2001 Targeting Tumor Angiogenesis with Gene Therapy
- 格式:pdf
- 大小:75.62 KB
- 文档页数:8
MCPIP1通过调节VEGFA-ERK途径促进胶质瘤细胞的增殖、迁移和血管生成摘要:MCPIP1是一种新型的转录因子,可通过调节多种信号通路影响不同细胞的生长、分化和凋亡等生物学过程。
本研究旨在探讨MCPIP1在胶质瘤细胞增殖、迁移和血管生成中的作用及其机制。
我们发现MCPIP1在人脑胶质瘤组织和细胞系中高表达,过表达MCPIP1可显著促进胶质瘤细胞的增殖和迁移,同时增强VEGFA/ERK信号通路的活性。
进一步实验发现,MCPIP1通过直接与VEGFA基因启动子结合,增强VEGFA的转录和表达,同时通过调节ERK1/2磷酸化水平影响VEGFA信号通路的下游效应,从而促进胶质瘤细胞的血管生成能力。
这些结果表明MCPIP1可能是促进胶质瘤发生和发展的一个新的靶点,有望成为预防和治疗胶质瘤的重要药物靶点。
关键词:胶质瘤,MCPIP1,VEGFA,ERK,血管生成。
Abstract: MCPIP1 is a novel transcription factor that can affect various biological processes such as growth, differentiation, and apoptosis in different cells by regulating multiple signaling pathways. This studyaims to investigate the role and mechanism of MCPIP1in glioma cell proliferation, migration, and angiogenesis. We found that MCPIP1 was highly expressed in human glioma tissues and cell lines. Overexpression of MCPIP1 significantly promoted gliomacell proliferation and migration, while enhancing the activity of the VEGFA/ERK signaling pathway. Further experiments revealed that MCPIP1 directly bound to the VEGFA gene promoter, increased VEGFA transcription and expression, and affected the downstream effects of the VEGFA signaling pathway by regulating the phosphorylation levels of ERK1/2, thereby promotingthe angiogenic ability of glioma cells. These results suggest that MCPIP1 may be a novel target forpromoting glioma occurrence and development, and may become an important drug target for prevention and treatment of glioma.Keywords: Glioma, MCPIP1, VEGFA, ERK, angiogenesisGlioma is a highly invasive and aggressive tumor that remains one of the most challenging diseases to treat. Therefore, there is an urgent need to identify new molecules and pathways that can be targeted to inhibit glioma growth and progression. In recent years, research has focused on the role of MCPIP1 in cancer, including its involvement in angiogenesis, which is a critical process for tumor development and progression.The study found that MCPIP1 promoted angiogenesis in glioma by upregulating the expression of VEGFA. The VEGFA gene is a key regulator of angiogenesis thatpromotes the growth of new blood vessels. Increased VEGFA expression is often observed in various types of tumors and is associated with poor prognosis.The study also revealed that MCPIP1 regulates the downstream effects of the VEGFA signaling pathway by regulating the phosphorylation levels of ERK1/2.ERK1/2 is an important signaling molecule that regulates many cellular processes, including cell proliferation, differentiation, and survival. In glioma, dysregulation of ERK1/2 signaling has been implicated in tumor growth and progression.Overall, these findings suggest that MCPIP1 may be a promising target for anti-angiogenic therapy in glioma. Targeting MCPIP1 could inhibit VEGFA expression and downstream signaling, thus suppressing angiogenesisand inhibiting tumor growth. Further studies are needed to explore the potential of MCPIP1 as a therapeutic target for gliomaIn addition to targeting MCPIP1, other approaches have been explored for anti-angiogenic therapy in glioma. One such approach is inhibition of vascularendothelial growth factor receptor 2 (VEGFR2), whichis the primary receptor for VEGFA. Inhibition of VEGFR2 reduces VEGFA-mediated angiogenesis and hasbeen shown to slow tumor growth in preclinical models of glioma. Clinical trials of VEGFR2 inhibitors, such as bevacizumab, have shown promise in treating recurrent glioblastoma, although there are concerns about resistance to this therapy and its effects on normal brain tissue.Another approach to anti-angiogenic therapy in glioma is targeting the perivascular niche, which is the microenvironment surrounding blood vessels in tumors. The perivascular niche is important for supporting tumor growth and angiogenesis, and has been shown to be associated with therapy resistance and tumor recurrence. Targeting the perivascular niche, either through direct targeting of perivascular cells or via inhibition of signaling pathways that regulate niche formation and maintenance, may be effective in inhibiting tumor growth and improving treatment outcomes.Additionally, some studies have investigated anti-angiogenic therapy in combination with other treatments, such as chemotherapy or immunotherapy. Preclinical studies have shown that combining anti-angiogenic therapy with chemotherapy can enhance tumor response and reduce resistance to chemotherapy. Clinical trials of this approach in glioblastoma haveyielded mixed results, but further investigation is ongoing. Combining anti-angiogenic therapy with immunotherapy, such as immune checkpoint inhibitors,is also being explored as a potential strategy to improve treatment outcomes in glioma.In conclusion, targeting angiogenesis is a promising approach to treating glioma, a highly vascularized and aggressive brain tumor. MCPIP1 is a novel target for anti-angiogenic therapy, and its inhibition may be effective in suppressing angiogenesis and inhibiting tumor growth. However, further studies are needed to fully understand the mechanisms of MCPIP1 in glioma and to explore its potential as a therapeutic target. Other approaches to anti-angiogenic therapy in glioma, including targeting VEGFR2, the perivascular niche, and combination with other treatments, are also being explored and may yield promising resultsIn addition to targeting MCPIP1, there are other promising approaches to anti-angiogenic therapy in glioma that are currently being explored. One such approach involves targeting VEGFR2, a receptor for vascular endothelial growth factor that plays acritical role in tumor angiogenesis. Bevacizumab, a monoclonal antibody that targets VEGF, has been approved for the treatment of recurrent glioblastomaand has shown promise in clinical trials. However, its effectiveness is limited by the development of resistance and the lack of overall survival benefit.Another approach to anti-angiogenic therapy in glioma is to target the perivascular niche, which is a microenvironment around blood vessels that supports tumor growth and development. The perivascular nicheis enriched with stem cells, immune cells, and extracellular matrix components that facilitate tumor cell survival and proliferation. Targeting the perivascular niche may disrupt the tumor microenvironment and improve the efficacy of anti-angiogenic therapy in glioma.Combination therapy is another strategy for enhancing the effectiveness of anti-angiogenic therapy in glioma. For instance, combining anti-angiogenic therapy with immunotherapy may enhance the antitumor immune response and produce a synergistic effect. Preclinical studies have shown that combining bevacizumab with immune checkpoint inhibitors or adoptive T celltherapy can improve survival and reduce tumor growthin glioma models. Similarly, combining anti-angiogenic therapy with chemotherapy or radiation therapy may enhance their cytotoxic effects and improve treatment outcomes.In summary, anti-angiogenic therapy has emerged as a promising strategy for the treatment of glioma, and the identification of novel targets such as MCPIP1 may further improve its efficacy. However, further research is needed to fully understand the mechanisms of tumor angiogenesis and to develop more effective and specific anti-angiogenic agents. Combination therapy and targeted approaches to the perivascular niche are also promising strategies for improving the efficacy of anti-angiogenic therapy in gliomaIn conclusion, anti-angiogenic therapy holds great potential for the treatment of glioma. MCPIP1 is a potential novel target that can improve the efficacy of this therapy. However, more research is necessary to understand tumor angiogenesis mechanisms and develop effective and specific anti-angiogenic agents. Combination therapy and targeted approaches to the perivascular niche are also promising strategies for improving the efficacy of anti-angiogenic therapy。
高危肾细胞癌术前新辅助靶向药物治疗邓建华;纪志刚【摘要】RCC is highly resistant to chemotherapy and radiotherapy,while its response to cytokine therapy is less than 20 %.During recent years,the introduction of new agents targeting tumor angiogenesis and intracellular pathways has dramatically changed the therapeutic approach forRCC.Preoperative neoadjuvant therapy with targeted drugs can minify volume of RCCs to meet the indication of surgery removal.Thus,the survival rate of RCC patients will be significantlyincreased.Furthermore,preoperative neoadjuvant therapy with TKI for high risk RCC patients can significantly decrease the size of tumor thrombus,increasing the efficiency of the nephron-sparing surgery(NSS) or radical nephrectomy.The preoperative neoadjuvant therapy with TKI combined surgery is of great effectiveness in high risk RCC patients among Chinese population.It is also necessary to recognize the limitations of this therapeutic method,more clinical trials are warranted to disclose its potency and efficacy.%肾细胞癌对化疗和放疗均不敏感,对细胞因子治疗的反应率亦不到20%.近年来,针对肿瘤血管生成抑制剂出现极大地改变了肾癌的治疗模式.通过术前新辅助靶向药物治疗高危肾癌的可以使得肿瘤缩小,增加手术切除成功率,提高肾癌患者的生存率.同时可显著降低肾癌瘤栓长度,可减少肿瘤负荷,帮助双侧肾癌及孤立行肾保留肾单位手术(NSS)或根治性切除术安全实施.但也必须认识到这种治疗方法的局限性,增加更多的临床研究获得更加可靠的数据.【期刊名称】《现代泌尿外科杂志》【年(卷),期】2018(023)006【总页数】4页(P405-408)【关键词】肾细胞癌;高危;术前新辅助;酪氨酸激酶抑制剂;术前【作者】邓建华;纪志刚【作者单位】中国医学科学院北京协和医院泌尿外科,北京100730;中国医学科学院北京协和医院泌尿外科,北京100730【正文语种】中文【中图分类】R737新辅助治疗(neoadjuvant chemotherapy,NCT)又称诱导化疗或术前化疗,以及使用靶向化学药物,是恶性肿瘤在局部治疗(手术或放疗)前给予全身化疗。
LBH589对人上皮性卵巢癌OVCAR-3细胞增殖的抑制作用及机制探讨晁宏图;邓君丽;马一鸣;王莉【摘要】目的:探讨新型组蛋白去乙酰化酶抑制剂LBH589对人上皮性卵巢癌OVCAR-3细胞增殖抑制和促进凋亡作用及其机制.方法:不同浓度LBH589处理细胞后,采用噻唑蓝(MTT)比色法检测对细胞增殖的影响;AO/EB(台盼蓝、吖啶橙溴化乙啶双染色法)检测细胞凋亡;Western blot 检测聚腺苷二磷酸核糖聚合酶(PARP)、Caspase-3、Bcl-2、Bax蛋白水平.结果:LBH589明显抑制OVCAR-3细胞增殖,48 h半数抑制浓度(IC50)为0.12μM.Western blot检测发现Caspsae-3、PARP-85kD剪切蛋白增加,Bax表达增加,Bcl-2表达减少.结论:LBH589在体外条件下能明显抑制卵巢癌细胞OVCAR-3细胞增殖,诱导细胞凋亡.%Objective: This study was designed to investigate the mechanism and effect ofLBH589, a novel histone deacetylase inhibitor, on the growth and apoptosis of the human ovarian cancer cell line OVCAR-3. Methods: The cells were treated with LBH589 at different concentrations. Cell proliferation was determined using the methyl thiazol tetrazolium assay. The apoptotic rate of the cells was detected by AO/EB double-staining. Protein expressions of polyADP-ribose polymerase (PARP), caspase-3, bel-2, and bax were analyzed by western blot assay. Results: LBH589 significantly inhibited the proliferation of OVCAR-3 cells at a concentration of 0.12 µM/L based on a 48 h half-inhibitory concentration (IC50). Western blot assay showed that the expressions of cleaved PARP-85KD, caspase-3, and bax were increased, but bcl-2 expression was downregulated.Conclusion: LBH589 in vitro can significantly inhibit the proliferation and induce apoptosis of the human ovarian cancer cell line OVCAR-3.【期刊名称】《中国肿瘤临床》【年(卷),期】2013(000)003【总页数】3页(P131-133)【关键词】LBH589;卵巢癌;细胞增殖;细胞凋亡【作者】晁宏图;邓君丽;马一鸣;王莉【作者单位】河南省肿瘤医院妇瘤科,郑州大学附属肿瘤医院郑州市450003;河南省肿瘤医院妇瘤科,郑州大学附属肿瘤医院郑州市450003;河南省肿瘤医院妇瘤科,郑州大学附属肿瘤医院郑州市450003;河南省肿瘤医院妇瘤科,郑州大学附属肿瘤医院郑州市450003【正文语种】中文上皮性卵巢癌是妇科恶性程度最高的肿瘤,大多数患者发现时已是晚期,多烯紫杉醇与铂类联合是治疗卵巢癌的金标准,但容易产生耐药。
研究肿瘤细胞与内皮细胞代谢中异常的代谢变化及关系- 经典论文Advances in drug development targeting tumor metabolism and the relevance with tumor angiogenesisAbstract:The reprogramming of cellular metabolism is one of the hallmarks of cancer,which is featured as enhanced glycolysis, glutamine metabolism and other biosynthetic activities. Given its crucial role in tumor development, targeting tumor cell metabolism has bee one of the hotspots in the research and development of antitumor drugs. Angiogenesis, a process related to a metabolic shift in endothelial cells, is another hallmark of cancer and supports the metabolic activities in tumor cells. In this article, the abnormal metabolic pathways in tumor cells and tumor endothelial cells were discussed and their relevance was further explored. The advance in therapeutic medicine targeting the metabolic pathways above was also summarized.Keyword:the reprogramming of tumor metabolism; endothelial metabolism; angiogenesis; aerobic glycolysis; glutamine metabolism;肿瘤能量代谢重编程是肿瘤发生发展过程中的一个重要特征,致/抑癌基因的突变及环境压力使肿瘤细胞的代谢网络发生重排,以适应生存与增殖的需要。
多肽修饰靶向给药系统在癌症治疗中的应用赵辰阳;范青【摘要】多肽作为一类重要生物活性物质,具有活性高、低免疫原性、毒性低、易于装载等特点广泛应用于癌症治疗中。
靶向给药系统可将药物选择性浓集定位于靶器官,靶组织,靶细胞中,将小分子多肽修饰于靶向给药系统表面,能在降低传统化疗药物毒副作用的同时提高治疗指数。
本文介绍了包括表皮生长因子专一肽,肿瘤新生血管靶向肽以及细胞穿膜肽等修饰的靶向给药系统在癌症治疗中的应用,表明多肽修饰药物给药系统在癌症治疗中具有很好的临床应用前景。
%As an important class of biologically active substances , peptides have high activity, low immunogenicity, low toxicity, easy to mount, now widely used in the cancer therapy .Targeting drug delivery systems can be selectively concen-trated and positioned in the target organs , the target tissues and the target cells .Use small peptides modified on the surface of targeting drug delivery systems , can reduce the toxic effects of traditional chemotherapy while enhancing the therapeutic index.Within this review, an overview on the applications of these peptides in targeting drug delivery systems is provided . Such as the specific peptides for epidermal growth factor , tumor angiogenesis targeting peptides and cell penetrating pep-tides.It showed that peptides modified targeting drug delivery systems have a good clinical application prospect in cancer therapy.【期刊名称】《大连医科大学学报》【年(卷),期】2014(000)001【总页数】4页(P88-91)【关键词】多肽;靶向;给药系统;表皮生长因子;肿瘤新生血管;细胞穿膜肽【作者】赵辰阳;范青【作者单位】大连医科大学附属第二医院药学部,辽宁大连116027;大连医科大学附属第二医院药学部,辽宁大连116027【正文语种】中文【中图分类】R944.9化疗作为癌症治疗的主要手段之一,因缺乏药理作用的专一性,对患者肿瘤组织及正常组织均造成严重的损伤。
寡糖的生物功能研究进展谢春锋1,王洁2*(1.南开大学药学院,天津300071;2. 中国食品发酵工业研究院,北京100027)摘要:寡糖具有奇特的结构和显著的生物功能,已成为目前糖化学和糖生物学领域的研究热点。
本文综述了近十年寡糖的生理功能研究进展,包括抗微生物、抗氧化、抗肿瘤、新生血管调节和增强免疫力等。
寡糖(oligosaccharides),亦称为寡聚糖或低聚糖,一般由2-10个单糖单元经苷键结合而成(分子量300-2000)。
寡糖常以稳定剂、膨松剂、免疫调节剂或益生素形式用于食品、饲料、药品或化妆品,常见的商品寡糖如低聚果糖、异麦芽低聚糖、麦芽低聚糖、低聚木糖、大豆寡糖、麦芽糖醇和低聚半乳糖等。
寡糖在人体内的生理功能有[1]:(1) 溶解于肠中形成粘胶,降低葡萄糖的吸收,因此不会导致血糖的增加或胰岛素的分泌;(2) 仅供应低量的能量,约为0-3 kcal/g;(3) 非致龋性;(4) 改善肠道内环境,增加有益菌群;(5) 抗腹泻;(6) 促进肠道对钙、镁和铁等矿物质的吸收;(7) 降低患现代“文明病”的发生概率,如心血管疾病、结肠癌等。
本文拟对近十年寡糖的生理功能研究进展作一综述,为研究和开发功能寡糖提供参考和科学依据。
1. 抗菌活性一些甜味食品中常含有易于被口腔致龋菌代谢的单糖或二糖,因此人们一直期望发现安全、低廉、非致龋性的甜味剂。
Seo等[2]以肠膜状明串珠菌(Leuconostoc mesenteroides) B-512 FMCM菌株的右旋糖酐蔗糖酶催化蔗糖(2.5~4 M))合成了耐酸耐热的寡糖(TASO),其不饱和度在2至11之间。
TASO可显著抑制致龋菌茸毛链球菌(Streptococcus sobrinus)的生长,因此可作为食品或饮料中强效的龋病抑制剂。
β-半乳糖基-海藻寡糖是由6-β-半乳糖基-海藻糖和4-β-半乳糖基-海藻糖组成的混合物(9:1,w/w)。
其亦具有抗龋作用,与蔗糖相比,该寡糖作用的茸毛链球菌仅有10%的生长[3]。
细胞内渗(intravasation)本综述由解螺旋学员Ewen负责整理(2017年12月)肿瘤的复发转移是肿瘤致死率高的原因之一,也是当今肿瘤研究需要攻克的难关。
肿瘤的转移是一个多步骤的级联反应,包括肿瘤细胞从原发灶的脱离(Dissemination)、内渗(Intravasation)进入血管、形成循环肿瘤细胞(Circulating tumor cells, CTCs)、外渗(Extravasaton)透过血管、在远处组织或器官形成微小克隆灶(Micrometastatic colonies),最终形成肉眼可见的转移灶1。
其中,内渗是肿瘤细胞进入循环系统至关重要的一步2。
内渗是指细胞通过内皮细胞的基底膜,进入到循环系统中3。
内渗存在于正常机体的生命活动中,比如,当人体被感染时候,处于淋巴组织内的免疫细胞需要通过内渗进入循环系统到达感染部位发挥功能4。
不仅如此,内渗也是肿瘤转移的一个复杂过程。
内渗进入血管有主动和被动两种方式,取决于肿瘤细胞的类型、血管结构和肿瘤细胞的周围微环境。
在肿瘤细胞发生内渗的过程中,肿瘤细胞接受到外部的信号刺激,改变细胞内骨架结构,使自身的运动能力增强,同时肿瘤细胞还会上调整合素和其他黏附分子的表达,协助自身穿过内皮细胞,进入循环系统5, 6。
越来越多的研究证实,导致肿瘤细胞内渗有许多因素,包括信号传递分子、蛋白酶类、和微环境中存在的其他细胞的交互作用等等。
这些因素协助肿瘤细胞突破基底膜,迁移,通过上皮细胞的连接和进入到循环系统中6。
TGFβ是其中一个能促进肿瘤细胞内渗的信号传递分子。
研究表明,TGFβ不仅能促进肿瘤细胞产生PDGFRβ受体,还能通过激活下游信号分子Lhx2,促进肿瘤分泌PDGF-B,形成PDGF-B/PDGFRβ自分泌环路,促进肿瘤细胞的内渗7。
同时,TGF-β激活的CSF1R自分泌环路也能激活肿瘤细胞发生内渗8。
MMPs和uPA/uPAR 都是能够促进肿瘤细胞内渗的蛋白酶类。
肿瘤细胞中乳酸的代谢途径介绍在肿瘤细胞中,乳酸代谢是一个重要的代谢途径。
乳酸产生和乳酸消耗对于肿瘤细胞的生存和增殖具有重要意义。
本文将深入探讨肿瘤细胞中乳酸的代谢途径,包括乳酸的产生、转运和消耗等方面。
乳酸的产生乳酸在肿瘤细胞中主要通过糖酵解途径产生。
当氧气供应不足时,无氧条件下,肿瘤细胞通过糖酵解将葡萄糖分解为乳酸和少量的ATP。
乳酸脱氢酶(LDH)是糖酵解途径中的关键酶,负责将产生的丙酮酸还原为乳酸。
乳酸产生速率的增加与肿瘤细胞的增殖速率呈正相关。
乳酸的转运乳酸产生后,肿瘤细胞需要将其转运出细胞,以维持细胞内酸碱平衡。
乳酸的转运主要依赖于乳酸转运蛋白(Monocarboxylate Transporters,MCTs)。
MCTs是一类跨膜蛋白,可以将乳酸和其它单羧酸共运输。
MCT1和MCT4是肿瘤细胞中最常见的乳酸转运蛋白。
MCT1主要负责乳酸的入细胞转运,而MCT4主要负责乳酸的出细胞转运。
乳酸转运蛋白的表达水平与肿瘤的侵袭性和预后呈相关关系。
乳酸的消耗肿瘤细胞产生的乳酸不仅可以通过转运蛋白转运出细胞,还可以被其他细胞或组织摄取和消耗。
乳酸的消耗主要依赖于MCTs的表达和功能,以及周围组织的代谢状态。
肿瘤周围的血管内皮细胞和平滑肌细胞表达有MCT1和MCT4,可以将乳酸摄取并进行消耗。
此外,肝脏和心脏等器官也可以通过乳酸脱氢酶将乳酸转化为葡萄糖,进行进一步的代谢。
乳酸代谢的调控乳酸代谢在肿瘤细胞中受到多种调控机制的影响。
乳酸转运蛋白的表达受到乳酸浓度、细胞内pH值和氧气浓度等因素的调节。
当肿瘤细胞内乳酸积累过多时,乳酸转运蛋白的表达会上调,以促进乳酸的转运和消耗。
此外,Wnt和HIF-1等信号通路也可以通过调控乳酸代谢相关基因的表达,影响乳酸代谢的速率和方向。
乳酸代谢与肿瘤发展乳酸代谢在肿瘤发展中发挥着重要的作用。
乳酸的积累可以导致肿瘤细胞内部的酸化,破坏细胞内环境平衡,从而促进肿瘤细胞的侵袭和转移。
・590・塞届鼬瘤堂苤查2Q塑生墓望鲞箜鱼翅望筮!坚翅靶向超声微泡在肿瘤诊断和治疗中的研究进展张久维综述程文审校【关键词】超声;靶向微泡;肿瘤【中图分类号】R445.1;I/981【文献标识码】Adoi:10.3969/j.iaan.1002-3070.2009.06.ff28近年来肿瘤的发病率有增高趋势,肿瘤的早期、无创、敏感而特异性的诊断和治疗对于降低死亡率及并发症的发生率至关重要。
近年研究发现,靶向超声微泡造影剂有望成为一种新的安全有效的载体…,从分子水平上介导肿瘤的靶向诊断和治疗。
靶向超声微泡是利用微泡表面特有的生物学性质或通过特殊处理将靶向配体与微泡表面连接,使其能够持续、定向地蓄积于靶组织,实现靶向超声分子成像与靶向治疗【2’3J。
靶向超声微泡经外周静脉注入后能靶向汇聚并较长时间滞留于靶组织或靶器官中,不但可达到从分子水平上行肿瘤显像和治疗的目的,且靶向超声微泡的出现弥补了全身使用微泡造影剂时,聚集于局部组织的微泡浓度低、微泡使用剂量大、花费高的缺憾。
最新的研究显示了靶向微泡在肿瘤诊断和治疗方面诱人的前景。
靶向微泡不仅可提高检测肿瘤的特异性和敏感性,同时可以靶向治疗肿瘤,但也存在很多问题亟待解决。
现就应用靶向超声微泡进行肿瘤显像及靶向微泡联合超声辐照进行肿瘤治疗的研究进展做一简要综述。
1肿瘤靶向性配体的选择及靶向超声微泡构建策略靶向超声微泡制备的关键是选择合适的配体,并选择最合适的修饰顺序、以最适宜的连接方法将该配体结合在普通微泡表面,从而成功制备出具有高效特舁性靶向结合能力的微泡。
肿瘤生长有很多特异性的标志,肿瘤的新生血管内皮表达大量的生长因子受体和黏附分子受体家族,靶向微泡造影剂就是根据这些特异性标志和血管内皮细胞受体构建而成。
因此,靶向肿瘤微泡的构建策略就是将能特异性识别这些生长因子受体和整合素受体的配体(抗体、肽类和维生素等)连接在微泡表面,从而制备出特异性靶向肿瘤的超声造影剂HJ,实现肿瘤的靶向分子成像。
MINIREVIEWTargeting Tumor Angiogenesis with Gene Therapy 1Qing-Rong Chen,Lei Zhang,Warren Gasper,and A.James Mixson 2Department of Pathology,University of Maryland at Baltimore,Baltimore,Maryland 21201Received June 22,2001,and revised form July 17,2001;published online September 17,2001A recent target of cancer gene therapy is tumor angiogenesis.An appealing feature of gene therapy targeting the tumor vasculature is that it is readily accessible,particularly when the carrier and its gene are administered systemically.Several gene-based viral and nonviral therapies that target tu-mor angiogenesis have demonstrated the “proof of principle”of antiangiogenic therapy in preclinical models.The utility of antiangiogenic gene therapy in a clinical setting will depend in large part on developing vectors with minimal toxicity and with increased in vivo transfection efficiency.In this re-view,we discuss the current status and future di-rections of antiangiogenic gene therapy.©2001Academic PressKey Words:gene therapy;antiangiogenesis;tu-mor;endothelial cells;VEGF;VEGFR2;angiopoietin 2;nonviral;viral;electroporation.Angiogenesis is a closely regulated physiological process.The human vasculature system of the aver-age person has more than a trillion endothelial cells (1).The lifetime of these normally quiescent cells in vessels surpasses 1000days (2),but once angiogen-esis is induced,endothelial cells can proliferate rap-idly.Except for the corpus luteum,most angiogene-sis in adults occurs in response to pathological insults from wounds or hypoxia (e.g.,stroke and tumors).Tumor growth is dependent on the development of new blood vessels (3).Once the diameter of the tu-mor exceeds 2mm,angiogenesis is required for fur-ther growth (4).Suppression of angiogenic inhibitors together with stimulation of angiogenic growth fac-tors is essential for tumor growth and progression (Fig.1).As a result,therapy targeting tumor vessels attempts either to reduce angiogenic growth factors or to increase angiogenic inhibitors within a tumor (5).Several antiangiogenic proteins/peptides have been shown to be effective in reducing tumor growth.Of particular note,angiostatin and endosta-tin have eliminated tumors in mice completely (6,7).In addition to its antitumor efficacy,an appealing feature of antiangiogenic therapy is that it may avoid the problem of acquired resistance to antican-cer drugs since endothelial cells of tumors have rel-ative genomic stability (8,9).Gene therapy is an attractive means of delivery-ing these antiangiogenic proteins to the tumor site.For most gene therapy approaches,the tumor vas-culature represents an obstacle for therapy to reach tumor cells.The tumor vessels,however,are readily accessible targets for antiangiogenic gene therapy,particularly when it is administered systemically.There are now many studies demonstrating some efficacy of antiangiogenic gene therapy in tumor-bearing animal models.Investigators have success-fully inhibited tumor growth by gene therapy ap-proaches designed to inhibit angiogenic inducers (VEGF,angiopoietin 2)and/or their receptors (10–13).Furthermore,gene therapy with angiogenic in-hibitors has also shown promise with tumor inhibi-tion in animal models (14–16)(Fig.2).Nevertheless,1Supported by the National Institutes of Health (CA70394).2To whom request for reprints should be addressed at Depart-ment of Pathology,University of Maryland at Baltimore,Room 7-59,Building-MSTF,10S.Pine Street,Baltimore,MD 21201.Fax:(410)706-8414.Molecular Genetics and Metabolism 74,120–127(2001)doi:10.1006/mgme.2001.3223,available online at on1201096-7192/01$35.00Copyright ©2001by Academic PressAll rights of reproduction in any form reserved.in vivo gene delivery carriers need improvement before these therapies are clinically useful.Although antiangiogenic gene therapy is an at-tractive gene therapy approach,is this approach more effective than directly injecting the peptide/protein?Several studies with recombinant antian-giogenic proteins have demonstrated marked re-gression of large tumors (6,7).Similar findings,to date,have not been reported with antiangiogenic gene therapy.Nevertheless,there are potential ad-vantages of gene delivery systems for these antian-giogenic proteins.First,gene therapy delivery sys-tems may circumvent the problems of adequate production of certain antiangiogenic proteins (e.g.,angiostatin or endostatin).Adequate production of these proteins by recombinant engineering methods has been problematic and may limit widespread ad-ministration of these proteins.Second,gene therapy can deliver the expressed proteins to the nucleus,cytosol,membrane,or extracellular space.With cur-rent technology,certain antiangiogenic proteins (e.g.,the transmembrane protein,VEGFR2-DN)are only effective if delivered by a gene therapy ap-proach.Thus,gene therapy expands the repertoire of proteins that can be utilized for antiangiogenic therapy.Third,if side effects from the antiangio-genic therapy occur,targeting the therapy to the tumor vessels would be critical.Although significant issues remain to be addressed concerning specificity of ligands for tumor vessels,targeting the tumor vessels with a gene delivery system is much more practical than peptide delivery systems.Because of these problems of protein delivery systems coupled with the improvement of gene delivery systems,we believe that continued investment of time and effort is warranted for the antiangiogenic gene therapy approach.Most studies of antiangiogenic gene therapy have shown their efficacy in preclinical models.Only p53and IL-12,two proteins with multiple tumor inhib-itory mechanisms in addition to antiangiogenesis,have progressed to clinical trial.In subsequent sec-tions,we review the various gene therapy targets and/or the genes encoding antiangiogenic proteins that investigators have used.VEGFVEGF and its cognate receptors (flt-1,or VEGFR1,flk-1or VEGFR2)are essential for tumor angiogen-esis.As a result,it is not surprising that these have frequently been targeted to limit tumor growth.An early study showing the utility ofantiangiogenicFIG.1.The endogenous regulators of tumor angiogenesis:The relative balance of activators and inhibitors of angiogenesis is important to maintain tumors in a quiescent state.Either reducing the inhibitor concentration or increasing the activator level can alter the balance and lead to the tumor angiogenesis and tumor growth.121TUMOR ANGIOGENESIS AND GENE THERAPYgene therapy targeted the VEGF receptor (VEGFR2).In this investigation,injection of a ret-rovirus-truncated VEGFR2construct together with tumor cells resulted in the inhibition of tumor growth (10).It has been suggested that this retrovi-rus-transduced receptor acts through a dominant-negative mechanism to inhibit the wild-type VEGF receptor.This study not only illustrated the poten-tial of this gene therapy approach but also confirmed the essential role of VEGF in tumor development.More recently,antiangiogenic therapy with the truncated VEGF receptor has been shown to sup-press angiogenesis and growth of an intracerebral glioma in a rat model (17).Consequently,rats with these intracerebral tumors survived significantly longer with treatment.In these two studies,the truncated VEGFR2inactivated VEGF-mediated sig-nal transduction by dimerizing with the wild-type receptor of the transfected cell.However,the clinical utility of this method may be limited owing to the low transfection efficiencies of the vector delivery systems injected directly into a tumor.One ap-proach to circumvent the low transfection efficiency of vector delivery systems is to engineer a secreted therapeutic protein.This was demonstrated in a mouse model where tumor cells transduced with a secreted soluble VEGF receptor (sVEGFR1)inhib-ited implantation of a primary tumor (13).Further-more,the mice displayed fewer lung metastases when tumor cells expressing elevated levels of sVEGFR1were injected intravenously.Alternatively,antisense strategies targetingVEGF or its receptor have shown efficacy in vivo .For example,stable transfection of tumor cells with plasmids expressing antisense VEGF did not affect the growth of tumor cells in vitro,but reduced tumor growth in vivo (11).Targeting VEGF with a retrovi-rus-producing antisense RNA also prolonged sur-vival in a glioma-bearing rat model (18).VEGF lev-els may also be decreased indirectly through inhibition of the adenosine 2B receptor with an an-tisense oligonucleotide (19).The major difficulty of targeting VEGF in vivo with antisense approaches either directly or indirectly is that if most of the VEGF is expressed in tumor cells and not in the tumor endothelial cells,it may be difficult to achieve adequate expression of the antisense construct.Re-cently,a ribozyme targeting VEGFR1,upregulated in mitogenic endothelial cells,has been reported to have significant antitumor activity in animal models (20).Angiopoietin 2In addition to VEGF,other ligands with receptors located specifically on endothelial cells have been targeted with antiangiogenic gene therapy.In-creased levels of angiopoietin 2,the ligand to the Tie2receptor,in concert with VEGF have been as-sociated with increased tumor angiogenesis.As a result,the ligand angiopoietin 2is a reasonable therapeutic target.An adenovirus expressing a sol-uble Tie 2receptor was recently shown to inhibit tumor growth by binding to angiopoietin 2when injected intravenously (12).With plasmaconcentra-FIG.2.Approaches of antiangiogenic gene therapy:Antiangiogenic gene therapy approaches can be categorized into those that suppress the angiogenic activators and those that enhance the angiogenic inhibitors.122CHEN ET AL.tions of the soluble recombinant Tie2receptor ex-ceeding1mg/ml for an8-day period in this study, established tumors treated by the intravenous in-jectable route were significantly reduced.In addi-tion,coadministration of the adenovirus-Tie2vector with tumor cells greatly reduced the number of lung metastases.A potential disadvantage of this ther-apy is that the adenovirus-Tie2construct may also inactivate angiopoietin1,a protein known primarily for its ability to stabilize blood vessels.CytokinesAlthough earlier in vivo antiangiogenic gene ther-apy experiments had demonstrated antitumor effi-cacy,gene therapy with the angiogenic inhibitor, platelet factor4(PF-4),was thefirst to show efficacy in a clinically relevant model.In this study,an ad-enovirus expressing PF-4(21)was injected into an established tumor and found to inhibit tumor growth of gliomas and prolong animal survival.PF-4 belongs to a family of C-X-C chemokines and may inhibit angiogenesis by altering the affinity of VEGF to its receptors(22).In addition to PF-4,other cytokines including IL-12and IP-10inhibit tumor growth by an antian-giogenic mechanism(23,24).IL-12can inhibit angio-genesis and reduce tumor growth(25),and this an-giostatic response is mediated by interferon-␥(IFN-␥)and IP-10(26,27).Mice receiving IL-12gene therapy were found to survive significantly longer (median survival,43days)than mice treated with control plasmid/lipid complexes(median survival of 35days).These data demonstrated that a nonviral IL-12gene therapy with a cationic liposome carrier could inhibit the development of lung metastases (28).IL-12may mediate its antiangiogenic effect through immune effector cells(29).In addition to its angiostatic mechanism of action,IL-12probably in-hibits tumor growth by several other mechanisms, and it is difficult to distinguish the relative roles of these mechanisms in reducing tumor growth. Angiostatin and EndostatinUnlike the inhibitors that target VEGF and its receptors,the specific molecular targets for the an-giogenic inhibitors,angiostatin and endostatin, have not been clearly identified.Nevertheless,pre-liminary data suggest that tropomyosin binds to endostatin and plays a role in its tumor inhibitory function(30),and it has recently been found that angiostatin binds tightly to an ATP synthase on the endothelial cell surface(31).Both angiostatin and endostatin polypeptides markedly inhibited a vari-ety of tumors in animal models(6,7).Moreover, there has been no evidence of resistance developing to endostatin in these tumor-bearing mice.Identifi-cation of the receptors for endostatin and angiosta-tin may lead to more potent antiangiogenic analogs. Several gene therapy studies with angiostatin and/or endostatin have shown antitumor efficacy in animal models(15,16,32–38).For the most part,pro-teins encoded by genes have been delivered intratu-morally or intravenously by adenoviruses to inhibit tumor growth.However,nonviral delivery via poly-vinylpyrrolidione(PVP),liposomes,or“naked”DNA injection has also demonstrated efficacy.With“na-ked”DNA intratumoral injections of endostatin not only were the tumors decreased as expected but interestingly,VEGF levels increased intratumor-ally.Thus,there appeared to be a compensatory increase in VEGF levels when endostatin was ad-ministered in this manner.Most of these preclinical studies have reported no toxicity with delivery of these peptides by gene therapy.In one study,how-ever,an adenoviral construct expressing endostatin or a fusion IgG/endostatin protein at high doses was associated with severe toxicity in recipient mice, including loss of weight,bleeding,and death(38). Although toxicity was not observed with injection of a control adenovirus that did not contain the en-dostatin,a control adenovirus expressing a protein other than endostatin was not included in these studies.It is possible that nonviral vectors such as cationic liposomes administered intravenously can achieve higher selectivity and less toxicity than their viral counterparts.In one preclinical study with cationic liposomes as a carrier,moderate reduction in the tumor size of mice bearing MDA-MB-435cells was observed after the third injection(15).Thurston et al.demonstrated that liposome:DNA complexes se-lectively target the mitogenic tumor endothelial cells rather than quiescent endothelial cells(39). Adding ligands that specifically target the tumor endothelial cells may further increase the specificity and efficacy of cationic liposomes(40,41).An alternative gene therapy strategy for increas-ing angiostatin levels in tumors was recently dem-onstrated by administering the gene encoding pan-creatic elastase I(42).Endothelial cells transduced with elastase showed marked growth inhibition in vitro,but the growth of transduced Lewis lung car-cinoma cells in vitro was not inhibited.However,123TUMOR ANGIOGENESIS AND GENE THERAPYLewis lung cells transduced with elastase and then implanted into mice showed reduced growth com-pared to controls.Increased levels of the antiangio-genic angiostatin,a breakdown product of plasmin-ogen,is the putative mechanism of elastase gene therapy.The Tumor Suppressor,p53In contrast to the preceding angiogenic inhibitors, which are secreted,p53is a nuclear transcriptional protein.Nevertheless,p53may induce secreted an-tiangiogenic proteins or may act directly with the cell cycle of transfected cells(e.g.,endothelial cells). Gene therapy with p53is one of the few gene ther-apy approaches showing efficacy in humans,but initially the antiangiogenic mechanism of p53was not considered(43).In one study,a retroviral vector containing the wild-type p53gene was used to trans-fer p53into human nonsmall cell lung cancers by direct injection(43).Of the nine patients with widely metastatic disease who were treated,tumors regressed in three patients and stabilized in two other patients.Some injected tumors regressed com-pletely despite a transfection efficiency of only20%. Thus,the authors concluded that apoptosis or cell cycle inhibition of the transfected tumor cell could not account for the amount of tumor inhibition ob-served.As a result,it is likely that there may be addi-tional mechanisms to explain tumor inhibition with p53gene therapy.A bystander effect has been sug-gested for the observed difference between the rela-tively low transfection efficiency of the tumors with p53and the reduction of tumor growth,and this effect appears to be due in part to inhibition of angiogenesis(33,44–46).We determined that lipo-somes in complex with a plasmid encoding p53in-hibited tumor growth in a mouse model(47),and subsequently found that blood vessel density was reduced with this therapy(44).Other studies with liposomes carrying p53plasmids have also shown reduction in tumor growth in mouse models (33,48,49).Besides liposomes,adenoviral and retro-viral carriers expressing the p53gene significantly reduced tumor growth and blood vessel density (45,46).Moreover,adenoviral vectors expressing p53significantly reduced VEGF levels within tu-mors(45).P53may also decrease angiogenesis by inhibiting the cell cycle and/or enhancing apoptosis of the endothelial cells(44),inhibiting differentia-tion of endothelial cells(46),or by inducing the antiangiogenic protein,thrombospondin1(33,50).ThrombospondinDameron et al.(51)first identified the binding site on the promoter of thrombospondin by which p53 induces thrombospondin,a secreted protein that po-tently inhibits angiogenesis.Similar to its inducer p53,thrombospondin can be an important prognos-tic indicator.Decreased thrombospondin-1(TSP-1) secreted by a variety of cell lines correlates with a more malignant phenotype.Indeed,the MDA-MB-435cell line,which is a very aggressive and meta-static breast cancer line in nude mice,secretes low levels of thrombospondin1when compared to less malignant breast cell lines tested(52).The antian-giogenic effects of TSP-1are mediated by the CD36 surface receptor.In vitro experiments in which TSP-1was used to reduce endothelial cell growth gave conflicting results.Nonetheless,peptides iso-lated from TSP-1clearly have antiangiogenic effects in vitro(53),but surprisingly,there have been no published reports demonstrating that these natural peptides have antitumor activity in vivo.It is un-clear if the reduced functional activity of these pep-tides in vivo is due to a short half-life.However, synthetic antiangiogenic thrombospondin peptides in the D-conformation do exhibit in vivo antitumor activity(54).In contrast to peptide therapy,three studies indi-cated that antiangiogenic gene therapy with TSP-1 or peptides derived from TSP-1has antitumor activ-ity(33,55–57).Initially,an ex vivo study demon-strated that a tumor clone expressing high levels of TSP-1peptide reduced tumor growth(55).Recently, an ex vivo study showed that tumor clones express-ing thrombospondin-2(TSP-2)had significantly more antitumor efficacy than clones expressing TSP-1.More impressively,tumor clones expressing TSP-1and-2were synergistic in their antitumor efficacy as demonstrated by the complete inhibition of tumor growth(57).In addition,p53and an anti-angiogenic peptide of TSP-1in complex with lipo-somes were found to be synergistic in their inhibi-tion of tumors(56).These liposome:DNA complexes were delivered intravenously at a site distant from the tumor.Tissue Inhibitory Metalloproteinases(TIMPS) Unlike the previously noted therapies,in vivo gene therapy with TIMPS has not been reported (58,59).In one study,adenovirus-mediated overex-pression of TIMP-1,2,and3inhibited invasion of several different tumor cells through an artificial124CHEN ET AL.basement membrane(58).Nevertheless,only TIMP-3promoted apoptosis,suggesting that this factor may be more effective in reducing tumor growth than either TIMP-1or TIMP-2.Potential Gene Therapy Inhibitors of Angiogenesis There are several angiogenic inhibitors that may be useful with in vivo gene therapy delivery sys-tems.For example,the relaxed form of antithrombin III has demonstrated marked antitumor efficacy but gene therapy with this protein has not yet been attempted.In addition,the kringle5domain of plas-minogen has also been found to be a potent inhibitor of angiogenesis and to be synergistic with angiosta-tin in reducing tumor growth(60–62);similar to antithrombin III,gene therapy with kringle5show-ing antitumor efficacy has not been reported.An-other promising angiogenic inhibitor,PEDF,has not yet demonstrated its antitumor efficacy by either the gene or protein therapy approaches.Pitfalls and Future GoalsSeveral studies have now shown that tumor inhi-bition occurs with antiangiogenic gene therapy. Nevertheless,little is known about the relative an-titumor efficacy of the genes encoding antiangio-genic proteins/peptides.Although comparison stud-ies of antitumor efficacy have been done with some antiangiogenic proteins,the results of protein and gene therapy may diverge because of differences in distribution,half-life,and specificity.Thus,al-though perhaps useful as an initial guide,sole reli-ance on antitumor efficacy of the protein for selec-tion of the appropriate gene may introduce an incorrect bias.Furthermore,it is evident that more studies are required to examine synergistic tumor inhibition between various combinations of genes encoding antiangiogenic proteins.In this context, studies examining the antiangiogenic mechanism of these proteins may facilitate determination of po-tent synergistic combinations.As with other gene therapy approaches,a signifi-cant limitation of antiangiogenic gene therapy lies in the vector delivery systems.Although there has been incremental improvement in these systems, precisely when an adequate vector system will be developed for antiangiogenic genes is difficult to forecast.Since proteins of antiangiogenic gene ther-apy may require secretion,they may be expressed by several tissues other than tumor endothelial cells. Consequently,secretion of antiangiogenic proteins allows the route of gene delivery to be varied,and intravenous,intratumoral,and intramuscular injec-tion have been used.In animal models,all three modes of delivery have demonstrated antitumor ef-ficacy with antiangiogenic genes.Whereas platform deliveries with AAV or electroporation offer the hope of high levels of systemic angiostatic proteins,there is concern that such angiostatic proteins may inter-rupt critical angiogenic processes.In contrast,deliv-ery of the angiostatic gene by intratumoral injec-tions or by tumor-specific carriers may circumvent these concerns.In addition to determining more potent vectors and angiogenic inhibitors,new approaches are on the horizon for antiangiogenic gene therapy.Vac-cines toward tumor angiogenesis are now being de-veloped with partially purified proteins or whole cell endothelial extracts from tumors.Results in preclin-ical models have been encouraging and success of this active immunization will depend on specificity toward tumor angiogenesis(63).A recent study showed that nearly80genes were increased in tu-mor endothelial cells compared to quiescent endo-thelial cells(64).Nevertheless,all of these genes (with the possible exception of tissue endothelial marker8)were also found to be expressed in endo-thelial cells involved in wound healing or the corpus luteum(64).Although the advantages of protein versus gene delivery of antiangiogenic vaccines will need to be ascertained,antiangiogenic vaccines offer new therapeutic opportunities for targeting tumor angiogenesis.REFERENCES1.Jaffe EA.Cell biology of endothelial cells.Hum Pathol18:234–239,1987.2.Denekamp J.Review article:Angiogenesis,neovascular pro-liferation and vascular pathophysiology as targets for cancer therapy.Br J Radiol66:181–196,1993.3.Folkman J.Tumor angiogenesis:Therapeutic implications.N Eng J Med333:1757–1763,1971.4.Folkman J.The vasculization of tumors.Sci Am234:58–73,1976.5.Folkman J.Antiangiogenic gene therapy.Proc Natl Acad SciUSA95:9064–9066,1998.6.O’Reilly MS,Holmgren L,Chen C,Folkman J.Angiostatininduces and sustains dormancy of human primary tumors in mice.Nature Med2:689–692,1996.7.O’Reilly MS,Boehm T,Shing Y,Fukai N,Vasios G,LaneWS,Flynn E,Birkhead JR,Olsen BR,Folkman J.Endosta-tin:An endogenous inhibitor of angiogenesis and tumor growth.Cell88:277–285,1997.8.Kerbel RS.Inhibition of tumor angiogenesis as a strategy to125TUMOR ANGIOGENESIS AND GENE THERAPYcircumvent acquired resistance to anticancer therapeutic agents.Bioessays13:31–36,1991.9.Boehm T,Folkman J,Browder T,O’Reilly MS.Antiangio-genic therapy of experimental cancer does not induce ac-quired drug resistance.Nature390:404–407,1997.lauer B,Shawver LK,Plate KH,Risau W,Ullrich A.Glioblastoma growth inhibited in vivo by a dominant-nega-tive Flk-1mutant.Nature367:576–579,1994.11.Saleh M,Stacker SA,Wilk AF.Inhibition of growth of C6glioma cells in vivo by expression of antisense vascular endothelial growth factor sequence.Cancer Res56:383–401, 1996.12.Lin P,Buxton JA,Acheson A,Radziejewski C,MaisonpierrePC,Yancopoulos GD,Channon KM,Hale LP,Dewhirst MW, George SE,Peters KG.Antiangiogenic gene therapy target-ing the endothelium-specific receptor tyrosine kinase Tie2.Proc Natl Acad Sci USA95:8829–8834,1998.13.Goldman CK,Kendall FL,Cabrera G,Soroceanu L,Heike Y,Gillespie GY,Siegel GP,Mao X,Bett AJ,Huckle WR, Thomas KA,Curiel DT.Paracrine expression of a native soluble vascular endothelial growth factor receptor inhibits tumor growth,metastasis,and mortality rate.Proc Natl Acad Sci USA95:8795–8800,1998.14.Griscelli F,Li H,Griscelli AB,Soria J,Opolon P.Soria C,Perricaudet M,Yeh P,Lu H.Angiostatin gene transfer: Inhibition of tumor growth in vivo by blockage of endothelial cell proliferation associated with a mitosis arrest.Proc Natl Acad Sci USA95:6367–6372,1998.15.Chen QR,Kumar D,Stass SA,Mixson AJ.Liposomes com-plexed to plasmids encoding angiostatin and endostatin in-hibit breast cancer in nude mice.Cancer Res59:3308–3312, 1999.16.Blezinger P,Wang J,Gondo M,Quezada A,Mehrens D,French M,Singhai A,Sullivan S,Rolland A,Ralston R,Min W.Systemic inhibition of tumor growth and tumor metas-tases by intramuscular administration of the endostatin gene.Nature Biotechnol17:623–632,1999.17.Machein MR,Risau W,Plate KH.Antiangiogenic gene ther-apy in a rat glioma model using a dominant-negative vas-cular endothelial growth factor receptor2.Hum Gene Ther 10:1117–1128,1999.18.Sasaki M,Wizigmann-Voos S,Risau W,Plate KH.Retrovi-rus producer cells encoding antisense VEGF prolong sur-vival of rats with intracranial GS9L gliomas.Int J Dev Neurosci17:579–591,1999.19.Grant MB,Tarnuzzer RW,Caballero S,Ozeck MJ,DavisMI,Spoerri PE,Feoktistov I,Biaggioni I,Shryock JC,Be-lardinelli L.Adenosine receptor activation induces vascular endothelial growth factor in human retinal endothelial cells.Circ Res85:699–706,1999.20.Weng DE,Usman N.Angiozyme:A novel angiogenesis in-hibitor.Curr Oncol Rep3:141–146,2001.21.Tanaka T,Manome Y,Wen P,Kufe DW,Fine HA.Viralvector-mediated transduction of a modified platelet factor4 cDNA inhibits angiogenesis and tumor growth.Nature Med 3:437–442,1997.22.Gengrinovitch S,Greenberg SM,Cohen T,Gitay-Goren H,Rockwell P,Maione TE,Neufeld G.Platelet factor-4inhibits the mitogenic activity of VEGF121and VEGF165usingseveral concurrent mechanisms.J Biol Chem270:15059–15065,1995.23.Boggio K,Di Carlo E,Rovero S,Cavallo F,Quaglino E,Lollini PL,Nanni P,Nicoletti G,Wolf S,Musiani P,Forni G.Ability of systemic interleukin-12to hamper progressive stages of mammary carcinogenesis in HER2/neu transgenic mice.Cancer Res60:359–364,2000.24.Sharpe RJ,Byers HR,Scott CF,Bauer SI,Maione TE.Growth inhibition of murine melanoma and human colon carcinoma by recombinant human platelet factor4.J Natl Cancer Inst82:848–853,1990.25.Voest EE,Kenyon BM,O’Reilly MS,D’Amato RJ,FolkmanJ.Inhibition of angiogenesis in vivo by interleukin12.J Natl Cancer Inst87:581–586,1995.26.Sgadari C,Angiolillo AL,Tosato G.Inhibition of angiogen-esis by interleukin-12is mediated by the interferon-induc-ible protein10.Blood87:3877–3882,1996.27.Angiolillo AL,Sgadari C,Tosato G.A role for the interferon-inducible protein10in inhibition of angiogenesis by inter-leukin-12.Ann N Y Acad Sci795:158–167,1996.28.Blezinger P,Freimark BD,Matar M,Wilson E,Singhal A,Min W,Nordstrom JL,Pericle F.Intratracheal administra-tion of interleukin12plasmid-cationic lipid complexes in-hibits murine lung metastases.Hum Gene Ther10:723–731, 1999.29.Yao L,Sgadari C,Furuke K,Bloom ET,Teruya-Feldstein J,Tosato G.Contribution of natural killer cells to inhibition of angiogenesis by interleukin-12.Blood93:1612–1621,1999.30.MacDonald NJ,Shivers WY,Plum SM,Sim BKL.Endosta-tin proteins binds tropomyosin:Potential for modulation of the anti-angiogenic activity of Endostatin protein.Presen-tations at the American Association of Cancer Research Conference on Molecular Targets and Cancer Therapeutics, 1999.31.Moser TL,Stack MS,Asplin I,Enghild JJ,Hojrup P,EverittL,Hubchak S,Schnaper HW,Pizzo SV.Angiostatin binds ATP synthase on the surface of human endothelial cells.Proc Natl Acad Sci USA96:2811–2816,1999.32.Cao Y,O’Reilly,MS,Marshall B,Flynn E,Ji RW,FolkmanJ.Expression of angiostatin cDNA in a murinefibrosarcoma suppresses primary tumor growth and produces long-term dormancy of metastases.J Clin Invest101:1055–1063,1998.33.Liu Y,Thor A,Shtivelman F,Cao Y,Tu G,Heath TD,DebsRJ.Systemic gene delivery expands the repertoire of effec-tive antiangiogenic agents.J Biol Chem274:13338–13344, 1999.34.Ding I,Sun JZ,Fenton B,Liu WM,Kimsely P,Okunieff P,Min W.Intratumoral administration of endostatin plasmid inhibits vascular growth and perfusion in MCa-4murine mammary carcinomas.Cancer Res61:526–531,2001.35.Feldman AL,Restifo NP,Alexander HR,Bartlett DL,HwuP,Seth P,Libutti SK.Antiangiogenic gene therapy of cancer utilizing a recombinant adenovirus to elevate systemic en-dostatin levels in mice.Cancer Res60:1503–1506,2000. 36.Regulier E,Paul S,Marigliano M,Kintz J,Poitevin Y,Ledoux C,Roecklin D,Cauet G,Calenda V,Homann HE.Adenovirus-mediated delivery of antiangiogenic genes as an antitumor approach.Cancer Gene Ther8:45–54,2001. 37.Sauter BV,Martinet O,Zhang WJ,Mandeli J,Woo SL.126CHEN ET AL.。