CIK细胞的体内外抗肿瘤作用及回输后体内分布特点研究
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:采用人宫颈癌细胞模型为研究对象。研究细胞因子诱导的杀伤细胞(cytokine-induced killer cells,CIK细胞)对人宫颈癌细胞及该细胞荷瘤小鼠的体内外抗肿瘤作用。分析将体外培养的CIK细胞回输体内后在不同组织器官的分布特点,为CIK细胞临床应用提供理论依据。
     方法:
     1 CIK细胞的制备及扩增采集健康志愿者外周血,经Ficoll密度梯度离心法收集外周血单个核细胞(Peripheral blood mononuclear cell, PBMC),按特定顺序加入IFN-γ、IL-2、抗CD3单抗和IL-1α,每3 d更换新鲜无血清培养基并补充IL-2,每7d测定细胞增殖反应,并绘制细胞生长曲线。经14 d培养后收集CIK细胞。
     2 CIK细胞表型及产生细胞因子测定用流式细胞技术检测CIK细胞表型分子的表达。用半定量RT-PCR法检测体外培养0、7、14、21、28、35 d时的CIK细胞IFN-γmRNA表达变化。
     3 CIK细胞的杀伤活性测定收集PBMC、体外经细胞因子诱导培养14d的CIK细胞及呈对数生长期的Hela-luc细胞,分别将CIK细胞和PBMC作为效应细胞,Hela-luc细胞作为靶细胞,以效靶比为20:1和40:1将两种细胞混合培养48 h,用MTT比色法测定CIK细胞和PBMC对Hela-luc细胞的杀伤作用。
     4经瑞氏-姬姆萨染色后,用光学显微镜观察CIK细胞对Hela-luc细胞作用后的细胞形态变化。
     5荷瘤小鼠模型的建立收集呈对数生长期的Hela-luc细胞,制成细胞悬液,调整细胞浓度为1×108/ml,于BALB/c裸鼠右侧胸背部皮下注射细胞悬液0.1ml,1×107/只,建立荷瘤小鼠模型。
     6 CIK细胞的体内抑瘤作用向裸鼠移植肿瘤细胞7 d后,于荷瘤小鼠的瘤旁分别注射CIK细胞(实验组)或PBS(对照组),每组6只。通过精诺真体内成像系统动态观察肿瘤大小变化及治疗效果。治疗结束后收集小鼠外周血、肿瘤组织、肺、肝、脾组织。
     7用ELISA法检测荷瘤小鼠外周血中IFN-γ的水平。
     8经HE染色,显微镜下观察肺、肝、脾的组织病理学变化以及有无肿瘤细胞浸润。
     9用羧基荧光素二醋酸盐琥珀酰亚胺酯(CFSE)标记CIK细胞,经激光共聚焦显微镜观察标记情况。
     10 CIK细胞在荷瘤小鼠体内的分布检测通过腹腔、瘤旁分别给荷瘤小鼠注射经CFSE标记的CIK细胞,对照组不进行任何处理。分别于注射CIK细胞后3、6、12、24 h留取外周血、肿瘤组织、肺、肝、脾。将上述各待测组织剪碎,制成单细胞悬液,一部分经流式细胞技术检测各组织中CFSE标记的CIK细胞含量;另一部分进行激光共聚焦显微镜观察肿瘤组织中CIK细胞的分布情况。
     结果:
     1将人外周血单个核细胞经不同细胞因子体外培养后,诱导分化的CIK细胞在培养至第7 d时开始快速增殖,14 d时达高峰,增殖倍数最高达100倍以上。
     2在培养过程中,CIK细胞CD3+、CD8+、CD3+CD56+ T细胞的百分率均大幅度增加,14 d时的表达率分别为92%、42.12%、63.34%。CD3+CD56+ T细胞的百分率与最初培养当天相比(1.10%)增加57.5倍。
     3体外诱导培养不同时间的CIK细胞IFN-γmRNA表达水平不断增加,在14 d时达到高峰。
     4以20:1、40:1的比例将效、靶细胞作用48 h后,CIK细胞对Hela-luc细胞的杀伤率分别为(51.16±2.64)%、(72.14%±4.21)%。PBMC对Hela-luc细胞的杀伤率则分别为(16.33±3.09)%和(21.26±2.71)%。
     5经瑞氏-姬姆萨染色显微镜下可见:正常靶细胞呈菱形或多边形生长,形态饱满,细胞核染色均匀;与CIK细胞共培养的靶细胞周围有多个效应细胞围绕,形成花环状,靶细胞膜疏松,胞浆中有空泡。
     6注射Hela-luc细胞7 d后,在裸鼠体内全部成瘤(成瘤率达100%),成功建立了荷瘤小鼠模型。
     7经CIK细胞治疗4次后,荷瘤小鼠的肿瘤明显小于对照组(p<0.05),经CIK细胞治疗的第5周、第8周,对肿瘤的总抑瘤率分别为47.18%、64.38%。
     8 CIK细胞治疗组荷瘤小鼠外周血中IFN-γ水平(61.92±6.49)pg/ml明显高于对照组(34.30±1.78)pg/ml(p﹤0.05)。
     9 CIK细胞治疗组和对照组荷瘤小鼠肿瘤组织无明显形态学差异;肝脏均可见完整汇管区,未见癌结节,但有大量炎细胞浸润;脾脏均可见癌结节。CIK细胞治疗组小鼠肺脏未见癌结节,但有炎细胞浸润,对照组小鼠肺脏出现大量癌结节。
     10 CFSE标记的CIK细胞经腹腔注射荷瘤小鼠3 h后,可在肺、肝、脾、外周血、肿瘤中观察到绿色荧光,以肺脏的分布浓度最高(32.22%),其次是肝脏,肿瘤组织只有9.44%;注射6 h时,小鼠的肺、肝、脾脏分布浓度逐渐下降,而外周血、肿瘤组织浓度逐渐增加;注射24 h时,肿瘤组织浓度达到最高(20.56%),而肺脏浓度最低(6.73%)。
     11 CFSE标记的CIK细胞经瘤旁注射荷瘤小鼠3 h后,同样可在肺、肝、脾、外周血、肿瘤中观察到绿色荧光,肺脏的分布浓度最高(36.83%),其次是肿瘤组织(25.75%);注射6 h时,外周血浓度达到最高(48.02%),其他器官浓度逐渐下降;注射24 h时,外周血浓度为23.41%,肿瘤组织为13.18%,肝、脾、肺中的浓度下降到3%以下。
     结论:
     1 CIK细胞在体内外对宫颈癌Hela-luc细胞均有较强的杀伤作用,在体内能明显抑制荷瘤小鼠肿瘤的生长,其作用机制可能与促进免疫细胞产生IFN-γ有关。
     2将CIK细胞经不同途径注射荷瘤小鼠后,可以广泛分布于全身器官,但在各脏器的分布与输注途径的不同有关。其中腹腔注射可能适用于体腔内肿瘤及恶性积液的治疗,而瘤旁注射可能适用于体表肿瘤的治疗。
Objective: The BALB/c nude mice inoculated with cervical cancer cell line Hela-luc were used as nude mouse xenograft model. To study the anti-tumor activity of cytokine-induced killer cells (CIK cells) against Hela-luc cells in vitro and in vivo. To analyse the characteristics of the distribution of CIK cells in cervical cancer nude mouse xenograft model. This research will provide theoretical for clinical application of CIK cells.
     Methods:
     1 The preparation and proliferation of CIK cells. The blood samples were obtained from anonymous healthy human volunteers. Peripheral blood mononuclear cells (PBMC) were isolated using standard Ficoll density gradient centrifugation. PBMC were induced to become CIK cells in vitro at the presence of interferon-gamma (IFN-γ), interleukin-2 (IL-2), CD3 monoclonal antibody (CD3McAb) and interleukin- 1α(IL-1α), in a certain sequence. CIK cells were incubated in a culture system that was renewed every 3 days with fresh medium include IL-2. To determinate proliferation response and draw curve of CIK cells growth every 7 days. After 14 days, CIK cells were collected.
     2 The phenotype and cytokine production of CIK cells were determined. Flow cytometry analysis was used to analyze the phenotype of CIK cells. The expression level of IFN-γmRNA in CIK cells was detected through semi-quantitative RT-PCR assay in 0, 7, 14, 21, 28, 35 days.
     3 Killing activity of CIK cells were determined in vitro. After 14 days, CIK cells by cytokines were collected in vitro. Logarithmic phase of Hela-luc cells were collected. CIK cells and PBMC were used as effector cells, respectively. Hela-luc cells were used as target cells. CIK cells and PBMC were added at specified effector: target (E: T) ratios (20:1, 40:1) and incubated after 48 hours, respectively. Killing activity of CIK cells and PBMC against Hela-luc cells was measured by MTT assay.
     4 After Wright-Giemsa’s stainning, morphological images of Hela-luc cells after CIK cells treated were observed by light microscope.
     5 The nude mouse was inoculated Hela-luc cells to establish tumor model. Logarithmic phase of Hela-luc cells were collected into single-cell suspension. To adjust cell concentration 1×108/ml. Hela-luc cells were injected in the BALB/c nude mice’s right department of shoulder (0.1ml, 1×107 once) to establish nude mouse xenograft model.
     6 Anti-tumor effect of CIK cells in vivo. After nude mice were injected tumor cells 7 days, the CIK cells (experiment group) or PBS (control group) was re-infused through tumor adjacent in tumor barring mice. Every group has 6 mice. Tumor changing and treatment effect were detected using in vivo Xenogen IVIS Imaging System. Peripheral blood of tumor barring mice, tumor tissue, lung, liver and spleen were collected after treatmented in two groups.
     7 The level of IFN-γin peripheral blood of tumor barring mice was detected by ELISA assay.
     8 After HE staining, the histopathological changed and tumor cells infiltrated of liver, lung and spleen were observed by microscope.
     9 CIK cells were labled by 5, 6-carboxyfluorescein diacetate succinimidyl ester (CFSE), and labled situation observed by laser scanning confocal microscope.
     10 Distribution of CIK cells in nude mouse xenograft model. CIK cells labled by CFSE were injected to tumor barring mice via peritoneal cavity or tumor adjacent. Control group didn’t do any treatment. Collected peripheral blood, tumor tissue, lung, liver and spleen in three groups after injected CIK cells 3 hours, 6 hours, 12 hours, 24 hours. Above-mentioned organizations were made of single-cell suspension. A part of single-cell suspension was detected CIK cells concontration by flow cytometry. Another part of single-cell suspension was observed distribution situation of CIK cells in tumor tissue by laser scanning confocal microscope.
     Results:
     1 The human peripheral blood mononuclear cells were cultured by different cytokines in vitro. Induced differentiation of CIK cells grew rapidly at 7 days and grew up to the peak at 14 days. The multiplication of CIK cells was highest reached 100 times.
     2 The CD3+, CD8+, CD3+CD56+ T cells’percentage were largely increased in the cells culturing process. And they expression rates may reach above 92%, 42.12%, 63.34% at 14 days, respectively. CD3+CD56+ T cells’percentage increased 57.5 times than beginning (1.10%).
     3 The expression level of IFN-γmRNA in CIK cells was gradually decreased in vitro cultured times. The expression level of IFN-γmRNA in CIK cells up to maximum was 14 days in cultured times. The expression level of IFN-γmRNA in CIK cells was very low in control group.
     4 At an effector-target cell ratio of 20: 1, 40: 1 after 48 hours, CIK cells showed higher cytotoxic effect for Hela-luc cells with the results are (51.16±2.64)% and (72.14%±4.21)% as measured by the MTT assay, respectively. The killing cytotoxic were (16.33±3.09)% and (21.26±2.71)% in PBMC, respectively.
     5 After Wright-Giemsa stainning, microscope observed: The normal Hela-luc cells were diamond-shaped or polygons, full shape, nuclear stainning were equal. A number of effector cells arounded the target cells to forming flower ring, and the target cell membrane changed loose, and cytoplasm had many vacuole in experiment group.
     6 After subcutaneous injected Hela-luc cells 7 days, the nude mice xenograft rate was 100%. Nude mice xenograft model were successful established.
     7 The tumor size in experiment group were smaller than that in control group after CIK cells treated (p<0.05). The inhibitory rates of the fifth week, eighth week were 47.18% and64.38%, respectiveily.
     8 The level of IFN-γin experiment group (61.92±6.49) pg/ml was higher than that in control group (34.30±1.78) pg/ml (p<0.05).
     9 The histopathological changed of tumor had no obviously difference between experiment group and control group. Alveola walls were integrity and cancer nodules didn’t find in liver, but there were many inflammatory cells in two groups. Cancer nodules were found in spleen in two groups. Many cancer nodules didn’t find in lung, but there were many inflammatory cells infiltrated in experiment group. Many tumor nodules were found in control group lung.
     10 Lung, liver, spleen, peripheral blood and tumor tissue were observed green fluorescence by laser scanning confocal microscope after injected CIK cells 3 hours via peritoneal cavity. The highest concentration of CIK cells was the lung (32.22%). Second was the liver. The concentration of CIK cells in tumor tissue was only 9.44%. After injected CIK cells 6 hours, the concentrations of CIK cells in lung, liver and spleen were gradually decreased; but the concentrations of CIK cells in peripheral blood and tumor tissue were gradually increased. After injected CIK cells 24 hours, the highest concentration of CIK cells was the tumor tissue (20.56%), and the lowest concentration of CIK cells was the lung (6.73%).
     11 Lung, liver, spleen, peripheral blood and tumor tissue were also observed green fluorescence by laser scanning confocal microscope after injected CIK cells 3 hours via tumor adjacent. The highest concentration of CIK cells was the lung (36.83%). Second was the tumor tissue (25.75%). After injected CIK cells 6 hours, the highest concentration of CIK cells was peripheral blood (48.02%), and the concentrations of CIK cells in other organs were gradually decreased. After injected CIK cells 24 hours, the concentration of CIK cells in peripheral blood was 23.41% and the concentration of CIK cells in tumor tissue was 13.18%; the concentration of CIK cells in liver, spleen and lung dropped to below 3%.
     Conclusions:
     1 CIK cells were highly efficient cytolytic effector cells which have a stronger significant suppression against growth of cervical cancer’s Hela-luc cells in vivo and in vitro. The CIK cells showed potent anti-tumor activity in animal experiment,and its mechanisms was maybe relate with IFN-γ.
     2 The CIK cells were extensively distributed to organs after injected via peritoneal cavity or tumor adjacent. There is relationship between distribution situation of CIK cells and infusion ways. Infusion of CIK cells via peritoneal cavity maybe suit for malignant tumor and malignant effusions in body cavity; and application by means of tumor adjacent should suit for body surface tumor.
引文
1黄体龙,杨跃煌.外周血单个核细胞冻存后诱导为CIK细胞及其抗肿瘤效应的研究[J].中国小儿血液及肿瘤杂志,2007,12(5):204-208.
    2 Schmidt-Wolf IG, Negrin RS, Kiem HP, et al. Use of a SCID mouse/human lymphoma model to evaluate cytokine-induced killer cells with potent antitumor cell activity. J Exp Med, 1991, 74:139-149.
    3 Kim HM, Lim J, Yoon YD, et al. Anti-tumor activity of ex vivo expanded cytokine-induced killer cells against human hepatocellular carcinoma. Int Immunopharmacol, 2007, 7: 1793-1801.
    4 Takayama T, Sekine T, Makuuchi M, et al. Adoptive immuno- therapy to lower postsurgical recurrence rates ofhepatocellular carcinoma:a randomised trial. Lancet, 2000, 356: 802-807.
    5 Kornacker M, Moldenhauer G, Herbst M, et al. Cytokine- induced killer cells against autologous CLL: direct cytotoxic effects and induction of immune accessory molecules by interferon-gamma. Int J Cancer, 2006, 119:1377-1382.
    6 Kim HM, Lim J, Park SK, et al. Antitumor activity of cytokine-induced killer cells against human lung cancer. Int Immunopharmacol, 2007, 7:1802-1807.
    7 Yang XJ, Huang JA, Lei W, et al. Antitumor effects of cocultured dendritic cells and cytokine-induced killer cells on lung cancer in vitro and in vivo. Ai Zheng, 2006, 25:1329-1333.
    8 Kim HM, Kang JS, Lim J, et al. Inhibition of human ovarian tumor growth by cytokine-induced killer cells. Arch Pharm Res, 2007, 30:1464-1470.
    9 Wang W, Epler J, Salazar LG, et al. Recognition of breast cancer cells by CD8+ cytotoxic T-cell clones specific for NY-BR-1. Cancer Res, 2006, 66:6826-6833.
    10 Sun S, Li XM, Li XD, et al. Studies on inducing apoptosis effects and mechanism of CIK cells for MGC-803 gastric cancer cell lines. Cancer Biother Radiopharm, 2005, 20:173- 180.
    11 Shibuya K, Mathers CD, Boschi-Pinto C, et al. Global and regional estimates of cancer mortality and incidence by site:Ⅱ. Results forthe global burden of disease 2000 [J]. BMCCancer, 2002, 2 (1) :37.
    12 Parkin DM, Bray F, Felay J, et al. Estimating the world cancer burden [J]. Int J Cancer, 2001, 94: 153-156.
    13刘曙正,张建营,王凯娟,等.河南省1985年至2004年宫颈癌死亡率趋势分析[J].郑州大学学报(医学版),2007, 42(2):353-355.
    14 Vernerism R, Ito M, Baker J, et al. Engineering hematopoietic grafts: purified allogeneic hematopoietic stem cells plus expanded CD8+NK-T cells in the treatment of lymphoma [J]. Biol Blood Marrow Transplant, 2001, 7(10):532-542.
    15 Linn YC, Wang SM, Hui KM.Comparative gene expression profiling of cytokine-induced killer cells in response to acute myloid leukemic and acute lymphoblastic leukemic stimulators using oligonucleotide arrays[J]. Exp Hematol, 2005, 33(6):671-681.
    16任欢,邢淑贤,徐红薇,等. CIK的体内外增殖及体内外杀瘤活性的实验研究.中国肿瘤生物治疗杂志,1999, 6(1): 17-21.
    17 Francis KP, Joh D, Bellinger-Kawahara C, et al. Monitoring bioluminescent staphylococcus aureus infections in living mice using a novel luxABCDE construct. Infect Immun, 2000, 68:3594-3600.
    18孙萍,王怡,孙志东,等.利用萤光素酶标记的肿瘤细胞建立肝癌原位移植模型及其活体成像.生物技术通讯, 2008, 19 (3):383-385.
    19 Wang X, Rosol M, Ge S, et al. Dynamic tracking of humanhematopoietic stem cell engraftment using in vivo bioluminescence imaging. Blood, 2003, 102:3478-3482.
    20张辉,赵群,左连富,等.细胞因子诱导的杀伤细胞对卵巢癌耐药细胞SKOV3/CDDP的作用机制.细胞与分子免疫学杂志,2007, 23(12):1167-1171.
    21 Hwan MK, Jaeseung L, Yeo DY,et al. Anti-tumor activity of ex vivo expanded cytokine-induced killer cells against human hepatocellular carcinoma. International Immunopharmacology, 2007, 7:1793-1801.
    22 Verneris MR, Kornacker M, Mailander V, et al. Resistance of ex vivo expanded CD3+CD56+ T cells to Fas-mediated apoptosis [J]. Cancer Immunol Immunother, 2000, 49(6): 336- 345.
    23 Brandacher G,Winkler C, Schroecksnadel K, et al. Antitumoral activity of interferon-gamma involved in impaired immune function in cancer patients[J]. Curr Drug Metab, 2006, 7(6):599-612.
    24陈婕,周佳青,王家东.γ-干扰素对喉癌细胞血管内皮生长因子-C表达的影响.临床耳鼻咽喉头颈外科杂志,2008, 22 (3):108-111.
    25 Kornacker M,Moldenhauer G,Herbst M,et al.Cytokine-induced killer cells against autologous CLL: direct cytotoxic effects and induction of immune accessory molecules by interferon-gamma.Int J Cancer , 2006, 119:1377–1382.
    26徐道华,夏永鹏,邱宗荫,等.γ干扰素增强CTL对宫颈癌细胞杀伤作用及其机制研究.免疫学杂志, 2005, 21(3):205-207.
    27 Gey A, Kumari P, Sambandam A, et al. Identification and characterisation of a group of cervical carcinoma patients with profound downregulation of intratumoral Type 1 ( IFNgamma) and Type 2 (IL-4) cytokine mRNA expression [J]. Eur J Cancer, 2003, 39 (5):595-603.
    1 Hoefel D, Grooby WL, Monis PT, et al. A comparative study of carboxyfluorescein diacetate and carboxyfluoresce- in diacetate succinimidyl ester as indicators of bacterial activity. J Microbiol Methods, 2003, 52:379- 388.
    2丘凌,何珊,黄瑞,等.荧光染料CFSE检测淋巴细胞增殖的研究.中国血液流变学杂志, 2005, 15(1):31-32.
    3陈蕾蕾,陈军浩,孙雪梅,等.荧光染料CFSE作为细胞标记的特性研究[J].细胞与分子免疫学杂志,2004, 20(2):140-141.
    4 Dumitriu IE, Mohr W, Kolowos W, et al. 5,6-Carboxy- fluorescein diacetate succnimidyl ester-labeled apoptotic and necrotic as well as detergenttreated cells can be traced in composite cell samples. J Anal Biochem, 2001, 299:247-252.
    5岳欣,李慧,于津浦,等.不同输注途径对CIK细胞治疗后的体内分布的影响.中国免疫学杂志, 2007, 23(3):224-228.
    6李东,崔磊,舒朝锋,等.荧光活性染料DiI标记观察人骨髓基质干细胞与部分脱钙骨粘附的实验研究[J].中华创伤骨科杂志,2004, 6(3):311-314.
    7 Paroo Z, Bollinger RA, Braasch DA, et al. Validating bioluminescence imaging as a high-throughput, quantitativemodality for assessing tumor burden [J]. Mol Imaging, 2004, 3(2):117-124.
    8姜晓兵,赵洪洋,周凤,等. EGFP在EGFP/GDNF融合基因修饰骨髓基质干细胞中的示踪作用[J].中华神经外科疾病研究杂志,2004, 3(6): 529-532.
    9安秀梅,岳欣,李慧,等.应用结肠癌动物模型探讨CIK细胞的抗肿瘤活性和体内分布特点.中国中西医结合外科杂志,2008, 14(3):260-264.
    10 Hazelrigg MR, Hirsch JI, Merchant RE. Distribution of adoptively transferred, tumor-sensitized lymphocytes in the glioma-bearing rat [J] . J Neurooncol, 2002, 60(2):143.
    11 Joseph S, Ronald A, Craig A, et al. Donor cell cycling, trafficking, and accumulation during adoptive immunotherapy for murine lung metastases [J]. Cancer Research, 2004, 64 (6): 2183.
    1 Xia CQ, Kao KJ. Monocyte-derived CDIa+ dendritic cells generated in two different culture systems: immunophenatypic and functional comparison. Scand J Immunol, 2003, 57(4):324-332.
    2 Keilholz U, Weber J, Finke JH, et al. Immunologic monitoring of cancer vaccine therapy: results of a workshop sponsored by the society for biological therapy. J Immunother, 2002, 25(2):97-138.
    3岳欣,李慧,于津浦,等.不同输注途径对CIK细胞治疗后的体内分布的影响.中国免疫学杂志,2007, 23(3):224-228.
    4孙耘玉,陈宝安.CIK细胞在恶性血液系统疾病中的应用研究.现代医学,2004, 32(5):347-349.
    5 Linn YC, Hui KM. Cytokine-induced killer cells : NK-like T cells with cytotolytic specificity against leukemia [J]. Leuk Lymphoma, 2003, 44(9):1457-1462.
    6 Kim HM, Lim J, Yoon YD, et al. Anti-tumor activity of ex vivo expanded cytokine-induced killer cells against human hepatocellular carcinoma. Int Immunopharmacol, 2007, 7:1793-1801.
    7 Takayama T, Sekine T, Makuuchi M, et al. Adoptiveimmunotherapy to lower postsurgical recurrence rates of hepatocellular carcinoma: a randomised trial. Lancet, 2000, 356:802-807.
    8 Kornacker M, Moldenhauer G, Herbst M, et al. Cytokine-induced killer cells against autologous CLL: direct cytotoxic effects and induction of immune accessory molecules by interferon-gamma. Int J Cancer, 2006, 119:1377-1382.
    9 Kim HM, Lim J, Park SK, et al. Antitumor activity of cytokine-induced killer cells against human lung cancer. Int Immunopharmacol, 2007, 7:1802-1807.
    10 Yang XJ, Huang JA, Lei W, et al. Antitumor effects of cocultured dendritic cells and cytokine-induced killer cells on lung cancer in vitro and in vivo. Ai Zheng, 2006, 25:1329-1333.
    11 Kim HM, Kang JS, Lim J, et al. Inhibition of human ovarian tumor growth by cytokine-induced killer cells. Arch Pharm Res, 2007, 30:1464-1470.
    12 WangW, Epler J, Salazar LG, et al. Recognition of breast cancer cells by CD8+ cytotoxic T-cell clones specific for NY-BR-1. Cancer Res, 2006, 66:6826-6833.
    13 Sun S, Li XM, Li XD, et al. Studies on inducing apoptosis effects and mechanism of CIK cells for MGC-803 gastric cancer cell lines. Cancer Biother Radiopharm, 2005, 20:173-180.
    14 Adisak W, Sakdipat S, Suradej H, et al. Effectiveosteosarcoma cytolysis using cytokine-induced killer cells pre-inoculated with tumor RNA-pulsed dendritic cells. Journal of orthopaedic Research, 2005, 23(6):1460-1466.
    15 Hwan MK, Jaeseung L, Song-Kyu P, et al. Antitumor activity of cytokine-induced killer cells against human lung cancer. International Immunopharmacology, 2007, 7: 1802-1807.
    16张辉,赵群,左连富,等.细胞因子诱导的杀伤细胞对卵巢癌耐药细胞SKOV3/CDDP的作用机制.细胞与分子免疫学杂志,2007, 23(12):1167-1171.
    17李可,吕章春,陈海祥,等.抗原致敏DC诱导CIK细胞对肺腺癌细胞的杀伤作用.肿瘤学杂志,2008, 14(4):310- 313.
    18张嵩,王恩忠,白春学,等.共培养的树突状细胞与CIK细胞治疗结肠癌血源性肺转移的实验研究[J].肿瘤,2003, 23(6):448-451.
    19张锦英,束永前,黄普文,等.抗原负载树突状细胞刺激细胞因子诱导的杀伤细胞治疗晚期胃癌的临床观察.中国生化药物杂志,2005, 26(6):336-338.
    20巩新建,刘军权,李玺,等.进展期胃癌术后自身CIK细胞和树突状细胞联合治疗的临床观察.中国肿瘤临床,2007, 34(14):803-806.
    21葛薇,李长虹,张伟,等.树突细胞与细胞因子诱导的杀伤细胞共培养增强其体内外抗肿瘤活性[J].中华血液学杂志,2004, 25(5):277-280.
    22蒋东霞,徐虹,胡杰英,等.人红白血病细胞来源的DC对细胞因子诱导的杀伤细胞逆转多药耐药的影响.实用诊断与治疗杂志,2007, 21(7):494-498.
    23湛海伦,高新,邱剑光,等.抗原致敏DC与CIK共培养体外抗肾癌效应的研究.中国病理生理杂志,2006, 22(10):1993-1996.
    24王欢,周芳坚,王其京,等.负载自体肿瘤细胞裂解物的DC疫苗联合CIK治疗晚期肾癌的临床观察—附10例报告[J].癌症,2006, 25(5):625-630.
    25孟琬如,糜若然.卵巢癌患者腹水来源树突状细胞强化CIK细胞对卵巢癌细胞的杀伤作用.现代妇产科进展,2005, 14(2):115-118.
    26 Wei XC, Zhai XH, Zhao WL, et al. Research on the biological activity and anti-tumor effect against lymphoma cells of DC-CIK cells.Chinese-German Journal of Clinical Oncology, 2008, 7(11):666-669.
    27应学翔,邹强,徐永华,等.负载Her-2多肽的DCIK细胞对乳腺癌细胞杀伤作用研究.分子细胞生物学报,2007, 40(4):193-198.
    28 Fields RC, Shimizu K, Mule JJ, et al. Murine dendritic cells pulsed with whole tumor lystes mediate potent antitumor immune responses in vitro. Proc Acad Sci USA, 1998, 95(16):9482-9487.
    29 Banchereau J, Briere F, Caux C, et al. Immunobiology of dendritic cells. Annu Rev Immunol, 2000, 18:761-811.
    30 Reid SD, Penna G, Adorini L, et al. The control of T cell responses by dentritic cell subsets. Curr Opin Immunol,2000, 12:114-121.
    31 Mehta BA, Schmidt-Wolf IG, Weissman IL, et al. Two pathways of exocytosis of cytoplasmic granule contents and target cell killing by cytoline-induced CD3+CD56+ killer cells [J]. Blood, 1995, 86(9):3493-3499.
    32 Brandacher G, Winkler C, Schroecksnadel K, et al. Antitumoral activity of interferon- gamma involved in impaired immune function in cancer patients [J]. Curr Drug Metab, 2006, 7:599-612.
    33陈婕,周佳青,王家东.γ-干扰素对喉癌细胞血管内皮生长因子-C表达的影响.临床耳鼻咽喉头颈外科杂志,2008, 22(3):108-111.
    34 Kornacker M, Moldenhauer G, Herbst M, et al. Cytokine-induced killer cells against autologous CLL: direct cytotoxic effects and induction of immune accessory molecules by interferon-gamma. Int J Cancer, 2006, 119 (6):1377-1378.
    35 Verneris MR, Kornacker M, Mailander V, et al. Resistance of ex vivo expanded CD3+CD56+T cells to Fas-mediated apoptosis[J]. Cancer Immunol Immunother, 2000, 49(6):336-345.
    36陈月,邹红岩,马云,等. IFN-γ促进树突状细胞分化成熟的实验研究.现代检验医学杂志,2008, 23(3):46-48.
    37 Marten A, Ziske C, Schottker B, et al. Interactions between dendritic cells and cytotkine-Induced killer cells lead to an activation of both populations [J]. J Immuno1, 2001, 24(6):502-510.
    38 Tsiatas ML, Gritzapis AD, Cacoullos NT, et al. A novel culture environment for generating mature human dentritic cells from peripheral blood CD14+ cells [J]. Anticancer Res, 2001, 21(2A):1199-1206.
    39 Liebowitz DN,Lee KP,June CH.Costimulatory approaches to adoptive immunotherapy[J]. Curr Opin Oncol, 1998, 10(6):533-541.