TW200533364A - Inhibitor of cytochrome P450 isozyme, CYP2C9 - Google Patents

Inhibitor of cytochrome P450 isozyme, CYP2C9 Download PDF

Info

Publication number
TW200533364A
TW200533364A TW93109325A TW93109325A TW200533364A TW 200533364 A TW200533364 A TW 200533364A TW 93109325 A TW93109325 A TW 93109325A TW 93109325 A TW93109325 A TW 93109325A TW 200533364 A TW200533364 A TW 200533364A
Authority
TW
Taiwan
Prior art keywords
inhibitor
drug
acid
cyp2c9
cytochrome
Prior art date
Application number
TW93109325A
Other languages
Chinese (zh)
Other versions
TWI299002B (en
Inventor
You-Pu Hu
hong-zhan Wang
Zheng-Hui Xiong
li-heng Bao
Original Assignee
Nat Defense Medical Ct Nat Defense University
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Nat Defense Medical Ct Nat Defense University filed Critical Nat Defense Medical Ct Nat Defense University
Priority to TW93109325A priority Critical patent/TW200533364A/en
Publication of TW200533364A publication Critical patent/TW200533364A/en
Application granted granted Critical
Publication of TWI299002B publication Critical patent/TWI299002B/zh

Links

Abstract

The present invention provides various kinds of inhibitors for cytochrome P450 isozyme, CYP2C9, wherein the inhibitor is selected from the group consisted of following compounds: tamarixetin, formononetin, isoliquritigenin, phloretin, luteolin, quercitrin, quercetin, myricetin, wongonin, puerarin, genistein, nordihydroguaiaretic acid, narigenin, capillarisin, chrysin, fisefin, eriodictyol, 6-gingerol, isorhamnetin, isoquercitrin, morin, (+)-taxifolin, isovitexin, 3-phenylpropyl acetate, oleanolic acid, ursolic acid, β-myrcene, cinnamic acid, luteolin-7-glucoside, liquiritin, (+)-limonene, homoorientin, swertiamarin, embelin, daidzein, poncirin, (–)-epicatechin, ergosterol. Among above, the natural product can be used to increase the bioavailability of drug.

Description

200533364 玖、發明說明: 〜 【發明所屬之技術領域】 本發明係提供細胞色素P450(cytochrome P450)的抑制劑,特別係關於 k供一種針對細胞色素P450同工酶(isozyme)CYP2C9之抑制劑。 【先前技術】200533364 发明 Description of the invention: ~ [Technical field to which the invention belongs] The present invention provides an inhibitor of cytochrome P450 (especially as an inhibitor of cytochrome P450 isozyme) CYP2C9. [Prior art]

Cytochrome P450 (P450)為體内藥物代謝過程中進行氧化還原反應之 重要酵素’ P450區分為多種亞型(subfamily),廣泛分佈在肝臟、腸道及其 他組織中(Krishna D· and Klotz U·,Extrahepatic metabolism of drugs in humans. Clinical Pharmacokinetics· 26:144-160, 1994),以進行 phase I reacti〇n 而利於代謝物排出體外,各種亞型及次亞型一般依據胺基酸序列相似度予幸 以區分(Slaughter R丄· and Edward D.J·,Recent advances: the cytochrome P450 enzymes· The Annals of Pharmacotherapy· 29:619-624, 1995),在哺乳動 物中相同亞族酵素彼此間基本上保有超過55%的胺基酸序列相似度,例 如、CYP2C9及CYP2C11或CYP1A1及CYP1A2,事實上阿拉伯數字相近 的單獨酵素間,往往具有高度胺基酸相似性,且基因座落於染色體 (chromosome)的位置也極為相近,以CYP2C9及CYP2C10為例,兩者僅 有兩個胺基酸的差別,而CYP3A3及CYP3A4兩者胺基酸序列有97.5 °/。相 同。因此其Cytochrome P450之酵素命名橫跨生物界各物種之間,包含動 物、植物及微生物,Cytochrome P450中文譯名為『細胞色素P450』,鲁 Cytochrome乃指一群含三價鐵離子並附著於細胞内質網的酵素蛋白質,此 含鐵蛋白結構及功能類似血紅素(hemoglobin)可負責電子轉移及能量釋 放,一般稱之為heme-group或prosthetic group,P450則表示當一氧化碳結 合於此酵素後’經可見光分光光谱儀檢測(Omura T. and Sato R.,The carbon monoxide-binding pigment of liver microsomes. The Journal of Biological Chemistry· 239:2370-2378, 1964),在波長450 nm上具有最大吸光係數,因 此定義為P450。 CYP450體内分佈: 200533364 對於細胞色素P450而言,鼠體及人體酵素分佈區域具有相當大相似 性,人類CYP450同工酶分佈區域相當廣泛(Zhang Q.Y.,Dunbar D” . Ostrowska A.? Zeisloft S.5 Yang J,, and Kaminsky L.S., Characterization of human small intestinal cytochromes P-450. Drug Metabolism and Disposition. ^ 27:804-809, 1999),人體内各型CYP450同工酶除CYP1A1之外大都分佈於 肝臟但含量比例各不相同(Waziers I·,Cugnenc P.H” Yang C.S·,Leroux J.P. and Beaune P.H., Cytochrome P450 isoenzymes, expoxide hydrolase and glutathione transferases in rat and human hepatic and extrahepatic tissues. The Journal of Pharmacology and Experimental Therapeutics. 253:387-394, 1990) » 其中CYP2C subfamily約佔1S.2%,而CY?3A4在腸道也具有相當高比例籲 (約為肝臟50%之含量);鼠體内情形與人類頗為相似,除Cyp2B1及cypiAi 之外,其餘各型同工酶也都以肝臟為主要分佈區域,同時在文獻中已知, 鼠體及人體中各型酵素之比例,雖因物種間差異性而具有相當程度的不 同,然而單就其酵素的生物功能仍具相當取代性,因此對於CYP2C之研究 就以Sprague Dawley大鼠(SD rat)之肝臟為主要酵素萃取來源。Cytochrome P450 (P450) is an important enzyme for redox reactions during drug metabolism in the body. P450 is divided into multiple subfamily and is widely distributed in the liver, intestine and other tissues (Krishna D. and Klotz U., Extrahepatic metabolism of drugs in humans. Clinical Pharmacokinetics · 26: 144-160, 1994), in order to carry out phase I reactin to facilitate the excretion of metabolites, various subtypes and subtypes are generally fortunate based on the similarity of amino acid sequences To distinguish (Slaughter R 丄 · and Edward DJ ·, Recent advances: the cytochrome P450 enzymes · The Annals of Pharmacotherapy · 29: 619-624, 1995), the same subfamily enzymes in mammals basically keep more than 55% from each other Amino acid sequence similarity, for example, CYP2C9 and CYP2C11 or CYP1A1 and CYP1A2. In fact, the individual enzymes with similar Arabic numbers often have a high degree of amino acid similarity, and the gene is located on the chromosome. Similar, taking CYP2C9 and CYP2C10 as examples, there are only two amino acid differences between the two, and the amino acid sequences of both CYP3A3 and CYP3A4 are: 97.5 ° /. the same. Therefore, the enzyme name of its Cytochrome P450 spans all species in the biological world, including animals, plants and microorganisms. Cytochrome P450 is translated into Chinese as "cytochrome P450". Cytochrome refers to a group of ferrous ions that are attached to the endoplasm of cells. Enzyme protein of the net, this ferritin-containing structure and function is similar to hemoglobin, which is responsible for electron transfer and energy release. It is generally called a heme-group or prosthetic group. P450 indicates that when carbon monoxide is bound to this enzyme, it is exposed to visible light. Spectral spectrometer detection (Omura T. and Sato R., The carbon monoxide-binding pigment of liver microsomes. The Journal of Biological Chemistry · 239: 2370-2378, 1964), which has a maximum absorption coefficient at a wavelength of 450 nm, is therefore defined For P450. CYP450 in vivo distribution: 200533364 For cytochrome P450, the distribution of enzymes in mice and humans is quite similar, and the distribution of isozymes in human CYP450 is quite extensive (Zhang QY, Dunbar D ". Ostrowska A.? Zeisloft S. 5 Yang J ,, and Kaminsky LS, Characterization of human small intestinal cytochromes P-450. Drug Metabolism and Disposition. ^ 27: 804-809, 1999), all types of CYP450 isoenzymes in the human body except CYP1A1 are distributed in the liver But the content ratios are different (Waziers I ·, Cugnenc PH "Yang CS ·, Leroux JP and Beaune PH, Cytochrome P450 isoenzymes, expoxide hydrolase and glutathione transferases in rat and human hepatic and extrahepatic tissues. The Journal of Pharmacology and Experimental Therapeutics. 253: 387-394, 1990) »Among them, CYP2C subfamily accounts for about 1S.2%, and CY? 3A4 also has a relatively high proportion in the intestine (approximately 50% of the liver content); the situation in rats is quite similar to that in humans. Similarly, with the exception of Cyp2B1 and cypiAi, all other isoenzymes also use the liver as the main distribution area. It is known that although the ratios of various types of enzymes in mice and humans are quite different due to differences between species, the biological functions of their enzymes are still quite replaceable. Therefore, the study of CYP2C is based on Sprague Dawley. The liver of SD rats is the main source of enzyme extraction.

人類CYP基因共發現有57種不同同工酶,並歸類為14種不同CYP 酵素家族(Nelson D.R·,Koymans L· and Kamataki Τ.,P450 superfamily: update on new sequences, gene mapping, accession numbers and nomenclature.A total of 57 different isozymes were found in the human CYP gene and classified into 14 different CYP enzyme families (Nelson DR ·, Koymans L · and Kamataki T., P450 superfamily: update on new sequences, gene mapping, accession numbers and nomenclature.

Pharmacogenetics· 6:1-42, 1996),分別為 CYP 1,2, 3, 4, 5, 7, 8, 11,17, 19, 21, 24, 27及51,其中CYPI〜3此三類家族酵素負責大部分對於藥物及外來物 質的代謝及解毒作用,而其餘十—種不同族類之酵素群則負責生物體之内 生性物質的代謝,諸如上述所提之贺爾蒙激素或固醇類物質等等…。 基因多型性·· 人類CYP2C家族目前已知具有四個確定的子型,分別為cyp2C8、 CYP2C9、CYP2C18及CYP2C19,四者胺基酸序列具有82%的相似度 (Miners J.O. and Birkett D.J,5 Cytochrome P4502C9: an enzyme of major mportance in human drug metabolism, British Journal of Clinical Pharmacology. 200533364 45:525媽1998),雖然胺基酸序列具有如此高相似度,然而各型酵素對於、 其專-性代謝之藥物卻有相當大差異,娜s年代即有報導指出cYp2c subfamity如同CYP2D6具有基因多型性特徵,臨床上也從事許多cyp2c 基因多型性研究,其中發現人類細胞色素對於藥物代謝之能力可歸納為兩 大類 Extensive metabolizers(EMs)及 Poor metabolizers(PMs),而基因型 相異的比例也隨人種各有不同,以(^僧⑽為例,白種人為削的人口約 佔2〜4 %,而亞洲人則約為2〇 %,因此更需注意對於酵素專一性代謝所產 生的藥物交互作用。 藥物代謝: 藥物經由各種給藥途徑吸收進入體内後,遵循動力學模式接續進行代鲁 謝及排除過程。藥物在體内的代謝方式主要分為一相反應 及二相反應(phase Π reaction),兩種機制皆可形成親水性代謝物,以利 藥物排出體外。混合功能單氧化酶(mixedmonooXygenase)是負責 體内phaselreaction的主要酵素,而p45〇則是屬於此型酵素,也就是經由 P450, P450 reductase,cytochrome b5等群組蛋白負責藥物的氧化還原及水解 反。声’其代谢機轉將詳述於下一節,而phase。Π reaction則主要為接合反 應(conjugation) ’可大致區分為六類(表一),其中以、 sulfation 及 glutathione conjugation 三種最為常見。 @ 表一、體内藥物一相及二相代謝反應(Shargel L·,and Yu A.B.C·, Hepatic elimination of drugs. Applied Biopharmaceutics and Pharmacokinetics. 4th ed” Appleton & Lange,Stamford,pp.353-398, 1999) 200533364 phase I reaction phase Π reaction (High energy intemiedate) Oxidation Aromatic hydroxylation Glucuronide conjugation (UDPGA) Aliphatic hydroxylation N-,O-oxidation Sulfate conjugation (PAPS) N-? O-dealkylation Deamination Glutathion conjugation (GSH) Reduction Azoreduction Acetylation (Acetyl coenzyme A) Nitroreduction Methylation (SAM) Alcohol dehydrogenase Hydrolysis Ester hydrolysis Amide hydrolysis 表一 UDPGA= uridine diphosphoglucuronic acid,PAPS== 3’-phosphoadenosine S^phosphosulfate, GSH= glutathione, SAM= S-adenoylmethionine “細胞色素P450同工酶中CYP2C subfamily之四個子型酵素雖各有不同 代謝藥物,但就臨床藥物代謝而言則以CYP2C9及CYP2C19為主,以肝臟 CYP2C含量比而言,兩者戶斤佔比例約為3 :1 (Venkatakrishnan K·,von Moltke L.L., Greenblatt D.J.5 Relative quantities of catalytically active CYP 2C9 and 2C19 in human liver microsomes: application of the relative activity factor approach. Journal of Pharmaceutical Sciences. 87:845-53, 1998),其中抑制胃酸 分泌的常用藥品Omeprazole即是CYP2C19的專一性代謝受質,至於 CYP2C9所負責代謝之藥物種類則涵蓋較廣泛,一般非固醇類抗發炎藥物 (NSAIDs)、降血糖藥物、抗凝血藥物都屬CYP2C9負責代謝之範疇,僅將 臨床上常見之藥物列於(表二),其中附帶一提的是,phenytoin及warfarin 200533364 都屬臨床上常用之治療指數低(na_細叩_d_)的藥物,一旦 ^個體間或外在魏的差異造成藥物口服·變異性,極有可能造成嚴重 藥物:1作用而導致不良治療後果,其中造成個體間吸收差異的i要原因之 就疋基因多難,基因多型性普赫在於不同人種之間,以CYP2D6為 例CYP2D6主要負責親脂性抗精神病藥物代謝,西方人約有η %的人口 比例為CYP2D6PM基因型,然而東方人為刚基因型者卻不到1%,因此 若使^般劑量之抗精神病藥物於PM患者,往往因藥物代謝減緩,使之 出現嚴重㈣用崎低其生活品質,如此更進—歩降低病人服藥順從性, 使得病情加速惡化,相同的結果;若使用治療指數低之藥物於pM患者, 可能得到更嚴重藥物副作用而造成醫療悲劇。 一要解決藥物在個體間的吸收差異,除了控制藥物吸收的量(例如長效控 釋劑型),再者最直接的方法就是控制藥物代謝的速率,當個體間藥物吸收 的量及代謝速率制-致性,則舒適#後,在理想狀況下應可得到 相似的藥物口服生體可用率,達到相同的洽療效果,也就可減少個體間吸 收的差異,_免副個的產生,除狀外尋找有效的細胞色素調控劑, 尚可藉由暫時性的酵素抑制,減緩高初度效應(flrst_pass effect)藥物之代 謝,然而有效的酵素調控劑必須兼顧安全的先決條件,日常生活中所食用 或藥用之天然物、中草藥即可符合此條件。天然物影響藥物口服生體可用 率最常見的,即是合併飲用葡萄柚汁造成藥物血中濃度大幅提升(Edgar et al.? Acute effects of drinking grapefruit juice on the pharmacokinetics and dynamics of felodipine—and its potential clinical relevance. European Journal of Clinical Pharmacology. 42:313-317, 1992; Lee et al.5 Grapefruit juice and its flavonoids inhibit 11 beta-hydroxysteroid dehydrogenase. Clinical Pharmacology and Therapeutics. 59:62^71, 1996; Kane et al.5 Drug-grapefruit juice interactions· Mayo Clinic Proceedings· 75(9):933-42, 2000)。 表二、CYP2C subfamily抑制、促進及負責代謝之臨床用藥(Rendic s., Summary of information on human CYP enzymes: human P450 metabolism 200533364 data. Drug Metabolism Reviews. 34:83-449, 2002)Pharmacogenetics 6: 1-42, 1996), which are CYP 1, 2, 3, 4, 5, 7, 8, 11, 17, 19, 21, 24, 27, and 51, among which the three families of CYPI ~ 3 Enzymes are responsible for most of the metabolism and detoxification of drugs and foreign substances, while the remaining ten-different types of enzyme groups are responsible for the metabolism of endogenous substances in the organism, such as the hormones or steroids mentioned above Matter, etc ... Genotypes · The human CYP2C family is currently known to have four defined subtypes, namely cyp2C8, CYP2C9, CYP2C18, and CYP2C19. The amino acid sequences of the four have 82% similarity (Miners JO and Birkett DJ, 5 Cytochrome P4502C9: an enzyme of major mportance in human drug metabolism, British Journal of Clinical Pharmacology. 200533364 45: 525 Ma 1998), although the amino acid sequence has such a high degree of similarity, however, all types of enzymes are However, there are considerable differences in drugs. It has been reported that cYp2c subfamity has genetic polymorphism characteristics like CYP2D6. It has also been clinically engaged in many cyp2c gene polymorphism studies. Among them, the ability of human cytochromes to drug metabolism can be summarized as There are two types of Extensive metabolizers (EMs) and Poor metabolizers (PMs), and the proportion of different genotypes also varies from race to race. Take (^ monk) as an example, the white population is about 2 to 4%, and Asians are about 20%, so more attention needs to be paid to drug interactions with enzyme-specific metabolism. Drug Metabolism: Drug Via After the drug is absorbed into the body through this route of administration, it follows the kinetic model to continue the process of deluchet and elimination. The metabolism of drugs in the body is mainly divided into one-phase reaction and two-phase reaction (phase Π reaction), both mechanisms are It can form hydrophilic metabolites to facilitate drug excretion. Mixed monooXygenase is the main enzyme responsible for phaselreaction in the body, and p45〇 belongs to this type of enzyme, that is, via P450, P450 reductase, cytochrome b5 Other groups of proteins are responsible for the redox and hydrolysis reactions of the drug. The sound of its metabolic mechanism will be described in the next section, and the phase. The Π reaction is mainly conjugation. It can be roughly divided into six categories (Table 1) Among them, three kinds, sulfation and glutathione conjugation are the most common. @ 表 一 、 One-phase and two-phase metabolic reactions in vivo (Shargel L ·, and Yu ABC ·, Hepatic elimination of drugs. Applied Biopharmaceutics and Pharmacokinetics. 4th ed ”Appleton & Lange, Stamford, pp. 353-398, 1999) 200533364 phase I reaction phase Π reaction (High energy intemiedate) Oxidation Aromatic hydroxylation Glucuronide conjugation (UDPGA) Aliphatic hydroxylation N-, O-oxidation Sulfate conjugation (PAPS) N-? O-dealkylation Deamination Glutathion conjugation (GSH) Reduction Azoreduction Acetylation (Acetyl coenzyduction Aethylation) ) Alcohol dehydrogenase Hydrolysis Ester hydrolysis Amide hydrolysis Table 1. UDPGA = uridine diphosphoglucuronic acid, PAPS = = 3'-phosphoadenosine S ^ phosphosulfate, GSH = glutathione, SAM = S-adenoylmethionine "CYP2C subfamily four subtypes Although enzymes have different metabolic drugs, but in terms of clinical drug metabolism, CYP2C9 and CYP2C19 are the mainstays. In terms of liver CYP2C content ratio, the proportion of the two households is about 3: 1 (Venkatakrishnan K ·, von Moltke LL , Greenblatt DJ5 Relative quantities of catalytically active CYP 2C9 and 2C19 in human liver microsomes: application of the relative activity factor approach. Journal of Pharmaceutical Sciences. 87: 845-53, 1998) Among them, Omeprazole, a commonly used drug that inhibits gastric acid secretion, is the specific metabolic substrate of CYP2C19. As for the types of drugs that CYP2C9 is responsible for metabolism, it covers a wide range. Generally, non-steroidal anti-inflammatory drugs (NSAIDs), hypoglycemic drugs, anticoagulants The drugs are all in the category of CYP2C9 responsible for metabolism. Only the commonly used drugs are listed in (Table 2). Among them, phenytoin and warfarin 200533364 are both commonly used in clinical treatment with low therapeutic index (na_ 细 叩 _d_ ) Drugs, once the differences between individuals or external Wei cause oral or variability of drugs, it is very likely to cause serious drugs: 1 effects and lead to adverse treatment consequences, among which the main reason for the difference in absorption between individuals is the gene It ’s difficult. The genetic polymorphism is between different races. Taking CYP2D6 as an example, CYP2D6 is mainly responsible for the lipophilic antipsychotic drug metabolism. About η% of the Western population has the CYP2D6PM genotype. Less than 1% of patients, so if the general dose of antipsychotic drugs is used in patients with PM, drug metabolism often slows down, causing severe application The quality of life is so much more advanced-it reduces the patient's medication compliance and accelerates the deterioration of the condition, the same result; if a drug with a low therapeutic index is used in pM patients, it may get more serious drug side effects and cause medical tragedy. First, to resolve the differences in drug absorption between individuals, in addition to controlling the amount of drug absorption (such as long-acting controlled release dosage forms), the most direct method is to control the rate of drug metabolism. When the amount of drug absorption and the rate of metabolism between individuals are controlled -致 性 , 则 温馨 # In the ideal situation, similar oral bioavailability of drugs should be obtained to achieve the same therapeutic effect, which can reduce the difference in absorption between individuals, _ avoid the occurrence of vices, except Looking for effective cytochrome regulators outside the state, you can still slow down the metabolism of high flrst_pass effect drugs by temporary enzyme inhibition. However, effective enzyme regulators must take into account the prerequisites for safety and consumption in daily life. Or medicinal natural substances and Chinese herbal medicines can meet this condition. The most common natural substance that affects the oral bioavailability of a drug is the significant increase in blood concentration of the drug when combined with grapefruit juice (Edgar et al.? Acute effects of drinking grapefruit juice on the pharmacokinetics and dynamics of felodipine—and its potential clinical relevance. European Journal of Clinical Pharmacology. 42: 313-317, 1992; Lee et al. 5 Grapefruit juice and its flavonoids inhibit 11 beta-hydroxysteroid dehydrogenase. Clinical Pharmacology and Therapeutics. 59: 62 ^ 71, 1996; Kane et al .5 Drug-grapefruit juice interactions. Mayo Clinic Proceedings. 75 (9): 933-42, 2000). Table 2. Clinical drugs for inhibiting, promoting and responsible for metabolism of CYP2C subfamily (Rendic s., Summary of information on human CYP enzymes: human P450 metabolism 200533364 data. Drug Metabolism Reviews. 34: 83-449, 2002)

Isoenzyme Substrate Inhibitor inducer CYP2C9 Tolbutamide Diclofenac Warfarin Phenytoin Torsemide Fluvastatin Losartan Celecoxib Meloxicam IsoniazideIsoenzyme Substrate Inhibitor inducer CYP2C9 Tolbutamide Diclofenac Warfarin Phenytoin Torsemide Fluvastatin Losartan Celecoxib Meloxicam Isoniazide

Fluconazole Ketoconazole Metronidazole Itraconazole Cimetidine Sulphaphenazole PhenylbutazoneFluconazole Ketoconazole Metronidazole Itraconazole Cimetidine Sulphaphenazole Phenylbutazone

Rifampin Phenobarbital Cabamazepine EthanolRifampin Phenobarbital Cabamazepine Ethanol

Valporic acid IbuprofenValporic acid Ibuprofen

Carvedilol NaproxanCarvedilol Naproxan

Ondansetron CYP2C19 Omeprazole Fluoxetine Imipramine Sertraline Diazepam Ritonavir Mephenytoin ClomipramineOndansetron CYP2C19 Omeprazole Fluoxetine Imipramine Sertraline Diazepam Ritonavir Mephenytoin Clomipramine

Rifampin HexobarbitalRifampin Hexobarbital

本《明選用大鼠肝微粒體酶為實驗模式,彻巾草藥及天然物中萃取 、、’成刀進行細胞色素體外抑制試驗,以降血糖藥物她说随此(〇如咖⑧) 之標的藥(9G%專—性受CYP2C9代謝)’以微粒體酶代謝抑 王又為師選指標,從中尋找有效的CYP2C9 R工酶調控劑,藉由此有 200533364 效拍調控劑達到提高藥物之生體可用率的目的。 【發明内容】 本發明。之第一方面,係為了增加藥物之生體可用率,而提供有效的細 胞色素P450之抑制劑,其中抑制劑係選自於下列化合物所組成群組: Tamarixetm、花黃素異黃酮㈣刪麵―)、异苷草元如〇1咖郝辟刪、 Phloretm、木犀草素(iuteolin卜解皮苷(Querdtrin)、槲皮素化_咖)、楊梅 樹皮素(myricetin)、漢黃芩素(Wongonin)、葛根素(以⑽如)、金雀異黃_ (Genistein)、正一|里癒瘡酸(N〇rdihydroguaiaretic acid)、柚皮素(Narigenin)、 茵陳色原酮(Capillarisin)、Chrysin、Fisefm、聖草素(eriodictyol)、6-薑辣素 (6-Gmgerol )、異鼠李素(isorhamnetin)、異槲皮苦(is〇quercitrin) 、Morin、 (+)-Taxifolin、異牡荊素(isovitexin)、乙酸苯丙酯(3_phenylpr〇pyi Acetate)、 齊墩果酸(Oleanolic acid )、熊果酸(ursolic acid)、β-月桂烯(β_ Myrcene )、桂 ψ 皮酸(cinnamic acid)、木犀草素;Ο·葡萄糖苷(Luteolin-7_Ghic〇side)、甘草萃 取(Liquiritin)、㈩檸檬精油((+)Limonene)、異荭草素(Homoorientin)、獐 牙菜苦(Swertiamarin)、Embelin、木質素異黃酮(Daid2ein)、枳屬(Poncirin )、 ㈠表兒茶素((-)-Epicatechin)、麥角固醇(erg0sterol)。, 該抑制劑係特別針對於細胞色素P450同工酶CYP2C9具有抑制作 用。該細胞色素(cytochrome)P450同工酶CYP2C9之抑制劑,較佳係選自於 下列化合物所組成群組:Tamarixetin、花黃素異黃酮(Formononetin)、异普 草元(isoliquritigenin)、Phloretin、木犀草素(luteolin)、懈皮苷(Quercitrin) ' 槲皮素(quercetin)、楊梅樹皮素(myricetin)、漢黃芩素(Wongonin)、葛根素 • < (Puerarin )、金雀異黃酮(Genistein)、正二經癒瘡酸(Nordihydroguaiaretic acid)、柚皮素(Narigenin) 〇 該細胞色素(cytochrome)P450同工酶CYP2C9之抑制劑,其中該抑制 劑更佳係為Tamarixetin。 本發明之第二方面係提供用於增加藥物之生體可用率的藥學組合物, 該組合物包含中藥藥引純成份及藥學上可接受之載劑。該中藥藥引純成份 11 200533364 可作為細胞色素(cytochrome)P450之抑制劑。 該中藥藥引純成份係選自於下列化合物所組成群組:Tamarixetin、花 黃素異黃酮(Formononetin)、异苷草元(isoliquritigenin)、Phloretin、木犀草 素(luteolin)、。懈皮苦(Quercitrin)、槲皮素(quercetin)、楊梅樹皮素(niyricetin)、 漢黃答素(Wongonin)、葛根素(Puerarin )、金雀異黃酮(Genistein)、正二經癒 瘡酸(Nordihydroguaiaretic acid)、柚皮素(Narigenin)、茵陳色原酮 (Capillarisin)、Chrysin、Fisefin、聖草素(eriodictyol)、6-薑辣素(6-Gingerol)、 異鼠李素(Isorhamnetin)、異槲皮苷(isoquercitrin)、Morin、(+)-Taxifolin、異 牡菊素(isovitexin)、乙酸苯丙 S旨(3_Phenylpropyl Acetate)、齊墩果酸(Oleanolic acid )、熊果酸(ursolic acid)、β-月桂稀(β- Myrcene )、桂皮酸(cinnamic acid)、。 木犀草素·7·〇-葡萄糖皆(Luteolin-T*Glucoside)、甘草萃取(Liquiritin)、(+) 杯檬精油((+)Limonene )、異蘇草素。(Homoorientin)、猜牙菜苦 (Swertiamarin)、Embelin、木質素異黃酮(Daidzein) ' 枳屬(Poncirin)、㈠ 表兒舍素(㈠-Epicatechin)、麥角固醇(ergosterol)。 該中藥藥引純成份係抑制細胞色素(cytochr〇me)P450同工酶CYP2C9。 該中藥藥引純成份較佳係選自於下列化合物所組成群組: Tamarixetin、花黃素異黃酮(Form〇n〇netin)、异苷草元、This "Ming selects rat liver microsomal enzymes as the experimental model, extracts herbal medicine and natural products, and conducts in vitro cytochrome inhibition tests to make blood sugar lowering drugs. She said that this is the standard drug (such as coffee) (9G% is exclusively metabolized by CYP2C9) 'Take microsomal enzyme metabolism and suppress the king as a teacher's selection indicator, from which to find effective CYP2C9 R engineering enzyme regulators, and by this 200533364 effect shoot regulators to improve the body of the drug The purpose of availability. SUMMARY OF THE INVENTION The present invention. The first aspect is to provide effective inhibitors of cytochrome P450 in order to increase the bioavailability of the drug. The inhibitor is selected from the group consisting of the following compounds: Tamarixetm, flavonoid isoflavones ―), Isoglycosides such as 〇1 coffee harpy, Phloretm, luteolin (iuteolin quercetin (Querdtrin), quercetinized _ coffee), myricetin, myricetin, Wongonin ), Puerarin (taken as Ru), Genistein, Nordihydroguaiaretic acid, Narigenin, Capillarisin, Chrysin , Fisefm, eriodictyol, 6-Gmgerol, isorhamnetin, isocercitrin, Morin, (+)-Taxifolin, Isobaric (Isovitexin), phenylpropyl acetate (3_phenylprOpyi Acetate), oleanolic acid, ursolic acid, β-myrcene, cinnamic acid , Luteolin; 0 · glucoside (Luteolin-7_Ghic〇side), licorice extract (Liquiritin), lime Oil ((+) Limonene), Homoorientin, Swertiamarin, Embelin, Lignin Isoflavones (Daid2ein), Poncirin, Epicatechin ((-)- Epicatechin), erg0sterol. This inhibitor is specifically targeted at the cytochrome P450 isoenzyme CYP2C9 with inhibitory effect. The inhibitor of cytochrome P450 isoenzyme CYP2C9 is preferably selected from the group consisting of the following compounds: Tamarixetin, Formononetin, Isoliquritigenin, Phloretin, Osmanthus Luteolin, Quercitrin 'quercetin, myricetin, wongonin, puerarin < (Puerarin), genistein , Nordihydroguaiaretic acid, naringin. The inhibitor of cytochrome P450 isoenzyme CYP2C9. The inhibitor is more preferably Tamarixetin. A second aspect of the present invention is to provide a pharmaceutical composition for increasing the bioavailability of a drug, the composition comprising a pure ingredient of a traditional Chinese medicine and a pharmaceutically acceptable carrier. The traditional Chinese medicine drug pure ingredients 11 200533364 can be used as an inhibitor of cytochrome P450. The pure ingredients of the traditional Chinese medicine are selected from the group consisting of the following compounds: Tamarixetin, Formononetin, Isoliquritigenin, Phloretin, luteolin, and luteolin. Quercitrin, quercetin, bayberry bark (niyricetin), wongonin, puerarin, genistein, nordihydroguaiaretic acid , Narigenin, Capillarisin, Chrysin, Fisefin, eriodictyol, 6-Gingerol, Isorhamnetin, Isoquercetin Isoquercitrin, Morin, (+)-Taxifolin, isovitexin, 3-Phenylpropyl Acetate, Oleanolic acid, ursolic acid, β- Laurel dilute (β-Myrcene), cinnamic acid (cinnamic acid),. Luteolin-7.0-glucose (Luteolin-T * Glucoside), licorice extract (Liquiritin), (+) cup lemon essential oil ((+) Limonene), isoxetolin. (Homoorientin), Swertiamarin, Embelin, Daidzein 'Poncirin, ㈠-Epicatechin, ergosterol. The pure component of this traditional Chinese medicine extract inhibits cytochrome P450 isoenzyme CYP2C9. The pure ingredients of the traditional Chinese medicine are preferably selected from the group consisting of the following compounds: Tamarixetin, flavonoid isoflavones (FormOnonetin), isoglycone,

Phloretin、木犀草素(luteolin)、懈皮苷(Querc〗trin)、槲皮素(职㈣ 樹皮素(myncetm)、漢黃芩素(Wongonin)、葛根素(puerarin )、金雀異黃酮 你以81師)、正二羥癒瘡酸(跑碰_〇酔丨批咖心)、袖皮素卿吻金)。 4中藥藥引純成份更佳係為Tamarixetin。 ”該組口物係包含tolbutamide之組合投藥,其中該中藥藥引純成份可增 力7tolbutamide之生體可用率。 该、、且口物係包含仙細伽之組合投藥,其中該中藥藥引純成份可增 加fluvastatin之生體可用率。 【實施方式】 本^月著眼於尋找CYP2C9賴控劑,依據文獻報導:以降血糖藥物 12 200533364 tolbutamide為模式藥,在大鼠 '兔、狗、豬及猴等動物取得之肝微粒體酶 體外實驗中發現以抑制模式(inhibition pattern)進行評估,狗與人類CYP2C9 的相似度最高,然而以酵素動力學角度觀察酵告活性及動力學參數 〇 € (Vmax/Km)大氣與人類則最為接近(Bogaards et al·,Determining the best animal model for human cytochrome P450 activities: a comparison of mouse, rat5 rabbit,dog,micropig,monkey and man· Xenobiotica· 30:1131-1152, 2000),若 觀察各種動物對於tolbutamide的代謝途徑(Dogterom P. & Rothuizen J., A species comparison of tolbutamide metabolism in precision-cut liver slices from rats and dogs· Drug Metabolism and Disposition· 21:705-709,1993)則發現大 鼠、兔及人類對於此藥遵循相同代謝途徑,所生成之代謝物也相同,然而 狗則屬不同代謝途徑。此外將人類及大鼠CYP2C9及CYP2C11多胜肽鏈之 妝基酸序列進行比較(http://ww.dmelson.utmem.edu/CytochromeP450/html) ,兩者之間具有73%胺基酸序列相同性,以功能性評估則具有84%生物功 能相似性(http://www.ncbLnlm.nih.gov/BLAST/),因此本發明選擇以大鼠為 理想動物模式,在體外及翻試驗中替代人類肝微粒麟,崎估藥物對 於CYP2C9的酵素抑制性。 、本^明即利用中藥純成分進行藥物體外代謝抑制實驗與小型動物活負 试驗進而仏f中藥藥引對低生體可用率。藥物之藥動性質影響,從中筛$ 具有細胞色素抑制潛力之中藥藥引。 。 較佳實施例之詳細說明 本發明將就下列實施例作進一步說明,然該等實施例僅為例示說明3 用’而不應被解釋為實施本發明之限制。 材料與方法: 分類tirr之中M引乃—般常财藥之化學純成分,其結構可大安 、為類· fiavones、flavanones、chalcones、isnf! t# ( microsomes preparation ) coumarins 〇 本發明之實驗動物為大鼠,因此進行藥物代謝之酵素也取自於鼠肝均質 13 200533364 二首先,動物犧牲後立即將肝臟取出,並置放於代的】.i5化钟' _麵声細溶液中,以清除組織中多餘的血液並降鮮以人甲 質直铺不再有塊 待均質完全後分裝於離心管内齙但應避免管壁結霜。 g 又疋1250〇 g初步離心20分鐘以去除多 餘的細胞外組織,離心後小心取出上層液,内含可溶性肝微粒體、肝細胞、 potassmm)溶液清洗管底之微粒體膠狀顆粒(peiiet),並清除管内殘存之離 心上π液’洗)條後將微粒體膠狀顆粒(pellet)取下,加入與肝等重之pH 7 4 〇·1Μ磷酸緩衝液,並進行第二次均質以得到濃度為1〇〇 % (w/w)微粒體 懸浮液。 細胞碎片、細胞核及粒腺體等胞器,實驗所需的細胞微粒體部分,可由初 步離:所付觸上層液進行超高速離心而製成:將適才所得之上層液分褒 於超同速離〜官’每㈣5〜6 m丨並將㉟製帽蓋旋緊,設^ 麟g超高速孀 離心2小時’離喊成後去除㈣上清液,並以U5 %氯化鉀(触则 製備完成之微粒體懸浮液,其蛋白質含量約為25 g/perml肝均質液, 將懸浮液儲放於-80°C冷凍庫,約可維持酵素活性八週而不致有影響性的 改變,經由上述對於微粒體酵素之製程所製備之懸浮液,即可應用於藥物 代謝之研究,實驗中所使用之微粒體酵素應盡快於時限内運用,以避免產春 生嚴重的誤差而錯判實驗結果。製備流程賤簡略說明如下: (1) Animal sacrifice (spinal cord dislocation); (2) 取肝臟; (3) 以KC1(U5%)清洗後秤重; ⑷剪碎後加入KC1(U5%),重量比(肝:KC1=1 : 2); ,,⑸均質完全; ⑹置入高速離心管,每管約12〜15ml ; ⑺高速離心 4t:520min512500g; 14 200533364 (8) 取上清液置入超高速離心管; „ (9) 超高速離心 4°C,2hr,l〇〇,〇〇〇g; (10) 棄置上清液,以KC1(U5%)清洗管内殘餘液體; (11) 將管内沉澱之pellet刮下; (12) 加入與肝等重之磷酸緩衝液pH 7.4 ; “(13)均質後分裝於 microcentrifuge tube 每管約 imi ; (14)置入-80 C冰箱冷藏。 2·以酵素活性體外實驗(CYP2C9 in vitro study )進行中藥藥弓|筛選· 微粒體酵素製備及蛋白質濃度測定完成後,即可針對酵素cYp2c進行 活性測定以及抑制劑的篩選,而在此之前必須先建立此酵素的實驗模式灯 找哥相關動力學參數’以決定體外試驗(in vitro study)的條件及反應環境。 tolbutamide是一種human CYP-2C9高專一性代謝之藥物,經活化代謝 後可轉變為帶有一親水性官能基的代謝物4-hydroxytolbutamide,並經證實 此反應為單一途徑,它種酵素不涉及其代謝過程,因此可視為是一理想實 驗藥物而能準確測知CYP2C9的活性,受質反應濃度設定為i g,可達酵 素飽合狀悲(Tang et al” Effect of albumin on phenytoin and tolbutamide metabolism in human liver microsonies: an impact more than protein binding.Phloretin, luteolin, Querc trin, quercetin (myncetm), wonongin, puerarin, genistein Teacher), dihydroguaiaretic acid (run bump _〇 酔 丨 batch coffee heart), sleeve skin Su Qing kiss gold). 4 The better component of traditional Chinese medicine is Tamarixetin. "This group of oral products contains a combination of tolbutamide, wherein the pure ingredients of the traditional Chinese medicine can increase the bioavailability of 7tolbutamide. The combination of oral and oral products containing xanbuta, wherein the traditional medicine is pure Ingredients can increase the bioavailability of fluvastatin. [Embodiment] This month, we focus on finding CYP2C9 lyostat, according to literature reports: the blood glucose lowering drug 12 200533364 tolbutamide is used as a model drug in rats' rabbits, dogs, pigs and monkeys In vitro experiments of liver microsomal enzymes obtained by animals and other animals were found to be evaluated in inhibition pattern. Dogs and humans have the highest similarity to CYP2C9. However, enzyme activity and kinetic parameters were observed from the perspective of enzyme kinetics. € (Vmax / Km) The atmosphere is closest to humans (Bogaards et al., Determining the best animal model for human cytochrome P450 activities: a comparison of mouse, rat5 rabbit, dog, micropig, monkey and man · Xenobiotica · 30: 1131-1152, 2000), if we observe the metabolic pathway of tolbutamide in various animals (Dogterom P. & Rothuizen J., A species comparison of t olbutamide metabolism in precision-cut liver slices from rats and dogs · Drug Metabolism and Disposition · 21: 705-709, 1993) found that rats, rabbits, and humans follow the same metabolic pathway for this drug, and the metabolites they produce are also the same, However, dogs belong to different metabolic pathways. In addition, the human and rat CYP2C9 and CYP2C11 polypeptide sequences are compared (http://ww.dmelson.utmem.edu/CytochromeP450/html). Has 73% amino acid sequence identity, and functional evaluation has 84% biological functional similarity (http://www.ncbLnlm.nih.gov/BLAST/), so the present invention chooses to use rats as the ideal animal model It replaces human liver microparticles in vitro and in vitro tests, and evaluates the enzyme inhibitory effect of drugs on CYP2C9. This test is to use the pure ingredients of traditional Chinese medicine for in vitro metabolism inhibition experiments and small animal live negative tests to further study Chinese medicine. Lead to low organism availability. The pharmacokinetic properties of the drug affect the screening of Chinese medicines with potential for cytochrome inhibition. . DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS The present invention will be further described with reference to the following examples, but these examples are only for illustrative purposes 3 and should not be construed as limitations to the practice of the present invention. Materials and methods: The classification of M Yinai, a common chemical compound in tirr, can be classified as Da'an, fiavones, flavanones, chalcones, isnf! T # (microsomes preparation) coumarins 〇 Experimental animals of the present invention It is a rat, so the enzyme for drug metabolism is also taken from rat liver homogenate 13 200533364 II First, the liver is taken out immediately after the animal is sacrificed and placed in the generation]. I5 化 钟 '_Face sound fine solution to remove The excess blood in the tissue is reduced and freshly spread with human nails. There is no longer a piece to be homogenized and then packed in a centrifuge tube. However, frost on the tube wall should be avoided. g and 1,250 g of preliminary centrifugation for 20 minutes to remove excess extracellular tissue. After centrifugation, carefully remove the supernatant, which contains soluble liver microsomes, hepatocytes, and potassmm) solution to clean the microsome gel particles (peiiet) at the bottom of the tube. After removing the remaining centrifugal π liquid 'washing' strips in the tube, remove the microsome gelatinous particles (pellet), add the same weight as the liver's pH 7 4 0 · 1M phosphate buffer, and perform a second homogenization to A microsome suspension was obtained at a concentration of 100% (w / w). Organelles such as cell debris, nuclei, and glandular glands, and the part of the cell microsomes required for the experiment can be prepared by preliminary separation: the contact with the upper layer fluid and ultra-high-speed centrifugation: the upper layer fluid obtained from the appropriate talent is divided into super-speed Isolate ~ officer '5 ~ 6 m each, and tighten the cap, set ^ Lin g ultra-high speed centrifugation for 2 hours, remove the supernatant solution, and use U5% potassium chloride (contact The prepared microsome suspension has a protein content of about 25 g / perml liver homogenate. The suspension can be stored in a -80 ° C freezer to maintain the enzyme activity for about eight weeks without affecting the changes. The suspension prepared by the process of microsomal enzymes can be applied to the study of drug metabolism, and the microsomal enzymes used in the experiments should be used within the time limit as soon as possible to avoid serious errors in the birth and misjudge the experimental results. Preparation Brief description of the process is as follows: (1) Animal sacrifice (spinal cord dislocation); (2) take the liver; (3) weigh after washing with KC1 (U5%); ⑷ add KC1 (U5%) after cutting, weight ratio (Liver: KC1 = 1: 2); ,, ⑸ homogeneous completely; High-speed centrifuge tubes, about 12 ~ 15ml per tube; ⑺ High-speed centrifugation 4t: 520min512500g; 14 200533364 (8) Take the supernatant and place it in an ultra-high-speed centrifuge tube; „(9) Ultra-high-speed centrifugation at 4 ° C, 2hr, l〇〇 (00); (10) Discard the supernatant and wash the remaining liquid in the tube with KC1 (U5%); (11) Scrape off the pellets in the tube; (12) Add a phosphate buffer solution equal to the liver pH 7.4 "(13) After homogenization, aliquot each tube in a microcentrifuge tube; (14) Place in -80 C refrigerator for refrigerating. 2. Perform traditional Chinese medicine bow | screening | screening | microparticles with CYP2C9 in vitro study After the body enzyme preparation and protein concentration measurement are completed, the activity measurement and inhibitor screening of the enzyme cYp2c can be performed. Before this, the experimental mode of this enzyme must be established to find the relevant kinetic parameters of the brother 'to determine the in vitro test (in in vitro study) conditions and reaction environment. tolbutamide is a highly specific metabolizing drug of human CYP-2C9, which can be transformed into a metabolite with a hydrophilic functional group 4-hydroxytolbutamide after activation and metabolism, and the reaction has been confirmed to be single Approach it This enzyme does not involve its metabolic process, so it can be regarded as an ideal experimental drug and can accurately detect the activity of CYP2C9. The concentration of the substrate response is set to ig, which can reach the enzyme saturation state (Tang et al ”Effect of albumin on phenytoin and tolbutamide metabolism in human liver microsonies: an impact more than protein binding.

Drug Metabolism and Disposition· 30:648-654, 2002) o 微粒體酵素活性反應環境設定如下(總體積==1 ml): (1) 0·1 Μ磷酸緩衝液ρΗ 7·4 ; · (2) 0·5 mg微粒體酵素蛋白; 》 (3) 5 mM氯化鎂溶液; (4) 10 mM葡萄糖麟酸; (5) 2 IU葡萄糖磷酸去氫腺; (6) 1 mM核酐二鱗酸腺酐; (7) 1 mM Tolbutamide ί '(8)l%MeOH〇 15 200533364 活性測定反應均於冰上操作以維持4乞環境,將輔酶加入完畢後,先放 置37°C水浴1分鐘,隨後加入模式藥開使計算反應時間,以湖汕m鹽 酸終止反應,並以2 mi二氣甲烷抽取,離心取下層溶劑,吹乾後以移動相 溶液溶解,再進行藥物分析。 反應時_設定,必須賴定受㈣度下,測定代謝物生成與反應時 門的相關:並取其具有正相關的線性區段,換算各時間點的代謝物生成 ϊ: j決定最適合進行分析的反應時間,而模式藥的反應起始濃度,則需 在固疋反應時_情況下,比較不同濃度受質經微粒體酵素代謝後所生成 的代謝物含量,財出《動力學參數Km及Vmax,以決定最適合分析的 反應初始濃度。 7模式藥及代谢物分析是利用高層液相分析儀(sw腿^ LC_1〇Ad), 吁實驗而。峨刀析則採用紫外光檢測器娜madzuSpD_1〇A)、波長設々 為230 ⑽卿et al,1988),層析管採用15〇 乂4 6臟,cμ Γ緩卿,卿-例溫咖 4.6, mI 2T5^ 144 B" J ^ ^ 進行酵素利l==conazole為正相控制組(posit—^ 濃产為10’制應,而確定反輕統的正雜 ’結果如圖一所示,4 %抑制效果μ 4^e=az°le可完_w_mes代舰力,而達101 〒樂#引抑制劑的篩選模式則 同濃度抑_以進行酵素活性^;!正相控制組·’較低、中、高三組耳 形,實驗過程中常需使用甲醇、乙'=、’然时藥藥引水溶解度不佳的價 種不同有機溶劑對於酵素活 :匕寺:機:劑’因此尚需測試名 比較,即可正確判斷中轉=11此扣除溶劑的變關時與控制翻 3·小型動物體内實驗引對於酵素活性的影響性。 降血糖樂物tolbutamide為模 體外實驗从鼠肝微粒麵 16 200533364 式藥進行巾賴引篩選,篩選結果將依抑制程度,選擇有效巾藥純成分進 行小型動物體内實驗,實驗動物仍以錢為動物模式,由於tdbutamide 口 服生體可料已高達9G%,®此並不適合胁㈣實驗,喊峨察藥物 在動物體内的吸收及代謝程度,本實驗選用降血脂藥fluvastatin (Lescol ⑨) 作為齄内试驗之模式藥,fluvastatin是第一種完全由化學合成之HMG—CoA reductase抑制劑,口服生體可用率約25〜30 %,專一性受CYP2C9代謝, fluvastatin sodium 口服吸收可達90 %,因此嚴重的初度效應是造成低生體 可严率的主要原因,此藥經肝臟代謝可生成四種不同代謝物(ScriptureetaL, 2001),其中CYP2C9負責代謝80 % fluvastatin,而其餘同工酶則負責另外 的 20 % 〇 大鼠貫驗前禁食乙日待麻醉後進行頸動脈插管,實驗組大鼠口服投予9 32 mg/kg Tamarixetin (溶於有機溶劑 DMSO, 10 mg/ml),控制組僅服 DMSO, 三十分鐘後兩組皆給予fluvastatin 1.5 mg/kg (溶於水,2 mg/ml),抽血點連同 空白檢品(Blank)共計十二點延續二十四小時,分別為〇, 1〇, 2〇, 40, 60, 120, 240, 360, 480, 720, 1080, 1440 mm,每點抽血約 0.5〜0.6 ml (可分離血漿約 250〜300 μΐ) ’取血之微量管預先加入2〇 μΐ 1〇 HJ肝素(heparin)以防止凝 血,血漿分離後避光置於_80°C冰箱冷藏。Drug Metabolism and Disposition · 30: 648-654, 2002) o Microsomal enzyme activity response environment is set as follows (total volume == 1 ml): (1) 0 · 1 Μ phosphate buffer ρΗ 7 · 4; · (2) 0.5 mg microsomal enzyme protein; (3) 5 mM magnesium chloride solution; (4) 10 mM gluconic acid; (5) 2 IU glucose phosphate dehydrogenation gland; (6) 1 mM nucleoside diphosphonic acid adenine ; (7) 1 mM Tolbutamide ί '(8) 1% MeOH〇15 200533364 The activity measurement reactions were all performed on ice to maintain the environment. After the coenzyme was added, it was placed in a 37 ° C water bath for 1 minute, and then added to the mode. The drug reaction time was calculated, and the reaction was terminated with Hushan m hydrochloric acid, and extracted with 2 mi of digas methane. The lower solvent was removed by centrifugation, and the mobile phase solution was dissolved after blow drying, and then the drug analysis was performed. Reaction time_set, must depend on the degree of acceptance, to determine the correlation between metabolite production and reaction time gate: take a linear segment with positive correlation and convert metabolite generation at each time point ϊ: j decides the most suitable for The reaction time of the analysis, and the initial concentration of the model drug need to be compared with the concentration of metabolites produced by the microsomal enzyme metabolism at different concentrations in the case of solid reaction. And Vmax to determine the initial reaction concentration that is most suitable for analysis. The analysis of 7-mode drugs and metabolites was performed using a high-level liquid phase analyzer (sw leg ^ LC_1〇Ad). Echocardiography uses an ultraviolet light detector (madzuSpD_1〇A), with a wavelength set to 230 (Qing et al, 1988), and a chromatography tube using 15 × 4 6 dirty, cμ Γ Qingqing, Qing-Example Wenka 4.6 , mI 2T5 ^ 144 B " J ^ ^ Enzyme benefit l == conazole is the normal phase control group (posit— ^ concentrated production is 10 'system, and the result of determining the anti-light system's positive impurity' is shown in Figure 1. 4% inhibitory effect μ 4 ^ e = az ° le can complete _w_mes generation of warship power, and the screening mode of up to 101 〒 乐 # primer inhibitor is the same concentration inhibition _ for enzyme activity ^ ;! Normal phase control group · ' The lower, middle, and high ear shapes are often used in the experiment. Methanol, B '=,' But when the drug is not good in water solubility, different organic solvents with different price are used for enzyme activity. By comparing the test names, you can correctly judge the transit = 11 when the deduction of the solvent is turned off and the control is turned 3. The effect of the small animal in vivo experiment on enzyme activity. Hypoglycemic tolbutamide is an in vitro experiment from rat liver microparticles Noodle 16 200533364-type medicine for towel index screening, the screening results will be based on the degree of inhibition, choose the effective ingredients of pure towel medicine for small In vivo experiments in animals, experimental animals still use money as the animal model. Since tdbutamide can be taken up to 9G% in oral organisms, it is not suitable for threatening experiments. The degree of absorption and metabolism of Echa drugs in animals is called. Fluvastatin (Lescol ⑨), a lipid-lowering drug, was selected as the model drug for intracranial tests. Fluvastatin is the first completely chemically synthesized HMG-CoA reductase inhibitor. The oral bioavailability is about 25-30%, and it is specifically affected by CYP2C9. Metabolism, fluvastatin sodium can be absorbed up to 90% orally. Therefore, the severe primary effect is the main reason for the low rate of biotic stricture. This drug can be metabolized by the liver to produce four different metabolites (ScriptureetaL, 2001), of which CYP2C9 is responsible for metabolism. 80% fluvastatin, while the remaining isoenzymes are responsible for the other 20%. Rats were fasted before the test for another day, and then carotid arterial intubation was performed after anesthesia. The rats in the experimental group were orally administered with 9 32 mg / kg Tamarixetin (soluble Organic solvent DMSO, 10 mg / ml), the control group was given DMSO only, and after 30 minutes, both groups were given fluvastatin 1.5 mg / kg (soluble in water, 2 mg / ml), and the blood samples were taken with blank control (Bl ank) A total of twelve o'clock lasts for twenty-four hours, which are 〇, 10, 20, 40, 60, 120, 240, 360, 480, 720, 1080, 1440 mm, and each point draws about 0.5 ~ 0.6 ml (Plasma can be separated from about 250 to 300 μΐ) 'The micro tube for taking blood is added with 20 μΐ 10HJ heparin (heparin) in advance to prevent blood clotting. After plasma separation, it should be refrigerated at _80 ° C in the refrigerator after being separated from light.

Fluvastatin血中濃度採用高效能液相層析儀(High Performance Liquid Chromatography,HPLC)進行分析,檢測器採用螢光分光光度計,激發光及 散射光分別設定為309 nm及390 nm,層析管採用5 μ,150 X 4.6 mm,C18 reverse phase column,移動相採用 〇·ΐΜ TBAF : 0·1 Μ 磷酸緩衝液(pH 6·0) : MeOH (15: 25: 60 v/v),控溫50°C環境下流速設定為1 ml/min,血液 檢品處理程序參照 Kalafsky et aL,(High-pbrformance liquid chromatographic method for the determination of fluvastatin in human plasma. Journal of Chromatography. 614:307-313,1993)及 Toreson et al”(Determination of fluvastatin enantiomers and the racemate in human blood plasma by liquid chromatography and fluorometric detection. Journal of Chromatography A. 17 200533364 729:13-18, 1996)之分析方法,如下所述: (1) 冰/東之檢品取出後,置於碎冰上解珠; (2) 取250 μ1血漿檢品製於螺旋試管; (3) 加入 50μ1 内標準品(celec〇xib 2〇 _ml ώ ; (4) 加入 Acetonitrile 250 μΐ 並震i 5,s Γ (5) 加入 0·5 M phosphate buffer ρΗ^^.Ο 250 μΐ ; ⑹加入 2·5 ml ΜΤΒΕ (methyl-tert七utyl ether), shaking 30 min ; (7) 取有機層溶液於平口試管,減壓濃縮將之揮發; (8) 以2〇〇 μΐ移動相溶液重新溶解; *⑼取出溶液以南速離心13000 rpm,5 min ; (10)取上清液150 μΐ注入HPLC。 實驗結果: 。 體外篩選中藥藥引HUCHE001〜HUCHE070,各成份抑制microsomes 代謝tolbutamide之程度依抑制劑濃度區分為1,1〇, 1〇〇 μΜ,部分中藥藥引 因溶解度不佳,僅以可溶濃度為最高抑制濃度,再依序稀釋為中、低設定 之濃度,實驗結果依抑制劑設定之高、中、低三種不同濃度區分為三類, 最佳抑制劑分別為:HUCHE009异苷草元(isdiquritigenin) 1〇〇 μΜ抑制 microsomes 達 95.5 %、HUCHE070 Tamarixetin 10 μΜ 抑制 88.2 %、 HUCHE037金雀異黃酮Genistein 1 μΜ抑制49·6 %,依中藥藥引編號整理 於表四〜表六。 表三、中藥藥引來源簡介 200533364 編號 純成分名稱 生藥來源 生藥名 HUCHE001 堯花素(Genkwanin) 茵陳蒿 Astemisiae Capillaris HUCHE002 芹菜素(apigenin) 洋甘菊 Chamomillae Flos HUCHE003 木犀草素(luteolin) 洋地黃 Digitals Folium HUCHE004 木犀草素-7-0-葡萄糖苷 (Luteolin-7-Glucoside) 洋地黃 Digitals Folium HUCHE005 異紅草素(Homoorientin) 當藥 Swerticie Herba HUCHE006 異牡荆素(isovitexin) 當藥 Swerttae Herba HUCHE007 新橙皮(Neohesperidin ) 松實 Anrantii Fructus Immatums HUCHE008 花黃素異黃酮(Formononetin) 黃耆 Astragali Radix HUCHE009 异發草元(isoliquritigenin) 黃耆 Astragali Radix HUCHE010 奈紛(kaempferol) 番瀉葉 Sennae Folium HUCHE011 異鼠李素(Isorhamnetin) 番瀉葉 Sennae Folium HUCHE012 異槲皮苷(isoquercitrin) 甘茶 Hydrangeae Dulcis Folium HUCHE013 (+)-表兒茶素(+)-epicatechin 児茶 Gambir HUCHE014 麥角固醇(ergosterol) 麥角. Ergota HUCHE015 (+)兒茶紛素⑴Catechin 芍藥 Paeoniae Radix HUCHE016 6-薑辣素(6- Gingerol) 薑 Zingiberis Rhizoma HUCHE017 甘草萃取(Liquiritin) 甘草 Glycyrrhizae Radix HUCHE018 ’乙酸苯丙酉旨3-Phenylpropyl Acetate 桂皮 Cinnamami Cortex HUCHE019 ㈠表兒茶素(-)-Epicatechin 余葉 Gambir HUCHE020 柚皮素(Narigenin) 枳實 Aurantii Fructus Immatums HUCHE021 繳形花内醋(Umbelliferone ) 枳實 Aurantii Fructus Immaturns HUCHE022 芸香素(Rutin) 槐花 Sophorae Flos HUCHE023 檸檬黃素(Hesperidin) 枳實 Aurantii Fructus Immaturus HUCHE024 香葉木(Diosmin) —— _ 一 HUCHE025 撥皮素(Hesperetin) 陳皮 Citri Reticulatae HUCHE026 漢黃答素(Wongonin) 黃芩 Scutellariae Radix HUCHE027 黃答苷(baicalin) 黃芩 Scutellariae Radix HUCHE028 黃答皆Baicalein 黃芩 Scutellariae Radix HUCHE029 葛根素(Puerarin) 私艮 Pueraria Radix HUCHE030 木質素異黃酮Daidzein 葛根 Pueraria Radix HUCHE031 大豆素(Daidzin ) 葛根 Pueraria Radix HUCHE032 懈皮皆(Quercitrin) 獬皮 Viscum Coloratum HUCHE033 槲皮素(quercetin) 正二羥癒瘡酸 獬皮 創木酸 Viscnm Coloratum HUCHE034 ~: (Nordihydroguaiaretic acid) 表三 19 200533364 表三、中藥藥引來源簡介(續) 編號 純成分名稱 生藥,來源 生藥名 HUCHE035 fe]陳色原麵I ( Capillarisin ) HUCHE036 獐牙菜苦(Swertiamarin) HUCHE037 金雀異黃酮Genistein HUCHE038 反- 肉桂醛 trans-Cinnamaldehyde HUCHE039 原兒茶酸(protocatechuic acid) HUCHE040 沒食子酸(gallic acid) HUCHE041 芍藥素(paeoniflorin) HUCHE042 聖草素(eriodictyol) HUCHE043 积屬(Poncirin) HUCHE044 黃酮 (a -Naphthoflavone ) HUCHE045 月桂烤(0 -Myrcene) HUCHE046 α-松油醇(α-terpineol) HUCHE047 (+)-檸檬精油(+) -Limonene HUCHE048 月桂醇 Lauryl Alcohol HUCHE049 十四烧酸乙脂(Ethyl Myristate) HUCHE050 桉樹紛(Cineole) HUCHE051 甘草甜素(glycyrrhizin) HUCHE052 齊墩果酸(Oleanolic acid ) HUCHE053 熊果酸(ursolic acid) HUCHE054 柚苦 Narigin HUCHE055 黃酮(yS-Naphthoflavone) HUCHE056 反-桂皮酸(trans-cinnamic acid) HUCHE061 Morin HUCHE062 (+)-Taxifolin HUCHE063 Chrysin HUCHE064 高良姜素(Galangin) HUCHE065 Fisefin HUCHE066 楊梅樹皮素(myricetin) HUCHE067 金聖草素(chrysoeriol) HUCHE068 Phloretin HUCHE069 Embelin HUCHE070 Tamarixetin HUCHE071 西阿多黃素(sciadopitysin) 桂皮 沒食子酸 芍藥 火刺木 积實 蔻花。蔻醇 1蒿莖桂 白細小月 蒿 陳藥根皮 茵當葛桂 小菫蔻 安腦樹 甘草 大棗 大棗 枳實 桂皮 桑白皮 芍藥 蜂膠 薑 芍藥’ 黃葵 傷寒草 蘋果 紫金牛 聖柳 銀杏The blood concentration of Fluvastatin was analyzed by High Performance Liquid Chromatography (HPLC). The detector was a fluorescence spectrophotometer. The excitation and scattered light were set to 309 nm and 390 nm, respectively. 5 μ, 150 X 4.6 mm, C18 reverse phase column, mobile phase was 〇 · Μ TBAF: 0 · 1 Μ phosphate buffer (pH 6.0 ·): MeOH (15: 25: 60 v / v), temperature control 50 The flow rate in ° C is set to 1 ml / min. For the blood test procedure, refer to Kalafsky et aL, (High-pbrformance liquid chromatographic method for the determination of fluvastatin in human plasma. Journal of Chromatography. 614: 307-313, 1993) And Toreson et al "(Determination of fluvastatin enantiomers and the racemate in human blood plasma by liquid chromatography and fluorometric detection. Journal of Chromatography A. 17 200533364 729: 13-18, 1996), as follows: (1) After the ice / dong test sample is taken out, put it on crushed ice to dissolve the beads; (2) Take 250 μ1 plasma test sample and make it in a spiral test tube; (3) Add 50 μ1 internal standard (c elec〇xib 2〇_ml ries; (4) Add Acetonitrile 250 μΐ and vibrate i 5, s Γ (5) add 0.5 M phosphate buffer ρΗ ^^. Ο 250 μΐ; ⑹ add 2.5 ml ΜΤΒΕ (methyl -tert seven utyl ether), shaking for 30 min; (7) Take the organic layer solution in a flat-mouth test tube, and concentrate it under reduced pressure to volatilize; (8) Re-dissolve with 200 μΐ mobile phase solution; * ⑼ Take the solution to the south speed Centrifuge at 13,000 rpm for 5 min; (10) Take the supernatant 150 μΐ and inject it into HPLC. Experimental results:. In vitro screening of traditional Chinese medicines cited HUCHE001 ~ HUCHE070, the degree to which each component inhibits the metabolism of microsomes tolbutamide is divided into 1,1. , 100μM, some traditional Chinese medicines have poor solubility, only the soluble concentration is the highest inhibitory concentration, and then diluted to the middle and low settings in order. The experimental results are set according to the inhibitor settings of high, medium and low. Different concentrations are divided into three categories. The best inhibitors are: HUCHE009 isdiquritigenin 100μM inhibits microsomes up to 95.5%, HUCHE070 Tamarixetin 10 μM inhibits 88.2%, HUCHE037 genistein isoflavone Genistein 1 μM inhibits 49 6% According to traditional Chinese medicine-numbered finishing shown in Table 4 to Table VI. Table 3. Introduction to Chinese Medicine Sources 200533364 No. Pure Ingredient Name Raw Medicine Source Raw Medicine Name HUCHE001 Genkwanin Astemisiae Capillaris HUCHE002 Apigenin Chamomillae Flos HUCHE003 Digitalis Folium HUCHE004 Luteolin-7-Glucoside Digitals Folium HUCHE005 Homoorientin Swerticie Herba HUCHE006 Isovitexin Swerttae Herba HUCHE007 New Orange Skin (Neeohesperidin) Anrantii Fructus Immatums HUCHE008 Formononetin Astragali Radix HUCHE009 Isoliquritigenin Astragali Radix HUCHE010 Kaempferol Sennae Folium Plum (Isorhamnetin) Senna Sennae Folium HUCHE012 isoquercitrin (Isoquercitrin) Sweet tea Hydrangeae Dulcis Folium HUCHE013 (+)-epicatechin (+)-epicatechin 児 茶 Gambir HUCHE014 Ergot ergot. Ergota HUCHE015 (+) Cate chin Paeoniae Radix HUCHE016 6-Gingerol Ginger Zingiberis Rhizoma HUCHE017 Liquiritin Glycyrrhizae Radix HUCHE018 'Phenylpropyl Acetate Cinnamami Cortex HUCHE019 ㈠ epicatechin (-) -Epicatechin Cosmos Gambir HUCHE020 Narigenin Aurantii Fructus Immatums HUCHE021 Umbelliferone Aurantii Fructus Immaturns HUCHE022 Rutin Sophorae Flos HUCHE023 Lemon Yellow Aurantii Fructus Immaturus HUCHE024 Diosmin —— _ HUCHE025 Hesperetin Citri Reticulatae HUCHE026 Wongonin Scutellariae Radix HUCHE027 Baicalin Scutellariae Radix HUCHE028 HUCHE029 Puerariain Pueraria Radix HUCHE030 Lignin Isoflavone Daidzein Pueraria Radix HUCHE031 Daidzin Pueraria Radix HUCHE032 Quercitrin) 獬 皮 Viscum Coloratum HUCHE033 Quercetin or quercetin Viscnm Coloratum HUCHE034 ~: (Nordihydroguaiaretic acid) Table III 19 200533364 Table III. Introduction to Chinese Medicine Sources (continued) No. Pure Ingredients Name of crude drug, source of raw drug name HUCHE035 fe] Chen Se original surface I (Capillarisin) HUCHE036 Swertiamarin HUCHE037 Genistein HUCHE038 trans-Cinnamaldehyde HUCHE039 protocatechuic acid HUCHE040 not Gallic acid HUCHE041 paeoniflorin HUCHE042 eriodictyol HUCHE043 Poncirin HUCHE044 flavones (a -Naphthoflavone) HUCHE045 laurel roast (0 -Myrcene) HUCHE046 α-terpineol (α-terpineol) terpineol) HUCHE047 (+)-Lemon essential oil (+) -Limonene HUCHE048 Lauryl Alcohol HUCHE049 Ethyl Myristate HUCHE050 Cineole HUCHE051 Glycyrrhizin HUCHE052 Oleanolic acid) HUCHE053 ursolic acid HUCHE054 Pomelo bitter Narigin HUCHE055 yS-Naphthoflavone HUCHE056 trans-cinnamic acid HUCHE061 Morin HUCHE062 (+)-Taxifolin HUCHE063 Chrysin HUCHE064 Galangin HUCHE065 Fisefin HUCHE066 Myrictin St. HUCHE0 Chrysoeriol HUCHE068 Phloretin HUCHE069 Embelin HUCHE070 Tamarixetin HUCHE071 sciadopitysin Cassia gallic acid paeony fire thorn wood sago flower. Myristyl alcohol 1 Artemisia sylvestris var. Artemis sylvestris Artemisia radix bark Yindang Gegui Berberis sylvestris scabra licorice Jujube Jujube Jujube Cinnamon mulberry peony Peony Propolis Ginger Paeonia oleifera Mallow typhus apple purple golden cow holy willow ginkgo

Artemisia Capillaris Swertiae Herba Puerariae RadixArtemisia Capillaris Swertiae Herba Puerariae Radix

Cinnamami Cortex Cinnamami CortexCinnamami Cortex Cinnamami Cortex

Paeoniae Radix Pyracantha Fortnneana Aurantii Fructus lmmaturusPaeoniae Radix Pyracantha Fortnneana Aurantii Fructus lmmaturus

Amomum cardamomum Cinae Flos Cardamomi FructusAmomum cardamomum Cinae Flos Cardamomi Fructus

Cardamomi FructusCardamomi Fructus

Cinae Flos Glycyrrhizae Radix Zizyphi Fructus Zizyphi Fructus Aurantii Fructus lmmaturusCinae Flos Glycyrrhizae Radix Zizyphi Fructus Zizyphi Fructus Aurantii Fructus lmmaturus

Cinnamoni CortexCinnamoni Cortex

Mori Radix Cortex Paeoniae Radix PropolisMori Radix Cortex Paeoniae Radix Propolis

Zingiberis Rhizoma Paeoniae Radix Hibiscus Abelmoschas Vernonia Cinerea AppleZingiberis Rhizoma Paeoniae Radix Hibiscus Abelmoschas Vernonia Cinerea Apple

Ardisia Squamulosa Tamarix Ramosissimci Ginko Biloba 表三(續) 20 200533364 表四、中藥藥引純成分100 μΜ對酵素活性之抑制程度 排名 純成分名稱 抑制劑濃度 % inhibition SD - Ketoconazole 100 μΜ 100.00 0.00 1 异苷草元(isoliquritigenin) 100 μΜ 95.47 0.15 2 Phloretin 100 μΜ 95.13 0.62 3 木犀草素(luteolin) 100 μΜ 93.20 0.94 4 槲皮素(quercetin) 100 μΜ 91.92 0.52 5 Tamarixetin Η)0μΜ 90.18 0.43 6 楊梅樹皮素(myricetin) 100 μΜ 88.84 3.37 7 漢黃芩素(Wongonin) 100 μΜ 84.03 2.22 8 金雀異黃酮(Genistein) 100 μΜ ^ 82.71 2.82 9 正二羥癒瘡酸 (Nordihydroguaiaretic acid) 100 μΜ 81.18 0.50 10 柚皮素(Narigenin) 100 μΜ 79.70 - 11 茵陳色原酮(Capillarisin) 100 μΜ 79.49 3.22 12 Chrysin 50 μΜ 75.11 6.05 13 Fisefin 100 μΜ » 72.89 3.37 f4 聖草素(eriodictyol) 100 μΜ 69.62 5.68 15 6-薑辣素(6- Gingerol) 100 μΜ 66.21 1.94 16 異鼠李素(Isorhamnetin) 75 μΜ 65.74 4.99 17 異榭皮苷(i soquerdtri η) 100 μΜ 61.80 15.60 18 花黃素異黃 9¾ (Formononetin) 50 μΜ 57.94 0.84 19 Morin 100 μΜ 51.00 4.55 20 (+)-Taxifolin 100 μΜ 50.47 10.38 21 異牡莉素(isovitexin) 100 μΜ 45.36 0.97 22 乙酸苯丙 S旨(3-Phenylpropyl Acetate ) 100 μΜ 42.62 2.00 23 齊墩果酸(Oleanolic acid) 100 μΜ 41.13 11.52 24 熊果酸(ursolic acid) 100 μΜ 38.47 3.37 25 葛根素(Puerarin) 100 μΜ 33.30 17.52 26 月桂烯(/3 -Myrcene) 100 μΜ " 29.85 4.31 27 反-桂皮酸(trans-cinnamic acid) 100 μΜ 26.10 3.57 28 木犀草素-7-0-葡萄糖苷 (Luteolin-7-Glucoside) 100 μΜ 25.08 1.57 29 甘草萃取(Liquiritin) 100 μΜ 24.77 8.72Ardisia Squamulosa Tamarix Ramosissimci Ginko Biloba Table III (continued) 20 200533364 Table IV. Inhibition of Enzyme Activity by Pure Ingredients of Chinese Medicine 100 μΜ Ranking of Pure Ingredient Name Inhibitor Concentration% inhibition SD-Ketoconazole 100 μΜ 100.00 0.00 1 Isoglycone (isoliquritigenin) 100 μM 95.47 0.15 2 Phloretin 100 μM 95.13 0.62 3 luteolin 100 μM 93.20 0.94 4 quercetin 100 μM 91.92 0.52 5 Tamarixetin Η) 0 μM 90.18 0.43 6 Myricetin 100 μM 88.84 3.37 7 Wongonin 100 μM 84.03 2.22 8 Genistein 100 μM ^ 82.71 2.82 9 Nordihydroguaiaretic acid 100 μM 81.18 0.50 10 Naringin 100 μΜ 79.70-11 Capillarisin 100 μΜ 79.49 3.22 12 Chrysin 50 μΜ 75.11 6.05 13 Fisefin 100 μΜ »72.89 3.37 f4 eriodictyol 100 μΜ 69.62 5.68 15 6-Gingerol 100 μM 66.21 1.94 16 Isorhamnetin 75 μM 65.74 4.99 17 isoxperidin ( i soquerdtri η) 100 μΜ 61.80 15.60 18 Formononetin 50 μΜ 57.94 0.84 19 Morin 100 μΜ 51.00 4.55 20 (+)-Taxifolin 100 μΜ 50.47 10.38 21 isovitexin 100 μΜ 45.36 0.97 22 3-Phenylpropyl Acetate 100 μΜ 42.62 2.00 23 Oleanic acid 100 μΜ 41.13 11.52 24 Ursolic acid 100 μΜ 38.47 3.37 25 Puerarin 100 μM 33.30 17.52 26 Myrcene 100 μM " 29.85 4.31 27 trans-cinnamic acid 100 μM 26.10 3.57 28 Luteolin-7-Glucoside 100 μΜ 25.08 1.57 29 Liquiritin 100 μΜ 24.77 8.72

表四 21 200533364 表四、中藥藥引纯成分100 μΜ對酵素活性之抑制程度(續) 排名 純成分名稱 抑制劑濃度 % inhibition SD 30 (+)-檸檬精油(+ ) -Limonene 100 μΜ 22.29 4.04 31 異紅草素(Homoorientin) 100 μΜ 20.19 11.59 32 獐牙菜苦(Swertiamarin) 100 μΜ 18.44 2.11 33 Embelin 50 μΜ 17.98 4.20 34 木質素異黃酮Daidzein 25 μΜ 15.74 3.24 35 积屬(Poncirin) β 100 μΜ 14.99 12.51 36 懈皮苦(Quercitrin) 100 μΜ 13.48 15.69 37 (-)表兒茶素㈠-Epicatechin 100 μΜ 5.44 4.90 38 甘草甜素(glycyrrhizin) 100 μΜ 4.87 2.73 39 麥角固醇(ergosterol) 30 μΜ 3.57 2.64 40 香葉木(Diosmin) 50 μΜ 3.51 1.99 41 (+)兒茶紛素(+)Catechin 100 μΜ -0.22 6.94 42 沒食子酸(gallic acid) 100 μΜ -0.97 16.40 43 大豆素(Daidzin ) 25 μΜ ΛΛ6 6.54 44 大豆素(Daidzin ) 100 μΜ -1.33 7.67 45 芍藥素(paeoniflorin) ’ 100 μΜ -1.77 3.49 46 繳形花内酯(Umbelliferone ) 100 μΜ -2.02 5.27 47 芸香素(Rutin) 100 μΜ -6.46 13.80 48 (+)-表兒茶素(+)-epicatechin 100 μΜ -11.54 0.77 49 柚苷 Narigin 100 μΜ -24.21 10.50 表四(續) 22 200533364 表五、中藥藥引纯成分10 μΜ對酵素活性之抑制程度 排名 純成分名稱 抑制劑濃度 % inhibition SD - Ketoconazole 10 μΜ 80.11 0.71 1 Tamarixetin 10 μΜ 88.12 0.69 2 芹菜素(apigenin) 25 μΜ 76.88 1.37 3 金雀異黃酮Genistein 10 μΜ 67.70 2.28 4 異鼠李素(Isorhamnetin) 10 μΜ 。 61.53 3.57 5 Chrysin 10 μΜ 60.62 2.07 6 漢黃答素(Wongonin) 10 μΜ 51.31 1.43 7 柚皮素(Narigenin) 10 μΜ 49.98 - 8 槲皮素(quercetin) 10 μΜ 44.80 2.37 9 齊墩果酸(Oleanolic acid) 10 μΜ 42.35 9.56 10 葛根素(Puerarin) 10 μΜ。 39.02 10.00 11 奈紛(kaempferol) 10 μΜ 38.29 15.43 12 木犀草素(luteolin) 10 μΜ 37.89 14.42 13 熊果酸(ursolic acid) 10 μΜ 37.46 3.31 14 異牡荆素(isovitexin) 10 μΜ 37.38 5.79 15 荛花素(Genkwanin) 黃酮 10 μΜ 37.37 3.64 16 (a -Naphthoflavone ) 10 μΜ 37.27 7.06 17 茵陳色原酮(Capillarisin) 10 μΜ 。 34.79 3.04 18 ' Phloretin 10 μΜ 34.41 7.95 19 ㈠表兒茶素(-)-Epicatechin 10 μΜ 33.75 13.74 20 (+)-Taxifolin 10 μΜ 31.16 8.11 21 花黃素異黃酮(Formononetin) 10 μΜ 30.57 3.69 22 异苷草元(isoliquritigenin) 10 μΜ 29.66 14.74 23 橙皮素(Hesperetin) 10 μΜ 29.09 。2.10 24 聖草素(eriodictyol) 10 μΜ 28.65 15.29 25 6-薑辣素(6- Gingerol) 10 μΜ 27.72 10.54 26 異槲皮苦(isoquercitrin) 10 μΜ 27.02 17.78 27 Fisefm 10 μΜ 26.52 7.25 28 懈皮皆(Quercitrin) 10 μΜ 21.10 15.81 29 甘草萃取(Liquiritin) 10 μΜ 18.35 1.97 表五 23 200533364 表五、中藥藥引纯成分10 μΜ對酵素活性之抑制程度(續) 排名 純成分名稱 抑制劑濃度 % inhibition SD 30 月桂烯(/3 -Myrcene ) 10 μΜ 16.60 6.31 31 獐牙菜苦(Swertiamarin) 10 μΜ 16.56 3.84 32 松屬(Poncirin) 10 μΜ 16.34 10.77 33 原兒茶酸(protocatechuic acid) 10 μΜ 16.22 L72 34 反-桂皮酸(trans-cinnamic acid) 10 μΜ 15.82 9.04 35 木質素異黃酮(Daidzein) 10 μΜ 13.45 4.49 36 Morin 10 μΜ 11.63 17.51 37 Embelin 10 μΜ 11.23 9.18 38 揚梅樹皮素(myricetin) 10 μΜ 10.57 13.21 39 (+)-檸檬精油(+ ) -Limonene 正 二 羥 癒 瘡 酸 10 μΜ 10.55 4.18 40 (Nordihydroguaiaretic acid) 10 μΜ 9.76。 5.26 41 麥角固醇(ergosterol) 10 μΜ 8.12 ^ 2.19 42 黃答苷(baicalin) 25 μΜ . 7.77 3.08 43 檸檬黃素(Hesperidin) 10 μΜ 6.68 332 44 (+)-表兒茶素(+)-epicatechin 10 μΜ 6.30 3.72 45 黃答皆Baicalein 25 μΜ 5.06 8.64 46 香葉木(Diosmin) 10 μΜ 4.70 0.75 47 黃酮(yS-Naphth〇flavone) 10 μΜ 4.64 3.02 48 異紅草素(Homoorientin) 10 μΜ 2.45 13.94 49" 甘草甜素(glycyrrhizin) 10 μΜ 2.23 4.65 50 芍藥素(paeoniflorin ) 木犀草素-ίο-葡萄糖苷 10 μΜ 0.70 3.50 51 (Luteolin-7-Glucoside) 10 μΜ -0.32 5.2〇 52 大豆素(Daidzin ) 10 μΜ ‘ -2.46 4.10 53 沒食子酸(gallic acid) 10 μΜ -2.47 10.16 54 繳形花内 3旨(Umbelliferone ) 10 μΜ -6.64 4.94 55 (+)兒茶盼素(+)Catechin 10 μΜ -8.46 3.53 56 ㈠表兒茶素㈠-Epicatechin 10 μΜ -8.61 5.95 57 柚 ^(Narigin) 10μΜ -13.25 4.33 58 芸香素(Rutin) 10 μΜ -13.97 14.31Table IV 21 200533364 Table IV. Inhibition of enzyme activity by 100 μM pure ingredients of traditional Chinese medicines (continued) Ranking pure ingredient name inhibitor concentration% inhibition SD 30 (+)-lemon essential oil (+) -Limonene 100 μM 22.29 4.04 31 Homoorientin 100 μM 20.19 11.59 32 Swertiamarin 100 μM 18.44 2.11 33 Embelin 50 μM 17.98 4.20 34 Lignin Isoflavone Daidzein 25 μM 15.74 3.24 35 Poncirin β 100 μM 14.99 12.51 36 Quercitrin 100 μM 13.48 15.69 37 (-) Epicatechin-Epicatechin 100 μM 5.44 4.90 38 Glycyrrhizin 100 μM 4.87 2.73 39 ergosterol 30 μM 3.57 2.64 40 Diosmin 50 μΜ 3.51 1.99 41 (+) Catechin 100 μM -0.22 6.94 42 gallic acid 100 μM -0.97 16.40 43 Daidzin 25 μΜ ΛΛ6 6.54 44 Daidzin 100 μM -1.33 7.67 45 paeoniflorin '100 μM -1.77 3.49 46 Umbelliferone 100 μM -2.02 5.27 47 Rutin Rutin) 100 μM -6.46 13.80 48 (+)-epicatechin (+)-epicatechin 100 μM -11.54 0.77 49 Naringin Narigin 100 μM -24.21 10.50 Table 4 (continued) 22 200533364 Table 5. Purified ingredients of traditional Chinese medicine Degree of inhibition of enzyme activity by 10 μΜ Ranking pure ingredient name Inhibitor concentration% inhibition SD-Ketoconazole 10 μΜ 80.11 0.71 1 Tamarixetin 10 μΜ 88.12 0.69 2 apigenin 25 μΜ 76.88 1.37 3 Genistein 10 μM 67.70 2.28 4 Isorhamnetin 10 μM. 61.53 3.57 5 Chrysin 10 μΜ 60.62 2.07 6 Wongonin 10 μΜ 51.31 1.43 7 Narigenin 10 μΜ 49.98-8 Quercetin 10 μΜ 44.80 2.37 9 Oleanic acid 10 μM 42.35 9.56 10 Puerarin 10 μM. 39.02 10.00 11 kaempferol 10 μΜ 38.29 15.43 12 luteolin 10 μΜ 37.89 14.42 13 ursolic acid 10 μΜ 37.46 3.31 14 isovitexin 10 μΜ 37.38 5.79 15 Genkwanin flavones 10 μM 37.37 3.64 16 (a-Naphthoflavone) 10 μM 37.27 7.06 17 Capillarisin 10 μM. 34.79 3.04 18 'Phloretin 10 μΜ 34.41 7.95 19 Epi-catechin (-)-Epicatechin 10 μΜ 33.75 13.74 20 (+)-Taxifolin 10 μΜ 31.16 8.11 21 Formononetin 10 μΜ 30.57 3.69 22 Isoglycoside Isoliquritigenin 10 μM 29.66 14.74 23 Hesperetin 10 μM 29.09. 2.10 24 eriodictyol 10 μΜ 28.65 15.29 25 6-Gingerol 10 μΜ 27.72 10.54 26 isoquercitrin 10 μΜ 27.02 17.78 27 Fisefm 10 μΜ 26.52 7.25 28 Quercitrin) 10 μΜ 21.10 15.81 29 Liquiritin 10 μΜ 18.35 1.97 Table 5 23 200533364 Table 5: Inhibition of Enzyme Activity by Pure Ingredients of Traditional Chinese Medicine 10 μΜ (Continued) Ranking Pure Ingredient Name Inhibitor Concentration% inhibition SD 30 Myrcene (/ 3 -Myrcene) 10 μΜ 16.60 6.31 31 Swertiamarin 10 μΜ 16.56 3.84 32 Poncirin 10 μΜ 16.34 10.77 33 protocatechuic acid 10 μΜ 16.22 L72 34 trans- Trans-cinnamic acid 10 μΜ 15.82 9.04 35 lignin isoflavone (Daidzein) 10 μΜ 13.45 4.49 36 Morin 10 μΜ 11.63 17.51 37 Embelin 10 μΜ 11.23 9.18 38 myricetin 10 μΜ 10.57 13.21 39 ( +)-Lemon essential oil (+)-Limonene 10 μM 10.55 4.18 40 (Nordihydroguaiaretic acid) 10 μM 9.76. 5.26 41 ergosterol 10 μM 8.12 ^ 2.19 42 baicalin 25 μM. 7.77 3.08 43 Hesperidin 10 μM 6.68 332 44 (+)-epicatechin (+)- epicatechin 10 μΜ 6.30 3.72 45 Huang Dajie Baicalein 25 μΜ 5.06 8.64 46 Diosmin 10 μΜ 4.70 0.75 47 yS-Naphth flavone 10 μΜ 4.64 3.02 48 Homoorientin 10 μΜ 2.45 13.94 49 & quot Glycyrrhizin 10 μM 2.23 4.65 50 paeoniflorin-Luo-glucoside 10 μM 0.70 3.50 51 (Luteolin-7-Glucoside) 10 μM -0.32 5.2〇52 Daidzin 10 μΜ '-2.46 4.10 53 gallic acid 10 μΜ -2.47 10.16 54 Umbelliferone 10 μΜ -6.64 4.94 55 (+) Catechin (+) Catechin 10 μΜ -8.46 3.53 56 Epiatechin-Epicatechin 10 μM -8.61 5.95 57 Citrus ^ (Narigin) 10 μM -13.25 4.33 58 Rutin 10 μM -13.97 14.31

表五(續) 24 200533364 表六、中藥藥引純成分1 μΜ對酵素活悻之抑制程度 排名 純成分名稱 抑制劑濃度 % inhibition SD - Ketoconazole 1 μΜ 45.88 。3·13 1 金雀異黃8¾ Genistein 1 μΜ 49.60 1.37 2 Tamarixetin 1 μΜ 41.96 6.63 3 葛根素(Puerarin ) 乙酸苯丙酯(3-Phenylpropyl ϊ μΜ 38.15 0.57 4 Acetate ) 1 μΜ 36.57 7.30 5 異牡荊素(isovitexin) 1 μΜ 35.56 7.96 6 熊果酸(ursolic acid) 1 μΜ 33.62 0.99 7 聖草素(eriodictyol) 1 μΜ 32.78 4.41 8 薨花素(Genkwanin) 1 μΜ 30.85 1.68 9 6-薑辣素(6- Gingerol) 1 μΜ 30.17 2.36 10 漢黃芩素(Wongonin) 1 μΜ 28.82 1.41 11 反-桂皮酸(trans-cinnamic acid) 1 μΜ 26.92 4.26 12 Embelin 1 μΜ 24.71 6.18 13 懈皮發(Quercitrin) 1 μΜ 24.19 . 1.71 14 月桂烤(/3 -Myrcene ) 1 μΜ » 24.06 3.08 15 Phloretin 1 μΜ 23.76 6.21 16 花黃素異黃嗣(Formononetin) 1 μΜ 23.33 0.43 Π 芹菜素(apigenin) 2.5 μΜ 21.69 1.37 18 異槲皮苦(isoquercitrin) 1 μΜ 20.94 1.96 19 原兒茶酸(protocatechuic acid) 1 μΜ 20.26 9.00 20 木犀草素(luteolin) 1 μΜ 。 20.09 21.27 21 異鼠李素(Isorhamnetin) 1 μΜ 19.63 6.32 22 茵陳色原酮(Capillarisin) 1 μΜ 19.33 7.81 23 甘草萃取(Liquiritin) 1 μΜ 18.10 9.70 24 (+)-表兒茶素CKKpicatechin ΙμΜ. 16.99 2.53 25 齊墩果酸(Oleanolic acid ) 1 μΜ 16.79 L67 26 獐牙菜苦(Swertiamarin ) 1 μΜ 16.33 。0.92 27 槲皮素(quercetin) 1 μΜ 15.11 1.03 28 Morin 1 μΜ 14.26 2.86 29 (+)-檸檬精油(+ ) -Limonene 1 μΜ 14.12 3.63Table 5 (continued) 24 200533364 Table 6. Degree of inhibition of enzyme activity by 1 μΜ of pure ingredients of traditional Chinese medicines Ranking Pure ingredient name Inhibitor concentration% inhibition SD-Ketoconazole 1 μΜ 45.88. 3.13 1 genistein 8¾ Genistein 1 μM 49.60 1.37 2 Tamarixetin 1 μM 41.96 6.63 3 Puerarin 3-Phenylpropyl ϊ μΜ 38.15 0.57 4 Acetate) 1 μM 36.57 7.30 5 isovitretin (isovitexin) 1 μΜ 35.56 7.96 6 ursolic acid 1 μΜ 33.62 0.99 7 eriodictyol 1 μΜ 32.78 4.41 8 Genkwanin 1 μΜ 30.85 1.68 9 6-Gingerin (6- Gingerol) 1 μΜ 30.17 2.36 10 Wongonin 1 μΜ 28.82 1.41 11 trans-cinnamic acid 1 μΜ 26.92 4.26 12 Embelin 1 μΜ 24.71 6.18 13 Quercitrin 1 μΜ 24.19 .1.71 14 Laurel (/ 3 -Myrcene) 1 μΜ »24.06 3.08 15 Phloretin 1 μΜ 23.76 6.21 16 Formononetin 1 μΜ 23.33 0.43 Π apigenin 2.5 μΜ 21.69 1.37 18 isoquercitrin) 1 μM 20.94 1.96 19 protocatechuic acid 1 μM 20.26 9.00 20 luteolin 1 μM. 20.09 21.27 21 Isorhamnetin 1 μM 19.63 6.32 22 Capillarisin 1 μM 19.33 7.81 23 Liquiritin 1 μM 18.10 9.70 24 (+)-epicatechin CKKpicatechin ΙμΜ. 16.99 2.53 25 Oleanic acid 1 μM 16.79 L67 26 Swertiamarin 1 μM 16.33. 0.92 27 quercetin 1 μM 15.11 1.03 28 Morin 1 μM 14.26 2.86 29 (+)-Lemon essential oil (+) -Limonene 1 μM 14.12 3.63

25 200533364 表六、中藥藥引纯成分1 μΜ對酵素活性之抑制程度(續) 排名 純成分名稱 抑制劑濃度 % inhibition SD 30 芍藥素(paeoniflorin) 木犀草素-7-0-葡萄糖皆 1 μΜ 10.11 4.34 31 (Luteol iri-7~Glucoside) 1 μΜ 937 。 3.17 32 枳屬(Pondrin) 1 μΜ 7.76 6.36 33 Chrysin 1 μΜ 6.86 2.17 34 Fisefin 1 μΜ 5.49 7.50 35 柚皮素(Narigenin) 1 μΜ 5.20 - 36 甘草甜素(glycyrrhizin) 1 μΜ 5.14 6.63 37 異紅草素(Homoorientin) 1 μΜ 3.37 8.22 38 檸檬黃素(Hesperidin) 1 μΜ ^ 2.57 2.07 39 黃酮(/3-Naphthoflavone) 1 μΜ 2.35 4.87 40 黃答* Baicalein 2.5 μΜ 1.76 2.53 41 香葉木(Diosmin) 1 μΜ 1,51 0.82 42 木質素異黃嗣Daidzein 1 μΜ 1.35 1.54 43 ㈠表兒茶素㈠-Epicatechin 1 μΜ .1.11 4.15 44 麥角固醇(ergosterol) 1 μΜ 1.00 0.59 45 大豆素(Daidzin ) 1 μΜ 0.95 3.51 46 异苷草元(isoliquritigenin) 1 μΜ 0.87 5.00 47 黃®(a-Naphthoflavone) 1 μΜ -0.05 6.26 48 (+)-Taxifolin 1 μΜ -1.29 8.16 49 芸香素(Rutin) 1 μΜ -2.59 12.71 50 沒食子酸(gallic acid) 1 μΜ -3.05 5.18 51 (+)兒茶紛素(+)Catechin ΙμΜ。 :3.05 0.78 52· 楊梅樹皮素(myricetin) 1 μΜ -3.19 16.64 53 撥皮素(Hesperetin) ΙμΜ -3.58 11.11 54 黃答苦(baicalin) 2.5 μΜ -5.36 6.97 55 繳形花内 _ ( Umbelliferone ) ΙμΜ -7.17 3.59 56 柚苷 Narigin 正二經癒瘡酸(Nordihydroguaiaretic ΙμΜ -11.48 2.10 57 acid) ΙμΜ -16.06 。 2·77 58 奈紛(kaempferol) ΙμΜ -2227 18.9625 200533364 Table 6. Inhibition of Enzyme Activity by Pure Ingredients of Chinese Medicine 1 μM (continued) Ranking Pure Ingredient Name Inhibitor Concentration% Inhibition SD 30 Paeoniflorin Luteolin-7-0-glucose 1 μM 10.11 4.34 31 (Luteol iri-7 ~ Glucoside) 1 μM 937. 3.17 32 Pondrin 1 μM 7.76 6.36 33 Chrysin 1 μM 6.86 2.17 34 Fisefin 1 μM 5.49 7.50 35 Narigenin 1 μM 5.20-36 glycyrrhizin 1 μM 5.14 6.63 37 Isorhodin (Homoorientin) 1 μΜ 3.37 8.22 38 Hesperidin 1 μΜ ^ 2.57 2.07 39 Flavones (/ 3-Naphthoflavone) 1 μΜ 2.35 4.87 40 Yellow A * Baicalein 2.5 μΜ 1.76 2.53 41 Diosmin 1 μΜ 1, 51 0.82 42 Lignin Isoxanthin Daidzein 1 μM 1.35 1.54 43 Epiatechin-Epicatechin 1 μM .1.11 4.15 44 ergosterol 1 μM 1.00 0.59 45 Daidzin 1 μM 0.95 3.51 46 Isoliquritigenin 1 μM 0.87 5.00 47 yellow (a-Naphthoflavone) 1 μM -0.05 6.26 48 (+)-Taxifolin 1 μM -1.29 8.16 49 Rutin 1 μM -2.59 12.71 50 Gallia Acid (gallic acid) 1 μM -3.05 5.18 51 (+) Catechin (+) Catechin 1 μM. : 3.05 0.78 52 · Myricetin 1 μΜ -3.19 16.64 53 Hesperetin ΙμΜ -3.58 11.11 54 baicalin 2.5 μΜ -5.36 6.97 55 (Umbelliferone) ΙμΜ- 7.17 3.59 56 Naringin Narigin Ordihydroguaiaretic 1 μM -11.48 2.10 57 acid 1 μM -16.06. 2.77 58 kaempferol ΙμΜ -2227 18.96

表六(續) 26 200533364 .·以統計檢定㈣dent T-test)各中藥藥引抑制μ·。·程度相對於· ⑽加丨g歷p是否達顯著性差異,中_引刚,叫丨_三種抑制濃度經筛 選所得抑制效果計名的巾賴㈣如圖二、圖三及圖四所示。 較佳實施例1之詳細說明 + 利用上述的實驗方法賴不同濃度的而咖_ Tamarixetin對於肝 微粒體酶韻tdbutamide之影響,妓祕件為·· tGlbutamide濃度為】 mM,Mi__蛋白質含量為0·5 mg,反應時間為7·5分鐘。測試絲顯 示編號HUCHE070Tamarixetin抑制劑,在高、中、低三種不同濃度下分別 具有90.2 %,88.1 %,42.0 %代謝抑制程度(如表七與圖五所示),可視為一Table 6 (continued) 26 200533364... (Stat T-test). · The degree is relative to the value of ⑽ plus 丨 g calendar p has a significant difference, medium _ cited Gang, called 丨 _ three kinds of inhibitory concentration of the inhibitory effect obtained by screening, the name of the towel is shown in Figures 2, 3 and 4 Show. Detailed description of the preferred embodiment 1 + The above-mentioned experimental method depends on the effects of different concentrations of tamarinxetin on the liver microsomal enzyme rhyme tdbutamide. The prostitutes are ... tGlbutamide concentration is mM, Mi__ protein content is 0 5 mg with a reaction time of 7.5 minutes. The test wire shows the number HUCHE070Tamarixetin inhibitor, which has 90.2%, 88.1%, and 42.0% metabolic inhibition levels at three different concentrations: high, medium, and low (as shown in Table 7 and Figure 5).

有效 CYP450 isoenzyme 調控劑。 表七、體外篩選中藥藥引HUCHE〇7〇 Tamarixetin不同濃度對於肝 微粒 體酶代謝tolbutamide之影響(n=3) 〇 濃度 4-ΟΗ (ng) % inhibition Control 368.5409±35.3091 0.0000 1 μΜ 213.5696 ±24.4309 41.9620 10Mm 43.10052 ±2.5372 88.1204 . 100 μΜ 35.49297±1.5825 90.1803 表七 ----jEffective CYP450 isoenzyme modulator. Table VII. In vitro screening of Chinese herbal medicines HUCHE〇7〇The effect of different concentrations of Tamarixetin on liver microsomal enzyme metabolism tolbutamide (n = 3) 〇Concentration 4-〇Η (ng)% inhibition Control 368.5409 ± 35.3091 0.0000 1 μΜ 213.5696 ± 24.4309 41.9620 10Mm 43.10052 ± 2.5372 88.1204. 100 μM 35.49297 ± 1.5825 90.1803 Table 7 ---- j

Tamarixetin對降血脂藥物fluvastatin於SD rat之口服吸收影響的實驗鈐春 果顯不’實驗組(表九)SD rat 口服fluvastatin 1·5 mg/kg合併使用jjuche〇7〇 西阿多黃素(sdad〇Pitysin)9.32 mg/kg (n=5),藥物血中濃度數據如表九,控 制·組(表八)SD rat (rv=7)合併服用溶解抑制劑之溶媒,兩組血中濃度經計算 後動力學參數列於表十,血中濃度經時曲線如圖六所示,由動力 學參數顯示,曲線下面積(areauncierthecurve,AUC)具有顯著性差異,最古 血中濃度(Cmax)實驗組為141·4 ± β·8 ng/m卜控制組為63」± 1〇4 ng/ml,濃度大約增加一倍,藥物清除率(c1t)及分佈體積(Vd)都具有顯 著性差異,表示肝臟酵素代謝藥物的能力已被明顯抑制,由於後段排除相 27 200533364 ,除常數(k)並沒有顯著差異,因此不改變體内藥物排除半衰期(Τι/〗),此 、'果顯示服用抑制劑HUCHEOTO Tamarixetin並不會怪久性的抑制_ 〆否性,而fluvastatin在動物體内仍可依循正常排除過程排出體P酵素代謝 表八、口服fluvastatin控制組SD rat血中濃度數據 —°Experimental study on the effect of Tamarixetin on oral absorption of the lipid-lowering drug fluvastatin in SD rats. Experimental group (Table 9) SD rat oral fluvastatin 1.5 mg / kg in combination with jjuche〇7〇 〇 Pitysin) 9.32 mg / kg (n = 5). The blood concentration data of the drug are shown in Table 9. The control group (Table 8) SD rat (rv = 7) was combined with a dissolution inhibitor solvent. The calculated kinetic parameters are listed in Table 10. The curve of blood concentration over time is shown in Figure 6. The kinetic parameters show that the area under the curve (areauncierthecurve (AUC)) has significant differences. The most ancient blood concentration (Cmax) experiment The control group was 141.4 ± β · 8 ng / m. The control group was 63 ″ ± 104 ng / ml. The concentration was approximately doubled. The drug clearance rate (c1t) and distribution volume (Vd) had significant differences. It indicates that the ability of liver enzymes to metabolize drugs has been significantly inhibited. Because of the exclusion phase 27 200533364 in the latter stage, there is no significant difference in the division constant (k), so it does not change the half-life of drug exclusion in the body (Ti /). Agent HUCHEOTO Tamarixetin does not blame for a long time _ No, but fluvastatin can still excrete P enzyme metabolism in the body according to the normal elimination process. Table VIII. SD rat blood concentration data of oral fluvastatin control group — °

Time ^ C_2 C"3 C"4 c-5 C-6 C-7 mean SB Concentration (ng/ml) CV〇/0 l〇 2〇 4〇 6〇 !2〇 24〇 36〇 48〇 720 13·56 16.39 19.78 24.70 0.27 8.94 16.41 14.3 3 〇 28.95 18.03 18.10 24.48 1.15 19.79 31.50 20.3 3.8 47.38 22.74 25.67 29.33 7.35 63.98 42.36 34.1 7q 69.48 24.74 34.71 41.24 7.57 99.41 44.17 45.9 \χ 4 61.69 29.13 43.91 54.63 11.19 98.41 41.14 48.6 i〇 4 54.64 38.66 66.97 73.42 14.48 68.72 38.28 50.7 8 1 40.68 45.74 60.75 83.84 20.41 50.80 27.07 47·〇 § 〇 37.37 57.30 45.05 54.92 21.57 37.68 24.92 39.8 5 2 22.45 37.37 25.63 39.30 21.75 18.47 15.40 25.8 35 1〇δ〇 16.11 35.39 22.58 36.80 14.10 10.67 11.68 21.1 〇 11·47 22.33 21.04 - 10.74 6.33 8.58 13.4 2/7 表八 55.2 49.1 54.5 65.9 56.6 42.1 45.2 34.4 35.6 52.6 51.Time ^ C_2 C " 3 C " 4 c-5 C-6 C-7 mean SB Concentration (ng / ml) CV〇 / 0 l〇2〇4〇6〇! 2〇24〇36〇48〇720 13 · 56 16.39 19.78 24.70 0.27 8.94 16.41 14.3 3 〇28.95 18.03 18.10 24.48 1.15 19.79 31.50 20.3 3.8 47.38 22.74 25.67 29.33 7.35 63.98 42.36 34.1 7q 69.48 24.74 34.71 41.24 7.57 99.41 44.17 45.9 \ χ 4 61.69 41.13 11.91 43.91 41.19 11.13 43.91 54.64 38.66 66.97 73.42 14.48 68.72 38.28 50.7 8 1 40.68 45.74 60.75 83.84 20.41 50.80 27.07 47 · 〇§ 〇37.37 57.30 45.05 54.92 21.57 37.68 24.92 39.8 5 2 22.45 37.37 25.63 39.30 21.75 18.47 15.40 25.8 35 10.5 δ. 10.67 11.68 21.1 〇11 · 47 22.33 21.04-10.74 6.33 8.58 13.4 2/7 Table 8 55.2 49.1 54.5 65.9 56.6 42.1 45.2 34.4 35.6 52.6 51.

表九、口服 fluvastatin 及 HUCHE070 Tamarixetin 實驗矣且 度數據 mt血中)農 28 200533364Table 9.Experimental data of oral fluvastatin and HUCHE070 Tamarixetin (mt blood) Agricultural 28 200533364

Concentration (ng/ml)Concentration (ng / ml)

Time (min) S-l S-2 S-3 S-4 S-5 mean SE C V % 10 59.54 18.79 29.52 100.09 25.45 46.68 15.07 72.2 〇 4 20 137.55 16.78 35.58 127.52 32.38 69.96 25.79 82.4 40 186.33 38.82 45.28 153.00 49.34 94.55 31.16 73.7 60 190.51 45.28 - 150.29 74.50 115.15 33.48 58.1 120 155.29 107.75 83.64 150.05 102.20 119.78 14.03 26.2 240 110.95 110.19 160.57 185.52 97.33 132.91 17.02。 28.6 360 88.95 9735 146.50 157.95 106.03 119.36 13.81 25.9 480 71.17 86.62 116.44 153.96 136.59 112.95 15.32 30.3 720 55.50 60.50 97.19 103.45 38.63 71.05 12.53 39.4 1080 28.45 - - 50.99 - 39.72 11.27 40.1 1440 27.61 - - 41.51 18.00 29.04 6.82 40.7 表九 表十、SD rat控制組及實驗組口服fluvastatin藥物動力學參數Time (min) Sl S-2 S-3 S-4 S-5 mean SE CV% 10 59.54 18.79 29.52 100.09 25.45 46.68 15.07 72.2 〇4 20 137.55 16.78 35.58 127.52 32.38 69.96 25.79 82.4 40 186.33 38.82 45.28 153.00 49.34 94.55 31.16 73.7 60 190.51 45.28-150.29 74.50 115.15 33.48 58.1 120 155.29 107.75 83.64 150.05 102.20 119.78 14.03 26.2 240 110.95 110.19 160.57 185.52 97.33 132.91 17.02. 28.6 360 88.95 9735 146.50 157.95 106.03 119.36 13.81 25.9 480 71.17 86.62 116.44 153.96 136.59 112.95 15.32 30.3 720 55.50 60.50 97.19 103.45 38.63 71.05 12.53 39.4 1080 28.45--50.99-39.72 11.27 40.1 1440 2761 10.1 9.40 1961. Fluvastatin pharmacokinetic parameters in SD, SD rat control group and experimental group

Fluvastatin (A) PK parameter (unit) Fluvastatin(5) control, n=7 with Tamarixetin? n=5 /βFluvastatin (A) PK parameter (unit) Fluvastatin (5) control, n = 7 with Tamarixetin? N = 5 / β

Cmax (ng/mL) 63.14 ± 10.36 141.40 ±15.76 2.4 Tmax (hr) 4.7 士 1.7 。 4.2 ± 1.1 0.9 AUCt (hr*ng/mL) 710.57 ± 8L55 1389.20^166.14* 2.0 AUCinf (hr*ng/mL) 949.86 ± 133.48 2281.00 ±386.56* 2.4 K (1/hr) 0.074 土 0.005 0.065 ± 0.009 0.9 Ti/2 (hr) 9.7 ±0.65 11.3i 1.33 1.2 Cl/F (mL/min/kg) 29.12 ±4.05 12.33 土 2.10** 0.4 Vz/F (mL/kg) 24846.64 ±4721.23 11163.54 ± 861.69* 0A AUMCinf (hr*hr*ng/mL) 15156.0 士 2864.6 42896.4 ± 12379.8 2.8 MRTinf (hr) 15.82 i 1.56 17.31 ±221 1.1 PK = pharmacokinetic,Data = mean 土 SE,*: p<0.05, **: P<0.01 表十 29 200533364 較佳實施例2之詳細說明 利用上述的實驗方法測試不同濃度的HUCHE009异苷草元 (isoliquritigenin)對於肝微粒體酶代謝tolbutamide之影響,其反應條件為: tolbutamide濃度為1 mM,Microsomes蛋白質含量為〇·5 mg,反應時間為 7··5分鐘。測試結果如表十一與圖七所示,編號HUCHE009异皆草元 〇〇叫111*丨%6!1丨11)抑制劑在高濃度下具有95.46 %代謝抑制程度,可視為_有 效 CYP450 isoenzyme 調控劑。 表十一、體外篩選中藥藥引HUCHE009异苷草元(is〇liqUritigenin)不同濃度 對於肝微粒體酶代謝tolbutamide之影響(n=3) 濃度 4-ΟΗ (ng) % inhibition Control 1 μΜ 10 μΜ 100 μΜ 374.8785 ±54.8521 0.0000 371.5965±18.7272 0.8737 263.4592±55.2455 29.6603 16.25213士0.5544 95.4680 表十一 較佳實施例3之詳細說明 利用上述的實驗方法測試不同濃度的HUCHE037金雀異黃酮 Genistein對於肝微粒體酶代謝之影響,其反應條件為· tolbutamide濃度為1 mM,Microsomes蛋白質含量為〇·5 mg,反應時間為 7·5分鐘。測試結果如表十二與圖八所示,編號金雀異黃酮 Gemstein抑制劑在高、中、低三種不同濃度下分別具有82.7 %,67.7 %,6 °/〇代謝抑制程度,可視為一有效CYP450 isoenzyme調控劑。 表十二、體外篩選中藥藥引HUCHE〇3 7金雀異黃ag Genistein不同濃度 對於肝微粒體酶代謝tolbutamide之影響(n=3) 30 200533364Cmax (ng / mL) 63.14 ± 10.36 141.40 ± 15.76 2.4 Tmax (hr) 4.7 ± 1.7. 4.2 ± 1.1 0.9 AUCt (hr * ng / mL) 710.57 ± 8L55 1389.20 ^ 166.14 * 2.0 AUCinf (hr * ng / mL) 949.86 ± 133.48 2281.00 ± 386.56 * 2.4 K (1 / hr) 0.074 soil 0.005 0.065 ± 0.009 0.9 Ti / 2 (hr) 9.7 ± 0.65 11.3i 1.33 1.2 Cl / F (mL / min / kg) 29.12 ± 4.05 12.33 Soil 2.10 ** 0.4 Vz / F (mL / kg) 24846.64 ± 4721.23 11163.54 ± 861.69 * 0A AUMCinf (hr * hr * ng / mL) 15156.0 ± 2864.6 42896.4 ± 12379.8 2.8 MRTinf (hr) 15.82 i 1.56 17.31 ± 221 1.1 PK = pharmacokinetic, Data = mean soil SE, *: p < 0.05, **: P < 0.01 Table 10 29 200533364 Detailed description of the preferred embodiment 2 The above experimental method is used to test the effects of different concentrations of HUCHE009 isoliquritigenin on liver microsomal enzyme metabolism tolbutamide. The reaction conditions are: tolbutamide concentration is 1 mM, Microsomes protein content It was 0.5 mg, and the reaction time was 7.5 minutes. The test results are shown in Table 11 and Figure 7. The number of HUCHE009 selenium grass 〇00 is 111 * 丨% 6! 1 丨 11) The inhibitor has a degree of metabolic inhibition of 95.46% at high concentrations, which can be regarded as _ effective CYP450 isoenzyme Regulators. Table 11: Effect of different concentrations of HUCHE009 isoliqUritigenin on the metabolism of tolbutamide in liver microsomal enzymes (n = 3) Concentration 4-〇Η (ng)% inhibition Control 1 μΜ 10 μΜ 100 μM 374.8785 ± 54.8521 0.0000 371.5965 ± 18.7272 0.8737 263.4592 ± 55.2455 29.6603 16.25213 ± 0.5544 95.4680 Table 11 Detailed description of Example 3 Use the above experimental method to test different concentrations of HUCHE037 Gin Isoflavone Genistein for liver microsomal enzyme metabolism The reaction conditions are: the tolbutamide concentration is 1 mM, the Microsomes protein content is 0.5 mg, and the reaction time is 7.5 minutes. The test results are shown in Table 12 and Figure 8. The numbered genistein isoflavone Gemstein inhibitors have 82.7%, 67.7%, and 6 ° / 〇 metabolic inhibition at three different concentrations: high, medium, and low, which can be regarded as effective. CYP450 isoenzyme modulator. Table 12. In vitro screening of traditional Chinese medicine cited HUCHE〇3 7 different effects of genistein ag Genistein at different concentrations on liver microsomal enzyme metabolism tolbutamide (n = 3) 30 200533364

Category 4-ΟΗ (ng) % inhibition Control 479.3314±56.4829 -- 0.0000 1 μΜ 241.2098土6.5885 49.5979 10 μΜ 154.311 士 10.9480 67.6979 100 μΜ 82.24342士 13.3679 82.7088 表十二 雖然本發明已藉由上述細說明以及較佳實施例來予以闊釋,本發明 =應被解釋為受之所_ ;減地’本發明實係涵蓋,#熟知此項技藝者 從本案發明說明書所揭示的技_容與·實絲考量時,可以做出的多種其 他不同等效變化及娜飾。鼠,林偏縣發明讀義下,大凡依本發 明申請專纖圍及發縣明書内容所作之鮮的較變化與修飾,皆應仍 屬本發明專利涵蓋之範圍内。 【圖式簡單說明】 圖一係為體外篩選Keto_zole影響肝微粒體酶代謝tdbutamide形成 44iydroxyt〇lbutamide 之比較圖。 圖-係為lGGpM中藥藥引纯成分抑制酵素代謝活性前十名之比較圖。 圖三係為ΙΟμΜ中藥藥引纯成分抑制酵素代謝活性前十名之比較圖。 圖四係為1 —中藥藥引純成分抑制酵素代謝活性前十名之比較圖。 圖五係為體外篩選中藥藥引抑制劑Tamarixetin影響肝微粒體酶代謝 tolbutamide 形成 4-hydroxytolbutamide 之比較圖。 圖六係為SD rat控制組(n=7)及實驗組(n二5) 口服fluvastatin血中濃 度經時變化圖。 、圖七係為體外篩選中藥藥引异苷草元(isoliquriti马enin)影響肝微粒體酶 代^射 tolbutamide 形成 4-hydroxytolbutamide 之比較圖。 31 200533364 圖八係為體外篩選中藥藥引抑制劑金雀異黃酮Genistein影響肝微粒體 酶代謝 tolbutamide 形成 4-hydroxytolbiitamide 之比較圖。 32Category 4-ΟΗ (ng)% inhibition Control 479.3314 ± 56.4829-0.0000 1 μΜ 241.2098 ± 6.5885 49.5979 10 μΜ 154.311 ± 10.9480 67.6979 100 μΜ 82.24342 ± 13.679 82.7088 Table 12 Although the present invention has been described by the above detailed description and better implementation Explained by way of example, the present invention = should be interpreted as being affected by; minus the ground 'the invention is covered by the present invention, #know when the skilled person from the technical disclosure of the invention specification disclosed in this case _ capacity and real silk consideration, There are many other different equivalent changes and decorations that can be made. Under the meaning of the invention of the rat and Lin Pianxian, all the relatively small changes and modifications made in the application for the special fiber circumference and the content of the county book according to the present invention should still fall within the scope of the invention patent. [Brief description of the figure] Figure 1 is a comparison chart of in vitro screening of Keto_zole affecting liver microsomal enzyme metabolism tdbutamide formation 44iydroxyt〇lbutamide. Figure-is a comparison chart of the top ten inhibitory enzyme metabolism activities of pure ingredients of lGGpM Chinese medicine. Figure 3 is a comparison chart of the top ten inhibitory enzyme metabolic activities of pure ingredients of 10 μM traditional Chinese medicine. Figure 4 is a comparison chart of the top ten of 1—Chinese herbal medicine pure ingredients inhibiting enzyme metabolism. Figure 5 is a comparison chart of the in vitro screening of the traditional Chinese medicine drug inhibitor Tamarixetin on liver microsomal enzyme metabolism tolbutamide to form 4-hydroxytolbutamide. Figure 6 shows the changes of blood concentration of fluvastatin in SD rat control group (n = 7) and experimental group (n = 2) over time. Figure 7 is a comparison chart of the in vitro screening of the traditional Chinese medicine isoliquriti horse enin on the effect of liver microsomal enzymes on tolbutamide to form 4-hydroxytolbutamide. 31 200533364 Figure 8 shows the comparison of in vitro screening of the traditional Chinese medicine drug inhibitor genistein (Genistein) on liver microsomes, enzyme metabolism, tolbutamide formation and 4-hydroxytolbiitamide. 32

Claims (1)

200533364 拾、申請專利範圍: 1. 一種細胞色素(cytochrome)P450同工酶CYP2C9,之抑制劑,其中抑制劑 係選自於下列化合物所組成群組:聖柳素(Tamarixetin)、花黃素異黃_ (Formononetin)、异苷草元(isoliquritigenin)、 Phloretin、木犀草素 (luteolin)、懈皮苦(Quercitrin)、槲皮素(quercetin)、楊梅樹皮素(myricetin)、 漢黃答素(Wongonin)、葛根素(Puerarin )、金雀異黃酮(Genistein)、正二 經癒瘡酸(Nordihydroguaiaretic acid)、柚皮素(Narigenin)、茵陳色原酮 (Capillarisin)、Chrysin、Fisefin、聖草素(eriodictyol)、6-薑辣素 6-Gingerol )、異鼠李素(Isorhamnetin)、異槲皮苷(isoquercitrin)、Morin、 (+)-Taxifolin、異牡荊素(isovitexin)、乙酸苯丙醋(3-Phenylpropyl Acetate)、 齊墩果酸(Oleanolic acid)、熊果酸(ursolic acid)、石-月桂豨(β-Myrcene )、 桂皮酸(cinnamic acid)、木厚草素-7-0•葡萄糖苦(Luteolin-7-Glucoside>、 甘草萃取(Liquiritin )、(+)檸檬精油((+)Limonene )、·異狂草素 (Homoorientin)、獐牙菜苦(Swertiamarin)、Embelin、木質素異黃酮 Daidzein、枳屬(Pondrin)、㈠表兒茶素(-)-Epicatechin、麥角固醇 (ergosterol) 〇 2·如申請專利範圍第1項之細胞色素(Cyt〇chrome)P450同工酶CYP2C9之 抑制劑’其中抑制劑係選自於下列化合物所組成群組:聖柳素 (Tamarixetin)、花黃素異黃酮(Formononetin)、异苷草元(isoliquritigenin)、 Phloretin、木犀草素(luteolin)、懈皮苷(Quercitrin)、槲皮素(quercetin)、楊 梅樹皮素(myricetin)、漢黃芩素(Wongonin)、葛根素(Puemrin )、金雀異 黃嗣(Genistein)、正二羥癒瘡酸(Nordihydroguaiaretic acid)、柚皮素 (Narigenin)。 3.如申請專利範圍第1項之細胞色素(cytocbrome)P450同工酶CYP2C9之 抑制劑’其中抑制劑係為聖柳素(Tamarixetin)。 4· 一種用以增加藥物之生體可用率的藥學組成物,其包含: (a)一藥學有效量之如申請專利範圍第1項的CYP2C9抑制劑;以及 33 200533364 (b)—藥學上可接受之載劑。 5. 如申請專利範圍第4項的藥學組成物,其中該藥學組成物係與另一種藥 學有效量的降血糖類藥物tolbutamide組合投藥: 6. 如申請專利範圍第4項的藥學組成物,°其中該藥學铒成物係與另一種藥 學有效量的降血脂類藥物fluvastatin組合投藥。200533364 The scope of patent application: 1. An inhibitor of cytochrome P450 isoenzyme CYP2C9, wherein the inhibitor is selected from the group consisting of the following compounds: Tamarixetin, xanthophyllin Yellow (Formononetin), isoliquritigenin, Phloretin, luteolin, Quercitrin, quercetin, myricetin, Wongonin, Puerarin, Genistein, Nordihydroguaiaretic acid, Narigenin, Capillarisin, Chrysin, Fisefin, eriodictyol , 6-Gingerol 6-Gingerol), Isorhamnetin, isoquercitrin, Morin, (+)-Taxifolin, isovitexin, phenylpropionate acetate (3- Phenylpropyl Acetate), oleanolic acid, ursolic acid, β-Myrcene, cinnamic acid, lignan-7-0 • glucose bitter ( Luteolin-7-Glucoside >, Liquiritin, (+ ) Lemon Essential Oil ((+) Limonene), Homoorientin, Swertiamarin, Embelin, Lignin Isoflavone Daidzein, Pondrin, Epiatechin (-)-Epicatechin Ergosterol 〇2. If the cytochrome (Cytochrome) P450 isoenzyme CYP2C9 inhibitor of item 1 of the scope of patent application 'inhibitor is selected from the group consisting of the following compounds: Tamarixetin, Formononetin, isoliquritigenin, Phloretin, luteolin, Quercitrin, quercetin, myricin myricetin), Wongonin, Puemrin, Genistein, Nordihydroguaiaretic acid, Narigenin. 3. The inhibitor of cytocbrome P450 isoenzyme CYP2C9 'according to item 1 of the scope of patent application, wherein the inhibitor is Tamarixetin. 4. A pharmaceutical composition for increasing the bioavailability of a drug, comprising: (a) a pharmaceutically effective amount of a CYP2C9 inhibitor such as the one in the scope of patent application; and 33 200533364 (b) —pharmaceutically acceptable Accepted vehicle. 5. If the pharmaceutical composition according to item 4 of the patent application is applied, wherein the pharmaceutical composition is administered in combination with another pharmaceutically effective amount of a hypoglycemic drug tolbutamide: 6. If the pharmaceutical composition according to item 4 of the patent application, ° The pharmaceutical composition is administered in combination with another pharmacologically effective amount of a lipid-lowering drug, fluvastatin. 3434
TW93109325A 2004-04-05 2004-04-05 Inhibitor of cytochrome P450 isozyme, CYP2C9 TW200533364A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
TW93109325A TW200533364A (en) 2004-04-05 2004-04-05 Inhibitor of cytochrome P450 isozyme, CYP2C9

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
TW93109325A TW200533364A (en) 2004-04-05 2004-04-05 Inhibitor of cytochrome P450 isozyme, CYP2C9

Publications (2)

Publication Number Publication Date
TW200533364A true TW200533364A (en) 2005-10-16
TWI299002B TWI299002B (en) 2008-07-21

Family

ID=45069563

Family Applications (1)

Application Number Title Priority Date Filing Date
TW93109325A TW200533364A (en) 2004-04-05 2004-04-05 Inhibitor of cytochrome P450 isozyme, CYP2C9

Country Status (1)

Country Link
TW (1) TW200533364A (en)

Also Published As

Publication number Publication date
TWI299002B (en) 2008-07-21

Similar Documents

Publication Publication Date Title
Choi et al. Yeast α-glucosidase inhibition by isoflavones from plants of Leguminosae as an in vitro alternative to acarbose
US5204369A (en) Method for the inhibition of aldh-i useful in the treatment of alcohol dependence or alcohol abuse
JP5535999B2 (en) Use of equol to treat androgen-mediated diseases
US20140343116A1 (en) Cytochrome p450 2c9 inhibitors
US20160184298A1 (en) Inhibitors and enhancers of uridine diphosphate-glucuronosyltransferase 2b (ugt2b)
JP5133064B2 (en) Inhibitors and promoters of uridine diphosphate (UDP) -glucuronyltransferase 2B (UGT2B)
Han et al. Effect of silymarin on the pharmacokinetics of losartan and its active metabolite E-3174 in healthy Chinese volunteers
JP2005535665A (en) Methods for preventing ischemia and reperfusion injury
CN109438536A (en) The application and preparation method of a kind of isoquercitin and its derivative
Keung Anti‐dipsotropic isoflavones: The potential therapeutic agents for alcohol dependence
Liu et al. Applications of Pueraria lobata in treating diabetics and reducing alcohol drinking
US6255497B1 (en) Method for the inhibition of ALDH-I useful in the treatment of alcohol dependence or alcohol abuse
Fan et al. Drug interaction study of flavonoids toward OATP1B1 and their 3D structure activity relationship analysis for predicting hepatoprotective effects
CN103391781A (en) New low side effect pharmaceutical composition containing antituberculosis drugs
CN102137671A (en) New low side effect pharmaceutical composition containing isoniazid
CN105859667A (en) Aryl benzofuran amidated derivatives and pharmaceutical use thereof
US7338941B2 (en) Pharmaceutical compositions for lowering blood glucose and blood cholesterol levels
Setchell et al. Pharmacokinetics of a slow-release formulation of soybean isoflavones in healthy postmenopausal women
Ueng et al. Alteration of the pharmacokinetics of theophylline by rutaecarpine, an alkaloid of the medicinal herb Evodia rutaecarpa, in rats
US20060040875A1 (en) Inhibitors and enhancers of uridine diphosphate-glucuronosyltransferase 2B (UGT2B)
TW200533364A (en) Inhibitor of cytochrome P450 isozyme, CYP2C9
Yan et al. Determination of puerarin in rat cortex by high‐performance liquid chromatography after intravenous administration of Puerariae flavonoids
CN107586284A (en) A kind of purposes of 2 arylbenzofuran analog derivative in gout medicine is prepared
CN105012294B (en) New application of the ellagic acid compounds in treatment antihyperuricemic disease drug is prepared
CN111249271A (en) Medicament for treating diabetes and preparation method and application thereof