用户名: 密码: 验证码:
纳米材料在抗肿瘤免疫治疗中的应用研究
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:近年来,抗肿瘤免疫疗法在肿瘤治疗中的应用日益受到医学界的重视,其中树突状细胞(dendritic cells,DC)疗法是抗肿瘤免疫治疗领域中的一个重要研究方向,已在多种肿瘤的临床治疗试验中显示出较好的应用前景。然而,DC抗肿瘤疗法的疗效尚有较大改进空间,因此仍需发展新的技术和方法以进一步增强DC所诱导的抗肿瘤免疫反应。碳纳米管(Carbon nanotubes, CNT)作为一种优良的载体可携带多种生物活性分子穿越生物膜,将DNA、RNA、多肽、蛋白、药物等带到效应器官及细胞内。在本研究中,我们评估了碳纳米管对肿瘤抗原的载体效应,构建了碳纳米管与肿瘤蛋白共价复合物(CNT-TumorP),用以刺激DC,在体外观察了碳纳米管是否会影响DC对肿瘤蛋白的吞噬,并评价了碳纳米管-肿瘤蛋白共价复合物对DC所诱导的抗肿瘤免疫反应的影响。
     方法:抽取健康志愿者外周血,常规分离外周血单个核细胞(peripheral blood mononuclear cells, PBMC),贴壁去悬浮法分离单核细胞后,加入含重组人巨噬细胞-粒细胞集落刺激因子(rhGM-CSF)和重组人白介素-4(rhIL-4)的RPMI 1640+10% fBS完全培养基中培养6天诱导出DC;用细胞裂解液裂解人乳腺癌细胞MCF-7,提取细胞裂解蛋白(tumor lysate protein),标记上FITC荧光,再与经过表面处理的多壁碳纳米管在催化剂碳二亚胺(EDAC)的作用下进行共价偶联;用碳纳米管-肿瘤蛋白的复合物刺激DC后,激光共聚焦显微镜下观察DC对肿瘤蛋白的吞噬,流式细胞术检测DC表面CD40、CD83、CD86、HLA-DR分子的表达水平;在体外用可溶性肿瘤抗原或碳纳米管-肿瘤抗原复合物负载的DC刺激PBMC,以MTS法检测PBMC对MCF-7细胞的特异性杀伤作用;将MCF-7细胞置于含各种浓度碳纳米管的培养基内,检测碳纳米管对细胞存活的影响。
     结果:1)人外周血单个核细胞在rhGM-CSF和rhIL-4因子的协同诱导下,可获得一定数量及功能正常的未成熟树突状细胞,20m1人外周血大约可诱导出2×106个DC;2)碳纳米管与裂解蛋白共价连接后,仍具有较好的分散性及稳定性;3)流式结果显示,肿瘤蛋白与碳纳米管结合后,DC对肿瘤蛋白的吞噬增加,平均荧光强度在肿瘤蛋白浓度为1μg/ml或0.1μg/ml时,分别提升了84%和95%(p<0.05);4)激光共聚焦显微镜显示,碳纳米管与肿瘤蛋白的复合物可被DC吞入细胞质中;5)碳纳米管对DC的表型不产生影响,不会促进DC的成熟;6)以碳纳米管-肿瘤蛋白的复合物刺激DC,可显著增强肿瘤蛋白的免疫原性,改善DC对淋巴细胞的活化,进而提升DC所诱导的淋巴细胞对靶肿瘤细胞的杀伤效应;7)碳纳米管-肿瘤蛋白的复合物所增强的抗肿瘤免疫效应具有抗原特异性,对其它细胞的杀伤作用无显著增强;8)所用碳纳米管的安全性较好,在0.0005-0.5μg/ml的浓度范围内,对MCF-7的存活不产生影响。
     结论:碳纳米管与肿瘤蛋白共价偶联后,在体外能促进DC对肿瘤蛋白的吞噬,并提升DC所诱导的淋巴细胞对肿瘤细胞的杀伤作用,因而有可能在基于DC的抗肿瘤免疫治疗中具有潜在的应用价值。
     目的:抗肿瘤免疫治疗作为一种重要的肿瘤辅助治疗手段,能有效提升传统治疗方法的疗效,改善病人的预后。然而,肿瘤免疫疗法中普遍存在的一个重要问题是相关肿瘤抗原的免疫原性往往较弱,导致治疗效果有限。提高肿瘤抗原免疫原性的一种直接有效的方法是应用免疫佐剂。传统铝佐剂尽管在人类疫苗中有七十余年的应用历史,但其在增强抗肿瘤免疫方面的效果一直不佳。在本研究中我们用纳米氧化铝作为肿瘤疫苗的佐剂,在小鼠体内比较纳米铝与传统铝佐剂在抗肿瘤免疫治疗中的效果,以此来评价纳米铝作为肿瘤疫苗佐剂的潜在应用价值。
     方法:用H22肝癌细胞系在BALB/c小鼠体内建立皮下肿瘤模型(DayO);H22细胞用丝裂霉素C灭活后作为肿瘤细胞疫苗;第7天(Day7)将荷瘤小鼠随机分为四个组,分别皮下注射生理盐水、肿瘤细胞疫苗(TCV)、肿瘤细胞疫苗联合氢氧化铝佐剂(TCV+Alum)或肿瘤细胞疫苗联合纳米铝佐剂(TCV+Nano-alum)进行免疫治疗,第14天再进行相同免疫一次。在两次免疫治疗之后(Day14),每四天测量小鼠肿瘤生长大小。实验末期检测小鼠外周血单个核细胞对肿瘤细胞的杀伤率,并取肿瘤组织进行病理切片观察淋巴细胞浸润和肿瘤坏死。
     结果:1)皮下肿瘤生长情况观察:TCV+Nano-alum组的平均肿瘤面积均明显小于对照组(p<0.05),除TCV+Alum组的肿瘤面积在第14天明显小于对照组外,其它治疗组与对照组相的平均肿瘤面积在所有检测点无差异;2)体外淋巴细胞毒性试验结果显示:对照组、TCV组、TCV+Alum组和TCV+Nano-alum组的肿瘤杀伤率分别是9.9±6.9%、30.1±3.09%、35.63±2.8%和67.96+9.54%,其中TCV+Nano-alum组显示了最强的肿瘤杀伤作用(p<0.05);3)肿瘤组织病理切片观察:治疗组比对照组有更多的淋巴细胞浸润,且TCV+Nano-alum组更可见肿瘤坏死。
     结论:纳米铝可提升肿瘤细胞疫苗的免疫治疗效果,因此有可能作为一种新型的佐剂在肿瘤的免疫治疗中发挥有效作用。
     目的:半抗原二硝基氟苯(dinitrophenyl, DNP)修饰后的肿瘤细胞疫苗(tumor cell vaccine, TCV)可明显提升针对恶性黑色素瘤的免疫反应,并改善患者预后。然而,DNP瘤苗修饰法能否促进针对其它肿瘤的免疫反应,仍有待于探索。本研究的目的,是评价DNP修饰的瘤苗是否会影响人淋巴细胞对人乳腺癌细胞和人肺癌细胞的杀伤效应。并将DNP与另一类广泛应用于肿瘤细胞疫苗的修饰物——新城疫病毒Ulster株(New Castle Disease Virus of Ulster Strain, NDV Ulster)在增强淋巴细胞对肿瘤细胞杀伤作用方面的效果进行比较。
     方法:人乳腺癌细胞系MCF-7和人肺癌细胞系H23用丝裂霉素C灭活,再经DNP/NDV修饰(或不修饰)后作为肿瘤疫苗;抽取健康志愿者外周血,用Ficoll密度梯度离心法分离单个核细胞(peripheral blood mononuclear cells, PBMC),将其用作杀伤肿瘤靶细胞的效应细胞。本试验将PBMC(其中富含的淋巴细胞发挥直接杀伤作用)分为三组:未经肿瘤疫苗刺激的淋巴细胞(T)经未修饰的肿瘤疫苗所刺激过的淋巴细胞(T+TCV),和经DNP(或NDV)修饰的肿瘤疫苗所刺激过的淋巴细胞(T+DNP-TCV或T+NDV-TCV)。淋巴细胞与各种肿瘤疫苗共同孵育三天后,用标准的MTS肿瘤抑制法检测淋巴细胞对靶细胞的杀伤。
     结果:1)比之对照组(T组),不论是未修饰的肿瘤疫苗(T+TCV组)还是DNP修饰过的肿瘤疫苗(T+DNP-TCV组),均诱导淋巴细胞产生了针对MCF-7或H23细胞更强的杀伤作用;2)在MCF-7为靶细胞时,DNP修饰过的肿瘤疫苗(T+DNP-TCV组)所诱导的肿瘤杀伤,要明显高于未经修饰的肿瘤疫苗(T+TCV组);3)DNP与NDV Ulster相比,两者增强抗肿瘤免疫反应的程度是相似的,且产生的杀伤作用不针对非肿瘤靶细胞。
     结论:DNP修饰的肿瘤疫苗能显著增强人淋巴细胞对乳腺癌细胞的特异性杀伤作用。提示DNP肿瘤疫苗修饰法除了可用于治疗恶性黑色素瘤之外,也可能在针对其它人类肿瘤的免疫治疗中发挥作用。
Objective:The significance of anti-tumor immunotherapy as an adjuvant cancer treatment has been well recognised. Among the practical immunotherapy technigues, dendritic cells (DC)-based therapy is considered an important approach that has attracted mainstream attention. Some of the prior clinical studies on DC-based therapy have generated promising results in treating malignancies such as glioblastoma or malignant melanoma. Nevertheless, the outcome of DC-based anticancer immunotherapy still needs further improvement. An important issue of DC-based therapy, therefore, is to develop new technologies that can further enhance the efficacy for DC stimulation by available tumor antigens. Carbon nanotubes (CNT) have been shown to have potential applications in multiple biomedical fields, especially as transporters for delivery of various bioactive molecules such as peptides, proteins, DNAs, RNAs, or drugs. In this study, we investigated whether the conjugates of CNT and tumor protein (CNT-TumorP) would influence the endocytosis of tumor antigen by human DC in vitro. We also evaluated the effects of CNT-TumorP on the capacity of DC to induce anticancer cytotoxicity in human lymphocytes.
     Methods:Human DC were cultured from fresh peripheral blood of healthy donors per standard protocol employing rhGM-CSF and rhIL-4. MCF7 human cancer cells were lysed and tumor lysate protein (TumorP) was extracted. The tumor protein was conjugated to the oxidized CNT in the presence of the coupling agent N-ethyl-N'-(3-dimethylaminopropyl) carbodiimide (EDAC). Tumor protein (TumorP) was labeled with fluorescein-5-isothiocyanate (TumorP-FITC), and then covalently conjugated to CNT (CNT-TumorP-FITC). Two groups of DC were co-incubated with TumorP-FITC and CNT-TumorP-FITC separately. The endocytosis of tumor protein by DC was evaluated by flow cytometry and confocal microscopy. Moreover, expression of CD40, CD83, CD86 and HLA-DR on immature DC after co-incubation with CNT, TumorP or CNT-TumorP was evaluated by flow cytometry. The lymphocytes'specific cytotoxiciy to MCF-7 was then evaluated after stimulated with CNT-TumorP loaded DC using a standard MTS cytotoxicity assay. Additionally, the toxicity of CNT in a wide range of concentrations was studied using MCF-7 cell line as target.
     Results:1) Approximately 2×106 immature DC were obtained by a routine protocol employing rhGM-CSF and rhIL-4, with mononuclear cells isolated from 20ml of peripheral human blood.2) The dispersibility and stability of CNT were well preserved after the conjugation.3) The flow cytometry analysis revealed that endocytosis of tumor protein by DC was significantly enhanced when TumorP was conjugated to CNT (p<0.05). The enhancements of mean fluorescence intensity were 84% and 95% for TumorP concentration of 1μg/ml and 0.1μg/ml, respectively.4) Confocal microscopy indicated that the CNT-PRO was taken into the DC.5) CNT showed no effect on the phenotype of DC and did not induce DC maturation.6) CNT-TumorP enhanced the function of DC to induce anticancer response in lymphocytes.7) The anticancer response, induced by CNT-TumorP, was specific to the tumor cells from which the TumorP was extracted, with no obvious cytotoxicity against other cell lines.8) CNT, through a range of concentration from 0.0005μg/ml to 0.5μg/ml, showed no influence on viability of MCF-7 cells.
     Conclusion:The conjugates of carbron nanotubes and tumor protein could enhance the endocytosis of tumor antigen by human DC and the capability of DC to induce anticancer response in vitro. The results suggest that CNT-based nanotechnology may have a prospective role in the development of more efficacious DC-based anticancer immunotherapy.
     Objective:Anti-tumor immunotherapy has the potential to significantly improve the prognosis of cancer treatment, though the efficacy of immunotherapy generally needs further improvement. One way to improve the efficacy is using immune adjuvants, but the adjuvants for anticancer immunotherapy have to be more potent than for prophylactic vaccines. The identification of novel and effective adjuvants is thus of practical importance for anticancer immunotherapy. Conventional aluminum has been used as adjuvant for more than 70 years in human vaccines; however, its efficacy for enhancing antitumor immune response is limited. Nanotechnology has the potential to generate new solutions for multiple biomedical problems. In this work, we studied whether aluminum oxide nanoparticles (nano-alum) would enhance the efficacy of an anti-tumor immunotherapy that employs tumor cell vaccine (TCV), in a murine model bearing the H22 liver cancer, to assess nano-alum's applicability as adjuvant in common anticancer immune regimens.
     Methods:BALB/c mice were inoculated with 2×106 live hepatoma H22 cells subcutaneously on day 0. Tumor cell vaccine (TCV) was prepared by inactivating H22 cells with Mitomycin C. On day 7, the mice with subcutaneous tumors were randomly divided into four groups:1) the control group,2) the TCV treatment group, 3) the TCV plus conventional alum (TCV+alum) treatment group, and 4) the TCV plus nano-alum (TCV+nano-alum) treatment group. On day 7 and 14, the treatment groups received two subcutaneous doses of designed immune treatment agents for triggering anti-tumor reactions. Tumor dimensions were monitored every 4 days after the second vaccination. At the end of the study, the cytotoxicity of mouse peripheral blood lymphocytes against H22 cells was evaluated in vitro. Histological slides of the tumor tissues were prepared for evaluation tumor necrosis and lymphocyte infiltration.
     Results:1) The average tumor size of the TCV+Nano-alum group was significantly smaller than that of the control group (p<0.05). Except for the TCV+Alum group which had smaller tumor size in comparison with the control group on day 14, no difference of the average tumor sizes was observed between the other groups.2) The cytotoxicity of the lymphocytes against H22 cells was 9.9±6.9% for the control group, 30.1±3.09% for the TCV group,35.63±2.8% for the TCV+alum group, and 67.96±9.54% for the TCV+nano-alum group. TCV+nano-alum group generated significantly higher anticancer cytotoxicity compared to the other treatment groups (p<0.05).3) Compared to the control group, there was more lymphocyte infiltration in the treatment groups by histological studies. Moreover, the tumor tissue of the TCV+nano-alum group developed obvious necrosis.
     Conclusion:Aluminum oxide nanoparticles enhanced the anticancer immune response induced by tumor cell vaccine. The results suggest that nano-alum may potentially serve as an effective adjuvant in anticaner immunotherapies.
     Objective:It has been reported that hapten dinitrophenyl (DNP) modified tumor cell vaccines (TCV) can enhance the immune response against malignant melanoma and improve the prognosis of patients. However, whether DNP-modified TCV can also induce effective immune reactions against other types of malignancy has not been well evaluated. In this study, we investigated DNP-modified TCV in the enhancement of lymphocytes'cytotoxicity against breast and lung cancer cells in vitro. Moreover, we compared the enhancement of DNP-modified TCV mediated lymphocytes' cytotoxicity with that of another widely used tumor vaccine modification agent, New Castle Disease Virus of Ulster Strain (NDV Ulster),-modified TCV.
     Methods:Human breast cancer cell line MCF7, lung cancer cell line H23 cells were inactivated by mitomycin C and modified with or without DNP/NDV. As the effector cells for the killing of targeted tumor cells, peripheral blood mononuclear cells (PBMC) from healthy human donors were obtained via standard Ficoll gradient centrifugation. Then PBMC were divided into three groups:control lymphocytes (T), tumor cell vaccine-stimulated lymphocytes (T+TCV), and DNP-or NDV-modified tumor cell vaccine-stimulated lymphocytes (T+DNP-TCV or T+NDV-TCV). After 3-day incubation of the lymphocytes with different modified tumor vaccines, cell killing was evaluated using a standard MTS cytotoxicity assay.
     Results:1) Compared to the control group (T), both DNP-modified TCV and unmodified TCV caused significant lymphocyte inhibition of MCF-7 cells and H23 cells.2) DNP-modified TCV (T+DNP-TCV) induced significantly higher lymphocyte inhibition of MCF-7 cells than that of unmodified TCV (T+TCV).3) Both DNP and NDV Ulster generated similar enhancement of anti-tumor cell immune reaction but not non-tumor cells.
     Conclusion:DNP-modified TCV enhances the specific cytotoxicity of lymphocytes against human breast cancer cells. Our findings suggest that DNP-modification of TCV may have the potential in immunotherapy against multiple types of human cancer in addition to malignant melanoma.
引文
1. Rosenberg, S.A., Progress in human tumour immunology and immunotherapy. Nature, 2001.411(6835):p.380-4.
    2. Takayama, T., et al., Adoptive immunotherapy to lower postsurgical recurrence rates of hepatocellular carcinoma:a randomised trial. Lancet,2000.356(9232):p.802-7.
    3. Vermorken, J.B., et al., Active specific immunotherapy for stage Ⅱ and stage Ⅲ human colon cancer:a randomised trial. Lancet,1999.353(9150):p.345-50.
    4. Berd, D., et al., Immunopharmacologic analysis of an autologous, hapten-modified human melanoma vaccine. J Clin Oncol,2004.22(3):p.403-15.
    5. Yu, J.S., et al., Vaccination with tumor lysate-pulsed dendritic cells elicits antigen-specific, cytotoxic T-cells in patients with malignant glioma. Cancer Res,2004. 64(14):p.4973-9.
    6. Salcedo, M., et al., Vaccination of melanoma patients using dendritic cells loaded with an allogeneic tumor cell lysate. Cancer Immunol Immunother,2006.55(7):p.819-29.
    7. Van Driessche, A., et al., Clinical-grade manufacturing of autologous mature mRNA-electroporated dendritic cells and safety testing in acute myeloid leukemia patients in a phase Ⅰ dose-escalation clinical trial. Cytotherapy,2009.11(5):p.653-68.
    8. Steinman, R.M. and Z.A. Cohn, Identification of a novel cell type in peripheral lymphoid organs of mice. Ⅰ. Morphology, quantitation, tissue distribution. J Exp Med,1973.137(5): p.1142-62.
    9. Banchereau, J. and R.M. Steinman, Dendritic cells and the control of immunity. Nature, 1998.392(6673):p.245-52.
    10. Ridolfi, L., et al., Unexpected high response rate to traditional therapy after dendritic cell-based vaccine in advanced melanoma:update of clinical outcome and subgroup analysis. Clin Dev Immunol,2010.2010:p.504979.
    11. Hirschowitz, E.A. and J.R. Yannelli, Immunotherapy for lung cancer. Proc Am Thorac Soc,2009.6(2):p.224-32.
    12. Heiser, A., et al., Autologous dendritic cells transfected with prostate-specific antigen RNA stimulate CTL responses against metastatic prostate tumors. J Clin Invest,2002. 109(3):p.409-17.
    13. Su, Z., et al., Telomerase mRNA-transfected dendritic cells stimulate antigen-specific CD8+and CD4+T cell responses in patients with metastatic prostate cancer. J Immunol, 2005.174(6):p.3798-807.
    14. Nestle, F.O., A. Farkas, and C. Conrad, Dendritic-cell-based therapeutic vaccination against cancer. Curr Opin Immunol,2005.17(2):p.163-9.
    15. Markovic, S.N., et al., Preparing clinical-grade myeloid dendritic cells by electroporation-mediated transfection of in vitro amplified tumor-derived mRNA and safety testing in stage Ⅳ malignant melanoma. J Transl Med,2006.4:p.35.
    16. Brossart, P., Dendritic cells in vaccination therapies of malignant diseases. Transfus Apher Sci,2002.27(2):p.183-6.
    17. Warger, T., et al., Synergistic activation of dendritic cells by combined Toll-like receptor ligation induces superior CTL responses in vivo. Blood,2006.108(2):p.544-50.
    18. Zobywalski, A., et al., Generation of clinical grade dendritic cells with capacity to produce biologically active IL-J2p70. J Transl Med,2007.5:p.18.
    19. Boullart, A.C., et al., Maturation of monocyte-derived dendritic cells with Toll-like receptor 3 and 7/8 ligands combined with prostaglandin E2 results in high interleukin-12 production and cell migration. Cancer Immunol Immunother,2008.57(11):p.1589-97.
    20. Mailliard, R.B., et al., alpha-type-1 polarized dendritic cells:a novel immunization tool with optimized CTL-inducing activity. Cancer Res,2004.64(17):p.5934-7.
    21. Hao, S., et al., Mature dendritic cells pulsed with exosomes stimulate efficient cytotoxic T-lymphocyte responses and antitumour immunity. Immunology,2007.120(1):p.90-102.
    22. Gilboa, E. and J. Vieweg, Cancer immunotherapy with mRNA-transfected dendritic cells. Immunol Rev,2004.199:p.251-63.
    23. Shortman, K., M.H. Lahoud, and I. Caminschi, Improving vaccines by targeting antigens to dendritic cells. Exp Mol Med,2009.41(2):p.61-6.
    24. Dominguez, A.L. and J. Lustgarten, Targeting the tumor microenvironment with anti-neu/anti-CD40 conjugated nanoparticles for the induction of antitumor immune responses. Vaccine,2010.28(5):p.1383-90.
    25. Adema, G.J., et al., Migration of dendritic cell based cancer vaccines:in vivo veritas? Curr Opin Immunol,2005.17(2):p.170-4.
    26. Verdijk, P., et al., Limited amounts of dendritic cells migrate into the T-cell area of lymph nodes but have high immune activating potential in melanoma patients. Clin Cancer Res, 2009.15(7):p.2531-40.
    27. Bianco, A., et al., Biomedical applications of functionalised carbon nanotubes. Chem Commun (Camb),2005(5):p.571-7.
    28. Martin, C.R. and P. Kohli, The emerging field of nanotube biotechnology. Nat Rev Drug Discov,2003.2(1):p.29-37.
    29. Heller, D.A., et al., Optical detection of DNA conformational polymorphism on single-walled carbon nanotubes. Science,2006.311(5760):p.508-11.
    30. Cai, D., et al., Highly efficient molecular delivery into mammalian cells using carbon nanotube spearing. Nat Methods,2005.2(6):p.449-54.
    31. Kam, N.W., et al., Carbon nanotubes as multifunctional biological transporters and near-infrared agents for selective cancer cell destruction. Proc Natl Acad Sci U S A, 2005.102(33):p.11600-5.
    32. Liu, Y., et al., Polyethylenimine-grafted multiwalled carbon nanotubes for secure noncovalent immobilization and efficient delivery of DNA. Angew Chem Int Ed Engl, 2005.44(30):p.4782-5.
    33. Pantarotto, D., et al., Immunization with peptide-functionalized carbon nanotubes enhances virus-specific neutralizing antibody responses. Chem Biol,2003.10(10):p. 961-6.
    34. Meng, J., et al., Carbon nanotubes conjugated to tumor lysate protein enhance the efficacy of an antitumor immunotherapy. Small,2008.4(9):p.1364-70.
    35. Valenti, L.E., et al., The adsorption-desorption process of bovine serum albumin on carbon nanotubes. J Colloid Interface Sci,2007.307(2):p.349-56.
    36. Lu Q, M.J., Huang G, et al, RNA Polymer Translocation with Single-Walled Carbon Nanotubes. Nano Lett,2004.4(12):p.2473-2477.
    37. Haas, C., et al., A tumor vaccine containing anti-CD3 and anti-CD28 bispecific antibodies triggers strong and durable antitumor activity in human lymphocytes. Int J Cancer,2006.118(3):p.658-67.
    38. Shen, H., et al., Enhanced and prolonged cross-presentation following endosomal escape of exogenous antigens encapsulated in biodegradable nanoparticles. Immunology,2006. 117(1):p.78-88.
    39. Banchereau, J., et al., Dendritic cells as vectors for therapy. Cell,2001.106(3):p.271-4.
    40. Jarnjak-Jankovic, S., et al., A full scale comparative study of methods for generation of functional Dendritic cells for use as cancer vaccines. BMC Cancer,2007.7:p.119.
    41. Waeckerle-Men, Y., et al., Encapsulation of proteins and peptides into biodegradable poly(D,L-lactide-co-glycolide) microspheres prolongs and enhances antigen presentation by human dendritic cells. Vaccine,2006.24(11):p.1847-57.
    42. Levy, F. and S. Colombetti, Promises and limitations of murine models in the development of anticancer T-cell vaccines. Int Rev Immunol,2006.25(5-6):p.269-95.
    43. Porter, A.E., et al., Direct imaging of single-walled carbon nanotubes in cells. Nat Nanotechnol,2007.2(11):p.713-7.
    1. Huang, L. and T. Ohno, Protective antitumor immunity induced by fixed tumor cells in combination with adjuvant in a murine hepatoma model. Cancer Lett,2003.202(2):p. 153-9.
    2. Mesa, C. and L.E. Fernandez, Challenges facing adjuvants for cancer immunotherapy. Immunol Cell Biol,2004.82(6):p.644-50.
    3. Lindblad, E.B., Aluminium adjuvants--in retrospect and prospect. Vaccine,2004. 22(27-28):p.3658-68.
    4. Duclos, P., Safety of immunisation and adverse events following vaccination against hepatitis B. Expert Opin Drug Saf,2003.2(3):p.225-31.
    5. Bordet, A.L., et al., [Post-vaccination granuloma due to aluminium hydroxide]. Ann Pathol,2001.21(2):p.149-52.
    6. Su, L., et al., Measuring injection-site pain associated with vaccine administration in adults:a randomised, double-blind, placebo-controlled clinical trial. J Epidemiol Biostat, 2000.5(6):p.359-65.
    7. Han, H.R. and H.M. Park, Effects of adjuvants on the immune response of staphylococcal alpha toxin and capsular polysaccharide (CPS) in rabbit. J Vet Med Sci,2000.62(3):p. 237-41.
    8. Berd, D., et al., Immunopharmacologic analysis of an autologous, hapten-modified human melanoma vaccine. J Clin Oncol,2004.22(3):p.403-15.
    9. Emens, L.A., Roadmap to a better therapeutic tumor vaccine. Int Rev Immunol,2006. 25(5-6):p.415-43.
    10. Williams, K.A., et al., Nanotechnology:carbon nanotubes with DNA recognition. Nature, 2002.420(6917):p.761.
    11. Heller, D.A., et al., Optical detection of DNA conformational polymorphism on single-walled carbon nanotubes. Science,2006.311(5760):p.508-11.
    12. Elder, J.B., C.Y. Liu, and M.L. Apuzzo, Neurosurgery in the realm of 10(-9), Part 2: applications of nanotechnology to neurosurgery--present and future. Neurosurgery,2008. 62(2):p.269-84; discussion 284-5.
    13. Jain, K.K., Nanomedicine:application of nanobiotechnology in medical practice. Med Princ Pract,2008.17(2):p.89-101.
    14. Cai, D., et al., Highly efficient molecular delivery into mammalian cells using carbon nanotube spearing. Nat Methods,2005.2(6):p.449-54.
    15. Fenske, D.B. and P.R. Cullis, Liposomal nanomedicines. Expert Opin Drug Deliv,2008. 5(1):p.25-44.
    16. Martin, C.R. and P. Kohli, The emerging field of nanotube biotechnology. Nat Rev Drug Discov,2003.2(1):p.29-37.
    17. Bianco, A., et al., Biomedical applications of functionalised carbon nanotubes. Chem Commun (Camb),2005(5):p.571-7.
    18. Liu, Y., et al., Polyethylenimine-grafted multiwalled carbon nanotubes for secure noncovalent immobilization and efficient delivery of DNA. Angew Chem Int Ed Engl, 2005.44(30):p.4782-5.
    19. Ni, Y., et al., Chemically functionalized water soluble single-walled carbon nanotubes modulate neurite outgrowth. J Nanosci Nanotechnol,2005.5(10):p.1707-12.
    20. Sun, Y.P., et al., Functionalized carbon nanotubes:properties and applications. Acc Chem Res,2002.35(12):p.1096-104.
    21. Zanello, L.P., et al., Bone cell proliferation on carbon nanotubes. Nano Lett,2006.6(3):p. 562-7.
    22. Bianco, A., K. Kostarelos, and M. Prato, Applications of carbon nanotubes in drug delivery. Curr Opin Chem Biol,2005.9(6):p.674-9.
    23. Hartman, K.B., L.J. Wilson, and M.G. Rosenblum, Detecting and treating cancer with nanotechnology. Mol Diagn Ther,2008.12(1):p.1-14.
    24. Fifis, T., et al., Size-dependent immunogenicity:therapeutic and protective properties of nano-vaccines against tumors. J Immunol,2004.173(5):p.3148-54.
    25. Farokhzad, O.C., et al., Targeted nanoparticle-aptamer bioconjugates for cancer chemotherapy in vivo. Proc Natl Acad Sci U S A,2006.103(16):p.6315-20.
    26. Frey, A., et al., Immunization of mice with peptomers covalently coupled to aluminum oxide nanoparticles. Vaccine,1999.17(23-24):p.3007-19.
    27.何萍,吕凤林,陈月等,合成纳米铝佐剂及其对乙型肝炎、狂犬病毒辅佐效应的研 究.免疫学杂志,2006.22(1):p.90-93.
    28.Luo, Y., et al., Immunotherapy of tumors with protein vaccine based on chicken homologous Tie-2. Clin Cancer Res,2006.12(6):p.1813-9.
    29. Vermorken, J.B., et al., Active specific immunotherapy for stage Ⅱ and stage Ⅲ human colon cancer:a randomised trial. Lancet,1999.353(9150):p.345-50.
    30. Schirrmacher, V., Clinical trials of antitumor vaccination with an autologous tumor cell vaccine modified by virus infection:improvement of patient survival based on improved antitumor immune memory. Cancer Immunol Immunother,2005.54(6):p.587-98.
    31. Takayama, T., et al., Adoptive immunotherapy to lower postsurgical recurrence rates of hepatocellular carcinoma:a randomised trial. Lancet,2000.356(9232):p.802-7.
    32. Yu, J.S., et al., Vaccination with tumor lysate-pulsed dendritic cells elicits antigen-specific, cytotoxic T-cells in patients with malignant glioma. Cancer Res,2004.64(14):p.4973-9.
    33. Rosenberg, S.A., et al., Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using high-dose bolus interleukin 2. Jama,1994.271(12):p.907-13.
    34. Baxevanis, C.N., S.A. Perez, and M. Papamichail, Cancer immunotherapy. Crit Rev Clin Lab Sci,2009.46(4):p.167-89.
    35. Ribas, A., Update on immunotherapy for melanoma. J Natl Compr Canc Netw,2006.4(7): p.687-94.
    36.何萍,吕凤林,任建敏等,铝佐剂机制及其纳米化前景世界华人消化杂志,2003.11(11):p.1764-1768.
    37. Copier, J. and A. Dalgleish, Overview of tumor cell-based vaccines. Int Rev Immunol, 2006.25(5-6):p.297-319.
    1. Rosenberg, S.A., Progress in human tumour immunology and immunotherapy. Nature, 2001.411(6835):p.380-4.
    2. Takayama, T., et al., Adoptive immunotherapy to lower postsurgical recurrence rates of hepatocellular carcinoma:a randomised trial. Lancet,2000.356(9232):p.802-7.
    3. Vermorken, J.B., et al., Active specific immunotherapy for stage Ⅱ and stage Ⅲ human colon cancer:a randomised trial. Lancet,1999.353(9150):p.345-50.
    4. Schirrmacher, V., Clinical trials of antitumor vaccination with an autologous tumor cell vaccine modified by virus infection:improvement of patient survival based on improved antitumor immune memory. Cancer Immunol Immunother,2005.54(6):p.587-98.
    5. Yu, J.S., et al., Vaccination with tumor lysate-pulsed dendritic cells elicits antigen-specific, cytotoxic T-cells in patients with malignant glioma. Cancer Res,2004. 64(14):p.4973-9.
    6. Berd, D., et al., Immunopharmacologic analysis of an autologous, hapten-modified human melanoma vaccine. J Clin Oncol,2004.22(3):p.403-15.
    7. Rosenberg, S.A., et al., Treatment of 283 consecutive patients with metastatic melanoma or renal cell cancer using high-dose bolus interleukin 2. Jama,1994.271(12):p.907-13.
    8. Rosenberg, S.A., et al., Durability of complete responses in patients with metastatic cancer treated with high-dose interleukin-2:identification of the antigens mediating response. Ann Surg,1998.228(3):p.307-19.
    9. Chatterjee, M., S. Draghici, and M.A. Tainsky, Immunotheranostics:breaking tolerance in immunotherapy using tumor autoantigens identified on protein microarrays. Curr Opin Drug Discov Devel,2006.9(3):p.380-5.
    10. Muller, A.J. and P.A. Scherle, Targeting the mechanisms of tumoral immune tolerance with small-molecule inhibitors. Nat Rev Cancer,2006.6(8):p.613-25.
    11. Yang, J.C. and R. Childs, Immunotherapy for renal cell cancer. J Clin Oncol,2006. 24(35):p.5576-83.
    12. Banchereau, J., et al., Dendritic cells as vectors for therapy. Cell,2001.106(3):p.271-4.
    13. Copier, J. and A. Dalgleish, Overview of tumor cell-based vaccines. Int Rev Immunol, 2006.25(5-6):p.297-319.
    14. Miller, S.D. and H.N. Claman, The induction of hapten-specific T cell tolerance by using hapten-modified lymphoid cells. I. Characteristics of tolerance induction. J Immunol, 1976.117(5 Pt 1):p.1519-26.
    15. Haas, C., et al., A tumor vaccine containing anti-CD3 and anti-CD28 bispecific antibodies triggers strong and durable antitumor activity in human lymphocytes. Int J Cancer,2006.118(3):p.658-67.
    16. V. Schirrmacher, Clinical trials of antitumor vaccination with an autologous tumor cell vaccine modified by virus infection:improvement of patient survival based on improved antitumor immune memory. Cancer Immunol. Immunother.,2005.54:p.587-598.
    17. Manne, J., et al., TCR rearrangement in lymphocytes infiltrating melanoma metastases after administration of autologous dinitrophenyl-modified vaccine. J Immunol,2002. 169(6):p.3407-12.
    18. Berd, D., et al., Autologous hapten-modified melanoma vaccine as postsurgical adjuvant treatment after resection of nodal metastases. J Clin Oncol,1997.15(6):p.2359-70.
    19. Berd, D., et al., Autologous, hapten-modified vaccine as a treatment for human cancers. Semin Oncol,1998.25(6):p.646-53.
    20. Berd, D., Autologous, hapten-modified vaccine as a treatment for human cancers. Vaccine,2001.19(17-19):p.2565-70.
    21. Berd, D., et al., Treatment of metastatic melanoma with autologous, hapten-modified melanoma vaccine:regression of pulmonary metastases. Int J Cancer,2001.94(4):p. 531-9.
    22. Berd, D., M-Vax:an autologous, hapten-modified vaccine for human cancer. Expert Rev Vaccines,2004.3(5):p.521-7.
    1. Banchereau, J. and R.M. Steinman, Dendritic cells and the control of immunity. Nature, 1998.392(6673):p.245-52.
    2. Dubois, B., et al., Measles virus exploits dendritic cells to suppress CD4+T-cell proliferation via expression of surface viral glycoproteins independently of T-cell trans-infection. Cell Immunol,2001.214(2):p.173-83.
    3. Mailliard, R.B., et al., Dendritic cells promote T-cell survival or death depending upon their maturation state and presentation of antigen. Immunol Invest,2000.29(2):p. 177-85.
    4. Livingston, P.O., Experimental and clinical studies with active specific immunotherapy. Prog Clin Biol Res,1989.288:p.309-21.
    5. Steinman, R.M. and Z.A. Cohn, Identification of a novel cell type in peripheral lymphoid organs of mice. I. Morphology, quantitation, tissue distribution. J Exp Med,1973.137(5): p.1142-62.
    6. Naik, S.H., et al., Development of plasmacytoid and conventional dendritic cell subtypes from single precursor cells derived in vitro and in vivo. Nat Immunol,2007.8(11):p. 1217-26.
    7. Sekar, D., B. Brune, and A. Weigert, Technical advance:Generation of human pDC equivalents from primary monocytes using Flt3-L and their functional validation under hypoxia. J Leukoc Biol,2010.88(2):p.413-24.
    8. Gandhi, R.T., et al., A randomized therapeutic vaccine trial of canarypox-HIV-pulsed dendritic cells vs. canarypox-HIV alone in HIV-1-infected patients on antiretroviral therapy. Vaccine,2009.27(43):p.6088-94.
    9. Redman, B.G., et al., Phase Ib trial assessing autologous, tumor-pulsed dendritic cells as a vaccine administered with or without IL-2 in patients with metastatic melanoma. J Immunother,2008.31(6):p.591-8.
    10. O'Neill, D.W. and N. Bhardwaj, Differentiation of peripheral blood monocytes into dendritic cells. Curr Protoc Immunol,2005. Chapter 22:p. Unit 22F 4.
    11. Nestle, F.O., A. Farkas, and C. Conrad, Dendritic-cell-based therapeutic vaccination against cancer. Curr Opin Immunol,2005.17(2):p.163-9.
    12. Dauer, M., et al., Mature dendritic cells derived from human monocytes within 48 hours: a novel strategy for dendritic cell differentiation from blood precursors. J Immunol,2003. 170(8):p.4069-76.
    13. Obermaier, B., et al., Development of a new protocol for 2-day generation of mature dendritic cells from human monocytes. Biol Proced Online,2003.5:p.197-203.
    14. Jarnjak-Jankovic, S., et al., A full scale comparative study of methods for generation of functional Dendritic cells for use as cancer vaccines. BMC Cancer,2007.7(1):p.119.
    15. Czerniecki, B.J., et al., Targeting HER-2/neu in early breast cancer development using dendritic cells with staged interleukin-12 burst secretion. Cancer Res,2007.67(4):p. 1842-52.
    16. Palucka, A.K., et al., Boosting vaccinations with peptide-pulsed CD34+ progenitor-derived dendritic cells can expand long-lived melanoma peptide-specific CD8+ T cells in patients with metastatic melanoma. J Immunother,2005.28(2):p. 158-68.
    17. Banchereau, J., et al., Immune and clinical responses in patients with metastatic melanoma to CD34(+) progenitor-derived dendritic cell vaccine. Cancer Res,2001. 61(17):p.6451-8.
    18. Marroquin, C.E., et al., Mobilization of dendritic cell precursors in patients with cancer by flt3 ligand allows the generation of higher yields of cultured dendritic cells. J Immunother,2002.25(3):p.278-88.
    19. Small, E.J., et al., Placebo-controlled phase Ⅲ trial of immunologic therapy with sipuleucel-T (APC8015) in patients with metastatic, asymptomatic hormone refractory prostate cancer. J Clin Oncol,2006.24(19):p.3089-94.
    20. Kantoff, P.W., et al., Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med,2010.363(5):p.411-22.
    21. Yasuda, T., et al., Superior anti-tumor protection and therapeutic efficacy of vaccination with allogeneic and semiallogeneic dendritic cell/tumor cell fusion hybrids for murine colon adenocarcinoma. Cancer Immunol Immunother,2007.56(7):p.1025-36.
    22. Dhodapkar, M.V., et al., Antigen-specific inhibition of effector T cell function in humans after injection of immature dendritic cells. J Exp Med,2001.193(2):p.233-8.
    23. Dhodapkar, M.V. and R.M. Steinman, Antigen-bearing immature dendritic cells induce peptide-specific CD8(+) regulatory T cells in vivo in humans. Blood,2002.100(1):p. 174-7.
    24. Gilboa, E., DC-based cancer vaccines. J Clin Invest,2007.117(5):p.1195-203.
    25. Banchereau, J. and A.K. Palucka, Dendritic cells as therapeutic vaccines against cancer. Nat Rev Immunol,2005.5(4):p.296-306.
    26. Jonuleit, H., et al., Pro-inflammatory cytokines and prostaglandins induce maturation of potent immunostimulatory dendritic cells under fetal calf serum-free conditions. Eur J Immunol,1997.27(12):p.3135-42.
    27. MartIn-Fontecha, A., et al., Regulation of dendritic cell migration to the draining lymph node:impact on T lymphocyte traffic and priming. J Exp Med,2003.198(4):p.615-21.
    28. Schadendorf, D., et al., Dacarbazine (DTIC) versus vaccination with autologous peptide-pulsed dendritic cells (DC) in first-line treatment of patients with metastatic melanoma:a randomized phase Ⅲ trial of the DC study group of the DeCOG. Ann Oncol, 2006.17(4):p.563-70.
    29. Jongmans, W., et al., Thl-polarizing capacity of clinical-grade dendritic cells is triggered by Ribomunyl but is compromised by PGE2:the importance of maturation cocktails. J Immunother,2005.28(5):p.480-7.
    30. Morelli, A.E. and A.W. Thomson, Dendritic cells under the spell of prostaglandins. Trends Immunol,2003.24(3):p.108-11.
    31. Scandella, E., et al., Prostaglandin E2 is a key factor for CCR7 surface expression and migration of monocyte-derived dendritic cells. Blood,2002.100(4):p.1354-61.
    32. Hoggatt, J., et al., Prostaglandin E2 enhances hematopoietic stem cell homing, survival, and proliferation. Blood,2009.113(22):p.5444-55.
    33. Mailliard, R.B., et al., alpha-type-1 polarized dendritic cells:a novel immunization tool with optimized CTL-inducing activity. Cancer Res,2004.64(17):p.5934-7.
    34. Warger, T., et al., Synergistic activation of dendritic cells by combined Toll-like receptor ligation induces superior CTL responses in vivo. Blood,2006.108(2):p.544-50.
    35. Sporri, R. and C. Reis e Sousa, Inflammatory mediators are insufficient for full dendritic cell activation and promote expansion of CD4+ T cell populations lacking helper function. Nat Immunol,2005.6(2):p.163-70.
    36. Zobywalski, A., et al., Generation of clinical grade dendritic cells with capacity to produce biologically active IL-12p70. J Transl Med,2007.5:p.18.
    37. Boullart, A.C., et al., Maturation of monocyte-derived dendritic cells with Toll-like receptor 3 and 7/8 ligands combined with prostaglandin E2 results in high interleukin-12 production and cell migration. Cancer Immunol Immunother,2008.57(11):p.1589-97.
    38. Nair, S., et al., Injection of immature dendritic cells into adjuvant-treated skin obviates the need for ex vivo maturation. J Immunol,2003.171(11):p.6275-82.
    39. Nencioni, A., et al., The use of dendritic cells in cancer immunotherapy. Crit Rev Oncol Hematol,2008.65(3):p.191-9.
    40. Tjoa, B.A., A.A. Elgamal, and G.P. Murphy, Vaccine therapy for prostate cancer. Urol Clin North Am,1999.26(2):p.365-74, ix.
    41. Schott, M., et al., Immunotherapy for medullary thyroid carcinoma by dendritic cell vaccination. J Clin Endocrinol Metab,2001.86(10):p.4965-9.
    42. Vambutas, A., et al., Therapeutic vaccination with papillomavirus E6 and E7 long peptides results in the control of both established virus-induced lesions and latently infected sites in a pre-clinical cottontail rabbit papillomavirus model. Vaccine,2005. 23(45):p.5271-80.
    43. Mule, J.J., Tumor vaccine strategies that employ dendritic cells and tumor lysates: experimental and clinical studies. Immunol Invest,2000.29(2):p.127-9.
    44. Bercovici, N., et al., Analysis and characterization of antitumor T-cell response after administration of dendritic cells loaded with allogeneic tumor lysate to metastatic melanoma patients. J Immunother,2008.31(1):p.101-12.
    45. Schwaab, T., et al., Clinical and immunologic effects of intranodal autologous tumor lysate-dendritic cell vaccine with Aldesleukin (Interleukin 2) and IFN-{alpha}2a therapy in metastatic renal cell carcinoma patients. Clin Cancer Res,2009.15(15):p.4986-92.
    46. Yu, J.S., et al., Vaccination with tumor lysate-pulsed dendritic cells elicits antigen-specific, cytotoxic T-cells in patients with malignant glioma. Cancer Res,2004.64(14):p.4973-9.
    47. Zitvogel, L., et al., Eradication of established murine tumors using a novel cell-free vaccine:dendritic cell-derived exosomes. Nat Med,1998.4(5):p.594-600.
    48. Matsumoto, K., et al., Exosomes secreted from monocyte-derived dendritic cells support in vitro naive CD4+ T cell survival through NF-(kappa)B activation. Cell Immunol,2004. 231(1-2):p.20-9.
    49. Hao, S., et al., Mature dendritic cells pulsed with exosomes stimulate efficient cytotoxic T-lymphocyte responses and antitumour immunity. Immunology,2007.120(1):p.90-102.
    50. Kim, G.Y., et al., Dendritic cell-tumor fusion vaccine prevents tumor growth in vivo. Biosci Biotechnol Biochem,2007.71(1):p.215-21.
    51. Koido, S., et al., Streptococcal preparation OK-432 promotes fusion efficiency and enhances induction of antigen-specific CTL by fusions of dendritic cells and colorectal cancer cells. J Immunol,2007.178(1):p.613-22.
    52. Boczkowski, D., et al., Dendritic cells pulsed with RNA are potent antigen-presenting cells in vitro and in vivo. J Exp Med,1996.184(2):p.465-72.
    53. Van Tendeloo, V.F., et al., Highly efficient gene delivery by mRNA electroporation in human hematopoietic cells:superiority to lipofection and passive pulsing of mRNA and to electroporation of plasmid cDNA for tumor antigen loading of dendritic cells. Blood, 2001.98(1):p.49-56.
    54. Gilboa, E. and J. Vieweg, Cancer immunotherapy with mRNA-transfected dendritic cells. Immunol Rev,2004.199:p.251-63.
    55. Heiser, A., et al., Induction of polyclonal prostate cancer-specific CTL using dendritic cells transfected with amplified tumor RNA. J Immunol,2001.166(5):p.2953-60.
    56. Boczkowski, D., et al., Induction of tumor immunity and cytotoxic T lymphocyte responses using dendritic cells transfected with messenger RNA amplified from tumor cells. Cancer Res,2000.60(4):p.1028-34.
    57. Kang, T.H., et al., Enhancing dendritic cell vaccine potency by combining a BAK/BAX siRNA-mediated antiapoptotic strategy to prolong dendritic cell life with an intracellular strategy to target antigen to lysosomal compartments. Int J Cancer,2007.120(8):p. 1696-703.
    58. Matsuyoshi, H., et al., Enhanced priming of antigen-specific CTLs in vivo by embryonic stem cell-derived dendritic cells expressing chemokine along with antigenic protein: application to antitumor vaccination. J Immunol,2004.172(2):p.776-86.
    59. Jack, A.M., et al., A novel dendritic cell-based cancer vaccine produces promising results in a syngenic CC-36 murine colon adenocarcinoma model. J Surg Res,2007.139(2):p. 164-9.
    60. Naka, T., et al., Tumor vaccine therapy against recrudescent tumor using dendritic cells simultaneously transfected with tumor RNA and granulocyte macrophage colony-stimulating factor RNA. Cancer Sci,2008.99(2):p.407-13.
    61. Shortman, K., M.H. Lahoud, and I. Caminschi, Improving vaccines by targeting antigens to dendritic cells. Exp Mol Med,2009.41(2):p.61-6.
    62. Tacken, P.J., R. Torensma, and C.G. Figdor, Targeting antigens to dendritic cells in vivo. Immunobiology,2006.211(6-8):p.599-608.
    63. Dominguez, A.L. and J. Lustgarten, Targeting the tumor microenvironment with anti-neu/anti-CD40 conjugated nanoparticles for the induction of antitumor immune responses. Vaccine,2010.28(5):p.1383-90.
    64. Ribas, A., et al., Multicenter phase Ⅱ study of matured dendritic cells pulsed with melanoma cell line lysates in patients with advanced melanoma. J Transl Med,2010.8:p. 89.
    65. Ridolfi, L., et al., Unexpected high response rate to traditional therapy after dendritic cell-based vaccine in advanced melanoma:update of clinical outcome and subgroup analysis. Clin Dev Immunol,2010.2010:p.504979.
    66. Hirschowitz, E.A. and J.R. Yannelli, Immunotherapy for lung cancer. Proc Am Thorac Soc,2009.6(2):p.224-32.
    67. Cranmer, L.D., K.T. Trevor, and E.M. Hersh, Clinical applications of dendritic cell vaccination in the treatment of cancer. Cancer Immunol Immunother,2004.53(4):p. 275-306.
    68. Lesterhuis, W.J., et al., Dendritic cell-based vaccines in cancer immunotherapy:an update on clinical and immunological results. Ann Oncol,2004.15 Suppl 4:p. ⅳ145-51.
    69. Fay, J.W., et al., Long-term outcomes in patients with metastatic melanoma vaccinated with melanoma peptide-pulsed CD34(+) progenitor-derived dendritic cells. Cancer Immunol Immunother,2006.55(10):p.1209-18.
    70. Heiser, A., et al., Autologous dendritic cells transfected with prostate-specific antigen RNA stimulate CTL responses against metastatic prostate tumors. J Clin Invest,2002. 109(3):p.409-17.
    71. Su, Z., et al., Telomerase mRNA-transfected dendritic cells stimulate antigen-specific CD8+ and CD4+ T cell responses in patients with metastatic prostate cancer. J Immunol, 2005.174(6):p.3798-807.
    72. Dannull, J., et al., Enhancement of vaccine-mediated antitumor immunity in cancer patients after depletion of regulatory T cells. J Clin Invest,2005.115(12):p.3623-33.
    73. Berger, R., et al., Phase I safety and pharmacokinetic study of CT-011, a humanized antibody interacting with PD-1, in patients with advanced hematologic malignancies. Clin Cancer Res,2008.14(10):p.3044-51.
    74. Yuan, J., et al., CTLA-4 blockade enhances polyfunctional NY-ESO-1 specific T cell responses in metastatic melanoma patients with clinical benefit. Proc Natl Acad Sci U S A,2008.105(51):p.20410-5.
    75. Shimizu, J., et al., Stimulation of CD25(+)CD4(+) regulatory T cells through GITR breaks immunological self-tolerance. Nat Immunol,2002.3(2):p.135-42.
    76. Ko, K., et al., Treatment of advanced tumors with agonistic anti-GITR mAb and its effects on tumor-infiltrating Foxp3+ CD25+CD4+ regulatory T cells. J Exp Med,2005.202(7): p.885-91.
    77. Lopez, M.N., et al., Prolonged survival of dendritic cell-vaccinated melanoma patients correlates with tumor-specific delayed type IV hyper sensitivity response and reduction of tumor growth factor beta-expressing T cells. J Clin Oncol,2009.27(6):p.945-52.
    78. Hanks, B.A., et al., Re-engineered CD40 receptor enables potent pharmacological activation of dendritic-cell cancer vaccines in vivo. Nat Med,2005.11(2):p.130-7.

© 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