放疗后残余肝癌侵袭转移潜能的变化、机制及干预的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
原发性肝癌(主要是肝细胞肝癌,hepatocellular carcinoma,HCC)是最常见的恶性肿瘤之一,预后极差,目前是世界第三位癌症杀手,而在中国则高居第二位。手术切除仍是治疗肝癌最有效的方法,但由于肝癌的一些特性,如多灶性,早期血管侵犯,合并肝硬化等,只有不到20%的肝癌患者就诊时适合手术治疗。放射治疗(以下简称放疗)用于肝癌的治疗已有半个世纪,近年随着适形放疗技术的出现及放射生物学观念的更新,放疗在肝癌治疗中的地位有所提高。但临床观察到即使接受根治剂量放射治疗后,仍有60%以上的患者有残存肿瘤存在,尽管放疗后肿瘤会缩小甚至消失,病情得到控制或缓解,但缓解期过后不少病人很快出现复发转移,而且进展迅速,预后较差。这一现象从临床上提出了许多问题,如放疗是否会促进残余肿瘤的播散转移,出于何种机制,如何干预以减少放疗后的复发转移并最终提高放疗疗效,等等。本课题即为此而设计,主要应用我所建立的转移性人肝癌MHCC97L裸鼠模型,模拟临床分割放疗,并主要观察放疗后残余肝癌的侵袭转移潜能的变化。通过实验新发现放疗结束后期残余肝癌的侵袭转移潜能增强,其机制主要是通过上调TMPRSS4表达从而诱导上皮一间质转化(Epithelial-mesenchymal transition,EMT)实现的。EMT是指上皮细胞向间质细胞转化的复杂过程,在此过程中上皮细胞的极性消失,迁移和运动能力增强,EMT是上皮来源的恶性肿瘤发生侵袭转移的重要原因。我们还发现放疗后单独使用TMPRSS4的抑制剂AEBSF或合并使用小剂量IFN-α和阿司匹林(Aspirin)可减少放疗后的肝内播散和转移,从而为进一步提高肝癌放疗疗效提供了线索。
     1.放射线照射可引起肝癌细胞MMP-2、VEGF表达增加及体外侵袭性增强,并与放射线剂量及时间相关
     分别以不同剂量的6mvX线照射MHCC97L细胞,各照射组吸收剂量分别为4Gy和8Gy,对照组(0Gy)除不接受X线照射外,其他处理与照射组相同。照射后24-48h MHCC97L细胞MMP-2和VEGF的表达或活性增强并与照射剂量及时间相关,组间差异具有统计学意义(P<0.05)。8Gy照射后48h其表达及活性最强,此后逐渐降低,照射后96h MMP-2活性低于非照射组。放射线照射对MHCC97L细胞体外侵袭能力的影响:0Gy(对照组)、4Gy和8GyX线照射后,MHCC97L细胞穿过Matrigel胶到达Transwell小室膜背面的细胞数分别为7.2±1.9、13.2±2.7和31.2±7.2,组间差异具有统计学意义(P<0.05)。放射线照射后96h,各组细胞穿膜数无显著差异(P>0.05)。提示放射线照射可使MHCC97L细胞体外侵袭性短期增强。
     2.Aspirin能抑制放射线照射后肝癌细胞MMP-2和VEGF的表达、活性及体外侵袭性的增强
     体外放射线照射前2h细胞培养基内加入Aspirin,使其终浓度为2mM,放疗后继续培养48h。在不抑制细胞增殖的前提下,与对照组相比,Aspirin能在mRNA水平和蛋白水平抑制放疗诱导的VEGF和MMP-2的表达增加和MMP-2活性的增强,以及由此导致的肝癌细胞侵袭性增强。
     3.放疗结束后残余肝癌体内侵袭转移潜能的变化规律
     为了在体内进一步验证放疗结束后残余肝癌侵袭转移能力的变化,我们将具有肺转移潜能的人MHCC97L肝癌组织块种植于BALB/c nu/nu雄性裸鼠肝脏,建立转移性人肝癌裸鼠原位模型。待种植后4周瘤体直径1cm左右时开始放疗。周一至周五每天照射2Gy,共计2周,总放疗剂量20Gy。分别于放疗结束后2天和30天处死放疗组及其对照组裸鼠,切除残余肝癌组织再次原位种植于正常裸鼠肝脏,观察6周处死。放疗结束后2d残余肝癌及其对照组肝癌切除再种植肿瘤大小分别为1.61±0.51cm~3和2.25±0.52cm~3(P<0.05),肺转移率分别为12.5%和66.7%(P<0.05),而放疗结束后30d残癌及其对照组肝癌切除再种植肿瘤大小分别为和2.60±0.61cm~3和2.15±0.71cm~3(P>0.05),肺转移率分别为100%和63.6%(P<0.05)。提示放疗结束后2d残癌生长受到抑制,而放疗结束后30d肿瘤生长速度已恢复到放疗前,甚或过之。放疗结束后30d残癌切除再种植后肿瘤在肝内呈播散性生长,肺转移率增加;而放疗结束后2d残癌切除再种组肿瘤呈外生性生长,无肝内播散,肺转移率降低。提示放疗结束后残余肝癌在体内的侵袭转移能力先减弱后增强。
     4.放疗结束后不同时段残余肝癌组织肿瘤侵袭转移相关基因表达差异的研究
     为了寻找放疗结束后残余肝癌在体内侵袭转移能力变化的相关机制,我们采用Super Array肿瘤侵袭转移相关基因芯片,对对照组、放疗结束后2d和放疗结束后30d残余肝癌组织肿瘤侵袭转移相关基因的表达谱进行分析。以上调或下调超过2倍为检测标准。与对照组相比,放疗结束后2d残余肝癌组织抑癌基因Kiss1和TIMP2表达上调。而放疗结束后30d残余肝癌组织癌基因TMPRSS4表达上调,抑癌基因BAI1表达下调。
     5.实时荧光定量PCR验证基因芯片结果和Western blot分析TMPRSS4在放疗前后不同时段的表达
     我们采用实时荧光定量PCR法验证基因芯片的结果。结果显示放疗结束后2d残癌组织Kiss1(P<0.01)和TIMP2(P<0.05)的mRNA表达显著增强,而放疗结束后30d残癌组织TMPRSS4 mRNA表达显著高于对照组及放疗结束后2d残癌组织(P<0.01),而BAI1mRNA表达变化无统计学意义(P>0.05)。由于放疗结束后30d残余肝癌的侵袭转移能力增强是影响放疗疗效的关键,我们用Western blot进一步验证放疗前后不同时段肝癌组织内TMPRSS4的蛋白含量,同样发现放疗结束后30d残癌组织内TMPRSS4蛋白含量显著高于对照组及放疗结束后2d肝癌组织。提示放疗结束后期TMPRSS4表达增加是其侵袭转移增强的重要原因。
     6.Western blot分析EMT相关分子及COX-2的变化
     根据文献报道,TMPRSS4表达增强能够抑制E-cadherin从而诱导EMT,我们进一步用Western blot检测上皮细胞的标志E-cadherin和其转录抑制因子SIP1,以及间质细胞的标志Vimentin和N-cadherin,发现放疗结束后30d残癌组织SIP1、Vimentin和N-cadherin的表达明显增加,E-cadherin的表达则显著减少。而TMPRSS4抑制剂AEBSF能抑制放疗诱导的SIP1和TMPRSS4的表达而增强E-cadherin的表达,提示放疗结束后30d残癌侵袭转移能力增强是通过上调TMPRSS4的表达,从而抑制E-cadherin和引起EMT实现的。此外,我们选取部分基因芯片未包括的与肿瘤侵袭转移相关的基因进行分析。Western blot结果显示,放疗结束后30d残癌组织COX-2的表达明显增强,由于COX-2的表达与E-cadherin负相关,因此COX-2表达增强也有助于EMT的发生。
     7.联合使用小剂量IFN-α和Aspirin或绿脓杆菌制剂能抑制肝癌在裸鼠体内的生长转移
     据我所前期实验结果,大剂量(1.5×10~7 U/kg·d)长期使用IFN-α能有效抑制转移性人肝癌裸鼠模型肿瘤的生长,并降低手术切除后的复发转移率。但部分患者长期使用大剂量IFN-α会出现白细胞减少等副作用,不能坚持使用影响疗效。鉴于放疗结束后期COX-2表达增加,我们选择联合使用IFN-α和COX-2抑制剂Aspirin观察其对肝癌在裸鼠体内生长转移的抑制作用。结果显示小剂量IFN-α(7.5×10~6 U/kg·d)+Aspirin(15mg/kg·d)可显著抑制肝癌在裸鼠体内的生长转移,其疗效与单独使用大剂量IFN-α(1.5×10~7 U/kg·d)无显著差异。此外,结果还显示绿脓杆菌制剂腹腔注射也可通过促进肿瘤细胞凋亡而显著抑制肿瘤生长转移。为此我们选择小剂量IFN-α(7.5×10~6 U/kg·d)+Aspirin(15mg/kg·d)和绿脓杆菌制剂作为干预放疗结束后期残余肝癌侵袭转移增加的治疗手段。
     8.联合使用小剂量IFN-α和Aspirin或绿脓杆菌制剂可降低放疗结束后期的肝内播散和转移
     将放疗结束后30d残癌组织切除再种植于裸鼠肝脏,分为对照组、小剂量IFN-α+Aspirin治疗组和绿脓杆菌制剂治疗组。与对照组相比,小剂量IFN-α+Aspirin治疗组和绿脓杆菌制剂治疗组肝内播散显著减少,而肺转移被完全抑制。Western blot及免疫组化结果显示小剂量IFN-α+Aspirin治疗组可显著减少放疗后残癌组织内的COX-2的表达,而增强E-cadherin的表达,从而逆转放疗结束后期TMPRSS4表达增强所诱导的EMT及侵袭转移的增加,显示出小剂量IFN-α和Aspirin在减少放疗结束后期残癌侵袭转移方面的临床应用价值。
     结论
     1.放疗结束后残余肝癌体内侵袭转移潜能随时间变化先降低后增强。
     2.放疗结束后残余肝癌TMPRSS4表达增加,进而抑制E-cadherin的表达和诱导EMT,是放疗结束后期残余肝癌侵袭转移能力增强的主要原因。
     3.联合使用小剂量IFN-α和Aspirin或单独使用TMPRSS4抑制剂AEBSF能有效抑制放疗后期COX-2和TMPRSS4表达的上调,增加E-cadherin的表达,抑制残癌的侵袭转移。绿脓杆菌制剂亦可通过促进肝癌细胞的凋亡来抑制放疗后的播散转移。
     4.联合使用小剂量IFN-α和Aspirin可成为临床肝癌患者放疗后减少复发转移有效实用的干预措施。
     应用价值
     1.明确放疗后残余肝癌体内侵袭转移潜能变化规律及其机制,有助于指导放疗后的综合治疗,从而降低放疗后肝癌的复发转移。
     2.联合应用小剂量IFN-α和Aspirin可显著降低放疗结束后期残癌在体内的侵袭转移,为临床提高肝癌放疗的疗效提供了线索。
     3.联合应用小剂量IFN-α和Aspirin可显著抑制肝癌的生长转移,有可能成为临床肝癌治疗的新方法。
     创新点
     1.首次发现放疗结束后残余肝癌体内侵袭转移潜能的变化规律及其机制,提出放疗后残癌TMPRSS4表达增加,进而抑制E-cadherin的表达和诱导EMT,是放疗结束后期残癌侵袭转移能力增强的主要原因。
     2.首次提出联合应用小剂量IFN-α和Aspirin可显著抑制肝癌的生长转移并降低放疗结束后期残癌的侵袭转移。
Hepatocellular carcinoma(HCC) is the third most common cause of death from cancer in the world and the second cancer killer in China.Surgical resection remains the most effective treatment for HCC.Unfortunately,less than 20%of patients with HCC are candidates for resection at the time of diagnosis because of multinodular pattern,vascular invasion and liver cirrhosis.Radiotherapy has been used to treat HCC for half century.Recently,with the advances of biology and technology,such as 3-dimensional conformal radiotherapy(3D-CRT),radiotherapy has become more important for the treatment of HCC.
     However,it is reported that even after radiotherapy with curative dosage,residual cancer can be detected in more than 60%of patients.We did observe that even tumor was "eradicated" after radiotherapy,but several months later some of the tumors relapsed or metastasized quickly with poor prognosis.Questions raise based on this observation:does radiotherapy promote the invasiveness and metastatic potential of residual cancer? What is the molecular background in this process? What can be done for intervention to improve the outcome of radiotherapy? Our present study was designed to clarify these issues by treating nude mice bearing human HCC orthotopic xenografts with fractionated radiotherapy.After treatment the invasiveness and metastatic potential of residual HCC was evaluated and possible molecular background was investigated.We found that radiation indeed promotes the invasion and metastasis of residual cancer through upregulation of TMPRSS4,which can induce epithelial-mesenchymal transition(EMT),a major determinant of metastasis. Administration of TMPRSS4 inhibitor AEBSF or combined use of IFN-αand aspirin can inhibit the invasion and metastasis caused by radiation,and may be a novel approach to improve outcome of radiotherapy for HCC.
     1.Radiation increased the invasiveness of MHCC97L cells in vitro through upregulation of MMP-2 and VEGF
     Alteration in the invasiveness of MHCC97L cells after radiation was examined using the Matrigel invasion assay.The invasiveness of MHCC97L cells were significantly increased after radiation at a time and dose-dependent manner,and reached the maximum at 48h after 8 Gy radiation.Forty eight hours after radiation at different dose(0、4、8Gy),the number of cells that passed through the transwell inserts was 7.2±1.9,13.2±2.7(P<0.05) and 31.2±7.2(P<0.005) respectively,while there was no significant difference 96 hours after radiation.This increase in invasiveness was associated with upregulation of expression or activity of MMP-2 and VEGF.The levels of protein for MMP-2 and VEGF were determined by Western blot analysis and showed a significant increase after exposure to 8 Gy for 48 hours.In order to see if the increase in MMP-2 and VEGF secretion correlates with changes in the expression of the corresponding genes,we tested their mRNA levels by quantitative RT-PCR.There was a dose-dependent increase in MMP-2 and VEGF mRNA expression and the maximal level was observed at dose of 8 Gy;MMP-2 activity was examined by zymographic analysis and was increased in a time and dose dependent manner,attaining a maximum at 48 hours after 8 Gy radiation.
     2.Aspirin inhibited the activity/expression of MMP-2 and VEGF and thereby suppressed radiation augmented invasiveness of MHCC97L cells.
     The invasiveness of MHCC97L cell was significantly enhanced after radiation at doses of 4 Gy and 8 Gy.However,when aspirin(2mM) was added before radiation, the invasion rate dropped near to the level of unirradiated cells.Furthermore,aspirin significantly inhibited the radiation induced increase in VEGF and MMP-2 secretion, as well as the MMP-2 activity.Taken together,these findings suggest that the concomitant use of aspirin during radiation could inhibit the radiation enhanced invasion through suppressing the expression or activity of VEGF and MMP-2.
     3.The invasion and metastasis of residual HCC after radiotherapy in vivo
     In order to further investigate the invasion and metastasis potential of residual HCC in vivo,metastatic animal model of human HCC was established by orthotopic implantation of histologically intact MHCC97L HCC tissue into the liver of nude (nu/nu) mice.Four weeks after implanation,the tumors were irradiated at a dose of 2Gy/d,from Monday to Friday for two weeks,and the total radiation dose was 20Gy. The mice were sacrificed 2 or 30 days afer radiation and the residual cancer were replanted into the liver of other nude mice.These mice were sacrificed 6 weeks after replantation.The replanted tumor volume of 2 days- postradiotherapy group and its control group are 1.61±0.51 and 2.25±0.52cm~3(p<0.05),and the incidence of lung metastasis was 12.5%and 66.7%(P<0.05);while tumor volume of 30 dayspostradiotherapy group and its control group are 2.60±0.61 and 2.15±0.71cm~3(p>0.05),and the incidence of lung metastasis was 100%and 63.6%(P<0.05), respectively.Significant tumor inhibition was only found between control and the 2 days-postradiotherapy group,but not between control and 30 days-postradiotherapy group.The incidence of lung metastasis and inhepatic dissemination were markedly increased in the 30 days-postradiotherapy group.These results suggested that 30 days after radiation the invasiveness and metastatic potential of residual HCC were significantly enhanced.
     4.Changes in the invasion and metastasis related gene expression profile of residual HCC in nude mice after radiotherapy.
     In order to investigate the mechanism involved in this process,invasion and metastasis related cDNA microarray was used to analyse changes of gene expression profile after radiotherapy.The results showed that KISS1 and TIMP2,two factors that inbibit invasion and metastasis,were down-regulated in residual cancer 2 days after radiotherapy,while TMPRSS4,which can promote invasion and metastasis,was significantly up-regulated in residual cancer 30 days after radiotherapy.Furthermore, BAI1 was down-regulated in residual cancer 30 days after radiotherapy.
     5.Validation of cDNA microarray results by quantitative RT-PCR and westen blot
     In order to verify the above-mentioned changes in KISS1,TIMP2,TMPRSS4 and BAIl from the cDNA microarray analyses,we tested the effect of radiation on their mRNA levels by quantitative RT-PCR.Compared with control group,the KISS 1(P<0.01) and TIMP2(P<0.05) mRNA expression in residual cancer 2 days postradiotherapy were significantly increased.The TMPRSS4 mRNA expression of residual cancer 30 days postradiotherapy was much higher than that of control group and residual cancer 2 days after radiotherapy(P<0.01),but there was no significant difference in BAI mRNA expression(P>0.05) between these groups.We further analysed the protein level of TMPRSS4 in these 3 groups and western blot result showed that TMPRSS4 protein was also significantly enhanced in residual cancer 30 days postradiotherapy.These findings indicate that TMPRSS4 is a key oncogene that related to radiation induced invasiveness and metastasis.
     6.Western blot analysis of EMT related molecules and COX-2
     According to literature,up-regulation of TMPRSS4 can inhibit the expression of E-cadherin and facilitate EMT,we further detected epithelial marker of E-cadherin and mesenchymal marker of Vimentain and N-cadherin.SIP1/ZEB2,an E-cadherin transcriptional repressor,was also detected by western blot.The results showed that expression of SIP1,Vimentain and N-cadherin was significantly increased while expression of E-cadherin was inhibited.Administration of TMPRSS4 inhibitor AEBSF inhibited the expression of TMPRSS4 and SIP1 and up-regulated the expression of E-cadherin,together with reduced invasion and metastasis in vivo after radiation.It is concluded that radiation promoted the invasiveness and metastasis through TMPRSS4 induced EMT.We also analysed some molecules that were not included in the cDNA microarray by western blot,and found that COX-2 expression was significantly increased in the residual cancer 30 days postradiotherapy.Since there is an inverse relationship between COX-2 and E-cadherin expression,enhanced expression of COX-2 also contributes to EMT.
     7.Combined administration of IFN-αand aspirin or pseudomonas aeruginosa vaccine(PA) inhibits the growth and metastasis of HCC in vivo
     We previously reported that High-dose(1.5×10~7 U/kg·d) and long-term therapy with IFN-αinhibits tumor growth and recurrence in nude mice bearing human HCC xenografts.However,in clinical practice some patients can not stand high-dose therapy with IFN-αbecause of the side effects.Since COX-2 expression is increased after radiation,we choose combined administration of low-dose IFN-α(7.5×10~6 U/kg·d) and aspirin(15mg/kg·d) to inhibit the growth and metastasis of HCC in vivo, which was as effective as high-dose IFN-α(1.5×10~7 U/kg·d).Furthermore, administration of PA intraperitoneally also significantly inhibit the growth and metastasis of HCC through induction of apoptosis.Thereby we choose combined administration of low-dose IFN-αand aspirin or PA to inhibit the radiation induced invasion and metastasis.
     8.Combined administration of low-dose IFN-αand aspirin or PA inhibits the dissemination and metastasis of residual cancer
     Thirty days afer radiation,the residual cancer were resected and replanted into the liver of other nude mice.The mice were divided into control group,low-dose IFN-αplus aspirin treatment group and PA treatment group.Six weeks after that,the mice were sacrificed.Low-dose IFN-αplus aspirin or PA treatment significantly decreased the dissemination in liver and completely inhibited the lung metastasis.These findings may provide novel approach to enhance the efficacy of radiotherapy for HCC.
     Conclusions
     1.The invasion and metastasis of HCC in vivo was inhibited in early phase and then enhanced in the late phase after radiation.
     2.Radiation enhanced the invasion and metastasis of residual cancer through upregulation of TMPRSS4,which inhibited the expression of E-cadherin and induced EMT.
     3.Combined administration of low-dose IFN-αand aspirin or TMPRSS4 inhibitor AEBSF inhibited the dissemination and metastasis of residual cancer through suppression of COX-2 and TMPRSS4 and upregulation of E-cadherin.PA can inhibit the dissemination and metastasis through inducing the apoptosis of HCC.
     4.Combined administration of low-dose IFN-αand Aspirin could be a potential therapeutic approach to inhibit radiation induced invasion and metastasis and improve the efficacy of radiotherapy for HCC.
     The potential application of this work
     1.Clarifying the molecular basis of radiation induced invasion and metastasis of residual HCC helps to instruct the combined therapy postradiotherapy.
     2.Combined administration of low-dose IFN-αand Aspirin significantly inhibited the dissemination and metastasis of residual HCC postradiotherapy,thereby provides a new clue to improve the outcome of radiotherapy.
     3.Combined administration of low-dose IFN-αand Aspirin significantly inhibited the growth and metastasis of HCC in vivo,thus provides a new approach for treatment of HCC.
     Originalities of this work
     1.For the first time to clarify the alteration and molecular basis of invasion and metastasis potential of residual HCC after radiation in vivo,and point out that upregulation of TMPRSS4 and induced EMT might play a major role.
     2.For the first time to point out that combined administration of low-dose IFN-αand aspirin significantly inhibited the growth and metastasis of HCC in vivo, and also suppressed the dissemination and metastasis of residual cancer postradiotherapy.
引文
1 Parkin DM, Bray F, Ferlay J, et al. Global cancer statistics, 2002. CA Cancer J Clin, 2005,55:74-108.
    2 Parkin DM. Global cancer statistics in the year 2000. Lancet Oncol, 2001,2:533-543.
    3 El-Serag HB. Hepatocellular carcinoma: an epidemiologic view. J Clin Gastroenterol, 2002,35:S72-78.
    4 Poon RTP, Fan ST, Tsang FHF, et al. Locoregional therapy for hepatocellular carcinoma: A critical review from the surgeon's perspective. Ann Surg, 2002,235:466-486.
    5 Seong J, Park HC, Han KH, et al. Clinical results and prognostic factors in radiotherapy for unresectable hepatocellular carcinoma: a retrospective study of 158 patients. Int J Radiat Oncol Biol Phys, 2003,55:329-336.
    6 Park W, Lim DH, Paik SW, et al. Local radiotherapy for patients with unresectable hepatocellular carcinoma. Int J Radiat Oncol Biol Phys, 2005,61:1143-1150.
    7 Zeng ZC,Fan J,Tang ZY, et al. A comparison of treatment combinations with and without radiotherapy for hepatocellular carcinoma with portal vein and/or inferior vena cava tumor thrombus. Int J Radiat Oncol Biol Phys, 2005,61:432-443.
    8 Zeng ZC, Jiang GL, Wang GM, et al. DNA-PKcs subunits in radiosensitization by hyperthermia on hepatocellular carcinoma hepG2 cell line. World J Gastroenterol, 2002,8:797-803.
    9 Moran JM,Elshaikh MA,Lawrence TS. Radiotherapy: what can be achieved by technical improvements in dose delivery?. Lancet Onco, 2005,6:51-58.
    10 Llovet JM, Bruix J. Systematic review of randomized trials for unresectable hepatocellular carcinoma: Chemoembolization improves survival. Hepatology, 2003,37:429-442.
    11 Kaplan HS, Murphy ED. The effect of local roentgen irradiation on the biological behavior of a transplantable mouse carcinoma. Increased frequency of pulmonary metastasis. J Natl Cancer Inst,1949, 9: 407-413.
    12 Baumann M, Becker S, Kruger HJ, et al. Flow cytometric determination of the time of metastasis during fractionated radiation therapy of the rat rhabdomyosarcoma R1H. Int J Radiat Biol, 1990,58:361-369.
    13 de Ruiter J, Cramer SJ, Lelieveld P, et al. Comparison of metastatic disease after local tumour treatment with radiotherapy or surgery in various tumour models. Eur J Cancer Clin Oncol, 1982,18:281 -289.
    14 Heisel MA, Laug WE, Stowe SM, et al. Effects of X-irradiation on artificial blood vessel wall degradation by invasive tumor cells. Cancer Res, 1984,44:2441-2445.
    15 Bonfil RD, Bustuoabad OD. Ruggiero RA, et al. Tumor necrosis can facilitate the appearance of metastases. Clin Exp Metastasis, 1988,6:121-129.
    16 Park JS, Qiao L, Su ZZ,et al. Ionizing radiation modulates vascular endothelial growth factor (VEGF) expression through multiple mitogen activated protein kinase dependent pathways. Oncogene, 2001,20:3266-3280.
    17 Brooks P C,Broek D,Roth J,et al. Differential effects of ionizing radiation on angiogenesis and matrix metalloproteinases (MMPs) activity in vitro and in vivo. Int J Radiat Oncol Biol Phys, 2001,51:156.
    18 Paley PJ, Goff BA, Minudri R, et al. The prognostic significance of radiation dose and residual tumor in the treatment of barrel-shaped endophytic cervical carcinoma. Gynecol Oncol, 2000,76:373-379.
    19 Babaian RJ, Kojima M, Saitoh M, et al. Detection of residual prostate cancer after external radiotherapy. Cancer, 1995,75:2153-2158.
    20 Kaliski A,Maggiorella L,Mathe D,et al. Inhibition of radiation-induced invasiveness and endothelial sustain by an MMP-inhibitor through interaction with radiation-enhanced MMP-system and VEGF release. Int J Radiat Oncol Biol Phys, 2004,60:S372.
    21 Wild-Bode C, Weller M, Rimner A,et al. Sublethal irradiation promotes migration and invasiveness of glioma cells: implications for radiotherapy of human glioblastoma. Cancer Res, 2001,61:2744-2750.
    22 Speake WJ, Dean RA, Kumar A,et al. Radiation induced MMP expression from rectal cancer is short lived but contributes to in vitro invasion. Eur J Surg Oncol, 2005,31:869-874.
    23 Qian LW, Mizumoto K, Urashima T,et al. Radiation-induced increase in invasive potential of human pancreatic cancer cells and its blockade by a matrix metalloproteinase inhibitor, CGS27023. Clin Cancer Res, 2002,8:1223-1227.
    24 Cheng JC, Chou CH, Kuo ML,et al. Radiation-enhanced hepatocellular carcinoma cell invasion with MMP-9 expression through PI3K/Akt/NF-kappaB signal transduction pathway. Oncogene, 2006,25:7009-7018.
    25 Citrin D, Menard C,Camphausen K. Combining radiotherapy and angiogenesis inhibitors: clinical trial design. Int J Radiat Oncol Biol Phys,64:15-25.
    26 Li Y,Tang ZY,Ye SL, et al. Establishment of a hepatocellular carcinoma cell line with unique metastatic characteristics through in vivo selection and screening for metastasis-related genes through cDNA microarray. J Cancer Res Clin Oncol, 2003,129:43-51.
    27 Weissmann G. Aspirin. Sci Am, 1991,264:84-90.
    28 Onoda JM, Piechocki MP, Honn KV. Radiation-induced increase in expression of the alpha IIb beta 3 integrin in melanoma cells: effects on metastatic potential. Radiat Res, 1992,130:281-288.
    29 Gliemroth J, Feyerabend T, Gerlach C, et al. Proliferation, migration, and invasion of human glioma cells exposed to fractionated radiotherapy in vitro. Neurosurg Rev, 2003,3:198-205.
    30 Bauman GS, Fisher BJ, McDonald W, et al. Effects of radiation on a three-dimensional model of malignant glioma invasion. Int J Devl Neuroscience, 1999:643-651.
    31 Hovinga KE, Stalpers LJ, van Bree C,et al. Radiation-enhanced vascular endothelial growth factor (VEGF) secretion in glioblastoma multiforme cell lines-a clue to radioresistance? J Neuro Oncol, 2005,74:99-103.
    32 Steiner HH, Karcher S, Mueller MM,et al. Autocrine pathways of the vascular endothelial growth factor (VEGF) in glioblastoma multiforme: clinical relevance of radiation-induced increase of VEGF levels. J Neuro Oncol, 2004,66:129-138.
    33 Zhao W, Goswami PC, Robbins ME. Radiation-induced up-regulation of MMP2 involves increased mRNA stability, redox modulation, and MAPK activation. Radiat Res, 2004,161:418-429.
    34 Pai R,Szabo IL,Soreghan BA,et al. PGE2 Stimulates VEGF Expression in Endothelial Cells via ERK2/JNK1 Signaling Pathways. Biochem Biophys Res Commun, 2001,286:923-928.
    35 Vartiainen N,Goldsteins G, Keksa-Goldsteine V, et al. Aspirin inhibits p44/42 mitogen-activated protein kinase and is protective against hypoxia/reoxygenation neuronal damage. Stroke, 2003,34:752-757.
    36 Murono S, Yoshizaki, Sato H. Aspirin inhibits tumor cell invasiveness induced by Epstein-Barr virus latent membrane protein 1 through suppression of matrix metalloproteinase-9 expression. Cancer Res, 2000,60:2555-2561.
    37 Gao Z, Zuberi A,Quon MJ,et al. Aspirin inhibits serine phosphorylation of insulin receptor substrate 1 in tumor necrosis factor-treated cells through targeting multiple serine kinases. J Biol Chem, 2003,278:24944-24950.
    38 Jiang MC, Liao CF, Lee PH. Aspirin inhibits matrix metalloproteinase-2 activity, increases E-cadherin production, and inhibits in vitro invasion of tumor cells. Biochem Biophys Res Commun, 2001,282:671-677.
    39 Kaliski A,Lassau N,Maggiorella L,et al. Antiangiogenic effect and tumor growth control achieved by an MMP-inhibitor combined to radiation in vivo, targeting radio-induced MMP-2 enhancement and VEGF modulation. Int J Radiat Oncol Biol Phys, 2004,60:S368-369.
    40 Yanamandra N, Kondraganti S, Srinivasula SM, et al. Activation of caspase-9 with irradiation inhibits invasion and angiogenesis in SNB19 human glioma cells. Oncogene, 2004,23:2339-2346.
    41 Jung H,Lee KP,Park SJ,et al. TMPRSS4 promotes invasion, migration and metastasis of human tumor cells by facilitating an epithelial-mesenchymal transition. Oncogene, 2007,Epub ahead of print.
    42 Dohadwala M, Yang SC, Luo J, et al. Cyclooxygenase-2-dependent regulation of E-cadherin: prostaglandin E(2) induces transcriptional repressors ZEBl and snail in non-small cell lung cancer. Cancer Res, 2006,66:5338-5345.
    43 Rao DS, Gui D, Koski ME, et al. An inverse relation between COX-2 and E-cadherin expression correlates with aggressive histologic features in prostate cancer. Appl Immunohistochem Mol Morphol, 2006,14:375-383.
    44 Okamoto A, Shirakawa T, Bito T, et al. Etodolac, a selective cyclooxygenase-2 inhibitor, induces upregulation of E-cadherin and has antitumor effect on human bladder cancer cells in vitro and in vivo. Urology, 2008,71:156-160.
    45 Camphausen K, Moses MA, Beecken WD, et al. Radiation therapy to a primary tumor accelerates metastatic growth in mice. Cancer Res, 2001,61:2207-2211.
    46 Rofstad EK, Mathiesen B, Galappathi K. Increased metastatic dissemination in human melanoma xenografts after subcurative radiation treatment: radiation-induced increase in fraction of hypoxic cells and hypoxia-induced up-regulation of urokinase-type plasminogen activator receptor. Cancer Res, 2004,64:13-18.
    47 Iwakawa M, Kaneko M, Ikebulkuro K. Enhanced metastasis after local therapy in a murine model using C-1300 neuroblastoma. J Pediatr Surg, 1999,34:1645-1646.
    48 von Essen CF. Radiation enhancement of metastasis: a review. Clin Exp Metastasis, 1991,9:77-104.
    49 Ohuchida K, Mizumoto K, Murakami M, et al. Radiation to stromal fibroblasts increases invasiveness of pancreatic cancer cells through tumor-stromal interactions. Cancer Res, 2004,64:3215-3222.
    50 Jadhav U, Mohanam S. Response of neuroblastoma cells to ionizing radiation: modulation of in vitro invasiveness and angiogenesis of human microvascular endothelial cells. Int J Oncol, 2006,29:1525-1531.
    51 Wild-Bode C, Weller M, Rimner A, et al. Sublethal irradiation promotes migration and invasiveness of glioma cells: implications for radiotherapy of human glioblastoma. Cancer Res, 2001,61:2744-2750.
    52 Park CM, Park MJ, Kwak HJ, et al. Ionizing Radiation Enhances Matrix Metalloproteinase-2 Secretion and Invasion of Glioma Cells through Src/Epidermal Growth Factor Receptor-Mediated p38/Akt and Phosphatidylinositol 3-Kinase/Akt Signaling Pathways. Cancer Res, 2006,66:8511-8519.
    53 Speake WJ, Dean RA, Kumar A, et al. Radiation induced MMP expression from rectal cancer is short lived but contributes to in vitro invasion. Eur J Surg Oncol, 2005,31:869-874.
    54 Wallrapp C, Hahnel S, Muller-Pillasch F, et al. A novel transmembrane serine protease (TMPRSS3) overexpressed in pancreatic cancer. Cancer Res, 2000,60:2602-2606.
    55 Kebebew E, Peng M, Reiff E, et al. ECM1 and TMPRSS4 are diagnostic markers of malignant thyroid neoplasms and improve the accuracy of fine needle aspiration biopsy. Ann Surg, 2005,242:353-61; discussion 361-363.
    56 Sawasaki T, Shigemasa K, Gu L, et al. The transmembrane protease serine (TMPRSS3/TADG-12) D variant: a potential candidate for diagnosis and therapeutic intervention in ovarian cancer. Tumour Biol, 2004,25:141-148.
    57 Wang H, Quah SY, Dong JM, et al. PRL-3 down-regulates PTEN expression and signals through PI3K to promote epithelial-mesenchymal transition. Cancer Res, 2007,67:2922-2926.
    58 Funasaka T, Hu H, Yanagawa T, et al. Down-regulation of phosphoglucose isomerase/autocrine motility factor results in mesenchymal-to-epithelial transition of human lung fibrosarcoma cells. Cancer Res, 2007,67:4236-4243.
    59 Huang HC, Hu CH, Tang MC, et al. Thymosin beta4 triggers an epithelial-mesenchymal transition in colorectal carcinoma by upregulating integrin-linked kinase. Oncogene, 2007,26:2781-2790.
    60 Tsukamoto H, Shibata K, Kajiyama H, et al. Irradiation-induced epithelial-mesenchymal transition (EMT) related to invasive potential in endometrial carcinoma cells. Gynecol Oncol, 2007,107:500-504.
    61 Andarawewa KL, Erickson AC, Chou WS, et al. Ionizing radiation predisposes nonmalignant human mammary epithelial cells to undergo transforming growth factor beta induced epithelial to mesenchymal transition. Cancer Res, 2007,67:8662-8670.
    62 Shahedi K, Lindstrom S, Zheng SL, et al. Genetic variation in the COX-2 gene and the association with prostate cancer risk. Int J Cancer, 2006,119:668-672.
    63 Singh B, Berry JA, Shoher A, et al. COX-2 involvement in breast cancer metastasis to bone. Oncogene, 2007,26:3789-796.
    64 Mascaux C, Martin B, Paesmans M, et al. Has Cox-2 a prognostic role in non-small-cell lung cancer? A systematic review of the literature with meta-analysis of the survival results. Br J Cancer, 2006,95:139-145.
    65 Potter JD, Ulrich CM. COX-2 and gastric cancer: More on inflammation and neoplasia. Gastroenterology, 2006,130:2198-2200.
    66 Buchanan FG, DuBois RN. Connecting COX-2 and Wnt in cancer. Cancer Cell, 2006,9:6-8.
    67 Siemann DW,Warrington JKH,Horsman MR. Targeting tumor blood vessels: an adjuvant strategy for radiation therapy. Radiother Oncol, 2000,57:5-12.
    68 Hawkins MA, Dawson LA. Radiation therapy for hepatocellular carcinoma: from palliation to cure. Cancer, 2006,106:1653-1663.
    69 Siemann DW, Warrington KH, Horsman MR. Targeting tumor blood vessels: an adjuvant strategy for radiation therapy. Radiother Oncol, 2000,57:5-12.
    70 Dische S, Anderson PJ, Sealy R, et al. Carcinoma of the cervix-anaemia, radiotherapy and hyperbaric oxygen. Br J Radiol, 1983,56:251-255.
    71 Overgaard J, Horsman MR. Modification of Hypoxia-Induced Radioresistance in Tumors by the Use of Oxygen and Sensitizers. Semin Radiat Oncol, 1996,6:10-21.
    72 Vaupel P, Kallinowski F, Okunieff P. Blood flow, oxygen and nutrient supply, and metabolic microenvironment of human tumors: a review. Cancer Res, 1989,49:6449-6465.
    73 Jain RK. Normalizing tumor vasculature with anti-angiogenic therapy: a new paradigm for combination therapy. Nat Med, 2001,7:987-989.
    74 Teicher BA, Holden SA, Ara G, et al. Influence of an anti-angiogenic treatment on 9L gliosarcoma: oxygenation and response to cytotoxic therapy. Int J Cancer, 1995,61:732-737.
    75 Lee CG, Heijn M, di Tomaso E, et al. Anti-Vascular endothelial growth factor treatment augments tumor radiation response under normoxic or hypoxic conditions. Cancer Res, 2000,60:5565-5570.
    76 Griffin RJ, Williams BW, Wild R, et al. Simultaneous inhibition of the receptor kinase activity of vascular endothelial, fibroblast, and platelet-derived growth factors suppresses tumor growth and enhances tumor radiation response. Cancer Res, 2002,62:1702-1706.
    77 Teicher BA, Holden SA, Ara G, et al. Potentiation of cytotoxic cancer therapies by TNP-470 alone and with other anti-angiogenic agents. Int J Cancer, 1994,57:920-925.
    78 Wachsberger P, Burd R, Dicker AP. Tumor response to ionizing radiation combined with antiangiogenesis or vascular targeting agents: exploring mechanisms of interaction. Clin Cancer Res, 2003,9:1957-1971.
    79 Benjamin LE, Golijanin D, Itin A, et al. Selective ablation of immature blood vessels in established human tumors follows vascular endothelial growth factor withdrawal. J Clin Invest, 1999,103:159-165.
    80 Gorski DH, Beckett MA, Jaskowiak NT,et al. Blockade of the vascular endothelial growth factor stress response increases the antitumor effects of ionising radiation. Cancer Res, 1999,59:3374-3378.
    81 Dicker AP. COX-2 inhibitors and cancer therapeutics: potential roles for inhibitors of COX-2 in combination with cytotoxic therapy: reports from a symposium held in conjunction with the Radiation Therapy Oncology Group June 2001 Meeting. Am J Clin Oncol, 2003,26:S46-47.
    82 Jones DA, Carlton DP, McIntyre TM, et al. Molecular cloning of human prostaglandin endoperoxide synthase type II and demonstration of expression in response to cytokines. J Biol Chem, 1993,268:9049-9054.
    83 Williams CS, DuBois RN. Prostaglandin endoperoxide synthase: why two isoforms? Am J Physiol, 1996,270:G393-400.
    84 Tsuji S, Tsujii M, Kawano S, et al. Cyclooxygenase-2 upregulation as a perigenetic change in carcinogenesis. J Exp Clin Cancer Res, 2001,20:117-129.
    85 Tsujii M, Kawano S, Tsuji S, et al. Cyclooxygenase regulates angiogenesis induced by colon cancer cells. Cell, 1998,93:705-716.
    86 Kishi K, Petersen S, Petersen C, et al. Preferential enhancement of tumor radioresponse by a cyclooxygenase-2 inhibitor. Cancer Res, 2000,60:1326-1331.
    87 Petersen C, Petersen S, Milas L, et al. Enhancement of intrinsic tumor cell radiosensitivity induced by a selective cyclooxygenase-2 inhibitor. Clin Cancer Res, 2000,6:2513-2520.
    88 Pyo H, Choy H, Amorino GP, et al. A selective cyclooxygenase-2 inhibitor, NS-398, enhances the effect of radiation in vitro and in vivo preferentially on the cells that express cyclooxygenase-2. Clin Cancer Res, 2001,7:2998-3005.
    89 Palayoor ST, Bump EA, Calderwood SK, et al. Combined antitumor effect of radiation and ibuprofen in human prostate carcinoma cells. Clin Cancer Res, 1998,4:763-771.
    90 Milas L, Furuta Y, Hunter N, et al. Dependence of indomethacin-induced potentiation of murine tumor radioresponse on tumor host immunocompetence. Cancer Res, 1990,50:4473-4477.
    91 Furuta Y, Hunter N, Barkley T Jr, et al. Increase in radioresponse of murine tumors by treatment with indomethacin. Cancer Res, 1988,48:3008-3013.
    92 Teicher BA, Bump EA, Palayoor ST, et al. Signal transduction inhibitors as modifiers of radiation therapy in human prostate carcinoma xenografts. Radiat Oncol Invest 1996;4:221-230.
    93 Keskek M, Gocmen E, Kilic M, et al. Increased expression of cyclooxygenase-2 (COX-2) in radiation-induced small bowel injury in rats. J Surg Res, 2006,135:76-84.
    94 Sonis ST, O'Donnell KE, Popat R, et al. The relationship between mucosal cyclooxygenase-2 (COX-2) expression and experimental radiation-induced mucositis. Oral Oncol, 2004,40:170-176.
    95 Amirghahari N, Harrison L, Smith M, et al. NS 398 radiosensitizes an HNSCC cell line by possibly inhibiting radiation-induced expression of COX-2. Int J Radiat Oncol Biol Phys, 2003,57:1405-1412.
    96 Steinauer KK, Gibbs I, Ning S, et al. Radiation induces upregulation of cyclooxygenase-2 (COX-2) protein in PC-3 cells. Int J Radiat Oncol Biol Phys, 2000,48:325-328.
    97 Brouty-Boye D, Zetter BR. Inhibition of cell motility by interferon. Science, 1980,208:516-518.
    98 Sidky YA, Borden EC. Inhibition of angiogenesis by interferons: effects on tumor-and lymphocyte-induced vascular responses. Cancer Res, 1987,47:5155-5161.
    99 Pestka S, Langer JA. Zoon KC, et al. Interferons and their actions. Annu Rev Biochem, 1987,56:727-777.
    100 Lengyel P. Biochemistry of interferons and their actions. Annu Rev Biochem, 1982,51:251-282.
    101 Patt YZ, Hassan MM, Lozano RD, et al. Phase II trial of systemic continuous fluorouracil and subcutaneous recombinant interferon Alfa-2b for treatment of hepatocellular carcinoma. J Clin Oncol, 2003,21:421-427.
    102 Obi S, Yoshida H, Toune R, et al. Combination therapy of intraarterial 5-fluorouracil and systemic interferon-alpha for advanced hepatocellular carcinoma with portal venous invasion. Cancer, 2006,106:1990-1997.
    103 von Marschall Z, Scholz A, Cramer T, et al. Effects of interferon alpha on vascular endothelial growth factor gene transcription and tumor angiogenesis. J Natl Cancer Inst, 2003,95:437-448.
    104 Huang SF, Kim SJ, Lee AT, et al. Inhibition of growth and metastasis of orthotopic human prostate cancer in athymic mice by combination therapy with Pegylated interferon-alpha-2b and docetaxel. Cancer Res, 2002,62:5720-5726.
    105 Hancock BW, Wheatley K, Harris S, et al. Adjuvant interferon in high-risk melanoma: the AIM HIGH Study-United Kingdom Coordinating Committee on Cancer Research randomized study of adjuvant low-dose extended-duration interferon Alfa-2a in high-risk resected malignant melanoma. J Clin Oncol, 2004,22:53-61.
    106 Wang L, Tang ZY, Qin LX, et al. High-dose and long-term therapy with interferon-alfa inhibits tumor growth and recurrence in nude mice bearing human hepatocellular carcinoma xenografts with high metastatic potential. Hepatology, 2000,32:43-48.
    107 Wang L, Wu WZ, Sun HC, et al. Mechanism of interferon alpha on inhibition of metastasis and angiogenesis of hepatocellular carcinoma after curative resection in nude mice. J Gastrointest Surg, 2003,7:587-594.
    108 Jiang MC,Liao CF,Lee PH. Aspirin inhibits matrix metalloproteinase-2 activity, increases E-cadherin production, and inhibits in vitro invasion of tumor cells. Biochem Biophys Res Commun, 2001,282:671-677.
    109 Abiru S, Nakao K, Ichikawa T, et al. Aspirin and NS-398 inhibit hepatocyte growth factor-induced invasiveness of human hepatoma cells. Hepatology, 2002,35:1117-1124.
    110 Nadar S, Blann AD, Lip GY. Effects of aspirin on intra-platelet vascular endothelial growth factor, angiopoietin-1, and p-selectin levels in hypertensive patients. Am J Hypertens, 2006,19:970-977.
    111 Escribano M, Molero L, Lopez-Farre A, et al. Aspirin inhibits endothelial nitric oxide synthase (eNOS) and Flk-1 (vascular endothelial growth factor receptor-2) prior to rat colon tumour development. Clin Sci (Lond), 2004,106:83-91.
    112 Gerrah R, Fogel M, Gilon D. Aspirin decreases vascular endothelial growth factor release during myocardial ischemia. Int J Cardiol, 2004,94:25-29.
    113 Drake JG, Becker JL. Aspirin-induced inhibition of ovarian tumor cell growth. Obstet Gynecol, 2002,100:677-682.
    114 Stark LA, Reid K, Sansom OJ, et al. Aspirin activates the NF-kappaB signalling pathway and induces apoptosis in intestinal neoplasia in two in vivo models of human colorectal cancer. Carcinogenesis, 2007,28:968-976.
    115 Din FV, Stark LA, Dunlop MG. Aspirin-induced nuclear translocation of NFkappaB and apoptosis in colorectal cancer is independent of p53 status and DNA mismatch repair proficiency. Br J Cancer, 2005,92:1137-1143.
    116 Flossmann E, Rothwell PM. Effect of aspirin on long-term risk of colorectal cancer: consistent evidence from randomised and observational studies. Lancet, 2007,369:1603-1613.
    117 Chan AT, Ogino S, Fuchs CS. Aspirin and the risk of colorectal cancer in relation to the expression of COX-2. N Engl J Med, 2007,356:2131-2142.
    118 Jayaprakash V, Menezes RJ, Javle MM, et al. Regular aspirin use and esophageal cancer risk. Int J Cancer, 2006,119:202-207.
    119 McCaffrey P. Aspirin use reduces skin-cancer risk. Lancet Oncol, 2006,7:16.
    120 Jacobs EJ, Rodriguez C, Mondul AM, et al. A large cohort study of aspirin and other nonsteroidal anti-inflammatory drugs and prostate cancer incidence. J Natl Cancer Inst, 2005,97:975-980.
    121 Schernhammer ES, Kang JH, Chan AT, et al. A prospective study of aspirin use and the risk of pancreatic cancer in women. J Natl Cancer Inst, 2004,96:22-28.
    122 Kiger N, Schulz J, Khalil A, et al. Delayed tumor growth caused by a combined treatment with BCG and Pseudomonas aeruginosa. Cancer Immunol Immunother, 1978,5:207-210.
    123 Mathe G,Florentin I,Bruley-Rosset M,et al.Heat-killed Pseudomonas aeruginosa as a systemic adjuvant in cancer immunotherapy.Biomedicine,1977,27:368-373.
    124 殷积斌.PA菌毛株菌苗膀胱灌注预防膀胱癌术后复发的研究.中华泌尿外科杂志,2001,10:597.
    125 Jia L,Wang C,Kong H,et al.Effect of PA-MSHA vaccine on plasma phospholipids metabolic profiling and the ratio of Th2/Thl cells within immune organ of mouse IgA nephropathy.J Pharm Biomed Anal,2007,43:646-654.
    126 Kaliski A,Deutsch E,Maggiorella L,et al.Ionizing radiation enhance tumor invasiveness which can be overcome by concurrent treatment which a MMP inhibitor.Radiother Oncol,2004,73:S325-326.
    127 Gliemroth J,Feyerabend T,Gerlach C,et al.Proliferation,migration,and invasion of human glioma cells exposed to fractionated radiotherapy in vitro.Neurosurg Rev,2003,26:198-205.
    128 Ferrara N,Henzel WJ.Pituitary follicular cells secrete a novel heparin-binding growth factor specific for vascular endothelial cells.Biochem Biophys Res Commun,1989,161:851-858.
    129 Donadio AC,Durand S,Remedi MM,et al.Evaluation of stromal metalloproteinases and vascular endothelial growth factors in a spontaneous metastasis model.Exp Mol Pathol,2005,79:259-264.
    130 White FC,Benehacene A,Scheele JS,et al.VEGF mRNA is stabilized by ras and tyrosine kinase oncogenes,as well as by UV radiation-evidence for divergent stabilization pathways.Growth Factors,1997,14:199-212.
    131 Miura Hideaki,Miyazaki Tom,Kuroda Masahiko,et al0 Increased expression of vascular endothelial growth factor in human hepatocellular carcinoma.J Hepatol,1997,27:854-861.
    132 Marcus S,Axel H,Ralf K,et al.Disruption of hepatocellular tight junctions by vascular endothelial growth factor(VEGF):a novel mechanism for tumor invasion.J Hepatol,2004,41:274-283.
    133 Melnyk O,Zimmerman M,Kim KJ,et al.Neutralizing antivascular endothelial growth factor antibody inhibits further growth of established prostate cancer and metastasis in a pre-clinical model.J Urol,1999,161:960-963.
    134 Wergin MC,Ballmer-Hofer K,Roos M,et al.Preliminary study of plasma vascular endothelial growth factor(VEGF) during low-and high-dose radiation therapy of dogs with spontaneous tumors. Vet Radiol Ultrasound, 2004,45:247-254.
    135 Trompezinski S, Pernet 1, Schmitt D, et al. UV radiation and prostaglandin E2 up-regulate vascular endothelial growth factor (VEGF) in cultured human fibroblasts. Inflamm Res, 2001,50:422-427.
    136 Brieger J, Schroeder P, Gosepath J,et al. VEGF-subtype specific protection of SCC and HUVECs from radiation induced cell death. Int J Mol Med, 2005,15:145-151.
    137 Choi SY, Kim MJ, Kang CM,et al. Activation of Bak and Bax through c-abl-protein kinase Cdelta-p38 MAPK signaling in response to ionizing radiation in human non-small cell lung cancer cells. J Biol Chem, 2006,281:7049-7059.
    138 Yacoub A, McKinstry R, Hinman D,et al. Epidermal growth factor and ionizing radiation up-regulate the DNA repair genes XRCC1 and ERCC1 in DU145 and LNCaP prostate carcinoma through MAPK signaling. Radiat Res, 2003,159:439-453.
    139 Yacoub A, Park JS, Qiao L,et al. MAPK dependence of DNA damage repair: ionizing radiation and the induction of expression of the DNA repair genes XRCC1 and ERCC1 in DU145 human prostate carcinoma cells in a MEK1/2 dependent fashion. Int J Radiat Biol, 2001,77:1067-1078.
    140 Dent P, Yacoub A, Fisher PB,et al. MAPK pathways in radiation responses. Oncogene, 2003,22:5885-5896.
    141 Reardon DB, Contessa JN, Mikkelsen RB, et al. Dominant negative EGFR-CD533 and inhibition of MAPK modify JNK1 activation and enhance radiation toxicity of human mammary carcinoma cells. Oncogene, 1999,18:4756-4766.
    142 Lund EL, Hog A, Olsen MW,et al. Differential regulation of VEGF, HIFlalpha and angiopoietin-1, -2 and -4 by hypoxia and ionizing radiation in human glioblastoma. Int J Cancer, 2004,108:833-838.
    143 Hoshi S, Nomoto K, Kuromitsu J,et al. High glucose induced VEGF expression via PKC and ERK in glomerular podocytes. Biochem Biophys Res Commun, 2002,290:177-184.
    144 Giuliani N, Lunghi P, Morandi F, et al. Downmodulation of ERK protein kinase activity inhibits VEGF secretion by human myeloma cells and myeloma-induced angiogenesis. Leukemia, 2004,18:628-635.
    145 Menu E, Kooijman R, Van Valckenborgh E,et al. Specific roles for the PI3K and the MEK-ERK pathway in IGF-1-stimulated chemotaxis, VEGF secretion and proliferation of multiple myeloma cells: study in the 5T33MM model. Br J Cancer, 2004,90:1076-1083.
    146 Calviello G, Di Nicuolo F, Gragnoli S,et al. n-3 PUFAs reduce VEGF expression in human colon cancer cells modulating the COX-2/PGE2 induced ERK-1 and -2 and HIF-1 alpha induction pathway. Carcinogenesis, 2004,25:2303-2310.
    147 Berra E, Pages G, Pouyssegur J. MAP kinases and hypoxia in the control of VEGF expression. Cancer Metastasis Rev, 2000,19:139-145.
    148 McCawley LJ, Matrisian LM. Matrix metalloproteinases: multifunctional contributors to tumor progression. Mol Med Today, 2000,6:149-156.
    149 Harada T, Arii S, Mise M, et al. Membrane-type matrix metalloproteinase-1(MT1-MMP) gene is overexpressed in highly invasive hepatocellular carcinomas. J Hepatol, 1998,28:231-239.
    150 Orlandoa M , Nathaliea T, Pierrea CJ , et al. In situ detection of matrix metalloproteinase-2 (MMP2) and the metalloproteinase inhibitor TIMP2 transcripts in human primary hepatocellular carcinoma and in liver metastasis. J Hepatol, 1997,26:593-605.
    151 Chang Chieh,Werb Zena. The many faces of metalloproteases: cell growth, invasion, angiogenesis and metastasis. Trends Cell Biol, 2001,1 l:S37-43.
    152 Liotta LA, Steeg PS, Stetler-Stevenson WG. Cancer metastasis and angiogenesis. An imbalance of positive and negative regulation. Cell, 1991,64:327-336.
    153 Arii Shigeki. Role of vascular endothelial growth factor on the invasive potential of hepatocellular carcinoma. J Hepatol, 2004,41:333-335.
    154 Foda Hussein D,Zucker Stanley. Matrix metalloproteinases in cancer invasion, metastasis and angiogenesis. Drug Discov Today, 2001,6:478-482.
    155 Araya J, Maruyama M, Sassa K, et al. Ionizing radiation enhances matrix metalloproteinase-2 production in human lung epithelial cells. Am J Physiol Lung Cell Mol Physiol, 2001,280:130-38.
    156 Tanimura S,Asato K,Fujishiro SH,et al. Specific blockade of the ERK pathway inhibits the invasiveness of tumor cells: down-regulation of matrix metalloproteinase-3/-9/-14 and CD44. Biochem Biophys Res Commun, 2003,304:801-806.
    157 Lakka SS, Jasti SL, Gondi C, et al. Downregulation of MMP-9 in ERK-mutated stable transfectants inhibits glioma invasion in vitro. Oncogene, 2002,21:5601-5608.
    158 Park MJ, Park IC, Lee HC,et al. Protein kinase C-alpha activation by phorbol ester induces secretion of gelatinase B/MMP-9 through ERK 1/2 pathway in capillary endothelial cells. Int J Oncol, 2003,22:137-143.
    159 Sandra D,Lucia M,Giulia T,et al. ERK1-2 and p38 MAPK regulate MMP/TIMP balance and function in response to thrombospondin-1 fragments in the microvascular endothelium. Life Sciences. 2004,74:2975-2985.
    160 Paolo S,Paola A,Silvano G,et al. Matrix metalloproteinase-2 and -9 are induced differently by doxorubicin in H9c2 cells: The role of MAP kinases and NAD(P)H Oxidase. Cardiovasc Res, 2006,69:736-745.
    161 Tanimura S, Nomura K, Ozaki K, et al. Prolonged nuclear retention of activated extracellular signal-regulated kinase 1/2 is required for hepatocyte growth factor-induced cell motility. J Biol Chem, 2002,277:28256-28264.
    162 Welch DR, Sakamaki T, Pioquinto R, et al. Transfection of constitutively active mitogen-activated protein/extracellular signal-regulated kinase kinase confers tumorigenic and metastatic potentials to NIH3T3 cells. Cancer Res, 2000,60:1552-1556.
    163 Husain S, Jafri F, Crosson CE. Acute effects of PGF2alpha on MMP-2 secretion from human ciliary muscle cells: a PKC- and ERK-dependent process. Invest Ophthalmol Vis Sci, 2005,46:1706-1713.
    164 Holvoet S, Vincent C, Schmitt D, et al. The inhibition of MAPK pathway is correlated with down-regulation of MMP-9 secretion induced by TNF- α in human keratinocytes. Exp Cell Res, 2003,290:108-119.
    165 Bryan GA,Alejandro M,Lin H,et al. Pioglitazone inhibits MMP-1 expression in vascular smooth muscle cells through a mitogen-activated protein kinase-independent mechanism. Atherosclerosis, 2005,178:249-256.
    166 Pintucci G, Yu PJ, Sharony R,et al. Induction of stromelysin-1 (MMP-3) by fibroblast growth factor-2 (FGF-2) in FGF-2-/- microvascular endothelial cells requires prolonged activation of extracellular signal-regulated kinases-1 and -2 (ERK-1/2). J Cell Biochem, 2003,90:1015-1025.
    167 Huntington JT, Shields JM, Der CJ, et al. Overexpression of Collagenase 1 (MMP-1) is mediated by the ERK pathway in invasive melanoma cells: role of BRAF mutation and fibroblast growth factor signaling. J Biol Chem, 2004,279:33168-33176.
    168 Tan X, Egami H, Abe M,et al. Involvement of MMP-7 in invasion of pancreatic cancer cells through activation of the EGFR mediated MEK-ERK signal transduction pathway. J Clin Pathol, 2005,58:1242-1248.
    169 Wang JL, Sun Y, Wu S. Gamma-irradiation induces matrix metalloproteinase II expression in a p53-dependent manner. Mol Carcinogen, 2000,27:252-258.
    170 Araya J, Maruyama M, Sassa K,et al. Ionizing radiation enhances matrix metalloproteinase-2 production in human lung epithelial cells. Am J Physiol Lung Cell Mol Physiol, 2001,280:L30-38.
    171 Li P, Gao Y, Ji Z,et al. Role of urokinase plasminogen activator and its receptor in metastasis and invasion of neuroblastoma. J Pediatr Surg, 2004,39:1512-1519.
    172 Rabbani SA, Xing RH. Role of urokinase (uPA) and its receptor (uPAR) in invasion and metastasis of hormone-dependent malignancies. Int J Oncol, 1998,12:911-920.
    173 Carriero MV, Del Vecchio S, Capozzoli M,et al. Urokinase receptor interacts with alpha(v)beta5 vitronectin receptor, promoting urokinase-dependent cell migration in breast cancer. Cancer Res, 1999,59:5307-5314.
    174 Wilson AJ, Gibson PR. Role of urokinase and its receptor in basal and stimulated colonic epithelial cell migration in vitro. Gut, 2000,47:105-111.
    175 Itoh YH, Nakatsugawa S, Oguri T,et al. Increase of cellular fibrinolysis in human lung cancer cell line by radiation: relationship between urokinase-type plasminogen activator (uPA) and metastasis and invasion. Nippon Acta Radiol, 1996,56:747-749.
    176 Rofstad EK, Mathiesen B, Galappathi K. Increased metastatic dissemination in human melanoma xenografts after subcurative radiation treatment: radiation-induced increase in fraction of hypoxic cells and hypoxia-induced up-regulation of urokinase-type plasminogen activator receptor. Cancer Res, 2004,64:13-18.
    177 Adair JC, Baldwin N, Kornfeld M,et al. Radiation-induced blood-brain barrier damage in astrocytoma: relation to elevated gelatinase B and urokinase. J Neuro Oncol, 1999,44:283-289.
    178 Richter KK,Fink LM,Hughes BM,et al. Differential effect of radiation on endothelial cell function in rectal cancer and normal rectum. Am J Surg, 1998,176:642-647.
    179 Torimura T,Ueno T,Kin M,et al. Integrin α 6 β 1 plays a significant role in the attachment of hepatoma cells to laminin. J Hepatol, 1999,31:734-740.
    180 Cordes N, Blaese MA, Meineke V,et al. Ionizing radiation induces up-regulation of functional beta1-integrin in human lung tumour cell lines in vitro. Int J Radiat Biol, 2002,78:347-357.
    181 Onoda JM, Kantak SS, Piechocki MP,et al. Inhibition of radiation-enhanced expression of integrin and metastatic potential in B16 melanoma cells by a lipoxygenase inhibitor. Radiat Res, 1994,140:410-418.
    182 Meineke V, Gilbertz KP, Schilperoort K,et al. Ionizing radiation modulates cell surface integrin expression and adhesion of COLO-320 cells to collagen and fibronectin in vitro. Strahlentherapie und Onkologie, 2002,178:709-714.
    183 Simon EL, Goel HL, Teider N,et al. High dose fractionated ionizing radiation inhibits prostate cancer cell adhesion and beta(1) integrin expression. Prostate, 2005,64:83-91.
    184 So-Young Hwang , Jae-Won Jung , Jae-Seob Jeong ,et al. Dominant-negative Rac increases both inherent and ionizing radiation-induced cell migration in C6 rat glioma cells. Int J Cancer, 2005,118:2056-2063.
    185 Gary G Zhai, Rajeev Malhotra, Meaghan Delaney,et al. Radiation Enhances the Invasive Potential of Primary Glioblastoma Cells via Activation of the Rho Signaling Pathway. J Neuro Oncol, 2005,76:227-237.
    186 Park SH,Cho HN,Lee SJ,et al. Hsp 25-induced radioresistance is associated with reduction of death by apoptosis: involvement of Bcl2 and the cell cycle. J Radiat Res, 2000,154:421-428.
    187 Hess C, Vuong V, Hegyi I,et al. Effect of VEGF receptor inhibitor PTK787/ZK222584 combined with ionizing radiation on endothelial cells and tumour growth. Br J Cancer, 2001,85:2010-2016.
    188 Gordon MS, Margolin K,Talpaz M ,et al. Phase I safety and pharmacokinetic study of recombinant human anti-vascular endothelial growth factor in patients with advanced cancer. J Clin Oncol, 2001,19:843-850.
    189 Kentaro I,Yasushi M,Naoya U,et al. Rapid inhibition of MAPK signaling and anti-proliferation effect via JAK/STAT signaling by interferon- α in hepatocellular carcinoma cell lines. Biochimica et Biophysica Acta, 2005,1745:401-410.
    190 Kaliski A, Maggiorella L, Cengel KA,et al. Angiogenesis and tumor growth inhibition by a matrix metalloproteinase inhibitor targeting radiation-induced invasion.Mol Cancer Ther,2005,4:1717-1728.
    191 Alessi DR,Cuenda A,Cohen P,et al.PD 098059 is a specific inhibitor of the activation of mitogen-activated protein kinase kinase in vitro and in vivo.J Biol Chem,1995,270:27489-27494.

© 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