibits certain metabolites and increases the safety of the compound.
  • each reaction is usually carried out in an inert solvent at room temperature to reflux temperature (eg Optimal ) proceed.
  • the reaction time is usually from 0.1 to 60 hours, preferably from 0.5 to 24 hours.
  • Phenol (2.0 g, 21.25 mmol) and sodium hydroxide (0.425 g, 10.63 mmol) were added to a microwave reaction flask, dissolved in 15 ml of heavy water, sealed, and placed in a microwave reactor at 180 ° C for 0.5 hour. The mixture was cooled to room temperature, and the mixture was acidified with dilute hydrochloric acid, and extracted with ethyl acetate 3-4 times. The organic phase was combined, washed with saturated brine, concentrated, and purified by silica gel column chromatography.
  • LC-MS (APCI): m / z 98.1 (M + 1) +.
  • Step 7 9- ⁇ (R)-2-[((R,S)- ⁇ [(S)-1-(isopropyloxycarbonyl)ethyl]amino ⁇ -2,4,6-d3-phenoxyphosphoryl) Synthesis of acyl)methoxy]propyl ⁇ adenine (Compound 7).
  • Step 8 9- ⁇ (R)-2-[((S)- ⁇ [(S)-1-(Isopropoxycarbonyl)ethyl]amino ⁇ -2,4,6-d3-phenoxyphosphoryl) Separation of methoxy]propyl ⁇ adenine (Compound T-1).
  • the racemic compound 7 (100 mg) was separated by a chiral supercritical fluid chromatography column (SFC) to obtain the target product T-1, which was dried to give a weight of 45 mg, yield: 90%.
  • LC-MS (APCI): m / z 480.5 (M + 1) +.
  • Step 3 9- ⁇ (R)-2-[((R,S)- ⁇ [(S)-1-(d7-isopropyloxycarbonyl)ethyl]amino ⁇ phenoxyphosphoryl)methoxy]propyl Synthesis of adenine (Compound 10).
  • Step 4 9- ⁇ (R)-2-[((S)- ⁇ [(S)-1-(d7-isopropyloxycarbonyl)ethyl]amino ⁇ phenoxyphosphoryl)methoxy]propyl ⁇ Separation of adenine (Compound T-2).
  • the racemic compound 10 (170 mg) was separated by a chiral supercritical fluid chromatography column (SFC) to obtain the target product T-2, which was dried to give a weight of 78.4 mg, yield: 46.1%.
  • LC-MS (APCI): m / z 484.5 (M + 1) +.
  • Phenol (2.0 g, 21.25 mmol), 5% Pt/C (0.4 g, 20 wt%) and 34 ml of heavy water were added to the reaction flask, and the hydrogen was replaced 3-4 times.
  • the reaction was carried out at room temperature for 24 hours, and the catalyst was removed by filtration.
  • Step 3 9- ⁇ (R)-2-[((R,S)- ⁇ [(S)-1-(isopropyloxycarbonyl)ethyl]amino ⁇ -d5-phenoxyphosphoryl)methoxy] Synthesis of propyl ⁇ adenine (Compound 13).
  • Step 4 9- ⁇ (R)-2-[((S)- ⁇ [(S)-1-(isopropyloxycarbonyl)ethyl]amino ⁇ -d5-phenoxyphosphoryl)methoxy]propyl ⁇ Separation of adenine (compound T-3).
  • Example 4 9- ⁇ (R)-2-[((S)- ⁇ [(S)-1-(isopropyloxycarbonyl)-d4-ethyl]amino ⁇ phenoxyphosphoryl)methoxy]propyl Adenine, the compound T-4, has the formula:
  • Step 2 9- ⁇ (R)-2-[((R,S)- ⁇ [(S)-1-(isopropyloxycarbonyl)-d4-ethyl]amino ⁇ phenoxyphosphoryl)methoxy] Synthesis of propyl ⁇ adenine (Compound 15).
  • Step 3 9- ⁇ (R)-2-[((S)- ⁇ [(S)-1-(isopropyloxycarbonyl)-d4-ethyl]amino ⁇ phenoxyphosphoryl)methoxy]propyl ⁇ Separation of adenine (Compound T-4).
  • the racemic compound 15 (150 mg) was separated by a chiral supercritical fluid chromatography column (SFC) to obtain the target product T-4, which was dried and weighed to obtain 53 mg, yield: 70.7%.
  • LC-MS (APCI): m / z 481.5 (M + 1) +.
  • Step 6 9- ⁇ (R)-2-[((R,S)- ⁇ [(S)-1-(Isopropoxycarbonyl)ethyl]amino ⁇ phenoxyphosphoryl)methoxy]propyl ⁇ Synthesis of -2,8-d2-adenine (Compound 21).
  • Step 7 9- ⁇ (R)-2-[((S)- ⁇ [(S)-1-(isopropyloxycarbonyl)ethyl]amino ⁇ phenoxyphosphoryl)methoxy]propyl ⁇ -2 Separation of 8-d2-adenine (Compound T-5).
  • Step 5 9- ⁇ (R)-2-[((R,S)- ⁇ [(S)-1-(Isopropoxycarbonyl)ethyl]amino ⁇ phenoxyphosphoryl)-d2-methoxy] Synthesis of propyl ⁇ adenine (Compound 26).
  • Step 6 9- ⁇ (R)-2-[((S)- ⁇ [(S)-1-(isopropyloxycarbonyl)ethyl]amino ⁇ phenoxyphosphoryl)-d2-methoxy]propyl ⁇ Separation of adenine (Compound T-6).
  • the racemic compound 26 (150 mg) was separated using a chiral supercritical fluid chromatography column (SFC) to obtain the objective product T-6, which was dried and weighed to yield 48 mg, yield: 64%.
  • LC-MS (APCI): m / z 479.1 (M + 1) +.
  • test compound and the reference compound will be diluted in DMSO and added to the cell culture plate.
  • the test compound and the reference compound will be tested at 8 concentrations, two duplicate wells.
  • HIV-1 and MT-4 cells were co-cultured for 1 h at 37 ° C in a 5% CO 2 incubator. The infected cells are then seeded in a cell culture plate at a density. The final concentration of DMSO in the cell culture medium was 0.5%. The cells were cultured for 5 days at 37 ° C in a 5% CO 2 incubator. The cells in the cytotoxicity test were uninfected MT-4 cells, and other experimental conditions were consistent with the antiviral activity experiments.
  • Cell viability assay Cell viability was determined by the cell activity assay reagent CellTiter-Glo (Promega). Raw data were used for compound anti-HIV-1 activity and cytotoxicity calculations. Compound dose response curve and the EC 50 and CC 50 values obtained by the analysis software GraphPad Prism, wherein, A represents EC 50 ⁇ 10nM, B represents 10nM ⁇ EC 50 ⁇ 100nM, C represents 100nM ⁇ EC 50 ⁇ 500nM, D represents EC 50 > 500 Nm; F means CC 50 > 10000 nM (as shown in Table 1 below).
  • the anti-hepatitis C virus activity of the compound was determined by detecting luciferase using Bright-Glo (Promega). Analysis of the data using GraphPad Prism software fitting curve 50 and EC 50 values were calculated and CC.
  • Anti-cell activity assay 20 compounds were tested for anti-HBV activity in HepG2.2.15 cells, and TDF was used as a positive control compound. On the first day, the cells were seeded into a 96-well plate, the compound was added to the cells for the next day, and the new compound-containing medium was replaced on the fifth day. On the eighth day, the supernatant was collected to extract DNA. The amount of HBV DNA was detected by quantitative PCR. The test compound and TDF were serially diluted 3 times, 8 concentration points, and 2 duplicate wells were determined in parallel. The final concentration of DMSO in the culture broth was 0.5%. The inhibition percentage is calculated as follows:
  • % inhibition rate (1 - copy number of HBV in the sample / copy number of HBV in the DMSO control group) ⁇ 100
  • EC 50 by the Graphpad Prism software (four parameter logistic equations) analysis, where I represents EC 50 ⁇ 5nM, II represents 5nM ⁇ EC 50 ⁇ 20nM, III represents 20nM ⁇ EC 50 ⁇ 100nM, IV represents EC 50> 100nM (as in Table 1 Shown).
  • Cytotoxicity test The compound plate and compound treatment procedure were consistent with the detection of anti-HIV activity. After six days of treatment of the cells, the cell viability was determined. Add Cell-titer Blue reagent to each well, incubate for 3 hours at 37 °C, read fluorescence values (560Ex/590Em); analyze data and calculate relative cell viability:
  • % cell viability (sample fluorescence reading - fluorescence reading of the culture control) Number) / (Fluorescence reading of DMSO control - fluorescence reading of culture control) x 100.
  • CC 50 value of the compound was calculated using GraphPad Prism software, and V represents CC 50 >200000 nM (as shown in Table 1 below).
  • the experimental results show that the compounds of the present invention have strong anti-HIV activity and HBV activity (both to nanomolar levels), compared with the newly-listed anti-HIV drug (GS7340) of the American pharmaceutical company Gilead Science Co., Ltd. Both anti-HIV activity and anti-HBV activity were comparable, and the anti-HBV activities of the example compounds T-5 and T-6 showed activity superior to GS7340. Furthermore, the compounds of the invention showed no toxicity (optimal CC 50 >200,000 nM) in the cell lines tested.
  • Microsomal experiments human liver microsomes: 0.5 mg/mL, Xenotech; rat liver microsomes: 0.5 mg/mL, Xenotech; coenzyme (NADPH/NADH): 1 mM, Sigma Life Science; magnesium chloride: 5 mM, 100 mM phosphate buffer Agent (pH 7.4).
  • phosphate buffer 100 mM, pH 7.4.
  • the pH was adjusted to 7.4, diluted 5 times with ultrapure water before use, and magnesium chloride was added to obtain a phosphate buffer (100 mM) containing 100 mM potassium phosphate, 3.3 mM magnesium chloride, and a pH of 7.4.
  • NADPH regeneration system containing 6.5 mM NADP, 16.5 mM G-6-P, 3 U/mL G-6-P D, 3.3 mM magnesium chloride was prepared and placed on wet ice before use.
  • Formulation stop solution acetonitrile solution containing 50 ng/mL propranolol hydrochloride and 200 ng/mL tolbutamide (internal standard). Take 25057.5 ⁇ L of phosphate buffer (pH 7.4) into a 50 mL centrifuge tube, add 812.5 ⁇ L of human liver microsomes, and mix to obtain a liver microsome dilution with a protein concentration of 0.625 mg/mL. 25057.5 ⁇ L of phosphate buffer (pH 7.4) was taken into a 50 mL centrifuge tube, and 812.5 ⁇ L of SD rat liver microsomes were added and mixed to obtain a liver microsome dilution having a protein concentration of 0.625 mg/mL.
  • the corresponding compound had a reaction concentration of 1 ⁇ M and a protein concentration of 0.5 mg/mL.
  • 100 ⁇ L of the reaction solution was taken at 10, 30, and 90 min, respectively, and added to the stopper, and the reaction was terminated by vortexing for 3 min.
  • the plate was centrifuged at 5000 x g for 10 min at 4 °C.
  • 100 ⁇ L of the supernatant was taken into a 96-well plate to which 100 ⁇ L of distilled water was previously added, mixed, and sample analysis was performed by LC-MS/MS.
  • the compound of the present invention has a longer half-life and a smaller clearance rate, and exhibits superior metabolic stability in both human liver microsomes and rat liver microsomes. Suitable as a drug against HIV or HBV.
  • SD rat grade SPF grade
  • Weight range 180 ⁇ 220g (actual weight range is 187 ⁇ 197g)
  • Group A was given 9- ⁇ (R)-2-[((S)- ⁇ [(S)-1-(isopropyloxycarbonyl)ethyl]amino ⁇ phenoxyphosphoryl)methoxy]propyl ⁇ gland ⁇ 3mg/kg
  • group B was given 3mg/kg of the compound of Example 1-6
  • blood was taken from the orbital vein of the rat for about 100-200L at 15min, 30min, 1, 2, 3, 5, 8 and 10h after administration.
  • the male SD rats were determined by the non-compartmental statistical moment theory to give 9- ⁇ (R)-2-[((S)- ⁇ [(S)-1-(isopropyloxycarbonyl)ethyl]amino ⁇ phenoxyphosphoryl)methoxy]propyl ⁇ adenine (3 mg/kg), the compound of Example 1-6 (3 mg/kg) Post-pharmacokinetic related parameters.
  • the compound of the present invention has superior activity and has excellent pharmacokinetic properties, and thus is more suitable as a compound for inhibiting nucleoside reverse transcriptase, and is therefore suitable for preparing a medicament for treating antiviral infection.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

L'invention concerne un composé d'adénine substituée et une composition pharmaceutique correspondante, le composé d'adénine substituée étant un composé de formule (I), ou une forme cristalline, un sel, promédicament, stéréoisomère, hydrate ou solvate associé pharmaceutiquement acceptable. Le composé selon la présente invention peut inhiber l'activité d'une transcriptase inverse de nucléoside et présente également de meilleures propriétés pharmacodynamiques/pharmacocinétiques ; le composé a une grande applicabilité, il est sans danger et peut être utilisé pour la préparation d'une composition pharmaceutique destinée au traitement des maladies associées aux infections virales, ce qui lui donne de grandes possibilités commerciales.
PCT/CN2017/074135 2016-03-01 2017-02-20 Composé d'adénine substituée et composition pharmaceutique correspondante WO2017148290A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201780003912.6A CN108350007B (zh) 2016-03-01 2017-02-20 一种取代的腺嘌呤化合物及其药物组合物

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201610114361 2016-03-01
CN201610114361.8 2016-03-01

Publications (1)

Publication Number Publication Date
WO2017148290A1 true WO2017148290A1 (fr) 2017-09-08

Family

ID=59743459

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2017/074135 WO2017148290A1 (fr) 2016-03-01 2017-02-20 Composé d'adénine substituée et composition pharmaceutique correspondante

Country Status (2)

Country Link
CN (1) CN108350007B (fr)
WO (1) WO2017148290A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108101943A (zh) * 2018-02-28 2018-06-01 顾世海 一种替诺福韦前药或可药用盐及其在医药上的应用
CN112778388A (zh) * 2021-01-21 2021-05-11 大连医科大学 一种核苷类似物及其制备方法和应用

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2018171665A1 (fr) * 2017-03-23 2018-09-27 四川好医生攀西药业有限责任公司 Analogue nucléotidique deutéré et son utilisation

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1443189A (zh) * 2000-07-21 2003-09-17 吉里德科学公司 核苷酸膦酸酯类似物前药及其筛选和制备方法
CN104327137A (zh) * 2014-11-07 2015-02-04 王彩琴 一种氘代索菲布韦及其用途
CN105254694A (zh) * 2014-07-14 2016-01-20 正大天晴药业集团股份有限公司 氘代核苷衍生物
CN105669751A (zh) * 2015-03-05 2016-06-15 洛阳聚慧医药科技有限公司 非环核苷酸磷酰胺类化合物及其盐的制备以及在抗病毒方面的应用
CN106167504A (zh) * 2015-11-04 2016-11-30 洛阳聚慧医药科技有限公司 非环核苷磷酰胺d‑氨基酸酯衍生物及其盐的制备以及在抗病毒方面的应用

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1443189A (zh) * 2000-07-21 2003-09-17 吉里德科学公司 核苷酸膦酸酯类似物前药及其筛选和制备方法
CN105254694A (zh) * 2014-07-14 2016-01-20 正大天晴药业集团股份有限公司 氘代核苷衍生物
CN104327137A (zh) * 2014-11-07 2015-02-04 王彩琴 一种氘代索菲布韦及其用途
CN104672288A (zh) * 2014-11-07 2015-06-03 王彩琴 一种氘代索菲布韦及其用途
CN105669751A (zh) * 2015-03-05 2016-06-15 洛阳聚慧医药科技有限公司 非环核苷酸磷酰胺类化合物及其盐的制备以及在抗病毒方面的应用
CN106167504A (zh) * 2015-11-04 2016-11-30 洛阳聚慧医药科技有限公司 非环核苷磷酰胺d‑氨基酸酯衍生物及其盐的制备以及在抗病毒方面的应用

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN108101943A (zh) * 2018-02-28 2018-06-01 顾世海 一种替诺福韦前药或可药用盐及其在医药上的应用
CN108101943B (zh) * 2018-02-28 2020-11-24 顾世海 一种替诺福韦前药或可药用盐及其在医药上的应用
CN112778388A (zh) * 2021-01-21 2021-05-11 大连医科大学 一种核苷类似物及其制备方法和应用
CN112778388B (zh) * 2021-01-21 2022-08-23 大连医科大学 一种核苷类似物及其制备方法和应用

Also Published As

Publication number Publication date
CN108350007B (zh) 2020-04-10
CN108350007A (zh) 2018-07-31

Similar Documents

Publication Publication Date Title
US11266666B2 (en) Methods for treating Filoviridae virus infections
KR102604975B1 (ko) (s)-7-(1-아크릴로일피페리딘-4-일)-2-(4-페녹시페닐)-4,5,6,7-테트라-하이드로피라졸로 [1,5-a] 피리미딘-3-카르복스아미드의 제조 및 그 용도
TWI389908B (zh) 抗病毒化合物類
EP2891658B1 (fr) Promédicament du ténofovir et utilisations pharmaceutiques associées
JP7066728B2 (ja) C型肝炎ウイルスの治療のためのヌクレオチドヘミ硫酸塩
CN110759908B (zh) 用于抑制Bcl-2蛋白的N-苯磺酰基苯甲酰胺类化合物及其组合物及应用
US10626118B2 (en) Pyrrolopyrimidine crystal for preparing JAK inhibitor
WO2017148290A1 (fr) Composé d'adénine substituée et composition pharmaceutique correspondante
KR102434764B1 (ko) 간 전송 항바이러스 전구체 약물 뉴클레오시드 시클로 포스페이트 에스테르 화합물 및 응용
KR102502749B1 (ko) 간 전달 엔테카비어 프로드러그 뉴클레오티드 시클로 포스페이트 화합물 및 응용
CN108350008B (zh) 一种新型的无环核苷类似物及其药物组合物
CN108676009B (zh) 作为her2酪氨酸激酶抑制剂的嘧啶衍生物及其应用
KR20060127906A (ko) 4'-치환된 카보버와 아바카비어 유도체 및 hiv와 hcv항바이러스 활성을 갖는 관련 화합물
CN108794517B (zh) 一种精氨酸酶抑制剂及其制备方法与用途
WO2021098850A1 (fr) Forme cristalline d'un inhibiteur de nucléoprotéine et son utilisation
CN111836818A (zh) 氨基磷(膦)酸缩醛和磷(膦)酸缩醛化合物
WO2023169572A1 (fr) Composé triazine, intermédiaire de celui-ci, procédé de préparation associé et utilisation correspondante
JP2000309598A (ja) 多剤結合型新規化合物、その製造法および用途
RU2666727C1 (ru) Ингибитор вируса гепатита В (ВГВ)
TWI382978B (zh) 化學化合物
TW202313066A (zh) 非天然構型之核苷酸前藥化合物
TW201736391A (zh) 作為人類鼻病毒抑制劑之炔基核苷類似物
WO2001038306A9 (fr) Nouveaux derives de la 3-nitropyridine et preparations pharmaceutiques les contenant
CN111484541A (zh) 双核苷酸前体药物及其制备方法
CN107849074A (zh) 一种核苷类似物的烷氧烷基酯前药及其应用

Legal Events

Date Code Title Description
NENP Non-entry into the national phase

Ref country code: DE

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

Ref document number: 17759141

Country of ref document: EP

Kind code of ref document: A1

122 Ep: pct application non-entry in european phase

Ref document number: 17759141

Country of ref document: EP

Kind code of ref document: A1