基于Endoglin靶大鼠胶质瘤的磁共振分子影像学实验研究
详细信息    本馆镜像全文|  推荐本文 |  |   获取CNKI官网全文
摘要
目的:合成制备基于Endoglin靶的空间稳定脂质体磁共振分子探针并评价其理化性质;各种对比剂大鼠胶质瘤磁共振分子影像学研究,验证探针特异性显示胶质瘤形态及边界的有效性。
     材料与方法:一)空间稳定脂质体的制备:(1)脂质体膜材料,二硬脂酰磷脂酰胆碱(DSPC).胆固醇(CHOL).甲氧基聚乙二醇-二硬脂酰乙醇胺(mPEG-DSPE).钆-二乙三胺五醋酸-二硬脂酰胺(Gd-DTPA-BSA)按36:36:3:25摩尔比例溶于氯仿甲醇混合溶剂(2:1容积)中,经40℃旋转蒸发,60℃水化,高压均质仪挤压过膜,制备包载顺磁性物质(Gd)的空间稳定脂质体(Gd-SLs);(2)评价脂质体的粒径、稳定性、钆含量、磁共振驰豫率及体外MR成像作用等性质。二)基于Endoglin靶的空间稳定免疫脂质体及预定位脂质体分子探针的制备:(1)Gd-SLs成膜过程中,加入膜材料吡啶二硫丙酰基-聚乙二醇(2000)-二硬脂酰乙醇胺(PDP-PEG(2000)-DSPE),制备表面修饰结合PDP基团脂质体(PDP-SLs), PDP-SLs脂质体经二硫苏糖醇(DTT)还原,得表面结合游离巯基的脂质体(HS-SLs):(2)CD105单克隆抗体(CD105~MAb)与丁二酰亚胺-4-(对-马来酰亚胺-苯基)-丁酸酯(SMPB)反应制备马来酰亚胺苯基丁酰基单克隆抗体(MPB-MAb);(3)将HS-SLs与MPB-MAb反应获得的表面连接抗体的空间稳定脂质体(MAb-SLs);(4)生物素氨基己糖酸-3-磺酸基-N-羟基琥珀酰亚胺酯(Sulfo-NHS-LC-biotin)与CD105-MAb反应可得生物素化单克隆抗体(Bio-MAb);(5)链霉亲和素结合空间稳定脂质体(SAv-SLs)的制备过程与MAb-SLs基本相同,将HS-SLs与马来酰亚胺苯基丁酰基链霉亲和素(MPB-SAv)作用,可得SAv-SLs;(6)测定免疫脂质体抗体蛋白密度及抗体结合率,各种脂质体探针透射电镜观察形态。三)大鼠胶质瘤新生血管内皮细胞Endoglin靶的MR分子成像:(1)大鼠C6胶质瘤细胞培养及胶质瘤动物模型的建立;(2)25只模型动物随机分为A-E共5组,每组5只,分别尾静脉注射钆喷酸葡胺(Gd-DTPA).空白脂质体(Gd-SLs).同种型对照IgG脂质体(IgG-SLs).结合Endoglin单克隆抗体的免疫脂质体探针(MAb-SLs)行MR成像,两步法预定位成像大鼠预先给予生物素化单克隆抗体(Bio-MAb)后再给予链霉亲和素脂质体探针(SAv- SLs)进行成像;(3)比较各组大鼠MR T1加权增强图像肿瘤强化农现的差异,分析各组动脉血管、正常脑组织及肌组织、肿瘤组织时间-信号增强特点;(4)比较各组间肿瘤强化程度及强化范围差异、比较组内肿瘤中央部及周边部强化差异;(5)比较各组间肿瘤强化形态的差异;(6)大鼠胶质瘤CD105免疫组织化学染色肿瘤中央部及周边部新生血管计数,比较两者差异。
     结果:一)所制备之钆结合空间稳定脂质体(Gd-SLs)经高压均质仪过膜后粒径控制于117.4±31.8nm;脂质体Gd载药率达87%~100%;常温下(26℃)Gd-SLs磁共振驰豫率为Gd-DTPA的1.16倍,37℃为1.25倍;体外MR成像显示Gd-SLs与Gd-DTPA两对比剂MR T1加权成像作用接近。二)空间稳定脂质体结合单克隆抗体后粒径由116.1nm±33.9nm增加至129.9nm±40.9nm,于4℃保存7、14、42天,平均粒径及分布变化较小;将Bio-MAb与SAv-SLs混合后,平均粒径显著增大(267nm±142.8nm),分布增宽;免疫脂质体抗体结合率为52~67%,相应抗体/磷脂比例约为47~60μg/μmol;透射电镜显示,脂质体为均匀大小囊泡样结构,将Bio-MAb与SAv-SLs混合后,SAv-SLs有相互聚集作用。三)荷瘤大鼠MR成像(1)动脉增强表现:注射Gd-DTPA后,动脉血液快速明显强化,即刻达峰值,2小时基本消退,时间-信号曲线呈快进-快出类型;B~E组大鼠注射相应分子探针后,血管强化表现接近,早期快速明显强化,强化峰值于20~60min出现,之后强化信号缓慢下降,于48小时恢复至增强前水平,其时间-信号曲线表现为快进-平台-缓出类型。(2)肿瘤强化表现:注射Gd-DTPA后,肿瘤迅速强化达峰值,20min强化即有减退,2小时强化信号约为峰值强度30%,24小时完全退出;注射Gd-SLs及IgG-SLs,肿瘤早期强化不明显,60~120min达峰值呈轻度强化,48小时强化信号为峰值强度42~58%;注射MAb-SLs及Bio-MAb/SAv-SLs两步法成像,肿瘤周边及中央信号迅速增高,于8小时达峰值,后缓慢下降,注射MAb-SLs后肿瘤中央周边强化程度相仿,而两步法成像周边强化明显高于中央部(P=0.032)。(3)肿瘤强化形态分析显示,注射Gd-DTPA整个肿瘤不均匀强化,Gd-SLs及IgG-SLs组以肿瘤边缘点状强化为主,MAb-SLs及Bio-MAb/SAv-SLs两步法成像肿瘤增强以边缘环形强化为主。免疫组织化学染色显示,CD105阳性微血管主要沿肿瘤周边分布。
     结论:(1)顺磁性空间稳定脂质体能有效提高磁共振分子探针体内成像信号,制备方法简单,实验操作可控性好,在一定温度范围内性质稳定,具有较高的磁共振弛豫作用,是理想的MR分子影像学对比剂载体。(2)免疫脂质体结合抗体方法简单,效果确切可靠,体外相关实验能够验证生物素-亲和素介导的两步法成像的有效性。(3)大鼠胶质瘤MR分子成像,临床用Gd-DTPA肿瘤强化周边及中央强度无差异,非特异性脂质体探针肿瘤强化程度低,肿瘤边界强化形态不完整,因此均不能用于肿瘤边界显示。MAb-SLs及Bio-MAb/SAv-SLs两步法成像显示肿瘤边界较明确,两步法成像强化程度更高,实验证明为特异性靶向成像作用,因此可以用来进行肿瘤边界分析。
Objectives:
     The objectives of this study was (1) to develop paramagnetic sterical stabilized liposomes conjugated with monoclonal antibodies targeted to Endoglin on neovascular endothelial cells and evaluate their physical characteristics. (2) to quantify and compare the magnitude of signal intensity enhancement on a rat glioma model applying different contrast agent or molecular tracer using a clinical MRI (3.0 Tesla) scanner. (3) to demonstrate the use of targeted paramagnetic nanoparticles to delineate the glioma margins.
     Materials and Methods:
     1. Preparation of paramagnetic sterical stabilized liposomes:(1) A mixture of the appropriate amounts of lipids (DSPC/CHOL/mPEG-DSPE/Gd-DTPA-BSA at a molar ratio of 36/36/3/25) was dissolved in chloroform/methanol 2/1 (v/v) and evaporated to dryness by rotary evaporation at 40℃. The lipid film was subsequently hydrated in HEPES-buffered saline at 60℃. The resulting lipid dispersion was extruded sequentially 10 times through polycarbonate membrane filters with a pore diameter of 400,200, and 100 using a high-pressure homogenizer. The temperature during extrusion was 60℃. The paramagnetic sterical stabilized liposomes were obtained. (2) The size, stability, Gd containing, magnetic resonance relaxivity of the liposomes was evaluated in vitro.
     2. Coupling the antibodies to liposomes and synthesizing of pretargeting reagents. (1) For monoclonal antibodies conjugated liposomes, PDP-PEG-DSPE were added to the lipid during the preparation of liposomes (at a molar ratio of mPEG-DSPE/PDP-PEG-DSPE=4/1). The pyridyldithio groups on the PDP-PEG-DSPE incorporated liposomes were reduced by adding DTT and the thiolated liposomes (HS-SLs) were obtained. (2) CD105 monoclonal antibodies reacted with SMPB to prepare the MPB-MAb (maleimidophenylbutyrate-MAb). (3) Thiolated liposomes were incubated with MPB-MAb. The antibodies conjugated immunoliposomes (MAb-SLs) were obtained. (4) Biotinylated monoclonal antibodies were synthesized by added Sulfo-NHS-LC-biotin to the antibodies. (5) The preparation of Streptavidin coupling liposomes (SAv-SLs) was similar to that of MAb-SLs. HS-SLs were incubated with MPB-SAv. Gel filtration or centrifugation was used to purify the product in these above-mentioned reactions. (6) The antibodies protein to lipid ratio which denoted the density of antibodies conjunct to the liposomes and antibodies coupling efficiency were evaluated. The morphology of liposomes was studied by Transmission electron microscopy (TEM).
     3. MR imaging in vivo. (1) 27 male Sprague-Dawley rats were inoculated with 1.5×106 C6 glioma cells in the right caudate nucleus to develop animal model. (2)25 tumor-bearing rats were classified into five groups randomly (5 rates pre group). Gd-DTPA, Gd-SLs, IgG-SLs, MAb-SLs were injected through tail veins respectively in corresponding groups. For two steps pretargeting group, Bio-MAb were administrated 24 hours before SAv-SLs contrast enhancement. (3) MR T1-weighted images of each group were compared to evaluate the enhancement features, the signal enhancement time-intensity curves of arteries, contralateral normal brain tissues, muscular tissues and tumor tissues were analyzed. (4)The degree of contrast enhancement and notable enhancement regions were compared between each group. The degree of enhancement of the tumor central and periphery were also compared for each tumor. (5) The enhancement morphology of the tumors was analyzed. (6) CD 105 immunohistochemistry was performed after MR imaging to detect the difference of microvessel density of the tumor central and periphery.
     Results:
     1. Features of Gd-SLs. The size of Gd-SLs was 117.4±31.8nm after homogenized. The incorporation efficiency of Gd-DTPA-BSA was 87%-100%. At 26℃(room temperature), the T1 relaxivity of Gd-SLs was 1.16 times that of the solution with free Gd-DTPA. It was 1.25 times at 37℃. MR T1 imaging of Gd-SLs in vitro presented similar enhancement effect to that of Gd-DTPA.
     2. Characterization of immunoliposomes. The mean size of immunoliposomes after antibodies coupling increased from 116.1nm±33.9nm (Gd-SLs) to 129.9nm±40.9nm (MAb-SLs) and the polydispersity index showed little increase. Hypothermia (4℃) showed little influence on the size of the immunoliposomes. The mean size of SAv-SLs increased dramatically to 267nm after incubated with Bio-MAb with a polydispersity index of 0.286. The antibodies coupling efficiency ranged from 52% to 67% and corresponding antibodies protein to lipid ratio was 47~60μg/μmol. Transmission electron microscopy revealed spherical structures of liposomes with homogeneous size. SAv-SLs aggregated after mixed with Bio-MAb.
     MR imaging and analysis. (1) Arterials enhancement:Cervical arterial showed instantly significantly enhancement after Gd-DTPA was given and declined dramatically after that. The signal intensity returned to precontrast level at 2-hr time point. Thus, time-intensity curve represented as quick washin-early washout pattern. The features of arterials enhancement after liposomes tracers (Gd-SLs. IgG-SLs. MAb-SLs, Bio-MAb/SAv-SLs) injection were similar. Notable contrast enhancement was detected soon after injection and continued to improve through 20 min. It decreased slowly and washout absolutely at 48-hr time point. The time-intensity curve was described as quick washin-platform-slow washout pattern. (2) Characteristics of tumor enhancement:The signal intensity of the tumor peaked soon after the Gd-DTPA injection and sharply decreased subsequently. The intensity was about 30% that of the peak at 2-hr time point and return to the precontrast level at 24-hr. After Gd-DTPA or IgG-DTPA was administrated, the tumor showed no enhancement at early phase. It reached to maximum at 60~120 min and appeared mildly enhancement. As for MAb-SLs and Bio-MAb/SAv-SLs groups, the central and periphery of tumor signal intensity increased post-injection and reached to peak at 8-hr time point. It decreased slowly after that. The signal enhancement of the central and periphery of tumor after MAb-SLs injection were not different. In contrast, the periphery enhancement was significantly higher than that of central in Bio-MAb/SAv-SLs group (P=0.032). (3) The morphology of enhancement:T1 weighted imaging revealed an intensive entire tumor heterogeneous enhancement after Gd-DTPA injection. As for Gd-SLs and IgG-SLs groups, dots or spots enhancement scattered in or around the tumor. Ring-like marked enhancement occurred predominately along the tumor periphery after MAb-SLs or Bio-MAb/SAv-SLs were given. Immunohistochemical assessments of the tumors corroborated that angiogenesis was predominately distributed along the tumor periphery.
     Conclusions:
     1. It is easy to prepare the paramagnetic sterical stabilized liposomes which present high relaxivity to improve the MR contrast signal. The liposomes are thermo-sensitive but stable at common temperature. It may potential serve as an ideally useful contrast agent for MR molecular imaging.
     2. The applied covalent coupling of targeting antibodies is efficient. The biotin-avidin mediate antibodies and liposomes crosslink are certified in vitro.
     3. MR imaging in a glioma rat model confirmed that:no difference was found in degree between the enhancement of tumor central and periphery after Gd-DTPA contrast, and the tumor showed low enhancement while using non-targeting liposomes. The results demonstrate the limitation of these contrast agents to define the tumor morphology. The tumor extent was well delineated after targeting liposomes (MAb-SLs and Bio-MAb/SAv-SLs) contrast enhancement and the specificity was supported by the competition study in vivo. Moreover, the two-step imaging using biotin-avidin interaction was certified to induce more intensive enhancement, which imply that this novel MR molecular tracer is more suitable for detecting the tumor margins.
引文
1 Weissleder R, Mahmood U. Molecular imaging [J]. Radiology,2001, 219(2):316-333
    2 Weissleder R. Molecular Imaging in Cancer [J]. Science,2006,312 (5777) 1168-1171
    3 Louie AY, Huber MM, Ahrens ET, et al. In vivo visualization of gene expression using magnetic resonance imaging [J]. Nat Biotechnol,2000,18(3):321-325
    4 Jendelova P, Herynek V, Urdzikova L, et al. Magnetic resonance tracking of transplanted bone marrow and embryonic stem cells labeled by iron oxide nanoparticles in rat brain and spinal cord [J]. J Neurosci Res,2004, 76(2):232-243
    5 Munoz R, Arias Y, Ferreras JM. Targeting a marker of the tumour neovasculature using a novel anti-human CD105-immunotoxin containing the non-toxic type 2 ribosome-inactivating protein nigrin b [J]. Cancer Letters,2007,256(1):73-80
    6 Anderson SA, Rader RK, Weatlin WF, et al. Magnetic resonance contrast enhancement of neovasculature with αvβ3-targeted nanoparticles [J]. Mag Reson Med,2000,44(3):433-439
    7 Cimpean AM, Raica M, Suciu C. CD105/smooth muscle actin double immunostaining discriminate between immature and mature tumor blood vessels [J]. Romanian Journal of Morphology & Embryology,2007,48(1):41-45.
    8 Wikstrom P, Lissbrant IF, Stantin P, et al. Endoglin (CD 105) is expressed on immature blood vessel and is a marker for survival in prostate cancer [J]. Prostate, 2002,51(4):268-275
    9 Gohary YM, Silverman JF, Olson PR, et al. Endoglin (CD105) and vascular endothelial growth factor as prognostic markers in prostatic adenocarcinoma [J]. American Journal of Clinical Pathology,2007,127(4):572-579
    10 Dakes JP, Garcia S, Carpentier S, et al. Prediction of metastasis risk (11 year follow-up) using VEGF-R1, VEGF-R2, Tie-2/Tek and CD105 expression in breast cancer (n=905) [J]. Br J Cancer,2004.90(6):1216-1221
    11 Takahashi N, Haba A, Matsuno F, et al. Antiangiogenic therapy of established tumors in human skin/sever combined immunodeficiency mouse chimeras by anti-endoglin (CD105) monoclonal antibodies, and synergy between anti-endoglin antibody and cyclophosphamide [J]. Cancer Res,2001. 61(21):7846-7854
    12 Yao Y, Kubota T, Takeuchi H, Sato K. Prognostic significance of microvessel density determined by an anti-CD 105/endoglin monoclonal antibody in astrocytic tumors:comparison with an anti-CD31 monoclonal antibody [J]. Neuropathology, 2005,25(3):201-206
    13 Behrem S, Zarkovic K, Eskinja N, Jonjic N. Endoglin is a better marker than CD31 in evaluation of angiogenesis in glioblastoma [J]. Croatian Medical Journal,2005,46(3):417-422
    14 Volkel T, Holig P, Merdan T, Muller R, Kontermann RE. Targeting of immunoliposomes to endothelial cells using a single-chain Fv fragment directed against human endoglin (CD 105) [J]. Biochimica Biophysica Acta,2004, 1663(1-2):158-166
    15 Ester F, Maresa A, Maria RN. Endoglin (CD105):a powerful therapeutic target on tumor-associated angiogenetic blood vessels [J]. Oncogene,2003, 22,6557-6563
    16 Cai W, Rao JH, Gambhir SS. et al. How molecular imaging is speeding up antiangiogenic drug development [J]. Mol Cancer Ther,2006,5(11):2624-2633
    17 Mulder WJM, Strijkers GJ, Habets JW, et al. MR molecular imaging and fluorescence microscopy for identification of activated tumor endothelium using a bimodal lipidic nanoparticle [J]. The FASEB Journal,2005,19:2008-2010
    18 Dong Zhang, Xiao-Yuan Feng, Tobias D. Henning, Li Wen, Wei-Yue Lu, Hong Pan, Xing Wu, Li-Guang Zou. MR imaging of tumor angiogenesis using sterically stabilized Gd-DTPA liposomes targeted to CD105 [J]. European Journal of Radiology,2009 (in press)
    19张冬,冯晓源,王从俭.磁显葡胺含量测定方法的建立[J].中国医学计算机成像杂志,2005,11(1):65-66
    20 Lee JH, Huh YM, Jun YW. Artificially engineered magnetic nanoparticles for ultra-sensitive molecular imaging [J]. Nature medicine,2007,13(1):95-99
    21 Kobayashi H, Kawamoto S, Star RA, et al. Activated clearance of a biotinylated macromolecular MRI contrast agent from the blood pool using an avidin chase [J]. Bioconjug Chem,2003,14(5):1044-1047
    22 Pan H, Han L, Chen W, et al. Targeting to tumor necrotic regions with biotinylated antibody and streptavidin modified liposomes [J]. Journal of Controlled Release,2008,125(3):228-233
    23 Duff SE, Li C, Garland JM, et al. CD105 is important for angiogenesis:evidence and potential applications [J]. FASEB,2003,17(9):984-992
    1. Cai, WB, Gambhir SS, and Chen XY. Molecular Imaging of Tumor Vasculature. Angiogenesis:in Vivo Systems [J]. Methods Enzymol.2008: 445:141-176.
    2. Costouros NG, Diehn FE, Libutti SK, Molecular imaging of tumor angiogenesis [J]. Journal of Cellular Biochemistry,2002:p.72-78.
    3. Sipkins, DA, Cheresh, DA, Kazemi, MR, et al. Detection of tumor angiogenesis in vivo by alphaVbeta3-targeted magnetic resonance imaging [J].Nat. Med.1998.4:623-626.
    4. Winter PM, Caruthers SD, Kassner A, et al. Molecular Imaging of angiogenesis in nascent vx-2 rabbit tumors using a novel alpha(v)beta(3)-targeted nanoparticle and 1.5 tesla magnetic resonance imaging [J]. Cancer Research.2003.63:5838-5843.
    5. Zhang D, Feng XY, Henning TD, et al. MR imaging of tumor angiogenesis using sterically stabilized Gd-GTPA liposomes targeted to CD105 [J]. European journal of radiology.2009; 70:180-189.
    6. Le UM, Cui ZR. Long-circulating gadolinium-encapsulated liposomes for potential application in tumor neutron capture therapy [J]. International journal of pharmaceutics.2006; 312:105-112.
    7. Mulder WJ, Strijkers GJ, Griffioen AW. et al. A Liposomal System for Contrast-Enhanced Magnetic Resonance Imaging of Molecular Targets [J]. Bioconjugate Chem.2004; 15:799-806.
    8. Van Tilborg GA, Strijkers GJ, Pouget EM, et al. Kinetics of Avidin-Induced Clearance of Biotinylated Bimodal Liposomes for Improved MR Molecular Imaging [J]. Magnetic Resonance in Medicine.2008; 60:1444-1456.
    9. Glφgard C, Stensrud G, Hovland R, et al. Liposomes as carriers of amphiphilic gadolinium chelates:the effect of membrane composition on incorporation efficacy and in vitro relaxivity [J]. International journal of pharmaceutics.2002; 233:131-140.
    10. Ayyagari AL, Zhang X. Ghaghada KB, et al. Long-circulating liposomal contrast agents for magnetic resonance imaging [J]. Magnetic resonance in medicine.2006; 55:1023-1029.
    11. Karathanasis E, Park J, Agarwal A, et al. MRI mediated, non-invasive tracking of intratumoral distribution of nanocarriers in rat glioma [J]. Nanotechnology.19:315101.
    12. Bangham AD, Standish MM, Watkins JC, et al. Diffusion of univalent ions across the lamellae of swollen phospholipids[J]. J Mol Biol.1965; 13: 238-252.
    13. Mulder WJ, Strijkers GJ, Habets JW, et al. MR molecular imaging and fluorescence microscopy for identification of activated tumor endothelium using a bimodal lipidic nanoparticle [J]. FASEB J.2005 19:2008-2010.
    14. Emanuel N, Kedar E, Bolotin EM, et al, Preparetion and characterization of Doxorubicin loaded sterically stabilized immunoliposomes [J]. Pharmaceutical Research.1996; 13:352-359.
    15. Allen T M, Hansen C, Martin F, et al. Liposomes containing synthetic lipid derivatives of poly(ethylene glycol) show prolonged circulation half-lives in vivo [J]. Biochim Biophys Acta.1991; 1066:29-36.
    16. Sipkins DA, Gijbels K, Tropper FD, et al. ICAM-1 expression in autoimmune encephalitis visualized using magnetic resonance imaging [J]. J Neuroimmunol.2000; 104:1-9.
    17. Storrs RW, Tropper FD, Li HY, et al. Paramagnetic polymerized liposomes as new recirculating MR contrast agents [J]. J Magn Reson Imaging.1995:5: 719-724.
    18. Leclercq F, Cohen-Ohana M, Mignet N, et al. Design, synthesis, and evaluation of gadolinium cationic lipids as tools for biodistribution studies of gene delivery complexes [J]. Bioconjugate Chem.2003; 14:112-119.
    19. Glogard C, Stensrud G, Hovland R, et al. Liposomes as carriers of amphiphilic gadolinium chelates:the effect of membrane composition on incorporation efficacy and in vitro relaxivity [J]. Int J Pharm.2002; 233: 131-140.
    20. Le UM. Cui ZR. Long-circulating gadolinium-encapsulated liposomes for potential application in tumor neutron capture therapy [J]. International Journal of Pharmaceutics.2006; 312:105-112.
    21. Winter PM, Caruthers SD, Kassner A, et al. Molecular Imaging of angiogenesis in nascent vx-2 rabbit tumors using a novel alpha(v)beta(3)-targeted nanoparticle and 1.5 tesla magnetic resonance imaging [J]. Cancer Research.2003.63:5838-5843.
    22. Hobbs SK, Monsky WL, Yuan F, et al., Regulation of transport pathways in tumor vessels:role of tumor type and microenvironment [J]. Proc Natl Acad Sci. U.S.A.1998; 95:4607-4612.
    23. Harashima H, Hiraiwa T, Ochi Y, et al. Size dependent liposome degradation in blood:in vivo/in vitro correlation by kinetic modeling [J]. J Drug Target. 1995; 3:253-261.
    1. Winter PM, Caruthers SD, Kassner A, et al. Molecular Imaging of angiogenesis in nascent vx-2 rabbit tumors using a novel alpha(v)beta(3)-targeted nanoparticle and 1.5 tesla magnetic resonance imaging [J]. Cancer Research.2003.63:5838-5843.
    2. Allen TM, Brandeis E. Hansen CB, et al. A new strategy for attachment of antibodies to sterically stabilized liposomes resulting in efficient targeting to cancer cells [J]. Biochimica et Biophysica Acta.1995.1237:99-108.
    3. Sipkins, DA, Cheresh, DA, Kazemi, MR, et al. Detection of tumor angiogenesis in vivo by alphaVbeta3-targeted magnetic resonance imaging [J]. Nat. Med.1998.4:623-626.
    4. Mulder WJ, Strijkers GJ, Habets JW, et al. MR molecular imaging and fluorescence microscopy for identification of activated tumor endothelium using a bimodal lipidic nanoparticle [J]. FASEB J.2005 19:2008-2010.
    5. Bertini I, Bianchini F, Calorini L, et al. Persistent contrast enhancement by sterically stabilized paramagnetic liposomes in murine melanoma [J]. Magn Reson Med.2004; 52:669-72.
    6. Le UM, Cui ZR. Long-circulating gadolinium-encapsulated liposomes for potential application in tumor neutron capture therapy [J]. International Journal of Pharmaceutics.2006.312:105-112.
    7. Paganelli G, Magnani P, Fazio F. Pretargeting of carcinomas with the avidin-biotin system [J]. Int J Biol Markers.1993;8:155-9.
    8. Martensson L, Nilsson R, Ohlsson T, et al. Improved tumor targeting and decreased normal tissue accumulation through extracorporeal affinity adsorption in a two-step pretargeting strategy [J]. Clin Cancer Res.2007; 15:5572s-5576s.
    9. Gasparri A, Moro M. Curnis F. et al. Tumor pretargeting with avidin improves the therapeutic index of biotinylated tumor necrosis factor alpha in mouse models [J]. Cancer Res.1999 15:59:2917-23.
    10. Li GP, Zhang H, Zhu CM, et al. Avidin-biotin system pretargeting radioimmunoimaging and radioimmunotherapy and its application in mouse model of human colon carcinoma [J]. World J Gastroenterol.2005; 28:6288-6294.
    11. Mulder WJ, Strijkers GJ, Griffioen AW, et al. A Liposomal System for Contrast-Enhanced Magnetic Resonance Imaging of Molecular Targets [J]. Bioconjugate Chem.2004.15:799-806.
    12. Hansen CB. Kao GY. Moase EH, et al. Attachment of antibodies to stcrically stabilized liposomes:evaluation, comparison and optimization of coupling procedures [J]. Biochim Biophys Acta.1995; 1239:133-44.
    13. Pan H. Han L, Chen W, et al. Targeting to tumor necrotic regions with biotinylated antibody and streptavidin modified liposomes [J]. J Control Release.2008;125:228-35.
    14. Mrsny RJ, Volwerk JJ, Griffith OH. et al. A simplified procedure for lipid phosphorus analysis shows that digestion rates vary with phospholipid structure [J]. Chem Phys Lipids.1986.39:185-191.
    15. Van Tilborg GA, Strijkers GJ, Pouget EM, et al. Kinetics of Avidin-Induced Clearance of Biotinylated Bimodal Liposomes for Improved MR Molecular Imaging [J]. Magnetic Resonance in Medicine.2008.60:1444-1456.
    16. Rudra A, Deepa RM, Ghosh MK, et al. Doxorubicin-loaded phosphatidylethanolamine-conjugated nanoliposomes:in vitro characterization and their accumulation in liver, kidneys, and lungs in rats [J]. Int J Nanomedicine.2010;5:811-23.
    17. Ravindran R. Bhowmick S. Das A. et al. Comparison of BCG, MPL and cationic liposome adjuvant systems in leishmanial antigen vaccine formulations against murine visceral leishmaniasis [J]. BMC Microbiol. 2010:10:181.
    18. Negishi Y, Omata D, Iijima H, et al. Preparation and characterization of laminin-derived peptide AG73-coated liposomes as a selective gene delivery tool [J]. Biol Pharm Bull.2010;33:1766-1769.
    19. Kamaly N, Miller AD. Paramagnetic Liposome Nanoparticles for Cellular and Tumour Imaging [J]. Int J Mol Sci.2010; 11:1759-1776.
    1. Cai W, Rao J, Gambhir SS, et al. How molecular imaging is speeding up anti-angiogenic drug development [J]. Mol Cancer Ther.2006;5:2624-2633.
    2. Iagaru A, Chen X, Gambhir SS. Molecular imaging can accelerate antiangiogenic drug development and testing [J]. Nat Clin Pract Oncol.2007;4:556-557.
    3. Chen XY, Park R, Hou YP. MicroPET imaging of brain tumor angiogenesis with 18F-labeled PEGylated RGD peptide [J]. Eur J Nucl Med Mol Imaging.2004; 31:1081-1089.
    4. Kaijzel EL, Van der Pluijm G, Clemens WGM. Whole-Body Optical Imaging in AnimalModels toAssess Cancer Development and Progression [J]. Clin Cancer Res 2007; 13:3490-3497.
    5. Winter PM, Caruthers SD. Kassner A, et al. Molecular Imaging of angiogenesis in nascent vx-2 rabbit tumors using a novel alpha(v)beta(3)-targeted nanoparticle and 1.5 tesla magnetic resonance imaging [J]. Cancer Research.2003.63: 5838-5843.
    6. Nagengast WB, Vries EG, Geke A. et al. In Vivo VEGF Imaging with Radiolabeled Bevacizumab in a Human Ovarian Tumor Xenograft [J]. J Nucl Med 2007:48:1313-1319.
    7. Backer MV, Levashova Z, Patel V. Molecular imaging of VEGF receptors in angiogenic vasculature with single-chain VEGF-based probes [J]. NATURE MEDICINE.2007.13:504-509.
    8. Bredow S, Lewin M, Hofmann B, et al. Imaging of tumour neovasculature by targeting the TGF-beta binding receptor endoglin [J]. Eur J Cancer.2000; 36:675-681.
    9. Bhaskar S. Tian FR. Stoeger T. et al. Multifunctional Nanocarriers for diagnostics, drug delivery and targeted treatment across blood-brain barrier:perspectives on tracking and neuroimaging [J]. Part Fibre Toxicol.2010; 7:3.
    10. Lockman PR, Mumper RJ, Khan MA, et al. Nanoparticle technology for drug delivery across the blood-brain barrier [J]. Drug Dev Ind Pharm.2002; 28(1):1-13.
    11. Weissleder R. Mahmood U. Molecular Imaging [J]. Radiology 2001: 219:316-333.
    12. Schmieder AH, Winter PM, Caruthers SD, et al. Molecular MR Imaging of Melanoma Angiogenesis With αvβ3-Targeted Paramagnetic Nanoparticles [J]. Magnetic Resonance in Medicine.2005; 53:621-627.
    13. Solorzano CC, Baker CH, Bruns CJ, et al. Inhibition of Growth and Metastasis of Human Pancreatic Cancer Growing in Nude Mice by PTK 787/ZK222584, an Inhibitor of the Vascular Endothelial Growth Factor Receptor Tyrosine Kinases [J]. Cancer Biother Radiopharm.2001:16:359-70.
    14. Price SJ, Jena R, Burnet NG, et al. Improved Delineation of Glioma Margins and Regions of Infiltration with the Use of Diffusion Tensor Imaging:An Image-Guided Biopsy Study [J]. AJNR Am J Neuroradiol.2006; 27:1969-74.
    15. Burger PC. Heinz ER, Shibata T, et al. Topographic anatomy and CT correlations in the untreated glioblastoma multiforme [J]. J Neurosurg.1988; 68:698-704.
    16. Lunsford LD, Martinez AJ, Latchaw RE, et al. Magnetic resonance imaging does not define tumor boundaries [J]. Acta Radiol Suppl.1986; 369:154-56.
    17. Watanabe M, Tanaka R, Takeda N, et al. Magnetic resonance imaging and histopathology of cerebral gliomas [J]. Neuroradiology.1992; 34:463-69.
    18. Schirner M, Menrad A, Stephens A, et al. Molecular imaging of tumor angiogenesis [J]. Ann N Y Acad Sci.2004;1014:67-75.
    19. Nelson SJ, Cha S. Imaging Glioblastoma Multiforme [J]. Cancer Journal. 2003;9:134-145.
    20. Pirzkall A, Li X, Oh J, et al.3DMRSIfor resected high-grade gliomas before RT: tumor extent according to metabolic activity in relation to MRI [J]. Int J Radiat Oncol Biol Phys.2004;59:126-37.
    21. Croteau D, Scarpace L, Hearshen D, et al. Correlation between magnetic resonance spectroscopy imaging and image-guided biopsies:semiquantitative and qualitative histopathological analyses of patients with untreated glioma[J]. Neurosurgery.2001:49:823-29.
    22. Price SJ. Jenad R. Burnet NG. et al. Improved Delineation of Glioma Margins and Regions of Infiltration with the Use of Diffusion Tensor Imaging:An Image-Guided Biopsy Study [J]. American Journal of Neuroradiology.2006; 27:1969-1974.
    23. Kono K, Inoue Y, Nakayama K, et al. The role of diffusion-weighted imaging in patients with brain tumors [J]. AJNR Am J Neuroradiol.2001; 22:1081-88.
    24. Lu S, Ahn D. Johnson G, et al. Peritumoral diffusion tensor imaging of highgrade gliomas and metastatic brain tumors [J]. AJNR Am J Neuroradiol.2003;24: 937-41.
    25. Tsuchiya K, Fujikawa A. Nakajima M. et al. Differentiation between solitary brain metastasis and high-grade glioma by diffusion tensor imaging [J]. Br J Radiology.2005;78:533-37.
    26. Pauleit D, Langen KJ, Floeth F, et al. Can the apparent diffusion coefficient be used as a noninvasive parameter to distinguish tumor tissue from peritumoral tissue in cerebral gliomas [J]? J Magn Reson Imaging.2004;20:758-64.
    27. Brat DJ, Mapstone TB. Malignant Glioma Physiology:Cellular Response to Hypoxia and Its Role in Tumor Progression [J]. Ann Intern Med. 2003;138:659-668.
    28. Fonsatti E, Del Vecchio L, Altomonte M, et al. Endoglin:an accessory component of the TGF-beta-binding receptor-complex with diagnostic, prognostic, and bioimmunotherapeutic potential in human malignancies [J]. J Cell Physiol.2001: 188:1-7.
    29. Wong SH, Hamel L, Chevalier S, et al. Endoglin expression on human microvascular endothelial cells association with betaglycan and formation of higher order complexes with TGF-beta signalling receptors [J]. Eur J Biochem. 2000; 267:5550-5560.
    30. Wikstrom P, Lissbrant IF, Stattin P, et al. Endoglin (CD105) is expressed on immature blood vessels and is a marker for survival in prostate cancer [J]. Prostate.2002; 51:268-275.
    31. Burrows FJ, Derbyshire EJ. Tazzari PL, et al. Up-regulation of endoglin on vascular endothelial cells in human solid tumors:implications for diagnosis and therapy. Clin Cancer Res.1995:1:1623-1634.
    32. Fonsatti E, Jekunen AP, Kairemo KJ, et al. Endoglin is a suitable target for efficient imaging of solid tumors:in vivo evidence in a canine mammary carcinoma model [J]. Clin Cancer Res.2000;6:2037-2043.
    33. Miller DW, Graulich W, Karges B, et al. Elevated expression of endoglin, a component of the TGF-beta-receptor complex, correlates with proliferation of tumor endothelial cells [J]. Int J Cancer.1999;81:568-572.
    34. Duff SE. Li C, Garland JM.CD105 is important for angiogenesis:evidence and potential applications [J]. FASEB J.2003; 17:984-92.
    1. Cai, W. and X. Chen, Anti-angiogenic cancer therapy based on integrin alpha(v)beta(3) antagonism. Anti-Cancer Agents in Medicinal Chemistry,2006. 6(5):p.407-428.
    2. Cai, W.B. and X.Y. Chen, Multimodality molecular imaging of tumor angiogenesis. Journal of Nuclear Medicine,2008.49:p.113S-128S.
    3. Provenzale, J.M., Imaging of angiogenesis:Clinical techniques and novel imaging methods. American Journal of Roentgenology,2007.188(1):p.11-23.
    4. Gwyther, S.J., New imaging techniques in cancer management. Annals of Oncology,2005.16:p.63-70.
    5. Choyke, P.L., A.J. Dwyer, and M.V. Knopp, Functional tumor imaging with dynamic contrast-enhanced magnetic resonance imaging. Journal of Magnetic Resonance Imaging,2003.17(5):p.509-520.
    6. Dimitrakopoulou-Strauss, A., L.G. Strauss, and C. Burger, Quantitative PET studies in pretreated melanoma patients:A comparison of 6-[F-18] fluoro-L-dopa with F-18-FDG and O-15-water using compartment and noncompartment analysis. Journal of Nuclear Medicine,2001.42(2):p. 248-256.
    7. Forsberg, F., et al., Assessment of angiogenesis:implications for ultrasound imaging. Ultrasonics,2004.42(1-9):p.325-330.
    8. Cai, W.B., S.S. Gambhir, and X.Y. Chen, Molecular Imaging of Tumor Vasculature. Angiogenesis:in Vivo Systems, Pt B,2008.445:p.141-+.
    9. Hood, J.D. and D.A. Cheresh, Role of integrins in cell invasion and migration. Nature Reviews Cancer,2002.2(2):p.91-+.
    10. Liu, S., Radiolabeled multimeric cyclic RGD peptides as integrin alpha(v)beta(3) targeted radiotracers for tumor imaging. Molecular Pharmaceutics,2006.3(5):p. 472-487.
    11. Cai, W.B., et al., How molecular imaging is speeding up antiangiogenic drug development. Molecular Cancer Therapeutics,2006.5(11):p.2624-2633.
    12. Cai, W.B. and X.Y. Chen, Nanoplatforms for targeted molecular imaging in living subjects. Small,2007.3:p.1840-1854.
    13. Haubner, R., et al., Noninvasive imaging of alpha(v)beta(3) integrin expression using F-18-labeled RGD-containing glycopeptide and positron emission tomography. Cancer Research,2001.61(5):p.1781-1785.
    14. Chen, X.Y., et al., F-18-labeled RGD peptide:initial evaluation for imaging brain tumor angiogenesis. Nuclear Medicine and Biology,2004.31(2):p. 179-189.
    15. Van Hagen, P.M., et al., Evaluation of a radiolabelled cyclic DTPA-RGD analogue for tumour imaging and radionuclide therapy. International Journal of Cancer,2000.90(4):p.186-198.
    16. Sivolapenko, G.B., et al., Imaging of metastatic melanoma utilising a technetium-99m labelled RGD-containing synthetic peptide. European Journal of Nuclear Medicine,1998.25(10):p.1383-1389.
    17. Chen, X.Y., et al., MicroPET imaging of brain tumor angiogenesis with F-18-labeled PEGylated RGD peptide. European Journal of Nuclear Medicine and Molecular Imaging,2004.31(8):p.1081-1089.
    18. Li, Z.B., et al.,64Cu-Labeled "Tetrameric and octameric RGD peptides for small-animal PET of Tumor alpha(v)beta(3) integrin expression. Journal of Nuclear Medicine,2007.48(7):p.1162-1171.
    19. Line, B.R., et al., Targeting tumor angiogenesis:Ccomparison of peptide and polymer-peptide conjugates. Journal of Nuclear Medicine,2005.46(9):p. 1552-1560.
    20. Liu, Z., et al., In vivo biodistribution and highly efficient tumour targeting of carbon nanotubes in mice. Nature Nanotechnology,2007.2(1):p.47-52.
    21. Ellegala, D.B., et al., Imaging tumor angiogenesis with contrast ultrasound and microbubbles targeted to alpha(v)beta(3). Circulation,2003.108(3):p.336-341.
    22. Chen, X.Y., P.S. Conti, and R.A. Moats, In vivo near-infrared fluorescence imaging of integrin a,alpha(v)beta(3) in brain tumor xenografts. Cancer Research,2004.64(21):p.8009-8014.
    23. Cai, W.B., et al., Peptide-labeled near-infrared quantum dots for imaging tumor vasculature in living subjects. Nano Letters,2006.6(4):p.669-676.
    24. Cai, W.B., et al., Are quantum dots ready for in vivo imaging in human subjects? Nanoscale Research Letters,2007.2(6):p.265-281.
    25. Costouros, N.G., F.E. Diehn, and S.K. Libutti, Molecular imaging of tumor angiogenesis. Journal of Cellular Biochemistry,2002:p.72-78.
    26. Sipkins, D.A., et al., Detection of tumor angiogenesis in vivo by alpha(v)beta(3)-targeted magnetic resonance imaging. Nature Medicine,1998. 4(5):p.623-626.
    27. Winter, P.M., et al., Molecular Imaging of angiogenesis in nascent vx-2 rabbit tumors using a novel alpha(v)beta(3)-targeted nanoparticle and 1.5 tesla magnetic resonance imaging. Cancer Research,2003.63(18):p.5838-5843.
    28. Zhang, C.F., et al., Specific targeting of tumor angiogenesis by RGD-conjugated ultrasmall superparamagnetic iron oxide particles using a clinical 1.5-T magnetic resonance scanner. Cancer Research,2007.67(4):p.1555-1562.
    29. Celec P, Yonemitsu Y. Vascular endothelial growth factor-basic science and its clinical implications. Pathophys.2004; 2:69-75.
    30. Hicklin, D.J. and L.M. Ellis, Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. Journal of Clinical Oncology,2005. 23(5):p.1011-1027.
    31. Ferrara, N., Vascular endothelial growth factor:Basic science and clinical progress. Endocrine Reviews,2004.25(4):p.581-611.
    32. Stollman, T.H., et al., Specific imaging of VEGF-A expression with radiolabeled anti-VEGF monoclonal antibody. International Journal of Cancer, 2008.122(10):p.2310-2314.
    33. Nagengast, W.B., et al., In vivo VEGF imaging with radiolabeled bevacizumab in a human ovarian tumor xenograft. Journal of Nuclear Medicine,2007.48(8): p.1313-1319.
    34. Li, S., et al., Imaging gastrointestinal tumours using vascular endothelial growth factor-165 (VEGF(165)) receptor scintigraphy. Annals of Oncology,2003.14(8): p.1274-1277.
    35. Li, S., et al., Iodine-123-vascular endothelial growth factor-165 (I-123-VEGF(165)) biodistribution, safety and radiation dosimetry in patients with pancreatic carcinoma. Quarterly Journal of Nuclear Medicine and Molecular Imaging,2004.48(3):p.198-206.
    36. Yoshimoto, M., et al., Radioiodinated VEGF to image tumor angiogenesis in a LS180 tumor xenograft model. Nuclear Medicine and Biology,2006.33(8):p. 963-969.
    37. Cai, W.B., et al., PET of vascular endothelial growth factor receptor expression. Journal of Nuclear Medicine,2006.47(12):p.2048-2056.
    38. Wang, H., et al., A new PET tracer specific for vascular endothelial growth factor receptor 2. European Journal of Nuclear Medicine and Molecular Imaging, 2007.34(12):p.2001-2010.
    39. Korpanty, G., et al., Monitoring response to anticancer therapy by targeting microbubbles to tumor vasculature. Clinical Cancer Research,2007.13(1):p. 323-330.
    40. Willmann, J.K., et al., US imaging of tumor angiogenesis with microbubbles targeted to vascular endothelial growth factor receptor type 2 in mice. Radiology. 2008.246(2):p.508-518.
    41. Becker, A., et al., Receptor-targeted optical imaging of tumors with near-infrared fluorescent ligands. Nature Biotechnology,2001.19(4):p. 327-331.
    42. Kaijzel, E.L., G. van der Pluijm, and C. Lowik, Whole-body optical Imaging in animal models to assess cancer development and progression. Clinical Cancer Research,2007.13(12):p.3490-3497.
    43. Backer, M.V., et al., Molecular imaging of VEGF receptors in angiogenic vasculature with single-chain VEGF-based probes. Nature Medicine,2007. 13(4):p.504-509.
    44. Cai, W.B., et al., Dual-function probe for PET and near-infrared fluorescence imaging of tumor vasculature. Journal of Nuclear Medicine,2007.48(11):p. 1862-1870.
    45. Lee, H.Y., et al., PET/MRI dual-modality tumor imaging using arginine-glycine-aspartic (RGD)-Conjugated radiolabeled iron oxide nanoparticles. Journal of Nuclear Medicine,2008.49(8):p.1371-1379.

© 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