前体药物—姜黄素二癸酸酯药代动力学研究及其纳米混悬剂工艺探索与评价
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
姜黄素是姜黄的主要成分之一,姜黄素作为色素、食品添加剂以及调味品已被广泛使用,各种实验模型证明姜黄素具有抗肿瘤、抗炎、抗氧化、保肝肾等药理活性;北大医学部库宝善等在郁金为主要成分的解郁丸中,发现并率先报道姜黄素具有明显的抗抑郁活性,并开展了深入的研究,认为选用姜黄素作为研究治疗抗肿瘤并伴有抑郁的候选药物具有极大的研究潜力和应用价值。但是由于姜黄素稳定性差,难溶于水,生物利用度低,体内代谢快,吸收过程发生转化,半衰期短等问题而限制了使用。如何解决其稳定性,提高其生物利用度,改善体内代谢及动力学问题成为研究的重点与难点。因此于凤华等人合成了姜黄素的酯类前体药物-姜黄素二癸酸酯(CurDD), CurDD吸收后酶解为姜黄素而发挥活性。本文主要采用高效液相色谱以及液质联用色谱技术,研究以整体实验动物给予姜黄素及其前体药物后的药代动力学特性,进行相关的药代动力学评价,并完成制剂成型与原料药的质量评价。
     首先,本文首次建立了高效、灵敏、稳定可靠的同时监测生物样品中姜黄素二癸酸酯和姜黄素的HPLC分析方法。本方法选用大黄素作为内标物,采用液液萃取同时加有高浓度表面活性剂的方法作为前处理步骤。结果显示,姜黄素二癸酸酯和姜黄素在各自的线性范围内呈现良好的线性,精密度、准确度、提取回收率均符合生物样品分析要求。同时,我们建立了HPLC-MS/MS分析方法测定姜黄素在大鼠血浆中的含量,可以检测血浆样品浓度在0.5ng/ml~20ng/ml范围内的低含量样品,且线性、精密度、准确度、提取回收率均符合生物样品分析要求。
     第二,我们研究了姜黄素二癸酸酯的体外生物转化。研究结果显示,姜黄素二癸酸酯可以转化为姜黄素,但是姜黄素的含量始终维持在100ng/ml左右,可以表明姜黄素在生成的同时又发生了自身的降解,而姜黄素二癸酸酯则作为姜黄素的一个储存仓库,缓慢释放,使其维持在一个稳定的浓度范围。
     第三,本文首次应用已经建立的HPLC方法,研究了姜黄素二癸酸酯和姜黄素在大鼠体内静脉注射后的药代动力学特性。静脉注射药代动力学评价显示姜黄素二癸酸酯可以使姜黄素的半衰期由6.6min延长至6h左右,而AUC值也提高近10倍,说明姜黄素二癸酸酯可以作为姜黄素的一个储存仓库具有缓慢释放姜黄素的作用。本文同时应用已建立的HPLC-MS/MS法,研究了姜黄素二癸酸酯和姜黄素在大鼠体内肌肉注射后的药代动力学特性。肌肉注射药代动力学评价显示姜黄素二癸酸酯可以使姜黄素的半衰期延长。
     第四,本文制备了平均粒径在1μm以下的姜黄素二癸酸酯纳米粒,单因素考察了采用单平行星式球磨仪各参数对纳米粒子粒径的影响,优化了工艺参数,并对纳米粒子进行相关的物理性质评价和稳定性评价。
     第五,本文研究了姜黄素二癸酸酯纳米/普通混悬注射油剂在大鼠肌肉注射部位的释放与消除。结果显示,姜黄素二癸酸酯纳米/普通混悬注射油剂,15d后释放均超过80%以上,而纳米混悬剂比普通混悬剂更能延长姜黄素二癸酸酯在注射部位的驻留时间。
     最后,本文又考察了姜黄素二癸酸酯纳米/普通混悬剂肌肉注射后在脑部的含量,结果发现,姜黄素二癸酸酯纳米混悬剂在脑部姜黄素二癸酸酯的含量显著高于普通混悬剂的含量,AUC值也提高4倍以上。
     本论文研究结果发现姜黄素二癸酸酯的药代动力学性质具有缓慢释放姜黄素的作用,能够显著延长姜黄素的半衰期和姜黄素在体内的作用时间,达到长效的作用,并为制剂的研发提供了一定的参考依据;而姜黄素二癸酸酯纳米混悬剂制备与评价结果显示,纳米混悬剂在缓释、长效方面比普通混悬剂具有显著的优势,并解决了姜黄素二癸酸酯在水和有机溶剂都难溶的难题,因此适合于姜黄素二癸酸酯长效抗抑郁制剂的开发和利用。
Curcumin is the major active ingredient of the rhizome of Curcuma longa L, which has been extensively used as a dietary spice and a coloring agent, as well as a medicinal herb in the Chinese and Ayurvedic medical systems for the treatment of flatulence, jaundice, menstrual difficulties, hematuria, hemorrhage and colic. The studies on curcumin and its analogues have drawn increasing attentions in recent years, due to the promising clinical applications and low toxicity and have documented that curcumin possesses various biological and pharmacological activities, including anti-oxidation, anti-inflammation, tumor prevention and anti-depression, etc.. Nonetheless, the pharmaceutical formulation development of curcumin remains to be a challenge due to its poor physicochemical and biopharmaceutical properties. As a polyphenolic compound, curcumin is water insoluble and unstable at aqueous medium, and exhibits very poor oral bioavailability and rapid systemic elimination. In order to improve the bioavailability and/or modify the pharmacokinetic parameters, numerous drug delivery techniques by loading curcumin into liposomes, nanoparticles, solid dispersion or self-microemulsifying system, forming curcumin-phospholipids or cyclodextrins complex, and synthesizing structural analogues of curcumin have been investigated.
     Recently, a prodrug of curcumin, curcumin didecanoate (CurDD), was prepared by esterification of the hydroxyl groups of curcumin with capric acid so as to provide sustained plasma levels of curcumin and to achieve long-acting therapeutic effects following administration as a depot.
     Firstly, a high-performance liquid chromatographic (HPLC) method has been developed for the simultaneous determination of curcumin and its prodrug, curcumin didecanoate (CurDD), in rat plasma. The analytes were extracted by ethyl acetate following the addition of sodium dodecyl sulfate, and separated on a reverse phase C18 column using a gradient mobile phase system of acetonitrile-tetrahydrofuran-water containing 0.1% formic acid. Detection by UV absorption at 425 nm gave a lower limit of quantitation (LLOQ) of 5 ng/ml and 10 ng/ml for curcumin and CurDD in 50μl of plasma, respectively. Intra-and inter-day precision of quality control samples except those at LLOQ were within 15% for curcumin and CurDD, respectively, and the accuracy for both compounds were between-6.07 and 8.88%. Meanwhile, an HPLC-MS/MS method has been established for the determination of curcumin in rat plasma. The LLOQ of curcumin were determined to be 0.5 ng/ml. The linearity, precision, accuracy, extraction recoveries were fit for purpose of biological sample analysis.
     Secondly, the results of biotransformation of CurDD in vitro showed that CurDD can be transformed into curcumin, and the concentrations of curcumin were maintained at about 100ng/ml. Meanwhile, curcumin may be not only released by CurDD, but also degraded by itself. The results suggested that CurDD may be used as a depot of curcumin to maintain a stable concentration.
     Thirdly, a simple HPLC method was applied to determine the plasma concentrations of CurDD and/or curcumin after CurDD or curcumin was intravenously administered to rats at a dose of 1 mg/kg (calculated as curcumin). Following IV administration of curcumin, the plasma concentrations of curcumin rapidly decreased to the LLOQ within 20 min with a t1/2 of-7 min. After IV administration of CurDD, the concentrations of both CurDD and curcumin declined slowly. The plasma concentration of curcumin converted from CurDD was detectable up to 24 h with a t1/2 of~400 min. When the AUC data of curcumin obtained by direct IV administration were compared with those converted from CurDD, the latter were surprisingly 6-fold higher than the former. An HPLC-MS/MS method has also been developed and validated for the determination of curcumin after CurDD or curcumin was intramuscularly administered to rats at a dose of 50 mg/kg (calculated as curcumin). The study also found that when entered into the blood, CurDD was converted to curcumin and markedly extended the apparent elimination half-life. These results suggested that CurDD might be utilized as a prodrug to provide sustained plasma levels of curcumin over a long duration when administered as a depot.
     Fourthly, nano-suspensions of CurDD was prepared by planetary ball milling with the median particle size being less than 1μm. The preparation processes were optimized for various parameters of planetary ball milling affecting the particle size. Moreover, the physical properties and stability of the nano-suspensions have also been evaluated.
     Fifthly, it has been assessed that the percentage of the dose of CurDD remaining at the injection site after intramuscular administration of CurDD nano-and normal-suspensions in peanut oil. After 15d, only less than 20% of CurDD was remained at the injection site, whether CurDD nano-or normal-suspensions. Compared with CurDD normal-suspensions, CurDD nano-suspensions have extremely extended the dwell time of CurDD at the injection site.
     Finally, the concentration of curcumin didecanoate (CurDD) in brain tissues was determined as a function of time following intramuscular of CurDD nano-and normal-suspensions to rats at doses of 50 mg/kg(calculated as curcumin). When the brain concentration and AUC data obtained by CurDD nano-suspensions were compared with normal-suspensions, the former were significantly higher than the latter.
引文
[1]Tang W and Eisenbrand G. Chinese drugs of plant origin[M]. Springer-Verlag:Berlin and Heidelberg, German,1992:401-415.
    [2]Eigner D and Scholz D. Ferula asa-foetida and curcuma longa in traditional medical treatment and diet in Nepal[J]. Journal of Ethnopharmacology,1999,67:1-6.
    [3]Abe Y, Hashimoto S and Horie T. Curcumin inhibition of inflammatory cytokine production by human peripheral blood monocytes and alveolar macrophages[J]. Pharmacological Research,1999,39:41-47.
    [4]Ak T and Grlgin I. Antioxidant and radical scavenging properties of curcumin[J]. Chemico-Biological Interactions,2008,174:27-37.
    [5]Xu Y, Ku BS, Yao HY, Lin YH, Ma X, Zhang YH and Li XJ. Antidepressant effects of curcumin in the forced swim test and olfactory bulbectomy models of depression in rats[J]. Pharmacology Biochemistry and Behavior,2005,82:200-206.
    [6]Xu Y, Ku BS, Yao HY, Lin YH, Ma X, Zhang YH and Li XJ. The effects of curcumin on depressive-like behaviors in mice[J]. European Journal of Pharmacology,2005,518: 40-46.
    [7]Bhutani MK, Bishnoi M and Kulkarni SK. Anti-depressant like effect of curcumin and its combination with piperine in unpredictable chronic stress-induced behavioral, biochemical and neurochemical changes[J]. Pharmacology Biochemistry and Behavior, 2009,92:39-43.
    [8]Chen DY, Shien JH, Tiley L, Chiou SS, Wang SY, Chang TJ, Lee YJ, Chan KW and Hsu WL. Curcumin inhibits influenza virus infection and haemagglutination activity[J]. Food Chemistry,2010,119:1346-1351.
    [9]Anand P, Kunnumakkara AB, Newman RA and Aggarwal BB. Bioavailability of curcumin:problems and promises[J]. Molecular Pharmaceutics,2007,4:807-818.
    [10]蒋永和,袁继承,沈志滨.姜黄属植物化学成分的研究进展[J].亚太传统医药,2009,2(2):124.
    [11]吴萍,许树长.姜黄素的研究进展及其在消化系统疾病中的应用前景[J].中国临床 营养杂志,2008,8(16):106-109.
    [12]安建斌,马景学.姜黄素的药理作用及其在眼科的应用研究进展[J].中国中医眼科杂志,2008,12(6):360
    [13]牛生洋,郝峰鸽,许秋亚.姜黄素的提取及应用研究进展[J].河南科技学院学报,2008,12(4):58.
    [14]刘钰,栾立标.姜黄素固体分散体的制备及体外溶出度测定[J].药学进展,2006,30(1):40-41.
    [15]黄秀旺,许建华,吴国华等.姜黄素固体分散体在小鼠体内的药代动力学[J].中国药理学通报,2008,24(11):1525.
    [16]汪洋,徐庆弟.包合技术在中药药剂学中的应用[J].中国药房,2008,19(3):226.
    [17]许汉林,孙芸,邵继征等.不同方法制备姜黄素脂质体的研究[J].中国中医药信息杂志,2006,13(3):51-52.
    [18]陆鹏,童强松.姜黄素前体药物的合成及其体外抗肿瘤活性研究[J].中国药理学通报,2006,22(3):321.
    [19]Anand P, Kunnumakkara AB, Newman RA, et al. Bioavailability of curcumin:problems and promises[J]. Mol Pharm,2007,4 (6):807-818.
    [20]Wahlstrom B and Blennow G. A study on the fate of curcumin in the rat[J]. Acta Pharmacol. Toxicol. (Copenhagen) 1978,43(2):86-92.
    [21]Ravindranath V and Chandrasekhara N. Absorption and tissue distribution of curcumin in rats[J]. Toxicology,1980,16(3):259-265.
    [22]Ravindranath V and Chandrasekhara N. Metabolism of curcumin studies with [3H]curcumin[J]. Toxicology,1981,22(4):337-344.
    [23]Shoba G, Joy D, Joseph T, Majeed M, Rajendran R, Srinivas P.S. Influence of piperine on the pharmacokinetics of curcumin in animals and human volunteers[J]. Planta Med,1998,64(4):353-356.
    [24]Pan MH, Huang TM, Lin JK. Biotransformation of curcumin through reduction and glucuronidation in mice[J]. Drug Metab Dispos,1999,27(4):486-494.
    [25]温彩霞,许建华,黄秀旺.姜黄素在小鼠体内各脏器的药代动力学研究[J].中药药理与临床,2007,23(4):25-26.
    [26]Hoehle SI, Pfeiffer E, Solyom AM, Metzler M. Metabolism of curcuminoids in tissue slices and subcellular fractions from rat liver[J]. J. Agric. Food Chem,2006,54(3):756-764.
    [27]Holder GM, Plummer JL, Ryan AJ. The metabolism and excretion of curcumin(1,7-bis-(4-hydroxy-3-methoxyphenyl)-1,6-heptadiene-3,5-dione) in the rat[J]. Xenobiotica,1978,8(12):761-768.
    [28]Ireson C, Orr S, Jones DJ, Verschoyle R, Lim CK, Luo JL, Howells L, Plummer S, Jukes R, Williams M, Steward WP and Gescher A. Characterization of metabolites of the chemopreventive agent curcumin in human and rat hepatocytes and in the rat in vivo, and evaluation of their ability to inhibit phorbol ester-induced prostaglandin E2 production[J]. Cancer Research,2001,61:1058-1064.
    [29]Zhang M, Deng CS, Zheng JJ, et al.Curcumin regulated shift fromTh1 to Th2 in trinitrobenzene sulphonic acid-induced chronic colitis[J]. Acta Pharmacol Sin,2006,27(8):1071-1077.
    [30]黄珈雯.姜黄素的抗肿瘤作用研究进展[J].甘肃中医,2008,21(1):55-56.
    [31]陈宏,陈宇英,张振书等.姜黄素抗大肠癌作用研究[J].肿瘤研究与临床,2006,18(1):4-7.
    [32]崔国惠,陈卫华,陈燕.姜黄素和肿瘤坏死因子α调节淋巴瘤细胞VEGF的表达及抗血管形成的研究[J].中国医院药学杂志,2008,28(23):1918-1919.
    [33]王勇,高大中.姜黄素防治心力衰竭的研究现状[J].中国药房,2007,18(33):2624-2626.
    [34]汪海慧,成扬.姜黄素药理作用的研究进展[J].上海中医药大学学报,2007,21(6):73-76.
    [35]向进见,田夫,沈世强等.姜黄素对大鼠肝缺血再灌注早期肝组织中氧自由基生成的影响[J].腹部外科,2008,21(5):315-316.
    [36]胡泰洪,蒋祥虎,朱锐等.姜黄素对实验性肝纤维化大鼠肝脏bax和bcl-2表达的影响[J].中国中西医结合消化杂志,2008,16(6):362-364.
    [37]郑虎.药物化学[M].北京:人民卫生出版社,2003:419.
    [38]何亮.前药的研究进展[J].世界临床药物,2006,27(1):55.
    [39]Mayer A, Francis RJ, Sharma SK, et al. A Phase I Study of Single Administration of Antibody-Directed Enzyme Prodrug Therapy with the Recombinant Anti-Carcinoembryonic Antigen Antibody-Enzyme Fusion Protein MFECP1and a Bis-Iodo Phenol Mustard Prodrug[J]. Clin Cancer Res,2006,12(21):6510-6516.
    [40]刘剑峰,徐文方.新药研究中的前药原理[J].齐鲁药事,2005,24(2):100.
    [41]何亮,李勤耕,田睿.前药载体研究新进展[J].中国药房,2007,18(2):141-142.
    [42]郭宗儒.药物设计中前药原理[J].药学学报,1979,14(9):566-575.
    [43]何训贵,高河勇,吕伟等.喜树碱及其衍生物的前药研究进展[J].药学进展,2005,29(4):146-149.
    [44]Cao ZS, Harris N, Kozielski A, et al. Alkyl esters of camptothecin and 9-nitrocamptothecin:synthesis, in vitro pharmacokinetics, toxicity, and antitumor activity[J]. J Med Chem,1998,41(1):31~37.
    [45]Cao ZS, Giovanella BC. Liposomal prodrugs comprising derivatives of camptothecin and methods of treating cancer using these prodrugs[P]. WO:2001008663, 2001-02-08.
    [46]Cao ZS, Giovanella BC. Preparation of aromatic esters of camptothecin and methods to treat cancers [P]. WO:2001009139,2001-02-08.
    [47]Vishnvvajjala R, Garzon A, burbey A. Preparation of water-soluble derivatives of camptothecin and their use as prodrug in cancer therapy[P]. US:104894,1990-03-01.
    [48]陈涛,王进,佛江涛等.聚乙二醇前体药物[J].世界最新医学信息文摘,2003,2(5):808.
    [49]王立升,周永红,刘百里等.苯乙哌嗪类化合物的合成及其抗变态,抗炎和镇痛活性[J].中国药物化学杂志,2000,10(1):5-8.
    [50]王淑月,王洪亮.前药原理与新药设计[J].河北工业科技,2003,20(1):56.
    [51]Boucher BA, Feler CA, Dean JC, et al. The safety, tolerability, and pharmacokinetics of fosphenytoin after intramuscular and intravenous administration in neurosurgery patients[J]. Pharmacotherapy,1996,16(4):638-645.
    [52]Wu MK. A New Classification of Prodrugs:Regulatory Perspectives[J]. Pharmaceuticals,2009,2(3),77-81.
    [53]刘文胜,罗维早,张志荣.N1-羧酰-5-氟尿嘧啶系列前体药物的体外降解动力学研究[J].中国医药工业杂志,2002,33(9):434.
    [54]王聪,王一腾,魏鑫.环磷酰胺的临床应用现状[J].临床和实验医学杂志,2008,7(8):179.
    [55]李安良,李正香.前药和药物靶向作用[J].中国药学杂志,2003,28(4):243-245.
    [56]马春来,焦正,应寅清.反相高效液相色谱法同时测定人血浆中磷苯妥英,苯妥英及其主要代谢产物4-羟苯妥英的浓度[J].中国药学杂志,2008,43(22):1743.
    [57]叶海,张灿,沈文斌,盛龙生等.灯盏乙素聚乙二醇前药的合成与表征[J].中国天然药物,2006,14(14):283-285.
    [58]陆鹏,童强松等.姜黄素前体药物的合成及其体外抗肿瘤活性研究[J].中国药理学通报,2006,22(3):321-324.
    [59]潘献柱,王林,温晓雪等.文拉法辛前药的脑靶向效应初探[J].安徽医科大学学报,2006,41(4):420-423.
    [60]周瑾等.结肠定位的糖皮质激素前体药物[J].中国药学杂志,2001,36(12):803.
    [61]周四元等.糖皮质激素前体药在大鼠胃肠道中定位转释的药代动力学[J].药学学报,2001,36(5):325-328.
    [62]李炜,章承继,仇缀百.阿片类前体药物的研究进展[J].中国药物化学杂志,2004,14(1):57.
    [63]魏树礼,张强.生物药剂学与药代动力学.北京大学医学出版社[M],2004:85-89.
    [64]李颖.中药药代动力学研究方法概况.安徽中医学院学报[J],2005,24(3):62-64.
    [65]李好枝.体内药物分析.中国医药科技出版社[M],2008:122-138.
    [66]耿魁魁,段贤春,夏伦祝.液质联用技术在中药药代动力学中的应用[J].安徽医药,2010,14(5):505-507.
    [67]胡海燕,朱馨乐,胡昊等.超高效液相色谱简介及应用比较[J].中国兽药杂志,2010,44(4):48-50.
    [68]吴剑威,赵润怀,陈波等.超高效液相色谱在中药领域的应用[J].中草药,2009,40:79-81.
    [69]艾华,王广基,顾轶等.超高效液相色谱在现代药代动力学中的应用[J].中国药科大学学报,2007,38(4):294-298.
    [70]Dongre VG, Karmuse PP, Rao PP, Kumar A. Development and validation of UPLC method for determination of primaquine phosphate and its impurities [J]. Journal of Pharmaceutical and Biomedical Analysis,2008,46:236-242.
    [71]白永涛,文红梅,彭彬,熊海伟UPLC法测定大鼠血浆中染料木素浓度及其药代动力学参数[J].中药新药与临床药理,2010,21(5):512-514.
    [72]Al-Dirbashi OY, Aboul-Enein HY, Jacob M, et al. UPLC-MS/MS determination of doxazosine in human plasma[J]. Anal Bioanal Chem,2006,385(8):1439-1443.
    [73]靳凤丹LC/MS/MS技术在药代动力学研究中的应用[J].高师理科学刊,2009,29(3):57-59.
    [74]杜玥,陈笑艳,杨汉煜等.液相色谱-串联质谱法测定大鼠血浆中的汉黄芩素[J].药学学报,2002,37:362-366.
    [75]余勤,梁茂植,南峰,向瑾,秦永平HPLC-MS/MS测定大鼠血浆及组织中盐酸戊乙奎醚浓度[J].四川大学学报,2010,41(1):153-157.
    [76]刘茜,霍舒怡,赵辉,杜红文,钟丽丽等HPLC-MS/MS测定赖诺普利血药浓度及其在药动学研究中的应用[J].中国现代应用药学,2010,27(2):158-159.
    [77]李媛媛,郝光涛,李海燕,高洪志HPLC-MS/MS测定人体血浆和尿液中帕洛诺司琼的浓度[J].中南药学,2010,8(2):119-122.
    [78]张庆,侯连兵,陈凌云,许军等HPLC-MS/MS测定人血浆阿奇霉素浓度及生物等效性研究[J].中国现代应用药学,2010,27(2):148-151.
    [79]楼永军,王峰,李会林HPLC-MS测定人血浆中奥美拉唑浓度及药动学[J].中国药学杂志,2010,45(4):292-294.
    [80]潘海燕.中药药代动力学研究综述[J].实用医技杂志,2008,15(33):4894-4897.
    [81]唐斌,王磊.药代动力学在中药研究中的应用进展[J].西南国防医药,2010,20(12):1380-1382.
    [82]刘萍,黄颖.中药复方的药代动力学研究方法和存在的若干问题[J].湖北中医学院学报,2010,12(6):64-65.
    [83]周丽娟,许利平,陈慧娟,白雪.中药复方药代动力学常用研究方法评价[J].中华中医药学刊,2007,25(5):1041-1042.
    [84]李东,王敏,谢静,张雁.中药药代动力学的研究方法[J].中国药业,2006,15(11):25-27.
    [85]高姗,徐康康.中药药代动力学研究进展[J].中国实用医药,2010,5(21):249-251.
    [86]Lipinski CA. Drug-like properties and the causes of poor solubility and poor permeability[J]. J Pharmacol Toxicol Methods,2000,44(1):235-249.
    [87]钱帅,张建军,高缘.纳米混悬剂研究进展[J].药学进展,2007,21(1):9-13.
    [88]Keck CM, Muller RH. Drug nanocrystals of poorly soluble drugs produced by high pressure homogenisation[J]. Eur J Pharm Biopharm,2006,62(1):3-16.
    [89]Krause KP, Muller RH. Production and characterization of highly concentrated nanosuspensions by high pressure homogenization[J]. Int J Pharm,2001,214(1-2): 21-24.
    [90]朱建芬,吴祥根.纳米混悬剂的制备方法及在药剂学中应用的研究进展[J].中国医药工业杂志,2006,37(3):196-200.
    [91]陈莉,汤态,陆伟根.纳米混悬剂粒径稳定性及其控制策略[J].世界临床药物,2010,31(4):245-249.
    [92]Peters K, Leitzke S, Diederichs JE, et al. Preparation of a clofazimine nanosuspension for intravenous use and evaluation of its therapeutic efficacy in murine Mycobacterium avium in-fection[J]. J Antimicrob Chemother,2000,45(1):77-83.
    [93]刘伟,王东凯.奥沙利铂磷脂复合物及其纳米混悬剂的制备[J].沈阳药科大学学报,2007,24(4):197-200.
    [94]闵智伟,鲁传华,吴鸿飞,完茂林.葛根素纳米水分散体系和混悬剂小鼠体内药代动力学过程比较[J].安徽中医学院学报,2009,28(1):52-55.
    [1]成晓静,刘华钢,赖茂祥.莪术的化学成分及药理作用研究概况[J].广西中医学院学报,2007,10(1):79.
    [2]雷云霞,孙立立,杨书斌等.高效液相色谱法测定郁金饮片中姜黄素的含量[J].中国药师,2007,10(6):604.
    [3]崔晶,翟光喜,赵宇.姜黄素微乳的体内吸收研究[J].中草药,2007,38(3):369.
    [4]Jayaprakasha GK, Jagan Mohan Rao L, Sakariah KK. Improved HPLC method for the determination of curcumin, demethoxycurcumin, and bisdemethoxycurcumin[J]. J Agric Food Chem,2002,50 (13):3668-3672.
    [5]Li J, Jiang Y, Wen J, et al. A rapid and simple HPLC method for the determination of curcumin in rat plasma:assay development, validation and application to a pharmacokinetic study of curcumin liposome[J]. Biomed Chromatogr,2009,23 (11):1201-1207.
    [6]Singh RS, Das U, Dimmock JR, et al. A general HPLC-UV method for the quantitative determination of curcumin analogues containing the 1,5-diaryl-3-oxo-1,4-pentadienyl pharmacophore in rat biomatrices[J]. J Chromatogr B Analyt Technol Biomed Life Sci,878(28):2796-2802.
    [7]Bhutani MK, Bishnoi M, Kulkarni SK. Anti-depressant like effect of curcumin and its combination with piperine in unpredictable chronic stress-induced behavioral, biochemical and neurochemical changes. Pharmacology [J], Biochemistry and Behavior, 2009,92(1):39-43.
    [8]Heath DD, Pruitt MA, Brenner DE, Rock CL. C urcumin in plasma and urine:quantitation by high-performance liquid chromatography[J]. Journal of Chromatography B,2003,783(1):287-295.
    [9]Pak Y, Patek R, Mayersohn M. Sensitive and rapid isocratic liquid chromatography method for the quantitation of curcumin in plasma[J]. Journal of Chromatography B,2003,796(1):339-346.
    [10]张建超,肖琪,程光明,卢金清,许汉林.分离纯化姜黄素的大孔树脂筛选[J].湖北中医杂志.2009,31(4):56-57.
    [11]韩刚,范颖,翟冠钰,董月,原海忠,林庆辉.姜黄素滴丸在大鼠体内药代动力学研究[J].中成药,2009,31(3):377-379.
    [12]许汉林,孙芸,邵继征,熊利容,张念,黄成刚.姜黄素脂质体在大鼠体内药代动力学研究[J].湖北中医学院学报,2007,9(1):42-43.
    [13]张继芬,侯世祥,刘惠莲,卢懿.小鼠血浆中姜黄素的含量测定及药动学研究[J].中国药学杂志,2007,42(4):288-290.
    [14]Liu AC, Lou HX, Zhao LX, Fan PH. Validated LC/MS/MS assay for curcumin and tetrahydrocurcumin in rat plasma and application to pharmacokinetic study of phospholipid complex of curcumin[J]. Journal of Pharmaceutical and Biomedical Analysis,2006,40(1):720-727.
    [15]刘安昌,娄红祥,赵丽霞.姜黄素在大鼠体内的代谢研究[J].上海中医药杂志,2008,42(8):74-77.
    [16]白文莉,张树云,郭菊玲等.比色法测定姜黄中有效成分的含量[J].中国药事,2007,21(10):840.
    [17]张苗,吕庆銮,岳宁宁等.酶催化-荧光猝灭法测定药物中的姜黄素[J].化学分析计量,2008,17(6):22.
    [18]李好枝.体内药物分析[M].中国医药科技出版社,2008:122-138.
    [19]Wang YJ, Pan MH, Cheng AL, Lin LI, Ho YS, Hsieh CY, Lin JK. Stability of curcumin in buffer solutions and characterization of its degradation products[J]. Journal of Pharmaceutical and Biomedical Analysis,1997,15:1867-1876.
    [1]Bundgaard H, Nielsen NM. Glycolamide ester as a novel biolabile prodrug type for non-steroidal anti-inflammatory carboxylic acid drugs [J]. Int J Pharm,1988,43:101-110.
    [2]张伟明,邓英杰,顾吉晋,董晓东等.酒石酸长春瑞滨及其脂质体中药物的降解动力学研究[J].中国医药导报,2009,6(23):23-24.
    [3]操锋,郭健新,平其能,邵云等.灯盏乙素酯类前药的合成,理化性质及降解研究[J].药学学报,2006,41(7):595-602.
    [4]王海凤,王东凯,张翠霞,肖玉婷等.吡硫醇丙酯前体药物的合成及其降解动力学[J].中国新药杂志,2007,16(18):1488-1491.
    [5]雷杰杰,田文艳,孙红卫,周欣羽等.齐多夫定棕榈酸酯及其脂质体在小鼠和大鼠血浆中的稳定性[J].沈阳药科大学学报,2007,24(1):13-15.
    [1]魏树礼,张强.生物药剂学与药代动力学[M].北京大学医学出版社,2004:85-89.
    [2]陈宏,陈宇英,张振书等.姜黄素抗大肠癌作用研究[J].肿瘤研究与临床,2006,18(1):4-7.
    [3]崔国惠, 陈卫华, 陈燕.姜黄素和肿瘤坏死因子α调节淋巴瘤细胞VEGF的表达及抗血管形成的研究[J].中国医院药学杂志,2008,28(23):1918-1919.
    [4]Xu Y, Ku BS, Yao HY, Lin YH, Ma X, Zhang YH and Li XJ. Antidepressant effects of curcumin in the forced swim test and olfactory bulbectomy models of depression in rats[J]. Pharmacology Biochemistry and Behavior 2005; 82:200-206.
    [5]Xu Y, Ku BS, Yao HY, Lin YH, Ma X, Zhang YH and Li XJ. The effects of curcumin on depressive-like behaviors in mice[J]. European Journal of Pharmacology 2005; 82: 200-206.
    [6]Bhutani MK, Bishnoi M and Kulkarni SK. Anti-depressant like effect of curcumin and its combination with piperine in unpredictable chronic stress-induced behavioral, biochemical and neurochemical changes[J]. Pharmacology Biochemistry and Behavior 2009; 88:511-522.
    [7]Wang Y, Pan M, Cheng A, Lin L, Ho Y, Hsieh C and Lin J. Stability of curcumin in buffer solutions and characterization of its degradation products[J]. Journal of Pharmaceutical and Biomedical Analysis 1997; 31:385-396.
    [8]温彩霞,丁延辉,许建华.姜黄素注射液在小鼠体内的药代动力学[J].中药药理与临床,2006,22(6):19-20.
    [9]韦晓瑜,陈世忠.姜黄素注射液在大鼠体内的药代动力学[J].北京大学学报,2003,35(3):230.
    [10]Luo JP, Hubbard JW and Midha KK. Studies on the mechanism of absorption of depot neuroleptics:fluphenazine decanoate in sesame oil[J]. Pharmaceutical Research 1997; 14: 1079-1084.
    [11]Li XG, Choi JS. Effect of genistein on the pharmacokinetics of paclitaxel administered orally or intravenously in rats[J]. International Journal of Pharmaceutics,2007,377: 188-193.
    [1]钱帅,张建军,高缘.纳米混悬剂研究进展[J].药学进展,2007,21(1):9-13.
    [2]Cornelia MK, Rainer HM. Drug nanocrystals of poorly soluble drugs produced by high pressure homogenisation [J]. Eur J Pharm Biopharm,2006,62 (1):3216.
    [3]Patravale VB, Date AA, Kulkarni RM. Nanosuspensions:a promising drug delivery strategy[J]. J Pharmacy and Pharmacology,2004,56:827-840.
    [4]Kazemian, Kazemeini M. Optimized experimental design for natural clinoptilolite zeolite ball milling to produce nano powders[J]. Powder Technology,2010,203(2):389-396.
    [5]Singh R, Lillard Jr JW. Nanoparticle-based targeted drug delivery[J]. Experimental and Molecular Pathology,2009,86:215-223.
    [6]吴义辉,伍玉甜.药物在辛醇-水体系分配系数的应用[J].广东药学,2000,10(4):12-14.
    [1]Senior J, Radomsky M郑俊民等译.可注射缓释制剂[M].化学工业出版社,2005:36-56.
    [1]王任烨, 徐英,蒋文高,姚海燕等.姜黄素对慢性应激大鼠不同脑区cAMP水平及腺苷酸环化酶活性的影响[J].中国药学杂志,2007,42(2):121-124.
    [2]施桂兰,库宝善,姚海燕.解郁丸对慢性应激大鼠HPA轴和免疫系统的影响[J].中国中药杂志,2007,32(15):1551-1554.
    [3]丁桂霞,胡大吉,李有田.抑郁症发病机理的研究进展[J].中华中医药学刊,2007,25(4):733-734.
    [4]杨楠,李秋波,闫玉忠.浅析抑郁症发病机理的研究现状[J].中国社区医师,2006,8(16):5-6.
    [1]Spiegel D, Giese-Davis J. Depression and Cancer:Mechanisms and Disease Progression[J]. Society of Biological Psychiatry,2003,54:269-282.
    [2]Massie MJ. Prevalence of Depression in Patients With Cancer[J]. Journal of the National Cancer Institute,2004,32:57-71.

© 2004-2018 中国地质图书馆版权所有 京ICP备05064691号 京公网安备11010802017129号

地址:北京市海淀区学院路29号 邮编:100083

电话:办公室:(+86 10)66554848;文献借阅、咨询服务、科技查新:66554700