用户名: 密码: 验证码:
Treg细胞和IL-18在脑胶质瘤中的作用研究
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:脑胶质瘤,尤其是高级别胶质瘤是颅内最常见的恶性肿瘤,肿瘤呈浸润性生长,手术很难将其全部切除,尽管结合放化疗等综合治疗,但胶质瘤患者的预后仍十分不理想,复发率高,高级别胶质瘤患者的生存时间甚至不足1年。随着分子生物学研究的不断进展,胶质瘤的免疫治疗逐步得到人们的重视。
     胶质瘤细胞的侵袭性与其复杂的病理生理学特点有关,其逃逸免疫监视能力限制了机体产生有效抗肿瘤免疫反应,调节性T细胞(CD4~+CD25~+Foxp3~+ T细胞,Treg)在肿瘤细胞免疫逃逸过程中扮演重要角色。Treg细胞具有免疫抑制作用,对维持机体免疫稳态发挥着重要作用。Treg细胞在抑制效应性T细胞功能的同时,也降低机体对肿瘤细胞的免疫反应,间接促进了肿瘤细胞的生长。在多种恶性肿瘤组织中(如乳腺癌,卵巢癌,肺癌,肝癌,恶性淋巴瘤等)有大量Treg细胞浸润,其在患者外周血淋巴细胞中的比例也明显升高。并且,Treg细胞的数量与肿瘤的发展和预后呈负相关,清除Treg细胞或抑制其功能有助于机体抗肿瘤免疫功能的恢复。
     Treg细胞分为天然型和诱导型,前者由胸腺产生,后者可在外周由非调节性T细胞转化而来。在肿瘤微环境中,异常增多的Treg细胞也分为两类,一种是在肿瘤细胞趋化下,由外周迁徙而来,一种是在肿瘤局部特定环境中由非调节性T细胞转化而来。浸润在肿瘤中的Treg细胞在肿瘤细胞作用下分化成熟,具有肿瘤特异性,可阻止效应性T细胞(主要是CD8~+和CD4~+T细胞)的功能。在肿瘤局部,Treg细胞和效应性T细胞在数量上的不平衡是引起肿瘤细胞免疫逃逸的重要因素。因此打破这种不平衡成为肿瘤免疫治疗新的策略。
     白细胞介素18(IL-18)作为前炎因子,常高表达在多种自身免疫疾病组织及体液中,如类风湿性关节炎、结节病、红斑狼疮等。在这些患者体内,Treg细胞明显减少,而效应性CD8~+T细胞功能由于失去Treg细胞的抑制作用而明显上调,导致自身免疫性疾病发生。IL-18的高表达应与Treg细胞的减少存在某种关联。此外,IL-18本身也具有明显抗肿瘤功能,包括诱导T细胞和NK细胞产生IFN-γ,增强NK细胞和CTL细胞的细胞毒性,促进活化的CD4~+T细胞分化成为辅助性T细胞和效应性T细胞。在肿瘤动物模型研究及临床试验中,IL-18已被证实具有抗肿瘤功能,但在肿瘤微环境中是否能够逆转Treg细胞的免疫抑制作用,尚缺乏研究报道。本研究旨在探讨Treg细胞在脑胶质瘤组织中的数量与肿瘤发展的关系,探讨其免疫抑制性功能的机制,以及IL-18对Treg细胞的调节作用。
     方法:
     1收集2007年9月至2009年9月在河北医科大学第四医院神经外科治疗的脑胶质瘤患者的外周血及肿瘤组织标本,共76例。同期因脑疝行脑内减压术获得的正常脑组织标本及健康人群外周血标本作为阴性对照。
     分离肿瘤组织及正常脑组织中浸润淋巴细胞及患者、健康对照者外周血单个核细胞。采用流式细胞术检测组织中及外周血中Treg细胞、CD4~+T CD8~+T细胞的含量及比率,分析其与脑胶质瘤患者临床、病理学特征及预后的关系。
     采用ELISA法测定脑胶质瘤患者外周血中TGF-β1的含量。采用逆转录聚合酶链反应(RT-PCR)技术定量检测外周血单个核细胞中Foxp3mRNA的含量,分析两者表达的关系。
     2收集2008年9月至2009年9月间在河北医科大学第四医院神经外科治疗的5例脑胶质瘤患者手术切除的新鲜肿瘤组织标本。采集同期健康志愿者外周血标本及行脑内减压术获得正常脑组织为对照。
     分别培养正常人脑星形胶质原代细胞及原代胶质瘤细胞,采用免疫组织化学染色法进行鉴定。胶质瘤组织体外原代培养细胞及星形胶质细胞体外原代培养均获成功,并经免疫组化染色鉴定、证实。分离组织中浸润淋巴细胞及外周血单个核细胞后,采用Mini MACS免疫磁珠分离技术获得组织和外周血中CD4~+CD25~+、CD4~+CD25-及CD8~+T细胞。经免疫磁珠两次分选后,CD4~+CD25~+T细胞的纯度达89~92%,CD4~+CD25-T细胞纯度在90%以上,CD8~+T细胞纯度达87%~ 94%。按照不同比例将CD4~+CD25~+ T和CD4~+CD25-T细胞或CD8~+T细胞共同培养,采用[3H]-TdR掺入法检测CD4~+CD25~+ T对CD4~+CD25-T细胞及CD8~+T细胞增殖的影响。
     采用Western Blot法对不同来源的CD4~+CD25~+T细胞中Foxp3、CTLA-4、GITR、CCR4和CCR8蛋白进行定量检测。采用ELISA法对胶质瘤组织原代培养细胞及不同来源CD4~+CD25~+T细胞培养上清中TGF-β1、CCL12和CCL22进行定量检测。
     3应用逆转录病毒载体LXSN,将插入IL-18基因的质粒感染PA317包装细胞,经G418筛选后获得表达IL-18分子的PA317阳性细胞克隆,用IL-18/PA317培养上清转染9L细胞,筛选出高表达IL-18的IL-18/9L细胞克隆,传代培养。
     将雄性F344大鼠随机分为2组,每组15只,分别颅内接种9L细胞和IL-18/9L细胞,每组分别在颅内接种肿瘤细胞后7天、14天、21天时各处死3只大鼠,每组剩余的6只大鼠用于观察生存期。
     采用逆转录聚合酶链反应(RT-PCR)技术定量检测各组大鼠脑胶质瘤组织中IL-18mRNA的表达。在颅内接种肿瘤细胞21天时,采用[3H]-TdR掺入法检测各组大鼠脾细胞增殖反应情况,ELISA法检测各组大鼠脾细胞IFN-γ的产生情况。采用流式细胞术检测各组胶质瘤组织中Treg细胞、CD4~+T、CD8~+T细胞的含量及比率,分析其与大鼠胶质瘤生长及生存期的关系。取接种9L细胞21天时的大鼠肿瘤组织,分离肿瘤组织中浸润淋巴细胞,利用Mini MACS免疫磁珠分离系统,分选CD4~+CD25~+T细胞和CD4~+CD25-T细胞,将两者共同培养,培养基中加入IL-18,观察CD4~+CD25~+T细胞免疫抑制功能的变化。
     结果:
     1胶质瘤患者肿瘤组织和外周血单个核细胞中Treg细胞比例变化:流式细胞学检测结果显示,星形胶质瘤患者外周血单个核细胞中CD4~+CD25~+Foxp3~+T细胞的含量为10.41±2.13%,明显高于健康人外周血(4.35±1.07%)(p<0.01)。脑胶质瘤组织中Treg细胞比例为35.45±2.47%,明显高于健康对照脑组织(Treg细胞含量2.53±0.75%)。胶质瘤组织中浸润的Treg细胞含量与肿瘤的恶性程度呈正相关,而与患者年龄、性别、肿瘤大小、部位无关。不同病理学级别胶质瘤组织中CD8~+T细胞比例无明显差别(p>0.05)。
     胶质瘤患者TIL或PBMC中Treg细胞比例变化:将胶质瘤患者TIL中Treg细胞所占比例的均值(M)作为阈值,分为高比值组(≥M)和低比值组(0.05)。而CD8~+T细胞在肿瘤组织的浸润数量亦与患者预后无关(p>0.05)。而以肿瘤组织中CD8~+T/Treg细胞比值作为观测指标时,其比值与胶质瘤患者预后呈正相关,高比值组患者的生存期明显长于低比值组,两者具有显著差异(p<0.05)。
     胶质瘤患者外周血单个核细胞Foxp3mRNA表达及TGF-β1含量:胶质瘤患者外周血单个核细胞中均可见Foxp3mRNA表达,明显高于健康人,并随胶质瘤恶性级别的增加而升高(p<0.05)。胶质瘤患者外周血中均可检测到TGF-β1,并随着肿瘤恶性程度的升高而增加。直线回归分析显示,外周血TGF-β1含量与PBMC中Foxp3mRNA的表达呈正相关(r=0.940,p<0.05)。
     2不同来源Treg细胞F0xp3、CTLA-4、GITR蛋白表达:Western Blot分析结果显示,不同来源的CD4~+CD25~+T细胞均检测到Foxp3、CTLA-4及GITR蛋白表达,而CD4~+CD25-T细胞无相应蛋白表达。源自胶质瘤患者外周血及肿瘤组织中的Treg细胞,Foxp3表达明显高于健康人。而在不同来源CD4~+CD25~+ T细胞之间,CTLA-4和GITR的表达并无明显区别(p>0.05)。源自胶质母细胞瘤患者TIL中的CD4~+ CD25~+T细胞高表达CCR4(0.64±0.21),显著高于健康者外周血中CD4~+CD25~+T细胞的含量(0.27±0.13)(p<0.05)(Fig2-8),但CCR8在胶质母细胞瘤患者中和健康者外周血中均未见明显表达。
     CD4~+CD25~+T细胞对CD4~+CD25-T及CD8~+T细胞增殖的影响:在抗人CD3单克隆抗体作用下,将不同比例CD4~+CD25~+T细胞和CD4~+CD25-T细胞及CD8~+T细胞共同培养,CD4~+CD25~+T细胞可明显抑制CD4~+CD25-T细胞及CD8~+T细胞的增殖。随着CD4~+CD25~+T细胞数量的增加,对CD4~+CD25-T细胞及CD8~+T细胞的抑制率逐步升高。
     原代胶质瘤细胞、不同来源CD4~+CD25~+T细胞培养上清中TGF-β1的水平:ELISA检测结果显示,体外培养的原代胶质瘤细胞及不同来源CD4~+CD25~+T细胞均分泌TGFβ1,而在正常人脑星形胶质细胞培养上清中,未检测到TGF-β1。在来源于胶质母细胞瘤患者TIL及PBMC的CD4~+ CD25~+T细胞中, TGF-β1的分泌量分别为6.21±1.14μg/ml和5.47±1.05μg/ml,两者无明显差异(p>0.05),但均明显高于健康者外周血中的CD4~+CD25~+T细胞(2.32±0.61μg/ml)(p<0.05)。原代胶质瘤细胞大量分泌CCL22(12.17±1.23μg /ml),明显高于CCL12(4.68±0.83μg/ml)(p<0.05)。
     3接种亲代鼠脑胶质瘤细胞9L的F344大鼠肿瘤组织中,CD4~+CD25~+ Foxp3~+T细胞的浸润数量明显增多,并呈现出时间依赖性。
     接种IL-18/9L和9L细胞大鼠脑肿瘤组织中IL-18mRNA表达:分别取接种IL-18/9L细胞和9L细胞21d时的大鼠脑肿瘤组织,经RT-PCR分析,结果表明:IL-18/9L细胞组大鼠脑肿瘤组织中有IL-18mRNA表达,而接种9L细胞组大鼠脑肿瘤组织中无IL-18mRNA表达,说明IL-18/9L细胞在荷瘤大鼠体内仍可表达IL-18。
     IL-18对大鼠脾细胞IFN-γ产生的影响:接种IL-18/9L细胞组大鼠脾细胞可产生高水平IFN-γ(178.2±13.1pg/ml),明显高于接种9L细胞组(40.7±5.3pg/ml)(p<0.01)。
     IL-18对CD4~+CD25~+ T细胞免疫抑制功能的影响:在不同CD4~+CD25~+T:CD4~+CD25-T细胞比例下,IL-18均不能影响CD4~+CD25~+ T细胞对CD4~+CD25-T细胞的增殖抑制,与对照组比较无显著差异(p>0.05)。
     接种IL-18/9L组大鼠生存期为66.3±5.02d,明显长于接种9L细胞组大鼠(43.5±3.87d)(p<0.05)。
     结论:
     1胶质瘤患者外周血、肿瘤组织中Treg细胞含量明显增加。胶质瘤组织中CD8~+T/Treg细胞比值与患者的预后呈正相关。Treg细胞在胶质瘤组织中的比例尚不能单独作为患者预后的指标。胶质瘤患者外周血中Treg细胞的数量与血浆TGF-β1的水平呈正相关。
     2 Treg细胞在胶质瘤中聚集和发挥免疫抑制作用的可能机制是:胶质瘤细胞通过CCL22/CCR4趋化作用,驱使Treg细胞由外周向肿瘤部位迁移,并在肿瘤部位通过分泌TGF-β1诱导非Treg细胞向Treg细胞转化和成熟、Treg细胞通过细胞-细胞接触和分泌抑制性细胞因子TGF-β1发挥免疫抑制作用,肿瘤组织中Treg细胞数量与其免疫抑制功能密切相关。
     3在荷9L/F344细胞大鼠脑胶质瘤模型中,Treg细胞数量明显增多,并呈现时间依赖性。在鼠脑胶质瘤模型中,IL-18可通过刺激脾细胞增殖,促进IFN-γ分泌,促进CD8~+T细胞在肿瘤部位的增殖,提高CD8~+ T/ Treg细胞的比率,解除Treg细胞在局部对效应性细胞的抑制作用可抑制鼠脑胶质瘤的生长,而IL-18在体外对Treg细胞增殖和功能无明显作用。
Objective: Gliomas, especially high-grade gliomas are the most common malignant brain tumor showed infiltrative growth. It’s very difficult to cut the tumor completely. The overall prognosis of glioma patients is still not ideal in spite of comprehensive treatment including radiotherapy and chemotherapy. The recurrence rate of glioma was high and the survival time for patients with high-grade glioma were even less than 1 year. With the continuous development of molecular biology, glioma immunotherapy is arousing people's attention gradually. The aggressive nature of this neoplasia is closely related to its complex pathophysiology. In particular, evasion of the immune system by gliomas limits effective anti-tumoral responses. Current evidence suggests a major role in the evasion of immune rejection by CD4~+CD25~+ regulatory T cells (Tregs).
     Tregs are lymphocytes that have a physiological role in the modulation of the immune response. Specifically, these cells prevent autoimmunity by inhibiting autoreactive effector T lymphocytes, which downregulates the anti-tumoral response and promotes the growth of tumor. Many studies have found a large population of Tregs were infiltrated in the tissue and blood of many malignancies including breast, ovarian, lung, liver carcinoma and malignant lymphoma. It’s suggested that the population of Tregs was correlated negatively with the progression and prognosis of tumor. Depletion of Tregs or inhibition of its function has been associated with recovery of anti-tumoral response.
     Tregs are divided into thymus-derived Tregs or natural Tregs (nTregs) and Inducible Tregs (iTregs). nTreg development takes place in the thymus and iTregs are very similar in function to nTregs but derive from non-Treg cells in the periphery under specific stimulation. The abnormal increasing Tregs in tumor microenvironment are also divided into two groups. One group includes Tregs migrated from periphery under the influence of chemokines. The other group are induced from CD4~+CD25–T cells upon the exposure to the suppressive cytokine milieu at the tumor site. Tregs separated from tumor infiltrated lymphocytes usually represent tumor-specific antigen. The differentiation and maturation of Tregs take place under the action of tumor cells, which prevent the function of effector T cells (mainly CD8~+, CD4~+). Imbalance between Tregs and conventional T cells play a role in the immune escape of tumor cells and recovery of imbalance may be a new strategy for tumor treatment.
     As pro-inflammatory cytokine, interleukin 18(IL-18) has also been implicated in multiple autoimmune associated disease, such as rheumatoid arthritis, sarcoidosis, lupus erythematosus. Tregs have been reported to be decreased or to have decreased functional activity in a number of autoimmune disorders, and it is possible that an imbalance of Tregs and effector T cells due to differential IL-18 signaling contributes to the loss of tolerance. Several preclinical studies have suggested that IL-18 may have significant antitumor effects. IL-18 enhances the production of IFN-γby T cells and NK cells and can augment the cytolytic activity of NK cells and cytotoxic T lymphocytes (CTL). IL-18 promotes the differentiation of activated CD4 T cells into helper effector cells of Th1 or Th2 type. It’s not clear, however, that IL-18 can manipulate the function of Tregs in tumor microenvironment.
     Aim of our investigation is to discuss the expression of Tregs in cancer tissue from patients with glioma, the correlation between the population of Tregs and progression of tumor, investigate the inhibitory mechanism of Tregs for immune fanction, analyze the manipulation of IL-18 on Tregs.
     Methods:
     1Tumor tissue and blood of 76 patients with astrocytoma were collected, who were operated in the department of neurosurgery of the 4th hospital affiliated to Hebei Medical University from September, 2007 to September, 2009. Normal brain tissue and blood were used for control, which were from patients with decompression surgery for herniation and health volunteers.
     TILs and PBMCs were separated from tumor tissue and blood. The frequency of Tregs, CD4~+T cells and CD8~+T cells and ratio between Tregs and other T cells in TILs and PBMC were detected by flow cytometry. Then, the frequency and ratio of Tregs were analyzed and observe the relationship between Tregs and clinical pathological characters, prognosis of patients with astrocytoma.
     Expression of TGF-β1 in periphery blood were analyzed by ELISA. The Foxp3mRNA in PBMC were detected by RT-PCR, and analyze the relationship between the expression of Foxp3mRNA and TGF-β1.
     2 Tumor tissue and blood of 5 patients with glioblastoma were collected, who were operated in the department of neurosurgery of the 4th hospital affiliated to Hebei Medical University from September, 2008 to September, 2009. Normal brain tissue and blood were used for control, which were from patients with decompression surgery for herniation and health volunteers. Primary glioma cells and primary normal astrocyte were cultured in vitro and identified by immunohistochemistry, respectively. Primary glioma cells and primary normal astrocytes were cultured successfully in vitro, and identified by immunohistochemistry. CD4~+CD25~+T cells, CD4~+CD25-T cells and CD8~+T cells from TIL and PBMC were isolated by immunomagnetic separation system. After magnetic activated cell sorting, the purity of CD4~+CD25-, CD8~+T cells was 89~92%, higher than 90% and 87~94%, respectively.CD4~+CD25-T cells were then co-cultured with CD4~+CD25~+T cells in different population. [3H]-TdR incorporation were used to determine the proliferation of CD4~+CD25-T cells.
     Expression of Foxp3, CTLA-4, GITR, CCR4 and CCR8 protein were detected by West Blot. The secretion of TGF-β1, CCL22 and CCL12 in the medium supernatant of primary glioma cells, primary normal astrocyte and different CD4~+CD25~+T cells.
     3 30 male Fisher344 rats were divided randomly into 2 groups: 9L group and IL-18/9L group.9L cells and IL-18/9L cells were inoculated into the right cerebral hemisphere, respectively. Three mice of each group were killed on the 7th, 14th and 21st day after inoculation of tumor cells, the spleens and tumors were obtained from mice. 6 rats remained of every group were observed for over survival time.
     Expression of IL-18mRNA in rat tumor tissue were detected by RT-PCR. [3H]-TdR incorporation were used to determine the proliferation of CD4~+CD25-T cells. The frequency of Tregs, CD4~+T cells and CD8~+T cells and ratio between Tregs and other T cells in TILs and PBMC were detected by flow cytometry on 21st day after inoculation of tumor cells. IFN-γproducted by splenocytes were detected with ELISA.
     On 21st day after inoculation of tumor cells, CD4~+CD25~+T cells, CD4~+CD25-T cells and CD8~+T cells from TIL and PBMC of rats were isolated by immunomagnetic separation system. CD4~+CD25-T cells were then co-cultured with CD4~+CD25~+T cells in different population and IL-18. [3H]-TdR incorporation were used to determine the proliferation of CD4~+CD25-T cells. The frequency and ratio of Tregs in TIL of every group on specified time were analyzed and observe the relationship between Tregs and tumor progression by flow cytometry.
     Results:
     1 The result of flow cytometry instructed that the mean frequency of Tregs in PBMC of patients with astrocytoma(10.41±2.13%) was higher than that of volunteers(4.35±1.07%, p<0.05). The mean frequency of Tregs in TIL of patients (35.45±2.47%) was higher than that of control(2.53±0.75%, p<0.05). In comparison to autologous patient blood, the frequency of Tregs in tumor tissue were correlated significantly with the malignancy of glioma but age, sex, tumor size and tumor location. There was not a significant difference of the frequency of CD8~+T cells in TILs according to pathological grade.
     In all patients were divided into higher group(≥M) and lower group(     Expression of Foxp3mRNA in blood of patients was significantly higher than in controls(p<0.05). Moreover, higher level of TGF-β1 in the blood of patients in comparison to the blood from control individuals(p<0.05). Expression of Foxp3mRNA in blood was correlated significantly with the level of TGF-β1(r=0.940, p<0.05).
     2 CD4~+CD25-, CD8~+T cells were respectively co-cultured with CD4~+CD25~+T cells in different population. With increasing population of CD4~+CD25~+T cells, the proliferation of CD4~+CD25-, CD8~+T cells was significantly suppressed by CD4~+CD25~+T cells.
     The result of ELISA showed that the level of TGF-β1 was observed in supernatant of primary glioma cells and different CD4~+CD25~+T cells but normal astrocytes. In comparison to CD4~+CD25~+T cells of health volunteers (2.32±0.61μg/ml), the higher secretion of TGF-β1 was observed in CD4~+CD25~+T cells from tumor tissue (6.21±1.14μg/ml) and autologous blood (5.47±1.05μg/ml). Moreover, the secretion of CCL22 was very higher than that ofCCL12.
     The result of West Blot instructed that higher level of CCR4, FOXP3 was measured in CD4~+CD25~+T cells from the TIL in comparison to that from control individuals (p<0.05). There was not a significant difference for CTLA-4, GITR between different CD4~+CD25~+T cells. Expression of CCR8 was not observed in any supernatant.
     3 For rats inoculated with 9L cells, a time dependent increase in the frequency of CD4~+CD25~+Foxp3~+Treg at the tumor bearing site was observed.
     Expression of IL-18mRNA was measured in IL-18/9L group but in 9L group, which indicated that IL-18/9L cell can represent IL-18 at site of tumor. The splenocytes from the rat inoculated with IL-18/9L cells can secret IFN-γ(178.2±13.1pg/ml) that were higher than with 9L cells (40.7±5.3pg/ml, p<0.05).
     In suppression assay, IL-18 failed to influence the inhibitory function of CD4~+CD25~+T cells in vitro.
     Compared with 9L group (43.5±3.87d), the mean overall survival time of IL-18/9L group (66.3±5.02d) was significant longer.
     Conclusion:
     1 Infiltration of Tregs is significantly higher in the tumor and blood of patients with astrocytoma. The ratio of CD8~+/Treg is a significant prognostic factor for the patients but Treg or CD8~+T cell alone. Expression of Foxp3mRNA in blood was correlated significantly with the level of TGF-β1 in patients with astrocytoma.
     2 Tregs infiltrated in glioma may migrated from periphere under the influence of CCR4/CCL22. Glioma cells can sectret high level TGF-β1 which may turn non-Tregs into Tregs. Cell-cell contact seems to be the important way by which Treg present inhibitory function in glioma. The population of Tregs play a important role in its function.
     3 For rats inoculated with 9L cells, a time dependent increase in the frequency of Treg at the tumor bearing site was observed. Although, IL-18 failed to influence the inhibitory function of CD4~+CD25~+T cells in vitro, it can increase the amount of CD8~+T cells, especially the ratio of CD8~+/Treg, promote the secretion of IFN-γand upregulate anti-tumor immunoresponse.
引文
1 Grauer O, Poschl P, Lohmeier A, et al. Toll-like receptor triggered dendritic cell maturation and IL-12secretion are necessary to overcome T-cell inhibition by gliomaassociated TGF-beta2. J Neurooncol, 2007,82, 151-161
    2 Platten M, Wick W, Weller M. Malignant glioma biology: role for TGF- beta in growth, motility, angiogenesis, and immune escape. Microsc Res Tech, 2001,52,401-410
    3 Nakano Y, Kuroda E, Kito T, et al. Induction of macrophagic prostaglandin E2 synthesis by glioma cells. J Neurosurg, 2006, 104, 574-582
    4 Wagner S, Czub S, Greif M, et al. Microglial/macrophage expression of interleukin 10 in human glioblastomas. Int J Cancer, 1999,82,12-16
    5 Wiendl H, Mitsdoerffer M, Hofmeister V, et al. A functional role of HLA- G expression in human gliomas: an alternative strategy of immune escape. J Immunol, 2002, 168,4772-4780
    6 Wiendl H, Mitsdoerffer M, Weller M. Hide-and-seek in the brain: a role for HLA-G mediating immune privilege for glioma cells. Semin Cancer Bio- l, 2003,13,343-351
    7 Hasegawa Y, Takanashi S, Kanehira Y, et al. Transforming growth factor beta level correlateswith angiogenesis tumor progression, and prognosis ofpat- ients with nonsmall cell lung carcinoma. Cancer, 2001, 91,964-971
    8 Curiel TJ, Coukos G, Zou L, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med, 2004, 10,942-949
    9 Lewkowicz P, Lewkowicz N, Sasiak A, et al. Lipopolysaccharide-activated CD4~+CD25~+T regulatory cells inhibit neutrophil function and promote their apoptosis and death [J]. J Immunol, 2006,177,7155-7163
    10 Von BoehmerH. Mechanisms of suppression by suppressor T cells. Nat Immuno, l 2005, 6,338-344
    11 El Andaloussi A, Lesniak MS. An increase in CD4~+CD25~+FOXP3~+ regulatory T cells in tumor-infiltrating lymphocytes of human glioblastoma multiforme. Neuro-oncol, 2006,8, 234-243
    12 Wood GW, Morantz RA. In vitro reversal of depressed T lymphocyte function in the peripheral blood of brain tumor patients. J Natl Cancer Inst, 1982,68,27-33
    13 Wood GW, Morantz RA. Depressed T lymphocyte function in brain tumor patients: monocytes as suppressor cells. J Neurooncol, 1983,1, 87-94
    14 Yamasaki T, Handa H, Yamashita J, et al. Characteristic immunological responses to an experimental mouse brain tumor. Cancer Res, 1983, 3, 4610-4617
    15 Curiel TJ, Coukos G, Zou L, et al. Specifc reruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. NatMed, 2004,18,942-949
    16 Yoshioka T, Miyamoto M, Cho Y, et al. Infiltrating regulatory T cell numbers is not a factor to predict patient’s survival in oesophageal squamous cell carcinoma. J Briti Med, 2008,98,1258-1263
    17 Eiiehi S, Sara HO, Jioung A, et al. Intraepithelial CD8~+ tumor-infiltrating lymphocytes and a high CD8~+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. PNAS, 2005,34,18538-18543
    18 Oshikiri T, Miyamoto M, Shiehinohe T, et al. Prognostic value of intratumoral CD8~+T lymphocyte in extrahepatic bile duct carcinoma asessential immune response. J Surg Oncol, 2003, 84,224-228
    19 Kronenberg M, Rudensky A. Regulation of immunity by self-reactive T cells. Nature, 2005, 435,598-604
    20 Sakaguchi S. Naturally arising Foxp3-expressing CD25~+CD4~+regulatory T cells in immunological tolerance to self and non-self. Nat Immunol, 2005, 6,345-352
    21 Strassmann G, Cole MD, Newman W. Regulation of colony-stimulating factor-1 dependent macrophage precursor proliferation by type beta transforming growth faetor. J Immunol, 1988,140,2645-2651
    22 Pyzik M, Piccirillo CA. TGF-beta1 modulates Foxp3 expression and regulatory activity in distinct CD4~+T cell subsets. J Leukoc Bio, 2007, 82, 335- 346
    23 Qiao M, Thornton AM, Shevach EM. CD4~+CD25~+regulatory T cells render naive CD4~+CD25–T cellsanergic and suppressive. Immunology, 2007, 120, 447-455
    24 Sehramm C, Protsehka M, Kohler HH, et al. Impairment of TGF-beta signaling in T cells increases susceptibility to experimental autoimmune heap- titisin miee. Am J Physiol Gastrointest Liver Physiol, 2003,284, 525-535
    25 Platten M, Wick W, Weller M. Malignant glioma biology: role for TGF-beta in growth, motility, angiogenesis, and immune escape. Microsc Res Tech, 2001,52,401-410
    26 Grauer O, Poschl P, Lohmeier A, et al. Toll-like receptor triggered dendritic cell maturation and IL-12 secretion are necessary to overcome T-cell inhibition by gliomaassociated TGF-beta1. J Neurooncol, 2007, 82,151-161
    1 Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science, 2003,299,1057-1061
    2 Fontenot JD, Rasmussen JP, Williams LM, et al. Regulatory T cell lineage specification by the forkhead transcription factor Foxp3. Immunity, 2005, 22,329-341
    3 Takahashi T, Tagami T, Yamazaki S, et al. Immunologic self-tolerance maintained by CD4~+CD25~+ regulatory T cells constitutively expressing cytotoxic T lymphocyte-associated antigen 4. Journal of experimental medical, 2000,192,303-310
    4 Shimizu J, Yamazaki S, Takahashi T, et al. Stimulation of CD4~+CD25~+ regulatory T cells through GITR breaks immunological self-tolerance. National Immunology, 2002,3,135-142
    5 McHugh RS, Whitters MJ, Piccirillo CA, et al. CD4~+ CD25~+ immuno regulatory T cells: gene expression analysis reveals a functional role for the glucocorticoid-induced TNF receptor. Immunity, 2002,16,311-312
    6 Fontenot JD, Rudensky AY. A well adapted regulatory contrivance: regulatory T cell development and the forkhead family transcription factor Foxp3. National Immunology, 2005,6,331-337
    7 Sutmuller RP, Morgan ME, Netea MG, et al. Toll like receptors on regulatory T cells: expanding immune regulation. Trends Immunology, 2006,27,387-393
    8 Sutmuller RP, den Brok MH, Kramer M, et al. Toll-like receptor 2 controls expansion and function of regulatory T cells. Journal of Clinical Investgation, 2006,116,485-494
    9 Woo EY, Chu CS, Goletz TJ, et al. Regulatory CD4~+CD25~+T cells in tumors from patients with early-stage non-small cell lung cancer andlate-stage ovarian cancer. Cancer Research, 2001,61,4766-4772
    10 Liyanage UK, Moore TT, Joo HG, et al. Prevalence of regulatory T cells is increased in peripheral blood and tumor microenvironment of patients with pancreas or breast adenocarcinoma. Journal of Immunology, 2002, 169, 27 56 -2761
    11 Curiel TJ, Coukos G, Zou L, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. National Medicine, 2004,10,942-949
    12 Viguier M, Lemaitre F, Verola O, et al. Foxp3 expressing CD4~+CD25 (high) regulatory T cells are overrepresented in human metastatic melanoma lymph nodes and inhibit the function of infiltrating T cells. Journal of Immunology, 2004,173,1444-1453
    13 Jones E, Dahm-Vicker M, Simon AK, et al. Depletion of CD25~+ regulatory cells results in suppression of melanoma growth and induction of autoreativity in mice. Cancer Immunology, 2002,21,1-3
    14 Wood GW, Morantz RA. In vitro reversal of depressed T lymphocyte function in the peripheral blood of brain tumor patients. J Nat Cancer Inst, 198 2,68,27-33
    15 Wood GW, Morantz RA. Depressed T lymphocyte function in brain tumor patients: monocytes as suppressor cells. J Neurooncol, 1983,1,87-94
    16 Yamasaki T, Handa H, Yamashita J, et al. Characteristic immunological responses to an experimental mouse brain tumor. Cancer Res, 1983,43, 46 10-4617
    17 Ito A, Shinkai M, Honda H, et al. Augmentation of MHC class I antigen presentation via heat shock protein expression by hyperthermia. Cancer Immunol Immunother, 2001,50,515-522
    18 Grauer O, Poschl P, Lohmeier A, et al. Toll-like receptor triggered dendritic cell maturation and IL-12 secretion are necessary to overcome T-cell inhibition by gliomaassociated TGF-beta2. J Neurooncol, 2007,82, 151- 161
    19 Platten M, Wick W, Weller M. Malignant glioma biology role for TGF-beta in growth, motility, angiogenesis, and immune escape. Microsc Res Tech, 2001,52,401-410
    20 Weller M, Fontana A. The failure of current immunotherapy for malignant glioma. Tumor-derived TGF-beta, T-cell apoptosis, and the immune privylege of the brain. Brain Res Brain Res Rev, 1995,21,128-151
    21 Lauro GM, Di Lorenzo N, Grossi M, et al. Prostaglandin E2 as an immune modulating factor released in vitro by human glioma cells. Acta Neuropathol (Berl), 1986,69,278-282
    22 Nakano Y, Kuroda E, Kito T, et al. Induction of macrophagic prostaglandin E2 synthesis by glioma cells. J Neurosurg, 2006, 104, 574-582
    23 Wagner S, Czub S, Greif M, et al. Microglial/macrophage expression of interleukin 10 in human glioblastomas. Int J Cancer, 1999,82,12-16
    24 El Andaloussi A, Han Y, Lesniak MS. Prolongation of survival following depletion of CD4~+ CD25~+ regulatory T cells in mice with experimental brain tumors. J Neurosurg, 2006,105,430-437
    25 El Andaloussi A, Lesniak MS. An increase in CD4~+CD25~+FOXP3~+ regulatory T cells in tumor-infiltrating lymphocytes of human glioblastoma multiforme. Neuro-oncol, 2006,8,234-243
    26 El Andaloussi A, Lesniak MS. CD4~+CD25~+FOXP3~+T-cell infiltration and heme oxygenase-1 expression correlate with tumor grade in human gliomas. J Neurooncol, 2007,83,145-152
    27 El Andaloussi A, Sonabend AM, Han Y, et al. Stimulation of TLR9 with CpG ODN enhances apoptosis of glioma and prolongs the survival of mice with experimental brain tumors. Glia, 2006,54,526-535
    28 Sakaguchi S. Naturally arising Foxp3-expressing CD4~+CD25~+regulatory T cells in immunological tolerance to self and non-self. Nat Immunol, 2005, 6,345-352
    29 Sanchez-Guajardo V, Tanchot C, O’Malley JT, et al. Agonist-driven development of CD4~+CD25~+FOXP3~+regulatory T cells requires a second sig- nalmediated by Stat6. J Immunol, 2007,178, 7550-7556
    30 Kronenberg M, Rudensky A. Regulation of immunity by self-reactive T cells. Nature, 2005, 435,598-604
    31 Birebent B, Lorho R, Lechartier H, et al. Suppressive properties of human CD4~+CD25~+regulatoryT cells are dependenton CTLA-4 expression. Eur J Immunol, 2004, 34,3485-3496
    32 Nakamura K, Kitani A, Strober W. Cell contact-dependent immune suppression by CD4~+CD25~+regulatory T cells is mediated by cell surface bound transforming growth factor beta. J Exp Med, 2001,194,629-644
    33 Pyzik M, Piccirillo CA. TGF-beta1 modulates Foxp3 expression and regulatory activity in distinct CD4~+T cell subsets. J Leukoc Bio, 2007, 82,335- 346
    34 Curiel TJ, Coukos G, Zou L, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med, 2004, 10, 942-949
    35 Zou L, Barnett B, Safah H, et al. Bonemarrow is a reservoir for CD4~+CD25~+regulatory T cells that traffic through CXCL12 /CXCR4 signals. Cancer Res, 2004, 64, 8451-8455
    1 Okamura H, Tsutsi H, Komatsu T, et al. Cloning of a new cytokine that induces IFN-gamma production by T cells. Nature, 1995,378,88-91
    2 Ushio S, Nambaa M, Okura T, et al. Cloning of the cDNA for human IFN-gamma-inducing factor, expression in Escherichia coli, and studies on the biologic activities of the protein. J Immunol, 1996,156,4274-4279
    3 Nakanishi K, Yoshimoto T, Tsutsui H, et al. Interleukin-18 regulates both Th1 and Th2 responses. Annu Rev Immunol, 2001,19,423-474
    4 Mathur AN, Chang HC, Zisoulis DG, et al. T-bet is a critical determinant in the instability of the IL-17-secreting T-helper phenotype. Blood, 2006,108, 1595-1601
    5 Mathur AN, Chang HC, Zisoulis DG, et al. Stat3 and Stat4 direct development of IL-17-secreting Th cells. J Immunol, 2007,178,4901-4907
    6 Marshall DJ, Rudnick KA, McCarthy SG, et al. Inerleukin-18 enhances Th1 immunity and tumor protection of a DNA vaccine. Vaccine, 2006,24, 244-253
    7 Luo Y, Zhou H, Mizutani M, et al. A DAN vaccine targeting Fos-related antigen 1 enhanced by IL-18 induces long-lived T-cell memory against tumor recurrence. Cancer Res, 2005,65,3419-3427
    8 Tatsumi T, Gambotto A, Robbins PD, et al. Interleukin 18 gene transfer expands the repertoire of antitumor Th1-type immunity elicited by dendritic cell-based vaccines in association with enhanced therapeutic efficacy. Cancer Res, 2002,62,5853-5858
    9 Xu B, Dong CY, Zhang F, et al. Synergistic antileukemia effect of combinational gene therapy using murine beta-defensin 2 and IL-18 in L1210 murine leukemia model. Gene Ther, 2007,14,1181-1187
    10 Boraschi D, Dinarello CA. IL-18 in autoimmunity: review. J Clin Invest, 2006, 17,224-252
    11 Pittoni V, Bombardieri M, Spinelli FR, et al. Anti-tumor necrosis factor alpha treatment of rheumatoid arthritis(infliximab) selectively down regulates the production of interleukin 18 but not of IL12 and IL13. Ann Rheum Dis, 2002,61,723-725
    12 Sutmuller RP, van Duivenvoorde LM, van Elsas A, et al. Synergism of cytotoxic T lymphocyte-associated antigen 4 blockade and depletion of CD25~+ regulatory T cells in antitumor therapy reveals alternative pathways for suppression of autoreactive cytotoxic T lymphocyte responses. J Exp Med, 2001,194, 823-832
    13 Woo EY, Chu CS, Goletz TJ, et al. Regulatory CD4~+CD25~+T cells in tumors from patients with early-stage non-small cell lung cancer and late stage ovarian cancer. Cancer Res, 2001,61,4766-4772
    14 Yu P, Lee Y, Liu W, et al. Intratumor depletion of CD41 cells unmasks tumor immunogenicity leading to the rejection of late-stage tumors. J Exp Med, 2005,201,779-791
    15 Hegmans JP, Hemmes A, Hammad H, et al. Mesothelioma environment comprises cytokines and Treg cells that suppress immune responses. Eur Respir J, 2006, 27l, 1086-1095.
    16 Jones E, Dahm-Vicker M, Simon AK, et al. Depletion of CD251 regulatory cells results in suppression of melanoma growth and induction of autoreactivity in mice. Cancer Immun, 2002,2,1
    17 Carroll RG, Carpenito Carmine X, Shan XC, et al. Distinct effect of IL-18 on the engraftment and function of human effector CD8~+T cells and regulatory T cell. PLOS ONE, 2008,9,1-10
    18 Dinarello CA. Interleukin-18 and the pathogenesis of inflammatory diseases. Semin Nephrol, 2007, 27, 98-114
    19 Calvani N, Tucci M, Richards HB, et al. Th1 cytokines in the pathogenesis of lupus nephritis: the role of IL-18. Autoimmun Rev, 2005,4,542-548
    20 Dai SM, Shan ZZ, Xu H, et al. Cellular targets of interleukin-18 in rheumatoid arthritis. Ann Rheum Dis, 2007, 66: 1411-1418
    21 Greene CM, Meachery G, Taggart CC, et al. Role of IL-18 in CD4~+T lymphocyte activation in sarcoidosis. J Immunol, 2000,165,4718-4724
    22 Tedeschi A, Lorini M, Suli C, et al. Serum interleukin-18 in patients withchronic ordinary urticaria: association with disease activity. Clin Exp Dermatol, 2007,32, 568-570
    23 Efthimiou P, Kontzias A, Ward CM, et al. Adult-onset Still’s disease: can recent advances in our understanding of its pathogenesis lead to targeted therapy? Nat Clin Pract Rheumatol, 2007, 3, 328-335
    24 Hultgren O, Andersson B, Hahn-Zoric M, et al. Serum oncentration of interleukin-18 is up-regulated in patients with ANCAassociated vasculitis. Autoimmunity, 2007, 40,529-531
    25 Lin YJ, Wan L, Sheu JJ, et al. A/C polymorphism in the interleukin-18 coding region among Taiwanese systemic lupus rythematosus patients. Lupus,2002, 17,124-127
    26 Takada H, Ohga S, Mizuno Y, et al. Oversecretion of IL-18 in haemophagocytic lymphohistiocytosis: a novel marker of disease activity. Br J Haemato, 1999, 106,182-189
    27 Viglietta V, Baecher-Allan C, Weiner HL, et al. Loss of functional suppression by CD4~+CD25~+ regulatory T cells in patients with multiple scleraosis. Journal of Experimental Medicine, 2004,14,971-979
    28 Tomohide T, Andrea G, Paul D, et al. Interleukin 18 gene transfer expands the repertoire of antitumor Th1-type immunity elicited by dendritic cell- based vaccines in association with enhanced therapeutic efficacy. Cancer Res, 2002,62,5853
    29 Marshall DJ, Rudnick KA, McCarthy SG, et al. Interleukin-18 enhances Th1 immunity and tumor protection of a DNA vaccine. Vaccine, 2006, 24, 244-253
    1 Viel CT, Moore TT, Liyanage UK, et al. Linehan DC: Depletion of CD4~+ CD25~+ regulatory T cells promotes a tumor-specificimmune response in pancreas cancer-bearing mice. AnnSurg Oncol, 2006,13,1252-1258
    2 Antony PA, Piccirillo CA, Akpinarli A, et al. CD8~+ T cellimmunity again- st a tumor/self-antigen is augmented byCD4~+T helper cells and hindered by naturally occurring T regulatory cells. J Immunol, 2005,174,2591-2601
    3 Linehan DC, Goedegebuure PS. CD25~+CD4~+ regulatory T-cells in cancer. Immunol Res, 2005, 32,155-168
    4 Hiur T, Kagamu H, Miur S, et al. Both regulatory T cells and antitumor effector T cells are primed in the same draining lymph nodes during tumor progression. J Immunol, 2005,175,5058-5066
    5 Tien AH, Xu L, Helgason CD. Altered immunity accompanies disease progression in a mouse model of prostate dysplasia. Cancer Res, 2005, 65, 2947-2955
    6 Liu JY, Zhang XS, Ding Y, et al. The changes of CD4~+CD25~+/CD4~+ proportion in spleen of tumor-bearing BALB/c mice. J Transl Med, 2005, 3,5
    7 Golgher D, Jones E, Powrie F, et al. Depletion of CD25~+regulatory cells uncovers immune responses to shared murine tumor rejection antigens. Eur J Immunol, 2002,32,3267-3275
    8 Liyanage UK, Goedegebuure PS, Moore TT, et al. Increase prevalence of regulatory T cells (Treg) is induced by pancreas adenocarcinoma. J Immunother, 2006, 29,416-424
    9 Ghiringhelli F, Lamonier N, Schmitt E, et al. CD4~+CD25~+ regulatory T cells suppress tumorimmunity but are sensitive to cyclophosphamidewhich allows immunotherapy of established tumors to be curative. Eur J Immunol, 2004,34,336-344
    10 Schabowsky RH, Madiredii S, Sharma R, et al. Targeting CD4~+CD25~+ Foxp3~+ regulatory T-cells for the augmentation of cancer immunotherapy. Curr Opin Invest Drugs, 2007,8,1002-1008
    11 El Andaloussi A, Han Y, Lesniak MS. Prolongation of survival following depletion of CD4~+CD25~+ regulatory T cells in mice with experimental brain tumors. J Neurosurg, 2006, 105,430-437
    12 Sutmuller RP, van Duivenvoorde LM, van Elsas A, et al. Synergism of cytotoxic T lymphocyte- associated antigen 4 blockade and depletion of CD25~+regulatory T cells in antitumor therapy reveals alternative pathways for suppression of autoreactive cytotoxic T lymphocyte responses. J Exp Med, 2001, 194,823-832
    13 Sharma S, Yang SC, Zhu L, et al. Tumor cy clooxygenase-2/prostaglandin E2-dependent promotion of FOXP3 expression and CD4~+CD25~+ T regu- latory cell activities in lung cancer. Cancer Res, 2005, 65,5211-5220
    14 Turk MJ, Guevara-Pati?o JA, Rizzuto GA, et al. Concomitant tumor imm- unityto a poorly immunogenic melanoma is prevented by regulatoryT cells. J Exp Med, 2004,200,771-782
    15 Lin YC, Chang LY, Huang CT, et al. Effector/memory but not naive regu- latory T cells are responsible for the loss of concomitant tumor immunity. J Immunol, 2009,182,6095-6104
    16 Valzasina B, Piconese S, Guiducci C, et al. Tumor-induced expansion of regulatory? T cells by conversionof CD4~+CD25-lymphocytes is thymus and proliferationpindependent. Cancer Res, 2006,66,4488-4895
    17 Lutsiak ME, Tagaya Y, Adams AJ, et al. Tumor-induced impairment of TCR signaling results incompromised functionality of tumor-infiltrating regulatoryT cells. J Immunol, 2008,180,5871-5881
    18 Mizukami Y, Kono K, Kawaguchi Y, et al. CCL17 and CCL22 chemo- kines withintumor environment are related to accumulation of Foxp3~+ regulatory T cells in gastric cancer. Int J Cancer, 2008,122,2286-2293
    19 Curiel TJ, Coukos G, Zou L, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nat Med, 2004,10,942-949
    20 Tzankov A, Meier C, Hirschmann P, et al. Correlation of high numbers of intratumoral FOXP3~+regulatory T cells with improved survival in germinal center-like diffuse large B-cell lymphoma, follicular lymphoma and classical Hodgkin’s lymphoma. Haematologica, 2008,93,193-200
    21 Ghiringhelli F, Larmonier N, Schmitt E, et al. CD4~+CD25~+regulatory T cells suppress tumor immunity but are sensitive to cyclophosphamide which allows immunotherapy of established tumors to be curative. Eur J Immunol,2004,34,336-344
    22 Ghiringhelli F, Menard C, Puig PE, et al. Metronomic cyclophosphamide regimen selectively depletes CD4~+CD25~+ regulatory T cells and restores T and NK effector functions in end stage cancer patients. Cancer Immunol Immunother, 2007,56, 641-648
    23 Lord R, Nair S, Schache A, et al. Low dose metronomic oral cyclophosphamide for hormone resistant prostate cancer: a Phase II study. J Urol, 2007,177, 2136-2140
    24 Audia S, Nicolas A, Cathelin D, et al. Increase of CD4~+CD25~+ regulatory T cells in the peripheral blood of patients with metastatic carcinoma: a Phase I clinical trial using cyclophosphamide and immunotherapy to eliminate CD4~+CD25~+ T lymphocytes. Clin Exp Immunol, 2007,150, 523-530
    25 Levitt ML, Kassem B, Gooding WE, et al. Phase I study of gemcitabine given weekly as a short infusion for non-small cell lung cancer: results and possible immune system-related mechanisms. Lung Cancer, 2004, 43, 335-344
    26 Plate JM, Plate AE, Shott S, et al. Effect of gemcitabine on immune cells in subjects with adenocarcinoma of the pancreas. Cancer Immunol Immunother, 2005,54, 915-925
    27 Correale P, Cusi MG, Tsang KY, et al. Chemo-Immunotherapy ofMetastatic Colorectal Carcinoma With Gemcitabine Plus FOLFOX 4 Followed by Subcutaneous Granulocyte Macrophage Colony-Stimulating Factor and Interleukin-2 Induces Strong Immunologic and Antitumor Activity in Metastatic Colon Cancer Patients. J Clin Oncol, 2005,23, 8950-8958
    28 Barni S, Lissoni P, Paolorossi F, et al. Mitoxantrone as a single agent in pretreated metastatic breast cancer: effects on T lymphocyte subsets and their relation to clinical response. Tumori, 1991, 77, 227-231
    29 Hegde U, Chhabra A, Chattopadhyay S, et al. Presence of low dose of fludarabine in cultures blocks regulatory T cell expansion and maintains tumor-specific cytotoxic T lymphocyte activity generated with peripheral blood lymphocytes. Pathobiology, 2008,75, 200-208
    30 Yaqub S, Henjum K, Mahic M, et al. Regulatory T cells in colorectal cancer patients suppress anti-tumor immune activity in a COX-2 dependent manner. Cancer Immunol Immunother, 2007,57,813-821
    31 Kreitman RJ, Wilson WH, White JD, et al. Phase I trial of recombinant immunotoxin anti-Tac(Fv)-PE38 (LMB-2) in patients with hematologic malignancies. J Clin Oncol, 2000,18, 1622-1636
    32 Talpur R, Jones DM, Alencar AJ, et al. CD25 Expression Is Correlated with Histological Grade and Response to Denileukin Diftitox in Cutaneous T-Cell Lymphoma.J Invest Dermatol, 2006,126, 575-583
    33 Mahnke K, Schonfeld K, Fondel S, et al. Depletion of CD4~+CD25~+ human regulatory T cells in vivo: kinetics of Treg depletion and alterations in immune functions in vivo and in vitro. Int J Cancer, 2007,120, 2723-2733
    34 Wei S, Kryczek I, Edwards RP, et al. Interleukin-2 Administration Alters the CD4~+FOXP3~+T-cell pool and tumor trafficking in patients with ovarian carcinoma. Cancer Res, 2007, 67,7487- 7494
    35 Attia P, Phan GQ, Maker AV, et al. Autoimmunity correlates with tumor regression in patients with metastatic melanoma treated with anti-cytotoxic T-lymphocyte antigen-4. J Clin Oncol, 2005b, 23,6043- 6053
    36 Maker AV, Phan GQ, Attia P, et al. Tumor regression and autoimmunityinpatients treated with cytotoxic T lymphocyte-associated antigen 4 blockade and interleukin 2: a phase I/II study. Ann Surg Oncol, 2005b, 12,1005-1016
    37 Menard C, Ghiringhelli F, Roux S, Chaput N, et al. Ctla-4 blockade confers lymphocyte resistance to regulatory T-cells in advanced melanoma:surrogate marker of efficacy of tremelimumab? Clin Cancer Res, 2008,14,5242- 5249
    38 Cranmer LD, Hersh E. The role of the CTLA4 blockade in the treatment of malignant melanoma. Cancer Invest, 2007,25, 613-631
    39 Kim JD, Choi BK, Bae JS, et al. Cloning and characterization of GITR ligand. Genes Immun, 2003,4, 564-569
    40 Turk MJ, Guevara-Patino JA, Rizzuto GA, et al. Concomitant tumor immunity to a poorly immunogenic melanoma is prevented by regulatory T cells. J Exp Med, 2004,200, 771-782
    41 Audia S, Nicolas A, Cathelin D, et al. Increase of CD4~+CD25~+ regulatory T cells in the peripheral blood of patients with metastatic carcinoma: a Phase I clinical trial using cyclophosphamide and immunotherapy to eliminate CD4~+CD25~+T lymphocytes. Clin Exp Immunol, 2007,150, 523-530
    42 Passalacqua R, Buti S, Lazza R. Bevacizumab plus low-doses immunotherapy plus chemotherapy in metastatic renal cell cancer (mRCC): Antitumor effects and variations of T-regulatory cells (Treg) and other T lymphocytes subsets. A study of the Italian Oncology Group for Clinical Research (GOIRC). J Clin Oncol, 2008,26,163-172
    43 Galustian C, Meyer B, Labarthe MC, et al. The anti-cancer agents lenalidomide and pomalidomide inhibit the proliferation and function of T regulatory cells. Cancer Immunol, 2008,17,142-145
    44 Peng G, Guo Z, Kiniwa Y, et al. Toll-like receptor 8-mediated reversal of CD4~+ regulatory T cell function. Science, 2005,309, 1380-1384
    45 Weinberg AD, Rivera MM, Prell R, et al. Engagement of the OX-40 receptor in vivo enhances antitumor immunity. J Immunol, 2000,164,2160- 2169
    46 Piconese S, Valzasina B, Colombo MP. OX40 triggering blocks suppression by regulatory T cells and facilitates tumor rejection. J Exp Med, 2008, 20,5
    1 Burnet FM. The concept of immune surveillance. Prog Exp Tumor Res, 1970, 13,1-27
    2 Robertson MJ. Cytokines in Human Health. Humana Press Totowa, NJ, 2007, 12,297-317
    3 Lanier LL. NK cell recognition. Annu. Rev Immunol, 2005, 23,225-274
    4 Taylor P, RMartinez PL, Stacey M, et al. Macrophage receptors and immune recognition.Annu. Rev Immunol, 2005, 23, 901-944
    5 Janeway CA, Travers P, Walport M, et al. Immunobiology: the immune system in health and disease. 6th ed. Garland Science: New York, 2005
    6 Paul WE Ed. Fundamental Immunology. 5th ed. Williams and Wilkin: Philadelphia, 2003
    7 Matzinger P. Tolerance, danger, and the extended family. Annu.Rev. Immunol, 1994, 12, 991-1045
    8 Smyth MJ, Cretney E, Kershaw MH, et al. Cytokines in cancer immunity and immunotherapy. Immunol Rev, 2004, 202,275-293
    9 Dougan M, Dranoff G. Immune therapy for cancer. Annu Rev Immunol, 2009, 27, 83-117
    10 Nakanishi K, Yoshimoto T, Tsutsui H, et al. Interleukin-18 regulates both Th1 and Th2 responses. Annu Rev Immunol, 2001, 19, 423-474
    11 Sims JE. IL-1 and IL-18 receptors, and their extended family. Curr Opin Immunol, 2002,14,117-122
    12 Dinarello CA. Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol, 2009,27,519-550
    13 Kim SH, Eisenstein M, Reznikov L, et al. Structural requirements of sixnaturally occurring isoforms of the IL-18 binding protein to inhibit IL-18. Proc Natl Acad Sci, 2000,97,1190-1195
    14 Gracie JA, Robertson SE, McInnes IB. Interleukin-18. J Leukoc Biol, 2003, 73, 213-224
    15 Okamura H, Tsutsui H, Komatsu T, et al. Cloning of a new cytokine that induces IFN-γproduction by T cells.Nature, 1995,378,88-91
    16 Robinson D, Shibuya K, Mui A, et al. IGIF does not drive Th1 development but synergizes with IL-12 for interferon-γproduction and activates IRAK and NF-κB. Immunity, 1997, 7, 571-581
    17 Reddy P. Interleukin-18: recent advances. Curr Opin Hematol, 2004, 11, 405-410
    18 Micallef MJ, Tanimoto T, Kohno K, et al. Interleukin 18 induces the sequential activation of natural killer cells and cytotoxic T lymphocytes to protect syngeneic mice from transplantation with Meth A sarcoma. Cancer Res, 1997, 57, 4557-4563
    19 Micallef MJ, Tanimoto T, Yoshida KK, et al. In vivo antitumor effects of murine interferon-γ-inducing factor / interleukin-18 in mice bearing syngeneic Meth A sarcoma malignant ascites. Cancer Immunol Immunother, 1997,43, 361-367
    20 Osaki T, Peron JM, Cai Q, et al. IFN-γ-inducing factor/IL-18 administration mediates IFN-γ- and IL-12-independent antitumor effects. J Immunol, 1998, 160, 1742-1749
    21 Hashimoto W, Osaki T, Okamura H, et al. Differential antitumor effects of administration of recombinant IL-18 or recombinant IL-12 are mediated primarily by Fas-Fas ligand- and perforin-induced tumor apoptosis, respectively. J Immunol, 1999,163, 583-589
    22 Iwasaki T, Yamashita K, Tsujimura T, et al. Interleukin-18 inhibits osteolytic bone metastasis by human lung cancer cells possibly through suppression of osteoclastic bone-resorption in nude mice. J Immunother, 2002, 25 (Suppl )1, S52-60
    23 Yamashita K, Iwasaki T, Tsujimura T, et al. Interleukin-18 inhibitslodging and subsequent growth of human multiple myeloma cells in the bone marrow. Oncol Rep, 2002, 9, 1237-1244
    24 Okamoto T, Yamada N, Tsujimura T, et al. Inhibition by interleukin-18 of the growth of Dunn osteosarcoma cells. J Interferon Cytokine Res, 2004, 24,161-167
    25 Akamatsu S, Arai N, Hanaya T, et al. Antitumor activity of interleukin-18 against the murine T-cell leukemia/lymphoma EL-4 in syngeneic mice. J Immunother, 2002, 25(Suppl 1), S28-34
    26 Wigginton JM, Komschlies KL, Back TC, et al. Administration of interleukin 12 with pulse interleukin 2 and the rapid and complete eradication of murine renal carcinoma. J Natl Cancer Inst, 1996, 88, 38-43
    27 Son YI, Dallal RM, Lotze MT. Combined treatment with interleukin-18 and low-dose interleukin-2 induced regression of a murine sarcoma and memory response. J Immunother, 2003, 26,234-240
    28 Redlinger RE, Mailliard RB, Lotze MT, et al. Synergistic interleukin-18 and low-dose interleukin-2 promote regression of established murine neuroblastoma in vivo. J Pediatr Surg, 2003, 38, 301-307
    29 Carson WE, Dierksheide JE, Jabbour S, et al. Coadminstration of interleukin-18 and interleukin-12 induces a fatal inflammatory response in mice: critical role of natural killer cell interferon-γproduction and STAT -mediated signal transduction. Blood, 2000, 96, 1465-1473
    30 Rodriguez MC, Reynolds D, Correa SG, et al. Coexpression of IL-18 strongly attenuates IL-12-induced systemic toxicity through a rapid induction of IL-10 without affecting its antitumor capacity. J Immunol, 2009, 183, 740-748
    31 Oshikawa K, Shi F, Rakhmilevich AL, et al. Synergistic inhibition of tumor growth in a murine mammary carcinoma model by combinational gene therapy using IL-12, pro-IL-18, and IL-1βconverting enzyme cDNA. Proc Natl Acad Sci, 1999, 96, 13351-13356
    32 Marshall DJ, Rudnick KA, McCarthy SG, et al. Interleukin-18 enhances Th1 immunity and tumor protection of a DNA vaccine. Vaccine, 2006, 24,244-253
    33 Zheng JN, Pei DS, Mao LJ, et al. Potent antitumor efficacy of interleukin-18 delivered by conditionally replicative adenovirus vector in renal cell carcinoma-bearing nude mice via inhibition of angiogenesis. Cancer Biol Ther, 2009, 8, 599-606
    34 Tomohide T, Andrea G, Paul D, et al. Interleukin 18 gene transfer expands the repertoire of antitumor Th1-type immunity elicited by dendritic cell- based vaccines in association with enhanced therapeutic efficacy. Cancer Res, 2002,62,5853
    35 Obeid M, Tesniere A, Ghiringhelli GM, et al. Calreticulin exposure dict- ates the immunogenicity of cancer cell death. Nat Med, 2007, 13, 54-61
    36 Alagkiozidis I, Facciabene A, Carpenito C, et al. Increased immunegenicity of surviving tumor cells enables cooperation between liposomal doxorubicin and IL-18. J Translat Med, 2009, 7, 104-112
    37 Okamoto I, Kohno K, Tanimoto T, et al. IL-18 prevents the development of chronic graft-versus-host disease in mice. J Immunol, 2000, 164, 6067 -6074
    38 Reddy P, Teshima T, Hildebrandt G, et al. Interleukin 18 preserves perforindependent graft-versus-leukemia effect after allogeneic bone marrow transplantation. Blood, 2002, 100, 3429-3431
    39 Reddy P, Teshima T, Kukuruga M, et al. Interleukin-18 regulates acute graft-versus-host disease by enhancing Fas-mediated donor T cell apoptosis. J Exp Med, 2001, 194, 1433-1440
    40 Min CK, Maeda Y, Lowler K, et al. Paradoxical effects of interleukin-18 on the severity of acute graft-versus-host disease mediated by CD4~+ and CD8~+ T-cell subsets after experimental allogeneic bone marrow transplantation. Blood, 2004, 104, 3393-3399
    41 Reddy P, Teshima T, Hildebrandt G, et al. Pretreatment of donors with interleukin-18 attenuates acute graft-versus-host disease via STAT6 and preserves graft-versus-leukemia effects. Blood, 2003, 101, 2877-2885
    42 Gratwohl A, Baldomero H, Aljurf M, et al. Hematopoietic stem celltransplantation: a global perspective. JAMA, 2010, 202, 1617-1624
    43 Klingemann HG, Phillips GL. Immunotherapy after bone marrow transplantation. Bone Marrow Transplant, 1991, 8, 73-81
    44 Robertson MJ, Pelloso D, Abonour R,et al. Interleukin-12 immunotherapy after autologous stem cell transplantation for hematologic malignancies. Clin Cancer Res, 2002, 8, 3383-3393
    45 Pelloso D, Cyran K, Timmons L, et al. Immunological consequences of interleukin 12 administration after autologous stem cell transplantation. Clin Cancer Res, 2004,10, 1935-1942
    46 Robertson MJ, Chang HC, Pelloso D, et al. Impaired interferon-γproduction as a consequence of STAT4 deficiency after autologous hematopoietic stem cell transplantation for lymphoma. Blood, 2005, 106, 963-970
    47 Chang HC, Han L, Goswami R, et al. Impaired development of human Th1 cells in patients with deficient expression of STAT4. Blood, 2009, 113, 5887-5890
    48 Zhang S, Kaplan MH. The p38 mitogen-activated protein kinase is requi- red for IL-12-induced IFN-γexpression. J Immunol, 2000,165, 1375-1380
    49 Robertson MJ, Mier JW, Logan T, et al. Clinical and biological effects of recombinant human interleukin-18 administered by intravenous infusion to patients with advanced cancer. Clin Cancer Res, 2006, 12, 4265-4273
    50 Robertson MJ, Kirkwood JM, Logan TF, et al. A dose-escalation study of recombinant human interleukin-18 using two different schedules of administration in patients with cancer. Clin Cancer Res, 2008, 14, 3462-3469
    51 Koch KM, Roman JR, Jaworski D, et al. PK and PD of recombinant human IL-18 (rhIL-18) administered IV in repeated cycles to patients with solid tumors. J. Clin. Oncol, 2005, 23, 174-181
    52 Lewis N, Gollob J, Cohen R, et al. Phase I dose escalation study to assess tolerability and pharmcokinetics of recombinant human interleukin-18 (rh IL-18) administered as fourteen daily subcutaneous injections in patientswith solid tumors. J Clin Oncol, 2004, 22, 185-190
    53 Tarhini AA, Millward M, Mainwaring P, et al. A phase 2, randomized study of SB-485232, rhIL-18, in patients with previously untreated metastatic melanoma. Cancer, 2009, 115, 859-868
    54 Robertson MJ, Kline J, Bauman J, et al. A phase I trial evaluating the saf- ety and biological activity of iboctadekin (rhIL-18) in combination with rituximab in patients with CD20~+ Bcell non-Hodgkin's lymphoma. J Clin Oncol, 2009, 27, 450-454

© 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