力达霉素抗胰腺癌作用的分子机制研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
胰腺癌是一种预后极差的消化系统恶性肿瘤,其死亡率高,5年生存率低于5%,发病率呈逐年增加趋势。由于胰腺癌发病隐袭,临床表现多无特异性,病情进展迅速,早期缺乏敏感和特异的诊断指标,大多数患者在确诊时已达晚期或有远处转移,只有10%~15%的患者有手术切除的机会,其中能根治者仅5%~7.5%,因此药物治疗对于胰腺癌患者来说是必不可少的治疗手段。
     力达霉素(Lidamycin,LDM)是本研究所从一株放线菌(Streptomyces globisporus C-1027)代谢产物中获得的对多种肿瘤细胞有强烈杀伤作用的大分子肽类抗肿瘤抗生素。力达霉素分子由一个酸性的辅基蛋白和一个烯二炔发色团组成,其中发色团是力达霉素的主要活性部分,具有断裂DNA的作用,而辅基蛋白的主要作用是保护发色团。大量研究表明,力达霉素具有极强的抗肿瘤活性,目前正在进行临床Ⅱ期试验研究。本课题选用人胰腺癌PANC-1和SW1990细胞株,探讨力达霉素对人胰腺癌细胞的影响及其作用机制,并通过人胰腺癌细胞SW1990裸鼠移植瘤实验,观察力达霉素在体内的抗胰腺癌作用。此外,本课题还初步研究了力达霉素与健择(Gemcitabine,吉西他滨)联合作用对胰腺癌细胞的影响,并探讨其作用机制,从而为力达霉素的临床应用提供理论依据。
     一、力达霉素作用于胰腺癌的体外实验研究
     1.MTT法检测力达霉素(LDM)对人胰腺癌PANC-1和SW1990细胞的增殖抑制作用。不同浓度的阿霉素(ADM)、丝裂霉素(MMC)、紫杉醇(Taxol)、吉西他滨(Gemcitabine)和LDM作用细胞48 h后,利用MTT法检测各种药物对胰腺癌细胞的增殖抑制作用。MTT结果显示,与临床常用化疗药物相比,LDM抑制细胞的增殖作用更强,对PANC-1和SWl990细胞的IC_(50)值分别为0.955±0.414 nM和0.426±0.212 nM,低于其他药物的10~3~10~4倍。
     2.Hoechst 33342荧光染色法和Annexin V-FITC/PI染色结合流式细胞仪检测细胞凋亡。不同浓度LDM作用细胞48h后,细胞凋亡比率逐渐增高,呈浓度依赖性,其中0.1nM LDM、1nM LDM、2 nM LDM和10 nM LDM作用于PANC-1细胞的细胞凋亡率分别为12.33%±0.66%、32.26%±1.58%、45.03%±3.16%和59.44%±4.04%,作用于SW1990细胞的细胞凋亡率分别为22.21%±0.28%、22.83%±0.26%、42.52%±0.67%和50.35%±0.56%,显著高于对照组的6.15%±1.47%和7.51%±0.45%(P<0.001)。荧光显微镜下可见1 nM、2 nM和10 nM LDM作用组的细胞出现染色质凝集、细胞核固缩、核碎裂、形成凋亡小体等典型的凋亡细胞形态学改变。
     3.利用PI单染并结合流式细胞仪检测LDM对细胞周期的影响。检测结果显示,1 nM LDM主要将细胞阻滞在G2/M期,2 nM LDM能引起S期和G2/M期阻滞,10 nM LDM则主要诱导S期阻滞。
     4.利用Transwell实验观察LDM对胰腺癌细胞的迁移和侵袭能力的影响。实验结果显示,LDM对PANC-1细胞和SW1990细胞的迁移和侵袭能力均有显著的抑制作用,并且LDM的作用浓度越大,抑制作用越强,呈现明显的浓度依赖关系。
     5.Western blot检测结果显示LDM能够减少基质金属蛋白酶-9(MMP-9)蛋白在胰腺癌细胞中的表达水平,并且随着LDM作用浓度的增加,抑制蛋白表达的作用亦随之加强,提示LDM通过降低细胞内MMP-9的含量而有效抑制细胞的迁移和侵袭能力,并呈剂量依赖性。
     6.Western blot和RT-PCR结果表明,LDM能够明显抑制胰腺癌细胞中VEGF蛋白的表达,但不影响细胞中VEGF mRNA的表达水平,说明LDM是在转录后的翻译水平对VEGF的表达进行调节。
     7.细胞中K-ras蛋白和mRNA表达水平,以及p-AKT和NF-κB蛋白含量都随LDM作用浓度的增高而降低,呈现一定的浓度依赖性。LDM对于细胞中的AKT蛋白含量没有影响,表明LDM能够抑制AKT蛋白的磷酸化过程。
     二、力达霉素作用于胰腺癌的体内实验研究
     1.利用人胰腺癌SW1990移植瘤裸鼠模型检测LDM的体内抗肿瘤作用。在瘤块接种后的第7天开始给药,每周1次,共2次。LDM通过尾静脉注射,给药剂量为0.02mg/kg和0.04 mg/kg;Gemcitabine通过腹腔注射,给药剂量为80mg/kg;对照组给予生理盐水。动物实验结果显示,LDM和Gemcitabine对SW1990细胞移植瘤的生长均有抑制作用,并且LDM显示出更强的抑瘤效果。实验第31天处死动物,剥取各组动物的瘤块进行比较,计算抑瘤率,其中Gemcitabine组的抑瘤率为38.87%,LDM 0.02 mg/kg剂量组和0.04 mg/kg剂量组的抑瘤率分别为65.71%和78.27%,与对照组相比具有显著性差异。
     2.肿瘤组织切片经HE染色,显微镜下可见,LDM作用组的肿瘤组织出现血管减少,肿瘤细胞坏死增多等病理学变化。
     3.利用脱氧核糖核酸末端转移酶介导的缺口末端标记(TUNEL)技术,对裸鼠肿瘤组织中的凋亡细胞进行观察和比较。TUNEL实验结果显示,与对照组和Gemcitabine组相比,LDM治疗组中凋亡细胞明显增多。
     4.免疫组化结果显示,LDM能够抑制肿瘤细胞中K-ras蛋白和CD31蛋白的表达,并呈现剂量效应关系,表明LDM具有抑制肿瘤新生血管生成的作用。
     三、力达霉素与吉西他滨联合作用于胰腺癌的体外实验研究
     目前,健择(Gemcitabine,吉西他滨)是临床上用于治疗胰腺癌的一线药物,健择与其他化疗药物合用治疗肿瘤亦得到广泛研究和应用,本课题对力达霉素联合健择对胰腺癌细胞的作用进行了研究,并探讨其主要作用机理。
     1.MTT结果显示,1 nM LDM与500 nM Gemcitabine联合作用时,能够明显抑制细胞增殖,其CDI<0.75,具有协同抗肿瘤作用。
     2.TUNEL实验结果显示,与单独用药相比较,1 nM LDM联合500 nM Gemcitabine作用于PANC-1和SW1990细胞,能够明显提高凋亡细胞的比率。流式细胞仪检测结果显示,二者联合用药作用PANC-1和SW1990细胞的细胞凋亡率分别为59.44%±1.54%和54.68%±2.62%,显著高于Gemcitabine单独作用组的18.48%±0.94%和25.79%±2.06%(P<0.05)。
     3.RT-PCR结果显示,500 nM Gemcitabine和1 nM LDM联合作用能够显著降低PANC-1和SW1990细胞中K-ras mRNA的表达水平。
     4.Western blot结果表明,LDM与Gemcitabine联合作用于PANC-1和SW1990细胞,能够减少细胞中K-ras蛋白的表达;500 nM Gemcitabine或1nM LDM单独作用时,使细胞中的NF-κB蛋白水平提高,但二者联用能够显著降低细胞中NF-κB的蛋白表达水平。
     5.利用荧光探针JC-1负载,通过共聚焦激光扫描显微镜观察和测定细胞线粒体膜电位的改变,结果表明,LDM与Gemcitabine联合作用能够明显降低细胞的线粒体膜电位。
     6.Caspase-3活性测定结果表明,LDM和Gemcitabine联合作用,能够导致PANC-1和SW1990细胞内的凋亡系统caspase-3蛋白酶激活;Western blot结果显示,LDM与Gemcitabine联合作用,能够显著降低PANC-1和SW1990细胞中Bcl-2的蛋白表达水平。
Pancreatic carcinoma is a highly malignant neoplasm characterized by locally advanced unresectable disease or metastasis at the time of diagnosis. Current therapies are largely ineffective in this disease and the 5-year survival rate remains dismal. Pancreatic cancer is now one of the most common causes of cancer death worldwide. Complete surgical resection remains the only potentially curative treatment modality for patients with pancreatic cancer. Unfortunately, the vast majority of patients present with unresectable or metastatic disease. Even among patients undergoing a potentially curative resection, many will develop metastatic disease. Therefore, the development of novel approaches for pancreatic cancer treatment is essential as tumor cells are highly resistant to most conventional chemotherapy drugs.
     Enediyne antibiotics have been focused on their potent antitumor activity due to their unique ability to damage the DNA of tumor cells by inducing single strand and/or double strand breaks through free radical attacks on the deoxyribose moieties in DNA. Lidamycin (LDM, also named as C-1027) is a member of the enediyne antibiotic family, which was produced by a Streptomyces globisporus C-1027 strain isolated in China. LDM molecule contains an enediyne chromophore (MW 843 Da) responsible for the extremely potent bioactivity and a noncovalently bound apoprotein (MW 10.5 kDa), which formed a hydrophobic pocket for protecting the chromophore. Lidamycin is currently being evaluated in phaseⅡclinical trials as a potential chemotherapeutic agent in China. As reported, LDM shows extremely potent cytotoxicity, anti-angiogenic activity and marked growth inhibition of transplantable tumors in mice. To determine the action of lidamycin on pancreatic cancer cells and to reveal its possible mechanism, we investigated the effects of lidamycin on two established pancreatic cell lines, PANC-1 and SW1990.
     1. Effects of lidamycin on PANC-1 and SW1990 cells cultured in vitro
     1.1 Inhibition of cell proliferation was measured by the MTT assays. A remarkable difference in chemosensitivity to lidamycin, mitomycin, adriamycin, taxol, and gemcitabine was found in both cell lines. The IC_(50) value of lidamycin for PANC-1 or SW1990 cells was 0.955±0.414nM or 0.426±0.212nM, and much lower than that of other drugs.
     1.2 Flow cytometry combined with FITC-Annexin V/PI staining showed that LDM at 1nM induced earlier apoptosis in both cell lines (the apoptotic ratio were 32.26%±1.58% and 22.83%±0.26% in PANC-1 and SW1990 cells, respectively). The rate of apoptosis was significant enhanced when cells were incubated with 2nM or 10nM LDM for 48h (45.03%±3.16% and 59.44%±4.04% in PANC-1 cells, 42.52%±0.67% and 50.35%±0.56% in SW1990 cells, respectively). After exposure to LDM for 48h, most cells presented typical morphologic changes of apoptosis such as chromatin condensation or shrunken nucleus by Hoechst33342 staining. Some condensed nuclei were observed when cells exposed to higher concentrations of LDM.
     1.3 Flow cytometric cell cycle analysis showed that LDM at 1nM induced G2/M arrest and at 2nM induced S and G2/M arrest. The accumulation of cells in S phase was induced by 10nM of LDM.
     1.4 The effect of LDM on the migration or invasion of PANC-1 and SW1990 cells through an extracellular matrix was examined using a transwell cell migration chamber assay. A dose-dependent reduction in cell migration or invasion in both cell lines was found after exposure to LDM.
     1.5 The expression of MMP-9 was determined by Western blot analysis. PANC-1 and SW1990 cells treated with LDM showed a dose-dependent decrease in the expression of MMP-9. The level of MMP-9 was markedly reduced by 1nM of LDM.
     1.6 The decrease of VEGF protein expression was observed in PANC-1 and SW1990 cells after exposure to LDM for 48h, Whereas the VEGF mRNA level was unaffected. The results indicated that LDM could affect VEGF on the translation phase.
     1.7 The result of RT-PCR showed that the levels of K-ras mRNA were decreased after treated with LDM at 2nM in PANC-1 cells and 1nM in SW1990 cells. Meanwhile, the levels of K-ras, phospho-Akt and NF-κB proteins decreased in a dose-dependent manner in either cell line, whereas the level of Akt protein was unaffected by LDM.
     2. In vivo antitumor activity of lidamycin
     2.1 Athymic female BALB/c (nu/nu) mice (20±2g, obtained from the Institute for Experimental Animals, Chinese Academy of Medical Sciences) at the age of 4-6 weeks were used for SW1990 human pancreatic tumor xenografts. Animals bearing s.c. tumors received by i.v. injection (200μl) either PBS (vehicle control) or LDM once a week for 2 weeks. The doses of LDM were 0.02mg/kg and 0.04mg/kg respectively. Animals received by i.p. injection (<300μl) of gemcitabine at a dose of 80mg/kg as the positive control. The mice were weighed and tumor sizes were measured with a vernier caliper and recorded every other day. Tumor volume was calculated using the formula: length×width~2×0.5. Growth of the established s.c. tumors in nude mice was decreased significantly when treated with LDM over a period of 3 weeks compared with control PBS-treated animals. Treatment with LDM at the dose of 0.02mg/kg and 0.04mg/kg inhibited the growth of pancreatic carcinoma SW1990 xenografts by 66% and 72%, respectively. The antitumor activity of LDM is more remarkable than that of gemcitabine. The body weights of animals showed no significant differences between control and treated groups.
     2.2 By hematoxylin and eosin staining, the pathologic changes of tumor tissues from SW1990 xenografted nude mice were observed. There were a decrease in tumor angiogenesis and an increase in necrosis of tumor cells in LDM-treated groups.
     2.3 The induction of apoptosis in the pancreatic tumors was evaluated by TUNEL (terminal deoxynucleotidyl transferase-mediated nick end labeling) assay. In tumors from control-treated mice, the median number of apoptotic cells was minimal. The number of apoptotic cells in tumors of LDM-treated mice increased.
     2.4 Vessel density in the tumors was determined by immunohistochemical staining with antibodies against CD31. The number of CD31-positive cells was significantly decreased in tumors from LDM-treated groups. Moreover, the level of K-ras protein in LDM-treated tumors showed a decreased trend by immunohistochemical examination.
     3. Effects on pancreatic carcinoma cells by the combination of lidamycin with gemcitabine
     Gemcitabine (2', 2'-difluorodeoxycytidine) is currently considered the optimal treatment for patients with newly diagnosed metastatic pancreatic cancer. Preclinical studies suggested that the combination of gemcitabine and other therapeutic agents may have greater activity against pancreatic cancer. In the present study, we investigated the effects on PANC-1 and SW1990 cells by the combination of lidamycin and gemcitabine.
     3.1 The effects of LDM alone or in combination with gemcitabine on the viability of pancreatic cancer cells were determined by the MTT assays. A significant reduction in proliferation was observed in PANC-1 and SW1990 cells treated with the combination of 1nM LDM with 500nM gemcitabine.
     3.2 Induction of apoptosis by lidamycin, gemcitabine, and the combination was measured by TUNEL assay and flow cytometry combined with FITC-Annexin V/PI staining. There was a significant potentiation of apoptosis observed in PANC-1 and SW1990 cells treated with gemcitabine and lidamycin compared with cells treated with gemcitabine alone.
     3.3 The level of mRNA encoding K-ras in PANC-1 and SW1990 cells treated with gemcitabine (500nM), lidamycin (1nM) and the combination was determined by reverse transcription-PCR analysis. The result showed that the K-ras mRNA level was significantly reduced by the combination of gemcitabine and lidamycin.
     3.4 In the PACN-1 and SW1990 cells, the combination of gemcitabine and lidamycin resulted in significant down-regulation of the K-ras and Bcl-2 compared with cells treated with gemcitabine or lidamycin alone. Gemcitabine treatment enhanced NF-κB level. However, treatment with lidamycin prevented the gemcitabine-induced NF-κB enhancement.
     3.5 Confocal microscopy analyses of cells stained with JC-1 clearly showed that PANC-1 and SW1990 cells have significantly reduced mitochondrial membrane potential (Δ_(ψm)) after treated with the combination of gemcitabine and lidamycin. Moreover, the caspase-3 activity significantly increased when cells were treated with the combination of gemcitabine and lidamycin.
引文
1. Jemal A, Tiwari RC, Murray T, et al. Cancer statistics, 2004. CA Cancer J Clin, 2004; 54(1): 8-29.
    2. Jemal A, Clegg LX, Ward E, et al. Annual report to the nation on the status of cancer, 1975-2001, with a special feature regarding survival. Cancer, 2004; 101:3-27.
    3. Schmidt CM, Powell ES, Yiannoutsos CT, et al. Pancreaticoduodenectomy: a 20-year experience in 516 patients. Arch Surg, 2004; 139:718-727.
    4. Wang L, Yang GH, Li H, et al. Pancreatic cancer mortality in China (1991-2000). World J Gastroenterol, 2003; 9(8): 1819-1823.
    5. Burris HA III, Moore MJ, Andersen J, et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol, 1997; 15:2403-2413.
    6. Nicolaou KC, Smith AL, Yue EW. Chemistry and biology of natural and designed enediynes. Proc Natl Acad Sci USA 1993; 90: 5881-5888.
    7. Hu JL, Xue YC, Xie MY, Zhang R. A new macromolecular antitumor antibiotic, C-1027. Ⅰ. Discovery, taxonomy of producing organism, fermentation and biological activity. J Antibiot, 1988; 41: 1575-1579.
    8. Otani T, Minami Y, Marunaka T, et al. A new macromolecular antitumor antibiotic, C-1027. Ⅱ. Isolation and physico-chemical properties. J Antibiot, 1988; 41: 1580-1585.
    9. Zhen YS, Ming XY, Yu B, et al. A new macromolecular antitumor antibiotic, C-1027. Ⅲ. Antitumor activity. J Antibiot, 1989; 42: 1294-1298.
    10.甄永苏,薛玉川,邵荣光.烯二炔类新抗生素C1027的抗肿瘤作用研究.中国抗生素杂志,1994;19:164-168.
    11. Xu YJ, Li DD, Zhen YS. Mode of action of C-1027, a new macromolecular antibiotic with highly potent cytotoxocity, on human hepatoma BEL-7402 cells. Cancer Chemother Pharmacol, 1990; 27: 41-46.
    12.甄红英,薛玉川,甄永苏.抗肿瘤抗生素C1027抑制肿瘤血管生成及其抗肿 瘤转移作用.中华医学杂志,1997;77:657-660.
    13.李军智,江敏,薛玉川,甄永苏.抗癌抗生素C1027与单克隆抗体片段偶联物的抗肝癌作用.药学学报,1993;28:260-265.
    14. Mchugh MM, Woynarowski JM, Gawron LS, et al. Effects of the DNA-damaging enediyne C-1027 on intracellular SV40 and genomic DNA in green monkey kidney BSC-1 cells. Biochemistry, 1995; 34(5): 1805-1814.
    15. Mchugh MM, Beerman TA. C-1027-induced alteration in Epstein-Barr viral DNA replication in latently infected cultured human Raji cells: relationship to DNA damage. Biochemistry, 1999; 38(21): 6962-6970.
    16. Lemoine NR, Jain S, Hughes CM, et al. Ki-ras oncogene activation in preinvasive pancreatic cancer. Gastroenterology, 1992; 102:230-236.
    17. Vimalachandran D, Ghaneh P, Costello E, et al. Genetics and prevention of pancreatic cancer. Cancer Control, 2004; 11(1): 6-14.
    18. Dong QG, Sclabas GM, Fujioka S, et al. The function of multiple IkappaB: NF-kappaB complexes in the resistance of cancer cells to Taxol-induced apoptosis. Oncogene, 2002; 21: 6510-6519.
    19. Fujioka S, Sclabas GM, Schmidt C, et al. Inhibition of constitutive NF-kappa B activity by I kappa B alpha M suppresses tumorigenesis. Oncogene, 2003; 22: 1365-1370.
    20. Sclabas GM, Uwagawa T, Schmidt C, et al. Nuclear factor kappa B activation is a potential target for preventing pancreatic carcinoma by aspirin. Cancer, 2005; 103: 2485-2490.
    21. Zhang Z and Rigas B. NF-kappaB, inflammation and pancreatic carcinogenesis: NF-kappaB as a chemoprevention target. Int J Oncol, 2006; 29: 185-192.
    22. Bramhall, SR. The matrix metalloproteinases and their inhibitors in pancreatic cancer: From molecular science to a clinical application. Int J Pancreatol, 1997; 21:1-12.
    23. Anan K, Morisaki T, Katano M, et al. Vascular endothelial growth factor and platelet-derived growth factor are potential angiogenic and metastatic factors in human breast cancer. Surgery, 1996; 119(3): 333-339.
    24. Ahmad T, Eisen T. Kinase inhibition with BAY 43-9006 in Renal Cell carcinoma. Clin Cancer Res, 2004; 10(18 Pt 2):6388S-6392S.
    25. Lennernas B, Albertsson P, et al. Chemotherapy and Antiangiogenesis. Acta Oncologica, 2003; 42(4): 294-303.
    26. Downward J. Targeting RAS signaling pathways in cancer therapy. Nat Rev Cancer, 2003; 3:11-22.
    27. Li D, Xie K, Wolff R, et al. Pancreatic cancer. Lancet, 2004; 363:1049-1057.
    28. Leach SD. Mouse models of pancreatic cancer: the fur is finally flying! Cancer Cell, 2004; 5:7-11.
    29. Almoguera C, Shibata D, Forrester K, et al. Most human carcinomas of the exocrine pancreas contain mutant c-K-ras gene. Cell, 1988; 53: 549-554.
    30. Osada M, Tolkacheva T, Li W, et al. Differential roles of Akt, Rac, and Ral in R-Ras-mediated cellular transformation, adhesion, and survival. Mol Cell Biol, 1999; 19: 6333-6344.
    31. Fleming JB, Shen GL, Hoiloway SE, et al. Molecular consequences of silencing mutant K-ras in pancreatic cancer cells: Justification for K-ras-directed therapy. Mol Cancer Res, 2005; 3:413-423.
    32. Arlt A, Gehrz a, Muerkoster S, et al. Role of NF-κB and Akt/PI3K in the resistance of pancreatic carcinoma cell lines against gemcitabine-induced cell death. Oncogene, 2003; 22:3243-3251.
    33. Wang CY, Mayo MW, Baldwin AS, et al. TNF-α and Cancer Therapy Induced Apoptosis: Potentiation by Inhibition of NF-κB. Science, 1996; 274: 784-787.
    34. Ting AY, Endy D. Decoding NF-κB Signaling. Science, 2002; 298: 1189-1190.
    35. Kucharczak J, Simmons MJ, Fan Y, et al. To be, or not to be: NF-κB is the answer-role of Rel/ NF-κB in the regulation of apoptosis. Oncogene, 2003; 22:8961-8982.
    36. Huxford T, Huang DB, Malek S, et al. The Crystal Structure of the I-κB / NF-κB Complex Reveals Mechanisms of NF-κB Inactivation. Cell, 1998; 95: 759-770.
    37. Sheng H, Shao J, Dubois RN. Akt/PKB activity is required for Ha-Ras-mediated transformation of intestinal epithelial cells. J Biol Chem, 2001; 276:14498-14504.
    38. Antwerp van DJ, Martin SJ, Kafri T, et al. Suppression of TNF-a Induced Apoptosis by NF-kB. Science, 1996; 274: 787-789.
    39. Mohammad RM, Banerjee S, Li Y, et al. Cisplatin-induced antitumor activity is potentiated by the soy isoflavone genistein in BxPC-3 pancreatic tumor xenografts. Cancer, 2006; 106:1260-1268.
    40. Yip-Schneider MT, Nakshatri H, Sweeney CJ, et al. Parthenolide and sulindac cooperate to mediate growth suppression and inhibit the nuclear factor-kappa B pathway in pancreatic carcinoma cells. Mol Cancer Ther, 2005; 4: 587-594.
    41. Liotta LA. Tumor invasion and metastases-role of the extracellular matrix: Rhoads Memorial Award Lecture. Cancer Res, 1986; 46 (1): 1-7.
    42. Bloomston M, Zervos EE and Rosemurgy AS. Matrix metalloproteinases and their role in pancreatic cancer: A review of preclinical studies and clinical trials. Ann Surg Oncol, 2002; 9: 668-674.
    43. Parsons SL, Watson SA, Brown PD, et al. Matrix metalloproteinases. Br J Surg, 1997; 84: 160-166.
    44. Egeblad M, Werb Z. New function for the matrix metalloproteinases in cancer progression. Nature Rev Cancer, 2002; 2: 161-174.
    45. Davies B, Waxman J, Wasan H, et al. Levels of matrix metalloproteinase in bladder cancer correlate with tumor grade and invasion. Cancer Res, 1993; 53:5365-5369.
    46. Bramhall SR Stamp GW, Dunn J, Lemoine NR, et al. Expression of collagenase (MMP2), stromelysin (MMP3) and tissue inhibitor of the metalloproteinases (TIMP) in pancreatic and ampullary disease. Br J Cancer, 1996; 73(8):972-978.
    47. Kuniyasu H, Ellis LM, Evans DB, et al. .Relative expression of E-cadherin and type IV collagenase genes predicts disease outcome in patients with resectable pancreatic carcinoma. Clin Cancer Res, 1999; 5(1):25-33.
    48. Eccles S A, Box GM, et al. Control of lymphatic and hematogenous metastasis of a rat mammary carcinoma by the matrix metalloproteinase inhibitor batimastat (BB94). Cancer Res, 1996; 56:2815-2822.
    49. Koivunen K, Arap W, Valtanen H, et al. Tumor targeting with a selective gelatinase inhibitor. Nat Biotechnol, 1999; 17(8):768-744.
    50. Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med, 2003; 9: 669-676.
    51. Plouet J, Moukadiri H. Characterization of the receptor to vasculotropin on bovine adrenal cortex-derived capillary endothelial cells. J Biol Chem, 1990; 265: 22071-22074.
    52. Vaisman N, Gospodarowicz D, Neufeld G. Characterization of the receptors for vascular endothelial growth factor. J Biol Chem, 1990; 265: 19461-19466.
    53. Lee J, Chow N, Want S, Huan S. Prognostic value of vascular endothelial growth factor expression in colorectal cancer patients. Eur J Cancer, 2000; 36: 748-753.
    54. Takahashi Y, Kitadai Y, Bucana CD, et al. Expression of vascular endothelial growth factor and its receptor, KDR, correlates with vascularity, metastasis, and proliferation of human colon cancer. Cancer Res, 1995; 55: 3964-3968.
    55. Maeda K, Chung YS, Ogawa Y, et al. Prognostic value of vascular endothelial growth factor expression in gastric carcinoma. Cancer, 1996; 77: 858-863.
    56. Takahashi Y, Cleary KR, Mai M, et al. Significance of vessel count and vascular endothelial growth factor and its receptor (KDR) in intestinal-type gastric cancer. Clin Cancer Res. 1996; 2: 1679-1684.
    57. Manders P, Beex LV, Tjan-Heijnen VC, et al. The prognostic value of vascular endothelial growth factor in 574 node-negative breast cancer patients who did not receive adjuvant systemic therapy. Br J Cancer, 2002; 87: 772-778.
    58. George DJ, Halabi S, Shepard TF, et al. Prognostic significance of plasma vascular endothelial growth factor levels in patients with hormone-refractory prostate cancer treated on Cancer and Leukemia Group B 9480. Clin Cancer Res, 2001; 7: 1932-1936.
    59. Gorski DH, Leal AD, Goydos JS. Differential expression of vascular endothelial growth factor-A isoforms at different stages of melanoma progression. J Am Coll Surg, 2003; 197: 408-418.
    60. Fujimoto K, Hosotani R, Wada M, et al. Expression of two angiogenic factors, vascular endothelial growth factor and platelet-derived endothelial cell growth factor in human pancreatic cancer, and its relationship to angiogenesis. Eur J Cancer, 1998; 34: 1439-1447.
    61.Ikeda N, Adachi M, Taki T, et al. Prognostic significance of angiogenesis in human pancreatic cancer. Br J Cancer, 1999; 79: 1553-1563.
    62. Nagase T, Kawata S, Nakajima H, et al. Effect of farnesyltransferase overexpression on cell growth and transformation. Int J Cancer 1999; 80: 126-133.
    63. Smith SE, Burris HA, Loehrer PJ, et al. Preliminary report of a phase II trial of gemcitabine combined with celecoxib for advanced pancreatic cancer. Proc Ann Meeting Am Soc Clin Oncol. 2003; 1502. Abstract.
    64. Tong WG, Ding XZ, Witt RC, et al. Lipoxygenase inhibitors attenuate growth of human pancreatic cancer xenografts and induce apoptosis through the mitochondrial pathway. Mol Cancer Ther. 2002; 1: 929-935.
    65. Muhammad WS. Anti-angiogenesis therapy in pancreatic carcinoma. J Pancreas, 2006; 7(2): 163-173.
    66. Burris HA III, Moore MJ, Andersen J, et al. Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol, 1997; 15: 2403-2413.
    67. Berardi R, Porfiri E, Scartozzi M, et al. Elderly patients with advanced non-small cell lung cancer: A phase II study with weekly cisplatin and gemcitabine. Oncology, 2003; 65: 198-203.
    68. Pino SM, Xiong HQ, McConkey D, et al. Novel therapies for pancreatic adenocarcinoma. Curr Gastroenterol Rep, 2004; 6: 119-125.
    69. Yardley DA. Gemcitabine and docetaxel in metastatic and neoadjuvant treatment of breast cancer. Semin Oncol, 2004; 31: 37-44.
    70. Van Cutsem E, van deVelde H, Karasek P, et al. Phase III trial of gemcitabine plus tipifarnib compared with gemcitabine plus placebo in advanced pancreatic cancer. J Clin Oncol, 2004; 22:1430-1438.
    71. Bramhall SR, Schulz J, Nemunaitis J, et al. A double-blind placebo-controlled, randomised study comparing gemcitabine and marimastat with gemcitabine and placebo as first line therapy in patients with advanced pancreatic cancer. Br J Cancer, 2002; 87:161-167.
    72. Berlin JD, Catalano P, Thomas JP, et al. Phase in study of gemcitabine in combination with fluorouracil versus gemcitabine alone in patients with advanced pancreatic carcinoma: Eastern Cooperative Oncology Group Trial E2297. J Clin Oncol, 2002; 20:3270-3275.
    73. Liptay S, Weber CK, Ludwig L, et al. Mitogenic and antiapoptotic role of constitutive NF-kappaB/Rel activity in pancreatic cancer. Int J Cancer, 2003; 105: 736 - 746
    74. Krajewski S, Thor AD, Edgerton SM, et al. Analysis of Bax and Bcl-2 expression in p53-immunopositive breast cancers. Clin Cancer Res, 1997; 3: 199-208.
    75. Brambilla E, Negoescu A, Gazzeri S, et al. Apoptosis-related factors p53, Bcl-2, and Bax in neuroendocrine lung tumors. Am J Pathol, 1996; 149: 1941-1952.
    76. Sinicrope FA, Ruan SB, Cleary KR, et al. Bcl-2 and p53 oncoprotein expression during colorectal tumorigenesis. Cancer Res, 1995; 55: 237-241.
    77. John DR, Orrenius. Role of mitochondria in toxic cell death. Toxicology, 2002; 182: 491-496.
    78. Freyer JP. Decreased mitochondrial function in quiescent cells isolated from multicellular tumor spheroids. J Cell Physiol, 1998; 176: 138-149.
    79. Hengartner MO. The biochemistry of apoptosis. Nature, 2000; 407: 770-776
    80. Blatt NB, Glick GD. Signaling pathways and effector mechanisms pre-programmed cell death. Bioorg Med Chem, 2001; 9(6): 1371-1384.
    81. Zhang XD, Gillespie SK, Hersey P. Staurosporine induces apoptosis of melanoma by both caspase-dependent and -independent apoptotic pathways. Mol Cancer Ther, 2004; 3(2): 187-197.
    82. Krajewski S, Tanaka S, Takayama S. Investigations of the subcellular distribution of the bcl-2 oncoprotein: residence in the nuclear envelope, endoplasmic reticulum, and outer mitochondria membranes. Cancer Res, 1993; 53: 4701-4714.
    83. Fahy BN, Schlieman MG, Mortenson MM, et al. Targeting BCL-2 overexpression in various human malignancies through NF-κB inhibition by the proteasome inhibitor bortezomib. Cancer Chemother Pharmacol, 2005; 56: 46-54.
    84. Mortenson MM, Schlieman MG., Virudachalam S, et al. Reduction in BCL-2 levels by 26S proteasome inhibition with bortezomib is associated with induction of apoptosis in small cell lung cancer. Lung Cancer, 2005; 49(2): 163-170.
    85. Deeb D, Jiang H, Gao X, et al. Curcumin (Diferuloyl-methane) sensitizes human prostate cancer cells to TRAIL/Apo2L-induced apoptosis by suppressing NF-{kappa}B via inhibition of pro-survival Akt signaling pathway. J Pharmacol Exp Ther, 2007; Feb 8 (on line).
    1. Nagase H. Activation mechanisms of matrix metalloproteinases. Biol Chem. 1997; 378: 151-159.
    2. Davies B, Waxman J, Wasan H, et al. Levels of matrix metalloproteinase in bladder cancer correlate with tumor grade and invasion. Cancer Res. 1993; 53:5365-5369.
    3. Bramhall SR, Stamp GW, Dunn J, et al. Expression of collagenase (MMP2), stromelysin (MMP3) and tissue inhibitor of the metalloproteinases (TIMP) in pancreatic and ampullary disease. Br J Cancer. 1996; 73(8): 972-978.
    4. Kuniyasu H, Ellis LM, Evans DB, et al.. Relative expression of E-cadherin and type Ⅳ collagenase genes predicts disease outcome in patients with resectable pancreatic carcinoma. Clin Cancer Res. 1999; 5(1): 25-33.
    5. Arteaga CL. The epidermal growth factor receptor: frommutant oncogene in nonhuman cancers to therapeutic target in human neoplasia. J Clin Oncol. 2001; 19: 32S-40S.
    6.谢一利,楼丽广.表皮生长因子受体信号通路及其耙向肿瘤治疗.国外医学肿瘤学分册,2005;32(5):361-364.
    7. Bowers G, Reardon D, Hewitt T, et al. The relative role of ErbB1-4 receptor tyrosine kinases in radiation signal transduction responses of human carcinoma cells. 2001; 20(11): 1388-1397.
    8. Newman L, Xia W, Yang HY, et al. Correlation of p27 protein expression with HER-2/neu expression in breast cancer. Mol Carcinog. 2001; 30(3): 169-175.
    9. Hognason T, Chatterjee S, Vartanian T, et al.Epidermal growrth factor receptor induced apoptosis: potentiation of ras signaling. FEBS Lett.2001; 491(1-2):9-15.
    10. Yaden Y, Sliwkowski MX. Untangling the ErbB signaling net work. Nat Rev Mol Cell Biol. 2001; 2(2): 127-137
    11. Klapper LN, Kirschbaum MH, Sela M, et al. Biochemical and clinical implications of the ErhB/HER signaling network of growth factor receptors. Adv Cancer Res. 2000; 77: 25-79
    12. Pedersen MW, Mehorn M, Damstrup I, et al. The type Ⅲ epidermal growth factor receptor mutation: biological significance and potential target for anti-cancer therapy. Ann Oncol. 2001; 12(6): 745-760.
    13. Tobita K, Kijima H, Dowaki S, et al. Epidermal growth factor receptor expression in human pancreatic cancer: Significance for liver metastasis. Int J Mol Med. 2003; 11: 305-309.
    14. Sclabas GM, Fujioka S, Schmidt C, et al. Restoring apoptosis in pancreatic cancer cells by targeting the nuclear factor kappaB signaling pathway with the antiepidermal growth factor antibody IMC-225. J Gastrointest Surg. 2003; 7: 37-43.
    15. Ciardiel!o F, Tortora G. A novel approach in the treatment of cancer: Targeting the epidermal growth factor receptor. Clin Cancer Res. 2001; 7(10): 2958-2970
    16. BaselgaJ, Albannell J, Monia MA, et al. Mechanism of action of trastuzumab and scientific update. Semin Oncol. 2001; 28 (5 Suppl 16): 4-11
    17. Kong WJ, Jiang JD. Advance in anti-tumor therapy targeting the EGFR family of receptor tyrosine kinases. Chinese Pharmacological Bulletin. 2003; 19(8): 847-851.
    18. Schmidt M, Maurer Gebhard M, Groner B, et al. Suppression of metastasis formation by a recombinant single chain antibody-tox in targeted to full-length and oncogenic variant EGF receptors. Oncogene. 1999; 18:1711-1721.
    19. Wakeling AE. Epidermal growth factor receptor tyrosine kinase inhibitors. Curr Opin Pharm. 2002; 2(4): 382-387.
    20. Reynolds LP, Redmer DA. Expression of the angiogenic factor, basic fibreblast growth factor and vascular endothelial growth factors. J Anim Sci. 1998; 76: 1671-1681.
    21. Senger DR, Van de Water L, Brown LF, et al. Vascular permeability factor (VPF VEGF) in tumor biology. Cancer Metatasis Rev. 1993; 12:303-324.
    22. Neufeld G, Gohen T, Gengrinovitch S, et al. Vascular endothelial growth factor (VEGF) and its receptors. FASEB -TI 1999; 13: 9-22.
    23. Ferrara J , Davis-Smyth T. The biology of vascular endothelial growth factor. Endocrine Rev. 1997; 18(1): 4-25.
    24. Kondo K, Hiratsuka S, Subbalakshmi E, et al. Genomic organization of the fit-1 gene encoding for vascular endothelial growth factor (VEGF) receptor-1 suggests an intimate evolutionary relationship between the 7-lg and 5-lg tyrosine kinase receptors. Gene. 1998; 208(2):297-305.
    25. Tanaka K, Yamaguchi S, Sawano A, et al. Characterization of the extra-cellular domain in vascular endothelial growth factor receptor-1 (Fit-1 tyrosine kinase). Jpn J Cancer Res. 1997; 88(9):867-876.
    26. Huang X, Gottstein C, Brekken RA, et al. Expression of soluble VEGF receptor-2 and characterization of its binding by surface plasmon resonance. Biochem Biophys Res Commun, 1998; 252(3):643-648.
    27. Soker S, Takashima S, Miao HQ, et al. Neuropilin-1 is expressed by endothelial and tumor cells as an isoform- specific receptor for vascular endothelial growth factor. Cell. 1998; 92(6):735-745.
    28.李向红,高玉彤,潘彦珞.血管内皮生长因子及其受体在肿瘤研究的概况.中华综合临床医学杂志,2004;6(6):146-149.
    29. Alon T, Hemo I, Itin A, et al. Vascular endothelial growth factor acts as a survival factor for newly formed retinal vessels and has implication for retinopathy of prematurity. Nat Med. 1995; 1:1024-1228.
    30. Shalaby F, Rossant J, Yamaguchi TP, et al. Failure of blood-island formation and vascular genesis in Flk-1 deficient mice. Nature, 1995; 376:62-66.
    31. Anan K, Morisaki T, Katano M, et al. Vascular endothelial growth factor and platelet-derived growth factor are potential angiogenic and metastatic factors in human breast cancer. Surgery. 1996; 119(3): 333-339.
    32. Van Trappen PO, Steele D, et al. Expression of vascular endothelial growth factor VEGF-C and VEGF-D and their receptor VEGFR-3 during different stages of cervical carcinogenesis. J Pathol. 2003; 201(4):544-554.
    33. Khorana AA, Ryan CK, Cox C, et al. Vascular endothelial growth factor, CD68 and epidermal growth factor receptor expression and survival in patients with Stage Ⅱ and Stage Ⅲ colon carcinoma: a role for the host response in prognosis. Cancer. 2003; 97(4): 960-968.
    34. Kabashima A, Maehara Y, Kakeji Y, et al. Over expression of vascular endothelial growth factor C is related to Iymphogenous metastasis in early gastric carcinoma. Oncology. 2001; 60(2): 146-150.
    35. Fontanini G, Faviana P, Lucchi M, et al. A high vascular count and over expression of vascular endothelial growth factor are associated with unfavourable prognosis in operated small cell lung carcinoma. Br J Cancer. 2002, 86(4): 558-563.
    36. Santos L, Costa C, Pereira S, et al. Neovascularisation is a prognostic factor of early recurrence in T1/G2 urothelial bladder tumors. Ann Oncol. 2003; 14(9): 1419-1424.
    37. Lim SC. Role of COX-2, VEGF and cyclin D1 in mammary infiltrating duct carcinoma. Oncol Rep. 2003; 10(5): 1241-1249.
    38. Wong C, Wellman TL, Lounsbury KM. VEGF and HIF-lalpha expression are increased in advanced stages of epithelial ovarian cancer. Gynecol Oncol. 2003; 91(3): 513-517.
    39.邱晓红,张卉,张继红.VEGF及MVD在子宫内膜癌中的表达.中国老年学杂志,2002;22(5):223-224.
    40. Brown LF, Berse B, Jackman, RW, et al. Expression of vascular permeability factor(vascular endothelial growth factor) and its receptors in adenocarcinomas of the gastrointestinal tract. Cancer Res. 1993; 53: 4727-4735.
    41. Itakura J, Ishiwata T, Friess H, et al. Enhanced expression of vascular endothelial growth factor in human pancreatic cancer correlates with local disease progression. Clin Cancer Res. 1997; 3:1309-1316.
    42. Niedergethmann M. High expression of vascular endothelial growth factor predicts early recurrence and poor prognosis after curative resection for ductal adenocarcinoma of the pancreas. Pancreas. 2002; 25(2):122-129.
    43. Karayinanakis AJ, Bolanaki H, Syrigos KN, et al. Serum vascular endothelial growth factor levels in pancreatic cancer patients correlated with advanced and metastatic disease and poor prognosis. Cancer Lett. 2003; 194(1): 119-124.
    44. Ikeda N, Adachi M, Taki T, et al. Prognostic significance of angiogenesis in human pancreatic cancer. British Journal of Cancer. 1999; 79 (9/10): 1553-1563.
    45. von Marschall Z, Cramer T, Hocker M, et al. Denovo expression of vascular endothelial growth factor in human pancreatic cancer: evidence for an autocrine mitogenic loop. Gastroenterology. 2000; 119:1358-1372.
    46. Buchler P, Reber HA, Buchler MW, et al. VEGFR-2 influrnce the prognosis of pancreatic cancer. Ann Surg. 2002; 236:738-749.
    47. Sev Y, Baba H, Fukuda T, et al. High expression of vascular endothelial growth factor is associated with liver metastasis and a poor prognosis for patients with ductal pancreatic adenocarcinoma. Cancer. 2000; 88:2239-2245.
    48. Leo J. Pancreatic cancer cell-derived vascular endothelial growth factor is biologically active in vitro and enhance tumorigenicity in vivo. Int J Cancer. 2001; 92(3):361-369.
    49. Cheryl H, Carmen C, Isaiah J. Angiogenesis and cancer metastasis: antiangiogenc therapy of human pancreatic adenocarcinoma. Int J Clin Oncol. 2001; 6:59-65.
    50. Ahmad T, Eisen T. Kinase inhibition with BAY 43-9006 in Renal Cell carcinoma.Clicinal Cancer Research. 2004; 10(18 Pt 2):6388S-6392S.
    51. Lennernas B, Albertsson P, et al. Chemotherapy and Antiangiogenesis. Acta Oncologica. 2003; 42(4): 294-303.
    52. Nagase T, Kawata S, Nakajima H, et al. Effect of farnesyltransferase overexpression on cell growth and transformation. Int J Cancer. 1999; 80: 126-133.
    53. Sebti SM, Hamilton AD. New approaches to anticancer drug design based on the inhibition of farnesyltransferase. Drug Dis Today. 1998; 3:26.
    54. Lobell RB, Kohl NE. Pre-clinical development of farnesyltransferase inhibitors. Cancer Meta Rev. 1998; 17:203-210.
    55. Liu M, Bryant MS, Chen J, et al. Antitumor activity of SCH66336, an orally bioavailable tricyclic inhibitor of farnesyl protein transferase, in human tumor xenograft models and wap-ras transgenic mice. Cancer Res. 1998; 58:4947-4956.
    56. Verma IM, Stevenson, JK, Schwarz, EM, et al. Rel/NF-κB /IκB family: intimate tales of association and dissociation. Genes Dev. 1995, 9: 2723-2735.
    57. Baldwin A, Jr. The NF-kB and IkB proteins: new discoveries and insights. Ann Rev Immunol. 1996; 14:694-681.
    58. Sen R, Baltimore D. Multiple nuclear factors interact with the immunoglobulin enhancer sequence. Cell.1986; 46:705-716.
    59. Piette J, Piret B. Multiple redox regulation in NF-kB transcription factor activation. Biol Chem. 1997; 378(11):1237-1245.
    60. Flohe L, Brigelius-Flohe R, Saliou C, et al. Packer L redox regulation of NF-kappaB activation. Free Radic Biol Med.1997; 22(6):1115-1126.
    61. May MJ, Ghosh S. Signal transduction through NF-kB. .Immunol Today. 1998; 19(2):80-88.
    62. Bellas RE, FitzGerald MJ, Fausto N, et al. Inhibition of NF-kB activity induces apoptosis in murine hepatocytes. Am J Pathol. 1997; 151:891-896.
    63. Wang CY, Mayo MW, Korneluk RG, et al. NF-κB antiapoptosis: Induction of TRAF1 and TRAF2 and c-IAP1 and c-IAP2 to suppress caspase-8 activation. Science. 1998; 281:1680-1683.
    64. Beg AA and Baltimore D. An essential role for NF-κB in preventing TNF-α-induced cell death.Science. 1996; 274:782-784.
    65. Wang CY, Mayo MW, Jr Baldwin AS. TNF- and cancer therapy-induced apoptosis: potentiation by inhibition of NF-κB. Science. 1996; 274:784-787.
    66. Van Antwerp DT, Martin ST, Kafri T, et al. Suppression of TNF-α-induced apoptosis by NF-κB. Science. 1996; 274:787-789.
    67. Brown K, Gerstberger, S, Carlson, L, et al. Control of IκBa proteolysis by site-specific, signal-induced phosphorylation. Science, 1995, 267: 1485-1488.
    68. Chen ZJ, Hagler J, Palombella VJ, et al. Signal-induced site-specific phosphorylation targets IκBa to the ubiquifin-proteasome pathway. Genes Dev. 1995; 9: 1586-1597.
    69. Didonato JA, Mercurio F, Karin, M. Phosphorylation of IκBa precedes but is not sufficient for its dissociation from NF-κB. Mol. Cell Biol. 1995; 15:1302-1311.
    70. Mercufio F, Zhu H, Murray B, et al. IKK-1, and IKK-2: cytokine-activated IκB kinases essential for NF-κB activation. Science. 1997; 278: 860-866.
    71. Woronicz JD, Gao X, Cao Z, et al. IκB kinase-β: NF-κB activation and complex formation with IκB kinase-α and NIK. Science. 1997; 278: 866-869.
    72. DiDonato JA, Hayakawa M, Rothwarf DM, et al. A cytokine-responsive IκB kinase that activates the transcription factor NF-κB. Nature. 1997; 388: 548-554.
    73. Regnier CH, Song HY, Gao X, et al. Identification and characterization of an IκB kinase. Cell. 1997; 90: 373-383.
    74. Zandi E, Rothwarf DM, Delhase M, et al. The IκB kinase complex (IKK) contains two kinase subunits, IKKa and IKKb, necessary for IκB phosphorylation and NF-κB activation. Cell. 1997; 91: 243-252.
    75. Fujioka S, Sclabas GM, Schmidt C, et al. Inhibition of constitutive NF-kappa B activity by I kappa B alpha M suppresses tumorigenesis. Oncogene. 2003; 22: 1365-1370.
    76. Dong QG, Sclabas GM, Fujioka S, et al. The function of multiple IkappaB: NF-kappaB complexes in the resistance of cancer cells to Taxol-induced apoptosis. Oncogene. 2002; 21: 6510-6519.
    77. Wang WX, Abbruzzese JL, Evans DB, et al. The nuclear factor-KB RelA transcription factor is constitutively activated in human pancreatic adenocarcinoma cells. Clin Cancer Res. 1999; 5:119-127.
    78. Sclabas GM, Uwagawa T, Schmidt C, et al. Nuclear factor kappa B activation is a potential target for preventing pancreatic carcinoma by aspirin. Cancer. 2005; 103: 2485-2490.
    79. Zhang Z and Rigas B. NF-kappaB, inflammation and pancreatic carcinogenesis: NF-kappaB as a chemoprevention target. Int J Oncol. 2006; 29: 185-192.
    80. Molina MA, Sitja-Arnau M, Lemoine MG, et al. Increased cyclooxygenase-2 expression in human pancreatic carcinomas and cell lines: growth inhibition by nonsteroidal anti-inflammatory drugs. Cancer Res. 1999; 59:4356-4362.
    81. Alexander A, Jens V, Maike B, et al. Inhibition of NF-kB sensitizes human pancreatic carcinoma cells to apoptosis induced by etoposide (VP16) or doxorubicin. Oncogene. 2001; 20:859-868.
    82. Banerjee S, Zhang YX, Ali S, et al. Molecular evidence for increased antitumor activity of gemcitabine by genistein in vitro and in vivo using an orthotopic model of pancreatic cancer. Cancer Res. 2005; 65:9064-9072.
    83. Mohammad RM, Banerjee S, Li Y, et al. Cisplatin-induced antitumor activity is potentiated by the soy isoflavone genistein in BxPC-3 pancreatic tumor xenografts. Cancer. 2006; 106:1260-1268.
    84. Yip-Schneider MT, Nakshatri H, Sweeney CJ, et al. Parthenolide and sulindac cooperate to mediate growth suppression and inhibit the nuclear factor-kappa B pathway in pancreatic carcinoma cells. Mol Cancer Ther. 2005; 4: 587-594.
    85. El-Rayes BF, Ali S, Ali IF, et al. Potentiation of the effect of erlotinib by genistein in pancreatic cancer: the role of Akt and nuclear factor-KB. Cancer Res. 2006; 66:10553-10559.
    86. Okami J, Yamamoto H, Fujiwara Y, et al. Overexpression of cyclooxygenase-2 in carcinoma of the pancreas. Clin Cancer Res. 1999; 5:2018-2024.
    87. Eibl G, Reber HA,Wente MN, et al. The selective cyclooxygenase-2 inhibitor nimesulide induces apoptosis in pancreatic cancer cells independent of COX-2. Pancreas. 2003; 26:33-41.
    88. Xiong HQ, Du M, Wolff RA, et al. Pharmacology study of celecoxib in combination with gemcitabine for advanced pancreatic cancer. Proc Annu Meet Am Soc Clin Oncol. 2002; 448. Abstract.
    89. Smith SE, Burris HA, Loehrer PJ, et al. Preliminary report of a phase II trial of gemcitabine combined with celecoxib for advanced pancreatic cancer. Proc Annu Meet Am Soc Clin Oncol. 2003; 1502. Abstract.
    90. Tong WG, Ding XZ, Witt RC, et al. Lipoxygenase inhibitors attenuate growth of human pancreatic cancer xenografts and induce apoptosis through the mitochondrial pathway. Mol Cancer Ther. 2002; 1:929-935.
    91. Motoi F, Sunamura M, Ding L, et al. Effective gene therapy for pancreatic cancer by cytokines mediated by restricted replication competent adenovirus. Hum Gene Ther. 2000; 11: 223-235.

© 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