CN110627774A - Anti-hepatic fibrosis compound, preparation method and application - Google Patents

Anti-hepatic fibrosis compound, preparation method and application Download PDF

Info

Publication number
CN110627774A
CN110627774A CN201910978213.4A CN201910978213A CN110627774A CN 110627774 A CN110627774 A CN 110627774A CN 201910978213 A CN201910978213 A CN 201910978213A CN 110627774 A CN110627774 A CN 110627774A
Authority
CN
China
Prior art keywords
chlorophenoxy
hepatic fibrosis
compound
preparing
preparation
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.)
Withdrawn
Application number
CN201910978213.4A
Other languages
Chinese (zh)
Inventor
艾观华
彭维杰
刘宇政
罗丹
冷红文
古超伦
赵林
赵芙蓉
刘玉婷
李婷婷
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Jiangxi Academy Of Medical Sciences
Nanchang University
Gannan Medical University
Original Assignee
Jiangxi Academy Of Medical Sciences
Nanchang University
Gannan Medical University
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 Jiangxi Academy Of Medical Sciences, Nanchang University, Gannan Medical University filed Critical Jiangxi Academy Of Medical Sciences
Priority to CN201910978213.4A priority Critical patent/CN110627774A/en
Publication of CN110627774A publication Critical patent/CN110627774A/en
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention belongs to the technical field of medicines, and particularly relates to an anti-hepatic fibrosis compound, a preparation method and application, wherein the structural formula is as follows:the medicine of the present invention can obviously improve hepatic fibrosis, improve liver function indexes, reduce the accumulation of extracellular matrixes such as collagen in the extracellular matrix, reduce the degree of hepatic fibrosis, and inhibit the formation and the development of hepatic fibrosis.

Description

Anti-hepatic fibrosis compound, preparation method and application
Technical Field
The invention belongs to the technical field of medicines, and particularly relates to an anti-hepatic fibrosis compound, a preparation method and application.
Background
Hepatic fibrosis is a process of diffuse excessive deposition of liver extracellular matrix (ECM) caused by various pathogenic factors, and is a common pathological pathway for most chronic liver diseases. Further progression of liver fibrosis can lead to cirrhosis of the liver and possibly even further hepatocellular carcinoma. Research proves that hepatic fibrosis can be reversed, and no definite anti-hepatic fibrosis treatment medicine is found clinically at present, so that the search for a treatment medicine capable of effectively reversing hepatic fibrosis has important clinical significance. Autotaxin (ATX) is a secreted glycoprotein, has phosphodiesterase and lysophospholipase D activities, and clinical case reports show that ATX in serum is positively correlated with hepatic fibrosis degree, and clinical studies also show that patients with viral infection (HBV and HCV) have increased serum ATX level, liver cirrhosis and liver cancer incidence; preliminary mechanism research shows that the active product LPA of ATX can activate hepatic stellate cells and promote collagen secretion. Therefore, ATX is also considered to be a potent serum marker of liver fibrosis and a target for developing anti-liver fibrosis drugs.
Research on autotaxin inhibitors has progressed, late rain sprout, etc., in central south medicine 2016, 9, 14 th, 9 th, and various ATX inhibitors are described, 1, ATX inhibitors of fatty alcohol phosphatidic acid structure, including enteron (darmstoff), cyclic phosphatidic acid (cPA), α -halomethylenephosphonic acid, alkyl aromatic methylenephosphonic acid, and inhibitors of tyrosine backbone structure, each of which consists of 1 electron-rich polar phosphate moiety and 1 hydrophobic chain; 2. small molecule ATX inhibitors including ATX inhibitors having a long linear or flexible structure, ATX inhibitors of rigid structural molecules, and metal ion chelating molecule ATX inhibitors; 3. other structures of ATX inhibitors.
Disclosure of Invention
The invention aims to solve the technical problem of providing an anti-hepatic fibrosis compound, a preparation method and application.
The invention comprises an anti-hepatic fibrosis compound, namely 2- ({5- [1- (4-chlorophenoxy) ethyl ] -4-phenyl-4H-1, 2, 4-triazol-3-yl } thio) -N- (1, 5-dimethyl-3-oxo-2-phenyl-2, 3-dihydro-1H-pyrazol-4-yl) acetamide, with the following structural formula:
the invention provides a preparation for resisting hepatic fibrosis, which comprises a compound for resisting hepatic fibrosis, and the preparation is tablets, coated tablets, sugar-coated pills, hard and soft gelatin capsules, solutions, emulsions, suspensions, nasal sprays, suppositories, ointments, gels or injections.
The preparation for resisting hepatic fibrosis can be a medicinal salt of a compound for resisting hepatic fibrosis, and can also be used in the form of a medicinal preparation. The pharmaceutical formulation may be administered internally in the following manner: such as orally (for example in the form of tablets, coated tablets, dragees, hard and soft gelatine capsules, solutions, emulsions or suspensions), nasally (for example in the form of nasal sprays) or rectally (for example in the form of suppositories) or topically ocularly (for example in the form of solutions, ointments, gels or water-soluble polymer inserts). Administration can also be effected parenterally, such as intramuscularly, intravenously or intraocularly (for example in the form of sterile injectable solutions).
The anti-hepatic fibrosis preparation can be processed with pharmaceutically inert, inorganic or organic adjuvants, and can be used for preparing tablet, coated tablet, sugar-coated pill, hard gelatin capsule, injection or topical preparation. Lactose, corn starch or derivatives thereof, talc, stearic acid or its salts and the like can be used, for example, as such auxiliary materials for tablets, dragees and hard gelatine capsules.
Suitable excipients for soft gelatine capsules are, for example, vegetable oils, waxes, fats, semi-solid substances, liquid polyols and the like. Suitable adjuvants for the preparation of solutions and syrups are, for example, water, polyols, sucrose, invert sugar, glucose and the like. Suitable excipients for injection solutions are, for example, water, alcohols, polyols, glycerol, vegetable oils and the like. Suitable excipients for suppositories are, for example, natural or hardened oils, waxes, fats, semi-solid or liquid polyols and the like. Suitable adjuvants for topical ocular formulations are, for example, cyclodextrins, mannitol, or many other carriers and excipients known in the art. In addition, the pharmaceutical preparations may contain preservatives, solubilizers, viscosity-increasing substances, stabilizers, wetting agents, emulsifiers, sweeteners, colorants, flavorants, salts for regulating the osmotic pressure, buffers, masking agents or antioxidants; they may also contain other therapeutically valuable substances.
The invention provides a preparation method of an anti-hepatic fibrosis compound, which comprises the following steps:
adding 4-chloroacetylamidoantipyrine, 5- (1- (4-chlorophenoxy) ethyl) -4-phenyl-4H-1, 2, 4-triazole-3-thiol and potassium carbonate into acetone, refluxing, filtering, evaporating filtrate to dryness, and recrystallizing residue to obtain compound for resisting hepatic fibrosis.
The preparation method of the 4-chloracetyl amino antipyrine comprises the steps of adding chloracetyl chloride and triethylamine into a dichloromethane solution containing the 4-amino antipyrine, and stirring at room temperature; then adding ice water into the solution for extraction, taking an organic layer, washing the organic layer by using a hydrochloric acid solution, then washing by using a saturated sodium bicarbonate aqueous solution, and then washing by using brine; after drying, removing the solvent to obtain a crude product; recrystallizing the crude product to obtain the 4-chloroacetylaminoaantipyrine.
The preparation method of the 5- (1- (4-chlorophenoxy) ethyl) -4-phenyl-4H-1, 2, 4-triazole-3-thiol comprises the steps of dissolving 2- (2- (4-chlorophenoxy) propionyl) -N-phenylhydrazine thiocarboxamide in a sodium hydroxide solution, heating and refluxing, cooling, adjusting the pH value to 2-3, filtering, and drying to obtain the 5- (1- (4-chlorophenoxy) ethyl) -4-phenyl-4H-1, 2, 4-triazole-3-thiol.
The 2- (2- (4-chlorophenoxy) propionyl) -N-phenylhydrazine thiocarboxamide is prepared by adding 2- (4-chlorophenoxy) propionylhydrazine and phenyl isothiocyanate to absolute ethyl alcohol, refluxing, cooling the mixture to room temperature, filtering, and concentrating the filtrate to obtain 2- (2- (4-chlorophenoxy) propionyl) -N-phenylhydrazine thiocarboxamide.
The preparation method of the 2- (4-chlorophenoxy) propionylhydrazine comprises the steps of dissolving 2- (4-chlorophenoxy) propionate in ethanol, and then adding a hydrazine water solution; the mixture was then stirred under reflux, concentrated in vacuo, and washed to give 2- (4-chlorophenoxy) propionylhydrazide.
The preparation method of the 2- (4-chlorophenoxy) propionic ester comprises the steps of dissolving the 2- (4-chlorophenoxy) propionic acid in methanol, and then slowly adding 98% concentrated sulfuric acid; stirring under reflux, cooling to room temperature, concentrating in vacuo, and dissolving the residue in dichloromethane, washing, drying, and removing the solvent to give 2- (4-chlorophenoxy) propionate.
The specific reaction route is as follows:
the invention provides application of an anti-hepatic fibrosis compound in preparing an ATX inhibitor, an anti-hepatic fibrosis preparation or a liver cancer treatment drug. Anti-hepatic fibrosis compounds when used as ATX inhibitors, anti-hepatic fibrosis preparations or drugs for treatment of liver cancer, the dosage may vary within wide ranges and will of course be adapted to the individual requirements in each particular case. Generally, in case of oral administration, a daily dose of about 0.1mg to 20mg per kg body weight, preferably about 0.5mg to 4mg per kg body weight (e.g. about 300mg per person), preferably divided into 1-3 individual doses which may e.g. consist of the same amount, should be suitable. In the case of topical administration, the formulation may contain from 0.001% to 15% by weight of the drug, and the required dose (which may be 0.1-25mg) may be administered by a single dose per day or week, or by multiple doses (2 to 4) per day, or by multiple doses per week. However, it will be clear that, when explicitly indicated, the upper or lower limits given herein may be exceeded.
The compound is an inhibitor specially aiming at human ATX enzyme, can well act on target enzyme ATX, regulates the in vivo LPA level by inhibiting ATX so as to play a role in preventing or treating hepatic fibrosis, and determines the IC of the compound through immunofluorescence detection50The value is 43.05 mu M, and the compound has better anti-hepatic fibrosis effect as shown in figures 3-5.
The compound is an ATX inhibitor, can inhibit lysophospholipase D from catalyzing and generating lysophosphatidic acid LPA by taking Lysophosphatidylcholine (LPC) as a substrate through inhibiting the activity of the lysophospholipase D, can be used as a regulator of LPA level and related signal transduction, is a specific inhibitor of ATX, can inhibit the generation or further aggravation of hepatic fibrosis through inhibiting the activity of the ATX, and has good clinical application prospect.
The compound can well improve the hepatic fibrosis of mice through mouse anti-fibrosis efficacy tests, and has a bright prospect in developing medicaments for treating the hepatic fibrosis in the future.
Drawings
FIG. 1 is a mass spectrum of the compound.
FIG. 2 shows the compound1H-NMR spectrum.
FIG. 3 shows CC1 used alone4Sirius red staining pattern of mouse liver tissue section of mouse liver injury model.
FIG. 4 shows the use of high dose (30 mg. kg)-1·d-1) Inventive drug treatment CC14Sirius red staining pattern of mouse liver tissue section of mouse liver injury model.
FIG. 5 shows the use of a low dose (15 mg. kg)-1·d-1) Inventive drug treatment CC14Sirius red staining pattern of mouse liver tissue section of mouse liver injury model.
Detailed Description
Example 1 preparation process conditions:
preparation of 2- (4-chlorophenoxy) propionic acid (2)
4-chlorophenol (12.80g, 100.0mmol) was dissolved in 150mL acetonitrile, followed by the addition of potassium carbonate (41.40g, 300.0 mmol). The mixture was stirred at 50 ℃ for 30 minutes, then 2-bromopropionic acid (15.20g, 100.0mmol) was added. The mixture was stirred at reflux overnight. After cooling to room temperature, the mixture was filtered and the filtrate was concentrated in vacuo. The residue was added to water and the pH was adjusted to 3 with 1N HCl and the resulting mixture was extracted with DCM (3 × 100 mL). The organic layer was washed with brine (100mL), dried over MgSO4, and concentrated in vacuo to give 17.10g of a colorless oil. The yield thereof was found to be 85%.
2.2 preparation of methyl (4-chlorophenoxy) propionate (3)
2- (4-chlorophenoxy) propionic acid was dissolved in 150mL of methanol, and 10mL of concentrated sulfuric acid (98 wt%) was slowly added. The mixture was then stirred at reflux overnight. After cooling to room temperature, the mixture was concentrated in vacuo and the residue was dissolved in DCM and washed with water (3 × 100mL) and again with saturated NaHCO3The aqueous solution (100mL) was washed with brine (100 mL). With MgSO4After drying, the solvent was removed to give 17.30g of a colorless oil, which was used in the next step without further purification. The yield thereof was found to be 95%.
3.2 preparation of (4-chlorophenoxy) propionylhydrazide (4)
2- (4-chlorophenoxy) propionate was dissolved in 150mL of ethanol, and then 15mL of an aqueous solution of hydrazine (80 wt% in water) was added. The mixture was then stirred at reflux for 3 hours. The mixture was then concentrated in vacuo and the residue was washed with ethanol. The obtained dried white solid was used to obtain 13.05g of 2- (4-chlorophenoxy) propionylhydrazide. The yield thereof was found to be 75%.
4.2 preparation of 2- (2- (4-chlorophenoxy) propionyl) -N-phenylhydrazine thiocarboxamide (5)
2- (4-chlorophenoxy) propionohydrazide (13.05g, 60.9mmol) and phenyl isothiocyanate (8.22g, 60.9mmol) were placed in 120mL of anhydrous ethanol and refluxed for 5 hours on a steam bath. The mixture was cooled to room temperature and then filtered. The filtrate was concentrated to give 18.15g of a light brown oil, which was used in the next step without further purification.
5.5- (1- (4-chlorophenoxy) ethyl) -4-phenyl-4H-1, 2, 4-triazole-3-thiol (6) 18.15g of 2- (2- (4-chlorophenoxy) propionyl) -N-phenylhydrazine thiocarboxamide were dissolved in 100ml of 2N sodium hydroxide solution and heated under reflux for 3 hours. Cooled, acidified to pH2-3 with 1N HCl and filtered to collect a white solid. After drying, 13.80g of a white solid are obtained. The yield thereof was found to be 76%.
Preparation of 2-chloro-N- (1, 5-dimethyl-3-oxo-2-phenyl-2, 3-dihydro-1H-pyrazol-4-yl) acetamide (8)
Chloroacetyl chloride (9.04g, 80mmol) and triethylamine (8.08g, 80mmol) were added to 150mL of a solution containing 4-aminoantipyrine (16.24g, 80mmol) in DCM and stirred at room temperature overnight. Then, a volume of ice water was added to the solution for extraction, the organic layer was taken and washed first with 1N HCl solution (3X 100mL) and then with saturated NaHCO3The aqueous solution (100mL) was washed, then brine (100mL) was added. With MgSO4After drying, the solvent was removed to obtain 25.15g of a green solid. The crude product was recrystallized from DCM/MTBE to give 13.83g of a white solid. The yield thereof was found to be 62%.
7. Preparation of 2- ((5- (1- (4-chlorophenoxy) ethyl) -4-phenyl-4H-1, 2, 4-triazol-3-yl) thio) -N- (1, 5-dimethyl-3) oxo-2-phenyl-2, 3-dihydro-1H-pyrazol-4-yl) acetamide (9)
A mixture of 4-chloroacetylamidoantipyrine (8.42g, 30.2mmol), 5- (1- (4-chlorophenoxy) ethyl) -4-phenyl-4H-1, 2, 4-triazole-3-thiol (10.00g, 30.2mmol) and anhydrous potassium carbonate (8.33g, 60.4mmol) were added to 150mL of acetone, refluxed for 4 hours, filtered and the filtrate evaporated to dryness. The residue was recrystallized from MeOH/MTBE to give 7.05g of a white solid. The yield thereof was found to be 40%.
As shown in FIGS. 1-2, FIG. 1 is the mass spectrum of the compound, and FIG. 2 is the mass spectrum of the compound1H-NMR spectrum.
Nuclear magnetic spectrum data of1H-NMR(400MHz,DMSO-d6):δ=1.58(3H,d,J=6.0Hz),2.07(3H,s), 3.04(3H,s),4.10(2H,s),5.54(1H,q),6.76(3H,d,J=8.8Hz),7.24(3H,d,J=9.2Hz),7.23–7.53 (10H,m),9.48(1H,s)。
Experimental example 1 human ATX enzyme inhibition assay
1. ATX inhibition was measured by fluorescence quenching assay using the specifically labeled substrate Amplex Red reagent, which was obtained by purchasing the kit.
2. The assay working solution was prepared as follows:
2.1 assay buffer (50mM Tris-HCl, 140mM NaCl, 5mM KCl, 1mM CaCl2、1mM MgCl2、 0.01%Triton-X-100、pH8.0;
2.2ATX solution: stock solutions of ATX (mouse ATX- β from nano Biological) (48nM, diluted in assay buffer) were diluted to a final concentration of 2nM in assay buffer at the time of assay;
2.3LPC substrate solution: LPC (16:0) substrate solid was weighed and diluted to 250nM final concentration in assay buffer.
2.4 detection method: ATX was detected using the Amplex Red kit, each reaction containing 50. mu.M Amplex Red reagent, 1U/mL HRP, 0.1U/mL choline oxidase, 250nM LPC and the amount of ATX diluted with 1 Xreaction buffer, and the reaction was incubated at 37 ℃ for 1 hour. Fluorescence was measured using a fluorescence microplate reader with an excitation wavelength of 530. + -. 12.5nm and an emission wavelength of 590. + -. 17.5 nm.
2.5A series of solutions of the compound of the present invention diluted with DMSO to 10mM, 8mM, 6.4mM, 5.12mM, 4.10mM, 3.28mM, 2.62mM, 2.10mM, 1.68mM, 1.34mM, 1.07mM were added to 2. mu.L to 2.4, respectively, of the kit solutionThe change in fluorescence values after addition of the compound was determined by the method of 2.4, and the IC of the compound was calculated from these data50The value was 43.05. mu.M.
Example 2 study of drug efficacy
1. Experimental materials and animals
1.1 drugs and reagents: glutamic-pyruvic transaminase determination kit (R1, R2) (MIVDRAY kit), glutamic-oxaloacetic transaminase determination kit R1, R2) (MIVDRAY kit), carbon tetrachloride (analytically pure), and rapeseed oil is prepared into 20% rapeseed oil solution before use.
1.2 Experimental animals: clean-grade Kunming mice, weighing 18-22g, Hunan Slek Jingda laboratory animals Co.
1.3 Experimental instruments: MIVDRAY BS-380 model full-automatic biochemical analyzer, EX 125DZH model electronic analytical balance (Aohaus instruments, Inc.), HC30L8R model high-speed refrigerated frozen centrifuge (Anhui Zhongzhou scientific instruments, Inc.).
1.4 tested drugs and treatment methods: taking the compound obtained in the preparation of example 1, the high dose group was diluted to 1.5mg/ml with 5% Tween 80 (diluted with 0.01mol/L PBS) and the low dose group was diluted to 0.75mg/ml with 5% Tween 80 (diluted with 0.01mol/L PBS).
2. Experimental methods
2.1 pairs of CC14Effects on hepatic fibrosis in mice
2.1.1 mouse CCl4Establishing a model for inducing hepatic fibrosis injury, and injecting 20% CC1 into abdominal cavity of each group except normal group4Rapeseed oil solution (5. mu.L/g body weight) was injected twice a week.
2.2.2 taking 48 male ICR mice, randomly dividing into 4 groups, normal group, model group, high dose group (30 mg. kg. group)-1·d-1) Low dose group (15 mg. kg)-1·d-1)。
2.2.3 Normal group: no drug administration, no model making, and normal diet and drinking water; model group: intraperitoneal injection of 20% CC1 alone4Rapeseed oil solution; administration group (high, low): besides the molding, the corresponding reagent solution prepared under item 1.4 should be injected into the abdominal cavity. This experiment givesThe drug combination is administered once a day and 6 times a week from the 14 th day of molding, and orbital bleeding and mice are sacrificed on the 34 th day of molding for a total of 33 days and 20 days for the normal group, the model group and the administration group. Centrifuging at 3000r/min for 10min after blood collection, collecting supernatant, and determining CC1 by kit operation method4ALT and AST contents in the blood serum of the liver injury mouse. The mice were sacrificed after blood collection, the livers were rapidly removed, the surrounding connective tissues were removed and fixed in 4% paraformaldehyde, paraffin-embedded, 4 μm-thick serial sections were stained with sirius red, and the condition of the collagen fibers in the liver tissues was observed under an optical microscope.
3. Results of the experiment
3.1 Effect of the Compounds on liver function in mice
The compound pair CC14The results of the serum indices for the mouse liver function influencing test are shown in the following table
TABLE 1 serum index data table for effect test of compound on mouse liver function
The results show that the liver function of a mouse in the carbon tetrachloride modeling group is obviously reduced, the detection results of the blood glutamic-pyruvic transaminase and the glutamic-oxalacetic transaminase are obviously increased, and the compound can reduce the level of serum transaminase and has a protective effect on the liver function.
3.2 this Compound on CCL4Induced effects of liver histomorphology in mice
As shown in FIGS. 3-5, the staining patterns of the liver tissue of mice with the compound at different doses in the model group showed significant reticular fibrosis. Compared with the model group mouse, the high-dose group and the low-dose group which are administrated have the advantages that the pathological condition of the liver is obviously improved, and the proliferation of the liver fibrous connective tissue is obviously reduced. The gold standard of the tissue section shows that the compound has exact prevention effect on the formation of hepatic fibrosis of the mice.

Claims (9)

1. An anti-hepatic fibrosis compound, characterized by the structural formula:
2. a preparation for combating liver fibrosis, comprising a compound according to claim 1 in the form of tablets, coated tablets, dragees, hard and soft gelatine capsules, solutions, emulsions, suspensions, nasal sprays, suppositories, ointments, gels or injections.
3. A method for preparing the anti-hepatic fibrosis compound of claim 1, comprising the steps of:
adding 4-chloroacetylamidoantipyrine, 5- (1- (4-chlorophenoxy) ethyl) -4-phenyl-4H-1, 2, 4-triazole-3-thiol and potassium carbonate into acetone, refluxing, filtering, evaporating filtrate to dryness, and recrystallizing residue to obtain compound for resisting hepatic fibrosis.
4. The method for preparing a compound according to claim 3, wherein the 4-chloroacetylamidoantipyrine is prepared by adding chloroacetyl chloride and triethylamine to a dichloromethane solution containing 4-aminoantipyrine, and stirring at room temperature; then adding ice water into the solution for extraction, taking an organic layer, washing the organic layer by using a hydrochloric acid solution, then washing by using a saturated sodium bicarbonate aqueous solution, and then washing by using brine; after drying, removing the solvent to obtain a crude product; recrystallizing the crude product to obtain the 4-chloroacetylaminoaantipyrine.
5. The method for preparing a hepatic fibrosis-resistant compound according to claim 3, wherein the 5- (1- (4-chlorophenoxy) ethyl) -4-phenyl-4H-1, 2, 4-triazole-3-thiol is prepared by dissolving 2- (2- (4-chlorophenoxy) propionyl) -N-phenylhydrazine thiocarboxamide in sodium hydroxide solution, heating under reflux, cooling to pH2-3, filtering, and drying to obtain 5- (1- (4-chlorophenoxy) ethyl) -4-phenyl-4H-1, 2, 4-triazole-3-thiol.
6. The method for preparing a compound according to claim 5, wherein the 2- (2- (4-chlorophenoxy) propionyl) -N-phenylhydrazine thiocarboxamide is prepared by adding 2- (4-chlorophenoxy) propionylhydrazine and phenyl isothiocyanate to absolute ethanol, refluxing, cooling the mixture to room temperature, filtering, and concentrating the filtrate to obtain 2- (2- (4-chlorophenoxy) propionyl) -N-phenylhydrazine thiocarboxamide.
7. The method for preparing a compound according to claim 6, wherein the 2- (4-chlorophenoxy) propionohydrazide is prepared by dissolving 2- (4-chlorophenoxy) propionate in ethanol, and adding hydrazine in water; the mixture was then stirred under reflux, concentrated in vacuo, and washed to give 2- (4-chlorophenoxy) propionylhydrazide.
8. The method for preparing a compound according to claim 7, wherein the 2- (4-chlorophenoxy) propionic acid ester is prepared by dissolving 2- (4-chlorophenoxy) propionic acid in methanol, and slowly adding 98% concentrated sulfuric acid; stirring under reflux, cooling to room temperature, concentrating in vacuo, and dissolving the residue in dichloromethane, washing, drying, and removing the solvent to give 2- (4-chlorophenoxy) propionate.
9. Use of the anti-hepatic fibrosis compound of claim 1 in preparing an ATX inhibitor, an anti-hepatic fibrosis preparation or a medicament for treating liver cancer.
CN201910978213.4A 2019-10-15 2019-10-15 Anti-hepatic fibrosis compound, preparation method and application Withdrawn CN110627774A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201910978213.4A CN110627774A (en) 2019-10-15 2019-10-15 Anti-hepatic fibrosis compound, preparation method and application

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201910978213.4A CN110627774A (en) 2019-10-15 2019-10-15 Anti-hepatic fibrosis compound, preparation method and application

Publications (1)

Publication Number Publication Date
CN110627774A true CN110627774A (en) 2019-12-31

Family

ID=68975112

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201910978213.4A Withdrawn CN110627774A (en) 2019-10-15 2019-10-15 Anti-hepatic fibrosis compound, preparation method and application

Country Status (1)

Country Link
CN (1) CN110627774A (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111187261A (en) * 2020-01-15 2020-05-22 沈阳药科大学 ATX inhibitor based on indole parent nucleus and preparation method and application thereof
CN111777597A (en) * 2020-07-27 2020-10-16 江西省医学科学院 Anti-hepatic fibrosis compound, preparation method and application
CN112514898A (en) * 2020-04-13 2021-03-19 辽宁先达农业科学有限公司 Method for preparing 2- (4-chlorophenoxy) -1-propanol
CN112679337A (en) * 2020-12-30 2021-04-20 锦州四海生物化学有限公司 Preparation method of (R) - (+) -2- (4-hydroxyphenoxy) propionic acid

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111187261A (en) * 2020-01-15 2020-05-22 沈阳药科大学 ATX inhibitor based on indole parent nucleus and preparation method and application thereof
CN111187261B (en) * 2020-01-15 2021-10-26 沈阳药科大学 ATX inhibitor based on indole parent nucleus and preparation method and application thereof
CN112514898A (en) * 2020-04-13 2021-03-19 辽宁先达农业科学有限公司 Method for preparing 2- (4-chlorophenoxy) -1-propanol
CN111777597A (en) * 2020-07-27 2020-10-16 江西省医学科学院 Anti-hepatic fibrosis compound, preparation method and application
CN112679337A (en) * 2020-12-30 2021-04-20 锦州四海生物化学有限公司 Preparation method of (R) - (+) -2- (4-hydroxyphenoxy) propionic acid

Similar Documents

Publication Publication Date Title
CN110627774A (en) Anti-hepatic fibrosis compound, preparation method and application
JP2798095B2 (en) Specific aminomethylphenylimidazole derivatives, a new class of dopamine receptor subtype specific ligands
BR112018005048B1 (en) uses of kinase inhibitors useful for the preparation of pharmaceutical compositions usable in the treatment of proliferative diseases
JP2005538096A (en) Antiviral 7-deaza D-nucleosides and methods of use thereof
EP4098258A1 (en) Pharmaceutical use of ketoamide-based compound
JP6407444B2 (en) Trifluoroacetohydrazide compounds, their preparation and pharmaceutical applications
CN106946792A (en) A kind of hydroxamic acid derivs of phthalazone and preparation method and application
WO2021055589A1 (en) Heteroaryl plasma kallikrein inhibitors
WO2003031434A1 (en) Compounds and methods
CN114671920B (en) Amide pentaacetyl geniposide derivative as well as preparation method and application thereof
EP2708534B1 (en) Thiazole compound and preparation method and use thereof
JP6644814B2 (en) C, O-spiroaryl glycoside compounds and their production and use
CN105732595B (en) PAR-1 inhibitor based on terpene derivant and preparation method thereof and the purposes in treating thrombotic diseases
KR20170103803A (en) Bifunctional compounds and use for reducing uric acid levels
CN115385819B (en) Rosmarinic acid bioelectrode isostere and preparation method and application thereof
CN111943906B (en) Amidine derivatives, preparation method, pharmaceutical composition and application thereof
CN111777597A (en) Anti-hepatic fibrosis compound, preparation method and application
CN105218479A (en) The new derivatives of 2-[3-cyano-4-isobutoxy phenyl]-4-methylthiazol-5-formic acid, its preparation method and application
EP2650292B1 (en) Thiazolamine derivative and use thereof as anti-picornaviral infection medicament
JP7534390B2 (en) SARM1 inhibitors
CN115515603A (en) Bicyclic and monocyclic nucleoside analogs for the treatment of hepatitis E
US20050256087A1 (en) Phosphonic acid diester compounds
CN109134419A (en) A kind of 2,2- dimethyl -1,3- dioxolanes analog derivative, preparation method and use
CN115043825B (en) Oxadiazole compound and preparation method, application and product thereof
CN110950773B (en) Biphenyldiphenol amide derivative and application thereof as anticancer drug

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
WW01 Invention patent application withdrawn after publication
WW01 Invention patent application withdrawn after publication

Application publication date: 20191231