Compound with HMG-CoA reductase inhibition activity, pharmaceutical composition and application

文档序号:695070 发布日期:2021-05-04 浏览:31次 中文

阅读说明:本技术 一种具有HMG-CoA还原酶抑制活性的化合物、药物组合物及应用 (Compound with HMG-CoA reductase inhibition activity, pharmaceutical composition and application ) 是由 金春华 朱勤丰 于 2019-10-29 设计创作,主要内容包括:本发明属于生物医药领域,具体公开了一种如式I所示的用于治疗和/或预防与HMG-CoA还原酶相关疾病的化合物或其药学上可接受的盐或酯。本发明还公开了含上述化合物的药物组合物。另外,本发明还提供了其作为HMG-CoA还原酶抑制剂以及在制备治疗和/或预防与HMG-CoA还原酶相关疾病的药物中的应用。本发明提供的化合物抑制HMG-CoA还原酶的活性较强,预期所制得的药物在治疗和/或预防血脂异常和动脉粥样硬化上效果较好,且该类化合物结构较简单,预期价格较便宜。此外,本发明提供的化合物与现有他汀类药物结构完全不同,可克服对现有他汀类药物的耐药的现象。(The invention belongs to the field of biological medicines, and particularly discloses a compound shown as a formula I and used for treating and/or preventing HMG-CoA reductase related diseases or pharmaceutically acceptable salt or ester thereof. The invention also discloses a pharmaceutical composition containing the compound. In addition, the invention also provides the HMG-CoA reductase inhibitor andthe application in the preparation of the medicine for treating and/or preventing HMG-CoA reductase related diseases. The compound provided by the invention has stronger activity for inhibiting HMG-CoA reductase, the prepared medicine is expected to have better effect on treating and/or preventing dyslipidemia and atherosclerosis, and the compound has simpler structure and lower expected price. In addition, the compound provided by the invention has a completely different structure from the existing statins, and can overcome the drug resistance phenomenon of the existing statins.)

1. A compound for treating and/or preventing HMG-CoA reductase related diseases as shown in formula I or its pharmaceutically acceptable salt or ester,

in the formula I, n is an integer of 1-3, R is C1~C4An alkyl group.

2. The compound of formula I or a pharmaceutically acceptable salt or ester thereof as claimed in claim 1 wherein R is C1~C4A linear alkyl group, preferably methyl, ethyl or n-propyl.

3. The compound of formula I or a pharmaceutically acceptable salt or ester thereof as claimed in claim 1 wherein n is 3;

and/or R is C1~C4A linear alkyl group, preferably methyl, ethyl or n-propyl.

4. The use of a compound of formula I, or a pharmaceutically acceptable salt or ester thereof, as defined in any one of claims 1 to 3, in the preparation of a medicament for the treatment and/or prevention of a disease associated with the need for inhibition of HMG-CoA reductase activity.

5. Use of a compound of formula I according to any one of claims 1 to 3, or a pharmaceutically acceptable salt or ester thereof, in the manufacture of a medicament for the treatment and/or prevention of dyslipidemia.

6. Use of a compound of formula I as defined in any one of claims 1 to 3, or a pharmaceutically acceptable salt or ester thereof, for the manufacture of a medicament for the treatment and/or prevention of hypercholesterolemia, hyper-low density lipoprotein cholesterolemia or hypertriglyceridemia.

7. Use of a compound of formula I or a pharmaceutically acceptable salt or ester thereof as claimed in any one of claims 1 to 3 in the manufacture of a medicament for the treatment and/or prevention of atherosclerosis.

8. A pharmaceutical composition containing the compound shown in the formula I or the pharmaceutically acceptable salt or ester thereof according to any one of claims 1 to 3, wherein the pharmaceutical composition is an injection administration preparation or an oral administration preparation and contains at least one pharmaceutical adjuvant.

9. The use of claim 8, wherein the injection preparation is small water injection, large infusion solution or powder injection; the oral administration preparation is a solid oral preparation or a liquid oral preparation.

10. The use according to claim 8, wherein the oral formulation is a solid oral formulation, and the solid oral formulation is a plain tablet, a coated tablet, a sugar-coated tablet, a capsule or a granule.

Technical Field

The invention relates to the field of biomedicine, in particular to a compound with HMG-CoA reductase inhibition activity, a pharmaceutical composition and application.

Technical Field

Dyslipidemia, commonly known as hyperlipidemia, generally refers to elevated serum levels of cholesterol (TC), low density lipoprotein cholesterol (LDL-C) and/or Triglycerides (TG), which are the most important risk factors for the cardiovascular system and are highly diseased and dangerous. Dyslipidemia is one of the important factors that causes Atherosclerosis (AS), a major cause of coronary heart disease, cerebral infarction, and peripheral vascular disease. Lipid metabolism disorder is the pathological basis of atherosclerosis, and is characterized in that affected arterial lesions start from intima, generally comprise lipid and complex carbohydrate accumulation, bleeding and thrombosis, further fibrous tissue hyperplasia and calcium deposition, and have gradual disintegration and calcification of middle layer of artery, so as to thicken and harden arterial wall and narrow blood vessel cavity. Lesions often involve large and medium muscle arteries, which supply tissues or organs that become ischemic or necrotic once they develop enough to occlude the lumen of the artery.

The 5 milestone tests in the 90 s of the 20 th century lay a foundation stone role of statins in treating hyperlipidemia. Numerous clinical trials have also fully demonstrated that statins lower LDL-C levels, thereby significantly reducing the risk of cardiovascular events. Clinical trial data in the global cholesterol treatment study cooperation group (CTT) in 2015 showed that for every 1mmol/L LDL-C reduction in statin monotherapy, the relative risk of cardiovascular events was reduced by 22% and 16% in men and women, respectively, and the global mortality was reduced by 10% and 9%, respectively. However, there is still more than 80% cardiovascular remaining risk, the main reasons include: the drug resistance to statins, the reduction of blood fat level by statin treatment can not reach the target index or be accompanied by severe mixed type hyperlipemia, and the like.

The main varieties of the statins are simvastatin, atorvastatin, rosuvastatin and the like. The action principle is that the action of reducing LDL-C and TG is achieved by inhibiting the activity of 3-hydroxy-3-methyl glutaryl coenzyme A reductase (HMG-CoA reductase for short). Because the existing statins have common diseases that the dosage is doubled but the lipid-lowering strength is only increased by 6 percent, and potential risks such as liver damage, rhabdomyolysis, new onset diabetes, nephrotoxicity and the like exist, the existing statins are generally considered to have difficulty in determining the residual risk by only improving the benefit and safety of the dosage. In order to make up for the clinical shortage of statins and increase the target achievement rate of lipid lowering, it is necessary to research new lipid lowering drugs.

Other lipid lowering agents have also been developed in the prior art, such as proprotein convertase subtilisin/kexin 9(PCSK9) inhibitors, cholesterol absorption inhibitors (ezetimibe), Cholesteryl Ester Transfer Protein (CETP) inhibitors, fibrates PPAR α receptor agonists (clofibrate, fenofibrate, gemfibrofibrate, etc.), high purity ethyl eicosapentaenoate, and the like. However, the other lipid-lowering drugs are still not ideal, and have the problems of poor curative effect, high price and the like, such as the PCSK9 inhibitor is very expensive.

Disclosure of Invention

Aiming at the problems of drug resistance, poor curative effect and high price of the blood fat reducing drugs in the prior art, the invention provides a compound with HMG-CoA reductase inhibitory activity and application of a pharmaceutical composition.

The invention adopts the following technical scheme: a compound for treating and/or preventing HMG-CoA reductase related diseases as shown in formula I or its pharmaceutically acceptable salt or ester,

in the formula I, n is an integer of 1-3, R is C1~C4An alkyl group.

Said "C1~C4The alkyl group refers to a group containing 1 to 4 carbon atoms formed by losing any one hydrogen atom on a paraffin molecule, wherein the paraffin molecule comprises straight-chain paraffin and branched-chain paraffin.

When n is 1,2 and 3, the structural formulas are respectively shown as the following formulas I-1, I-2 and I-3:

preferably, the structure of the compound or the pharmaceutically acceptable salt or ester thereof is shown as the formula I-3.

Preferably, R is C1~C4The alkyl group, R is more preferably methyl, ethyl or n-propyl. In the formula I, compounds in which n is an integer of 1-3 and R is methyl, ethyl or n-propyl are shown in the following table:

chemical name n R
Compd 1 5-hydroxy-6- (1-acetyl) -1, 3-benzodioxole 1 CH3
Compd 2 5-hydroxy-6- (1-propionyl) -1, 3-benzodioxole 1 CH2CH3
Compd 3 5-hydroxy-6- (1-butyryl) -1, 3-benzeneBenzodioxolane 1 CH2CH2CH3
Compd 4 6-hydroxy-7- (1-acetyl) -1, 4-benzodioxole 2 CH3
Compd 5 6-hydroxy-7- (1-propionyl) -1, 4-benzodioxole 2 CH2CH3
Compd 6 6-hydroxy-7- (1-butyryl) -1, 4-benzodioxole 2 CH2CH2CH3
Compd 7 7-hydroxy-8- (1-acetyl) -1, 5-benzodioxole 3 CH3
Compd 8 7-hydroxy-8- (1-propionyl) -1, 5-benzodioxocycloheptane 3 CH2CH3
Compd 9 7-hydroxy-8- (1-butyryl) -1, 5-benzodioxole cycloheptaAlkane (I) and its preparation method 3 CH2CH2CH3

Preferably, n is 3 and R is C1~C4A linear alkyl group. More preferably, n is 3 and R is methyl, ethyl or n-propyl. The target product obtained at this time is a novel compound, Compd 7-9, which has a better inhibitory activity against HMG-CoA reductase.

The specific synthetic route for the compounds of formula I above is as follows, wherein n and R in formulae II and III are defined in accordance with formula I:

the invention also provides application of the compound shown as the formula I or pharmaceutically acceptable salt or ester thereof as an HMG-CoA reductase inhibitor and in preparation of medicaments related to diseases needing to inhibit the activity of HMG-CoA reductase (3-hydroxy-3-methylglutaryl coenzyme A reductase ).

The invention also provides application of the compound shown as the formula I or pharmaceutically acceptable salt or ester thereof in preparing a medicament for treating and/or preventing dyslipidemia.

The invention also provides application of the compound shown as the formula I or pharmaceutically acceptable salt or ester thereof in preparing a medicament for treating and/or preventing hypercholesterolemia, high low density lipoprotein cholesterolemia or hypertriglyceridemia.

The invention also provides application of the compound shown as the formula I or pharmaceutically acceptable salt or ester thereof in preparing a medicament for treating and/or preventing atherosclerosis.

The invention also provides a pharmaceutical composition containing the compound shown as the formula I or pharmaceutically acceptable salt or ester thereof, wherein the pharmaceutical composition is an injection administration preparation or an oral administration preparation and contains at least one pharmaceutical adjuvant.

The injection administration preparation is preferably small water injection, large infusion or powder injection.

The oral administration preparation is preferably a solid oral preparation or a liquid oral preparation. Most preferably a solid oral dosage formulation. Further preferably a plain tablet, a coated tablet, a sugar-coated tablet, a capsule or a granule.

The mass percentage of the compound shown in the formula I or the pharmaceutically acceptable salt or ester thereof in the pharmaceutical composition is 0.1-99.9%, preferably 1-99%. The mass percentage refers to the percentage of the compound shown in the formula I or the pharmaceutically acceptable salt or ester thereof in the total mass of the pharmaceutical composition. The sum of the mass fractions of the compound shown in the formula I or the pharmaceutically acceptable salt or ester thereof and the pharmaceutic adjuvant is 100%.

The choice of the pharmaceutical excipients depends on the administration route and the action characteristics, and is usually a filler, a diluent, a binder, a wetting agent, a disintegrating agent, a lubricant, a coating agent, an emulsifying agent or a suspending agent, preferably a pharmaceutical excipient approved for use by the administrative registration.

The pharmaceutical compositions of the present invention may be prepared according to the disclosure using any method known to those skilled in the art, such as conventional mixing, dissolving, granulating, emulsifying, levigating, encapsulating, entrapping, lyophilizing, tableting, and coating processes.

The above preferred conditions can be combined arbitrarily to obtain preferred embodiments of the present invention without departing from the common general knowledge in the art.

Compared with the prior art, the invention has the following technical effects: the compound provided by the invention has stronger activity for inhibiting HMG-CoA reductase, and the prepared medicament is expected to have better effect on treating and/or preventing dyslipidemia and atherosclerosis; in addition, the compounds have simpler structures and are expected to be cheaper. The compound provided by the invention has a structure completely different from that of the existing statins, and can overcome the drug resistance phenomenon of the existing statins.

Detailed Description

The invention is further illustrated by the following examples, which are not intended to limit the scope of the invention. The following examples are experimental methods not specifically mentioned, and reagents and starting materials used in the following examples are commercially available according to conventional methods and conditions.

Example 1

(1) Synthesis of compounds of formula II:

wherein n is an integer of 1 to 3, the structure of the compound is shown as the following formula II-1 when n is 1, the structure of the compound is shown as the following formula II-2 when n is 2, and the structure of the compound is shown as the following formula II-3 when n is 3:

the compound of formula II-1 is sesamol, which is commercially available and is used as is. The compounds of formula II-2 and formula II-3 are prepared according to the prior art [ Nippon Kagaku Kaishi (1977), (6), 925-7; JP 52133984 discloses a method of producing a compound by reacting 1,2, 4-benzenetriol with 1, 2-dichloroethane or 1, 3-dichloropropane, and the specific reaction method is as follows.

Preparation of a Compound of formula II-3: placing 12.6g of 1, 2.4-benzenetriol, 11.3g of 1, 3-dichloropropane, 8.8g of sodium hydroxide and 63g of DMSO (dimethyl sulfoxide) in a reaction bottle, heating to 120 ℃ and 130 ℃ under the condition of stirring for reaction for 6 hours, cooling to room temperature, adjusting the pH value to 4-5 by using concentrated hydrochloric acid, adding 315g of water, stirring to filter out solids, and drying to obtain 15g of a product of a compound shown in a formula II-3 for later use.

Preparation of the Compound of formula II-2: the procedure was essentially identical to that described above for the preparation of the compound of formula II-3, except that 9.9g of 1, 2-dichloroethane was used instead of 11.3g of 1, 3-dichloropropane to carry out the reaction and 13.9g of the compound of formula II-2 was obtained for further use.

(2) Synthesis of target product

Placing 15mmol of anhydrous aluminum trichloride and 80ml of dichloromethane into a reaction bottle, stirring and dissolving, cooling to 0 ℃, adding 12mmol of acetyl chloride, and dropwise adding a solution of sesamol 10 mmol/dichloromethane 20 ml. After the dripping is finished, the reaction is maintained at 0-5 ℃ for 0.5 hour, cooling is removed, stirring is carried out at room temperature, and the reaction is monitored by TLC until the reaction is complete. 100ml of 1.5N hydrochloric acid was added dropwise, the aqueous phase was separated, the organic phase was washed twice with water, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated to dryness. And (4) performing silica gel column chromatography on the residue, and eluting by gradient of petroleum ether-methyl tert-butyl ether mixed solution to obtain the Compd 1.

Compd 2 was prepared as described above for Compd1 except that propionyl chloride was used instead of acetyl chloride.

Compd 3 was prepared as described above for Compd1 except that butyryl chloride was used instead of acetyl chloride.

Compd 4 was prepared as described above for Compd1 except that the compound of formula II-2 was used in place of sesamol.

Compd5 was prepared as described above for Compd1 except that the compound of formula II-2 was used in place of sesamol and propionyl chloride in place of acetyl chloride.

Compd6 was prepared according to the above method for Compd1 except that the compound of formula II-2 was used instead of sesamol and butyryl chloride was used instead of acetyl chloride.

Compd7 was prepared according to the procedure described for Compd1 above except that the compound of formula II-3 was used in place of sesamol.

Compd 8 was prepared as described for Compd1 above except that the compound of formula II-3 was used in place of sesamol and propionyl chloride in place of acetyl chloride.

Compd9 was prepared by the method described for Compd1, except that the compound of formula II-3 was used in place of sesamol and butyryl chloride in place of acetyl chloride.

The structural analysis results of the prepared Compd1-9 are as follows:

Compd 1:1H-NMR(400MHz,CDCl3):13.09(s,OH);7.21(s,1,arom.H);6.54(s,1.arom.H);6.07(s,2H,OCH2O);2.52(s,3H,COCH3).HR-EI-MS:m/z[M+H]+181.0495(calcd for C9H8O4,180.0423).

Compd 2:1H-NMR(400MHz,CDCl3):13.08(s,OH);7.07(s,1 arom.H);6.43(s,1 arom.H);6.01(s,2H,OCH2O);2.81(t,2H,CH2CO);1.01(t,3H,CH3).HR-EI-MS:m/z[M+H]+195.0648(calcd for C10H10O4,194.0579).

Compd 3:1H-NMR(400MHz,CDCl3):13.17(s,OH);7.08(s,1 arom.H);6.44(s,1 arom.H);5.97(s,2H,OCH2O);2.88(q,2H,CH2CO);1.69-1.79(m,2H,CH2CH3),1.21(t,3H,CH3).HR-EI-MS:m/z[M+H]+209.0823(calcd for C11H12O4,208.0736).

Compd 4.1H-NMR(400MHz,CDCl3):13.07(s,OH);7.13(s,1 arom.H);6.40(s,1 arom.H);4.35–4.28(m,2H,OCH2CH2O);4.25–4.07(m,2H,OCH2CH2O);2.52(s,3H,COCH3).HR-EI-MS:m/z[M+H]+195.0653(calcd for C10H10O4,194.0579).

Compd 5.1H-NMR(400MHz,CDCl3):13.06(s,OH);7.08(s,1 arom.H);6.40(s,1 arom.H);4.34–4.27(m,2H,OCH2CH2O);4.23–4.05(m,2H,OCH2CH2O);2.89(q,COCH2);1.09(t,CH2CH3).HR-EI-MS:m/z[M+H]+209.0818(calcd for C11H12O4,208.0736).

Compd 6.1H-NMR(400MHz,CDCl3):13.08(s,OH);7.10(s,1 arom.H);6.40(s,1 arom.H);4.36–4.28(m,2H,OCH2CH2O);4.26–4.07(m,2H,OCH2CH2O);2.86(q,2H,COCH2);1.70-1.79(m,2H,CH2CH3),1.23(t,3H,CH3).HR-EI-MS:m/z[M+H]+223.1004(calcd for C12H14O4,222.0892).

Compd 7.1H-NMR(400MHz,CDCl3):13.07(s,OH);7.13(s,1 arom.H);6.40(s,1 arom.H);4.06–4.04(t,2H,OCH2CH2);3.98–3.96(t,2H,OCH2CH2);2.52(s,3H,COCH3),2.06-2.01(m,2H,CH2CH2O).HR-EI-MS:m/z[M+H]+209.0815(calcd for C11H12O4,208.0736).

Compd 8.1H-NMR(400MHz,CDCl3):13.06(s,OH);7.08(s,1 arom.H);6.40(s,1 arom.H);4.06–4.04(t,2H,OCH2CH2);3.98–3.96(t,2H,OCH2CH2);2.89(q,COCH2);2.06-2.00(m,2H,CH2CH2O);1.09(t,CH3CH2).HR-EI-MS:m/z[M+H]+223.1002(calcd for C12H14O4,222.0892).

Compd 9.1H-NMR(400MHz,CDCl3):13.08(s,OH);7.10(s,1 arom.H);6.40(s,1 arom.H);4.06–4.04(t,2H,OCH2CH2);3.98–3.96(t,2H,OCH2CH2);2.86(q,2H,COCH2),2.05-2.01(m,2H,CH2CH2O),1.70-1.79(m,2H,CH2),1.21(t,3H,Me)..HR-EI-MS:m/z[M+H]+237.1123(calcd for C13H16O4,236.1049).

physical properties of the melting points of the obtained Compd1 to comp 9 were observed, and the melting points of the Compd1 to comp 9 were measured according to a conventional method, and the obtained physical properties and melting points are shown in table 1 below.

TABLE 1

Example 2: compd1-9 assay for inhibition of human HMG-CoA reductase Activity

NADPH, human HMG-CoA reductase, HMGRh assay kit and positive control simvastatin and dimethyl sulfoxide were all used from Sigma, USA.

The specific experiment is carried out according to the reagent specification: HMG-CoA 0.13mM, HMGRh 1 μ L, test sample were mixed with 50mM Tris-HCl buffer pH 7.5, and the volume was made to 100 μ L. Incubate at 37 ℃ for 15 minutes, add NADPH 0.13mM, and continuously test for 10 minutes. NADPH oxidative absorbance was measured at a wavelength of 340nm with a spectrophotometer, and the activity of the enzyme was measured from the rate of decrease of absorbance, and the concentration of the sample required to inhibit HMG-CoA reductase by 50% was calculated. The sample application experiment should be performed at least 6 times, and the sample application experiment should be performed 2 times without sample application. Each unit of enzyme activity is defined as the amount of enzyme required to catalyze the oxidation of 1mmol of NADPH per minute. As shown in Table 2, it is clear from Table 2 that all of Compd1 to 9 exhibit a strong inhibition of HMG-CoA reductase activity.

TABLE 2 results of experiments for inhibiting the activity of human HMG-CoA reductase

Example 3: blood lipid lowering experiment

Representative compounds, Compd1, Compd5, Compd9, were selected for in vivo lipid lowering assays in mice. Animal rooms, laboratories and experiments were performed according to the GLP standard.

The medicine is prepared as follows:

tyloxapol: preparing 80mg/ml solution by using normal saline;

simvastatin: preparing a solution of 2mg/ml by using normal saline;

reagent testing: solutions of 1mg/ml, 2mg/ml and 5mg/ml were prepared with physiological saline.

And (3) testing:

healthy male ICR mice, weighing 18-22g, were freely fed and watered, and were randomly divided into a blank group, a model group, a statin group, and a trial group. The test group was divided into three dose groups of 12 animals per drug. The three doses of the reagent group are respectively 5mg/kg, 10mg/kg and 25mg/kg, and the dose of the statin group is 10 mg/kg. Administration mode IP, administration volume 100 μ l. The blank group was given physiological saline and the model group was given Tyloxapol at a dose of 400 mg/kg. The statin group and the test drug group were administered Tyloxapol400mg/kg first, and simvastatin or the test drug was administered 1 hour later. After 24 hours of administration, blood was collected from the orbit, centrifuged at 13000rpm, and serum was collected to detect TC, LDL-C, TG and HDL-C. The detection is carried out according to the kit instruction, the data is statistically analyzed by using Excel and GraphPad Prism, the comparison between the experimental group and the control group adopts non-matching t test, and the difference with the statistical significance is that P is less than 0.05. The results are shown in Table 3.

TABLE 3

As shown in Table 3, Compd1, Compd5 and Compd9 have good lipid-lowering effect on hyperlipidemia caused by Tyloxapol in mice, and the effect of lowering total cholesterol, LDL-C and TG is better than that of simvastatin, and the dosage-effect relationship is good.

10页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:一种硫酸沙丁胺醇中间体的合成方法

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!

技术分类