WO2014012236A1 - Dabigatran etexilate derivative and preparation method and application thereof - Google Patents

Dabigatran etexilate derivative and preparation method and application thereof Download PDF

Info

Publication number
WO2014012236A1
WO2014012236A1 PCT/CN2012/078897 CN2012078897W WO2014012236A1 WO 2014012236 A1 WO2014012236 A1 WO 2014012236A1 CN 2012078897 W CN2012078897 W CN 2012078897W WO 2014012236 A1 WO2014012236 A1 WO 2014012236A1
Authority
WO
WIPO (PCT)
Prior art keywords
formula
derivative
pharmaceutically acceptable
compound
acceptable salt
Prior art date
Application number
PCT/CN2012/078897
Other languages
French (fr)
Chinese (zh)
Inventor
王志岩
Original Assignee
北京普禄德医药科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 北京普禄德医药科技有限公司 filed Critical 北京普禄德医药科技有限公司
Priority to PCT/CN2012/078897 priority Critical patent/WO2014012236A1/en
Publication of WO2014012236A1 publication Critical patent/WO2014012236A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • 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

Definitions

  • the present invention relates to the field of medicine, and in particular, the present invention relates to an ester derivative of dabigatran having thrombin inhibitor activity, a process for preparing the derivative, a pharmaceutical composition comprising the derivative, and the Use of derivatives and pharmaceutical compositions in the preparation of thrombin inhibitors and treatment related diseases.
  • Scenery technology relates to an ester derivative of dabigatran having thrombin inhibitor activity, a process for preparing the derivative, a pharmaceutical composition comprising the derivative, and the Use of derivatives and pharmaceutical compositions in the preparation of thrombin inhibitors and treatment related diseases.
  • Dabigatran (see formula 1 ⁇ 2) is a selective high-efficiency thrombin inhibitor, but due to its strong alkaline sulfhydryl group, it is not absorbed orally:
  • the free carboxyl groups in the dabigatran group have been converted to ethyl esters, and the thiol groups have been converted to hexyl aminodecanoate to obtain the diester prodrug dabigatran etexilate.
  • Dabigatran etexilate (hereinafter referred to as "the compound of the formula 15") is orally absorbed from the gastrointestinal tract and then converted into an active form of dabigatran (formula 1 ⁇ 2) in vivo to exert an anticoagulant effect.
  • the first oral thrombin inhibitor to prevent deep vein thrombosis and pulmonary embolism after artificial joint replacement.
  • studies have shown that the oral bioavailability of dabigatran etexilate is still relatively low. Therefore, there remains a need in the art to develop ester derivatives of novel dabigatran, which are safe, have high oral bioavailability, and have significant anticoagulant effects. Summary of the invention
  • an object of the present invention to provide an ester derivative of dabigatran having a thrombin inhibitor activity, or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or external thereof. Racemic mixture to meet current demand for antithrombin inhibitor drugs.
  • Another object of the present invention is to provide a process for the preparation of the above-mentioned ester derivative of dabigatran or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof.
  • Still another object of the present invention is to provide a pharmaceutical composition
  • a pharmaceutical composition comprising the ester derivative of dabigatran or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof as an active ingredient .
  • a further object of the present invention is to provide an pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture or pharmaceutical composition of the dabigatran group ester derivative or pharmaceutically acceptable use.
  • the present invention provides a derivative of dabigatran etexilate of formula I or a pharmaceutically acceptable salt thereof, dissolved
  • Ri is hydrogen or ( ⁇ -( 5 alkyl); I II
  • R 2 is R 3 , wherein R 3 and R 4 are independently hydrogen or dC 5 alkyl, n is
  • R 5 is Ci-C 8 alkyl or an optionally substituted C r C 8 alkyl.
  • the present invention provides a derivative of dabigatran etexilate or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof, wherein is dC 5 alkyl;
  • R 3 and R 4 are independently hydrogen or an alkyl group of dC 3 ;
  • n 1;
  • it is ( ⁇ -(3 6 alkyl or optionally substituted dC 6 alkyl).
  • the present invention provides a derivative of dabigatran etexilate or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof, wherein ( ⁇ - ( 3 alkyl group, preferably -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , further preferably -CH 2 CH 3 ;
  • the present invention provides a derivative of dabigatran etexilate or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof, wherein R 5 is ( ⁇ -( 6 alkyl, preferably -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , -C(CH 3 ) 3 , -CH(CH 3 )CH 2 CH 3 , -CH 2 CH(CH 3 )CH 3 , -CH 2 CH 2 CH 2 CH 3 ,
  • -CH 2 CH 2 CH 2 CH 2 CH 3 further preferably -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , more preferably -CH(CH 3 ) 2 .
  • the invention provides a derivative of dabigatran etexilate or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof, said dabigatran
  • the ester derivative is shown in the following formula 12:
  • the present invention provides a process for the preparation of the above derivative of dabigatran etexilate or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof, the preparation method
  • the invention provides a process for the preparation of a derivative of dabigatran etexilate or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof, said preparation
  • the method includes the following steps:
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising a derivative of dabigatran etexilate according to the present invention, or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer thereof or A racemic mixture, and a pharmaceutically acceptable excipient.
  • the pharmaceutically acceptable salt means that the compound of the present invention can form a pharmaceutically acceptable salt with an inorganic or organic acid such as hydrochloric acid, hydrobromic acid, hydroiodic acid, nitric acid, perchloric acid, sulfuric acid or phosphoric acid;
  • the organic acid is, for example, sulfonic acid, trifluorosulfonium sulfonic acid, ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, fumaric acid, oxalic acid, maleic acid, citric acid.
  • the solvate is, for example, a hydrate, an alcoholate or the like.
  • the pharmaceutically acceptable excipients in the pharmaceutical composition may include one or more of the following depending on the particular dosage form and mode of administration: diluents, solubilizers, disintegrants, suspending agents, lubricants, binders , fillers, flavoring agents, sweeteners, antioxidants, surfactants, preservatives, encapsulants, and pigments.
  • the pharmaceutical composition may be in any dosage form for clinical administration, such as tablets, suppositories, dispersible tablets, enteric coated tablets, chewable tablets, orally disintegrating tablets, capsules, dragees, granules, dry powders, oral solutions, injections.
  • a small needle, a lyophilized powder for injection or a large infusion preferably an oral dosage form or an injection Type.
  • the present invention provides the above-described derivative of dabigatran etexilate or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof or the above pharmaceutical composition for preparing thrombin inhibition Use in the class of drugs.
  • the present invention provides a method for treating, preventing or delaying the following diseases: venous thrombosis (including deep vein thrombosis and pulmonary embolism), stroke in patients with atrial fibrillation (AF), and acute coronary syndrome (ACS) a heart attack of a patient, the method comprising administering to a patient in need of treatment a therapeutically effective amount of a derivative of dabigatran etexilate according to the invention or a pharmaceutically acceptable salt, solvate, polymorph thereof, enantiomer thereof A body or racemic mixture or a pharmaceutical composition according to the invention.
  • venous thrombosis including deep vein thrombosis and pulmonary embolism
  • AF atrial fibrillation
  • ACS acute coronary syndrome
  • the derivative of dabigatran etexilate provided by the present invention or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof or the pharmaceutical composition provided by the present invention may be combined with other therapies Or the therapeutic agents are co-administered.
  • the mode of administration can be simultaneous, sequential or at certain time intervals.
  • the dose of the compound or pharmaceutical composition required to effect a therapeutic, prophylactic or prolonged action will generally depend on the particular compound being administered, the patient, the particular disease or condition and its severity, the route of administration and frequency, and the like, and The specific situation is judged.
  • the dose may be from 1 to 100 mg/day, preferably from 100 to 300 mg/day, further preferably from 300 mg/day; the dose may be divided into from 1 to 1 Two doses are administered, preferably two times.
  • the present invention provides a novel compound having thrombin inhibitor activity.
  • the novel dabigatran etexilate derivatives of the present invention have higher oral bioavailability and stronger anticoagulant inhibition than the existing dabigatran etexilate (Formula 15), and thus are more suitable for preparation.
  • Fig. 1 shows the results of pharmacokinetic experiments of the compounds of II, 12, 15 and 1 ⁇ 2 in Example 4;
  • Fig. 2 shows the results of aPTT experiments of the compounds of II, 12 and 15 in Example 5. The best way to implement the invention
  • the present invention is further described in detail with reference to the preferred embodiments of the present invention.
  • the experimental methods in the following examples are conventional methods unless otherwise specified.
  • the raw materials, reagent materials and the like used in the following examples can be purchased from a conventional biochemical reagent store or a pharmaceutical business enterprise unless otherwise specified.
  • a compound of the formula IVII was prepared in the same manner as in Example 1, followed by a stirred solution of the compound of the formula ⁇ (420 mg, 3.11 mmol), DMAP (20 mg) and TEA (0.45 mL) in THF (30 mL). Two drops of the compound of formula IV were added. The mixture was stirred at 0 °C for 10 minutes and then quenched with MeOH. The solvent was removed and the crude was purified by EtOAc EtOAc (EtOAc)
  • This example examined the in vitro liver microsome stability of the compound of formula II of the present invention, a compound of formula 12, which was compared to a known compound of formula 15 by detecting the formation of dabigatran.
  • Test compound a compound of formula 11 , a compound of formula 12, and a compound of formula 15;
  • Control compound verapamil.
  • Microsomes Human liver microsomes and rat liver microsomes were purchased from CellzDirect (Invitrogen); stored at -80 °C prior to use.
  • a mother liquid was prepared according to Table 1, and then a test compound or a control compound was added so that the final concentration of these compounds in the reaction system was 2 ⁇ M.
  • the mixed solution was then preheated at 37 ° C for 2 minutes.
  • NADPH was added to the mixed solution to a final concentration of 1 mM, and then the reaction system was placed at 37 °C. The same volume of ultrapure water was added to the blank control instead of NADPH.
  • Mobile phase 0.1% aqueous solution of citric acid (B) and 0.1% citric acid-acetonitrile (A); elution procedure is 0 ⁇ 2min, mobile phase A is 5 ⁇ 100%, mobile phase B is 95% ⁇ 0%; 2 ⁇ 2.2min, mobile phase A is 100%, mobile phase B is 0%; 2.2 ⁇ 2.4min, mobile phase A is 100% ⁇ 5%, mobile phase B is 0% ⁇ 95%; 2.4 ⁇ 3min, mobile phase A At 5%, mobile phase B is 95%.
  • Collision gas 6 L/min; Curtain gas: 30 L/min; Atomizing gas: 50 L/min; Auxiliary gas: 50 L/min; Temperature: 500 ° C; Spray voltage: 4500 v.
  • Test compound II formula in human liver or rat liver microsome system with NADPH
  • the compound of formula 12 and the compound of formula 15 produced a percentage of dabigatran.
  • the results are shown in Table 2 and Table 3 ( (%), day, ,, ,
  • the II compound, the 12 compound, the 15 compound and the 16 compound were each dissolved in a blank solution (30% PEG-400 and physiological saline) at a concentration of lg/L.
  • the experimental animals were male SD rats, 6 to 8 weeks old, weighing 190-215 g, purchased from Beijing Weili Tonghua Experimental Animal Technology Co., Ltd.
  • the SD rats were randomly divided into 4 groups, 3 animals in each group.
  • the doses and routes of administration of each group of rats are shown in Table 4.
  • Group 3 15 Compound 10 Oral 3 Group 4 16 Compound 1 By intravenous 3 SD rats were fasted for 16 hours prior to the pharmacokinetic test. A single dose of the compound or blank solution was then administered intravenously (1 mg/kg) or orally (10 mg/kg) as indicated in Table 4.
  • the jugular vein puncture method was used to collect 200 uL of blood at the time of administration, wherein for the intravenously administered animal group, 0, 15 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, 8 hours, and 24 after administration. Blood was collected at an hour; for the group of animals orally administered, blood was collected at 0, 15 minutes, 30 minutes, 45 minutes, 75 minutes, 135 minutes, 4 hours, 8 hours, and 24 hours after administration. Blood samples were collected in a sample tube with EDTA, and the blood samples were immediately centrifuged at 4000 rpm for 5 minutes at 4 ° C, then the plasma was transferred to another sample tube and stored at -20 ° C.
  • Quantitative method internal standard method
  • Example 5 In vivo pharmacodynamic test - aPPT coagulation inhibition experiment
  • This example examined the in vivo efficacy, i.e., clotting activity, of a compound of formula II, a compound of formula 12, and a known compound of formula 15 of the present invention.
  • Compound of formula II 16.0 mg of the compound of formula II is dissolved in 8.0 mL of 30% PEG400 solution to a final concentration of 2.0 mg/mL;
  • Compound of formula 12 16.2 mg of the compound of formula 12 is dissolved in 8.1 mL of 30% PEG400 solution to a final concentration of 2.0 mg/mL;
  • Compound of formula 15 15.8 mg of the compound of formula 15 is dissolved in 7.9 mL of 30% PEG400 solution to a final concentration of 2.0 mg/mL;
  • the experimental animals were male SD rats, and the body weights were randomly divided into 4 groups of 5 animals each.
  • Group 1 was a blank control group and 30% PEG400 solution was administered.
  • the doses and routes of administration of each group of rats are shown in Table 6.
  • the Coagulometer measures aPPT.
  • the data from Table 7 indicates that the aPTT can be prolonged compared to the blank control group, Group 1, the compound of Formula 15 and the compound of Formula 12 of the present invention, and the anti-clotting inhibitory effect of the compound of Formula 12 provided by the present invention is 2 of the compound of Formula 15 Double, the effect is better.

Abstract

The present invention provides a dabigatran etexilate derivative shown as a formula (I), or pharmaceutically acceptable salt, a solvate, a polymorph, an antipode, or a racemic mixture thereof. In the formula (I), R1 is hydrogen or C1 to C5 alkyl, R2 is a formula (II), R3 and R4 are independently hydrogen or C1 to C5 alkyl, n is 0 or 1, and R5 is C1 to C8 alkyl or optionally substituted C1 to C8 alkyl. The compound has the activity of a thrombin inhibitor. The present invention also provides a preparation method for the compound, a compound-containing medical composition, and application of the compound and the medicinal composition to preparation of thrombin inhibitor medicines and treatment of related diseases.

Description

达比加群的酯衍生物及其制备方法和用途 技术领域  Ester derivative of dabigatran group, preparation method and use thereof
本发明属于医药领域, 具体而言, 本发明涉及具有凝血酶抑制剂活性的 达比加群的酯衍生物、 所述衍生物的制备方法、 包含所述衍生物的药物组合 物、以及所述衍生物和药物组合物在制备凝血酶抑制剂类药物和治疗相关疾 病中的用途。 皆景技术  The present invention relates to the field of medicine, and in particular, the present invention relates to an ester derivative of dabigatran having thrombin inhibitor activity, a process for preparing the derivative, a pharmaceutical composition comprising the derivative, and the Use of derivatives and pharmaceutical compositions in the preparation of thrombin inhibitors and treatment related diseases. Scenery technology
达比加群( Dabigatran, 见下式 ½ )是一种选择性的高效凝血酶抑制剂, 但是由于其存在强碱性脒基, 口服无法吸收:  Dabigatran (see formula 1⁄2) is a selective high-efficiency thrombin inhibitor, but due to its strong alkaline sulfhydryl group, it is not absorbed orally:
Figure imgf000002_0001
为了提高达比加群的生物利用度, 已经分別将达比加群分子中的游离羧 基转化成乙酯, 脒基转化成氨基曱酸己酯, 得到其双酯前体药物达比加群酯 ( Dabigatran Etexilat
Figure imgf000002_0001
In order to improve the bioavailability of dabigatran, the free carboxyl groups in the dabigatran group have been converted to ethyl esters, and the thiol groups have been converted to hexyl aminodecanoate to obtain the diester prodrug dabigatran etexilate. ( Dabigatran Etexilat
Figure imgf000002_0002
Figure imgf000002_0002
达比加群酯(下文称为 "式 15化合物" )经口服后, 从胃肠道吸收, 然后在体内转化为活性形式的达比加群(式 ½ ) , 进而发挥抗凝血作用。 其 于 2008年上市后, 称为首个口服凝血酶抑制剂, 用于预防人工关节置换术 后并发深静脉血栓形成和肺动脉栓塞。 但是, 研究表明达比加群酯的口服生 物利用度仍比较低。 因此, 本领域仍然需要开发安全、 具有较高口服生物利用度且抗凝血作 用明显的新型达比加群的酯衍生物。 发明内容 Dabigatran etexilate (hereinafter referred to as "the compound of the formula 15") is orally absorbed from the gastrointestinal tract and then converted into an active form of dabigatran (formula 1⁄2) in vivo to exert an anticoagulant effect. After its market launch in 2008, it was called the first oral thrombin inhibitor to prevent deep vein thrombosis and pulmonary embolism after artificial joint replacement. However, studies have shown that the oral bioavailability of dabigatran etexilate is still relatively low. Therefore, there remains a need in the art to develop ester derivatives of novel dabigatran, which are safe, have high oral bioavailability, and have significant anticoagulant effects. Summary of the invention
针对上述技术问题, 本发明的一个目的在于提供一种具有凝血酶抑制 剂活性的达比加群的酯衍生物或其药物可接受的盐、 溶剂化物、 多晶型体、 对映体或外消旋混合物, 以满足目前对于抗凝血酶抑制剂类药物的需求。  In view of the above technical problems, it is an object of the present invention to provide an ester derivative of dabigatran having a thrombin inhibitor activity, or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or external thereof. Racemic mixture to meet current demand for antithrombin inhibitor drugs.
本发明的另一个目的在于提供所述达比加群的酯衍生物或其药物可接 受的盐、 溶剂化物、 多晶型体、 对映体或外消旋混合物的制备方法。  Another object of the present invention is to provide a process for the preparation of the above-mentioned ester derivative of dabigatran or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof.
本发明的又一个目的在于提供以所述达比加群的酯衍生物或其药物可 接受的盐、 溶剂化物、 多晶型体、 对映体或外消旋混合物为活性成分的药 物组合物。  Still another object of the present invention is to provide a pharmaceutical composition comprising the ester derivative of dabigatran or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof as an active ingredient .
本发明的再一个目的在于提供所述达比加群的酯衍生物或其药物可接 受的盐、 溶剂化物、 多晶型体、 对映体或外消旋混合物或药物组合物在制 药方面的用途。  A further object of the present invention is to provide an pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture or pharmaceutical composition of the dabigatran group ester derivative or pharmaceutically acceptable use.
本发明的还一个目的在于提供采用所述达比加群的酯衍生物或其药物 可接受的盐、 溶剂化物、 多晶型体、 对映体或外消旋混合物或者采用所述 药物组合物用于治疗相关疾病的方法。  It is still another object of the present invention to provide an ester derivative of the dabigatran or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof or to employ the pharmaceutical composition A method for treating a related disease.
为了实现上述目的, 本发明采取的技术方案如下:  In order to achieve the above object, the technical solution adopted by the present invention is as follows:
一方面, 本发明提供式 I所示的达比加群酯的衍生物或其药物可接受 的盐、 溶  In one aspect, the present invention provides a derivative of dabigatran etexilate of formula I or a pharmaceutically acceptable salt thereof, dissolved
Figure imgf000003_0001
其中
Figure imgf000003_0001
among them
Ri是氢或(^-( 5的烷基; I II Ri is hydrogen or (^-( 5 alkyl); I II
- C-(0) -C - R5 - C-(0) -C - R 5
R2R3 , 其中 R3和 R4独立地是氢或 d-C5的烷基, n为R 2 is R 3 , wherein R 3 and R 4 are independently hydrogen or dC 5 alkyl, n is
0或 1 , R5是 Ci-C8烷基或任选地取代的 CrC8烷基。 0 or 1, R 5 is Ci-C 8 alkyl or an optionally substituted C r C 8 alkyl.
根据本发明的一些实施方式, 本发明提供达比加群酯的衍生物或其药 物可接受的盐、 溶剂化物、 多晶型体、 对映体或外消旋混合物, 其中, 是 d-C5的烷基; According to some embodiments of the present invention, the present invention provides a derivative of dabigatran etexilate or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof, wherein is dC 5 alkyl;
优选地, R3和 R4独立地是氢或 d-C3的烷基; Preferably, R 3 and R 4 are independently hydrogen or an alkyl group of dC 3 ;
优选地, n为 1 ;  Preferably, n is 1;
优选地, 是(^-(36烷基或任选地取代的 d-C6烷基。 Preferably, it is (^-(3 6 alkyl or optionally substituted dC 6 alkyl).
根据本发明的一些实施方式, 本发明提供达比加群酯的衍生物或其药 物可接受的盐、 溶剂化物、 多晶型体、 对映体或外消旋混合物, 其中, 是(^-( 3的烷基, 优选 -CH3、 -CH2CH3、 -CH2CH2CH3、 -CH(CH3)2, 进一步 优选 -CH2CH3; According to some embodiments of the present invention, the present invention provides a derivative of dabigatran etexilate or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof, wherein (^- ( 3 alkyl group, preferably -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , further preferably -CH 2 CH 3 ;
优选地, 和 独立地是氢。  Preferably, and independently is hydrogen.
根据本发明的一些实施方式, 本发明提供达比加群酯的衍生物或其药 物可接受的盐、 溶剂化物、 多晶型体、 对映体或外消旋混合物, 其中, R5 是(^-( 6的烷基,优选 -CH3、 -CH2CH3、 -CH2CH2CH3、 -CH(CH3)2、 -C(CH3)3、 -CH(CH3)CH2CH3、 -CH2CH(CH3)CH3、 -CH2CH2CH2CH3According to some embodiments of the present invention, the present invention provides a derivative of dabigatran etexilate or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof, wherein R 5 is ( ^-( 6 alkyl, preferably -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , -C(CH 3 ) 3 , -CH(CH 3 )CH 2 CH 3 , -CH 2 CH(CH 3 )CH 3 , -CH 2 CH 2 CH 2 CH 3 ,
-CH2CH2CH2CH2CH3,进一步优选 -CH3、 -CH2CH3、 -CH2CH2CH3、 -CH(CH3)2, 更优选 -CH(CH3)2-CH 2 CH 2 CH 2 CH 2 CH 3 , further preferably -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , more preferably -CH(CH 3 ) 2 .
根据本发明的具体实施方式, 本发明提供达比加群酯的衍生物或其药 物可接受的盐、 溶剂化物、 多晶型体、 对映体或外消旋混合物, 所述达比 加群酯的衍生物如下式 12所示:  According to a particular embodiment of the invention, the invention provides a derivative of dabigatran etexilate or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof, said dabigatran The ester derivative is shown in the following formula 12:
Figure imgf000004_0001
12。
Figure imgf000004_0001
12.
另一方面, 本发明还提供上述达比加群酯的衍生物或其药物可接受的 盐、 溶剂化物、 多晶型体、 对映体或外消旋混合物的制备方法, 所述制备 方  In another aspect, the present invention provides a process for the preparation of the above derivative of dabigatran etexilate or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof, the preparation method
Figure imgf000005_0001
Figure imgf000005_0001
la X  La X
其中, 1 6是?、 Cl、 Br或 I , R3至 R5和 n如说明书上文所定义。 根据本发明的具体实施方式, 本发明提供达比加群酯的衍生物或其药 物可接受的盐、溶剂化物、 多晶型体、对映体或外消旋混合物的制备方法, 所述制备方法包括以下步骤: Among them, 1 6 is? , Cl, Br or I, R 3 to R 5 and n are as defined above in the specification. According to a particular embodiment of the invention, the invention provides a process for the preparation of a derivative of dabigatran etexilate or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof, said preparation The method includes the following steps:
物:  Object:
Figure imgf000005_0002
Figure imgf000005_0002
III IVII  III IVII
( 2 M吏式 IV所示化合物在 ZnCl2存在下与(HCHO)n反应生成式 V所 示 4t合物: (2 M 吏 Compound of formula IV is reacted with (HCHO)n in the presence of ZnCl 2 to form a 4t compound of formula V:
Figure imgf000005_0003
Figure imgf000005_0003
IV V  IV V
( 3 )使式 V所示化合物与式 Nal反应生成式 VI所示化合物:
Figure imgf000005_0004
(3) reacting a compound of formula V with a compound of formula Nal to form a compound of formula VI:
Figure imgf000005_0004
V VI 以及 V VI as well as
( 4 M吏式 IVII所示化合物与式 VI所示化合物反应生成式 12所示化合  ( 4 M 吏 Formula IVII is reacted with a compound of formula VI to form a compound of formula 12
Figure imgf000006_0001
Figure imgf000006_0001
12。  12.
再一方面, 本发明提供一种药物组合物, 该药物组合物包含根据本发 明的达比加群酯的衍生物或其药物可接受的盐、 溶剂化物、 多晶型体、 对 映体或外消旋混合物, 以及药学上可接受的辅料。  In a further aspect, the present invention provides a pharmaceutical composition comprising a derivative of dabigatran etexilate according to the present invention, or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer thereof or A racemic mixture, and a pharmaceutically acceptable excipient.
所述药物可接受的盐是指本发明的化合物可以与无机酸或有机酸形 成可药用盐, 其中无机酸诸如盐酸、 氢溴酸、 氢碘酸、 硝酸、 高氯酸、 硫 酸或磷酸; 所述有机酸诸如曱磺酸、 三氟曱磺酸、 乙磺酸、 苯磺酸、 对曱苯 磺酸、 富马酸、 草酸、 马来酸、 柠檬酸。  The pharmaceutically acceptable salt means that the compound of the present invention can form a pharmaceutically acceptable salt with an inorganic or organic acid such as hydrochloric acid, hydrobromic acid, hydroiodic acid, nitric acid, perchloric acid, sulfuric acid or phosphoric acid; The organic acid is, for example, sulfonic acid, trifluorosulfonium sulfonic acid, ethanesulfonic acid, benzenesulfonic acid, p-toluenesulfonic acid, fumaric acid, oxalic acid, maleic acid, citric acid.
所述溶剂化物例如水合物、 醇合物等。  The solvate is, for example, a hydrate, an alcoholate or the like.
选择和制备药物可接受的盐和溶剂化物等是本领域公知技术。  The selection and preparation of pharmaceutically acceptable salts and solvates and the like are well known in the art.
根据具体剂型和施用方式, 所述药物组合物中的药学上可接受的辅料 可以包括下述的一种或多种: 稀释剂、 增溶剂、 崩解剂、 悬浮剂、 润滑剂、 粘合剂、 填充剂、 矫味剂、 甜味剂、 抗氧化剂、 表面活性剂、 防腐剂、 包 裹剂、 和色素等。  The pharmaceutically acceptable excipients in the pharmaceutical composition may include one or more of the following depending on the particular dosage form and mode of administration: diluents, solubilizers, disintegrants, suspending agents, lubricants, binders , fillers, flavoring agents, sweeteners, antioxidants, surfactants, preservatives, encapsulants, and pigments.
所述药物组合物可以为临床施用的任何剂型, 例如片剂、 栓剂、 分散 片、 肠溶片、 咀嚼片、 口崩片、 胶嚢、 糖衣剂、 颗粒剂、 干粉剂、 口服溶 液剂、 注射用小针、 注射用冻干粉针或大输液, 优选为口服剂型或注射剂 型。 The pharmaceutical composition may be in any dosage form for clinical administration, such as tablets, suppositories, dispersible tablets, enteric coated tablets, chewable tablets, orally disintegrating tablets, capsules, dragees, granules, dry powders, oral solutions, injections. Use a small needle, a lyophilized powder for injection or a large infusion, preferably an oral dosage form or an injection Type.
又一方面, 本发明提供上述达比加群酯的衍生物或其药物可接受的 盐、 溶剂化物、 多晶型体、 对映体或外消旋混合物或上述药物组合物在制 备凝血酶抑制剂类药物中的用途。  In still another aspect, the present invention provides the above-described derivative of dabigatran etexilate or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof or the above pharmaceutical composition for preparing thrombin inhibition Use in the class of drugs.
还一方面, 本发明提供一种用于治疗、 预防或延緩下述疾病的方法: 静脉血栓(包括深静脉血栓和肺栓塞) 、 心房颤动(AF )病人的中风和急 性冠状动脉综合征(ACS ) 病人的心脏发病, 所述方法包括给予有治疗需 要的患者治疗有效量的根据本发明的达比加群酯的衍生物或其药物可接 受的盐、 溶剂化物、 多晶型体、 对映体或外消旋混合物或根据本发明的药 物组合物。并且,本发明提供的达比加群酯的衍生物或其药物可接受的盐、 溶剂化物、 多晶型体、 对映体或外消旋混合物或者本发明提供的药物组合 物可以与其它疗法或治疗剂共同施用。 施用方式可以为同时、 顺序或以一 定时间间隔进行。  In still another aspect, the present invention provides a method for treating, preventing or delaying the following diseases: venous thrombosis (including deep vein thrombosis and pulmonary embolism), stroke in patients with atrial fibrillation (AF), and acute coronary syndrome (ACS) a heart attack of a patient, the method comprising administering to a patient in need of treatment a therapeutically effective amount of a derivative of dabigatran etexilate according to the invention or a pharmaceutically acceptable salt, solvate, polymorph thereof, enantiomer thereof A body or racemic mixture or a pharmaceutical composition according to the invention. Furthermore, the derivative of dabigatran etexilate provided by the present invention or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof or the pharmaceutical composition provided by the present invention may be combined with other therapies Or the therapeutic agents are co-administered. The mode of administration can be simultaneous, sequential or at certain time intervals.
实施治疗、 预防或延緩等作用所需的化合物或药物组合物的剂量通常 取决于施用的具体化合物、 患者、 具体疾病或病症及其严重程度、 给药途 径和频率等, 并且需要由主治医师根据具体情况判定。 例如, 在通过口服 途径施用本发明提供的化合物或药物组合物时, 其剂量可为 1至 lOOOmg/ 天, 优选 100至 300mg/天, 进一步优选 300mg/天; 所述剂量可以分每曰 1 至 2次给药, 优选 2次。  The dose of the compound or pharmaceutical composition required to effect a therapeutic, prophylactic or prolonged action will generally depend on the particular compound being administered, the patient, the particular disease or condition and its severity, the route of administration and frequency, and the like, and The specific situation is judged. For example, when the compound or pharmaceutical composition provided by the present invention is administered by an oral route, the dose may be from 1 to 100 mg/day, preferably from 100 to 300 mg/day, further preferably from 300 mg/day; the dose may be divided into from 1 to 1 Two doses are administered, preferably two times.
综上所述, 本发明提供了一种新型的具有凝血酶抑制剂活性的化合物。 实验证明, 相比现有达比加群酯(式 15 ) , 本发明的新型达比加群酯衍生物 口服生物利用率更高, 且具有更强的抗凝血抑制作用, 因此更适合制成多种 剂型的药物, 以用于治疗相关疾病。 附图说明  In summary, the present invention provides a novel compound having thrombin inhibitor activity. Experiments have shown that the novel dabigatran etexilate derivatives of the present invention have higher oral bioavailability and stronger anticoagulant inhibition than the existing dabigatran etexilate (Formula 15), and thus are more suitable for preparation. A variety of dosage forms of drugs for the treatment of related diseases. DRAWINGS
以下, 结合附图来详细说明本发明的实施例:  Hereinafter, embodiments of the present invention will be described in detail with reference to the accompanying drawings:
图 1为实施例 4中 II、 12、 15和½化合物的药代动力学实验结果; 图 2为实施例 5中 II、 12和 15化合物的 aPTT实验结果。 实施发明的最佳方式  BRIEF DESCRIPTION OF THE DRAWINGS Fig. 1 shows the results of pharmacokinetic experiments of the compounds of II, 12, 15 and 1⁄2 in Example 4; Fig. 2 shows the results of aPTT experiments of the compounds of II, 12 and 15 in Example 5. The best way to implement the invention
下面结合具体实施方式对本发明进行进一步的详细描述, 给出的实施 例仅为了阐明本发明, 而不是为了限制本发明的范围。 下述实施例中的实验方法, 如无特殊说明, 均为常规方法。 下述实施 例中所用的药材原料、 试剂材料等, 如无特殊说明, 均可自常规生化试剂 商店或药品经营企业购买得到。 The present invention is further described in detail with reference to the preferred embodiments of the present invention. The experimental methods in the following examples are conventional methods unless otherwise specified. The raw materials, reagent materials and the like used in the following examples can be purchased from a conventional biochemical reagent store or a pharmaceutical business enterprise unless otherwise specified.
Figure imgf000008_0001
Figure imgf000008_0001
III IVII  III IVII
向式 III (2.0 g, 4.15 mmol)在 EtOH (50 mL)中的搅拌溶液中加入盐酸羟 胺 (1.72 g, 24.9 mmol)和 DIPEA (4.36 mL, 24.9 mmol)。在 90。C下搅拌混合物 16小时。将溶液冷却至室温, 然后在减压下去除溶剂。在 EA (50mL)和水 (20 mL)之间分配残余物, 分离有机层。 用 EA (2x30 mL)进一步萃取水相合并有 机提取物。 经 NaS04干燥、 过滤并浓缩。 在硅胶柱 MW1207上纯化粗产物
Figure imgf000008_0002
To a stirred solution of the formula III (2.0 g, 4.15 mmol) in EtOAc (50 mL), EtOAc (1. At 90. The mixture was stirred at C for 16 hours. The solution was cooled to room temperature and then the solvent was removed under reduced pressure. The residue was partitioned between EA (50 mL) and water (20 mL). The aqueous phase was further extracted with EA (2 x 30 mL) and the organic extracts were combined. Dry over NaS04, filter and concentrate. Purification of crude product on silica gel column MW1207
Figure imgf000008_0002
1H NMR (DMSO-d6, 400 MHz) δ 9.24 (s, 1H), 8.38-8.39 (m, 1H), 7.37-7.56 (m, 5H), 7.09-7.15 (m, 2H), 6.88 (d, J = 8.0 Hz, 1H), 6.69 (d, J = 8.8 Hz, 2H), 6.44 (t, J = 5.2 Hz, 1H), 5.56 (br s, 2H), 4.51 (d, J = 5.6 Hz, 2H), 4.22 (t, J = 7.2 Hz, 2H), 3.97 (q, J = 7.2 Hz, 2H), 3.76 (s, 3H), 2.66-2.69 (m, 2H), 1.11 (t, J = 7.2 Hz, 3H). ESI-LCMS: m/z516 [M + H] +. 1H NMR (DMSO-d6, 400 MHz) δ 9.24 (s, 1H), 8.38-8.39 (m, 1H), 7.37-7.56 (m, 5H), 7.09-7.15 (m, 2H), 6.88 (d, J = 8.0 Hz, 1H), 6.69 (d, J = 8.8 Hz, 2H), 6.44 (t, J = 5.2 Hz, 1H), 5.56 (br s, 2H), 4.51 (d, J = 5.6 Hz, 2H) , 4.22 (t, J = 7.2 Hz, 2H), 3.97 (q, J = 7.2 Hz, 2H), 3.76 (s, 3H), 2.66-2.69 (m, 2H), 1.11 (t, J = 7.2 Hz, 3H). ESI-LCMS: m/z 516 [M + H] +.
步骤 (2) :  Step (2):
〇 〇 〇 〇
(HCHO)n ZnCl2 (HCHO)n ZnCl 2
CI CI 〇  CI CI 〇
IV V IV V
在室温、 氮气下搅拌式1¥化合物(212§,2.011101)、 聚曱醛 (60.1 g, 2 mol) 和 ZnCl2(0.5g,cat.)的混合物 40分钟, 然后在 50-60 oC下搅拌 24小时。 经 分馏得到纯的式 V化合物, 为无色的油 (215, 77.9%)。 A mixture of the compound (212 § , 2.011101), polyfurfural (60.1 g, 2 mol) and ZnCl 2 (0.5 g, cat.) was stirred at room temperature under nitrogen for 40 minutes and then stirred at 50-60 °C. 24 hours. Fractional distillation gave the pure compound of formula V as a colourless oil (215, 77.9%).
步骤 (3) :  Step (3):
〇 〇  〇 〇
Nal  Nal
C1 〇 〇  C1 〇 〇
V VI 在 30 oC下将式 V化合物(14.0 g, 0.103 mol)和碘化钠 (27.75 g, 0.185 mol) 在 MeCN(50mL)中的混合物搅拌 5小时。然后用二氯曱烷 (100 mL)和水 (100 mL)稀释溶液, 用 2%Na2S203水溶液洗涤有机层。 在真空下浓缩, 得到式V VI A mixture of the compound of the formula V (14.0 g, 0.103 mol) and sodium iodide (27.75 g, 0.185 mol) in MeCN (50 mL) was stirred at 30 °C for 5 hours. The solution was then diluted with dichloromethane (100 mL) and water (100 mL) and the organic layer was washed with 2% Na 2 S 2 3 aqueous. Concentrate under vacuum
VI化合物, 为发黄色的油(19.6 g, 77%)。 Compound VI, a yellow oil (19.6 g, 77%).
Figure imgf000009_0001
Figure imgf000009_0001
IVII  IVII
Figure imgf000009_0002
Figure imgf000009_0002
12 向式 IVn化合物 (550 mg, 0.95 mmol)和 Cs2C03 (919 mg, 2.82 mmol) 在 DMF (20 mL)中的搅拌溶液中加入式 VI化合物 (429 mg, 1.88 mmol)。 在室温 下搅拌混合物 22小时,然后用水 (50 mL)和 CHC13 (100 mL)稀释。用水 (3x100 mL)进一步洗涤有机层, 然后经 Na2S04干燥合并的有机层并浓缩。 通过 RP HPLC (中性 MeCN和水 )纯化粗产物, 得到式 12化合物, 为白色固体 (22.2 mg, 3.4%) 12 IVn compound of formula (550 mg, 0.95 mmol) and Cs 2 C0 3 (919 mg, 2.82 mmol) was added the compound of formula VI (429 mg, 1.88 mmol) in DMF (20 mL) stirred solution. The mixture was stirred at room temperature for 22 hours and then diluted with water (50 mL) and CHCI ( 3 mL). The organic layer was further washed with water (3x100 mL), the organic layer was then dried over Na 2 S0 4 dried and concentrated. The crude product was purified by EtOAc EtOAc (EtOAc:EtOAc)
:H NMR (DMSO-d6, 400 MHz) δ 8.38 (br s, 1H), 7.37-7.56 (m, 5H), 7.10-7.16 (m, 2H), 6.88 (d, J= 8.0 Hz, 1H), 6.71 (d, J= 8.8 Hz, 2H), 6.57 (br s, 1H), 5.58 (br s, 2H), 4.52 (br s, 2H), 4.22 (t, J = 6.8 Hz, 2H), 3.95-4.00 (m, 2H), 3.76 (s, 3H), 2.68-2.70 (m, 2H), 2.49 (s, 1H), 1.06-1.14 (m, 9H). : H NMR (DMSO-d6, 400 MHz) δ 8.38 (br s, 1H), 7.37-7.56 (m, 5H), 7.10-7.16 (m, 2H), 6.88 (d, J = 8.0 Hz, 1H), 6.71 (d, J= 8.8 Hz, 2H), 6.57 (br s, 1H), 5.58 (br s, 2H), 4.52 (br s, 2H), 4.22 (t, J = 6.8 Hz, 2H), 3.95- 4.00 (m, 2H), 3.76 (s, 3H), 2.68-2.70 (m, 2H), 2.49 (s, 1H), 1.06-1.14 (m, 9H).
ESI-LCMS: m/z 616 [M + H]+. ESI-LCMS: m/z 616 [M + H] + .
Figure imgf000010_0001
Figure imgf000010_0001
II  II
采用和实施例 1相同的方法制备式 IVII化合物,然后在 o °c下向式 ινπ 化合物 (420 mg, 3.11 mmol)、 DMAP (20 mg)和 TEA (0.45 mL)在 THF (30mL) 的搅拌溶液中加入 2滴式 IV化合物。在 0 °C下搅拌混合物 10分钟,然后用 MeOH淬灭反应。 去除溶剂, 通过 RP HPLC (中性 MeCN和水 ) 纯化粗产 物, 得到式 II化合物, 为白色固体 (22.4 mg, 2.13%)。  A compound of the formula IVII was prepared in the same manner as in Example 1, followed by a stirred solution of the compound of the formula ινπ (420 mg, 3.11 mmol), DMAP (20 mg) and TEA (0.45 mL) in THF (30 mL). Two drops of the compound of formula IV were added. The mixture was stirred at 0 °C for 10 minutes and then quenched with MeOH. The solvent was removed and the crude was purified by EtOAc EtOAc (EtOAc)
:H NMR (DMSO-d6,400 MHz) ί58.38 (d, J= 3.6 Hz, 1H), 7.38-7.56 (m, 5H), 7.12-7.16 (m, 2H), 6.88 (d, J= 8.0 Hz, 1H), 6.74 (d, J= 8.4 Hz, 2H), 6.66 (br s, 1H), 6.42 (br s, 2H), 4.55 (d, J= 4.0 Hz, 2H), 4.21 (t, J= 7.6 Hz, 2H), 3.97 (q, J= 7.2 Hz, 2H), 3.75 (s, 3H), 2.66-2.75 (m, 3H), 1.21 (t, J= 7.2 Hz, 3H). : H NMR (DMSO-d6,400 MHz) ί58.38 (d, J= 3.6 Hz, 1H), 7.38-7.56 (m, 5H), 7.12-7.16 (m, 2H), 6.88 (d, J= 8.0 Hz, 1H), 6.74 (d, J= 8.4 Hz, 2H), 6.66 (br s, 1H), 6.42 (br s, 2H ), 4.55 (d, J= 4.0 Hz, 2H), 4.21 (t, J= 7.6 Hz, 2H), 3.97 (q, J= 7.2 Hz, 2H), 3.75 (s, 3H), 2.66-2.75 (m , 3H), 1.21 (t, J= 7.2 Hz, 3H).
ESI-LCMS: m/z 608 [M + Na]+. 实施例 3: 体外稳定性试验 ESI-LCMS: m/z 608 [M + Na] + . Example 3: In vitro stability test
本实施例检测了本发明的式 II化合物、 式 12化合物的体外肝微粒体稳 定性, 其中通过检测达比加群的生成, 与已知的式 15化合物进行了比较。  This example examined the in vitro liver microsome stability of the compound of formula II of the present invention, a compound of formula 12, which was compared to a known compound of formula 15 by detecting the formation of dabigatran.
测试化合物: 式 11化合物、 式 12化合物和式 15化合物;  Test compound: a compound of formula 11 , a compound of formula 12, and a compound of formula 15;
对照化合物: 维拉帕米 ( verapamil ) 。  Control compound: verapamil.
微粒体: 人肝微粒体和大鼠肝微粒体购自 CellzDirect ( Invitrogen ) ; 使用前储存于 -80°C。  Microsomes: Human liver microsomes and rat liver microsomes were purchased from CellzDirect (Invitrogen); stored at -80 °C prior to use.
方法:  Method:
1 )根据表 1所示制备母液, 然后加入测试化合物或对照化合物, 使这 些化合物在反应体系中的最终浓度为 2μΜ。 然后在 37°C下预热混合溶液 2 分钟。  1) A mother liquid was prepared according to Table 1, and then a test compound or a control compound was added so that the final concentration of these compounds in the reaction system was 2 μM. The mixed solution was then preheated at 37 ° C for 2 minutes.
表 1. 母液的制备  Table 1. Preparation of mother liquor
Figure imgf000011_0001
Figure imgf000011_0001
2 ) 向混合溶液中加入 NADPH, 使其最终浓度为 ImM, 然后将反应体 系置于 37°C下。 空白对照中加入同样体积的超纯水代替 NADPH。  2) NADPH was added to the mixed solution to a final concentration of 1 mM, and then the reaction system was placed at 37 °C. The same volume of ultrapure water was added to the blank control instead of NADPH.
3 )在 0、 15、 30、 45和 60分钟时从反应体系中取出 50μ 的等分试样, 并加入 3倍体积的冷曱醇终止反应。 以 16000g离心该等份试样 10分钟以沉 淀蛋白。 将 lOO L的上清液用于 LC/MS/MS分析, 从而测定剩余的测试化 合物和对照化合物的量。 检测一式两份地进行。  3) A 50 μ aliquot was taken from the reaction system at 0, 15, 30, 45 and 60 minutes, and the reaction was stopped by adding 3 volumes of cold methanol. The aliquots were centrifuged at 16000 g for 10 minutes to precipitate the protein. The supernatant of 100 L was used for LC/MS/MS analysis to determine the amount of the remaining test compound and control compound. Detection was performed in duplicate.
LC分析采用的仪器和条件为:  The instruments and conditions used for LC analysis are:
Shimadzu (Degasser DGU - 20A3, Solvent Delivery Unit LC - 20ADXR, Shimadzu (Degasser DGU - 20A3, Solvent Delivery Unit LC - 20ADXR,
System Controller CBM - 20A, Column Oven CTO - 1 OAS VP), CTC Analytics HTC PAL - XT System System Controller CBM - 20A, Column Oven CTO - 1 OAS VP), CTC Analytics HTC PAL - XT System
柱子: Phenomenex 5μ C 18(2) (2.0x50mm)  Column: Phenomenex 5μ C 18(2) (2.0x50mm)
流动相: 0.1 %曱酸水溶液( B )和 0.1 %曱酸 -乙腈( A ) ; 洗脱程序为 0~2min, 流动相 A为 5~100%, 流动相 B为 95%~0%; 2~2.2min, 流动相 A为 100%, 流动相 B为 0%; 2.2~2.4min, 流动相 A为 100%~5%, 流动 相 B为 0%~95%; 2.4~3min, 流动相 A为 5%, 流动相 B为 95%。  Mobile phase: 0.1% aqueous solution of citric acid (B) and 0.1% citric acid-acetonitrile (A); elution procedure is 0~2min, mobile phase A is 5~100%, mobile phase B is 95%~0%; 2 ~2.2min, mobile phase A is 100%, mobile phase B is 0%; 2.2~2.4min, mobile phase A is 100%~5%, mobile phase B is 0%~95%; 2.4~3min, mobile phase A At 5%, mobile phase B is 95%.
流速: 0.5mL/min;  Flow rate: 0.5mL/min;
柱温: 25 °C ;  Column temperature: 25 °C;
上样体积: ΐθμΐ^  Loading volume: ΐθμΐ^
MS/MS分析采用的仪器和条件为:  The instruments and conditions used for MS/MS analysis are:
AB API4000 LC/MS/MS instrument  AB API4000 LC/MS/MS instrument
源: Turbo spray  Source: Turbo spray
电离模式: ESI  Ionization mode: ESI
扫描类型: MRM  Scan Type: MRM
碰撞气: 6 L/min; 帘气: 30 L/min; 雾化气: 50 L/min; 辅助气: 50 L/min; 温度: 500°C ; 喷射电压: 4500 v。  Collision gas: 6 L/min; Curtain gas: 30 L/min; Atomizing gas: 50 L/min; Auxiliary gas: 50 L/min; Temperature: 500 ° C; Spray voltage: 4500 v.
检测结果:  Test results:
在存在 NADPH的人肝或大鼠肝微粒体体系中, 测试式 II化合物、 式 Test compound II, formula in human liver or rat liver microsome system with NADPH
12化合物和式 15化合物生成达比加群的百分比, 结果见表 2和表 3( ( % ) 、 日 、 、 , The compound of formula 12 and the compound of formula 15 produced a percentage of dabigatran. The results are shown in Table 2 and Table 3 ( (%), day, ,, ,
^^样品 达比加群% ( 100 nm ) 达比加群% ( 1 μΜ ) 式 11 式 12 式 15 式 11 式 12 式 15 时间(分  ^^Sample Dabigatran% (100 nm) Dabigatran Group% (1 μΜ) Formula 11 Formula 12 Formula 15 Formula 11 Formula 12 Style 15 Time
空白 0.0 0.0 0.0 0.0 0.0 0.0  Blank 0.0 0.0 0.0 0.0 0.0 0.0
T=60 115.7 100.0 51.1 13.3 11.5 5.9  T=60 115.7 100.0 51.1 13.3 11.5 5.9
Τ=45 81.5 74.8 56.2 9.4 8.6 6.4  Τ=45 81.5 74.8 56.2 9.4 8.6 6.4
Τ=30 74.6 71.1 70.6 8.6 8.2 8.1  Τ=30 74.6 71.1 70.6 8.6 8.2 8.1
Τ=15 41.2 42.9 57.1 4.7 4.9 6.6  Τ=15 41.2 42.9 57.1 4.7 4.9 6.6
Τ=5 28.3 41.7 76.5 3.3 4.8 8.8  Τ=5 28.3 41.7 76.5 3.3 4.8 8.8
Τ=0 1.0 1.5 2.1 0.1 0.2 0.2  Τ=0 1.0 1.5 2.1 0.1 0.2 0.2
T=NCF 7.1 32.6 176.0 0.8 3.7 20.2 表 3. 化合物在大鼠肝微粒体中在不同时间生成的达比加群百分比 (%) 样品 达比加群% ( 100 nm ) 达比加群% ( 1 μΜ ) 式 11 式 12 式 15 式 11 式 12 式 15 时间(分 T=NCF 7.1 32.6 176.0 0.8 3.7 20.2 Table 3. Percentage of dabigatran (%) of compounds produced in rat liver microsomes at different times. Sample dabigatran group (100 nm) dabigatran group % (1 μΜ) formula 11 formula 12 formula 15 11 Equation 12 Equation 15 Time (minutes
空白 0.0 0.0 0.0 0.0 0.0 0.0 Blank 0.0 0.0 0.0 0.0 0.0 0.0
T=60 852.7 636.9 140.9 1 1 1.7 83.4 18.5T=60 852.7 636.9 140.9 1 1 1.7 83.4 18.5
Τ=45 866.5 639.8 177.3 1 13.5 83.8 23.2Τ=45 866.5 639.8 177.3 1 13.5 83.8 23.2
Τ=30 886.6 654.4 230.3 1 16.2 85.7 30.2Τ=30 886.6 654.4 230.3 1 16.2 85.7 30.2
Τ=15 600.8 509.6 219.2 78.7 66.8 28.7Τ=15 600.8 509.6 219.2 78.7 66.8 28.7
Τ=5 496.7 387.1 278.4 65.1 50.7 36.5Τ=5 496.7 387.1 278.4 65.1 50.7 36.5
Τ=0 16.6 10.8 7.9 2.2 1.4 1.0Τ=0 16.6 10.8 7.9 2.2 1.4 1.0
T=NCF 14.7 37.0 347.6 1.9 4.8 45.5 从表 2和表 3的数据可以看到, 在存在 NADPH的人肝微粒体和大鼠肝微 粒体体系中, 本发明的式 II化合物、 式 12化合物经转化生成的达比加群百分 比远高于现有的达比加群酯 (式 5 ) 。 实施例 4: 体内药代动力学试验 T=NCF 14.7 37.0 347.6 1.9 4.8 45.5 It can be seen from the data of Tables 2 and 3 that the compound of the formula II and the compound of the formula 12 of the present invention are transformed in the human liver microsome and the rat liver microsome system in which NADPH is present. The percentage of dabigatran group produced is much higher than the existing dabigatran group (formula 5). Example 4: In vivo pharmacokinetic test
本实施例检测了本发明的 11化合物、 12化合物和 16化合物以及已知达 比加群酯(式 15 ) 的体内药代动力学。  This example examined the in vivo pharmacokinetics of the 11 compounds, 12 compounds and 16 compounds of the present invention and known dabigatran etexilate (Formula 15).
方法:  Method:
将 II化合物、 12化合物、 15化合物和 16化合物分別以 lg/L的浓度溶于 空白溶液( 30% PEG-400和生理盐水 ) 中。  The II compound, the 12 compound, the 15 compound and the 16 compound were each dissolved in a blank solution (30% PEG-400 and physiological saline) at a concentration of lg/L.
实验动物为雄性 SD大鼠, 6至 8周龄, 体重 190-215克, 购自北京维 利通华实验动物技术有限公司。 基于 SD大鼠体重随机分成 4组, 每组 3 只动物。 各组大鼠的给药剂量和途径见表 4。  The experimental animals were male SD rats, 6 to 8 weeks old, weighing 190-215 g, purchased from Beijing Weili Tonghua Experimental Animal Technology Co., Ltd. The SD rats were randomly divided into 4 groups, 3 animals in each group. The doses and routes of administration of each group of rats are shown in Table 4.
表 4. 药代动力学试验分组及给药情况  Table 4. Grouping and administration of pharmacokinetic tests
Figure imgf000013_0001
组 3 15化合物 10 口服 3 组 4 16化合物 1 经静脉 3 在药代动力学试验前,将 SD大鼠禁食 16小时。 然后按照表 4中所示 经静脉( 1 mg/kg )或口服( 10 mg/kg )单个剂量的化合物或空白溶液。 采 取颈静脉穿刺的方式在给药后定时收集血液 200uL, 其中对于经静脉给药 的动物组, 在给药后 0、 15分钟、 30分钟、 1小时、 2小时、 4小时、 8小 时和 24小时收集血液; 对于口服给药的动物组, 在给药后 0、 15分钟、 30分钟、 45分钟、 75分钟、 135分钟、 4小时、 8小时和 24小时收集血 液。 将血样收集于具有 EDTA的样品管中, 立即在 4°C下以 4000rpm离心 血样 5分钟, 然后将血浆转移到另一个样品管中, 储存于 -20摄氏度下。
Figure imgf000013_0001
Group 3 15 Compound 10 Oral 3 Group 4 16 Compound 1 By intravenous 3 SD rats were fasted for 16 hours prior to the pharmacokinetic test. A single dose of the compound or blank solution was then administered intravenously (1 mg/kg) or orally (10 mg/kg) as indicated in Table 4. The jugular vein puncture method was used to collect 200 uL of blood at the time of administration, wherein for the intravenously administered animal group, 0, 15 minutes, 30 minutes, 1 hour, 2 hours, 4 hours, 8 hours, and 24 after administration. Blood was collected at an hour; for the group of animals orally administered, blood was collected at 0, 15 minutes, 30 minutes, 45 minutes, 75 minutes, 135 minutes, 4 hours, 8 hours, and 24 hours after administration. Blood samples were collected in a sample tube with EDTA, and the blood samples were immediately centrifuged at 4000 rpm for 5 minutes at 4 ° C, then the plasma was transferred to another sample tube and stored at -20 ° C.
对样品进行药代动力学检验, 采用的方法和仪器如下:  The samples were subjected to pharmacokinetic tests using the following methods and instruments:
HPLC: Shimadzu (DGV-20A3, Serial NO: SSI-3-0536; LC-20AD Serial HPLC: Shimadzu (DGV-20A3, Serial NO: SSI-3-0536; LC-20AD Serial
NO: L20104551674 USB and L20104551673 USB;), CTC Analytics HTC PAL System (Serial NO: 4353); NO: L20104551674 USB and L20104551673 USB;), CTC Analytics HTC PAL System (Serial NO: 4353);
MS: AB API4000 Q Trap LC/MS/MS instrument (Serial NO. AR19020706) 柱子: Phenomenex Luna 5μ C18 (2.0x50 mm)  MS: AB API4000 Q Trap LC/MS/MS instrument (Serial NO. AR19020706) Column: Phenomenex Luna 5μ C18 (2.0x50 mm)
流动相: 100%乙腈 (2mM乙酸铵)和 100%水 (2mM 乙酸铵) Mobile phase: 100% acetonitrile ( 2 mM ammonium acetate) and 100% water ( 2 mM ammonium acetate)
定量方法: 内标法  Quantitative method: internal standard method
II化合物、 12化合物、 15化合物和½化合物的药代动力学图谱分別见图 1。 其比较结果见表 5。  The pharmacokinetic profiles of the II compound, the 12 compound, the 15 compound, and the 1⁄2 compound are shown in Figure 1, respectively. The comparison results are shown in Table 5.
表 5. II化合物、 12化合物、 15化合物和½化合物的药代动力学数据比较  Table 5. Comparison of pharmacokinetic data of II compounds, 12 compounds, 15 compounds and 1⁄2 compounds
Figure imgf000014_0001
Figure imgf000015_0001
Figure imgf000014_0001
Figure imgf000015_0001
NA: 数据未得。  NA: The data is not available.
比较 12化合物和已知达比加群酯化合物 15 , 表 5中的药代动力学数据表 明, 12化合物在体内产生 ½的半衰期明显长于 15化合物, 达到了 15化合物的 2 倍; 并且, 在口服利用率方面, 12化合物实现了 21.3%的利用率, 同样比 15 化合物的 11.2%的利用率优越。  Comparing the compound of 12 with the known dabigatran etexilate compound 15, the pharmacokinetic data in Table 5 indicates that the 12 compound has a half-life of 1⁄2 in vivo significantly longer than 15 compounds, which is twice as high as 15 compounds; In terms of utilization, 12 compounds achieved 21.3% utilization, which was also superior to 11.2% utilization of 15 compounds.
比较 II化合物和 15化合物, 表 5数据表明, II化合物和 15化合物在体内的 药代特征比较相似。 实施例 5: 体内药效学试验—— aPPT凝血抑制实验  Comparing the II compound with the 15 compound, the data in Table 5 shows that the pharmacological characteristics of the II compound and the 15 compound are similar in vivo. Example 5: In vivo pharmacodynamic test - aPPT coagulation inhibition experiment
本实施例检测了本发明的式 II化合物、 式 12化合物以及已知的式 15化 合物的体内药效, 即凝血活性。  This example examined the in vivo efficacy, i.e., clotting activity, of a compound of formula II, a compound of formula 12, and a known compound of formula 15 of the present invention.
测试化合物及处理:  Test compounds and treatment:
式 II化合物: 将 16.0mg式 II化合物溶解于 8.0mL 30%的 PEG400溶液 中, 终浓度为 2.0mg/mL;  Compound of formula II: 16.0 mg of the compound of formula II is dissolved in 8.0 mL of 30% PEG400 solution to a final concentration of 2.0 mg/mL;
式 12化合物: 将 16.2mg式 12化合物溶解于 8.1mL 30%的 PEG400溶液 中, 终浓度为 2.0mg/mL;  Compound of formula 12: 16.2 mg of the compound of formula 12 is dissolved in 8.1 mL of 30% PEG400 solution to a final concentration of 2.0 mg/mL;
式 15化合物: 将 15.8mg式 15化合物溶解于 7.9mL 30%的 PEG400溶液 中, 终浓度为 2.0mg/mL;  Compound of formula 15: 15.8 mg of the compound of formula 15 is dissolved in 7.9 mL of 30% PEG400 solution to a final concentration of 2.0 mg/mL;
实验动物及处理:  Laboratory animals and treatment:
实验动物为雄性 SD大鼠, 体重随机分成 4组, 每组 5只动物。 其中 组 1为空白对照组, 施用 30%的 PEG400溶液。 各组大鼠的给药剂量和途 径见表 6。  The experimental animals were male SD rats, and the body weights were randomly divided into 4 groups of 5 animals each. Group 1 was a blank control group and 30% PEG400 solution was administered. The doses and routes of administration of each group of rats are shown in Table 6.
表 6. aPPT凝血抑制实验分组及给药情况  Table 6. Grouping and administration of aPPT coagulation inhibition experiments
Figure imgf000015_0002
组 2 式 11化合物 10 口服 5 组 3 式 12化合物 10 口服 5 组 4 式 15化合物 10 口服 5 在试验之前, 大鼠禁食 16小时, 然后口服空白溶液或化合物。 在给药后 30分钟, 经心脏穿刺将血样收集到含抗凝剂的试管中。 立刻在 4°C下 1800g离 心 5分钟, 获取血浆。 通过使用 Instrumentation Laboratory ACL9000
Figure imgf000015_0002
Group 2 Formula 11 Compound 10 Oral 5 Group 3 Formula 12 Compound 10 Oral 5 Group 4 Formula 15 Compound 10 Oral 5 Prior to the test, the rats were fasted for 16 hours and then orally administered with a blank solution or compound. At 30 minutes after administration, blood samples were collected by cardiac puncture into tubes containing anticoagulants. Immediately, centrifugation was performed at 1800 g for 5 minutes at 4 ° C to obtain plasma. By using the Instrumentation Laboratory ACL9000
Coagulometer测量 aPPT。  The Coagulometer measures aPPT.
进行统计分析, 显著性水平设定为 P<0.05。 对研究设计的所有测量参数 计算平均值和标准差。用软件 GraphPad Prism 5.0进行组间的多重比较后的单 因素方差分析(ANOVA ) 。  For statistical analysis, the significance level was set to P < 0.05. Calculate the mean and standard deviation for all measurement parameters of the study design. One-way analysis of variance (ANOVA) was performed with multiple comparisons between groups using the software GraphPad Prism 5.0.
结果见表 7和图 2。  The results are shown in Table 7 and Figure 2.
表 7· aPPT测定结果( Mean ± SD )  Table 7· aPPT measurement results ( Mean ± SD )
Figure imgf000016_0001
Figure imgf000016_0001
注: 相对于组 1 , ** p<0.01  Note: ** p<0.01 relative to group 1
从表 7数据表明, 相比空白对照组即组 1 , 式 15化合物和本发明的式 12化 合物都可以延长 aPTT,并且本发明提供的式 12化合物抗凝血的抑制效果是式 15化合物的 2倍, 作用更优。  The data from Table 7 indicates that the aPTT can be prolonged compared to the blank control group, Group 1, the compound of Formula 15 and the compound of Formula 12 of the present invention, and the anti-clotting inhibitory effect of the compound of Formula 12 provided by the present invention is 2 of the compound of Formula 15 Double, the effect is better.

Claims

权 利 要 求 Rights request
1、一种式 I所示的达比加群酯的衍生物或其药物可接受的盐、溶剂化 物、 多晶 1. A derivative of dabigatran etexilate represented by formula I or its pharmaceutically acceptable salt, solvate, polymorph
Figure imgf000017_0001
其特征在于,
Figure imgf000017_0001
It is characterized by,
Ri是氢或(^-( 5的烷基; Ri is hydrogen or (^-( 5 alkyl;
R4 R 4〇
I II I II
- C-(0) -C - R - C-(0) -C - R
, 其中 R3和 R4独立地是氢或 d-C5的烷基, n为 0或 1 , R5是 c c8烷基或任选地取代的 crc8烷基。 , where R 3 and R 4 are independently hydrogen or dC 5 alkyl, n is 0 or 1, and R 5 is cc 8 alkyl or optionally substituted cc 8 alkyl.
2、 根据权利要求 1所述的达比加群酯的衍生物或其药物可接受的盐、 溶剂化物、 多晶型体、 对映体或外消旋混合物, 其特征在于, !^是^-^ 的烷基; 2. The derivative of dabigatran etexilate or its pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture according to claim 1, characterized in that,! ^is an alkyl group of ^-^;
优选地, R3和 R4独立地是氢或 d-C3的烷基; Preferably, R 3 and R 4 are independently hydrogen or dC 3 alkyl;
优选地, n为 1 ; Preferably, n is 1;
优选地, 是(^-(36烷基或任选地取代的 d-C6烷基。 Preferably, is C-(3 6 alkyl or optionally substituted dC 6 alkyl.
3、 根据权利要求 1或 2所述的达比加群酯的衍生物或其药物可接受 的盐、 溶剂化物、 多晶型体、 对映体或外消旋混合物, 其特征在于, 是(^-( 3的烷基, 优选 -CH3、 -CH2CH3、 -CH2CH2CH3、 -CH(CH3)2, 进一步 优选 -CH2CH3; 3. The derivative of dabigatran etexilate or its pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture according to claim 1 or 2, characterized in that, is ( The alkyl group of -( 3 is preferably -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , and further preferably -CH 2 CH 3 ;
优选地, 和 独立地是氢。 Preferably, and are independently hydrogen.
4、 根据权利要求 1至 3中任一项所述的达比加群酯的衍生物或其药 物可接受的盐、 溶剂化物、 多晶型体、 对映体或外消旋混合物, 其特征在 于, 是(^-( 6的烷基, 优选 -CH3、 -CH2CH3、 -CH2CH2CH3、 -CH(CH3)2、 -C(CH3)3、 -CH(CH3)CH2CH3、 -CH2CH(CH3)CH3、 -CH2CH2CH2CH3 4. The derivative of dabigatran etexilate or its pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture according to any one of claims 1 to 3, characterized by exist is an alkyl group of (^-( 6 , preferably -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , -C(CH 3 ) 3 , -CH( CH 3 )CH 2 CH 3 , -CH 2 CH(CH 3 )CH 3 , -CH 2 CH 2 CH 2 CH 3 ,
-CH2CH2CH2CH2CH3,进一步优选 -CH3、 -CH2CH3、 -CH2CH2CH3、 -CH(CH3)2, 更优选 -CH(CH3)2-CH 2 CH 2 CH 2 CH 2 CH 3 , more preferably -CH 3 , -CH 2 CH 3 , -CH 2 CH 2 CH 3 , -CH(CH 3 ) 2 , more preferably -CH(CH 3 ) 2 .
5、 根据权利要求 1至 4中任一项所述的达比加群酯的衍生物或其药 物可接受的盐、 溶剂化物、 多晶型体、 对映体或外消旋混合物, 其特征在 于, 所示达比加群酯的衍生物如下式 12所示: 5. The derivative of dabigatran etexilate or its pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture according to any one of claims 1 to 4, characterized by That is, the derivative of dabigatran etexilate is shown in the following formula 12:
Figure imgf000018_0001
Figure imgf000018_0001
12。 12.
6、 一种权利要求 1至 5中任一项所述的达比加群酯的衍生物或其药 物可接受的盐、溶剂化物、 多晶型体、对映体或外消旋混合物的制备方法, 所 : 6. Preparation of a dabigatran etexilate derivative or a pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture thereof according to any one of claims 1 to 5 method, so:
Figure imgf000018_0002
Figure imgf000018_0002
la X la
其中, 1 6是?、 Cl、 Br或 I, R3至 R5和 n如权利要求 1至 4中任 一项所定义。 Among them, 1 6 are? , Cl, Br or I, R 3 to R 5 and n are as defined in any one of claims 1 to 4.
7、 一种权利要求 5所述的达比加群酯的衍生物或其药物可接受的盐、 溶剂化物、 多晶型体、 对映体或外消旋混合物的制备方法, 所述制备方法 包括以下步骤: 7. A method for preparing the derivative of dabigatran etexilate or its pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture according to claim 5, the preparation method Includes the following steps:
( 1 )使式 III所示化合物与 NH2OH反应生成式 IVII所示化合物:
Figure imgf000019_0001
(1) React the compound represented by formula III with NH 2 OH to generate the compound represented by formula IVII:
Figure imgf000019_0001
III IVII III IVII
( 2 Μ吏式 IV所示化合物在 ZnCl2存在下与(HCHO)n反应生成式 V所 示化合物: (2M The compound represented by formula IV reacts with (HCHO)n in the presence of ZnCl 2 to generate the compound represented by formula V:
〇 〇 〇 〇
(HCHO)n ZnCl: (HCHO)nZnCl:
CI Cl 〇 CI Cl 〇
IV V IV V
( 3 )使式 V所示化合物与式 Nal反应生成式 VI所示化合物: (3) React the compound represented by formula V with formula Nal to generate the compound represented by formula VI:
ClCl
Figure imgf000019_0002
Figure imgf000019_0002
以及 as well as
物反应生成式 12所示化合 The reaction produces the compound shown in formula 12
Figure imgf000019_0003
Figure imgf000019_0003
12' 12'
8、 一种药物组合物, 该药物组合物包含根据权利要求 1至 5中任一 项所述的达比加群酯的衍生物或其药物可接受的盐、溶剂化物、多晶型体、 对映体或外消旋混合物, 以及药学上可接受的辅料。 8. A pharmaceutical composition comprising the derivative of dabigatran etexilate according to any one of claims 1 to 5 or a pharmaceutically acceptable salt, solvate, or polymorph thereof, Enantiomeric or racemic mixtures, and pharmaceutically acceptable excipients.
9、 根据权利要求 1至 5中任一项所述的达比加群酯的衍生物或其药 物可接受的盐、 溶剂化物、 多晶型体、 对映体或外消旋混合物或根据权利 要求 8所述的药物组合物在制备凝血酶抑制剂类药物中的用途。 9. The derivative of dabigatran etexilate or its pharmaceutically acceptable salt, solvate, polymorph, enantiomer or racemic mixture according to any one of claims 1 to 5 or according to the rights Use of the pharmaceutical composition described in claim 8 in the preparation of thrombin inhibitor drugs.
10、 一种用于治疗、 预防或延緩下述疾病的方法: 静脉血栓(包括深 静脉血栓和肺栓塞)、 心房颤动(AF )病人的中风和急性冠状动脉综合征 10. A method for treating, preventing or delaying the following diseases: venous thrombosis (including deep vein thrombosis and pulmonary embolism), stroke and acute coronary syndrome in patients with atrial fibrillation (AF)
( ACS ) 病人的心脏发病, 所述方法包括给予有治疗需要的患者治疗有效 量的根据权利要求 1至 5中任一项所述的达比加群酯的衍生物或其药物可 接受的盐、 溶剂化物、 多晶型体、 对映体或外消旋混合物或根据权利要求 9所述的药物组合物。 (ACS) cardiac disease in a patient, the method includes administering to a patient in need of treatment a therapeutically effective amount of the derivative of dabigatran etexilate or a pharmaceutically acceptable salt thereof according to any one of claims 1 to 5 , solvates, polymorphs, enantiomers or racemic mixtures or a pharmaceutical composition according to claim 9.
PCT/CN2012/078897 2012-07-19 2012-07-19 Dabigatran etexilate derivative and preparation method and application thereof WO2014012236A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/CN2012/078897 WO2014012236A1 (en) 2012-07-19 2012-07-19 Dabigatran etexilate derivative and preparation method and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2012/078897 WO2014012236A1 (en) 2012-07-19 2012-07-19 Dabigatran etexilate derivative and preparation method and application thereof

Publications (1)

Publication Number Publication Date
WO2014012236A1 true WO2014012236A1 (en) 2014-01-23

Family

ID=49948176

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2012/078897 WO2014012236A1 (en) 2012-07-19 2012-07-19 Dabigatran etexilate derivative and preparation method and application thereof

Country Status (1)

Country Link
WO (1) WO2014012236A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3247332A1 (en) * 2015-01-20 2017-11-29 Incarda Therapeutics, Inc. Unit aerosol doses for anticoagulation

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1675193A (en) * 2002-08-02 2005-09-28 贝林格尔英格海姆法玛两合公司 Prodrugs of 1-methyl-2-(4-amidinophenylaminomethyl)-benzimidazol-5-yl-carboxylic acid-(n-2-pyridyl-n-2-hydroxycarbonylethyl)-amide
WO2006045756A1 (en) * 2004-10-25 2006-05-04 Boehringer Ingelheim International Gmbh Use of dipyridamole in combination with antithrombotics for treatment and prevention of thromboembolic diseases

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1675193A (en) * 2002-08-02 2005-09-28 贝林格尔英格海姆法玛两合公司 Prodrugs of 1-methyl-2-(4-amidinophenylaminomethyl)-benzimidazol-5-yl-carboxylic acid-(n-2-pyridyl-n-2-hydroxycarbonylethyl)-amide
WO2006045756A1 (en) * 2004-10-25 2006-05-04 Boehringer Ingelheim International Gmbh Use of dipyridamole in combination with antithrombotics for treatment and prevention of thromboembolic diseases

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3247332A1 (en) * 2015-01-20 2017-11-29 Incarda Therapeutics, Inc. Unit aerosol doses for anticoagulation
US10668015B2 (en) 2015-01-20 2020-06-02 Incarda Therapeutics, Inc. Unit aerosol doses for anticoagulation
GB2542064B (en) * 2015-01-20 2020-11-18 Univ California Unit aerosol doses for anticoagulation
US11547663B2 (en) 2015-01-20 2023-01-10 Incarda Therapeutics, Inc. Unit aerosol doses for anticoagulation

Similar Documents

Publication Publication Date Title
WO2016041489A1 (en) Indoleamine-2,3-dioxygenase inhibitor and preparation method therefor
JP2017165751A (en) Inhibitors of hepatitis b virus covalently closed circular dna formation and their method of use
TWI771272B (en) Use of clemizole compounds for prevention and treatment of liver cancer
JP6272626B2 (en) Cannabinoid receptor-mediated compounds
MX2010010999A (en) Crystalline salt forms of antifolate compounds and methods of manufacturing thereof.
KR20160058098A (en) Thienopiperidine derivative and use thereof
WO2016196646A1 (en) Cannabinoid receptor mediating compounds
JP2005232179A (en) Antiarrythmic agent containing 3-phenylsulfonyl-3,7-diazabicyclo[3,3,1]nonane compound
CA3106839A1 (en) Inhibitors of hepatitis b virus
CN110698415A (en) Myosin inhibitor and preparation method and application thereof
BR112020025701A2 (en) PRODUCTION PROCESS AND INTERMEDIARIES FOR A PIRROLEUM COMPOUND [2,3-D] PYRIMIDINE AND ITS USE
CN114805478A (en) Deuterated peptidomimetic compound and application thereof
JP2010505747A (en) Selective antagonist of A2A adenosine receptor
US9862678B2 (en) 3,4-diaminobenzenesulfonamide derivatives for inhibiting cell death
JP2000063363A (en) New triazole derivative
US9062023B2 (en) Heterocycle compounds and uses thereof
CN102766134B (en) Dabigatran etexilate derivative and preparation method and application thereof
WO2014012236A1 (en) Dabigatran etexilate derivative and preparation method and application thereof
US9556196B2 (en) Anti-platelet compound addition salt
AU2003296091A1 (en) Optically active dihydropyridine derivative
TW201002687A (en) Phenyl-alkylpiperazines with TNF-modulating activity
WO2014043895A1 (en) 2-hydroxyl tetrahydro thienopyridine derivative with optical activity and preparation method and use thereof
WO2016019849A1 (en) Dabigatran carboalkoxy derivative, preparation method therefor, and pharmaceutical use thereof
TW201400470A (en) 4-alkanoylamino-3-pyrazolone derivatives
TWI682928B (en) Dabigatran alkyl ester derivative, its preparation method and use in pharmacy

Legal Events

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

Ref document number: 12881369

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 12881369

Country of ref document: EP

Kind code of ref document: A1