用户名: 密码: 验证码:
ARHI基因对胰腺癌微环境血管生成的影响及相关分子机制的研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
胰腺癌是恶性程度最高的消化系统肿瘤之一,是西方国家第四大癌症死亡原因;在我国,胰腺癌的发病率逐年上升,在所有肿瘤中居第7位。由于早期诊断困难,胰腺癌患者确诊时约50%出现全身转移;由于缺少有效治疗手段,5年生存率仅6%,中位生存时间仅为4-6个月。
     肿瘤的生长和转移与肿瘤血管生成密切相关,当肿瘤大小超过1~2mm3时,其继续生长依赖于新生血管生成。研究发现多种信号分子参与肿瘤血管生成,其中CXCL8和VEGF是最重要的促血管生成因子,同时多种趋化因子和受体及相关信号通路的参与亦影响肿瘤血管生成。针对胰腺癌血管生成的研究将有利于发现胰腺癌治疗的新靶点。
     ARHI基因是一个母源性印迹基因,是Ras超家族成员之一;与大部分Ras家族成员不同,ARHI基因是一个抑癌基因,其编码的蛋白在人类多种组织中如卵巢、乳腺中表达,但在乳腺癌、卵巢癌中表达下调。北京协和医院消化内科实验室的前期研究发现,ARHI基因是胰腺癌抑癌基因,可以抑制胰腺癌细胞的增殖、促进细胞周期停滞、诱导细胞凋亡及自噬发生和抑制细胞迁移。ARHI基因是胰腺癌发生发展重要抑癌基因,然而肿瘤细胞的生长和转移受到微环境的调控,而在这种调控中血管生成是重要的关键环节,并且是目前和未来治疗的重要靶点。因此探讨ARHI基因对胰腺癌肿瘤血管生成的调控作用及其机制有重要意义,目前尚未见报道。
     本研究目的:通过人胰腺癌组织、胰腺癌细胞株、裸鼠胰腺癌荷瘤,探讨ARHI基因对胰腺癌血管生成的影响及相关机制,探讨ARHI基因对肿瘤微环境中趋化因子和受体的影响及意义。为进一步研究胰腺癌发病机制和抗肿瘤血管生成治疗在胰腺癌中的应用,提供重要的理论依据。
     第一部分ARHI基因对人胰腺癌组织血管生成的影响及其相关临床与病理意义
     目的:探讨ARHI基因与胰腺癌组织血管密度、血管生成相关蛋白和临床与病理的相关性
     方法:1.收集23例胰腺癌组织和27例正常胰腺组织标本,登记临床病理资料。2.应用免疫组化方法检测ARHI、CD34、VEGF和CXCL8蛋白在胰腺癌组织和正常胰腺组织中的表达。3.分析各蛋白表达和胰腺癌临床与病理因素的关系。4.分析ARHI与MVD、VEGF、CXCL8四者之间的相互关系。
     结果:1.ARHI蛋白在正常胰腺组织和胰腺癌组织中表达的阳性率分别为88.9%和21.7%,两者有显著差异;正常胰腺组织MVD值显著低于胰腺癌组织,分别为27.84±2.01和43.47±6.44;正常胰腺组织VEGF蛋白表达阳性率(44.4%)显著低于胰腺癌组织中VEGF (100%),两者比较有显著统计学差异;CXCL8蛋白在正常胰腺组织和胰腺癌组织中表达的阳性率分别为44.4%和100%,两者比较有显著统计学差异。2. ARHI的表达与肿瘤分化程度呈相关性,肿瘤分化程度越低,ARHI表达缺失率越高;CXCL8的表达与CA199水平呈相关性,CA199水平越高,CXCL8阳性表达率越高,与肿瘤分化程度呈相关性,肿瘤分化程度越低,CXCL8阳性表达率越高;VEGF的表达与年龄、性别、吸烟史、糖尿病史、血清CA199水平、血清CA242水平、临床分期、分化程度、肿瘤大小、肿瘤位置、淋巴结转移、血管浸润均无相关性。3.胰腺癌中MVD与CXCL8的表达呈正相关,MVD与VEGF、 CXCL8与VEGF、ARHI与MVD、ARHI与VEGF、ARHI与CXCL8的表达均无显著相关性。4.在所有统计病例中,CXCL8与VEGF的表达呈正相关,ARHI与MVD、VEGF及CXCL8的表达呈负相关。
     小结:1)在胰腺癌组织中,ARHI表达缺失或显著下调,MVD值显著增多,VEGF和CXCL8表达显著上调。2) ARHI、VEGF和CXCL8与肿瘤分化程度相关;MVD与血清CA199水平、临床分期、分化程度、肿瘤大小、淋巴结转移均呈正相关。3)在所有统计病例中,CXCL8与VEGF的表达呈正相关,ARHI与MVD、 VEGF及CXCL8的表达呈负相关。
     第二部分胰腺癌细胞株和裸鼠胰腺癌荷瘤验证ARHI基因对肿瘤血管生成的影响及相关机制的初探
     目的:探讨ARHI基因对胰腺癌血管生成的影响及相关机制。
     方法:1.应用携带ARHI基因的质粒pIRES2-ARHI-EGFP和pLNCX2-ARHI-EGFP,采用脂质体瞬时转染胰腺癌PANC-1细胞系,并用G418筛选建立稳定转染的多克隆细胞株。2.验证ARHI基因表达情况,用MTT法检测ARHI基因对胰腺癌细胞增殖的影响。3.通过Real-time PCR和Western blot检测胰腺癌细胞株中ARHI基因转染对mTOR、SOSC3、Stat3、NF-κB、VEGF、CXCL8mRNA和(或)蛋白表达的影响。4.将ARHI稳定转染PANC-1细胞株、空载体稳定转染PANC-1细胞株和未转染PANC-1细胞株接种于裸鼠腋窝皮下,比较三组裸鼠体重变化、肿瘤重量和体积。5.荷瘤标本一部分进行病理学检测,通过免疫组化检测ARHI、 Ki-67、CD34、VEGF、CXCL8蛋白的表达;一部分标本通过Real-time验证ARHI基因对mTOR、SOSC3、Stat3、NF-κB、VEGF、CXCL8mRNA表达的影响。
     结果:1.G418筛选4周收集稳定转染胰腺癌细胞,通过RT-PCR和Western blot方法检测,在ARHI基因稳定转染组可见ARHI mRNA和蛋白表达,而空载体和对照组无表达。2.慢病毒质粒pLNCX2-ARHI-EGFP转染效率高,荧光更强,用于后续实验。3.MTT结果显示,pLNCX2-ARHI-EGFP组稳定转染多克隆细胞株比pLNCX2-EGFP组稳定转染细胞株增殖缓慢。4. pLNCX2-ARHI-EGFP组mTOR、 Stat3、NF-κB、VEGF、CXCL8mRNA表达比pLNCX2-EGFP组显著降低,SOCS3mRNA表达显著升高;p-mTOR、p-Stat3、NF-κB、CXCL8蛋白表达显著降低。5.接种后所有裸鼠均有一定程度的消瘦,pLNCX2-ARHI-EGFP组消瘦显著,肿瘤质量和体积显著偏小。6. pLNCX2-ARHI-EGFP组裸鼠荷瘤中ARHI mRNA和蛋白表达阳性,pLNCX2-EGFP组和对照组无表达,证明胰腺癌荷瘤成功建立。7.pLNCX2-ARHI-EGFP组裸鼠荷瘤中Ki-67蛋白表达显著下降,MVD显著减少,VEGF和CXCL8蛋白表达显著下降。8. pLNCX2-ARHI-EGFP组裸鼠荷瘤中mTOR、 Stat3、NF-κB、VEGF、CXCL8mRNA表达显著下降,SOCS3mRNA表达显著升高,与细胞结果一致。
     小结:1)成功构建稳定表达ARHI基因的多克隆PANC-1细胞株。2) ARHI基因再表达可以抑制胰腺癌细胞的增殖。3)通过裸鼠胰腺癌荷瘤的建立,在动物水平验证了ARHI基因对胰腺癌生长及肿瘤血管生成的抑制作用。4) ARHI基因抑制胰腺癌细胞和裸鼠荷瘤中VEGF、CXCL8的表达,ARHI基因可能通过mTOR/NF-KB和STAT3信号通路途径抑制CXCL8和VEGF的表达,进而抑制胰腺癌血管生成。
     第三部分ARHI基因对胰腺癌细胞中血管生成相关趋化因子和受体表达的影响
     目的:探讨ARHI基因通过对肿瘤微环境中趋化因子和受体的作用,对胰腺癌生长、侵袭转移、免疫和血管生成的影响。
     方法:1.通过Real-time PCR Array检测稳定转染ARHI基因对84个趋化因子和受体mRNA表达水平的影响。2.挑选与肿瘤血管生成相关的趋化因子和受体,设计引物在ARHI基因稳定转染细胞株和胰腺癌裸鼠荷瘤中进行验证。
     结果:1. PCR Array结果:1) pLNCX2-ARHI-EGFP组中有36个基因mRNA表达下调,9个基因mRNA表达上调;2)与肿瘤血管生成相关基因:促肿瘤血管生成基因中,CCL2、CXCL1、CXCL2、CXCL8、CXCL12和CXCR4表达显著下调,CXCL3、CCR1和CCR2表达轻度下调;抗肿瘤血管生成基因中,CXCL9、 CXCL10、CXCL11和CXCR3表达显著上调,以CXCL9最明显,达27.8倍;3)与肿瘤转移和侵袭相关基因:CXCL12和CXCR4表达显著下调,MMP-2表达轻度下调;与对远端器官定位浸润和潜伏相关基因:CXCL12、CXCR4和CCR7表达显著下调,CXCR5表达轻度下调;与肿瘤免疫调节相关基因:CXCL8、CXCR1和CCR7表达显著下调,其中以CXCR1尤为明显,达-41.3倍。2.验证结果显示,pLNCX2-ARHI-EGFP组较pLNCX2-EGFP组CXCL8、CXCL1和CXCR4mRNA表达显著下降,CXCR3mRNA表达显著上调,与PCR Array结果一致;CXCR2表达显著下降。3. pLNCX2-ARHI-EGFP组裸鼠荷瘤较pLNCX2-EGFP组裸鼠荷瘤中CXCL8、CXCL1、CXCR4和CXCR2mRNA表达显著下降,CXCR3mRNA表达显著上调,与细胞结果一致。
     小结:ARHI基因影响胰腺癌细胞趋化因子和受体的表达,提示ARHI基因抑制胰腺癌生长作用部分通过趋化因子及其受体调节肿瘤微环境的血管生成和免疫而实现的。
     结论:
     1. ARHI在78.3%胰腺癌组织中表达缺失或下调,再次验证ARHI基因是胰腺癌抑癌基因。而ARHI蛋白表达与MVD、VEGF及CXCL8蛋白表达呈负相关。
     2. ARHI基因抑制胰腺癌细胞株增殖,抑制胰腺癌裸鼠荷瘤的生长,抑制胰腺癌裸鼠荷瘤中血管生成和相关蛋白VEGF和CXCL8的表达。
     3. ARHI基因抑制肿瘤中血管生成和相关蛋白VEGF和CXCL8可能通过mTOR/NF-KB和STAT3信号传导通路,进而抑制胰腺癌血管生成。
     4. ARHI基因促进或抑制肿瘤微环境中多种趋化因子和受体,进而影响胰腺癌生长和转移。
Pancreatic cancer is one of the most common gastrointestinal malignant tumors, which has become the fourth cause of cancer death in western countries. Recently, the incidence of pancreatic cancer is increasing in China, which are ranked the7th among all of the cancers. For the low early diagnosis rate, half of the patients have developed systemic metastasis. Overall, pancreatic cancer carries an unfavorable prognosis with short survival rate and poor quality life due to lacking of effective treatment. The5-year overall survival rate is less than6%and the median survival time is4-6months.
     It is known that tumor angiogenesis is critical to tumor growth and metastasis. When the tumor is growing with a size larger than1-2mm3, its growth is angiogenesis-dependent. Many studies showed that a number of signaling molecules were involved in tumor angiogenesis, including CXCL8and VEGF as pro-angiogenesis factors. Furthermore, some chemokines and their receptors could regulate tumor angiogenesis.
     ARHI, a maternally imprinted gene belongs to the Ras superfamily is a tumor suppressor gene, which appeared high expression in ovarian and breast normal tissues, but down-regulated expression in ovarian and breast carcinomas. A series of studies in our group showed that ARHI was a tumor suppressor gene in pancreatic cancer. Re-expression of ARHI in pancreatic cancer cells can inhibit cell proliferation and invasion, induce apoptosis, autophagy and cell cycle arrest, and inhibit cell mobility. ARHI is an importment tumor suprressor gene in the occurrence and development of pancreatic cancer. Actually, the growth and metastasis of tumor cells is regulated by the tumor microenvironment, in which angiogenesis play a key role. Angiogenesis has become the important target for the current and future treatment strategy. Therefore, the study for ARHI, angiogenesis and related mechanisms has the important significance. Until now, there is no report to study ARHI-induced angiogenesis in pancreatic cancer.
     The aim of this study was to disclose the effect of ARHI on angiogenesis and related mechanism in pancreatic cancer and to analyze the impact of ARHI on chemokines and receptors in tumor microenvironment by human pancreatic cancer tissues, cell lines, and pancreatic cancer xenografted in Nude Mice. According to the above researches, we hope to find more theoretical basis of pathogenesis and feasibility of antiangiogenic therapy in pancreatic cancer for the further study.
     PART I:ARHI and angiogenesis-related protein in pancreatic cancer and its clinicopathological significance.
     Aim:To explore the relationship between pancreatic cancer and angiogenesis, and illustrate the role of ARHI in the pancreatic angiogenesis.
     Methods:1.23cases of pancreatic cancer and27cases of normal pancreas with clinical data were collected and recorded.2. Immuohistochemical technique was performed to detect the expression of ARHI, CD34, VEGF and CXCL8in the specimens.3. The relationship between the expression of above proteins and clinicopathological factors of pancreatic cancer was analyzed.4. The relationship among ARHI, MVD, VEGF and CXCL8analyzed.
     Results:1. The positive protein expression rate of ARHI in normal pancreas and pancreatic cancer tissue was88.9%and21.7%, there was a significant difference between the two groups. MVD in normal pancreas and pancreatic cancer are27.84±2.01and43.47±6.44, there was a significant difference. The positive expression rate of VEGF in normal pancreas and pancreatic cancer was44.4%and100%, there was a significant difference. The positive expression rate of CXCL8in normal pancreas and pancreatic cancer was44.4%and100%, there was a significant difference.2. ARHI expression showed significant negative correlation with tumor differentiation. CXCL8expression showed significant positive correlation with CA199and tumor differentiation. The expression of VEGF protein showed a significant correlation with age, gender, smoking history, drinking history, history of diabetes, serum CA199level, serum CA242level, clinical stage, tumor differentiation, tumor size, tumor location, lymphatic metastasis and vessel infiltration.3. In pancreatic cancer cases, MVD showed significant positive correlation with CXCL8, and there were no significant correlation on MVD with VEGF, CXCL8with VEGF, ARHI with MVD, ARHI with VEGF, ARHI with CXCL8.4. In all cases, CXCL8showed significant positive correlation with VEGF, ARHI showed significant negative correlation with MVD, VEGF and CXCL8.
     Summary:1) ARHI expression in pancreatic cancer was loss or significantly down-regulated, however, MVD, VEGF and CXCL8showed a significant high expression in pancreatic cancer.2) ARHI expression had significant correlation with tumor differentiation. MVD showed a significant correlation with serum CA199level, clinical stage, tumor differentiation, tumor size and lymphatic metastasis.3) In all cases, CXCL8showed significant positive correlation with VEGF, and ARHI showed significant negative correlation with MVD, VEGF and CXCL8.
     PART II:The effect of ARHI on angiogenesis of pancreatic cancer cell lines and xenografted in nude mice and the related mechanisms Aim:To investigate the effect of ARHI on angiogenesis of stably transfected PANC-1cell line and pancreatic cancer xenografted in Nude Mice, and to explore the related mechanisms.
     Methods:1. Both plasmids of pIRES2-ARHI-EGFP and pLNCX2-ARHI-EGFP were transfected into human pancreatic cancer cell line PANC-1by lipofectamine2000, the polyclonal stable PANC-1cell line with G418was selected.2. The expressions of ARHI mRNA and protein in pLNCX2-ARHI-EGFP group were analyzed by RT-PCR and Western blot methods respectively. MTT assay were used to determine the effect of ARHI on the cell proliferation.3. The expressions of mTOR, SOSC3, Stat3, NF-κB, VEGF and CXCL8were detected at the mRNA level by Real-time PCR.4. The cells with pLNCX2-ARHI-EGFP, pLNCX2-EGFP and control were injected subcutaneously into nude mice to establish the pancreatic cancer xenografted model. The tumor growth, size, and body weight change of nude mice were recorded and compared.5. The protein expression of ARHI, Ki-67, CD34, VEGF and CXCL8protein in xenografted tumor tissues were studied by immunohistochemistry. The expressions of mTOR, SOSC3, Stat3, NF-κB, VEGF and CXCL8were detected at the mRNA level in xenografted tumor tissues by Real-time PCR.
     Results:1. After4weeks of G418selection, the polyclonal stable transfected PANC-1cell line was identified with ARHI mRNA and protein expression by RT-PCR and western blot.2. The stable cell lines transfected with lentivirus plasmid pLNCX2-ARHI-EGFP were chose for further studies due to the stronger fluorescence density.3. The MTT assay showed that ARHI could inhibit cell proliferation in pLNCX2-ARHI-EGFP group with a significant difference compared with that in vector group at the third day.4. The expression of mTOR, Stat3, NF-κB, VEGF and CXCL8at the mRNA level were significantly down-regulated in pLNCX2-ARHI-EGFP group compared with the polyclonal vector stable cell line, while the SOSC3gene was significantly up-regulated. Western blot analysis showed that the expression of p-mTOR, p-Stat3, NF-κB and CXCL8proteins were significantly down-regulated in pLNCX2-ARHI-EGFP group.5. The growth of tumor in nude mice was much slower in pLNCX2-ARHI-EGFP group than the vector group and control group, meanwhile, the size of tumors of ARHI group were also smaller than the other two groups, and the body weight change of vector group and control group were more obvious than pLNCX2-ARHI-EGFP group.6. Pancreatic cancer xenografted models was successfully established with ARHI positive expression.7. In pLNCX2-ARHI-EGFP group xenografted model, the ARHI protein expressed in the higher level than that in vector group and control group, while the Ki-67index, MVD (characterized with CD34), VEGF and CXCL8protein expressed in lower level inversely.8. The expression of mTOR, Stat3, NF-κB, VEGF and CXCL8at the mRNA level were significantly down-regulated in pLNCX2-ARHI-EGFP group xenografted model than the vector group, while while the SOSC3gene was significantly up-regulated, and the result was in accordance with that in the cell level.
     Summary:1) The polyclonal stable transfected PANC-1cell line which could express ARHI was successfully established.2) Re-expression of ARHI in stable transfected PANC-1cell line could inhibit proliferation of pancreatic cancer cells.3) The pancreatic cancer xenografted nude mice model was successfully established, and the data in the animal levels showed that ARHI gene had an inhibition effect on tumor growth and angiogenesis in pancreatic cancer.4) ARHI gene may inhibit the expression of CXCL8and VEGF by means of mTOR/NF-KB and STAT3signaling pathway, eventually inhibit angiogenesis of pancreatic cancer.
     Part Ⅲ:The effect of ARHI transfection on chemokines and receptors gene expression profile
     Aim:To explore the effect of ARHI transfection on the chemokines and receptors gene expression profile of PANC-1cells.
     Methods:1. Expression profile, including84chemokines and receptors related genes was detected by using quantitative real-time PCR based RT2Profiler(?) PCR Array.2. Real-time PCR was performed to detect mRNA expression of CXCL8, CXCL1, CXCR4, CXCR3and CXCR2in stable transfected cell lines and xenografted cancer tissues.
     Results:1. PCR Array:1) In PANC-1cells transfected with pLNCX2-ARHI-EGFP, the expression levels of mRNA of36genes were down-regulated, and9genes were up-regulated.2) Among these genes related with tumor angiogenesis, CCL2,CXCL1,CXCL2,CXCL8,CXCL12,CXCR4were significantly down-regulated, CXCL3,CCR1,CCR2were slightly down-regulated. CXCL9,CXCL10,CXCL11,CXCR3were significantly up-regulated, and the increase of CXCL9was the most significant (-27.8fold).3) Among these genes related with tumor metastasis and invasion, CXCL12and CXCR4were significantly down-regulated, MMP-2was slightly down-regulated. Among these genes related with location of organ infiltration and latency, CXCL12,CXCR4,CCR7were significantly down-regulated, CXCR5was slightly down-regulated. Among these gene with tumor immunity, CXCL8,CXCR1,CCR7were significantly down-regulated, and decrease of CXCR1was the most significant (-41.3fold).2. The expression of CXCL8, CXCL1and CXCR4at the mRNA level were significantly down-regulated in pLNCX2-ARHI-EGFP group compared with the vector group, while the CXCR3gene was significantly up-regulated, the result was the same as PCR Array. We also found the expression of CXCR2was down-regulated in pLNCX2-ARHI-EGFP group compared with the vector group.3. In xenografted cancer tissues, the expression of CXCL8, CXCL1, CXCR4and CXCR2at the mRNA level were significantly down-regulated in pLNCX2-ARHI-EGFP group compared with the vector group, while the CXCR3gene was significantly up-regulated, and the result was in accordance with that in the cell level.
     Summary:ARHI gene could influence the expression of many chemokines and receptors, which suggests that the mechanisms of ARHI gene inhibiting tumor growth partly correlate with chemokine and receptors to regulating tumor angiogenesis and immunity in microenvironment in pancreatic cancer.
     Conclusions:
     1) The expression of ARHI gene in pancreatic cancer was loss or significantly down-regulated, which verified the inhibition effect of ARHI gene. ARHI showed significant negative correlation with MVD, VEGF and CXCL8.
     2) Re-expression of ARHI gene could inhibit proliferation of pancreatic cancer cells. ARHI gene could inhibit the growth and angiogenesis of xenografted cancer tissues, as well as the expression of VEGF and CXCL8.
     3) ARHI gene may inhibit the expression of CXCL8and VEGF by means of mTOR/NF-KB and STAT3signaling pathway, eventually inhibit angiogenesis of pancreatic cancer.
     4) ARHI gene could influence expression of many chemokines and receptors, and then inhibit tumor growth and metastasis.
引文
[1]Siegel R, Naishadham D, Jemal A. Cancer statistics,2013[J]. CA:a cancer journal for clinicians,2013,63(1):11-30.
    [2]Vincent A, Herman J, Schulick R, et al. Pancreatic cancer[J]. The Lancet,2011, 378(9791):607-620.
    [3]Stathis A, Moore M J. Advanced pancreatic carcinoma:current treatment and future challenges[J]. Nature Reviews Clinical Oncology,2010,7(3):163-172.
    [4]Yu Y, Xu F, Peng H, et al. NOEY2 (ARHI), an imprinted putative tumor suppressor gene in ovarian and breast carcinomas[J]. Proceedings of the National Academy of Sciences,1999,96(1):214-219.
    [5]Hisatomi H, Nagao K, Wakita K, et al. ARHI/NOEY2 inactivation may be important in breast tumor pathogenesis[J]. Oncology,2002,62(2):136-140.
    [6]卢朝晖,陈杰.ARHI在胰腺癌组织中mRNA水平及蛋白表达[J].中国医学科学院学报,2001,23(4):324-327.
    [7]杨红,陈原稼.新的候选抑癌基因NOEY2在胰腺肿瘤中的表达[J].胰腺病学,2002,2(1):28-30.
    [8]Lu X, Qian J, Yu Y, et al. Expression of the tumor suppressor ARHI inhibits the growth of pancreatic cancer cells by inducing G1 cell cycle arrest[J]. Oncology reports,2009,22(3):635-640.
    [9]Yang H, Lu X, Qian J, et al. Imprinted tumor suppressor gene ARHI induces apoptosis correlated with changes in DNA methylation in pancreatic cancer cells[J]. Molecular medicine reports,2010,3(4):581.
    [10]Itakura J, Ishiwata T, Shen B, et al. Concomitant over-expression of vascular endothelial growth factor and its receptors in pancreatic cancer[J]. International journal of cancer,2000,85(1):27-34.
    [11]Matsuo Y, Ochi N, Sawai H, et al. CXCL8/IL-8 and CXCL12/SDF-1α co-operatively promote invasiveness and angiogenesis in pancreatic cancer [J]. International Journal of Cancer,2009,124(4):853-861.
    [12]Zhao X, Li J, Zhuo J, et al. Reexpression of ARHI inhibits tumor growth and angiogenesis and impairs the mTOR/VEGF pathway in hepatocellular carcinoma[J]. Biochemical and biophysical research communications,2010,403(3):417-421.
    [13]Lu Z, Luo R Z, Lu Y, et al. The tumor suppressor gene ARHI regulates autophagy and tumor dormancy in human ovarian cancer cells[J]. The Journal of clinical investigation,2008,118(12):3917.
    [14]Rosen D G, Wang L, Jain AN, et al. Expression of the tumor suppressor gene ARHI in epithelial ovarian cancer is associated with increased expression of p21WAF1/CIP1 and prolonged progression-free survival[J]. Clinical Cancer Research,2004,10(19):6559-6566.
    [15]Weidner N, Semple JP, Welch WR, et al. Tumor angiogenesis and metastasis-correlation in invasive breast carcinoma[J]. New England Journal of Medicine,1991,324(1):1-8.
    [16]Folkman J, Hochberg M. Self-regulation of growth in three dimensions[J]. The Journal of experimental medicine,1973,138(4):745-753.
    [17]Peng H, Xu F, Pershad R, et al. ARHI is the center of allelic deletion on chromosome Ip31 in ovarian and breast cancers[J]. International Journal of Cancer,2000,86(5): 690-694.
    [18]Yuan J, Luo R Z, Fujii S, et al. Aberrant methylation and silencing of ARHI, an imprinted tumor suppressor gene in which the function is lost in breast cancers[J]. Cancer research,2003,63(14):4174-4180.
    [19]Yu Y, Fujii S, Yuan J, et al. Epigenetic regulation of ARHI in breast and ovarian cancer cells[J]. Annals of the New York Academy of Sciences,2003,983(1): 268-277.
    [20]鲁嘉,钱家鸣,杨红,等.小ras同源蛋白I基因在胰腺癌中的抑癌作用及其对表皮生长因子-Ras-Raf-促分裂原活化蛋白激酶/细胞外信号调节激酶信号转导通路的影响[J].中国医学科学院学报,2012,34(3):197-201.
    [21]Hernandez S L, Banerjee D, Garcia A, et al. Notch and VEGF pathways play distinct but complementary roles in tumor angiogenesis[J]. Vascular cell,2013,5(1):17.
    [22]Tsuzuki Y, Carreira C M, Bockhorn M, et al. Pancreas microenvironment promotes VEGF expression and tumor growth:novel window models for pancreatic tumor angiogenesis and microcirculation[J]. Laboratory investigation,2001,81(10): 1439-1451.
    [23]Kopfstein L, Veikkola T, Djonov V G, et al. Distinct roles of vascular endothelial growth factor-D in lymphangiogenesis and metastasis[J]. The American journal of pathology,2007,170(4):1348-1361.
    [24]Dvorak H F, Brown L F, Detmar M, et al. Vascular permeability factor/vascular endothelial growth factor, microvascular hyperpermeability, and angiogenesis[J]. The American journal of pathology,1995,146(5):1029.
    [25]Ikeda N, Adachi M, Taki T, et al. Prognostic significance of angiogenesis in human pancreatic cancer[J]. British journal of cancer,1999,79(9-10):1553.
    [26]Georgiadou D, Sergentanis T N, Sakellariou S, et al. VEGF and Id-1 in pancreatic adenocarcinoma:prognostic significance and impact on angiogenesis[J]. European Journal of Surgical Oncology (EJSO),2014.
    [27]Huang C, Huang R, Chang W, et al. The expression and clinical significance of pSTAT3, VEGF and VEGF-C in pancreatic adenocarcinoma[J]. Neoplasma,2011, 59(1):52-61.
    [28]Kim L C, Song L, Haura E B. Src kinases as therapeutic targets for cancer[J]. Nature Reviews Clinical Oncology,2009,6(10):587-595.
    [29]Sugimoto M, Yamaoka Y, Furuta T. Influence of interleukin polymorphisms on development of gastric cancer and peptic ulcer[J]. World journal of gastroenterology: WJG,2010,16(10):1188.
    [30]Mizukami Y, Jo W S, Duerr E M, et al. Induction of interleukin-8 preserves the angiogenic response in HIF-la-deficient colon cancer cells[J]. Nature medicine, 2005,11(9):992-997.
    [31]Lee D F, Kuo H P, Chen C T, et al. IKKp suppression of TSC1 links inflammation and tumor angiogenesis via the mTOR pathway[J]. Cell,2007,130(3):440-455.
    [32]Huang S, Robinson J B, DeGuzman A, et al. Blockade of nuclear factor-κB signaling inhibits angiogenesis and tumorigenicity of human ovarian cancer cells by suppressing expression of vascular endothelial growth factor and interleukin 8[J]. Cancer Research,2000,60(19):5334-5339.
    [33]Wei D, Le X, Zheng L, et al. Stat3 activation regulates the expression of vascular endothelial growth factor and human pancreatic cancer angiogenesis and metastasis[J]. Oncogene,2003,22(3):319-329.
    [1]Lee D F, Kuo H P, Chen C T, et al. IKKβ suppression of TSC1 links inflammation and tumor angiogenesis via the mTOR pathway[J]. Cell,2007,130(3):440-455.
    [2]Huang S, Robinson J B, DeGuzman A, et al. Blockade of nuclear factor-κB signaling inhibits angiogenesis and tumorigenicity of human ovarian cancer cells by suppressing expression of vascular endothelial growth factor and interleukin 8[J]. Cancer Research,2000,60(19):5334-5339.
    [3]Wei D, Le X, Zheng L, et al. Stat3 activation regulates the expression of vascular endothelial growth factor and human pancreatic cancer angiogenesis and metastasis[J]. Oncogene,2003,22(3):319-329.
    [4]Huang C, Huang R, Chang W, et al. The expression and clinical significance of pSTAT3, VEGF and VEGF-C in pancreatic adenocarcinoma[J]. Neoplasma,2011, 59(1):52-61.
    [5]Rosen D G, Wang L, Jain AN, et al. Expression of the tumor suppressor gene ARHI in epithelial ovarian cancer is associated with increased expression of p21WAFl/CIPl and prolonged progression-free survival[J]. Clinical Cancer Research,2004,10(19):6559-6566.
    [6]Weidner N, Semple JP, Welch WR, et al. Tumor angiogenesis and metastasis-correlation in invasive breast carcinoma[J]. New England Journal of Medicine,1991,324(1):1-8.
    [7]Paku S, Paweletz N. First steps of tumor-related angiogenesis[J]. Laboratory investigation; a journal of technical methods and pathology,1991,65(3):334-346.
    [8]BarSu A, Ruiz-Sauri A, Valencia G, et al. High microvessel density in pancreatic ductal adenocarcinoma is associated with high grade[J]. Virchows Archiv,2013, 462(5):541-546.
    [9]Zhao X, Li J, Zhuo J, et al. Reexpression of ARHI inhibits tumor growth and angiogenesis and impairs the mTOR/VEGF pathway in hepatocellular carcinoma[J]. Biochemical and biophysical research communications,2010,403(3):417-421.
    [10]闫长青,赵玉沛。血管生成因子VEGF, bFGF, endostati在胰腺癌细胞中的表达[J].中华外科杂志,2009,47(10):787-790.
    [11]Itakura J, Ishiwata T, Shen B, et al. Concomitant over-expression of vascular endothelial growth factor and its receptors in pancreatic cancer[J]. International journal of cancer,2000,85(1):27-34.
    [12]Hotz H G, Reber H A, Hotz B, et al. Angiogenesis inhibitor TNP-470 reduces human pancreatic cancer growth[J]. Journal of Gastrointestinal Surgery,2001,5(2): 131-138.
    [13]Wente M N, Keane M P, Burdick M D, et al. Blockade of the chemokine receptor CXCR2 inhibits pancreatic cancer cell-induced angiogenesis[J]. Cancer letters,2006, 241(2):221-227.
    [14]Matsuo Y, Ochi N, Sawai H, et al. CXCL8/IL-8 and CXCL12/SDF-la co-operatively promote invasiveness and angiogenesis in pancreatic cancer[J]. International Journal of Cancer,2009,124(4):853-861.
    [15]Xiong HQ, Abbruzzese J L, Lin E, et al. NF-κB activity blockade impairs the angiogenic potential of human pancreatic cancer cells[J]. International journal of cancer,2004,108(2):181-188.
    [16]Bonavia R, Inda M M, Vandenberg S, et al. EGFRvIII promotes glioma angiogenesis and growth through the NF-κB, interleukin-8 pathway[J]. Oncogene,2012,31(36): 4054-4066.
    [17]Karar J, Maity A. PI3K/AKT/mTOR pathway in angiogenesis[J]. Frontiers in molecular neuroscience,2011,4.
    [18]Chen J, Wang J, Lin L, et al. Inhibition of STAT3 signaling pathway by nitidine chloride suppressed the angiogenesis and growth of human gastric cancer[J]. Molecular cancer therapeutics,2012,11(2):277-287.
    [19]Henkel T, Machleidt T, Alkalay I, et al. Rapid proteolysis of IκB-α is necessary for activation of transcription factor NF-kB [J].1993.
    [20]Xie T X, Xia Z, Zhang N, et al. Constitutive NF-κB activity regulates the expression of VEGF and IL-8 and tumor angiogenesis of human glioblastoma[J]. Oncology reports,2010,23(3):725.
    [21]Shibata A, Nagaya T, Imai T, et al. Inhibition of NF-κB activity decreases the VEGF mRNA expression in MDA-MB-231 breast cancer cells[J]. Breast cancer research and treatment,2002,73(3):237-243.
    [22]Frost P, Shi Y, Hoang B, et al. AKT activity regulates the ability of mTOR inhibitors to prevent angiogenesis and VEGF expression in multiple myeloma cells [J]. Oncogene,2007,26(16):2255-2262.
    [23]Wei D, Le X, Zheng L, et al. Stat3 activation regulates the expression of vascular endothelial growth factor and human pancreatic cancer angiogenesis and metastasis[J]. Oncogene,2003,22(3):319-329.
    [24]Xie T, Huang F J, Aldape K D, et al. Activation of stat3 in human melanoma promoes brain metastasis [J]. Cancer research,2006,66(6):3188-3196
    [25]Fridman W H, Dieu-Nosjean M C, Pages F, et al. The immune microenvironment of human tumors:general significance and clinical impact[J]. Cancer Microenvironment,2013,6(2):117-122.
    [26]Lorusso G, Ruegg C. The tumor microenvironment and its contribution to tumor evolution toward metastasis[J]. Histochemistry and cell biology,2008,130(6): 1091-1103.
    [27]Zlotnik A, Yoshie O. The chemokine superfamily revisited[J]. Immunity,2012, 36(5):705-716.
    [28]Keeley E C, Mehrad B, Strieter R M. Chemokines as mediators of tumor angiogenesis and neovascularization[J]. Experimental cell research,2011,317(5): 685-690.
    [29]Zhao X, Li J, Zhuo J, et al. Reexpression of ARHI inhibits tumor growth and angiogenesis and impairs the mTOR/VEGF pathway in hepatocellular carcinoma[J]. Biochemical and biophysical research communications,2010, 403(3):417-421.
    [30]Teicher B A, Fricker S P. CXCL12 (SDF-1)/CXCR4 pathway in cancer[J]. Clinical Cancer Research,2010,16(11):2927-2931.
    [31]Miiller A, Homey B, Soto H, et al. Involvement of chemokine receptors in breast cancer metastasis[J]. nature,2001,410(6824):50-56.
    [32]Hassan S, Buchanan M, Jahan K, et al. CXCR4 peptide antagonist inhibits primary breast tumor growth, metastasis and enhances the efficacy of anti-VEGF treatment or docetaxel in a transgenic mouse model[J]. International Journal of Cancer,2011,129(1):225-232.
    [33]John A, Tuszynski G. The role of matrix metalloproteinases in tumor angiogenesis and tumor metastasis[J]. Pathology oncology research,2001,7(1): 14-23.
    [34]Tang C H, Tsai C C. CCL2 increases MMP-9 expression and cell motility in human chondrosarcoma cells via the Ras/Raf/MEK/ERK/NF-KB signaling pathway[J]. Biochemical pharmacology,2012,83(3):335-344.
    [35]Zlotnik A, Burkhardt A M, Homey B. Homeostatic chemokine receptors and organ-specific metastasis[J]. Nature Reviews Immunology,2011,11(9):597-606.
    [36]Allavena P, Germano G, Marchesi F, et al. Chemokines in cancer related inflammation[J]. Experimental cell research,2011,317(5):664-673.
    [37]Sperveslage J, Frank S, Heneweer C, et al.251:The role of chemokine receptor CCR7 and Its ligands CCL19 and CCL21 in tumor progression of pancreatic ductal adenocarcinoma[J]. Cytokine,2013,63(3):302.
    [38]Mehrad B, Keane M P, Strieter R M. Chemokines as mediators of angiogenesis[J]. THROMBOSIS AND HAEMOSTASIS-STUTTGART-,2007,97(5):755.
    [39]Matsuo Y, Raimondo M, Woodward T A, et al. CXC-chemokine/CXCR2 biological axis promotes angiogenesis in vitro and in vivo in pancreatic cancer[J]. International Journal of Cancer,2009,125(5):1027-1037.
    [40]Campanella G S V, Colvin R A, Luster A D. CXCL10 can inhibit endothelial cell proliferation independently of CXCR3[J]. PLoS One,2010,5(9):e12700.
    [41]Zhao B, Cui K, Wang C L, et al. The chemotactic interaction between CCL21 and its receptor, CCR7, facilitates the progression of pancreatic cancer via induction of angiogenesis and lymphangiogenesis[J]. Journal of hepato-biliary-pancreatic sciences,2011,18(6):821-828.
    [42]Sounni N E, Noel A. Targeting the tumor microenvironment for cancer therapy[J]. Clinical chemistry,2013,59(1):85-93.
    [43]Shojaei F. Anti-angiogenesis therapy in cancer:current challenges and future perspectives[J]. Cancer letters,2012,320(2):130-137.
    [44]Baker C H, Solorzano C C, Fidler I J. Blockade of vascular endothelial growth factor receptor and epidermal growth factor receptor signaling for therapy of metastatic human pancreatic cancer[J]. Cancer research,2002,62(7):1996-2003.
    [45]Ferrara N, Hillan K J, Gerber H P, et al. Discovery and development of bevacizumab, an anti-VEGF antibody for treating cancer[J]. Nature reviews Drug discovery,2004,3(5):391-400.
    [46]Siu L L, Awada A, Takimoto C H, et al. Phase I trial of sorafenib and gemcitabine in advanced solid tumors with an expanded cohort in advanced pancreatic cancer[J]. Clinical cancer research,2006,12(1):144-151.
    [47]Medioni J, Choueiri T K, Zinzindohoue F, et al. Response of renal cell carcinoma pancreatic metastasis to sunitinib treatment:a retrospective analysis[J]. The Journal of urology,2009,181(6):2470-2475.
    [48]Awasthi N, Zhang C, Schwarz A M, et al. Enhancement of Nab-paclitaxel Antitumor Activity through Addition of Multitargeting Antiangiogenic Agents in Experimental Pancreatic Cancer[J]. Molecular cancer therapeutics,2014: molcanther.0361.2013.
    [49]Kindler H L, Friberg G, Singh D A, et al. Phase II trial of bevacizumab plus gemcitabine in patients with advanced pancreatic cancer[J]. Journal of Clinical Oncology,2005,23(31):8033-8040.
    [50]Van Cutsem E, Vervenne W L, Bennouna J, et al. Phase III trial of bevacizumab in combination with gemcitabine and erlotinib in patients with metastatic pancreatic cancer[J]. Journal of clinical oncology,2009,27(13):2231-2237.
    [51]Ma J X, Sun Y L, Wang Y Q, et al. Triptolide Induces Apoptosis and Inhibits the Growth and Angiogenesis of Human Pancreatic Cancer Cells by Downregulating COX-2 and VEGF[J]. Oncology Research Featuring Preclinical and Clinical Cancer Therapeutics,2013,20(8):359-368.
    [52]Li Q, Weina P, Hickman M. The Use of Artemisinin Compounds as Angiogenesis Inhibitors to Treat Cancer[J].2013.
    [53]Chen H, Sun B, Pan S, et al. Dihydroartemisinin inhibits growth of pancreatic cancer cells in vitro and in vivo[J]. Anti-cancer drugs,2009,20(2):131-140.
    [54]Singh S, Sadanandam A, Nannuru K C, et al. Small-molecule antagonists for CXCR2 and CXCR1 inhibit human melanoma growth by decreasing tumor cell proliferation, survival, and angiogenesis[J]. Clinical Cancer Research,2009, 15(7):2380-2386.
    [55]Varney M L, Singh S, Li A, et al. Small molecule antagonists for CXCR2 and CXCR1 inhibit human colon cancer liver metastases[J]. Cancer letters,2011, 300(2):180-188.
    [56]Gil M, Seshadri M, Komorowski M P, et al. Targeting CXCL12/CXCR4 signaling with oncolytic virotherapy disrupts tumor vasculature and inhibits breast cancer metastases[J]. Proceedings of the National Academy of Sciences, 2013,110(14):E1291-E1300.
    [57]Wong D, Kandagatla P, Korz W, et al. Targeting CXCR4 with CTCE-9908 inhibits prostate tumor metastasis[J]. BMC urology,2014,14(1):12.
    [58]Ruehlmann J M, Xiang R, Niethammer A G, et al. MIG (CXCL9) chemokine gene therapy combines with antibody-cytokine fusion protein to suppress growth and dissemination of murine colon carcinoma[J]. Cancer research,2001,61(23): 8498-8503.
    [59]Gorbachev A, Kish D, Fairchild R. Modulating CXCR3 chemokine expression in the tumor microenvironment as a therapeutic approach for skin cancers(P2074)[J]. The Journal of Immunology,2013,190:132.27.
    [60]Xie K, Wei D, Huang S. Transcriptional anti-angiogenesis therapy of human pancreatic cancer[J]. Cytokine & growth factor reviews,2006,17(3):147-156.
    [61]Turkson J, Sebti S, Jove R, et al. Stat3 inhibitor having anti-cancer activity and methods:U.S. Patent 8,609,639[P].2013-12-17.
    [62]Chien W, Lee D H, Zheng Y, et al. Growth inhibition of pancreatic cancer cells by Histone Deacetylase inhibitor belinostat through suppression of multiple pathways including HIF, NFkB, and mTOR signaling in vitro and in vivo[J]. Molecular carcinogenesis,2013.
    [63]Ray-Coquard I, Favier L, Weber B, et al. Everolimus as second-or third-line treatment of advanced endometrial cancer:ENDORAD, a phase II trial of GINECO[J]. British journal of cancer,2013,108(9):1771-1777.
    [64]Hayman T J, Wahba A, Rath B H, et al. The ATP-Competitive mTOR Inhibitor INK128 enhances in vitro and in vivo radiosensitivity of pancreatic carcinoma cells[J]. Clinical Cancer Research,2014,20(1):110-119.
    [1]Folkman J. Role of angiogenesis in tumor growth and metastasis[C]//Seminars in oncology. WB Saunders,2002,29(6):15-18.
    [2]Carmeliet P, Jain R K. Angiogenesis in cancer and other diseases[J]. Nature,2000, 407(6801):249-257.
    [3]Bonecchi R, Galliera E, Borroni E M, et al. Chemokines and chemokine receptors: an overview[J]. Frontiers in bioscience:a journal and virtual library,2008,14: 540-551.
    [4]Dimberg A. Chemokines in angiogenesis[M]//The Chemokine System in Experimental and Clinical Hematology. Springer Berlin Heidelberg,2010:59-80.
    [5]Singh S, Sadanandam A, Singh R K. Chemokines in tumor angiogenesis and metastasis[J]. Cancer and Metastasis Reviews,2007,26(3-4):453-467.
    [6]Bosisio D, Salvi V, Gagliostro V, et al. Angiogenic and Antiangiogenic Chemokines[J].2013.
    [7]Howard O M, Ben-Baruch A, Oppenheim J J. Chemokines:progress toward identifying molecular targets for therapeutic agents[J]. Trends in biotechnology, 1996,14(2):46-51.
    [8]Lee H J, Song I C, Yun H J, et al. CXC chemokines and chemokine receptors in gastric cancer:From basic findings towards therapeutic targeting[J]. World journal of gastroenterology:WJG,2014,20(7):1681.
    [9]Rollins B J. Chemokines [J]. Blood,1997,90(3):909-928.
    [10]Zlotnik A, Yoshie O. The chemokine superfamily revisited[J]. Immunity,2012, 36(5):705-716.
    [11]Rossi D, Zlotnik A. The biology of chemokines and their receptors[J]. Annual review of immunology,2000,18(1):217-242.
    [12]Mantovani A, Allavena P, Sica A, et al. Cancer-related inflammation[J]. Nature,2008, 454(7203):436-444.
    [13]Bernardini G, Ribatti D, Spinetti G, et al. Analysis of the role of chemokines in angiogenesis[J]. Journal of immunological methods,2003,273(1):83-101.
    [14]Zlotnik A. Chemokines and cancer[J]. International journal of cancer,2006,119(9): 2026-2029.
    [15]Balkwill F R. The chemokine system and cancer[J]. The Journal of pathology,2012, 226(2):148-157.
    [16]Lee H J, Song I C, Yun H J, et al. CXC chemokines and chemokine receptors in gastric cancer:From basic findings towards therapeutic targeting[J]. World journal of gastroenterology:WJG,2014,20(7):1681.
    [17]Marinissen M J, Gutkind J S. G-protein-coupled receptors and signaling networks: emerging paradigms[J]. Trends in pharmacological sciences,2001,22(7):368-376.
    [18]Burger J A, Tsukada N, Burger M, et al. Blood-derived nurse-like cells protect chronic lymphocytic leukemia B cells from spontaneous apoptosis through stromal cell-derived factor-1[J]. Blood,2000,96(8):2655-2663.
    [19]Teicher, B.A. and S.P. Fricker, CXCL12 (SDF-1)/CXCR4 pathway in cancer. Clin Cancer Res,2010.16(11):p.2927-31.
    [20]Muller A, Homey B, Soto H, et al. Involvement of chemokine receptors in breast cancer metastasis[J]. nature,2001,410(6824):50-56.
    [21]Zlotnik, A., A.M. Burkhardt, and B. Homey, Homeostatic chemokine receptors and organ-specific metastasis. Nat Rev Immunol,2011.11(9):p.597-606.
    [22]Allavena, P., et al., Chemokines in cancer related inflammation. Exp Cell Res,2011. 317(5):p.664-73.
    [23]Sperveslage J, Frank S, Heneweer C, et al.251:The role of chemokine receptor CCR7 and Its ligands CCL19 and CCL21 in tumor progression of pancreatic ductal adenocarcinoma[J]. Cytokine,2013,63(3):302.
    [24]Rubin J B. Chemokine signaling in cancer:one hump or two?[C]//Seminars in cancer biology. Academic Press,2009,19(2):116-122.
    [25]Keeley E C, Mehrad B, Strieter R M. Chemokines as mediators of tumor angiogenesis and neovascularization[J]. Experimental cell research,2011,317(5): 685-690.
    [26]陈飞兰,张华蓉,徐承平,等.SDF-1/CXCR4轴活化诱导人内皮祖细胞增殖,迁移及管型形成[J].基础医学与临床,2008,28(5):428-431.
    [27]Romagnani P, Annunziato F, Lasagni L, et al. Cell cycle-dependent expression of CXC chemokine receptor 3 by endothelial cells mediates angiostatic activity[J]. Journal of Clinical Investigation,2001,107(1):53-63.
    [28]Kim W J H. Cellular signaling in tissue regeneration[J]. Yonsei medical journal, 2000,41(6):692-703.
    [29]Addison C L, Daniel T O, Burdick M D, et al. The CXC chemokine receptor 2, CXCR2, is the putative receptor for ELR+CXC chemokine-induced angiogenic activity[J]. The Journal of Immunology,2000,165(9):5269-5277.
    [30]Lai Y, Liu X, Zeng Y, et al. Interleukin-8 induces the endothelial cell migration through the Racl/RhoA-p38MAPK pathway[J]. Cell,2012,51(8):38MAPK.
    [31]Lai Y, Liu X, Zeng Y, et al. Interleukin-8 induces the endothelial cell migration through the Racl/RhoA-p38MAPK pathway[J]. Cell,2012,51(8):38MAPK.
    [32]Grunewald M, Avraham I, Dor Y, et al. VEGF-induced adult neovascularization: recruitment, retention, and role of accessory cells[J]. Cell,2006,124(1):175-189
    [33]Ping Y, Yao X, Jiang J, et al. The chemokine CXCL12 and its receptor CXCR4 promote glioma stem cell-mediated VEGF production and tumour angiogenesis via PI3K/AKT signalling[J]. The Journal of pathology,2011,224(3):344-354.
    [34]Jouan V, Canron X, Alemany M, et al. Inhibition of In vitro angiogenesis by platelet factor-4-derived peptides and mechanism of action[J]. Blood,1999,94(3):984-993.
    [35]Li M, Ransohoff R M. The roles of chemokine CXCL12 in embryonic and brain tumor angiogenesis[C]//Seminars in cancer biology. Academic Press,2009,19(2): 111-115.
    [36]Sparmann A, Bar-Sagi D. Ras-induced interleukin-8 expression plays a critical role in tumor growth and angiogenesis[J]. Cancer cell,2004,6(5):447-458.
    [37]Mizukami Y, Jo W S, Duerr E M, et al. Induction of interleukin-8 preserves the angiogenic response in HIF-la-deficient colon cancer cells[J]. Nature medicine, 2005,11(9):992-997.
    [38]Matsuo Y, Ochi N, Sawai H, et al. CXCL8/IL-8 and CXCL12/SDF-la co-operatively promote invasiveness and angiogenesis in pancreatic cancer[J]. International Journal of Cancer,2009,124(4):853-861.
    [39]Matsuo Y, Raimondo M, Woodward T A, et al. CXC-chemokine/CXCR2 biological axis promotes angiogenesis in vitro and in vivo in pancreatic cancer[J]. International Journal of Cancer,2009,125(5):1027-1037.
    [40]Hattermann K, Mentlein R. An infernal trio:the chemokine CXCL12 and its receptors CXCR4 and CXCR7 in tumor biology [J]. Annals of Anatomy-Anatomischer Anzeiger,2013,195(2):103-110.
    [41]Romain B, Hachet-Haas M, Rohr S, et al. Hypoxia differentially regulated CXCR4 and CXCR7 signaling in colon cancer[J]. Molecular cancer,2014,13(1):58.
    [42]Liang Z, Brooks J, Willard M, et al. CXCR4/CXCL12 axis promotes VEGF-mediated tumor angiogenesis through Akt signaling pathway[J]. Biochemical and biophysical research communications,2007,359(3):716-722.
    [43]Maishi N, Ohga N, Hida Y, et al. CXCR7:a novel tumor endothelial marker in renal cell carcinoma[J]. Pathology international,2012,62(5):309-317.
    [44]Gil M, Seshadri M, Komorowski M P, et al. Targeting CXCL12/CXCR4 signaling with oncolytic virotherapy disrupts tumor vasculature and inhibits breast cancer metastases[J]. Proceedings of the National Academy of Sciences,2013,110(14): E1291-E1300.
    [45]Miao Z, Luker K E, Summers B C, et al. CXCR7 (RDC1) promotes breast and lung tumor growth in vivo and is expressed on tumor-associated vasculature[J]. Proceedings of the National Academy of Sciences,2007,104(40):15735-15740.
    [46]Liu C, Luo D, Reynolds B A, et al. Chemokine receptor CXCR3 promotes growth of glioma[J]. Carcinogenesis,2011,32(2):129-137.
    [47]Campanella G S V, Colvin R A, Luster A D. CXCL10 can inhibit endothelial cell proliferation independently of CXCR3[J]. PLoS One,2010,5(9):e12700.
    [48]Billottet C, Quemener C, Bikfalvi A. CXCR3, a double-edged sword in tumor progression and angiogenesis[J]. Biochimica et Biophysica Acta (BBA)-Reviews on Cancer,2013,1836(2):287-295.
    [49]Van Raemdonck K, Gouwy M, Lepers S A, et al. CXCL4L1 and CXCL4 signaling in human lymphatic and microvascular endothelial cells and activated lymphocytes: involvement of mitogen-activated protein (MAP) kinases, Src and p70S6 kinase[J]. Angiogenesis,2014:1-10.
    [50]Zhang J, Patel L, Pienta K J. CC chemokine ligand 2 (CCL2) promotes prostate cancer tumorigenesis and metastasis[J]. Cytokine & growth factor reviews,2010, 21(1):41-48.
    [51]Sperveslage J, Frank S, Heneweer C, et al.251:The role of chemokine receptor CCR7 and Its ligands CCL19 and CCL21 in tumor progression of pancreatic ductal adenocarcinoma[J]. Cytokine,2013,63(3):302.
    [52]Zheng J, Yang M, Shao J, et al. Chemokine receptor CX3CR1 contributes to macrophage survival in tumor metastasis[J]. Molecular cancer,2013,12(1):141.
    [53]Bosisio D, Salvi V, Gagliostro V, et al. Angiogenic and Antiangiogenic Chemokines[J].2013.
    [54]Singh S, Sadanandam A, Nannuru K C, et al. Small-molecule antagonists for CXCR2 and CXCR1 inhibit human melanoma growth by decreasing tumor cell proliferation, survival, and angiogenesis[J]. Clinical Cancer Research,2009,15(7): 2380-2386.
    [55]Varaey M L, Singh S, Li A, et al. Small molecule antagonists for CXCR2 and CXCR1 inhibit human colon cancer liver metastases[J]. Cancer letters,2011,300(2): 180-188.
    [56]Sun X, Charbonneau C, Wei L, et al. CXCR4-targeted therapy inhibits VEGF expression and chondrosarcoma angiogenesis and metastasis [J]. Molecular cancer therapeutics,2013,12(7):1163-1170.
    [57]Gil M, Seshadri M, Komorowski M P, et al. Targeting CXCL12/CXCR4 signaling with oncolytic virotherapy disrupts tumor vasculature and inhibits breast cancer metastases[J]. Proceedings of the National Academy of Sciences,2013,110(14): E1291-E1300.
    [58]Wong D, Kandagatla P, Korz W, et al. Targeting CXCR4 with CTCE-9908 inhibits prostate tumor metastasis[J]. BMC urology,2014,14(1):12.
    [59]Billottet C, Quemener C, Bikfalvi A. CXCR3, a double-edged sword in tumor progression and angiogenesis[J]. Biochimica et Biophysica Acta (BBA)-Reviews on Cancer,2013,1836(2):287-295.
    [60]Gorbachev A, Kish D, Fairchild R. Modulating CXCR3 chemokine expression in the tumor microenvironment as a therapeutic approach for skin cancers(P2074)[J]. The Journal of Immunology,2013,190:132.27.
    [61]Somovilla-Crespo B, Alfonso-Perez M, Cuesta-Mateos C, et al. Anti-CCR7 therapy exerts a potent anti-tumor activity in a xenograft model of human mantle cell lymphoma[J]. Journal of hematology & oncology,2013,6(1):1-14.
    [62]Jin Y, Arita M, Zhang Q, et al. Anti-angiogenesis effect of the novel anti-inflammatory and pro-resolving lipid mediators[J]. Investigative ophthalmology & visual science,2009,50(10):4743-4752.

© 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