干扰素Tα对肝癌复发再治疗疗效及相关机理的实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
肝癌(主要是肝细胞癌,hepatocellular carcinoma,HCC)是全球第三位癌症杀手,在我国更高居第二位。手术切除仍为肝癌最有效的疗法,但即使根治性切除,5年内仍有60%~70%的转移复发率,成为进一步提高疗效的瓶颈。对部分不能切除肝癌,局部治疗是一种选择,但同样遇到高转移复发率。肝癌是多血管的实体瘤,为此,抗血管生成是一条有希望的途径。我所王鲁等发现干扰素α(IFNα)通过抑制血管生成而减少肝癌的转移复发,其机理是下调血管内皮细胞生长因子(VEGF)。临床随机对照研究亦证实IFN-α可推迟肝癌术后的复发,成为预防肝癌术后复发的常规之一。我们的临床随机分组研究发现肝癌切除术后给予IFNα治疗可延长患者的生存,推迟复发;但与对照组相比复发率并未降低(用药期间生存率提高了,停药一个阶段后生存率才降低到对照组水平)。因此我们推测IFNα在治疗早期可抑制复发病灶生长,但在后期,肝癌可能对IFNα治疗产生耐药性,因此肿瘤重获生长。如能弄清肝癌对IFNα治疗耐药的机制,将有助于设计新的治疗方案,克服肝癌对IFNα的耐药,以提高IFNα治疗肝癌的疗效。
     本课题是在此基础上,通过我所建立的高转移人肝癌原位移植裸鼠模型LCI-D20,模拟临床治疗模式,探讨IFNα治疗一段时间停药后是否能够继续使用,再次应用是否能够达到和第一次同样的治疗效果(目的在于回答临床工作中产生的问题,即是否可以对肝癌术后应用IFNα治疗18个月停药后复发病人再次使用IFNα治疗),通过对IFNα第一、二次治疗前、后血管生成相关基因表达谱异同的分析和验证,分析复发肝癌对再次应用IFNα治疗所产生逃逸的分子机制,试图找到克服肝癌对治疗逃逸的方法。
     1.IFNα停药后再治疗不能有效地抑制肝癌生长和降低微血管密度MVD
     根据我所前期的实验结果,IFNα在剂量为1.5×10.~7 .U/kg/d时能有效地抑制高转移人肝癌原位移植裸鼠模型LCI-D20的生长。本次实验中这一剂量在最初治疗的20天中,IFNα治疗组和对照组在肿瘤大小(0.27±0.19 g versus0.68±0.24 g)、微血管密度[22/HP(95%CI=15-29)versus 46/HP(95%CI=32-60)]和血清VEGF浓度(24.5±8.7 pg/ml versus 41.6±12.0 pg/ml)等方面均有显著差异,P<0.05。停药20天后,肿瘤大小、微血管密度和血清VEGF浓度在两组间的差别已没有统计学意义(P>0.05)。按照实验设计再次给予IFNα治疗(1.5×10.~7 .U/kg/d)20天,与对照组相比,再次治疗组的血清VEGF浓度明显减少(58.0±13.7 pg/ml versus 92.8±24.3 pg/ml,P=0.012),但肿瘤大小(3.22±0.62g,3.81±0.92 g)和微血管密度[61/HP(95%CI=48-74),70/HP(95%CI=59-81)]与第一次治疗不同,没有明显的减少(P>0.05)。
     2.肝癌前负荷在一定范围内对IFNα的治疗没有明显影响
     考虑到IFNα第一次和第二次治疗时肝癌的重量不同,为了证实给药前肝癌的重量是否对IFNα治疗产生影响,一组高转移人肝癌原位移植裸鼠模型LCI-D20在无任何干预的情况下生长30天,此时肿瘤大小与IFNα第二次治疗时相近(约1.5g),给予IFNα治疗(1.5×10.~7 .U/kg/d)20天,与对照组相比,肿瘤重量(2.28±0.63 g versus 3.90±0.80 g)、微血管密度[39/HP(95%CI=30-48)versus 67/HP(95%CI=45-89)]和血清VEGF水平(44.9±10.6 pg/ml versus76.1±15.6 pg/ml)均显著降低(P<0.05)。
     3.IFNα治疗不同时间点血管生成相关基因表达差异的研究
     利用Super Array血管生成相关基因芯片,我们对IFNα第一次治疗前、后和第二次治疗前、后血管生成相关基因的表达谱进行了分析。与治疗前相比,IFNα第一次治疗后有多个促血管生成因子表达下调,如VEGF、angiogenin、bFGF、PDGF-A、TGF、TNF、G-CSF、EGF、IGF、IL-8。IFNα第二次治疗后,除PDGF-A基因表达上调外,其他几个促血管生成因子表达再次下调。
     4.Real-time PER和Western blot分析VEGF165和PDGF-A在IFNα第一、二次治疗前、后的表达。
     考虑到血管周细胞对血管内皮细胞具有重要的支持和信号传导作用,而PDGF/PDGF受体在周细胞的信号传导中具有重要意义,因此将下一步实验研究的重点放到了VEGF165和PDGF-A基因和蛋白水平的检测上。通过探针法real-timePCR和Western blot检测发现,VEGF165在第一次和第二次IFNα治疗过程中表达均下调,并且伴有VEGF受体2(Flk-1)磷酸化水平的降低。这些结果表明VEGF/VEGF受体信号传导通路在前后两次治疗中均被有效地抑制。PDGF-A在第一次治疗中表达下调,在第二次治疗中表达反而上调。
     5.IFNα第二次治疗联合PDGF受体抑制剂imatinib显著降低肿瘤的大小和微血管密度。
     为了验证PDGF-A上调是IFNα第二次治疗效果差的原因,在IFNα第二次治疗过程中联合应用了PDGF/PDGF受体抑制剂imatinib(100mg/kg/d)治疗20天。结果表明联合用药组的肿瘤大小(2.23±0.43 g)、微血管密度[42/HP (95%CI=32-52)],与IFNα单纯再次治疗组[3.22±0.62 g,61/HP (95%CI=48-77)]相比均显著降低,更显著低于对照组[3.81±0.92 g,70/HP (95%CI=59-81)]。
     6.缺氧诱导因子(HIF)1α、2α在各组中和不同细胞系缺氧状态中的表达
     肿瘤的非限制性增生导致瘤体内缺氧,促使HIF-1α、HIF-2α表达上调。其中HIF-1α是VEGF高表达的主要转录因子。Western blot检测结果显示,IFN-α第一次治疗后HIF-1α、HIF-2α的表达水平均显著降低,停药20天后HIF-1α、HIF-2α的表达水平则明显升高。与对照组相比,HIF-1α表达水平在IFNα再次治疗时仍可以被有效地下调;与第一次IFNα治疗不同,HIF-2α的表达水平在IFNα再次治疗时不再被下调。在正常氧浓度培养条件下,Hela、MHCC-97L、MHCC-97H细胞均只表达较低的HIF1α、HIF2α。当在低氧浓度(1%O_2)下培养5h后,HIF1α、HIF2α表达在3种细胞系中均显著升高。
     7.超声造影能够显示抗血管治疗对肝癌血流灌注的影响。
     治疗组裸鼠在给药前、给药的第3天、8天、18天、32天分别进行微泡超声造影,利用软件给出造影强度随时间变化的造影时间强度曲线(TIC曲线),通过公式求出主要参数:产生显影的时间(AT)、达到峰值强度的时间(TTP)及峰值强度(PI)、曲线下面积(AUC)等数据。在拟合回归分析曲线中给出基础强度BI,缩放因子α1,下降曲率α2,阶段曲率α3。通过统计分析发现,在对照组随着肿瘤的增大,AUC有由低到高、再逐渐降低的趋势,而VEGF受体抑制剂Pazopanib治疗组则逐渐降低,给药8天后两组相比有统计学差异。AT在对照组随着时间的延长急剧降低,在治疗组降低的速度明显缓慢。统计分析显示,AUC与MVD正相关,AT与MVD负相关,PI与MVD没有相关性。
     结论
     1.IFNα第二次应用治疗肝癌时不能有效地抑制肿瘤的生长和降低微血管密度,肝癌对IFNα再次治疗产生逃逸。
     2.IFNα治疗高转移裸鼠人肝癌的疗效不受初始治疗时肿瘤负荷影响。
     3.促血管生长因子PDGF-A在IFNα再次治疗高转移裸鼠人肝癌时高表达,与IFNα再次治疗抑瘤效果差有显著的相关性,此时肝癌血管生成由VEGF依赖转化为PDGF-A依赖。IFNα再次治疗高转移裸鼠人肝癌时联合应用PDGF受体抑制剂imatinib可显著提高治疗效果。
     4.超声造影技术能够较好地反映肿瘤生长和治疗过程中血流灌注的变化,时间强度曲线下面积(AUC)和造影剂开始显示的时间(AT)分别与微血管密度正相关和负相关,两者有可能作为抗血管生成治疗疗效检测的指标。
     应用价值
     1.IFNα先后两次治疗高转移裸鼠人肝癌前后不同时间点血管生成相关基因表达谱发生改变,有利于对抗肝癌血管生成治疗耐药机制的深入研究。
     2.IFNα再次治疗高转移裸鼠人肝癌时联合PDGF受体抑制剂imatinib可显著提高治疗效果,为临床合理选择抗血管生成药物的使用提供了线索。
     3.超声造影具有较强的可操作性和较好的依从性,有助于动态观察抗血管治疗的效果,使及时发现耐药性和选择新的抗血管生成药物成为可能。
     创新点
     1.首次发现并提出促血管生长因子PDGF-A在IFNα再次治疗高转移裸鼠人肝癌时高表达,血管生成由VEGF依赖转化为PDGF-A依赖,与肝癌对IFNα再次治疗产生逃逸密切相关。
     2.首次提出IFNα再次治疗肝癌时联合PDGF受体抑制剂imatinib可显著提高治疗效果。
     3.首次应用超声造影动态检测抗肝癌血管治疗的效果,并提出AUC与MVD正相关,AT与MVD负相关。
Hepatocellular carcinoma (HCC), one of the most common malignanciesworldwide and the second cancer killer in China, is a hypervascular tumor, andabundant and tortuous blood vessels distinguish HCC from benign lesions inangiography. The first clues to the endogenous angiogenesis inhibitors came with theobservations that interferon. (IFN-α) may inhibit endothelial cell chemotaxis andproliferation, and IFN-. has been proven to effectively inhibit HCC growth andrecurrence, especially when combined with cytotoxic drugs. Although severalsignaling pathways are involved in the angiogenesis, attention has been focused onthe vascular endothelial growth factor (VEGF) family, especially VEGF-A. Thecritical involvement of VEGF-A in vascular development and angiogenesis is to bindand activate of VEGF receptor-2, which stimulates endothelial migration andproliferation. IFN-. inhibits tumor cells production of VEGF and endothelial cellmotility, and demostrates direct and indirect anti-angiogenic activity. Nowadays,IFN-. is applied in the treatment of HCC, renal cancer, neuroendocrine tumors,prostate cancer, malignant melanoma, chronic myelogenous leukemia, and superficialbladder carcinoma,
     We previously reported that IFN-. inhibited tumor growth and recurrence innude mice bearing human HCC xenografts, the same is true of the nude mice withhuman HCC after curative resection, mediated by anti-angiogenic effect throughdown-regulation of VEGF but not basic fibroblast growth factor (bFGF). In theLCI-D20 model, anti-angiogenic activity was a more important factor than theanti-proliferative effect of p48-ISGF, protein deficiency, which plays a pivotal role inthe IFN-αassociated signal transduction pathway. Despite the conspicuous decreasein tumor burden and vasculature induced by the treatment with IFN-., little is knownabout the reversibility of these changes after IFN-. therapy is discontinued.
     A randomized control trial showed that 18 months' post-operative IFN-.treatment delayed recurrence and prolonged patients' survival. Unfortunately, tumorsrecurred in a number of patients shortly after IFN-αtreatment was discontinued. Thespeed of vascular regrowth in HCC after IFN-. therapy is discontinued is clinicallyrelevant and may influence the use of IFN-.. Based on the results of our clinical trial and pilot study, this experimental design investigates whether relapse is inevitable,and whether restarted IFN-. treatment is effective. The results demonstrate that withvascular regrowth after IFN-. therapy is discontinued, PDGF-A expression increasesdramatically when IFN-αtreatment is restarted. There is no obvious decrease intumor burden and microvessel density (MVD). Pericytes are another vascular celltype that provides endothelial cells with crucial survival signals, and PDGF/PDGFRpathway regulated pericyte homeostasis, so we postulate that up-regulation ofPDGF-A is responsible for the failure of restarted IFN-αtreatment for HCC. To testthis hypothesis, we added imatinib (Gleevec, Novartis, Basel, Switzerland), thePDGF-receptor inhibitor, to the restarted IFN-αtreatment course, and resulted insignificant inhibition of tumor growth and angiogenesis. Overall, the results indicatethat the up-regulation of PDGF-A was responsible for the failure in the restartedIFN-αtreatment when tumors recurred after the first IFN-αtherapy was discontinued,and long-term disease control can be best achieved by combination therapy withimatinib in an IFN-αrestarted treatment course.
     1. Restarted IFN-αtreatment was less effective in suppressing tumor growth than the first treatment course
     When IFN-αtreatment group was compared with the control group, a significantdecrease of tumor weight was observed (0.27±0.19 g versus 0.68±0.24g), MVD wasalso markedly decreased [22/HP(95% CI=15-29) versus 46/HP (95% CI=32-60)].Furthermore, ELISA results showed that the serum VEGF levels were lower in theIFN-αtreatment group than in control group A (24.5±8.7 pg/ml versus 41.6±12.0pg/ml), P<0.05. When IFN-αtreatment was discontinued for 20 days, there was nosignificant difference in the tumor weight between the follow -up of control group andfollow-up of IFN-αtreatment group. Furthermore, the restarted IFN-αtreatment didnot result in significant inhibitory effect on tumor growth and MVD when comparewith control group [restrated group F: 3.22±0.62 g, 61/HP (95% CI=48-74), controlgroup E: 3.81±0.92 g, 70/HP (95% CI=59-81)], P>0.05.
     2. Response of IFN-αtreatment was not influenced by pretreatment tumor weight.
     The poorer tumor response to the restarted treatment of IFN-αmay have resultedfrom a larger tumor load. To test this possibility, tumors were allowed to grow for 35days when the tumor weight was about 1.5 g (the same size as the tumor before the restarted IFN-αtreatment). IFN-αtreatment for 20 days resulted in a significantreduction of tumor weight compared with control group G, being 2.28±0.63 g versus3.90±0.80 g (P=0.003).
     3. Changes in the angiogenesis-related gene expression profile of human HCC in nude mice treated by IFN-α.
     In two independent cDNA microarray analyses, the results showed that IFN-αtreatment down-regulated several pro-angiogenic factors, including VEGF,angiogenin, bFGF, PDGF-A, TGF, TNF, G-CSF, EGF, IGF, and IL-8 in the firsttreatment course. All the pro-angiogenic factors except PDGF-A were againdown-regulated in restarted IFN-αtreatment group compared with control group.
     4. Effects of IFN-αon the expression of VEGF165, PDGF-A and VEGF receptor 2.
     RT-PCR with primers showed two transcripts whose base pair lengthcorresponded to VEGF165 and VEGF121, and a minor transcript corresponding toVEGF189. However, Western blotting results showed no immunoreactive proteinsother than VEGF165. These data indicated that IFN-αdown-regulated the expressionof VEGF165.
     To verify the results from the cDNA microarray analyses, we used RT-PCR andWestern blot assays to study the expression of VEGF165 and PDGF-A. The results ofRT-PCR showed that IFN-αdecreased the VEGF165 expression in the first (IFN-αtreatment group) and restarted IFN-αtreatment course (restarted IFN-αtreatmentgroup) compared with their co ntrol groups, respectively, which corresponds with thechange in serum VEGF concentration.
     Western blot assays for VEGF165 of HCC tissues under reducing conditionsshowed two bands of 23 and 21 kD, respectively, which is consistent with twoglycosylation variants of VEGF165, as reported previously. No other isoforms ofVEGF were detected. Tumors from IFN-αtreatment group expressed a lower level ofVEGF165 than those of control group in the first treatment course. Similarly, in thesecond treatment course, the VEGF165 concentration of restarted IFN-αtreatmentgroup was lower than that of restarted control group. Thus, VEGF165 expression inthe first and second treatment courses was down-regulated by IFN-αtreatment.
     The results of RT-PCR show that PDGF-A expression diminished in tumorstreated with IFN-αcompared with the control group in the first treatment course, andincreased in the restarted IFN-αtreatment group compared with its control group. These results are consistent with the cDNA microarray analysis. Western blot assaysshowed that PDGF-A in IFN-αtreatment group was down-regulated significantlycompared with its control group. In contrast, restarted IFN-αtreatment group had ahigher PDGF-A expression level than its control group in the second treatment course
     To assess the inhibiting effect of IFN-αon the VEGFR2 signaling, thephosphorylation status of VEGFR2 was determined using antibodies that recognize aphosphorylation site in the VEGFR2 kinase domain of tyrosine 951 (Y951), whichplays a critical role in pathological angiogenesis. Western blot assays results showedthat VEGFR2 phosphorylation of Y951 was detected at a high level in control groups,but at a very low level in IFN-αtreatment group and restarted IFN-αtreatment group.Thus, both the VEGF165 level and VEGFR2 phosphorylation demonstrated that theIFN-αretains its ability to inhibit VEGF signaling in the first and second treatmentcourses.
     5. Blockage of PDGF signaling by imatinib significantly improved the efficacy of restarted IFN-αtreatment.
     Combination treatment with imatinib (PDGF receptor inhibitor) and IFN-αresulted in a significant decrease in tumor burden in the second treatment coursecompared to IFN-αtreatment alone (P=0.01) and imatinib treatment alone (P=0.018), which was accompanied by a decrease in angiogenesis estimated by MVD(P=0.007 for IFN-αtreatment alone; P=0.004 for imatinib treatment alone).
     6. The high expression of transcription factor HIF-2αin the restarted IFN-αtreatment may trigger the high expression of PDGF-A.
     The HeLa and MHCC07-H、MHCC07-L cells exposed to hypoxic conditionsexpressed high level of hypoxia inducible factor-1αand 2αcompared with cells in thenormoxic conditions. HIF-1αand HIF-2αare both undergo rapid hypoxia-inducedprotein stabilization and bind identical target DNA sequences. In the first IFN-αtreatment course, the expression of HIF-1αand HIF-2αin the IFN-αtreatment groupB is lower than in the control group A; in the restarted IFN-αtreatment group F, theexpression of HIF-1αwas down regulated again compared with control group E,while HIF-2αmaintained high level in the restarted IFN-αtreatment group F.
     7. Correlation of contrast agent flow and immunohistochemical results.
     Contrast-enhanced ultrasonography were performed at day 9, 12, 17, 27, and 42.For the entire cohort (treated and control tumors), AUC and AT were positively andnegatively correlated with MVD and the tumor volume and weight.
     Conclusions
     1. Restarted IFN-αtreatment was less effective in suppressing tumor growththan the first treatment course by evasion of antiangiogenic targeting of VEGFsignaling.
     2. Response of IFN-αtreatment was not influenced by pretreatment tumorweight.
     3. Up regulation of platelet -derived growth factor-Ais responsible for the failureof restarted interferonαtreatment in hepatocellular carcinoma, and blockage ofPDGF signaling by imatinib significantly improved the efficacy of restarted IFN-αtreatment.
     4. Quantification of intratumor flow of ultrasonographic contrast agent atgray-scele imaging shows promise for monitoring tumor vascular response toanti-angiogenic therapy.
     The potential application of this work
     1. The differential expressed genes, in the first and restrated IFN-αtreatment,identified by angiogenesis associated cDNA microarray analysis provide a valuableresource for basic and clinical studies of angiogenesis in the study of HCC metastasis.
     2. Restarted IFN-αtreatment was not effective because of up-regulation ofPDGF-A and the VEGF signal pathway remained inhibited. The combination ofPDGF-A inhibitor improved the restarted IFN-αtreatment, and may be a tractableclinical strategy for treating recurrent HCC.
     3. Quantification of intratumoral flow of ultrasonographic contrast agent atgray-scale imaging shows promise for monitoring tumor vascular response toanti-angiogenic therapy.
     Originalities of this work
     1. Analysed and confirmed, for the first time, that restarted IFN-αtreatment isnot effective in inhibiting HCC growth because of up-regulation of PDGF-A, and thecombination of PDGF receptor inhibitor imatinib improved the restarted IFN-αtreatment.
     2. Established the platform of using contrast-enhanced ultrasonography toassess the response of anti-angiogenic agents on the HCC in the LCI-D20 model, andfound the AUC and AT were positively and negatively correlated with current histologic indices for quantifying angiogenesis MVD.
引文
1 Tang, ZY, Ye, SL, Liu, YK, Qin, LX, Sun, HC, Ye, QH, Wang, L, Zhou, J, Qiu, S J, Li, Y, Ji, XN, Liu, H, Xia, JL, Wu, ZQ, Fan, J, Ma, ZC, Zhou, XD, Lin, ZY and Liu, KD. A decade's studies on metastasis of hepatocellular carcinoma. J Cancer Res Clin Oncol, 2004, 130: 187-96.
    2 Parkin, DM. Global cancer statistics in the year 2000. Lancet Oncol, 2001, 2: 533-43.
    3 El-Serag, HB. Hepatocellular carcinoma: an epidemiologic view. J Clin Gastroenterol, 2002, 35: S72-8.
    4 Tang, ZY. Hepatocellular carcinoma surgery—review of the past and prospects for the 21st century. J Surg Oncol, 2005, 91: 95-6.
    5 Zhou, XD, Tang, ZY, Yu, YQ, Yang, BH, Lu, JZ, Lin, ZY, Ma, ZC and 7hang, BH. Recurrence after resection of alpha-fetoprotein-positive hepatocellular carcinoma. J Cancer Res Clin Oncol, 1994, 120: 369-73.
    6 Sun, HC, Li, XM, Xue, Q, Chen, J, Gao, DM and Tang, ZY. Study of angiogeuesis induced by metastatic and non-metastatic liver cancer by corneal mieropocket model in nude mice. World J Gastroenterol, 1999, 5: 116-118.
    7 Folkman, J. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med, 1995, 1:27-31.
    8 Folkman, J. Angiogenesis. Annu Rev Med, 2006, 57: 1-18.
    9 Folkman, J. Is angiogenesis an organizing principle in biology and medicine? J Pediatr Surg, 2007, 42: 1-11.
    10 Kerbel, RS. Tumor angiogenesis: past, present and the near future. Carcinogenesis, 2000, 21: 505-15.
    11 Folkman, J, Hatmfeldt, P and Hlatky, L. Cancer: looking outside the genome. Nat Rev Mol Cell Biol, 2000, 1: 76-9.
    12 Hisaka, T, Yano, H, Ogasawara, S, Momosaki, S, Nishida, N, Takemoto, Y, Kojiro, S, Katafuchi, Y and Kojiro, M. Interferon-alphaConl suppresses proliferation of liver cancer cell lines in vitro and in vivo. J Hepatol, 2004, 41: 782-9.
    13 Maheshwari, RK, Srikantan, V, Bhartiya, D, Kleinman, HK and Grant, DS. Differential effects of interferon gamma and alpha on in vitro model of angiogenesis. J Cell Physiol, 1991, 146: 164-9.
    14 Pestka, S, Langer, JA, Zoon, KC and Samuel, CE. Interferons and their actions. Annu Rev Biochem, 1987, 56: 727-77.
    15 Brouty-Boye, D and Zetter, BR. Inhibition of cell motility by interferon. Science, 1980, 208: 516-8.
    16 Sidky, YA and Borden, EC. Inhibition of angiogenesis by interferons: effects on tumor-and lymphocyte-induced vascular responses. Cancer Res, 1987, 47: 5155-61.
    17 Patt, YZ, Hassan, MM, Lozano, RD, Brown, TD, Vauthey, JN, Curley, SA and Ellis, LM. Phase Ⅱ trial of systemic continuous fluorouracil and subcutaneous recombinant interferon Alfa-2b for treatment of hepatocellular carcinoma. J Clin Oncol, 2003, 21: 421-7.
    18 Obi, S, Yoshida, H, Toune, R, Unuma, T, Kanda, M, Sato, S, Tateishi, R, Teratani, T, Shiina, S and Omata, M. Combination therapy of intraarterial 5-fluorouracil and systemic interferon-alpha for advanced hepatocellular carcinoma with portal venous invasion. Cancer, 2006, 106: 1990-7.
    19 Sun, HC, Tang, ZY, Wang, L, Qin, LX, Ma, ZC, Ye, QH, Zhang, BH, Qian, YB, Wu, ZQ, Fan, J, Zhou, XD, Zhou, J, Qiu, SJ and Shen, YE Postoperative interferon alpha treatment postponed recurrence and improved overall survival in patients after curative resection of HBV-related hepatocellular carcinoma: a randomized clinical trial. J Cancer Res Clin Oncol, 2006, 132: 458-65.
    20 Ravaud, A and Dilhuydy, MS. Interferon alpha for the treatment of advanced renal cancer. Expert Opin Biol Ther, 2005, 5: 749-62.
    21 von Marschall, Z, Scholz, A, Cramer, T, Schafer, G, Schirner, M, Oberg, K, Wiedenmann, B, Hocker, M and Rosewicz, S. Effects of interferon alpha on vascular endothelial growth factor gene transcription and tumor angiogenesis. J Natl Cancer Inst, 2003, 95: 437-48.
    22 Huang, SF, Kim, SJ, Lee, AT, Karashima, T, Bucana, C, Kedar, D, Sweeney, P, Mian, B, Fan, D, Shepherd, D, Fidler, IJ, Dinney, CP and Killion, JJ. 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: 5720-6.
    23 Hancock, BW, Wheatley, K, Harris, S, Ives, N, Harrison, G, Horsman, JM, Middleton, MR, Thatcher, N, Lorigan, PC, Marsden, JR, Burrows, L and Gore, M. 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.
    24 Sun, FX, Tang, ZY, Lui, KD, Ye, SL, Xue, Q, Gao, DM and Ma, ZC. Establishment of a metastatic model of human hepatocellular carcinoma in nude mice via orthotopic implantation of histologically intact tissues. Int J Cancer, 1996, 66: 239-43.
    25 Wang, L, Tang, ZY, Qin, LX, Wu, XF, Sun, HC, Xue, Q and Ye, SL. High-dose and long-term therapy with interferon-alfa inhibits tumor growth and recurrence in nude mice bearing human hepatoceUular carcinoma xenografts with high metastatic potential. Hepatology, 2000, 32. 43-8.
    26 Wang, L, Wu, WZ, Sun, HC, Wu, XF, Qin, LX, Liu, YK, Liu, KD and Tang, ZY. Mechanism of interferon alpha on inhibition of metastasis and angiogenesis of hepatocellular carcinoma after curative resection in nude mice. J Gastrointest Surg, 2003, 7: 587-94.
    27 Hurwitz, H, Fehrenbacher, L, Novotny, W, Cartwright, T, Hainsworth, J, Helm, W, Berlin, J, Baron, A, Griffing, S, Holmgren, E, Ferrara, N, Fyfe, G, Rogers, B, Ross, R and Kabbinavar, F. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med, 2004, 350: 2335-42.
    28 Motzer, RJ, Michaelson, MD, Redman, BG, Hudes, GR, Wilding, G, Figlin, RA, Ginsberg, MS, Kim, ST, Baum, CM, DePrimo, SE, Li, JZ, Bello, CL, Theuer, CP, George, DJ and Rini, BI. Activity of SU11248, a multitargeted inhibitor of vascular endothelial growth factor receptor and platelet-derived growth factor receptor, in patients with metastatic renal cell carcinoma. J Clin Oncol, 2006, 24: 16-24.
    29 Escudier, B, Eisen, T, Stadler, WM, Szczylik, C, Oudard, S, Siebels, M, Negrier, S, Chevreau, C, Solska, E, Desai, AA, Rolland, F, Demkow, T, Hutson, TE, Gore, M, Freeman, S, Schwartz, B, Shah, M, Simantov, R and Bukowski, RM. Sorafenib in advanced clear-cell renal-cell carcinoma. N Engl J Med, 2007, 356: 125-34.
    30 Jubb, AM, Oates, AJ, Holden, S and Koeppen, H. Predicting benefit from antiangiogenic agents in malignancy. Nat Rev Cancer, 2006, 6: 626-35.
    31 Czekierdowski, A, Szymanski, M, Szumilo, J and Kotarski, J. [Color Doppler blood flow measurements and microvessel density assessment in ovarian tumors]. Ginekol Pol, 2003, 74: 695-700.
    32 Buadu, LD, Murakami, J, Murayama, S, Hashiguchi, N, Toyoshima, S, Sakai, S, Yabuuchi, H, Masuda, K, Kurold, S and Ohno, S. Colour Doppler sonography of breast masses: a multiparameter analysis. Clin Radiol, 1997, 52: 917-23.
    33 Chaudhari, MH, Forsberg, F, Voodarla, A, Saikali, FN, Goonewardene, S, Needleman, L, Finkel, GC and Goldberg, BB. Breast tumor vascularity identified by contrast enhanced ultrasound and pathology: initial results. Ultrasonics, 2000, 38: 105-9.
    34 Weidner, N. Current pathologic methods for measuring intratumoral microvessel density within breast carcinoma and other solid tumors. Breast Cancer Res Treat, 1995, 36: 169-80.
    35 Wu, WZ, Sun, HC, Gao, YQ, Li, Y, Wang, I., Zhou, K, Liu, KD, Iliakis, G and Tang, ZY. Reduction in p48-ISGFgamma levels confers resistance to interferon-alpha2a in MHCC97 cells. Oncology, 2004, 67: 428-40.
    36 de Bont, ES, Fidler, V, Meeuwsen, T, Scherpen, F, Hahlen, K and Kamps, WA. Vascular endothelial growth factor secretion is an independent prognostic factor for relapse, free survival in pediatric acute myeloid leukemia patients. Clin Cancer Res, 2002, 8: 2856-61.
    37 Folkman, J, Cole, P and Zimmerman, S. Tumor behavior in isolated perfused organs: in vitro growth and metastases of biopsy material in rabbit thyroid and canine intestinal segment. Ann Surg, 1966, 164: 491-502.
    38 Sun, HC, Tang, ZY, Li, XM, Zhou, YN, Sun, BR and Ma, ZC. Microvessel density of hepatocellular carcinoma: its relationship with prognosis. J Cancer Res Clin Oncol, 1999, 125 419-26.
    39 Viloria-Petit, A, Crombet, T, Jothy, S, Hicklin, D, Bohlen, P, Schlaeppi, JM, Rak, J and Kerbel, RS. Acquired resistance to the antitumor effect of epidermal growth factor receptor-blocking antibodies in vivo: a role for altered tumor angiogenesis. Cancer Res, 2001, 61: 5090-101.
    40 Casanovas, O, Hicklin, DJ, Bergers, G and Hanahan, D. Drug esistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell, 2005, 8: 299-309.
    41 Kerbel, RS. Therapeutic implications of intrinsic or induced angiogenic growth factor redundancy in tumors revealed. Cancer Cell, 2005, 8: 269-71.
    42 Relf, M, LeJeune, S, Scott, PA, Fox, S, Smith, K, Leek, R, Moghaddam, A, Whitehouse, R, Bicknell, R and Harris, AL. Expression of the angiogenic factors vascular endothelial cell growth factor, acidic and basic fibroblast growth factor, tumor growth factor beta-1, platelet-derived endothelial cell growth factor, placenta growth factor, and pleiotrophin in human primary breast cancer and its relation to angiogenesis. Cancer Res, 1997, 57: 963-9.
    43 Drevs, J, Zirrgiebel, U, Schmidt-Gersbach, CI, Mross, K, Medinger, M, Lee, L, Pinheiro, J, Wood, J, Thomas, AL, Unger, C, Henry, A, Steward, WP, Laurent, D, Lebwohl, D, Dugan, M and Marine, D. Soluble markers for the assessment of biological activity with PTK787/ZK 222584 (PTK/ZK), a vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitor in patients with advanced colorectal cancer from two phase I trials. Ann Oncol, 2005, 16: 558-65.
    44 Allinen, M, Beroukhim, R, Cai, L, Brennan, C, Lahti-Domenici, J, Huang, H, Porter, D, Hu, M, Chin, L, Richardson, A, Schnitt, S, Sellers, WR and Polyak, K. Molecular characterization of the tumor microenvironment in breast cancer. Cancer Cell, 2004, 6: 17-32.
    45 Katano, M, Nakamura, M, Fujimoto, K, Miyazaki, K and Morisaki, T. Prognostic value of platelet-derived growth factor-A (PDGF-A) in gastric carcinoma. Ann Surg, 1998, 227: 365-71.
    46 Heinrich, MC, Corless, CL, Duensing, A, McGreevey, L, Chen, CJ, Joseph, N, Singer, S, Griffith, DJ, Haley, A, Town, A, Demetri, GD, Fletcher, CD and Fletcher, JA. PDGFRA activating mutations in gastrointestinal stromal tumors. Science, 2003, 299. 708-10.
    47 Gerhardt, H and Betsholtz, C. Endothelial-pericyte interactions in angiogenesis. Cell Tissue Res, 2003, 314: 15-23.
    48 Pietras, K and Hanahan, D. A multitargeted, metronomic, and maximum-tolerated dose "chemo-switch" regimen is antiangiogenic, producing objective responses and survival benefit in a mouse model of cancer. J Clin Oncol, 2005, 23: 939-52.
    49 Pooh, RT, Lau, CP, Cheung, ST, Yu, WC and Fan, ST. Quantitative correlation of serum levels and tumor expression of vascular endothelial growth factor in patients with hepatocellular carcinoma. Cancer Res, 2003, 63: 3121-6.
    50 Neufeld, G, Cohen, T, Gengrinovitch, S and Poltorak, Z. Vascular endothelial growth factor (VEGF) and its receptors. Faseb J, 1999, 13: 9-22.
    51 Seghezzi, G, Patel, S, Ren, C J, Gualandris, A, Pintucci, G, Robbins, ES, Shapiro, RL, Galloway, AC, Rifkin, DB and Mignatti, P. Fibroblast growth factor-2 (FGF-2) induces vascular endothelial growth factor (VEGF) expression in the endothelial cells of forming capillaries: an autocrine mechanism contributing to angiogenesls. J Cell Biol, 1998, 141: 1659-73.
    52 Rosen, LS. Clinical experience with angiogenesis signaling inhibitors: focus on vascular endothelial growth factor (VEGF) blockers. Cancer Control, 2002, 9: 36-44.
    53 Carmeliet, P. VEGF as a key mediator of angiogenesis in cancer. Oncology, 2005, 69 Suppl 3: 4-10.
    54 Karpanen, T, Wirzenius, M, Makinen, T, Veikkola, T, Haisma, HJ, Achen, MG, Stacker, SA, Pytowski, B, Yla-Herttuala, S and Alitalo, K. Lymphangiogenic growth factor responsiveness is modulated by postnatal lymphatic vessel maturation. Am J Pathol, 2006, 169: 708-18.
    55 Paavonen, K, Puolakkainen, P, Jussila, L, Jahkola, T and Alitalo, K. Vascular endothelial growth factor receptor-3 in lymphangiogenesis in wound healing. Am J Pathol, 2000, 156: 1499-504.
    56 Sctmadig, ID and Blanke, CD. Gastrointestinal stromal tumors: imatinib and beyond. Curr Treat Options Oncol, 2006, 7: 427-37.
    57 Joensuu, H and Dimitrijevic, S. Tyrosine kinase inhibitor imatinib (STI571) as an anticancer agent for solid tumours. Ann Med, 2001, 32: 451-5.
    58 Carroll, M, Ohno-Jones, S, Tamura, S, Buchdunger, E, Zimmermann, J, Lydon, NB, Gilliland, DG and Druker, BJ. CGP 57148, atyrosine kinase inhibitor, inhibits the growth of ceils expressing BCR-ABL, TEL-ABL, and TEL-PDGFR fusion proteins. Blood, 1997, 90: 4947-52.
    59 Uchida, T, Rossignol, F, Matthay, MA, Mounier, R, Couette, S, Clottes, E and Clerici, C. Prolonged hypoxia differentially regulates hypoxia-inducible factor (HIF)-lalpha and HIF-2alpha expression in lung epithelial cells: implication of natural antisense HIF-1alpha. J Biol Chem, 2004, 279: 14871-8.
    60 WiUett, CG, Boucher, Y, di Tomaso, E, Duda, DG, Munn, LL, Tong, RT, Chung, DC, Sahani, DV, Kalva, SP, Kozin, SV, Mino, M, Cohen, KS, Scadden, DT, Hartford, AC, Fischman, AJ, Clark, JW, Ryan, DP, Zhu, AX, Blaszkowsky, LS, Chen, HX, Shellito, PC, Lauwers, GY and Jain, RK. Direct evidence that the VEGF-specific antibody bevacizumab has antivascular effects in human rectal cancer. Nat Med, 2004, 10: 145-7.
    61 Shih, T and Lindley, C. Bevacizumab: an angiogenesis inhibitor for the treatment of solid malignancies. Clin Ther, 2006, 28: 1779-802.
    62 Karhoff, D, Sauer, S, Schrader, J, Arnold, R, Fendrich, V, Bartsch, DK and Horsch, D. Rap1/B-Raf Signaling Is Activated in Neuroendocrine Tumors of the Digestive Tract and Raf Kinase Inhibition Constitutes a Putative Therapeutic Target. Neuroendocrinology, 2007,
    63 Kamat, AA, Kiln, TJ, Landen, CN, Jr., Lu, C, Han, LY, Lin, YG, Merritt, WM, Thaker, PH, Gershenson, DM, Bischoff, FZ, Heymach, JV, Jaffe, RB, Coleman, RL and Sood, AK. Metronomic chemotherapy enhances the efficacy of antivascular therapy in ovarian cancer. Cancer Res, 2007, 67: 281-8.
    64 Miller, JC, Pien, HH, Sahani, D, Sorensen, AG and Thrall, JH. Imaging angiogenesis: applications and potential for drug development. J Natl Cancer Inst, 2005, 97: 172-87.
    65 Hormigo, A, Gutin, PH and Rafii, S. Tracking normalization of brain tumor vasculature by magnetic imaging and proangiogenic biomarkers. Cancer Cell, 2007, 11: 6-8.
    66 Meijerink, MR, van Cruijsen, H, Hoekman, K, Kater, M, van Schaik, C, van Waesberghe, JH, Giaccone, G and Manoliu, RA. The use of perfusion CT for the evaluation of therapy combining AZD2171 with gefitinib in cancer patients. Eur Radiol, 2006,
    67 McCarville, MB, Streck, CJ, Dickson, PV, Li, CS, Nathwani, AC and Davidoff, AM. Angiogenesis inhibitors in a murine neuroblastoma model: quantitative assessment of intratumoral blood flow with contrast-enhanced gray-scale US. Radiology, 2006, 240: 73-81.
    68 Garcia, Y, Breen, A, Burugapalli, K, Dockery, P and Pandit, A. Stereological methods to assess tissue response for tissue-engineered scaffolds. Biomaterials, 2007, 28: 175-86.
    69 Phongkitkarun, S, Kobayashi, S, Kan, Z, Lee, TY and Charnsangavej, C. Quantification of angiogenesis by functional computed tomography in a Matrigel model in rats. Acad Radiol, 2004, 11: 573-82.
    70 Guidi, L, De Franco, A, De Vitis, I, Armuzzi, A, Semeraro, S, Roberto, I, Papa, A, Bock, E, Gasbarrini, G and Fedeli, G. Contrast-enhanced ultrasonography with SonoVue after infliximab therapy in Crohn's disease. Eur Rev Med Pharmacol Sci, 2006, 10: 23-6.
    71 Torzilli, G. Adverse effects associated with SonoVue use. Expert Opin Drug Saf, 2005, 4: 399-401.
    72 Li, J, Dong, B, Yu, X, Wang, X and Li, C. Grey-scale contrast enhancement in rabbit liver with SonoVue at different doses. Ultrasound Med Biol, 2005, 31: 185-90.
    73 Broillet, A, Hantson, J, Ruegg, C, Messager, T and Schneider, M. Assessment of microvascular perfusion changes in a rat breast tumor model using SonoVue to monitor the effects of different anti-angiogenic therapies. Acad Radiol, 2005, 12 Suppl 1: S28-33.
    74 Ye, QH, Qin, LX, Forgues, M, He, P, Kim, JW, Peng, AC, Simon, R, Li, Y, Robles, AI, Chert, Y, Ma, ZC, Wu, ZQ, Ye, SL, Liu, YK, Tang, ZY and Wang, XW. Predicting hepatitis B virus-positive metastatic hepatocellular carcinomas using gene expression profilhg and supervised machine learning. Nat Med, 2003, 9: 416-23.
    75 Podar, K, Tonon, G, Sattler, M, Tai, YT, LegouiU, S, Yasui, H, Ishitsuka, K, Kumar, S, Kumar, R, Pandite, LN, Hideshima, T, Chauhan, D and Anderson, KC. The small-molecule VEGF receptor inhibitor pazopanib (GW786034B) targets both tumor and endothelial cells in multiple myeloma. Proc Natl Acad Sci U S A, 2006, 103: 19478-83.
    76 Niho, S, Kubota, K, Goto, K, Yoh, K, Ohmatsu, H, Kakinuma, R, Saijo, N and Nishiwaki, Y. First-line single agent treatment with gefitinib in patients with advanced non-small-cell lung cancer: a phase Ⅱ study. J Clin Oncol, 2006, 24: 64-9.
    77 Jubb, AM, Hurwitz, HI, Bai, W, Holmgren, EB, Tobin, P, Guerrero, AS, Kabbinavar, F, Holden, SN, Novotny, WF, Frantz, GD, Hillan, KJ and Koeppen, H. Impact of vascular endothelial growth factor-A expression, thrombospondin-2 expression, and microvessel density on the treatment effect of bevacizumab in metastatic colorectal cancer. J Clin Oncol, 2006, 24: 217-27.
    78 Des Guetz, G, Uzzan, B, Nicolas, P, Cucherat, M, Morere, JF, Benamouzig, R, Breau, JL and Perret, GY. Microvessel density and VEGF expression are prognostic factors in colorectal cancer. Meta-analysis of the literature. Br J Cancer, 2006, 94: 1823-32.
    79 Shen, PQ, Chen, ZL, Xiao, FC and Shen, XD. [Relationship of angiopoietins expression with microvessel density (MVD) in human colorectal tumors]. Zhejiang Da Xue Xue Bao Yi Xue Ban, 2006, 35: 194-8, 203.
    80 Valente, G, Mamo, C, Bena, A, Prudente, E, Cavaliere, C, Kerim, S, Nicotra, G, Comino, A, Palestro, G, Isidoro, C and Beatrice, F. Prognostic significance of microvessel density and vascular endothelial growth factor expression in sinonasal carcinomas. Hum Pathol, 2006, 37: 391-400.
    81 Yao, L, Li, X, Lu, X and Yin, W. [The significance of microvessel density and CD34 expression in nasal polyps]. Lin Chuang Er Bi Yah Hou Ke Za Zhi, 2005, 19: 913-5.
    82 El-Assal, ON, Yamanoi, A, Soda, Y, Yamaguchi, M, Igarashi, M, Yamamoto, A, Nabika, T and Nagasue, N. Clinical significance of microvessel density and vascular endothelial growth factor expression in hepatocellular carcinoma and surrounding liver: possible involvement of vascular endothelial growth factor in the angiogenesis of cirrhotic liver. Hepatology, 1998, 27: 1554-62.
    83 Ker, CG, Chen, HY, Juan, CC, Lo, HW, Shen, YY, Chen, JS, Lee, KT and Sheen, PC. Role of angiogenesis in hepatitis and hepatocellular carcinoma. Hepatogastroenterology, 1999, 46: 646-50.
    84 Forsberg, F, Ro, RJ, Potoczek, M, Liu, JB, Merritt, CR, James, KM, Dicker, AP and Nazarian, LN. Assessment of angiogenesis: implications for ultrasound imaging. Ultrasonics, 2004, 42 325-30.
    85 Ferrara, KW, Merritt, CR, Burns, PN, Foster, FS, Mattrey, RF and Wickline, SA. Evaluation of tumor angiogenesis with US: imaging, Doppler, and contrast agents. Acad Radiol, 2000, 7: 824-39.
    86 Forsberg, F, Merton, DA, Liu, JB, Needleman, L and Goldberg, BB. Clinical applications of ultrasound contrast agents. Ultrasonics, 1998, 36: 695-701.
    87 Reynolds, K, Farzaneh, F, Collins, WP, Campbell, S, Bourne, TH, Lawton, F, Moghaddam, A, Harris, AL and Bicknell, R. Association of ovarian malignancy with expression of platelet-derived endothelial cell growth factor. J Natl Cancer hast, 1994, 86 1234-8.
    88 Hata, K, Nagami, H, Iida, K, Miyazaki, K and Collins, WP. Expression of thymidine phosphorylase in malignant ovarian tumors: correlation with microvessel density and an ultrasound-derived index of angiogenesis. Ultrasound Obstet Gynecol, 1998, 12: 201-6.
    89 Lee, WJ, Chu, JS, Holing, SJ, Chung, MF, Wang, SM and Chert, KM. Breast cancer angiogenesis: a quantitative morphologic and Doppler imaging study. Ann Surg Oncol, 1995, 2: 246-51.
    90 Sterns, EE, SenGupta, S, Saunders, F and Zee, B. Vascularity demonstrated by Doppler ultrasound and immunohistochemistry in invasive ductal carcinoma of the breast. Breast Cancer Res Treat, 1996, 40: 197-203.
    91 Lamer, S, Sigal, R, Lassau, N, Bosq, J, Frouin, F, Di Paola, M, Mamelle, G, Leclere, J, Bittoun, J and Di Paola, R. Radiologic assessment of intranodal vascularity in head and neck squamous cell carcinoma. Correlation with histologic vascular density. Invest Radiol, 1996, 31: 673-9.
    1 Bergers, G, Javaherian, K, Lo, KM, Folkman, J and Hanahan, D. Effects of angiogenesis inhibitors on multistage carcinogenesis in mice. Science, 1999, 284: 808-12.
    2 Yang, JC, Haworth, L, Sherry, RM, Hwu, P, Schwartzentruber, DJ, Topalian, SL, Steinberg, SM, Chen, HX and Rosenberg, SA. A randomized trial of bevacizumab, an anti-vascular endothelial growth factor antibody, for metastatic renal cancer. N Engl J Med, 2003, 349: 427-34.
    3 Folkman, J. Angiogenesis. Annu Rev Med, 2006, 57: 1-18.
    4 Carmeliet P, JR. Angiogenesis in cancer and other diseases. Nature., 2000, 407: 249-257.
    5 Ferrara, N. Vascular endothelial growth factor: basic science and clinical progress. Endocr Rev, 2004, 25: 581-611.
    6 Niho, S, Kubota, K, Goto, K, Yoh, K, Ohmatsu, H, Kakinuma, R, Saijo, N and Nishiwaki, Y. First-line single agent treatment with gefitinib in patients with advanced non-small-cell lung cancer: a phase Ⅱ study. J Clin Oacol, 2006, 24: 64-9.
    7 Hurwitz, H, Fehrenbacher, L, Novotny, W, Cartwright, T, Hainsworth, J, Heim, W, Berlin, J, Baron, A, Griffing, S, Holmgren, E, Ferrara, N, Fyfe, G, Rogers, B, Ross, R and Kabbinavar, F. Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer. N Engl J Med, 2004, 350: 2335-42.
    8 Motzer, RJ, Michaelson, MD, Redman, BG, Hudes, GR, Wilding, G, Figlin, RA, Ginsberg, MS, Kim, ST, Baum, CM, DePrimo, SE, Li, JZ, Bello, CL, Theuer, CP, George, DJ and Riri, BI. Activity of SU11248, a multitargeted inhibitor of vascular endothelial growth factor receptor and platelet-derived growth factor receptor, in patients with metastatic renal cell carcinoma. J Clin Oncol, 2006, 24: 16-24.
    9 Escudier, B, Eisen, T, Stadler, WM, Szczylik, C, Oudard, S, Siebels, M, Negrier, S, Chevreau, C, Solska, E, Desai, AA, Rolland, F, Demkow, T, Hutson, TE, Gore, M, Freeman, S, Schwartz, B, Shan, M, Simantov, R and Bukowski, RM. Sorafenib in advanced clearcell renal-cell carcinoma. N Engl J Med, 2007, 356: 125-34.
    10 Jubb, AM, Hurwitz, HI, Bai, W, Holmgren, EB, Tobin, P, Guerrero, AS, Kabbinavar, F, Holden, SN, Novotny, WF, Frantz, GD, Hillan, KJ and Koeppen, H. Impact of vascular endothelial growth factor-A expression, tlarombospondin-2 expression, and microvessel density on the treatment effect of bevacizumab in metastatic colorectal cancer. J Clin Oncol, 2006, 24: 217-27.
    11 Jayson, GC, Zweit, J, Jackson, A, Mulatero, C, Julyan, P, Ranson, M, Broughton, L, Wagstaff, J, Hakannson, L, Groenewegen, G, Bailey, J, Smith, N, Hastings, D, Lawrance, J, Haroon, H, Ward, T, McGown, AT, Tang, M, Levitt, D, Marreaud, S, Lehmann, FF, Herold, M and Zwierzina, H. Molecular imaging and biological evaluation of HuMV833 anti, VEGF antibody: implications for trial design of antiangiogenic antibodies. J Natl Cancer last, 2002, 94: 1484-93.
    12 Jubb, AM, Oates, AJ, Holden, S and Koeppen, H. Predicting benefit from antiangiogenic agents in malignancy. Nat Rev Cancer, 2006, 6: 626-35.
    13 Relf, M, LeJeune, S, Scott, PA, Fox, S, Smith, K, Leek, R, Moghaddam, A, Whitehouse, R, Bicknell, R and Harris, AL. Expression of the angiogenic factors vascular endothelial cell growth factor, acidic and basic fibroblast growth factor, tumor growth factor betal, platelet-derived endothelial cell growth factor, placenta growth factor, and pleiotrophin in human primary breast cancer and its relation to angiogenesis. Cancer Res, 1997, 57: 963-9.
    14 Drevs, J, Zirrgiebel, U, Sdmaidt-Gersbach, CI, Mross, K, Medinger, M, Lee, L, Pirheiro, J, Wood, J, Thomas, AL, Unger, C, Henry, A, Steward, WP, Laurent, D, Lebwohl, D, Dugan, M and Marme, D. Soluble markers for the assessment of biological activity with PTK787/ZK 222584 (PTK/ZK), a vascular endothelial growth factor receptor (VEGFR) tyrosine kinase inhibitor in patients with advanced colorectal cancer from two phase Ⅰ trials. Ann Oncol, 2005, 16: 558-65.
    15 Allinen, M, Beroukhim, R, Cai, L, Brennan, C, Lahti-Domenici, J, Huang, H, Porter, D, Hu, M, Chin, L, Richardson, A, Schnitt, S, Sellers, WR and Polyak, K. Molecular characterization of the tumor microenvironment in breast cancer. Cancer Cell, 2004, 6: 17-32.
    16 Miller, KD, Sweeney, CJ and Sledge, GW, Jr. The Snark is a Boojum: the continuing problem of drug resistance in the antiangiogenic era. Ann Oncol, 2003, 14: 20-8.
    17 Schrag, D. The price tag on progress-chemotherapy for colorectal cancer. N Engl J Med, 2004, 351: 317-9.
    18 Hladovec, J. Circulating endothelial cells as a sign of vessel wall lesions. Physiol Bohemoslov, 1978, 27: 140-4.
    19 Shaked, Y, Bertolini, F, Man, S, Rogers, MS, Cervi, D, Foutz, T, Rawn, K, Voskas, D, Dumont, DJ, Ben-David, Y, Lawler, J, Henkin, J, Huber, J, Hicklin, DJ, D'Amato, RJ and Kerbel, RS. Genetic heterogeneity of the vasculogenic phenotype parallels angiogenesis; Implications for cellular surrogate marker analysis of antiangiogenesis. Cancer Cell, 2005, 7: 101-11.
    20 Blann, AD, Woywodt, A, Bertolini, F, Bull, TM, Buyon, JP, Clancy, RM, Haubitz, M, Hebbel, RP, Lip, GY, Mancuso, P, Sampol, J, Solovey, A and Dignat-George, F. Circulating endothelial cells. Biomarker of vascular disease. Thromb Haemost, 2005, 93: 228-35.
    21 Johnson, MR, Wang, K, Smith, JB, Heslin, MJ and Diasio, RB. Ouantitation of dihydropyrimidine dehydrogenase expression by real-time reverse transcription polymerase chain reaction. Anal Biochem, 2000, 278: 175-84.
    22 Capillo, M, Mancuso, P, Gobbi, A, Monestiroli, S, Pruned, G, Dell'Agnola, C, Martinelli, G, Shultz, L and Bertolini, F. Continuous infusion of endostatin inhibits differentiation, mobilization, and clonogenic potential of endothelial cell progenitors. Clin Cancer Res, 2003, 9: 377-82.
    23 Monestiroli, S, Mancuso, P, Burlini, A, Pruneri, G, Dell'Agnola, C, Gobbi, A, Martinelli, G and Bertolini, F. Kinetics and viability of circulating endothelial cells as surrogate angiogenesis marker in an animal model of human lymphoma. Cancer Res, 2001, 61: 4341-4.
    24 Asahara, T, Murohara, T, Sullivan, A, Silver, M, van der Zee, R, Li, T, Witzenbichler, B, Schatteman, G and Isner, JM. Isolation of putative progenitor endothelial cells for angiogenesis. Science, 1997, 275: 964-7.
    25 Asahara, T, Takahashi, T, Masuda, H, Kalka, C, Chen, D, Iwaguro, H, Inai, Y, Silver, M and Isner, JM. VEGF contributes to postnatal neovascularization by mobilizing bone marrow-derived endothelial progenitor cells. Embo J, 1999, 18: 3964-72.
    26 Beaudry, P, Force, J, Naumov, GN, Wang, A, Baker, CH, Ryan, A, Soker, S, Johnson, BE, Folkman, J and Heymach, JV. Differential effects of vascular endothelial growth factor receptor-2 inhibitor ZD6474 on circulating endothelial progenitors and mature circulating endothelial cells: implications for use as a surrogate marker of antiangiogenic activity. Clin Cancer Res, 2005, 11: 3514-22.
    27 Rafii, S, Lyden, D, Benezra, R, Hattori, K and Heissig, B. Vascular and haematopoietic stem cells: novel targets for anti-angiogenesis therapy? Nat Rev Cancer, 2002, 2: 826-35.
    28 Ruzinova, MB, Schoer, RA, Gerald, W, Egan, JE, Pandolfi, PP, Rafii, S, Manova, K, Mittal, V and Benezra, R. Effect of angiogenesis inhibitiou by Id loss and the contribution of bone-marrow-derived endothelial cells in spontaneous routine tumors. Cancer Cell, 2003, 4: 277-89.
    29 Garcia-Barros, M, Paris, F, Cordon-Cardo, C, Lyden, D, Rafii, S, Haimovitz-Friedman, A, Fuks, Z and Kolesnick, R. Tumor response to radiotherapy regulated by endothelial cell apoptosis. Science, 2003, 300: 1155-9.
    30 De Palma, M, Venneri, MA, Roca, C and Naldini, L. Targeting exogenous genes to tumor angiogenesis by transplantation of genetically modified hematopoietic stem cells. Nat Med, 2003, 9: 789-95.
    31 Peters, BA, Diaz, LA, Polyak, K, Meszler, L, Romans, K, Guinan, EC, Antin, JH, Myerson, D, Hamilton, SR, Vogelstein, B, Kinzler, KW and Lengauer, C. Contribution of bone marrow-derived endothelial cells to human tumor vasculature. Nat Med, 2005, 11: 261-2.
    32 Keyes, M, Dudek, A, Jahagirdar, B, Koodie, L, Marker, PH and Verfaillie, CM. Origin of endothelial progenitors in human postnatal bone marrow. J Clin Invest, 2002, 109: 337-46.
    33 Sipkins, DA, Cheresh, DA, Kazemi, MR, Nevin, LM, Bednarski, MD and Li, KC. Detection of tumor angiogenesis in vivo by alpha Vbeta3-targeted magnetic resonance imaging. Nat Med, 1998, 4: 623-6.
    34 Gillies, RJ, Bhujwalla, ZM, Evelhoch, J, Garwood, M, Neeman, M, Robinson, SP, Sotak, CH and Van Der Sanden, B. Applications of magnetic resonance in model systems: tumor biology and physiology. Neoplasia, 2000, 2: 139-51.
    35 Miles, KA. Measurement of tissue perfusion by dynamic computed tomography. Br J Radiol, 1991, 64: 409-12.
    36 Peters, AM, Gunasekera, RD, Henderson, BL, Brown, J, Lavender, JP, De Souza, M, Ash, JM and Gilday, DL. Noninvasive measurement of blood flow and extraction fraction. Nucl Med Commun, 1987, 8: 823-37.
    37 Hamberg, LM, Hunter, GJ, Maynard, KI, Owen, C, Morris, PP, Putman, CM, Ogilvy, C and Gonzalez, RG. Functional CT perfusion imaging in predicting the extent of cerebral infarction from a 3-hour middle cerebral arterial occlusion in a primate stroke model. AJNR Am J Neuroradiol, 2002, 23: 1013-21.
    38 Fuentes, MA, Keith, CJ, Griffiths, M, Durbndge, G and Miles, KA. Hepatic haemodynamics: interrelationships between contrast enhancement and perfusion on CT and Doppler perfusion indices. Br J Radiol, 2002, 75: 17-23.
    39 Blomley, MJ, McBride, A, Mohammedtagi, S, Albrecht, T, Harvey, CJ, Jager, R, Standfield, NJ and Dawson, P. Functional renal perfusion imaging with colour mapping: is it a useful adjunct to spiral CT of in the assessment of abdominal aortic aneurysm (AAA)? Eur J Radiol, 1999, 30: 214-20.
    40 Dugdale, PE, Miles, KA, Bunce, I, Kelley, BB and Leggett, DA. CT measurement of perfusion and permeability within lymphoma masses and its ability to assess grade, activity, and chemotherapeutic response. J Comput Assist Tomogr, 1999, 23: 540-7.
    41 Harvey, CJ, Blomley, MJ, Dawson, P, Morgan, JA, Dooher, A, Deponte, J, Vernon, CC and Price, P. Functional CT imaging of the acute hyperemic response to radiation therapy of the prostate gland: early experience. J Comput Assist Tomogr, 2001, 25: 43-9.
    42 Harvey, C, Morgan, J, Blomley, M, Dooher, A, de Souza, N and Dawson, P. Tumor responses to radiation therapy: use of dynamic contrast material-enhanced CT to monitor functional and anatomical indices. Acad Radiol, 2002, 9 Suppl 1: $215-9.
    43 Schutt, EG, Klein, DH, Mattrey, RM and Riess, JG. Injectable microbubbles as contrast agents for diagnostic ultrasound imaging: the key role of perfluorochemicals. Angew Chem Int Ed Engl, 2003, 42: 3218-35.
    44 Lanza, GM and Wickline, SA. Targeted ultrasonic contrast agents for molecular imaging and therapy. Prog Cardiovasc Dis, 2001, 44: 13-31.
    45 Ogura, O, Takebayashi, Y, Sameshima, T, Maeda, S, Yamada, K, Hata, K, Akiba, S and Aikou T. Preoperative assessment of vascularity by color Doppler ultrasonography in human rectal carcinoma. Dis Colon Rectum, 2001, 44: 538-46; discussion 546-8.
    46 Buadu, LD, Murakami, J, Murayama, S, Hashiguchi, N, Toyoshima, S, Sakai, S, Yabuuchi, H, Masuda, K, Kuroki, S and Ohno, S. Colour Doppler sonography of breast masses: a multiparameter analysis. Clin Radiol, 1997, 52: 917-23.
    47 Lee, WJ, Chu, JS, Houng, S J, Chung, MF, Wang, SM and Chen, KM. Breast cancer angiogenesis: a quantitative morphologic and Doppler imaging study. Ann Surg Oncol, 1995, 2: 246-51.
    48 Sterns, EE, SenGupta, S, Sannders, F and Zee, B. Vascularity demonstrated by Doppler ultrasound and immunohistochemistry in invasive ductal carcinoma of the breast. Breast Cancer Res Treat, 1996, 40: 197-203.
    49 Lamer, S, Sigal, R, Lassau, N, Bosq, J, Frouin, F, Di Paola, M, Mamelle, G, Leclere, J, Bittoun, J and Di Paola, R. Radiologic assessment of intranodal vascularity in head and neck squamous cell carcinoma. Correlation with histologic vascular density. Invest Radiol, 1996, 31: 673-9.
    50 Fleischer, AC, Wojcicki, WE, Donnelly, EF, Pickens, DR, Thirsk, G, Thurman, GB and Hellerqvist, CG. Quantified color Doppler sonography of tumor vascularity in an animal model. J Ultrasound Med, 1999, 18: 547-51.
    51 Taylor, KJ, Ramos, I, Carter, D, Morse, SS, Shower, D and Fortune, K. Correlation of Doppler US tumor signals with neovascular mot phologic features. Radiology, 1988, 166: 57-62.
    52 Forsberg, F, Merton, DA, Liu, JB, Nee dleman, L and Goldberg, BB. Clinical applications of ultrasound contrast agents. Ultrasonics, 1998, 36: 695-701.
    53 Chaudhari, MH, Forsberg, F, Voodarla, A, Saikali, FN, Goonewardene, S, Needleman, L, Finkel, GC and Goldberg, BB. Breast tumor vascularity identified by contrast enhanced ultrasound and pathology: initial results. Ultrasonics, 2000, 38: 105-9.
    54 Reynolds, K, Farzaneh, F, Collins, WP, Campbell, S, Bourne, TH, Lawton, F, Moghaddam, A, Harris, AL and Bicknell, R. Association of ovarian malignancy with expression of platelet-derived endothelial cell growth factor. J Natl Cancer Inst, 1994, 86: 1234-8.
    55 Hata, K, Nagami, H, Iida, K, Miyazaki, K and Collins, WP. Expression of thymidine phosphorylase in malignant ovarian tumors: correhtion with microvessel density and an ultrasound-derived index of angiogenesis. Ultrasound Obstet Gynecol, 1998, 12: 201-6.

© 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