CN112778371B - Thienopyridine derivative and preparation method and application thereof - Google Patents

Thienopyridine derivative and preparation method and application thereof Download PDF

Info

Publication number
CN112778371B
CN112778371B CN201911072807.5A CN201911072807A CN112778371B CN 112778371 B CN112778371 B CN 112778371B CN 201911072807 A CN201911072807 A CN 201911072807A CN 112778371 B CN112778371 B CN 112778371B
Authority
CN
China
Prior art keywords
compound
formula
aqueous solution
preparation
solvent
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.)
Active
Application number
CN201911072807.5A
Other languages
Chinese (zh)
Other versions
CN112778371A (en
Inventor
杨成
陆华龙
刘晓鹏
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.)
Huachuang Synthetic Pharmaceutical Co ltd
Original Assignee
Huachuang Synthetic Pharmaceutical Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Huachuang Synthetic Pharmaceutical Co ltd filed Critical Huachuang Synthetic Pharmaceutical Co ltd
Priority to CN201911072807.5A priority Critical patent/CN112778371B/en
Publication of CN112778371A publication Critical patent/CN112778371A/en
Application granted granted Critical
Publication of CN112778371B publication Critical patent/CN112778371B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • 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 System
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/02Antithrombotic agents; Anticoagulants; Platelet aggregation inhibitors
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis

Abstract

The invention relates to a thienopyridine derivative and a preparation method and application thereof, wherein the thienopyridine derivative has a structure shown in the following formula (I), wherein the groups R1, R2, R3, R4, X1, X2 or X3 and m, n and w are defined in the claims, the invention also comprises a preparation method of the compound shown in the formula (I), medicinal salts, solvent compounds, polymorphs, enantiomers or racemic mixtures thereof, a medicinal composition containing the same and application thereof as medicines, in particular to the preparation method for preventing or treating atherosclerosis diseases, myocardial infarction, stroke, ischemic cerebral thrombosis, peripheral arterial diseases, acute coronary syndrome or thrombosis after coronary intervention.

Description

Thienopyridine derivative and preparation method and application thereof
Technical Field
The invention relates to the field of pharmaceutical chemistry, in particular to a thienopyridine derivative, a preparation method thereof and application thereof in pharmacy, and particularly relates to application of the thienopyridine derivative in preparing medicines for preventing or treating diseases related to thrombus and embolism.
Background
Thrombotic diseases are various diseases in which a blood vessel lumen is narrowed and occluded by thrombus and a main organ is ischemic and infarcted to cause dysfunction. Factors that lead to thrombosis are the adhesion and aggregation of platelets on the surface of the damaged vessel wall, blood stasis, activation of coagulation factors, promotion of thrombin formation and low fibrinolytic activity. Drugs used clinically for thrombus treatment can be classified into 3 classes: antiplatelet agents, anticoagulants and thrombolytics. The antiplatelet medicine has both therapeutic and preventive effects and is a main category of antithrombotic medicines. The antiplatelet medicine can inhibit the adhesion, aggregation and release of platelet, prevent thrombosis and prevent and treat cardiac and cerebral ischemic diseases and peripheral thromboembolic diseases. Antiplatelet drugs are currently divided into three generations: aspirin is the first generation, ticlopidine is the second generation (thiophene pyridine, a class of antiplatelet drugs with adenosine diphosphate receptor as a target point, the most widely used antiplatelet aggregation, antithrombotic drugs/non-thiophene pyridine in clinic at present), such as clopidogrel/prasugrel, and platelet membrane glycoprotein IIb/IIIa receptor antagonists are the third generation. As a second generation thiophene pyridine derivative, the P2Y12-ADP receptor antagonist clopidogrel (clopidogrel) has better safety than ticlopidine, and aspirin and clopidogrel are the standard combination of the current anti-platelet treatment and become gold standards of the anti-thrombosis drug treatment. However, the combination of the two drugs resulted in an increased bleeding rate.
Clopidogrel and prasugrel are P2Y12-ADP receptor antagonists with higher selectivity. ADP binds to platelets through 3 receptors on the platelet membrane during platelet activation: the P2X1 receptor, the P2Y1 receptor and the P2Y12 receptor play an important role. The P2Y12 receptor belongs to a member of the GPCR family, and after ADP binds to the P2Y12 receptor, the two subunits of Gi protein (αgi, βγ) are exposed, the αgi subunits reduce cAMP by inhibiting adenylate cyclase, resulting in activation of the platelet glycoprotein IIb/IIIa complex, and the βγ subunits activate phosphatidylinositol 3 kinase through a range of intracellular signaling, resulting in platelet aggregation. P2Y12-ADP receptor antagonists bind to P2Y12 receptor competitively or non-competitively, reduce the binding site of ADP, reduce platelet aggregation, and play an antithrombotic role.
Another disadvantage of clopidogrel is the resistance of clopidogrel which is of increasing concern. Clinically, clopidogrel resistance (Clopidogrel resistance) is a very common phenomenon with a rate of 4% -30% in white people, lower in african black people and highest in asian people, possibly up to 55%. After the clopidogrel resistance occurs, the consequences are very serious, and the cardiovascular event and the death rate are greatly increased. Patients with clopidogrel resistance in the treatment process are more prone to suffer from acute and subacute intrastent thrombosis, the mortality rate of thrombotic event patients is up to 15% -45%, and the rate of myocardial infarction again is up to 60% -70%.
The mechanism of the generation of clopidogrel resistance is complex, and the mechanism of the mechanism which is known to be CytochromeP450 enzyme activity. Studies have shown that clopidogrel is first absorbed into the blood from the gastrointestinal tract after oral administration. In blood, 85% of clopidogrel is directly metabolized by esterase into inactive metabolites to be discharged out of the body, wherein only 15% of clopidogrel is metabolized by CytochromeP450 enzyme, and enzymes involved in the metabolism include CYP3A4, CYP3A5, CYP2C9, CYP1A2, CYP2B6 and CYP2C19 to form thiolactone, and then the thiolactone is metabolized into metabolites with anticoagulant activity by CYP3A4 enzyme to exert anticoagulant effect. More and more researches show that the CYP450 enzyme system in the liver of a patient individual with clopidogrel resistance has weaker or absent function, so that clopidogrel cannot be metabolized into thiolactone after entering the body, and is further metabolized into active metabolites to exert the efficacy.
Aiming at the defects existing in the prior art, the invention aims to synthesize a series of novel thiophene pyridine derivative compounds by structurally modifying clopidogrel and prasugrel, the compounds are rapidly metabolized into effective metabolites after entering the body, the metabolism of CYP450 enzyme systems is successfully avoided, the active compounds can be directly metabolized to play the drug effect, and meanwhile, the thienopyridine derivatives have better water solubility and can be well mutually dissolved with clinically used diluent media (5% glucose injection and 0.9% sodium chloride injection) after being prepared into injections.
Disclosure of Invention
In order to overcome the defects in the prior art, the invention provides a compound shown in a thienopyridine derivative formula (I), a preparation method of the compound, a pharmaceutical composition containing the compound and application of the compound in preparing medicines for preventing or treating thrombosis and embolism related diseases.
The above object of the present invention is achieved by the following means.
In one aspect, the present invention provides a compound of formula (I);
wherein: n is an integer of 0 to 1, m is an integer of 0 to 1, and w is an integer of 0 to 12.
R1 is hydrogen or halogen;
r2 is hydrogen, hydroxy or C1-C10 alkyl;
r3 or R4 is independently hydrogen;
x1, X2 and X3 represent hydrogen, alkali metal, amino acid, choline and meglumine.
The invention also provides application of the compound or stereoisomers, hydrates, deuterides, solvates, crystal forms, metabolites and pharmaceutically acceptable salts thereof in preparing medicaments for preventing or treating thrombosis and embolism related diseases.
According to some embodiments of the present invention there is provided a thienopyridine derivative or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof, wherein the thienopyridine derivative comprises the following compound:
in another aspect, the present invention provides a method for preparing a compound represented by the above formula (I), the method comprising:
(a) Reacting a compound shown in a formula A with a compound shown in a formula B in the presence of inert gas, without a solvent or in an organic solvent A to form a compound shown in a formula C;
(b) Hydrolyzing the compound represented by formula C formed in step (a) with a solvent B to form a compound represented by formula D;
(c) Reacting the compound shown in the formula D obtained in the step (b) with a condensing agent, phosphate and a catalyst in a solvent C, wherein the compound is shown in the formula (I);
wherein the radicals R1, R2, R3, R4, X1, X2 or X3 and m, n and w are as defined above.
In the above preparation method, in the step (a), the inert gas is selected from one or more of nitrogen, helium and argon, preferably nitrogen or argon.
In the above preparation method, in the step (a), the organic solvent a is selected from one or more of aromatic hydrocarbons, halogenated hydrocarbons, nitriles, ketones, ethers, triethylamine, diethylamine, pyridine, 1-methylimidazole, N-diisopropylethylamine and esters, preferably ethyl acetate, acetonitrile, tetrahydrofuran, dichloromethane, toluene, acetone, triethylamine, 1-methylimidazole, pyridine or chloroform.
In the above preparation method, in step (B), the solvent B is selected from one or more of water, an aqueous alkaline solution, and an aqueous organic solvent solution. The alkaline aqueous solution is preferably sodium hydroxide aqueous solution, ammonia water, potassium hydroxide aqueous solution, sodium bicarbonate aqueous solution or sodium carbonate aqueous solution, and the organic solvent aqueous solution is selected from dichloromethane aqueous solution, acetonitrile aqueous solution, tetrahydrofuran aqueous solution or acetone aqueous solution.
In the above preparation method, in the step (C), the solvent C is selected from one or more of water, aromatic hydrocarbons, halogenated hydrocarbons, nitriles, ketones, alcohols, ethers, esters, dimethylformamide, dimethylacetamide; preferably one or more selected from the group consisting of water, ethanol, acetonitrile, acetone, tetrahydrofuran, ethyl acetate, methylene chloride, toluene and butanone.
In the above preparation method, in the step (c), the condensing agent is selected from one or more of 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride, N ' -carbonyldiimidazole, N ' -diisopropylcarbodiimide, 1, 8-diazabicyclo [5.4.0] undec-7-ene, N ' -dicyclohexyl-carbodiimide, p-toluenesulfonyl chloride.
In the above preparation method, in the step (C), the phosphate is selected from one or more of triethylamine phosphate, monopotassium phosphate, monoammonium phosphate, disodium phosphate, dipotassium phosphate, sodium dihydrogen phosphate, diammonium phosphate, polyphosphoric acid, and triethylamine pyrophosphate.
In the preparation method, in the step (C), the catalyst is selected from one or more of triphenylphosphine, 2' -dithiodipyridine, manganese chloride, zinc chloride, magnesium sulfate, magnesium chloride and calcium chloride.
In the above preparation method, in the step (a), the reaction temperature is-10 to 25 ℃, preferably-5 to 10 ℃.
In the above preparation method, in the step (b), the hydrolysis temperature is from-20 to 100 ℃, preferably from 0 to 60 ℃.
In the above preparation method, in the step (c), the reaction temperature is-10 to 40 ℃, preferably-5 to 30 ℃.
In the above preparation method, in the step (a), the molar ratio between the compound represented by the formula a and the compound represented by the formula B is 1:1.0 to 20.0, preferably 1:2.25 to 10.0.
In the above preparation method, in the step (c), the molar ratio between the compound represented by the formula D and the condensing agent is 1:1 to 10, preferably 1:4.
in the above preparation method, in the step (C), the molar ratio between the compound represented by formula E and the phosphate is 1:1 to 10, preferably 1:2 to 4.
In the above preparation method, in the step (C), the molar ratio between the compound represented by formula E and the catalyst is 1:1 to 10, preferably 1:2 to 4.
In still another aspect, the present invention further provides a pharmaceutical composition comprising a compound of formula I above and a pharmaceutically acceptable excipient.
Preferably, the pharmaceutical composition is a tablet, suppository, dispersible tablet, enteric-coated tablet, chewable tablet, orally disintegrating tablet, capsule, sugar-coated agent, granule, dry powder, oral solution, small injection needle, lyophilized powder injection or large infusion.
Preferably, the pharmaceutically acceptable excipients are selected from one or more of the following: pH adjusters, diluents, solubilizers, excipients, disintegrants, suspending agents, lubricants, binders, fillers, flavoring agents, sweeteners, antioxidants, surfactants, preservatives, coating agents and pigments.
The dosage and method of use of the compounds of the invention will depend upon a number of factors including the age, weight, sex, health condition, nutritional status, the strength of activity of the compound, the time of use, the rate of metabolism, the severity of the condition and the subjective judgment of the clinician. The dosage of the medicine is preferably between 2 and 1200mg/kg, and the dosage of the medicine is preferably between 0.2 and 300 mg/kg for 24 hours, and the medicine can also be used in a mode of multiple administration.
Compared with the prior art, the invention has at least the following beneficial effects:
firstly, the solubility of the compound shown in the formula I is better than prasugrel, clopidogrel, prasugrel Lei Linsuan ester and clopidogrel phosphate;
secondly, the compound shown in the formula I has small clinical application irritation, stronger platelet aggregation resistance and stronger platelet aggregation inhibition effect;
thirdly, the compound shown in the formula I can be well mutually dissolved with a clinically used dilution medium (5% glucose injection and 0.9% sodium chloride injection) after being prepared into an injection, and opalescence turbidity can not be generated, so that the compatibility is good; the other prasugrel Lei Linsuan ester and clopidogrel Lei Linsuan ester salts have poor solubility with 0.9% sodium chloride injection, and generate opalescence turbidity after dissolution, thus being not suitable for clinical use.
Fourth, when the thienopyridine derivatives of formula I herein are administered in vivo, they act as "prodrugs" and are converted to the biologically active parent compound in the presence of alkaline phosphatase. The thienopyridine derivatives of formula I herein have superior water solubility, making the prodrugs suitable for oral, topical and parenteral administration.
Detailed Description
The following detailed description of the invention is provided in connection with the accompanying drawings that are presented to illustrate the invention and not to limit the scope thereof.
Example 1: preparation of Compound 1
The preparation method comprises the following steps:
compound a (10.00 g,27.18 mmol) was weighed into a dry 250-mL three-necked flask, methylene chloride (100 mL) was added under nitrogen protection, the temperature was reduced to 0-5 ℃, phosphorus oxychloride (12.55 g,81.55 mmol) of compound B was slowly added, the addition was completed for about 1min, the reaction was 6-h-10 h, and the reaction was completed. The reaction was judged to be complete by in-process HPLC.
The reaction solution compound C was added dropwise to 150mL pure water at 0℃under control of hydrolysis temperature of 0℃to 5℃and stirring of 3. 3 h, the organic phase and the aqueous phase were transferred to a separating funnel for extraction, the solid phase was dissolved with methanol (150. 150 mL) and combined with the extracted organic phase, then 200 to 300 mesh silica gel was poured thereinto for about 25g, a rotary evaporator was used to evaporate the solvent and mix the sample, the evaporated silica gel was poured into a column (diameter: 4.5 cm) containing 25. 25 cm silica gel, the objective product was eluted from the column, the collected solvent was removed by rotary evaporation to give a yellow solid, 150mL of water and 50mL of methylene chloride were added, extraction was carried out, the organic phase was dried with anhydrous sodium sulfate, and then the compound D6.93 g was obtained by evaporation with a rotary evaporator, yield was 69.3%.
6.0g of compound D is placed in a dry 250mL three-necked flask, 100mL of acetonitrile is added, the mixture is stirred uniformly, 6.3g of 1-methylimidazole, 8.46g of 2,2' -dithiodipyridine and 11.58g of triphenylphosphine are added at room temperature, the mixture is reacted for 1 hour at room temperature, 1.7g of triethylamine phosphate is slowly added, and the reaction is judged whether to be finished by HPLC (high Performance liquid chromatography) in the reaction process.
The reaction mixture was evaporated to dryness under reduced pressure at 40℃and 100ml of acetone was added thereto, followed by crystallization at 0℃for 10 hours, filtration and drying to give 18.6g of (Compound 1). (moisture 0.57%)
Example 2: preparation of Compound 2
The preparation method comprises the following steps:
compound a (10.00 g,29.60 mmol) was weighed into a dry 250-mL three-necked flask, ethyl acetate (100 mL) was added under nitrogen protection, the temperature was reduced to 0-5 ℃, compound B phosphorus oxychloride (13.00 g,88.80 mmol) was slowly added, the addition was completed for about 1min, the reaction was completed for 6-10 h. The reaction was judged to be complete by in-process HPLC.
The reaction solution compound C was added dropwise to 150mL pure water at 0℃under control of hydrolysis temperature 0℃to 5℃and stirring 3. 3 h, the organic phase and the aqueous phase were transferred to a separating funnel for extraction, the solid phase was dissolved with methanol (150 mL) and combined with the extracted organic phase, then 200 to 300 mesh silica gel was poured thereinto for about 25g, a rotary evaporator was used to evaporate the solvent and mix the sample, the evaporated silica gel was poured into a column (diameter 4.5 cm) containing 25 cm silica gel, the objective product was eluted from the column, the collected solvent was removed by rotary evaporation to give a yellow solid, 150mL of water and 50mL of methylene chloride were added, extraction was carried out, the organic phase was dried with anhydrous sodium sulfate, and then 7.06g of compound D was obtained by evaporation using a rotary evaporator, yield 70.6%.
6.0g of compound D and 1.0g of triethylamine are placed in a dry 100mL three-necked flask, 50mL of dichloromethane is added, the mixture is stirred uniformly, N-carbonyl diimidazole is added at room temperature, the mixture is stirred for 2 hours for reaction, 3.0g of triethylamine phosphate, 2.4g of manganese chloride and 3.0g of magnesium sulfate are slowly added, and HPLC (high Performance liquid chromatography) judges whether the reaction is completed or not in the reaction process.
The reaction mixture was evaporated to dryness under reduced pressure at 40℃and 100ml of acetone was added thereto, followed by crystallization at 0℃for 10 hours, filtration and drying to give 13.2g of (Compound 2). (moisture 0.99%)
Example 3: preparation of Compound 4
10g of (compound 2) is placed in a 250ml reaction bottle, 160ml of methanol, 0.85g of sodium hydroxide and the mixture are added, stirred and dissolved at 50 ℃, filtered, cooled to 0 ℃ for crystallization for 12 hours, filtered and dried at 40 ℃ for 4 hours, thus obtaining 12.3g of (compound 4). (moisture: 3.06%).
Example 4: preparation of Compound 5
The preparation method comprises the following steps:
compound a (10.00 g,27.69 mmol) was weighed into a dry 250-mL three-necked flask, ethyl acetate (100 mL) was added under nitrogen protection, the temperature was reduced to 0-5 ℃, compound B phosphorus oxychloride (12.65 g,83.07 mmol) was slowly added, the addition was completed for about 1min, the reaction was completed for 6-10 h. The reaction was judged to be complete by in-process HPLC.
The reaction solution compound C was added dropwise to 150mL pure water at 0℃under control of hydrolysis temperature of 0℃to 5℃and stirring of 3. 3 h, the organic phase and the aqueous phase were transferred to a separating funnel for extraction, the solid phase was dissolved with methanol (150. 150 mL) and combined with the extracted organic phase, then 200 to 300 mesh silica gel was poured thereinto for about 25g, a rotary evaporator was used to evaporate the solvent and mix the sample, the evaporated silica gel was poured into a column (diameter: 4.5 cm) containing 25. 25 cm of silica gel, the objective product was eluted from the column, the collected solvent was removed by rotary evaporation to give a yellow solid, 150mL of water and 50mL of methylene chloride were added, extraction was carried out, the organic phase was dried with anhydrous sodium sulfate, and then the compound D6.56 g was obtained by evaporation with a rotary evaporator, yield was 65.6%.
6.0g of compound D is placed in a dry 250mL three-necked flask, 100mL of acetonitrile is added, the mixture is stirred uniformly, 6.2g of 1-methylimidazole, 8.16g of 2,2' -dithiodipyridine and 11.88g of triphenylphosphine are added at room temperature, the mixture is reacted for 1 hour at room temperature, 1.8g of triethylamine phosphate is slowly added, and the reaction is judged whether to be finished by HPLC (high Performance liquid chromatography) in the reaction process.
The reaction mixture was evaporated to dryness under reduced pressure at 40℃and 100ml of acetone was added thereto, followed by crystallization at 0℃for 10 hours, filtration and drying to give 12.6g of (Compound 5). (moisture 1.13%)
Example 5: preparation of Compound 8
The preparation method comprises the following steps:
compound a (10.00 g,27.69 mmol) was weighed into a dry 250-mL three-necked flask, ethyl acetate (100 mL) was added under nitrogen protection, the temperature was reduced to 0-5 ℃, compound B phosphorus oxychloride (12.65 g,83.07 mmol) was slowly added, the addition was completed for about 1min, the reaction was completed for 6-10 h. The reaction was judged to be complete by in-process HPLC.
The reaction solution compound C was added dropwise to 150mL pure water at 0℃under control of hydrolysis temperature of 0℃to 5℃and stirring of 3. 3 h, the organic phase and the aqueous phase were transferred to a separating funnel for extraction, the solid phase was dissolved with methanol (150. 150 mL) and combined with the extracted organic phase, then 200 to 300 mesh silica gel was poured thereinto for about 25g, a rotary evaporator was used to evaporate the solvent and mix the sample, the evaporated silica gel was poured into a column (diameter: 4.5 cm) containing 25. 25 cm of silica gel, the objective product was eluted from the column, the collected solvent was removed by rotary evaporation to give a yellow solid, 150mL of water and 50mL of methylene chloride were added, extraction was carried out, the organic phase was dried with anhydrous sodium sulfate, and then the compound D6.56 g was obtained by evaporation with a rotary evaporator, yield was 65.6%.
6.0g of compound D is placed in a dry 250mL three-necked flask, 100mL of acetonitrile is added, the mixture is stirred uniformly, 6.2g of 1-methylimidazole, 8.16g of 2,2' -dithiodipyridine and 11.88g of triphenylphosphine are added at room temperature, the mixture is reacted for 1 hour at room temperature, 1.8g of triethylamine phosphate is slowly added, and the reaction is judged whether to be finished by HPLC (high Performance liquid chromatography) in the reaction process.
The reaction mixture was evaporated to dryness under reduced pressure at 40℃and 100ml of acetone was added thereto, followed by crystallization at 0℃for 10 hours, filtration and drying to give 11.6g of a solid.
Putting 10g into a 250ml reaction bottle, adding 160ml of methanol and 2.15g of sodium hydroxide, stirring and dissolving at 50 ℃, filtering, cooling to 0 ℃ for crystallization for 12 hours, filtering, and drying at 40 ℃ for 4 hours to obtain (compound 4) 8.5 g. (moisture: 2.12%).
Example 6: preparation of Compound 9
20g of (compound 5) is placed in a 250ml reaction bottle, 160ml of methanol, 15.5g of choline hydroxide methanol solution (0.2 g/g) is added, stirred and dissolved at 50 ℃, filtered, cooled to 0 ℃ for crystallization for 12 hours, filtered and dried at 40 ℃ for 4 hours, and 16.7g of (compound 9) is obtained. (moisture: 7.17%).
Example 7: preparation of Compound 10
Putting 20g of compound 2 into a 250ml reaction bottle, adding 160ml of 20% methanol aqueous solution, stirring and dissolving at 50 ℃, filtering, cooling to 0 ℃ for crystallization for 12 hours, filtering, and drying at 40 ℃ until the water content is within the range of 9-10%, thus obtaining 15.6g of (compound 10). (moisture: 15.33%).
Example 8: preparation of tablets
Prescription:
compound 2 50g
Starch 20g
Microcrystalline cellulose 30g
5% povidone K30 10g
Magnesium stearate 1g
The preparation method comprises the following steps: crushing the compound 2 and sieving the crushed compound with a 80-mesh sieve; weighing starch with a prescription amount, compound 2 with a prescription amount and microcrystalline cellulose, and uniformly mixing. The materials are made into soft materials by 5 percent povidone K30 solution, and are granulated by a 20-mesh sieve, and are dried at the temperature of 40 to 60 ℃ until the moisture in the granules is about 5 percent. Sieving with 20 mesh sieve, granulating, adding magnesium stearate with prescribed amount, mixing, measuring intermediate content, and weighing; tabletting.
Example 9: preparation of granule
Prescription:
compound 4 50g
Starch 20g
Sucralose 30g
5% povidone K30 10g
The preparation method comprises the following steps: crushing the compound 4 and sieving the crushed compound with a 80-mesh sieve; weighing starch with a prescription amount, compound 4 with a prescription amount and sucralose, and uniformly mixing. The materials are made into soft materials by 5 percent povidone K30 solution, and are granulated by a 20-mesh sieve, and are dried at the temperature of 40 to 60 ℃ until the moisture in the granules is about 5 percent. Sieving with 20 mesh sieve, grading, measuring intermediate content, and bagging.
Example 10: preparation of Compound 4 for injection
Prescription:
compound 4 100g
PH regulator 4.0-9.0
Glucose 30g
Water and its preparation method 2000ml
The preparation method comprises the following steps: adding water for injection in batch volume, weighing compound 4 and glucose in prescription amount, stirring to dissolve thoroughly, adjusting pH to 4.0-9.0 with pH regulator, filtering with 0.22um microporous membrane, and packaging; freeze drying, capping and packaging.
EXAMPLE 11 preparation of Compound 9 for injection
Prescription:
compound 9 100g
PH regulator 4.0-9.0
Excipient 20g
Water and its preparation method 2000ml
The preparation method comprises the following steps: adding water for injection in batch volume, weighing compound 9 and excipient in prescription amount, stirring to dissolve thoroughly, adjusting pH to 4.0-9.0 with pH regulator, filtering with microporous membrane of 0.22um, and packaging; freeze drying, capping and packaging.
Comparative example 1: preparation of Compound C of example 4 of patent application CN200910023773.0
37.3 g (0.1 mol) of 2- [2- (hydroxy) -6, 7-dihydrothieno [3,2-c ] pyridine-5 (4H) -yl ] -1-cyclopropyl-2- (2-fluorophenyl) ethanone is dissolved in 2000ml of ethyl acetate, 30.6 (0.2 mol) of phosphorus oxychloride is dropwise added under ice bath cooling, the reaction is carried out for 5 hours at room temperature, 50ml of distilled water is slowly added under ice bath cooling, the reaction is stirred for 1 hour, concentrated sodium hydroxide solution is slowly dropwise added to adjust the pH to about 4, the mixture is evaporated to dryness, and the methanol is desalted and evaporated again to obtain the compound C20.8g.
Comparative example 2: preparation of Compound V of example 1 of patent application CN201210355789.3
Synthetic scheme
Step 1: synthesis of (R) - (2-chloro-phenyl) -hydroxy-acetic acid methyl ester (2)
To a stirred solution of compound 1 (18.6 g,0.1 mol) in methanol (100 mL) was added concentrated sulfuric acid (2 mL).
The mixture was then heated at reflux, 3 times better, and excess methanol was removed under vacuum. The oily residue was taken up in 200mL of dichloromethane, then washed with 10% aqueous potassium carbonate (240 mL), the organic solvent extracted, dried and then concentrated under vacuum to give compound 2 (19 g, 95%) as a colourless oil.
Step 2: synthesis of (R) - (2-chloro-phenyl) - (4-nitro-benzenesulfonyloxy) -acetic acid methyl ester (4)
/>
To a stirred solution of compound 2 (12 g,60 mmol) and TEA (10.7 mL) in anhydrous DCM (150 mL) at 0deg.C and N2 was added dropwise a solution of compound 3 (19.6 g,90 mmol) in DCM (50 mL). The mixture was then stirred for 3 hours. The reaction was quenched with water (100 mL), the organic layer was separated, washed with brine, dried over anhydrous Na2SO4, and then concentrated to dryness. The residue was purified by column on silica gel (PE: ea=100:1-10:1) to give compound 4 (13 g, 56%).
Step 3: (S) - (2-chloro-phenyl) - (2-hydroxy-6, 7-dihydro-4H-thiophene [3,2-c ] picoline
Synthesis of methyl pyridin-5-yl) -acetate (6)
Compound 4 (9.8 g,25.4 mmol), compound 5 (6.1 g,31.8 mmol) and N2 were stirred at room temperature
A mixture of KHCO3 (6.36 g,63.6 mmol) in anhydrous CH3CN (50 mL) for 15h. The salt was then turned over and the filtrate was concentrated to dryness. The residue was purified by silica gel column chromatography (PE: ea=50:1-3:1) to give compound 6, which was recrystallized from ethanol to give a white solid (2.3 g, 54%) (percent enantiomeric excess: about 80%). Chemically pure compound 6 was isolated by SFC to give an optically pure product (1.71 g) (percent enantiomeric excess: 100%) as a white solid.
Step 4: (S) - (2-chloro-phenyl) - [2- (dimethoxy-phosphoryloxy) -6, 7-dihydro-4H-thiophene
Synthesis of [3,2-c ] pyridin-5-yl ] -acetic acid methyl ester (8)
To a stirred solution of compound 6 (500 mg,1.48 mmol) and TEA (600 mg,5.92 mmol) in anhydrous DCM (5 mL) at 0℃and N2 was added dropwise compound 7 (265 mg,1.84 mmol). The mixture was then stirred at room temperature for 12 hours and the reaction quenched with water. The organic layer was washed with NaHCO3 solution and brine, dried over anhydrous Na2SO4, and concentrated to dryness. The residue was purified by column on silica gel (PE: ea=30:1-1:1) to give compound 7 (248 mg, 38%) as a white solid.
Step 5: (S) - (2-chloro-phenyl) - (2-phosphonooxy-6, 7-dihydro-4H-thiophene [3,2-c ] pyridine
Synthesis of methyl pyridin-5-yl) -acetate (Compound of formula V)
To a solution of compound 8 (450 mg,1.01 mmol) in anhydrous DCM (10 mL) at room temperature and N2 was added
TMSBr (1.54 mg,10.1 mmol). The mixture was then stirred for 2 hours and the solvent was removed under reduced pressure. The residue was redissolved in methanol and concentrated to dryness. The residue was isolated by HPLC (TFA) system to give the compound of formula V (110 mg, 26%) as a white solid.
Comparative example 3: preparation of Compound VI of example 2 of patent application CN201210355789.3
The preparation scheme is as follows:
step 1: synthesis of (S) - (2-chloro-phenyl) - [2- (di-tert-butoxy-phosphoryloxy-methoxy) -6, 7-dihydro-4H-thiophene [3,2-c ] pyridin-5-yl ] -acetic acid methyl ester (10)
Compound 6 (480 mg,1.41 mmol) prepared in step 3 of example 1 and at-78℃and N2
LHMDS (4.23 mL, 1.0M in THF, 2.85 mmol) was added dropwise to a stirred solution of NaI (430 mg,2.82 mmol) in anhydrous THF (3 mL), then the mixture was stirred at room temperature for 30 min, after which compound 9 (540 mg,2.11 mmol) was added and stirred for an additional 10 h. The reaction was quenched with NH4Cl solution and extracted with EA. The organic layer was washed with NaHCO3 solution and brine, dried over anhydrous Na2SO4, and concentrated to dryness. The residue was purified by column on silica gel (PE: ea=50:1-3:1) to give compound 10 (52 mg, 7%) as a white solid.
Step 2: synthesis of (S) - (2-chloro-phenyl) - (2-phosphonooxymethoxy-6, 7-dihydro-4H-thiophene [3,2-c ] pyridin-5-yl) -acetic acid methyl ester (Compound of formula VI)
To a solution of compound 10 (900 mg,1.61 mmol) in anhydrous DCM (5 mL) at 0deg.C was added
TFA (3 mL). The mixture was then stirred for 30 minutes, the dissolution removed under reduced pressure, and concentrated to dryness. The residue was isolated by HPLC (TFA) system to give the compound of formula VI (98 mg, 14%) as a white solid.
Comparative example 4: preparation of Compound I-2 of example 5 of patent application CN201010624329.7
Methyl (2S) -2- (2-chlorophenyl) -2- (2-oxo-5, 6,7 a-tetrahydrothieno [3,2-c ] pyridyl) acetate (IV-1) (6.5 g) was reacted with acetic anhydride (3.6 ml) to prepare methyl (S) -2- (2-acetoxy-6, 7-dihydrothieno [3,2-c ] pyridin-5 (4H) -yl) -2- (2-chlorophenyl) -acetate (I-2) (6.8 g),
yield 93%, white solid obtained by ethanol recrystallization, mp: 73. ee=98.9% (chiral HPLC analysis conditions: chiralpak IC 4.6 mm. Times.250 mm; column temperature: 25 ℃ C.; mobile phase: 92% n-hexane/8% tetrahydrofuran/0.1% diethylamine; flow rate: 0.5 ml/min; detection wavelength: UV 254 nm), [ alpha ]]D23 = +45.00° (c = 1.0,CH 3 OH) ;
Comparative example 5: preparation of Compound A of example 1 of patent application CN200910023773.0
The reaction equation:
37.3 g (0.1 mol) of 2- [2- (hydroxy) -6, 7-dihydrothieno [3,2-c ] pyridin-5 (4H) -yl ] -1-cyclopropyl-2- (2-fluorophenyl) ethanone are dissolved in 2000ml of ethyl acetate, 26.3g (0.1 mol) of triphenylphosphine and 17.52g (0.1 mol) of BOC-glycine are added, 17.42g (0.1 mol) of diethyl azodicarboxylate (DEAD) are added dropwise with stirring and reacted at room temperature for 5H, filtration is carried out, dry HCl gas is introduced into the filtrate until saturation, stirring is carried out at room temperature for 5H, a large amount of white solid is precipitated, filtration is carried out, and the filter cake is washed with a small amount of ethyl acetate and dried to obtain 13.8g of compound A hydrochloride.
After 10g of compound hydrochloride was dissolved in 100ml of purified water, a saturated sodium carbonate solution was slowly dropped to a pH of 10 to 10.5, and 500ml of ethyl acetate was used for extraction three times each time, the extracts were combined, dried over anhydrous sodium sulfate and evaporated under reduced pressure to obtain 5g of compound A.
Comparative example 6: synthesis of 5- ((S) - (methoxycarbonyl) (2-chlorophenyl) methyl) -4,5,6, 7-tetrahydrothieno [3,2-c ] pyridin-2-yl 2, 6-diaminohexanoic acid (Compound 1-1) of example 1 of patent application CN201510518276.3
N alpha-t-Butoxycarbonyl-N epsilon-t-Butoxycarbonyl-L-lysine (22 mmol,7.6 g), N' -dicyclohexylcarbodiimide (4.4 mmol,0.91 g), 4-dimethylaminopyridine (2.2 mmol,0.3 g) were dissolved in anhydrous dichloromethane (30 ml) under water bath. Followed by addition of (S) - (2-chloro-phenyl) - (2-hydroxy-6, 7-dihydro-4H-thiapin [3,2-c ] pyridin-5-yl) -acetic acid methyl ester (26 mmol,8.76 g) and stirring overnight at ambient temperature. Filtering to remove solid insoluble substances, and evaporating filtrate to dryness; diethyl ether was added to the residue and insoluble impurities in the solution were removed by filtration. Concentrating the filtrate, rapidly passing through column with diethyl ether as developing agent, and collecting product fraction. Concentrating under reduced pressure to obtain 6.5g of product; the yield of compound 1 was 74.2%.
Comparative example 7: example 1 Synthesis of Compound 1 of patent application CN 201610216287.0
The chloroform solution of 45g chloroacetic acid was cooled with an ice bath to lower the temperature of the solution to below 5 ℃, then 11.5 g of compound (II) was added in portions, the internal temperature was controlled to not exceed 10 ℃, the bath temperature was naturally raised to room temperature after the addition was completed, and the reaction was stirred for 4 hours, insoluble matters were filtered off, chloroform was washed with a small amount of chloroform, chloroform solutions were combined, washed with water twice, dried over anhydrous sodium sulfate, and concentrated to dryness. The residue was separated by HPLC to give compound 1.
Experimental example 1: solubility study
The compounds of the present invention, comparative example 1, comparative example 2, comparative example 3, comparative example 4, prasugrel, clopidogrel bisulfate were respectively tested for solubility in water (25 ℃) and the test results are shown in the following tables.
Solubility data of test compounds
Compounds of formula (I) Solubility of
Compound 1 52mg/ml
Compound 2 80mg/ml
Compound 3 95mg/ml
Compound 4 160mg/ml
Compound 5 85mg/ml
Compound 6 90mg/ml
Compound 7 85mg/ml
Compound 8 180mg/ml
Compound 9 132mg/ml
Compound 10 72mg/ml
Comparative example 1 Less than 30mg/ml
Comparative example 2 Less than 30mg/ml
Comparative example 3 Less than 30mg/ml
Comparative example 4 Less than 30mg/ml
Comparative example 5 Less than 30mg/ml
Comparative example 6 Less than 30mg/ml
Comparative example 7 Less than 30mg/ml
Prasugrel Less than 5mg/ml
Clopidogrel bisulfate Less than 30mg/ml
From the experimental results: the compound of the invention has higher solubility, are all higher than the solubility of comparative example 1, comparative example 2, comparative example 3, comparative example 4, comparative example 5, comparative example 6, comparative example 7, prasugrel, clopidogrel bisulfate in water. The same drug loading amount, the solubility of the compound of the present invention is better than that of comparative examples 1, 2, 3, 4,5,6,7, prasugrel and clopidogrel, and thus the compound of the present invention is easier to prepare into parenteral formulations without adding a cosolvent to improve the water solubility thereof.
Experimental example 2: research of re-dissolution experiment
The compound of the present invention was tested for reconstitution with comparative example 1, comparative example 2, comparative example 3, comparative example 4, comparative example 5, comparative example 6, comparative example 7 in 0.9% sodium chloride injection (50-100 ml), respectively. The test results are shown in the following table.
Comparative reconstitution results of Compounds in 50ml of 0.9% sodium chloride injection
Comparative reconstitution results of Compounds in 100ml of 0.9% sodium chloride injection
From the experimental results: the compound and the medicinal salt thereof prepared by the invention are still colorless and clear after being placed in a 0.9% sodium chloride solution for 8 hours, but comparative examples 1, 2, 3, 4,5,6 and 7 are obviously turbid after being placed for 2 hours, and a large amount of sediment is generated after being placed for 8 hours, so that the result shows that the compound of the invention can be well dissolved in a 0.9% sodium chloride injection, does not generate turbid sediment, is beneficial to clinical application, reduces clinical medication risk and has high safety coefficient.
Experimental example 3: the experimental examples are given in the following table for the experimental experiments of the influence factors of the compounds of the present invention.
Influence factor test results
Conclusion: the compound prepared by the invention has no obvious change in various investigation indexes compared with 0 day after being placed for 10 days under the conditions of high temperature, high humidity and illumination, which proves that the compound has stable property.
Experimental example 4: the stability test of the compounds of the present invention and the comparative examples at room temperature is shown in the following table.
Stability test results
From the experimental results, the stability of the compound of the invention is better than that of comparative examples 1, 3, 4 and 7, and the compound is favorable for the storage of industrial production.
Experimental example 5: anti-platelet aggregation Activity assay
Medicine and preparation: the positive medicine is clopidogrel sulfate and prasugrel. The inventive compounds were prepared as in comparative examples 1, 2, 3, 4,5,6,7 (prepared as in the above examples).
Suspensions were formulated for administration to animals in 0.5% CMC-Na (sodium carboxymethyl cellulose).
Animals: male SD rats, weighing about 250 g.
Instrument: centrifuge (80-2 bench low speed centrifuge) and full-automatic platelet aggregation tester
(STELLEXLG-PAPER-1 platelet aggregation analyzer) and the like.
The method comprises the following steps: the compounds of the present invention were tested for pharmacological activity against platelet aggregation by reference to the BORN turbidimetry (Nature, 1962, 194 (4832): 927). Platelet Rich Plasma (PRP) was stirred with the addition of the pro-coagulant Adenosine Diphosphate (ADP) to aggregate the platelets. The aggregation of platelets causes a change in optical density, which can be detected by a spectrophotometer. This experiment allows to evaluate the platelet aggregation effect of the test compounds administered in vivo or in vitro.
Anti-platelet aggregation Activity assay: male SD rats, weighing about 250g, were given clopidogrel sulfate and test drug (uniform suspension of 0.5% CMC-Na, drug concentration 1 mg/ml) by oral gavage, and the same volume of 0.5% CMC-Na by oral gavage in a blank group at a dose of 10mg/kg or 3 mg/kg. After 2 hours, the orbit was bled, 3.8% sodium citrate was anticoagulated, and the whole blood to anticoagulant ratio was 9:1, centrifuged at 1000rpm for 7min to prepare Platelet Rich Plasma (PRP). PRP was modulated with Platelet Poor Plasma (PPP) to maintain platelet counts at 2x 106/ml. PRP was added to a test cup and incubated at 37℃for 10min, zeroed with PRP and 100% with PPP, ADP (final concentration 5. Mu.M) as inducer, and the percent aggregation of platelets was determined by turbidimetry with a platelet aggregation meter and statistically compared by t-test. The platelet aggregation inhibition rate was calculated as follows: platelet aggregation inhibition (%) = [1- (percent drug administration tube aggregation/percent control tube aggregation) ] x100%.
TABLE 1 platelet aggregation inhibition after oral administration of test compounds to rats
Results: platelet aggregation inhibition by nephelometry after oral administration of the test compounds was measured in rats, and some of the experimental results are shown in table 1. The results show that most of the tested compounds showed stronger anti-platelet aggregation activity than clopidogrel bisulfate, prasugrel, comparative example 1, comparative example 2, comparative example 3, comparative example 4, comparative example 5, comparative example 6, comparative example 7, and had stronger platelet aggregation inhibition effect.
Experimental example 6: vascular irritation test of intravenous administration of Compounds of the invention to mice
The vascular irritation of the compounds of the present invention was experimentally compared with those of the compounds prepared in comparative examples 1 to 7.
Method and results
60 healthy intact rabbits are taken, the compound of the invention is intravenously injected into one side margin of each rabbit according to a sterile operation method, the equal volume of sterilized normal saline is intravenously injected into the other side margin as a control, the administration volume is 1ml/kg (corresponding to clinical administration dose), the intravenous injection speed is 1 ml/min, and the intravenous injection is carried out for 3 consecutive days and 1 time per day (3 administration). Before each administration, the situation of venous stasis, bleeding, edema, necrosis and the like of the vein and surrounding tissues at the edge of the ear is observed, then the carotid artery is exsanguinated to kill the rabbit, the rabbit ear is cut off from the auricular root part, the rabbit ear is fixed by 10% formalin, pathological section examination is carried out, and whether the endothelial cells of the vein at the edge of the ear are denatured and necrotic, congestion or thrombosis exists in the lumen, inflammatory cell infiltration exists in the wall of the tube and the surrounding tissues and the like are observed.
The results show that: after intravenous injection of the compound of the present invention with the compounds prepared in comparative examples 1 to 7 and sterilized physiological saline for 3 consecutive days from the rabbit ear vein, no obvious pathological changes were observed in the rabbit ear and surrounding tissues except for the injection site, as observed visually during the administration period and 24 hours after the last administration.
Conclusion: the compound of the invention has no obvious stimulation effect on the rabbit ear vein after continuous intravenous injection for 3 days, and the compounds prepared in comparative examples 1-7 have obvious stimulation effect on the rabbit ear vein after continuous intravenous injection for 3 days.

Claims (8)

1. A thienopyridine derivative characterized by the following structural formula:
2. a thienopyridine derivative characterized by the following structural formula:
3. a process for the preparation of a thienopyridine derivative according to claim 1 or 2, characterized in that the process comprises:
(a) Reacting a compound shown in a formula A with a compound shown in a formula B in the presence of inert gas, without a solvent or in an organic solvent A to form a compound shown in a formula C;
formula A is:
formula B is:
formula C is:
(b) Hydrolyzing the compound represented by formula C formed in step (a) with a solvent B to form a compound represented by formula D;
formula D is:
(c) Reacting the compound shown in the formula D obtained in the step (b) with a condensing agent, phosphate and a catalyst in a solvent C to obtain the following compound:
4. a method of preparing according to claim 3, wherein in step (a), the inert gas is nitrogen or argon;
in step (a), the organic solvent a is ethyl acetate, acetonitrile, tetrahydrofuran, dichloromethane, toluene, acetone, triethylamine, 1-methylimidazole, pyridine or chloroform;
in step (B), the solvent B is selected from one or more of water, an aqueous alkaline solution, and an aqueous organic solvent solution;
the alkaline aqueous solution is sodium hydroxide aqueous solution, ammonia water, potassium hydroxide aqueous solution, sodium bicarbonate aqueous solution or sodium carbonate aqueous solution, and the organic solvent aqueous solution is dichloromethane aqueous solution, acetonitrile aqueous solution, tetrahydrofuran aqueous solution or acetone aqueous solution;
in step (C), the solvent C is selected from one or more of water, ethanol, acetonitrile, acetone, tetrahydrofuran, ethyl acetate, dichloromethane, toluene, and butanone;
in step (c), the condensing agent is selected from one or more of 1-ethyl- (3-dimethylaminopropyl) carbodiimide hydrochloride, N ' -carbonyldiimidazole, N ' -diisopropylcarbodiimide, 1, 8-diazabicyclo [5.4.0] undec-7-ene, N ' -dicyclohexylcarbodiimide;
in step (c), the phosphate is selected from one or more of triethylamine salt, potassium dihydrogen phosphate, ammonium dihydrogen phosphate, disodium hydrogen phosphate, dipotassium hydrogen phosphate, sodium dihydrogen phosphate, diammonium hydrogen phosphate, polyphosphoric acid, triethylamine salt of pyrophosphoric acid;
in the step (c), the catalyst is selected from one or more of manganese chloride and zinc chloride.
5. The process according to claim 3 or 4, wherein in step (a), the reaction temperature is-10 ℃ to 25 ℃;
in the step (b), the hydrolysis temperature is-20-100 ℃;
in step (c), the reaction temperature is-10 to 40 ℃.
6. The production method according to claim 3 or 4, wherein in the step (a), a molar ratio between the compound represented by the formula a and the compound represented by the formula B is 1:1.0 to 20.0;
in the step (c), the molar ratio between the compound shown in the formula D and the condensing agent is 1:1-10;
in the step (c), the molar ratio between the compound shown in the formula D and the phosphate is 1:1-10;
in the step (c), the molar ratio between the compound shown in the formula D and the catalyst is 1:1-10.
7. Use of a compound according to claim 1 or 2 for the manufacture of a medicament for thrombo-and embolic related diseases.
8. A pharmaceutical composition comprising a compound according to claim 1 or 2 and a pharmaceutically acceptable adjuvant; the pharmaceutical composition is a tablet, a suppository, a dispersible tablet, an enteric coated tablet, a chewable tablet, an orally disintegrating tablet, a capsule, a sugar-coated agent, a granule, a dry powder, an oral solution, a small-volume injection, a freeze-dried powder injection for injection or a large infusion; the pharmaceutically acceptable auxiliary materials are selected from one or more of the following: pH adjusters, diluents, disintegrants, suspending agents, excipients, lubricants, binders, fillers, flavoring agents, sweeteners, antioxidants, preservatives, encapsulating agents and pigments.
CN201911072807.5A 2019-11-05 2019-11-05 Thienopyridine derivative and preparation method and application thereof Active CN112778371B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201911072807.5A CN112778371B (en) 2019-11-05 2019-11-05 Thienopyridine derivative and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN201911072807.5A CN112778371B (en) 2019-11-05 2019-11-05 Thienopyridine derivative and preparation method and application thereof

Publications (2)

Publication Number Publication Date
CN112778371A CN112778371A (en) 2021-05-11
CN112778371B true CN112778371B (en) 2024-01-30

Family

ID=75747524

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201911072807.5A Active CN112778371B (en) 2019-11-05 2019-11-05 Thienopyridine derivative and preparation method and application thereof

Country Status (1)

Country Link
CN (1) CN112778371B (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102002053A (en) * 2009-09-02 2011-04-06 陕西合成药业有限公司 Tetrahydro thienopyridine derivative for treating
CN103665042A (en) * 2012-09-21 2014-03-26 北京普禄德医药科技有限公司 Optically active 2-hydroxyltetrahydrothienopyridine derivative as well as preparation method and use thereof
CN104418891A (en) * 2013-08-28 2015-03-18 江苏威凯尔医药科技有限公司 Preparation method and medical application of water-soluble 2-hydroxyl tetrahydrothienopyridine derivatives
CN104447867A (en) * 2013-09-17 2015-03-25 天士力控股集团有限公司 Thienopiperidine derivative, preparation method and application thereof
CN107266497A (en) * 2016-04-08 2017-10-20 陕西合成药业股份有限公司 A kind of purposes of phosphoryl carboxylic acid derivative its preparation method and Pharmaceutical composition

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN102002053A (en) * 2009-09-02 2011-04-06 陕西合成药业有限公司 Tetrahydro thienopyridine derivative for treating
CN103665042A (en) * 2012-09-21 2014-03-26 北京普禄德医药科技有限公司 Optically active 2-hydroxyltetrahydrothienopyridine derivative as well as preparation method and use thereof
CN104418891A (en) * 2013-08-28 2015-03-18 江苏威凯尔医药科技有限公司 Preparation method and medical application of water-soluble 2-hydroxyl tetrahydrothienopyridine derivatives
CN104447867A (en) * 2013-09-17 2015-03-25 天士力控股集团有限公司 Thienopiperidine derivative, preparation method and application thereof
CN107266497A (en) * 2016-04-08 2017-10-20 陕西合成药业股份有限公司 A kind of purposes of phosphoryl carboxylic acid derivative its preparation method and Pharmaceutical composition

Also Published As

Publication number Publication date
CN112778371A (en) 2021-05-11

Similar Documents

Publication Publication Date Title
FI87216C (en) Process for the preparation of a right-turning enantiomer of methyl alpha-5- (4,5,6,7-tetrahydro (3,2-c) thienopyridyl) - (2-chlorophenyl) acetate
KR101453181B1 (en) Thienopyridine ester derivative containing cyano group, preparation method, use and composition thereof
CA2808520C (en) Methods for the treatment or prophylaxis of thrombosis or embolism
KR20070052780A (en) Crystalline clopidogrel hydrobromide and processes for preparation thereof
CN101402641A (en) Oxime derivatives containing thienopyridine, preparation method and application thereof
JP2021534106A (en) Substituted thienopyrol as a PAD4 inhibitor
WO2011035077A1 (en) Novel compounds and therapeutic use thereof for protein kinase inhibition
JP2003246735A (en) Medicine comprising acid addition salt of hydropyridine derivative
KR20110045034A (en) Prasugrel hydrogen sulfate and pharmaceutical compositions and uses thereof
CN112778371B (en) Thienopyridine derivative and preparation method and application thereof
CN106554368A (en) Thienopyridine analog derivative and its production and use
CN106554303A (en) Thienopyridine analog derivative and its production and use
CN106467546A (en) Clopidogrel derivant and its production and use
AU2016282124B2 (en) Deuterated thienopiperidine derivatives, manufacturing method, and application thereof
KR101552691B1 (en) Novel anti-platelet compound addition salt
CN102268012B (en) Thiazole derivative and preparation method and application thereof
WO2005021481A1 (en) N, n’-dibenzyl ethylenediamine salt pf 2-(alpha-hidroxypentyl) benzoic acid and its preparing process and usage
CN102796093B (en) Thiomorpholine-containing pyrrole derivatives and their preparation method and use
JP2020533401A (en) Thienopyridine derivative containing unsaturated aliphatic olefinic bond, its preparation method and use
CN102351878B (en) Isoxazole derivatives as well as preparation method and application thereof
JP3049284B2 (en) Hydantoin derivatives and preventive and therapeutic agents for diabetic complications and cardiovascular diseases using the same as active ingredients
CN106554302A (en) Thienopyridine analog derivative and its production and use
RU2526624C2 (en) Thienopyridine ester derivative, containing cyanogroup, method of its obtaining, its application and based on it composition
JPH11180980A (en) 1-furoyl-substituted piperazine derivative, medicine containing the same as active ingredient and intermediate for producing the same
KR100742134B1 (en) Pharmaceutical composition comprising crystalline s-(+)-methyl-2-(2-chlorophenyl)-2-(6,7-dihydrothieno[3,2-c]pyridin-5(4h)-yl)acetate.camsylate

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
GR01 Patent grant
GR01 Patent grant