赖氨大黄酸的制备及其抗肿瘤作用机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
恶性肿瘤是人类健康的第二大杀手,在某些地区已上升为头号杀手,世界每年死于恶性肿瘤的人数呈逐年递增的趋势。开发新型抗肿瘤药物将是人类战胜恶性肿瘤的重要手段之一。近年来,随着免疫学和分子生物学的发展,人们对肿瘤形成过程中的受体、基因和信号转导有了比较深入的了解,在此基础上发展的靶向治疗获得了突破性的进展,靶向药物的研究是当前临床研究最活跃的领域之一,其中靶向蛋白酪氨酸激酶的药物已经成为研究的热点。蛋白酪氨酸激酶(PTKs)是一种能选择性地使不同底物的酪氨酸残基磷酸化的一种酶,大致可分为受体酪氨酸激酶、非受体酪氨酸激酶、IR和Janus激酶等。在病理条件下PTKs会持续活化,阻断其对细胞分化、生长和凋亡等的调节功能,诱发肿瘤。
     据报道受体PTKs家族中超过50%的成员发生与人类恶性肿瘤相关的突变或者过度表达,其中30%的成员与人类实体肿瘤相关。EGFR家族是受体PTKs中重要成员,其包括4个成员,分别是EGFR(ErbB-1,HER-1)、HER-2(ErbB-2)、HER-3(ErbB-3)和HER-4(ErbB-4)。在全世界每年诊断的150万新增乳腺肿瘤患者中,27%—30%与EGFR的过度表达有关,20%—25%与HER-2的过度表达有关。单纯针对EGFR或者HER-2的单克隆抗体或蛋白酪氨酸激酶抑制剂(TKI)存在很大局限性,针对单靶点的药物在肿瘤治疗方面有效率不足30%,虽然与其它药物联用可以在一定程度上提高药物疗效,但是通过受体酪氨酸激酶活性的相互补充,很快产生耐药。因此开发针对EGFR和HER-2双靶点的酪氨酸激酶抑制剂将会提高肿瘤治疗的疗效。蛋白酪氨酸激酶的抑制剂—大黄素能够通过抑制HER-2的自身磷酸化和交互磷酸化,阻断ErbB-2的酪氨酸激酶活性。对HER-2过度表达的乳腺癌细胞采用大黄素处理能够降低酪氨酸的过度磷酸化,提高细胞对化疗药物紫杉醇的敏感性,本文研究同为大黄蒽醌类化合物的大黄酸是否对EGFR和HER-2蛋白酪氨酸激酶活性起到双重抑制作用。
     大量文献报道大黄中的三种蒽醌类化合物大黄酸、大黄素和芦荟大黄素能够抑制肿瘤细胞增殖、促进肿瘤细胞凋亡、抑制肿瘤血管形成、逆转肿瘤细胞多药耐药等多方面的抗肿瘤活性。但是由于其水溶性低,生物利用度差,目前除应用其泻下作用外,其它药理活性还没有应用于临床。大黄酸与大黄中其它蒽醌类化合物最显著的区别就是大黄酸存在一个游离的羧基,因此我们选择大黄酸的羧基进行结构改造。
     第一部分以增加水溶性为目标的大黄酸衍生物制备
     目的:通过对大黄酸进行结构改造,制备出水溶性好的大黄酸衍生物或其盐。
     方法:方法一,在特定反应条件下,通过大黄酸与赖氨酸反应生成赖氨酰大黄酸,借助赖氨酸存在的游离氨基和羧基增加赖氨酰大黄酸在水中的溶解度。方法二,在一定反应条件下,通过赖氨酸与大黄酸之间的酸碱反应生成稳定的盐,增加大黄酸在水中的溶解度。通过MTT方法检测产物的体外抗肿瘤活性。
     结果:按照方法一获得了黄色和紫红色两种产物,其中黄色产物不溶于水,紫红色产物水溶性好,对两种产物进行了红外光谱测定,证明两种产物均为大黄酰赖氨酸甲酯,应用MTT方法检测了这两种产物体外抗肿瘤活性,发现二者的体外抗肿瘤活性远远低于大黄酸,而且其产率也比较低,分别为6%和9%,因此没有进行进一步研究。按照方法二获得了水溶性好的紫色产物,经过紫外分光光度法、HPLC法和红外光谱法验证此产物为赖氨大黄酸盐(RHL),通过MTT方法证实RHL与大黄酸具有同等的抑制肿瘤细胞增殖活性。
     结论:大黄酰赖氨酸甲酯虽然能够部分改善大黄酸水溶性差的缺点,但是其体外抗肿瘤活性远远低于大黄酸,没有对其进一步研究。RHL不仅解决了大黄酸水溶性难的问题,而且其抗肿瘤活性与大黄酸相当,有进一步开发成抗肿瘤药物的潜力。
     第二部分赖氨大黄酸体外抗肿瘤活性及分子机制研究
     目的:观察RHL体外抗肿瘤作用,探讨其分子作用机制。
     方法:通过MTT检测细胞增殖,Western blot检测受体PTKs通路蛋白表达水平及其磷酸化水平的变化,免疫细胞化学检测EGFR和HER-2蛋白表达水平变化,RT-PCR检测HER-2在基因转录水平上的变化,流式细胞技术(FACS)检测细胞凋亡和细胞周期。
     结果:RHL抑制不同来源的乳腺癌细胞系(MCF-7、SK-Br-3和MDA-MB-231)、卵巢癌SK-OV-3细胞系和人脐静脉内皮细胞(HUVEC)增殖,IC_(50)分别为95μmol/L、80μmol/L、110μmol/L、85μmol/L和200μmol/L。而且RHL在抑制肿瘤细胞增殖方面与大黄酸相近。对RHL抗肿瘤机制研究中发现:在EGFR高表达和HER-2低表达的MCF-7细胞,其通过抑制EGFR的磷酸化及其下游Raf/MEK/ERK信号通路的磷酸化抑制细胞增殖,并且其与紫杉醇等其它化疗药物联合发挥协同抗肿瘤作用;在EGFR和HER-2高表达的SK-Br-3细胞,其能够抑制EGFR和HER-2蛋白表达和蛋白磷酸化水平,并且能够诱导肿瘤细胞通过p53和p21途径发生细胞凋亡。
     结论:RHL是新型的靶向EGFR和HER-2双靶点的受体蛋白酪氨酸激酶抑制剂。通过抑制细胞增殖和诱导细胞凋亡途径发挥抗肿瘤作用。
     第三部分赖氨大黄酸体内抗肿瘤活性及其与紫杉醇的协同作用
     目的:观察RHL体内抗肿瘤活性和探讨其与紫杉醇协同抗肿瘤作用的机制。
     方法:建立小鼠肝癌和人类肿瘤裸鼠移植模型,分别采用灌胃、腹腔注射途径给药,观察小鼠对药物的耐受情况和药物的抗肿瘤活性。并通过免疫组化方法观察肿瘤组织EGFR蛋白磷酸化水平变化。
     结果:RHL单独用药有一定的抑制肿瘤生长作用。采用腹腔注射方法给药优于灌胃给药。其与紫杉醇合用,在小鼠H22肝癌和MCF-7乳腺癌裸鼠移植瘤模型均表现出明显的协同作用。免疫组化研究结果表明RHL能够抑制由紫杉醇引起的EGFR和ERK蛋白磷酸化水平升高。
     结论:RHL体内应用抑制小鼠H22肝癌和人类乳腺癌裸鼠移植瘤生长,腹腔注射途径给药比灌胃给药有更高抑瘤率。在小鼠H22肝癌和人类乳腺癌裸鼠移植瘤,RHL与Taxol联合有协同抗肿瘤作用。
     总结:本文通过对大黄酸的改造获得了水溶性好的RHL。RHL抑制乳腺癌细胞和卵巢癌细胞的增殖,并且在抑制肿瘤细胞增殖方面与大黄酸相近。进一步研究发现RHL抑制EGFR和HER-2蛋白磷酸化水平,是靶向EGFR和HER-2双靶点的小分子TKI。其通过抑制EGFR高表达的MCF-7细胞株细胞增殖,和诱导EGFR和HER-2高表达的SK-Br-3细胞株细胞凋亡发挥抗肿瘤活性。其与紫杉醇等化疗药联合应用能增强化疗药的抗肿瘤作用,有望成为肿瘤化疗药或者辅助化疗药。
Malignancy is the second cause of death, and in some areas it has been the first one. The mortality of malignancy increases yearly in the world. The important way for human to overcome malignancy is to explore new anti-tumor drugs. With the development of immunology and molecular biology, a number of receptors, genes and signal transduction pathways involved in tumor generation had been reported. Based on this discovery, targeted therapy of tumor has been made a breakthrough. The development of targeted drugs is being one of the most attractive fields in tumor research and therapy. The drugs targeting protein tyrosine kinase had already been the focus point. A tyrosine kinase is an enzyme that can transfer a phosphate group from ATP to a tyrosine residue in a protein, including receptor PTKs, non-receptor PTKs, IR and Janus. In the abnormal condition, PTKs are constructively activated, consequently blocking the regulation of cell differentiation, growth and apoptosis, and inducing tumor progression.
     More than 50% of the known receptor PTKs have been found to be either mutated or overexpressed in association with human malignancies and 30% of them have been implicated in human solid toumors. EGFR family, an important group of PTKs, includes four members: EGFR (ErbB-1), HER-2 (ErbB-2), HER-3 (ErbB-3) and HER-4 (ErbB-4). There are one million and five hundred thousand new cases of breast cancer around the world every year, 27%—30% of them with EGFR overexpression and 20%—25% of them with HER-2 overexpression. Therapeutic use of protein tyrosine kinase inhibitors (TKIs) and monoclonal antibody targeting EGFR or HER-2 have some limitations. The efficacy of an anti-tumor drug that targets only a single point is less than 30%. The anti-tumor activity can be improved by the combination of chemotherapeutic agents; however, resistance occours soon through the mutual compensation of receptor PTKs. So, dual receptor tyrosine kinase inhibitors targeting both EGFR and HER-2 may increase the response rate. Emodin, a tyrosine kinase inhibitor, suppresses HER-2 atuo-phosphorylation and trans-phosphorylation and blocks tyrosine kinase activity of ErbB-2. Emodin can inhibit the growth of HER-2 -overexpressing tumors in mice and also sensitize these tumors to taxol. In this article, we studied if rhein analog showed dual inhibition to EGFR and HER-2.
     Rhein, emodin and aloe-emodin are the main anthraquinone derivatives of rhubarb. It was reported that the anthraquinone derivatives of rhubarb could inhibit tumor cell proliferation, induce tumor cell apoptosis, block blood vessel formation and even reverse multi-drug resistance. However, all of them have poor water solubility and poor bioavailability. Their therapeutic applications are limited in cathartic. The most significant difference between rhein and other anthraquinone derivatives is that rhein has a free carboxyl group. Therefore structural modification can be directed to the free carboxyl group.
     PartⅠPreparation of rhein analog with high solubility in water
     Purpose: To prepare rhein derivatives easily dissolving in water.
     Methods: One is the preparation of lysyl-rhein. The carboxyl group of rhein reacts with the amino group of lysine in special condition to form lysyl-rhein, which improves its solubility in water depending on the free amino group and the free carboxyl group of lysine. Another is the preparation of rhein lysinate (RHL). The carboxyl group of rhein reacts with the amino group of lysine in room temperature to form RHL, which easily dissolves in water. By MTT assay, we observed the anti-tumor activities of these new products in vitro.
     Results: We obtained two new products by the first method. One was yellow and did not dissolve in water. Another was purple and easy to dissolve. These two compounds were proved to be rheinyl lysine methyl ester, by infrared spectrum analysis. The anti-tumor efficacy of these two compounds was far less than that of rhein and the yield of them was very low, only 6% and 9% respectively. No further study was conducted. By second mthod we obtained a purple compound with good solubility. It was proved to be RHL by violet spectrophotometry, HPLC, and infrared spectrum analysis. By MTT assay, RHL had the same anti-tumor activity as rhein (in DMSO solution).
     Conclusions: Although rheinyl lysine methyl ester could partly improve the solubility, the anti-tumor activity was far less than rhein. No further study was conducted. RHL not only easily dissolved in water but also showed the same anti-tumor activity as rhein. That warrants further development of RHL.
     PartⅡThe anti-tumor activity of rhein lysinate and its molecular mechanism in tumor treatment in vitro
     Purpose: To observe the anti-tumor activity of RHL and investigate its molecular mechanism.
     Methods: By MTT assay investigating tumor cell proliferation; by Western blot investigating the phosphorylation of protein and the expression level of proteins in PTKs signaling pathway; by immunocytochemistry observing the expression level of EGFR and HER-2; by RT-PCR observing HER-2 change in transcriptional level; by FACS observing cell apoptosis and cell cycle arrest.
     Results: RHL inhibited the proliferation of different breast cancer cells (MCF-7, SK-Br-3 and MDA-MB-231), ovarian cancer cells (SK-OV-3) and HUVEC. The IC_(50) was 95μmol/L, 80μmol/L, 110μmol/L, 85μmol/L and 200μmol/L, respectively. It had the same anti-prliferation with rhein. In MCF-7 cells with EGFR overexpression and without HER-2 overexpression, RHL inhibited the phosphorylation of EGFR, the phosphorylation of downstream Raf/MEK/ERK signaling pathway and showed synergistic effect with chemotherapeutic drugs such as taxol. In SK-Br-3 cells with both EGFR and HER-2 overexpression, RHL inhibited the phosphorylation of EGFR and HER-2 and induced cell apoptosis through p53 and p21 pathway.
     Conclusions: RHL was a new dual target protein tyrosine kinase inhibitor aiming directly at EGFR and HER-2. RHL not only inhibited tumor cell proliferation but also induced tumor cell apoptosis.
     PartⅢThe anti-tumor activity of rhein lysinate and its synergism with taxol in tumor treatment in vivo
     Purpose: To observe anti-tumor activity of RHL and explore its synergistic effect with taxol. Methods: Establishing the model of mouse H22 liver cancer and human MCF-7 breast cancer xenografts in athymic mice, administering RHL by intragastric administration and intraperitoneal injection, observing the tolerance in mice and the therapeutic effect of RHL, and observing the phosphorylation of EGFR in tumor tissue by immunohistochemistry method.
     Results: RHL inhibited tumor growth. The efficacy of RHL administered by intraperitoneal injection was better than by intragastric administration. RHL in combination with taxol showed the synergistic effect in inhibiting tumor growth in mouse H22 liver cancer and MCF-7 breast cancer xenografts in athymic mice. The result of immunohistochemistry demonstrated that RHL partly abrogated the increase of the phosphorylation of EGFR and ERK induced by taxol.
     Conclusions: RHL inhibited tumor growth of mouse liver cancer H22 and human breast cancer MCF-7 xenografts in athymic mice. The efficacy of RHL administered by intraperitoneal injection is better than that by intragastric administration. It had a synergistic anti-tumor activity with taxol.
     In summary, we obtained a new compound - RHL. That is easily dissolved in water. RHL inhibited the proliferation of breast cancer and ovarian cancer cells and had the same potency with rhein. Our results indicated that RHL, as a new dual tyrosine kinase inhibitor, was capable of repressing the tyrosine kinase activity of EGFR and HER-2, accordingly inhibiting the proliferation of EGFR overexpressing MCF-7 breast cancer cells and inducing cell apoptosis of EGFR and HER-2 overexpressing SK-Br-3 breast cancer cells. Moreover, it sensitized tumor cells to chemotherapeutic drugs such as taxol. These results may have important therapeutic implications in cancer chemotherapy.
引文
1.Okutani D,Lodyga M,Han B,et al.Src protein tyrosine kinase family and acute inflammatory responses.Am J Physiol Lung Cell Mol Physiol.2006;291(2):L129-141.
    2.Le MN,Kohanski RA,Wang LH,et al.Dual mechanism of signal transducer and activator of transcription 5 activation by the insulin receptor.Mol Endocrinol.2002;16(12):2764-2779.
    3.毛志强,项伟中,俞雁,等.蛋白酪氨酸激酶小分子抑制剂研究进展.中国药物化学杂志.2005;15(2):121-129.
    4.Bianco R,Gelardi T,Damiano V,et al.Rational bases for the development of EGFR inhibitors for cancer treatment.Int J Biochem Cell Biol.2007;39:1416-1431.
    5.Marmor MD,Skaria KB,Yarden Y.Signal transduction and oncogenesis by ErbB/HER receptors.Int J Radiat Oncol Biol Phys.2004;58(3):903-913.
    6.李文杰,朱孝峰,成志毅.靶向HER-2受体酪氨酸激酶抗肿瘤药物研究进展.中国药学杂志.2005;40(15):1121-1124.
    7.Yang XR,Sherman ME,Rimm DL,et al.Differences in risk factors for breast cancer molecular subtypes in a population-based study.Cancer Epidemiol Biomarkers Prev.2007;16(3):439-443.
    8.Daniele L,Macri L,Schena M,et al.Predicting gefitinib responsiveness in lung cancer by fluorescence in situ hybridization/chromogenic in situ hybridization analysis of EGFR and HER-2 in biopsy and cytology specimens.Mol Cancer Ther.2007;6(4):1223-1229.
    9.Liu FS.Molecular carcinogenesis of endometrial cancer.Taiwan J Obstet Gynecol.2007;46(1):26-32.
    10.Yu D,Liu B,Tan M,et al.Overespression of c-erbB-2/neu in breast cancer cells confers increased resistance to Taxol via mdr-1-independent mechanisms.Oncogene.1996;13(6):1359-1365.
    11.Yu D,Liu B,Jing T,et al.Overexpression of both p185 c-erbB-2 and p170 mdr-1renders breast cancer cells highly resistant to Taxol.Oncogene.1998;16(16):2087-2094.
    12.朱孝峰,刘宗潮,谢冰芬,等.SUCI02选择性抑制HER-2受体酪氨酸激酶磷酸化及其对HER-2过表达乳腺癌.癌症.2003;22(8):790-794.
    13.封宇飞,曾春兰.EGFR和ErbB-2双重酪氨酸激酶抑制剂拉帕替尼二甲苯磺酸酯的研究进展.中国新药杂志.2007;16(23):1990-1993.
    14.Sliwkowski MX,Lofgren JA,Lewis GD,et al.Nonclinical studies addressing the mechanism of action of Trastuzumab(Herceptin).Semin Onco1.1999;26(4 suppl 12):60-70.
    15.Konecny GE,Pegram MD,Venkatesan N,et al.Activity of the dual kinase inhibitor lapatinib(GW572016) against HER-2-overexpressing and trastuzumab -treated breast cancer cells.Cancer Res.2006;66(3):1630-1639.
    16.Zhang L,Lau YK,Xi L,et al.Tyrosine kinase inhibitors,emodin and its derivative repress HER-2-induced celular transformation and metastasis- associated properties.Oncogene.1998;16(22):2855-2863.
    17.郑虎.《药物化学》.第四版,北京:人民卫生出版社,2000:279-283.
    18.郑俊华,果德安.大黄的现代研究.北京:北京大学医学出版社,2007:3-5.
    19.陈英玉,郑合勇,胡建达,等.大黄素抑制HL-60/ADR耐药细胞增殖和诱导凋亡的作用.中国实验血液学杂志.2007;15(5):955-960.
    20.李家宁,吕福祯,肖金玲.大黄素对人肺腺癌细胞Anip973增殖周期及凋亡基因的影响.中国中西医结合杂志.2006;26(11):1015-1020.
    21.Su YT,Chang HL,Shyue SK,et al.Emodin induces apoptosis in human lung adenocarcinoma cells through a reactive oxygen species-dependent mitochondrial signaling pathway.Biochem Pharmacol.2005;70(2):229-241。
    22.Shieh DE,Chen YY,Yen MH,et al.Emodin-induced apoptosis through p53-dependent pathway in human hepatoma cells.Life Sci.2004;74(18):2279-2290.
    23.Huang Q,Shen HM,Shui G,et al.Emodin inhibits tumor cell adhesion through disruption of the membrane lipid Raft-associated integrin signaling pathway.Cancer Res.2006;66(11):5807-5815。
    24.Huang Q,Shen HM,Ong CN.Emodin inhibits tumor cell migration through suppression of the phosphatidylinositol 3-kinase-Cdc42/Racl pathway.Cell Mol Life Sci.2005;62(10):1167-1175。
    25.Kaneshiro T,Morioka T,Inamine M,et al.Anthraquinone derivative emodin inhibits tumor-associated angiogenesis through inhibition of extracellular signal-regulated kinase 1/2 phosphorylation.Eur J Pharmacol.2006;553(1-3):46-53.
    26.Kwak H J,Park M J,Park CM,et al.Emodin inhibits vascular endothelial growth factor-A-induced angiogenesis by blocking receptor-2(KDR/F1k- 1 )phosphorylation.Int J Cancer.2006;118(11):2711-2720.
    27.Wang XH,Wu SY,Zhen YS.Inhibitory effects of emodin on angiogenesis.Acta Pharmacol Sin.2004;39(4):254-258.
    28.Acevedo-Duncan M,Russell C,Patel S,et al.Aloe-emodin modulates PKC isozymes,inhibits proliferation,and induces apoptosis in U-373MG glioma cells.Int Immunopharmacol.2004;4(14):1775-1784.
    29.Guo JM,Xiao BX,Liu Q,et al.Anticancer effect of aloe-emodin on cervical cancer cells involves G2/M arrest and induction of differentiation.Acta Pharmacol Sin.2007;28(12):1991-1995.
    30.Lin JG,Chen GW,Li TM,et al.Aloe-emodin induces apoptosis in T24 human bladder cancer cells through the p53 dependent apoptotic pathway.J Urol.2006;175(1):343-347.
    31.Lian LH,Park EJ,Piao HS,et al.Aloe emodin-induced apoptosis in t-HSC/C1-6cells involves a mitochondria-mediated pathway.Basic Clin Pharmacol Toxicol.2005;96(6):495-502.
    32.Cárdenas C,Quesada AR,Medina MA.Evaluation of the anti-angiogenic effect of aloe-emodin.Cell Mol Life Sci.2006;63(24):3083-3089.
    33.Lin ML,Chen SS,Lu YC,et al.Rhein induces apoptosis through induction of endoplasmic reticulum stress and Ca~(2+)-dependent mitochondrial death pathway in human nasopharyngeal carcinoma cells.Anticancer Res.2007;27(5A):3313-3322.
    34.Ip SW,Weng YS,Lin SY,et al.The role of Ca~(2+) on rhein-induced apoptosis in human cervical cancer Ca Ski cells.Anticancer Res.2007;27(1A):379-89.
    35.Lin S,Fujii M,Hou DX.Rhein induces apoptosis in HL-60 cells via reactive oxygen species-independent mitochondrial death pathway.Arch Biochem Biophys.2003;418(2):99-107.
    36.Zhang L,Lau YK,Xia W,et al.Tyrosine kinase inhibitor emodin suppresses growth of HER-2-overexpressing breast cancer cells in athymic mice and sensitizes these cells to the inhibitory effect of paclitaxel.Clin Cancer Res.1999;5(2):343-353.
    37.Kuo PL,Hsu YL,Ng LT,et al.Rhein inhibits the growth and induces the apoptosis of Hep G2 cells.Planta Med.2004;70:12-16.
    38.龚伟,黎磊石,孙骅,等.大黄酸对糖尿病大鼠转化生长因子β及其受体表达的影响.肾脏病与透析肾移植杂志.2006;15(2):101-111.
    39.朱加明,刘志红,黄燕飞,等.大黄酸对db/db小鼠糖尿病肾病疗效的观察.肾脏病与透析肾移植杂志.2002;11(1):3-10。
    40.郭美姿,徐海荣,李孝生.大黄酸对小鼠急性肝损伤的影响.中医药研究.2002;18(1):37-38.
    41.郭美姿,李孝生,沈鼎明,等.大黄酸对大鼠肝纤维化形成的影响.中华肝病杂志.2003;11(1):26-29.
    42.刘凯,郑海生.大黄酸的药理作用研究述略.中医药学刊.2004;22(9):1732-1734.
    43.郭啸华,刘志红,彭艾,等.大黄酸对2型糖尿病肾病大鼠疗效观察.中华肝病杂志.2002;18(4):280-284.
    44.张学亮,郭啸华,刘志红,等.大黄酸对低剂量链脲佐菌素肥胖糖尿病大鼠糖尿病肾病的影响.中华内分泌代谢杂志.2005;21(6):563-565.
    45.纪泽泉,黄翠雯,梁成结,等.大黄酸对。肾小球硬化肾皮质凋亡蛋白酶-3活性及细胞凋亡的影响.中华医学杂志.2005;85(26):1836-1841.
    1.Huang Q,Lu G,Shen HM,et al.Anti-cancer properties of anthraquinones from rhubarb.Med Res Rev.2007;27:609-630.
    2.丁岩,梁永拒,陆豫等.大黄素蒽醌衍生物介导KB及KBv200细胞氧化损伤的研究.中草药.2004;35(11):1259-1262.
    3.郑虎.《药物化学》.第四版,北京:人民卫生出版社,2000:279-283.
    4.李敏,李丽霞,刘渝,等.大黄研究进展.世界科学技术-中医药现代化.2006;8(4):34-39.
    5.Bayer health care ag,Franckowiak Gerhard,Ledwoch wolfram,et al.Stable active ingredient complex of salts of the o-acetylsalicylic acid with basic amino acids and glycine:Deutsch,WO/2006/128600[p].2006-07-12.
    6.Gerhard Franckowiak,Hubert Appolt,Gregor leifker,et al.Stable salts of the o-acetylsalicylic acid with basic amino acids:USA,US6773724B2[p].2004-08-10.
    7.山田羲次,宫原匠一郎,河野一二.赖氨酸阿司匹林结晶的制造方法:日本,昭56-113741[p].1981-09-07.
    1.Baselga J,Albanell J.Mechanism of action of anti-HER2 monoclonal antibodies.Ann Oncol,2001;12 Suppl 1:S35-41.
    2.Stephens P,Hunter C,Bignell G,et al.Lung cancer:intragenic ERBB-2 kinase mutations in tumours.Nature,2004;431(7008):525-526.
    3.Cobleigh MA,Vogel CL,Tripathy D,et al.Multinational study of the efficacy and safety of humanized anti-HER-2 monoclonal antibody in women who have HER-2-overexpressing metastatic breast cancer that has progressed after chemotherapy for metastatic disease.J Clin Oncol,1999;17(9):2639-2648
    4.Vogel CL,Cobleigh MA,Tripathy D,et al.Efficacy and safety of trastuzumab as a single agent in first-line treatment of HER-2-overexpressing metastatic breast cancer.J Clin Oncol,2002;20(3):719-726.
    5.Slamon D J,Leyland-Jones B,Shak S,et al.Use of chemotherapy plus a monoclonal antibody against HER-2 for metastatic breast cancer that overexpresses HER-2.N Engl J Med,2001;344(11):783-792.
    6.Motoyama AB,Hynes NE,Lane HA.The efficacy of ErbB receptor-targeted anticancer therapeutics is influenced by the availability of epidermal growth factor-related peptides.Cancer Res,2002;62(11):3151-3158.
    7.Agus DB,Akita RW,Fox WD,et al.Targeting ligand-activated ErbB-2 signaling inhibits breast and prostate tumor growth.Cancer Cell,2002;2(2):127-137.
    8.Cho HS,Mason K,Ramyar KX,et al.Structure of the extracellular region of HER-2 alone and in complex with the Herceptin Fab.Nature,2003; 421(6924):756-760.
    9.Okutani D,Lodyga M,Han B,et al.Src protein tyrosine kinase family and acute inflammatory responses.Am J Physiol Lung Cell Mol Physiol.2006;291(2):L129-141.
    10.封宇飞,曾春兰.EGFR和ErbB-2双重酪氨酸激酶抑制剂拉帕替尼二甲苯磺酸酯的研究进展.中国新药杂志.2007;16(23):1990-1993.
    11.Nakamura H,Takamori S,Fujii T,et al.Cooperative cell-growth inhibition by ombination treatment with ZD1839(Iressa) and trastuzumab(Herceptin) in on-small-cell lung cancer.Cancer-Lett.2005;230(1):33-46.
    12.Chan CT,Metz MZ,Kane SE.Differential sensitivities of trastuzumab (Herceptin)-resistant human breast cancer cells to phosphoinositide-3 kinase(PI3K)and epidermal growth factor receptor(EGFR) kinase inhibitors.Breast Cancer Res Treat.2005;91(2):187-201.
    13.Konecny GE,Pegram MD,Venkatesan N,et al.Activity of the dual kinase inhibitor lapatinib(GW572016) against HER - 2 - overexpressing and trastuzumab -treated breast cancer cells.Cancer Res.2006;66(3):1630-1639.
    14.Coley HM,Shotton CF,Ajose-Adeogun A,et al.Receptor tyrosine kinase(RTK)inhibition is effective in chemosensitising EGFR-expressing drug resistant human ovarian cancer cell lines when used in combination with cytotoxic agents.Biochem Pharmacol 2006;72:941-948.
    15.Qiu L,Di W,Jiang Q,et al.Targeted inhibition of transient activation of the EGFR-mediated cell survival pathway enhances paclitaxel-induced ovarian cancer cell death.Int J Oncol 2005;27:1441-1448.
    16.Qiu L,Zhou C,Sun Y,et al.Paclitaxel and ceramide synergistically induce cell death with transient activation of EGFR and ERK pathway in pancreatic cancer cells.Oncol Rep 2006;16:907-913.
    17.Kuo PL,Hsu YL,Ng LT,et al.Rhein inhibits the growth and induces the apoptosis of Hep G2 cells.Planta Med 2004;70:12-16.
    18.Ip SW,Weng YS,Lin SY,et al.The role of Ca~(2+) on rhein-induced apoptosis in human cervical cancer Ca Ski cells.Anticancer Res.2007;27(1A):379-389.
    19.Lin ML,Chen SS,Lu YC,et al.Rhein induces apoptosis through induction of endoplasmic reticulum stress and Ca~(2+)-dependent mitochondrial death pathway in human nasopharyngeal carcinoma cells.Anticancer Res.2007;27(5A):3313-3322.
    20.刘梁英,李孝生,万晓强.川芎嗪和大黄酸对肝细胞增殖和凋亡的影响.中华肝病杂志.2006;14(3):219-221.
    21.万晓强,李孝生,曾庆贵,刘梁英.大黄酸对TNF-α和TG诱导L-02肝细胞凋亡的影响.胃肠病学和肝病学杂志.2006;15(4):383-386.
    1.El Etreby MF,Liang Y.Effect of antiprogestins and tamoxifen on growth inhibition of MCF-7 human breast cancer cells in nude mice.Breast Cancer Res Treat.1998;49:109-117.
    2.Damaraju VL,Bouffard DY,Wong CK,et al.Synergistic activity of troxacitabine (Troxatyl~(TM)) and gemcitabine in pancreatic cancer.BMC Cancer.2007;7:121
    3.Zhang L,Lau YK,Xia W,et al.Tyrosine kinase inhibitor emodin suppresses growth of HER-2/neu-overexpressing breast cancer cells in athymic mice and sensitizes these cells to the inhibitory effect of paclitaxel.Clin Cancer Res.1999;5:343-353.
    4.Su JL,Shih JY,Yen ML,et al.Cyclooxygenase-2 induces EP1- and HER-2/Neu-dependent vascular endothelial growth factor-C up-regulation:a novel mechanism of lymphangiogenesis in lung adenocarcinoma.Cancer research.2004;64:554-564.
    5.Coley HM,Shotton CF,Ajose-Adeogun A,et al.Receptor tyrosine kinase(RTK)inhibition is effective in chemosensitising EGFR-expressing drug resistant human ovarian cancer cell lines when used in combination with cytotoxic agents.Biochem Pharmacol.2006;72:941-948.
    6.Qiu L,Di W,Jiang Q,et al.Targeted inhibition of transient activation of the EGFR-mediated cell survival pathway enhances paclitaxel-induced ovarian cancer cell death.J Oncol.2005;27:1441-1448.
    7.Qiu L,Zhou C,Sun Y,et al.Paclitaxel and ceramide synergistically induce cell death with transient activation of EGFR and ERK pathway in pancreatic cancer cells.Oncol Rep.2006;16:907-913.
    1.Huang Q,Lu G,Shen HM,et al.Anti-Cancer Properties of Anthraquinones from Rhubarb.Med Res Rev.2007;27(5):609-630
    2.段淑娥,李敏.中草药中蒽醌化合物的研究进展.西安文理学院学报.2005;8(1):25-28.
    3.李敏,李丽霞,刘渝,等.大黄研究进展.世界科学技术-中医药现代化.2006;8(4):34-39.
    4.郑虎.《药物化学》.第四版,北京:人民卫生出版社,2000:279-283.
    5.Pecere T,Gazzola MV,Mucignat C,et al.Aloe-emodin is a new type of anti-cancer agent with selective activity against neuroectodermal tumors.Cancer Res.2000;60(11):2800-2804.
    6.富泽龙,刘晓霓,张承玉,等.芦荟大黄素对人胃癌细胞株SGC-7901的增殖抑制作用.牡丹江医学院学报.2006;27(4):8-11.
    7.Cha TL,Qiu L,Chen CT,et al.Emodin down-regulates androgen receptor and inhibits prostate cancer cell growth.Cancer Res.2005;65(6):2287-2295.
    8.黄绿叶,胡建达,陈鑫基,等.C-myc在大黄素抑制HL-60细胞增殖及诱导凋亡中的作用.中华血液学杂志.2005;6(6):348-351.
    9.邱学德,刘乔保,孙力莉,等.大黄素对膀胱肿瘤细胞株BIU87和724的作用研究.云南医药.2004;25(3):192-194.
    10.Lee N J,Choi JH,Koo BS,et al.Antimutagenicity and cytotoxicity of the constituents from the aerial parts ofrumex acetosa.Biol.Pharm.Bull.2005;28(11):2158-2161.
    11.朱峰,刘新光,梁念慈.大黄素、芹菜素抑制人卵巢癌细胞侵袭的体外实验研究.癌症.2003;24(4):358-362.
    12.刘剑波,高学岗,连涛,等.大黄素在体外诱导人肝癌细胞HepG2发生凋亡 的初步研究.癌症.2003;22(12):1280-1283.
    13.王心华,甄永苏.大黄素抑制人高转移巨细胞肺癌PG细胞的肿瘤转移相关性质.癌症.2001;20(8):789-793.
    14.姜晓峰,甄永苏.大黄素逆转肿瘤细胞多药抗药性的作用.药学学报.1999;34(3):164-167.
    15.Lee HZ,Hsu SL,Liu MC,et al.Effects and mechanisms of aloe-emodin on cell death in human lung squamous cell carcinoma.Eur J Pharmacol.2001;431(3):287-295.
    16.Kuo PL,Lin TC,Lin CC.The antiproliferative activity of aloe-emodin is through p53-dependent and p21-dependent apoptotic pathway in human hepatoma cell lines.Life Sci.2002;71(16):1879-1892.
    17.Yeh FT,Wu CH,Lee HZ.Signaling pathway for aloe-emodin-induced apoptosis in human H460 lung nonsmall carcinoma cell.Int J Cancer.2003;106(1 ):26-33.
    18.黄云虹,甄永苏.大黄酸诱导肿瘤细胞凋亡及与丝裂霉素的协同作用.药学学报.2001;36(5):334-338.
    19.Su YT,Chang HL,Shyue SK,et al.Emodin induces apoptosis in human lung adenocarcinoma cells through a reactive oxygen species-dependent mitochondrial signaling pathway.Biochem Pharmacol.2005;70(2):229-241.
    20.Chen YC,Shen SC,Lee WR,et al.Emodin induces apoptosis in human promyeloleukemic HL-60 cells accompanied by activation of caspase-3 cascade but independent of reactive oxygen species production.Biochem Pharmacol.2002;64(12):1713-1724.
    21.Srinivas G,Anto R J,Srinivas P,et al.Emodin induces apoptosis of human cervical cancer cells through poly(ADP-ribose) polymerase cleavage and activation of caspase-9.Eur J Pharmacol.2003;473(2-3):117-125.
    22.李家宁,吕福祯,肖金玲.大黄素对人肺腺癌细胞Anip973增殖周期及凋亡基因的影响.中国中西医结合杂志.2006;26(11):1015-1017.
    23.Wang C,Wu X,Chen M,et al.Emodin induces apoptosis through caspase-3-dependent pathway in HK-2 cells.Toxicology.2007;231(2-3):120-128.
    24.Shieh DE,Chen YY,Yen MH,et al.Emodin-induced apoptosis through p53-dependent pathway in human hepatoma cells.Life Sci.2004;74(18):2279-2290.
    25.Lee HZ,Lin C J,Yang WH,et al.Aloe-emodin induced DNA damage through generation of reactive oxygen species in human lung carcinoma cells.Cancer Lett.2006;239(1):55-63.
    26.Lian LH,Park E J,Piao HS,et al.Aloe Emodin-induced apoptosis in t-HSC/C1-6cells involves a mitochondria-mediated pathway.Basic Clin Pharmacol Toxicol.2005;96(6):495-502.
    27.Chen HC,Hsieh WT,Chang WC,et al.Aloe-emodin induced in vitro G2/M arrest of cell cycle in human promyelocytic leukemia HL-60 cells.Food Chem Toxicol.2004;42:42(8):1251-1257.
    28.Lin JG,Chen GW,Li TM,et al.Aloe-emodin induces apoptosis in T24 human bladder cancer cells through the p53 dependent apoptotic pathway.J Urol.2006;175(1):343-347.
    29.Mijatovic S,Maksimovic-Ivanic D,Radovic J,et al.Aloe-emodin decreases the ERK-dependent anticancer activity of cisplatin.Cell Mol Life Sci.2005;62(11):1275-1282.
    30.Mijatovic S,Maksimovic-Ivanic D,Radovic J,et al.Anti-glioma action of aloe emodin:the role of ERK inhibition.Cell Mol Life Sci.2005;62(5):589-598.
    31.Kuo PL,Hsu YL,Ng LT,et al.Rhein inhibits the growth and induces the apoptosis of Hep G2 cells.Planta Med.2004;70(1):12-16.
    32.Bironaite D,Ollinger K.The hepatotoxicity of rhein involves impairment of mitochondrial functions.Chem Biol Interact 1997;103(1):35-50.
    33.Kagedal K,Bironaite D,Ollinger K.Anthraquinone cytotoxicity and apoptosis in primary cultures of rat hepatocytes.Free Radic Res.1999;31(5):419-428.
    34.徐丽敏,毛舒和.大黄酸对colo-16细胞线粒体的作用.天津医科大学学报.1999;5(4):109-110.
    35.Lin S,Fujii M,Hou DX.Rhein induces apoptosis in HL-60 cells via reactive oxygen species-independent mitochondrial death pathway.Arch Biochem Biophys.2003;418(2):99-107.
    36.Raimondi F,Santoro P,Maiuri L,et al.Reactive nitrogen species modulate the effects of rhein,an active component of senna laxatives,on human epithelium in vitro.J Pediatr Gastroenterol Nutr.2002;34(5):529-534.
    37.Castiglione S,Fanciulli M,Bruno T,et al.Rhein inhibits glucose uptake in Ehrlich ascites tumor cells by alteration of membrane-associated functions.Anticancer Drugs.1993;4(3):407-414.
    38.万晓强,李孝生,曾庆贵,等.大黄酸对TNF-α和TG诱导L-02肝细胞凋亡的影响.胃肠病学和肝病学杂志.2006;15(4):383-386.
    39.王心华,吴淑英,甄永苏.大黄素对血管生成的抑制作用.药学学报.2004;39(4):254-258.
    40.吕盈盈,钱家鸣,邱辉忠.大黄素对结肠癌血管内皮生长因子受体的影响及对结肠癌抑制作用的研究.中国中西医结合消化杂志.2005;13(5):310-313.
    41.Kaneshiro T,Morioka T,Inamine M,et al.Anthraquinone derivative emodin inhibits tumor-associated angiogenesis through inhibition of extracellular signal-regulated kinase 1/2 phosphorylation.Eur J Pharmacol.2006;553(1-3):46-53.
    42.Kwak H J,Park MJ,Park CM,et al.Emodin inhibits vascular endothelial growth factor-A-induced angiogenesis by blocking receptor-2(KDR/Flk-1)phosphorylation.Int.J.Cancer.2006;118(11):2711-2720.
    43.Cárdenas C,Quesada AR,Medina MA.Evaluation of the anti-angiogenic effect of aloeoemodin.Cell Mol Life Sci.2006;63(24):3083-3089.
    44.王振军,赵博.大黄酸在抑制血管生成中的应用[P].专利申请号:02159686.7
    45.Lin S,Li J J,Fujii M.Rhein inhibits TPA-induced activator protein-1 activation and cell transformation by blocking the JNK-dependent pathway.Int J Oncol.2003;22(4):829-833.
    46.Huang Q,Shen HM,Ong CN.Emodin inhibits tumor cell migration through suppression of the phosphatidylinositol 3-kinase-Cdc42/Racl pathway.Cell Mol Life Sci.2005;62(10):1167-1175.
    47.Kim MS,Park MJ,Kim SJ et al.Emodin suppresses hyaluronic acid-induced MMP-9 secretion and invasion of glioma cells.Int J Oncol.2005;27(3):839-846.
    48.Huang Q,Shen HM,Ong CN,et al.Inhibitory effect of emodin on tumor invasion through suppression of activator protein-1 and nuclear factor-kB.Biochem Pharmacol.2004;68(2):361-371.
    49.陆长虹,李杰,郭伟剑,等.大黄素对乳腺癌多药耐药细胞株MCF-7/Adr的耐药逆转作用.临床肿瘤学杂志.2004;9(4):340-343.
    50.胡凯文,侯丽,陈信义,等.大黄酸/大黄素抗多药耐药肿瘤细胞研究.中国中医基础医学杂志.1998;4(11):19-20.
    51.van Gorkom BA,Timmer-Bosscha H,de Jong S,et al.Cytotoxicity of rhein,the active metabolite of sennoside laxatives,is reduced by multidrug resistance-associated protein.Br J Cancer.2002;86(9):1494-1500.
    52.Zhang L,Hung MC.Sensitization of HER-2-overexpressing non-small cell lung cancer cells to chemotherapeutic drugs by tyrosine kinase inhibitor emodin.Oncogene.1996;12(3):571-576.
    53.Zhang L,Lau YK,Xia W,et al.Tyrosine kinase inhibitor emodin suppresses growth of HER-2-overexpressing breast cancer cells in athymic mice and sensitizes these cells to the inhibitory effect of paclitaxel.Clin Cancer Res.1999;5(2):343-353.
    54.陈玉英,杨洁,胡庆沈,等.大黄素增强砷剂对食道癌细胞的促凋亡作用.上海交通大学学报(医学版).2006;26(11):1227-1232.
    55.Jing YW,Yi J,Chert YY,et al.Dicoumarol alters cellular redox state and inhibits nuclear factor kappa B to enhance arsenic trioxide-induced apoptosis.Acta Biochim Biophys Sin(Shanghai).2004;36(3):235-242.
    56.Garg AK,Buchholz TA,Aggarwal BB.Chemosensitization and radiosensitization of tumors by plant polyphenols.Antioxid Redox Signal.2005;7(11-12):1630-1647.
    57.Fenig E,Nordenberg J,Beery E,et al.Combined effect of aloe-emodin and chemotherapeutic agents on the proliferation of an adherent variant cell line of Merkel cell carcinoma.Oncol Rep.2004;11(1):213-217.
    58.Fanciulli M,Gentile FP,Bruno T,et al.Inhibition of membrane redox activity by rhein and adriamycin in human glioma cells.Anticancer Drugs.1992;3(6):615-621.
    59.Floridi A,Pulselli R,Gentile FP,et al.Rhein enhances the effect of adriamycin on mitochondrial respiration by increasing antibiotic-membrane interaction.Biochem Pharmacol.1994;47(10):1781-1788.
    60.Yang J,Li H,Chen YY,et al.Anthraquinones sensitize tumor cells to arsenic cytotoxicity in vitro and in vivo via reactive oxygen species-mediated dual regulation of apoptosis.Free Radic Biol Med.2004;37(12):2027-2041.
    61.Kwak HJ,Park MJ,Park CM,et al.Emodin inhibits vascular endothelial growth factor-A-induced angiogenesis by blocking receptor-2(KDR/Flk-1)phosphorylation.Int J Cancer.2006;118(11 ):2711-2720.
    62.Koyama J,Morita I,Tagahara K,et al.Chemopreventive effects of emodin and cassiamin B in mouse skin carcinogenesis.Cancer Lett.2002;182(2):135-139
    63.袁利超,程延安,党双锁,等.大黄素、黄芪多糖对大鼠实验性肝癌的预防作用. 中国药房.2005;16(8):574-576.
    64.Kupchan SM.Novel plant-derived tumor inhibitors and their mechanisms of action.Cancer Treat Rep.1976;60(8):1115-1126.
    65.Kupchan SM,Karim A.Tumor inhibitors.114.Aloe-emodin:Antileukemic principle isolated from Rhamnus frangula L.Lloydia.1976;39(4):223-224.
    66.孙红,王浩.Her-2与乳腺癌研究进展.现代肿瘤医学.2006;14(3):365-367.
    67.Zhang L,Chang C J,Bacus SS et al.Suppressed transformation and induced differentiation of HER-2/neu-overexpressing breast cancer cells by emodin.Cancer Res.1995;55(17):3890-3896.
    68.Zhang L,Lau YK,Xi L,et al.Tyrosine kinase inhibitors,emodin and its derivative repress HER-2-induced cellular transformation and metastasis-associated properties.Oneogene.1998;16(22):2855-2863.
    69.张刚,沐贤友.NF-κB及其相关分子研究进展.国外医学(临床生物化学与检验学分册).2002;23(5):246-248.
    70.Kumar A,Dhawan S,Aggarwal BB.Emodin(3-methyl-1,6,8-trihydroxyanthra quinone) inhibits TNF induced NF-kappaB activation,IkappaB degradation,and expression of cell surface adhesion proteins in human vascular endothelial cells.Oncogene.1998;17(7):913-918.
    71.Li HL,Chen HL,Li H,et al.Regulatory effects of emodin on NF-kappaB activation and inflammatory cytokine expression in RAW264.7 macrophages.Int J Mol Med.2005;16(1):41-47.
    72.Mendes AF,Caramona MM,de Carvalho AP,et al.Diacerhein and rhein prevent interleukin-lbetainduced nuclear factor-kappaB activation by inhibiting the degradation of inhibitor kappaB-alpha.Pharmacol Toxicol.2002;91(1):22-28.
    73.Chen Y,Yang L,Lee TJ.Oroxylin A inhibition of lipopolysaccharide-induced iNOS and COX-2 gene expression via suppression of nuclear factor-kappaB activation.Biochem Pharmacol.2000;59(11):1445-1457.
    74.Mijatovic S,Maksimovic-Ivanic D,Radovic J,et al.Aloe-emodin prevents cytokine-induced tumor cell death:The inhibition of auto-toxic nitric oxide release as a potential mechanism.Cell Mol Life Sci.2004;61(14):1805-1815.
    75.许少峰,付丽.p53研究的新进展.中华病理学杂志.2004;33(6):559-561.
    76.姚逸临。肿瘤抑制基因p53的研究进展.广东医学。2006;27(8):1263-1265.
    77.Pecere T,Sarinella F,Salata C,et al.Involvement of p53 in specific anti-neuroectodermal tumor activity of aloe-emodin.Int J Cancer.2003;106(6):836-847.
    78.邹循锋,鄢业鸿.线粒体P细胞色素C介导的凋亡通路的研究进展.医学综述.2007;13(1):16-19.
    79.Ip SW,Weng YS,Lin SY,et al.The role of Ca~(2+) on rhein-induced apoptosis in human cervical cancer Ca Ski cells.Anticancer Res.2007;27(1A):379-389.
    80.Bironaite D,Ollinger K.The hepatotoxicity of rhein involves impairment of mitochondrial functions.Chem Biol Interact.1997;103(1):35-50.
    81.Lee HZ.Protein kinase C involvement in aloe-emodin and emodin-induced apoptosis in lung carcinoma cell.Br J Pharmacol.2001;134(5):1093-103.

© 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