用户名: 密码: 验证码:
阻断转化生长因子β信号通路的人肿瘤特异性淋巴毒细胞对个体化肾癌荷瘤免疫重建SCID鼠的免疫治疗作用研究
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:细胞毒性T淋巴细胞(CD8+CTL细胞)是一类具有CD8+表面标志、受MHC I类分子限制性杀伤功能的T淋巴细胞,其重要功能是可以特异性地杀伤靶细胞,在肿瘤免疫中,体内杀伤肿瘤的最终效应细胞CD8+T淋巴细胞,但其杀伤肿瘤的能力经常被肿瘤分泌的免疫抑制因子所抑制,如转化生长因子-β(TGF-β)。在本研究中,我们应用TGF-β不敏感的CD8+T淋巴细胞过继性输注至肾癌个体化荷瘤免疫重建重度免疫缺陷(SCID)鼠体内,来评价其对肾癌的治疗效果。我们应用肿瘤裂解物负载的人树突状(DC)细胞,与CD8+T淋巴细胞共培养以激发其杀伤活性,用包含显性负相II型TGF-β受体(TβRIIDN)基因的逆转录病毒感染CD8+T淋巴细胞,使其对TGF-β信号通路不敏感。检测转染后CD8+T淋巴细胞表型变化及体外杀伤作用,将这种TGF-β不敏感的CD8+T淋巴细胞过继性输注至免疫重建后的个体化肾癌荷瘤SCID鼠体内,评价其抗肿瘤免疫治疗作用及机制。
     方法:1)构建含有显性负相TGF-βII型受体(TβRIIDN)质粒的逆转录病毒:含有TβRIIDN质粒的逆转录病毒转染293包装细胞,32℃下共转染12小时,10% DMEM培养基37℃下孵化过夜,PBS漂洗后同样条件再次转染293细胞24小时,收集上清,获得含有TβRIIDN重组逆转录病毒。
     2)人肾癌原代细胞系的建立:无菌条件下将新鲜切取的肾癌组织剪成1.0cm3小块,包埋于裸鼠双侧腋下,4周后肿瘤组织生长至2-3cm3大小时,连续传代3次以上后取组织块培养,用胶原酶II及胰酶消化,得到上皮样肿瘤细胞,连续传代50次以上,得到稳定传代的肾癌原代细胞系。
     3)SCID-beige鼠免疫重建(Hu-PBMC-SCID):取患者外周血5-10ml,用淋巴细胞分离液分离外周血单核细胞(PBMC),用PBS重悬成0.3ml细胞悬液,腹腔注射至6-8周龄SCID-beige鼠体内,注射前SCID鼠接受剂量为3.5 Gy的钴60照射,4周后尾静脉取血,酶联免疫吸附法(ELISA)检测血清中人IgG含量。
     4)检测肾癌组织及肾癌原代细胞系中TGF-β1表达:用免疫组织化学及免疫荧光方法检测肾癌手术标本、裸鼠成瘤组织以及肾癌细胞系中TGF-β1表达。
     5)肿瘤特异性CD8+T淋巴细胞的分离培养与激活:取肾癌患者外周血,分离获得PBMC细胞,加入含rhIL-4(1000U/ml)、GM-CSF(1000U/ml)的完全培养基,隔日半量换液,在第6d梯度离心收获非粘附细胞为非成熟的DC细胞。反复冻融法获得肾癌细胞裂解物,裂解物反复刺激冲击非成熟的DC细胞,同时用免疫磁珠法分离外周血获取CD8+T淋巴细胞,将负载肾癌抗原的DC细胞与CD8+T淋巴细胞,同时加入rhIL-2(500U/ml)共培养,获得肿瘤特异性CD8+T淋巴细胞。
     6)肿瘤特异性CD8+T淋巴细胞转染:获取TGF-β不敏感CD8+T细胞(TβRIIDN CD8+T细胞):含有TβRIIDN和GFP基因重组逆转录病毒转染负载肾癌抗原的CD8+T细胞5%CO2、37℃下孵化48小时,获得表达TGF-β不敏感CD8+T细胞,流式细胞仪检测CD8+T淋巴细胞表型CD27,CD45RA表达。
     7)TβRIIDN- CD8+T淋巴细胞体外杀伤实验:将未经活化的CD8+T淋巴细胞(na?ve CD8+T淋巴细胞)以及TβRIIDN CD8+T淋巴细胞与相关肾癌细胞系共培养,51Cr释放实验测定其细胞杀伤作用,人前列腺癌细胞系PC-3作为对照靶细胞。
     8)TβRIIDN- CD8+T淋巴细胞体内抗肿瘤评价:建立肾癌细胞系皮下荷瘤免疫重建SCID-beige鼠模型,免疫重建SCID-beige鼠15只,随机分为3组,每组5只,皮下注射肾癌细胞5×106个,注射14天后肿瘤长至2-3mm时,分别腹腔注射TβRIIDN- CD8+T细胞(1×107),na?ve CD8+T细胞(1×107),以及PBS(0.5ml),一周后重复注射一次;同时建立肺转移Hu-PBMC-SCID鼠模型,Hu-PBMC-SCID 15只,随机分为3组,每组5只,分别尾静脉注射肾癌细胞5×106个,于注射后第7天分别腹腔注射TβRIIDN- CD8+T细胞(1×107),na?ve CD8+T细胞(1×107),以及PBS(0.5ml),一周后重复注射一次,观察至第40天各组荷瘤小鼠的肿瘤体积,重量的改变及生存情况,同时用TUNEL法检测肿瘤组织凋亡情况。
     9)小鼠体内细胞因子检测:尾静脉取小鼠血清行酶联免疫吸附(ELISA)测定细胞因子IFN-γ变化情况。10)肿瘤组织凋亡检测:取肿瘤组织,用原位末端标记法(TUNEL)检测肿瘤凋亡
     结果:
     1)建立了5例肾癌原代细胞系:共取68例肾癌手术标本接种裸鼠,5例肿瘤标本在裸鼠体内生长并可在体外稳定传代超过100次,手术接种成功率7.3 %。
     2)肾癌组织及细胞系中TGF-β1表达:TGF-β1在肾癌组织及细胞系的胞浆及细胞膜中呈强表达。
     3)Hu-PBMC-SCID鼠血清中人免疫球蛋白(IgG)的含量:110只用患者PBMC进行免疫重建的SCID-beige鼠中,75只可以检测到IgG表达,免疫重建成功率68.2%,IgG表达水平在0.8-2.2mg/ml之间,各组表达水平无明显统计学差异(P>0.05)。
     4)流式细胞仪分析:免疫磁珠分离CD8+T淋巴细胞纯度为98%。对GFP荧光分析提示TβRIIDN和GFP的逆转录病毒转染效率分别为92.13%和91.95%。对两种CD8+T淋巴细胞表型分子CD27,CD45RA的分析提示TβRIIDN CD8+T细胞优势表达CD27+CD45RA-(78.6±6.7%),为效应性T细胞表型,而na?ve CD8+T细胞优势表达CD27+CD45RA+(60.5±16.2%),为未活化T细胞表型。
     5)51Cr释放实验测定显示TβRIIDN CD8+T淋巴细胞对肾癌细胞杀伤活性较na?veCD8+T细胞强,在E:T为100:1时,其杀伤活性为75.5%,而na?veCD8+T细胞仅为15.8%。两种CD8+T淋巴细胞对人前列腺癌细胞PC-3无明显杀伤作用,结果表明阻断的TGF-β信号通路能增加CD8+T细胞肿瘤特异性杀伤活性。
     6)在成功构建的皮下荷瘤的SCID鼠模型分别接种两种CD8+T细胞,TβRIIDN CD8+T细胞治疗组肿瘤组织重量和体积均较na?veCD8+T细胞治疗组有明显差异(p<0.001),而在小鼠肺转移模型中,TβRIIDN CD8+T细胞治疗组其肺转移灶明显少于na?veCD8+T细胞治疗组,其生存期也明显延长(p<0.05)。TUNEL检测发现TβRIIDN CD8+T细胞治疗组多数肿瘤细胞发生凋亡,而na?veCD8+T细胞治疗组并未发现明显凋亡。这些结果说明TGF-β不敏感的CD8+T细胞可以发挥其特异性抗肿瘤活性,提高肾癌皮下荷瘤及肺转移小鼠的生存率,降低肺转移率。
     7)接种两种CD8+T细胞的荷瘤小鼠体内可以测出IFN-γ基础水平,接种TβRIIDN CD8+T细胞其荷瘤小鼠体内IFN-γ水平升高更为明显(P<0.05)。
     结论:
     1)成功构建了含有TβRIIDN质粒的逆转录病毒;
     2)成功建立了5例肾癌原代细胞系,发现肾癌中存在有TGF-β1高表达。
     3)成功用患者外周血单核细胞(PBMC)对SCID-beige鼠进行人免疫系统重建,并可检测到人IgG表达。
     4)用肾癌细胞裂解产物作为抗原负载DC,与CD8+T淋巴细胞共培养,诱导出了肾癌特异性的CD8+T淋巴细胞;
     5)使用修饰后TβRII基因转染肾癌特异性的CD8+T淋巴细胞,使TβRII显性负相表达,阻断TGF-β信号通路;
     6)TβRIIDN CD8+T淋巴细胞呈效应性T细胞表型;
     7)TGF-β不敏感的CD8+T淋巴细胞能明显抑制肾癌皮下荷瘤小鼠肿瘤生长,并提高肺转移小鼠的生存率,诱导肿瘤凋亡。
     8)TGF-β不敏感的CD8+T淋巴细胞具有肿瘤特异性杀伤靶细胞作用。
     9) TGF-β不敏感的CD8+T淋巴细胞输注的荷瘤小鼠体内IFN-γ水平明显升高。
Objective
     CD8+ T lymphocyte cells are the T cells which have the CD8+ phenotype,MHC-I restricted cytotoxic function. In tumor immunotherapy,the functions of CD8+ T lymphocyte cells are often suppressed by the transforming growth factor-β(TGF-β) . TGF-βis a potent immunosuppressant. Adoptive transfer murine-derived, tumor-reactive, TGF-β-insensitive CD8+ T cells into tumor challenged mice has shown potent antitumor responses. The present study was conducted to a one-to-one adoptive transfer strategy to treat tumor-bearing severe combined immunodeficient (SCID/beige) mouse. The SCID/beige mice were humanized with PBMC from renal cell carcinoma (RCC) patients (Hu-PBMC-SCID) before adoptive transfer. Autologous CD8+ T cells were expanded ex vivo with using autologous patient’s dendritic cells (DCs) pulsed with the tumor-lysate and rendered TGF-β-insensitive by dominant-negative TGF-βtype II receptor(TβRIIDN). Flow cytometry analysis showed the TGF-β-insensitive CD8+ T cells were the effector CD8+ cells (CD27-CDRA+). Then, adoptive transfer autologous TGF-β-insensitive CD8+ T cell into tumor-bearing Hu-PBMC-SCID mice can induce powerful tumor-specific cytotoxic T lymphocyte (CTL) responses, induced tumor apoptosis, suppressed lung metastasis and prolonged survival times. This one-to-one adoptive transfer strategy provides a scientific rationale for expected clinical investigation in the treatment of renal cell carcinoma.
     Materials and methods
     1. Production of infectious TβRIIDN-GFP retrovirus
     TβRIIDN was excised from pcDNA3-TβRIIDN by BamHI/EcoRI digestion and inserted into the pMig-inteRIIDNl ribosomal entry sequence-green fluorescence protein (herein designated MSCV-GFP) vector by first linearizing pMig with EcoRI and ligating an EcoRI/BamHI adapter (5′-AATTGGATCCGCGGCCGCG-3′, 3′-CCTAGGCGCCGGCGCTTAA-5′). These clones were designated as MSCV-TβRIIDN and were screened by sequencing for correct orientation and insert numbers.Pantropic GP293 retroviral packaging cells (Clontech, San Diego, CA) were seeded at a density of 2.5×106 per T-25 collagen I-coated flask (Biocoat; BD Biosciences, Mountain View, CA) and incubated for 24 h before plasmid transfection in antibiotic-free 10% DMEM (Gibco, Grand Island, NY). A mixture of 2μg retroviral plasmid and 2μg vesicular stomatitis virus envelope G protein (VSV-G) envelope plasmid was cotransfected in serum-free DMEM by using Lipofectamine-Plus (Invitrogen, Carlsbad, CA) according to the manufacturer’s protocols. Briefly, the cells were transfected for 12 h followed by the addition of an equivalent volume of 10% DMEM and incubation for an additional 12 h. The supeRIIDNtant was then aspirated, the cells were rinsed gently in PBS, and 3 ml fresh 10% DMEM was added to each flask. After 24 h, the virus-containing supeRIIDNtant was collected and used to infect target cells.
     2. Establish five renal cell carcinoma cell lines
     Specimens were obtained from 68 patients who were diagnosed with RCC and underwent radical nephrectomy between September 2005 and December 2007.The study protocol was approved by the Ethics Committee of the Xijing Hospital, Fourth Military Medical University. Informed consent was obtained from all participants.Total of 68 fresh RCC tissues were obtained after operation and implanted subcutaneously to nude mice as described previously .Five kinds of RCC specimens remained engrafted successfully 1 month after transplantation. Histological section from each initial tissue and xenograft specimen was subjected to hematoxylin and eosin (H&E) analysis. Then, five kinds of human RCC cell lines were obtained from the xenograft transplanted in the nude mice. These RCC cell lines was maintained in a complete medium (CM) containing RPMI-1640 medium (HyClone, Logan, UT) supplemented with 10% heat-inactivated fetal bovine serum (FBS; GIBCO, Gaithersburg, MD), 2 mmol/L L-glutamine, 50μmmol/L 2-mercaptoethanol, 100 units/ml penicillin, and 100μg/ml streptomycin (Sigma, St Louis, MO).
     3. Establish the Hu-PBMC-SCID mice model
     Male or female SCID/beige mice 6-8 weeks old were obtained from the Laboratory Animal Research Center of the Fourth Military Medical University and housed in sterile filter-top caged placed in a laminar backflow-cabinet under specific pathogen-free conditions. The SCID-beige mice were divided medially with five groups received PBMCs injection from five patients. One day before PBMCs injection, mice were sublethally irradiated with 3.5 Gy ([60Co] source Gammatron F 80S, Simens, Germany). Autologous PBMCs purified from each patient’s blood using a Ficoll-HyPaque (Pharmacia, New Jersey, USA) gradient after platelet depletion and washing, each mouse received 0.3 ml of the PBMCs (2×107cells) suspended in PBS via intra-peritoneal injection .
     4. Immunohistochemical and immunofluorescence analysis for TGF-β1 expression in RCC tumor tissues and cell lines
     Five kinds of patient’s RCC original and xenograft specimens were performed by immunohistochemical analysis for TGF-β1 expression. Briefly, Paraffin-embedded sections (4μm) were deparaffinized and rehydrated. After quenched endogenous peroxidase and blocking step performed, primary anti-TGF-β1 mAb and goat-anti-mouse second antibody (anti-TGF-β1 mAb, 1:100; second antibody 1:500; Abcam Biotechnology, Cambridge, UK) were incubated. Peroxidase substrate solution 3, 3’-diaminobenzidine was used for direct staining. Counter-staining was done with 10 % hematoxylin. Nonimmune murine antibody was used for negative control sections. For immunofluorescence analysis, cells were incubated with TGF-β1 mAb for 2h, stained with FITC-conjugated anti-mouse IgG (1:1000, Abcam Biotechnology) for 1h. The nuclei were stained with 100 ng/ml DAPI (4',6-diamidino-2-phenylindole) and were examined by fluorescence microscopy (Nikon Corp., Tokyo, Japan).
     5. Generation of patient autologous tumor reactive TGF-β-insensitive CD8+ T cells
     With the use of CD8+ Microbeads (Miltenyi Biotec, Bergisch Gladbach, Germany), patient’s CD8+ T cells were positively selected from whole blood with a purity of more than 98 %. CD8+ T cells was expanded with using autologous patient’s DCs pulsed with the tumor-lysate in the presence of recombinant human interleukin-2( IL-2;500 unit/ml, PeproTech, London, England).There were two types of CD8+ T cells:1) tumor-reactive TGF-β-insensitive CD8+ T cells which rendered insensitive to TGF-βby infection with TβRIIDN-green fluorescent protein (GFP)–containing retrovirus. 2) naive CD8+ T cells isolated from PBMC without any treatment.
     6. Flow cytometric analysis TGF-β-insensitive CD8+ T cell characterization
     Immunophenotypical characterization of TGF-β-insensitive CD8+ T cell and naive CD8+ T cell were performed using fluorescein isothiocyanate (FITC)-conjugated anti-CD27mAb, anti-CD45RA mAb before administration to the mice. Cells stained with appropriate mAbs in PBS, 0.2% BSA, 50μM EDTA for 20 min at 4 0C and either directly analyzed or sorted into defined populations on a FACSVantae SE, using CellQuest software (BD Bioscience).
     7. 51 Chromium release assays
     The two types of CD8+ T cells were subjected to a standard 51Chromium- release assay. RCC cell lines and another irrelevant cell line, human prostate carcinoma cell line, PC-3 cells were used as targets. Briefly, target cells were labeled with 100μCi 51Cr/105 cells. Different groups of CD8+ T cells were added to U-bottom plates containing 5,000 cells /well with various effectors to target (E/T) ratios ranging from 1:1 to 100:1. Equal volumes of RPMI-1640 and 1 mol/L HCl were added to other wells as the negative and positive controls, respectively. After a 4-hour incubation, 100μl of supernatants was harvested from each well and the 51Cr released was measured using a gamma-counter. The percent cell lysis was calculated according to the formula: percent specific 51Cr-Release= (Experimental Release– Spontaneous Release)×100/ (Maximum Release– Spontaneous Release)].
     8. Adoptive transfer TGF-β-insensitive CD8+ T cell in tumor bearing Hu- PBMC-SCID mice
     The Hu-PBMC-SCID mice received an injection in the right flank of 5×106 RCC patient’s autologous tumor cell line (Day 0).Tumors developed approximately 2-3 mm in diameters 14 days later. At day 14, adoptive transfer with patient’s autologous TGF-β-insensitive CD8+ T cells were done in the tumor-bearing Hu-PBMC-SCID mice. Three groups (5 mice per group) received intraperitoneal injection with different types of adoptive transfer composed of TGF-β-insensitive CD8+ T cells (1×107), naive CD8+ T cells (1×107), PBS (0.5ml), respectively. The vaccination was repeated on day 21.Tumor growth and animal survival was monitored daily after vaccine.The mice pulmonary metastasis model was also prepared by a single injection of 5×106 RCC in the tail vein. On day 7,the tumor-bearing mice (n=5/group)were inoculated with two types of CD8+ T cells vaccines (1×107 cells) and PBS via intraperitoneal injection, respectively. Forty days after adoptive transfer, all mice were sacrificed and the tumors were isolated for evaluation of the volume (volume=length×width2×π/6), weight and histological analysis. Other tissues such as spleen, pulmonary were also harvested.
     9. ELISA assay for INF-γ
     The sera of the 3 above mentioned mouse groups were harvested. The serum levels of INF-γwere determined using an ELISA kit (R&D Systems, Minneapolis, MN) according to the protocol. Serum was stored at -70℃until the assay.
     10. TUNEL staining for tumor apoptosis
     Paraffin-embedded tumor sections were used for apoptosis assay. The nuclear and terminaldeoxynucleotidyl transferase mediated dUTP nick end labeling(TUNEL) apoptosis assay kit(R﹠D system, Minneapolis, MN) were done as described previously
     11. Statistical analysis
     Numerical data were expressed as mean±standard deviation (SD). ANOVA and chi-square tests were performed to determine the differences in the means among the various treatment groups. P < 0.05 was considered statistically significant. The SPSS 12.0 software package (SPSS Inc., Chicago, IL) was used for analysis. The Kaplan-Meier survival curve was analyzed by the log-rank test with the Graphpad Prism 4.02 software (Graphpad Software Inc., San Diego, CA).
     Results
     1. Establishment and characterization of RCC cell lines Five kinds of human RCC cell lines were established successfully from specimens inoculated nude mice. All RCC cell lines stably cultured more than 100 passages after cryopreserved and thawed. The histological analysis of xenograft in mice was similar to the histological evaluation of the original patient tissue specimen
     2. TGF-β1 expression in RCC xenograft in mice and cell lines
     A representative result of the immunohistochemistry for TGF-β1 in xenograft in mice is shown in Fig.2A-B.In these cancerous tissues, TGF-β1 was found strong expression either in the cytoplasm or on the plasma membrane of the neoplastic cell. This result was also confirmed by immunofluorescent staining in RCC cell lines.
     3. Appearance of human immunoglobulins in Hu-PBMC-SCID mice
     Four weeks after PBMCs injection, human immunoglobulins could be detected in 75 of 110 (68.2%) Hu-PBMC-SCID mice sera. The IgG levels of each group averaged between 0.8-2.2 mg/ml which in agreement with results of previous studies.There were no significant differences in the success rate of PBMC engraftment and the levels of IgG were similar between each group of mice (P>0.05). Furthermore, no severe xenogenic graft versus host disease (GVHD) was observed.
     4. Phenotypic analysis of TGF-β-insensitive and naive CD8+ T cells
     Flow cytometry analysis showed that the expression of co-stimulatory molecules CD27 and CD45RA in two types of CD8+ T cells were different. In the TGF-β-insensitive CD8+ T cells, the dominant phenotype was the CD27+CD45RA-(78.6±6.7%),the effector CD8+ T cell phenotype. But in naive CD8+ T cells, most of the cells showed the CD27+CD45RA+ phenotype(60.5±16.2%), the unprimed CD8+ T cell phenotype.
     5. TGF-β-insensitive CD8+ T cells showed superior anti-tumor responses in vitro
     The specific tumor-killing ability of the autologous TGF-β-insensitive CD8+ T cells was shown by the in vitro CTL assay. We found that the TGF-β-insensitive CD8+ T cells displayed 5-fold greater tumor-killing activity than the na?ve CD8+ T cells((75.5% vs 15.8% at an E:T cell ratio of 100:1,).When incubated with an irrelevant cell line, PC-3 cell, no apparent lytic activity was observed.
     6. TGF-β-insensitive CD8+ T cells showed superior anti-tumor responses in vivo
     In the group treated with TGF-β-insensitive CD8+ T cells ,the average tumor volumes and tumor weights was significantly decreased than the group treated na?ve CD8+ T cells and PBS group (P<0.05). Interestingly, in the mice with pulmonary metastasis, all the animals died in the PBS treated group before day 30, there were 4 of 5 mice died in the na?ve CD8+ T treated group before day 32 of the experiment due to poor health conditions, while all the mice survived in the TGF-β-insensitive CD8+ T cell treated group at the end of the experiment. According to the long-rank test, there were significant differences among three groups (p<0.05).
     7. TGF-β-insensitive CD8+ T cells induced high serum levels of INF-γ
     The serum levels of INF-γexhibited increases in the TGF-β-insensitive CD8+ T cells treated group, but in the na?ve CD8+ T cell and PBS treated group, the expression levels of INF-γwere negligible. The differences of INF-γserum levels between TGF-β-insensitive CD8+ T cells, na?ve CD8+ T cell group and PBS group were significant (Fig.7. p<0.01). The greater increase in levels of serum INF-γobserved in the TGF-β-insensitive CD8+ T cell treated group indicated that immune cells were most strongly activated in these hosts.
     8. TGF-β-insensitive CD8+ T cell induced tumor cell apoptosis
     The TUNEL assay confirmed that autologous TGF-β-insensitive CD8+ T cell could induce tumor cell apoptosis in the TβRIIDN group. But in the other two groups, no apparent apoptotic cells were observed in tumor tissues.
     Conclusion
     1. We successfully constructed a retrovirus containing dominant-negative TGF-βtype II receptor (TβRIIDN).
     2. We successfully established five RCC cell lines and detected the high expression of TGF-β1 in RCC.
     3. Established the Hu-PBMC-SCID mouse models successfully,and found human immunoglobulins expression in these mice.
     4. Incubated with tumor lysate loaded-DCs pulsed with CD8+T cells can induce and activate RCC-specific CD8+T cells.
     5. RCC-specific CD8+T cells were rendered TGF-βinsensitive by infecting with a retrovirus containing dominant-negative TGF-βtype II receptor (TβRIIDN), leading to the blockade of TGF-βsignals to members of the Smad protein family.
     6. The TGF-β-insensitive RCC-specific CD8+T cells were the effector CD8+T phenotype .
     7. TGF-βinsensitive TP- CD8+T cells suppressed tumor growth ,induced tumor apoptosis,and increased survival rate of pulmonary metastases mice.
     8. The most potent CTL response was induced by the TGF-?-insensitive CD8+T cells in vitro (75.4% killing activity at an effector:target cell ratio of 100:1). No apparent lysis was observed against irrelevant PC-3 cells.
     9. TGF-βinsensitive TP- CD8+T cells induced higher IFN-γlevel in vivo.
引文
[1] Blobe GC, Schiemann WP, Lodish HF. Role of transforming growth factor beta in human disease. N Engl J Med ,2000,342:1350-1358
    [2] Wrana JL, Attisano L, Wieser R, et al:Mechanism of activation of the TGF-beta receptor.Nature ,1994,370:341-347
    [3] Inman GJ, Nicolas FJ, Hill CS: Nucleocytoplasmic shuttling of Smads 2, 3, and 4 permitssensing of TGF-beta receptor activity. Mol Cell, 2002,10:283-294,
    [4] Fink SP, Swinler SE, Lutterbaugh JD, et al: Transforming growth factor-beta-induced growth inhibition in a Smad4 mutant colon adenoma cell line. Cancer Res, 2001,61:256-260
    [5] Frolik CA, Dart LL, Meyers CA, et al: Purification and initial characterization of a type beta transforming growth factor from human placenta. Proc Natl Acad Sci U S A ,1983,80:3676- 3680
    [6] Hannon GJ, Beach D: P15INK4B is a potential effector of TGF-beta-induced cell cycle arrest. Nature 1994,371:257-261
    [7] Pietenpol JA, Stein RW, Moran E, et al: TGF-beta 1 inhibition of c-myc transcription and growth in keratinocytes is abrogated by viral transforming proteins with pRB binding domains.Cell ,1990,61:777-785
    [8] Petritsch C, Beug H, Balmain A, et al: TGF-beta inhibits p70 S6 kinase via protein phosphatase2A to induce G(1) arrest. Genes Dev, 2000,14:3093-3101
    [9] Fink SP, Swinler SE, Lutterbaugh JD, et al: Transforming growth factor-beta-induced growth inhibition in a Smad4 mutant colon adenoma cell line. Cancer Res ,2001,61:256-260
    [10] Nicolas FJ, Hill CS: Attenuation of the TGF-beta-Smad signaling pathwayin pancreatic tumor cells confers resistance to TGF-betainduced growth arrest. Oncogene, 2003,22:3698-3711
    [11] Yakymovych I, Ten Dijke P, Heldin CH, et al: Regulation of Smad signaling by protein kinase C. Faseb J, 2001,15:553-555
    [12] Jonson T, Albrechtsson E, Axelson J, et al: Altered expression of TGFB receptors and mitogenic effects of TGFB in pancreatic carcinomas.Int J Oncol ,2001,19:71-81
    [13] Park BJ, Park JI, Byun DS, et al: Mitogenic conversion of transforming growth factorbeta1 effect by oncogenic Ha-Ras-induced activation of the mitogen-activated protein kinase signaling pathway in human prostate cancer. Cancer Res,2000, 60:3031-3038
    [14] Seifert RA, Coats SA, Raines EW, et al: Platelet-derived growth factor (PDGF) receptor alpha-subunit mutant and reconstituted cell lines demonstrate that transforming growth factorbeta can be mitogenic through PDGF A-chaindependent and -independent pathways. J Biol Chem ,1994,269:13951-13955
    [15] Piek E, Heldin CH, Ten Dijke P: Specificity, diversity, and regulation in TGF-beta superfamily signaling. FASEB J ,1999,13:2105-2124
    [16] Schiemann WP, Blobe GC, Kalume DE, et al: Context-specific effects of fibulin-5 (DANCE/EVEC) on cell proliferation, motility, and invasion. Fibulin-5 is induced by transforming growth factor-beta and affects protein kinase cascades. J Biol Chem ,2002,277:27367-27377
    [17] Blobe GC, Schiemann WP, Lodish HF: Role of transforming growth factor beta in human disease. N Engl J Med ,2000,342:1350-1358
    [18] Hahn WC: Role of telomeres and telomerase in the pathogenesis of human cancer. J Clin Oncol ,2003,21:2034-2043
    [19] Lin SY, Elledge SJ: Multiple tumor suppressor pathways negatively regulate telomerase. Cell ,2003, 113:881-889
    [20] Comijn J, Berx G, Vermassen P, et al: The two-handed E box binding zinc finger protein SIP1 downregulates E-cadherin and induces invasion. Mol Cell, 2001,7:1267-1278
    [21] Stampfer MR, Garbe J, Levine G, et al: Expression of the telomerase catalytic subunit, hTERT, induces resistance to transforming growth factor beta growth inhibition in p16INK4A(-) human mammary epithelial cells. Proc Natl Acad Sci U S A, 2001,98:4498-4503
    [22] Yamamura Y, Hua X, Bergelson S, et al: Critical role of smads and AP-1 complex in TGFbeta- dependent apoptosis. J Biol Chem,2000,275: 36295-36302,
    [23] Lallemand F, Mazars A, Prunier C, et al: Smad7 inhibits the survival nuclear factor kappaB and potentiates apoptosis in epithelial cells.Oncogene ,2001,20:879-884
    [24] Inman GJ, Allday MJ: Apoptosis induced by TGF-beta 1 in Burkitt’s lymphoma cells is caspase 8 dependent but is death receptor independent. J Immunol 2000,165:2500-2510
    [25] Hagimoto N, Kuwano K, Inoshima I, et al: TGF-beta 1 as an enhancer of Fas-mediated apoptosis of lung epithelial cells. J Immunol,2002,168: 6470-6478
    [26] Remy I, Montmarquette A, Michnick SW: PKB/Akt modulates TGF-beta signalling through a direct interaction with Smad3. Nat Cell Biol, 2004,6:358-365
    [27] Pepper MS: Transforming growth factorbeta: Vasculogenesis, angiogenesis, and vessel wall integrity. Cytokine Growth Factor Rev,1997, 8:21- 43
    [28] Larsson J, Goumans MJ, Sjostrand LJ, et al: Abnormal angiogenesis but intact hematopoietic potential in TGF-beta type I receptordeficient mice. Embo J ,2001,20:1663-1673
    [29] Johnson DW, Berg JN, Baldwin MA, et al: Mutations in the activin receptor-like kinase 1gene in hereditary haemorrhagic telangiectasia type 2. Nat Genet ,2000,13:189-195
    [30] Burrows FJ, Derbyshire EJ, Tazzari PL, et al: Up-regulation of endoglin on vascular endothelial cells in human solid tumors: Implications for diagnosis and therapy. Clin Cancer Res,1995,1:1623-1634
    [31] Yamamoto T, Kozawa O, Tanabe K, et al: Involvement of p38 MAP kinase in TGF-betastimulated VEGF synthesis in aortic smooth muscle cells. J Cell Biochem 2001,82:591-598
    [32] Lamouille S, Mallet C, Feige JJ, et al: Activin receptor-like kinase 1 is implicated in the maturation phase of angiogenesis. Blood 2002,100: 4495-4501
    [33] Goumans MJ, Valdimarsdottir G, Itoh S, et al: Activin receptor-like kinase (ALK)1 is an antagonistic mediator of lateral TGFbeta/ALK5 signaling. Mol Cell 2003,12:817-828
    [34] Kirkbride KC, Blobe GC: Inhibiting the TGF-beta signalling pathway as a means of cancer immunotherapy. Expert Opin Biol Ther ,2003,3:251- 261
    [35] Leach DR, Krummel MF, Allison JP: Enhancement of antitumor immunity by CTLA-4 blockade. Science, 1996,271:1734-1736
    [36] Brandes ME, Wakefield LM, Wahl SM: Modulation of monocyte type I transforming growth factor-beta receptors by inflammatory stimuli. J Biol Chem, 1991,266:19697-19703
    [37] Kehrl JH, Wakefield LM, Roberts AB, et al: Production of transforming growth factor beta by human T lymphocytes and its potential role in the regulation of T cell growth. J Exp Med, 1986,163:1037-1050
    [38] Gorelik L, Flavell RA: Transforming growth factor-beta in T-cell biology. Nat Rev Immunol ,2002,2:46-53
    [39] Nakamura K, Kitani A, Strober W: Cell contact-dependent immunosup-pression by CD4(+)CD25(+) regulatory T cells is mediated by cell surface-bound transforming growth factor beta. J Exp Med,2001,194: 629-644
    [40] Shull MM, Ormsby I, Kier AB, et al: Targeted disruption of the mouse transforming growth factor-beta 1 gene results in multifocal inflammatory disease. Nature,1992, 359:693-699
    [41] Gorelik L, Flavell RA: Abrogation of TGFbeta signaling in T cells leads to spontaneous T cell differentiation and autoimmune disease. Immunity, 2000 ,12:171-181
    [42] Bogdan C, Paik J, Vodovotz Y, et al: Contrasting mechanisms for suppression of macrophage cytokine release by transforming growth factor-beta and interleukin-10. J Biol Chem,1992, 267:23301-23308
    [43] Riedl E, Strobl H, Majdic O, et al: TGFbeta 1 promotes in vitro generation of dendritic cells by protecting progenitor cells from apoptosis. J Immunol ,1997,158:1591-1597
    [44] Borkowski TA, Letterio JJ, Mackall CL, et al: Langerhans cells in the TGF beta 1 null mouse. Adv Exp Med Biol ,1997,417:307-310
    [45] Kinzler KW, Vogelstein B: Cancersusceptibility genes: Gatekeepers and caretakers.Nature ,1997,386:761-763
    [46] Glick A, Popescu N, Alexander V, et al: Defects in transforming growth factor-beta signaling cooperate with a Ras oncogene to cause rapid aneuploidy and malignant transformation of mouse keratinocytes. Proc Natl Acad Sci U S A ,1999,96:14949-14954
    [47] Takaku K, Oshima M, Miyoshi H, et al: Intestinal tumorigenesis in compound mutant mice of both Dpc4 (Smad4) and Apc genes. Cell ,1998,92:645-656
    [48] Furuhashi M, Yagi K, Yamamoto H, et al: Axin facilitates Smad3 activation in the transforming growth factor beta signaling pathway. MolCell Biol ,2001,21:5132-5141
    [49] Nishita M, Hashimoto MK, Ogata S, et al: Interaction between Wnt and TGF-beta signalling pathways during formation of Spemann’s organizer. Nature ,2000,403:781-785
    [50] Murray NR, Weems C, Chen L, et al: Protein kinase C betaII and TGFbetaRII in omega-3 fatty acid-mediated inhibition of colon carcinogenesis. J Cell Biol, 2002,157:915-920
    [51] Siegel PM, Shu W, Cardiff RD, et al: Transforming growth factor beta signaling impairs Neu-induced mammary tumorigenesis while promoting pulmonary metastasis. Proc Natl Acad Sci U S A, 2003, 100:8430-8435
    [52] McEarchern JA, Kobie JJ, Mack V, et al: Invasion and metastasis of a mammary tumor involves TGF-beta signaling. Int J Cancer , 2001,91:76-82
    [53] Thiery JP, Chopin D: Epithelial cell plasticity in development and tumor progression.Cancer Metastasis Rev ,1999,18:31-42
    [54] Piek E, Moustakas A, Kurisaki A, et al: TGF-(beta) type I receptor/ALK-5 and Smad proteins mediate epithelial to mesenchymal transdifferentiation in NMuMG breast epithelial cells.J Cell Sci ,1999112:4557-4568
    [55] Reeves R, Edberg DD, Li Y: Architectural transcription factor HMGI(Y) promotes tumor progression and mesenchymal transition of human epithelial cells. Mol Cell Biol 2001,21:575-594
    [56] Eickelberg O, Centrella M, Reiss M, et al: Betaglycan inhibits TGF-beta signaling by preventing type I-type II receptor complex formation: Glycosaminoglycan modifications alter betaglycan function. J Biol Chem ,2002,277:823-829
    [57] Copland JA, Luxon BA, Ajani L, et al: Genomic profiling identifies alterations in TGFbeta signaling through loss of TGFbeta receptor expression in human renal cell carcinogenesis and progression.Oncogene,2003,22:6109-6118
    [58] Tsushima H, Ito N, Tamura S, et al: Circulating transforming growth factor beta 1 as a predictor of liver metastasis after resection in colorectal cancer. Clin Cancer Res ,2001,7:1258-1262
    [59] Shariat SF, Kim JH, Andrews B, et al: Preoperative plasma levels of transforming growth factor beta(1) strongly predict clinical outcome in patients with bladder carcinoma. Cancer ,2001,92:2985-2992
    [60] Shariat SF, Shalev M, Menesses-Diaz A, et al: Preoperative plasma levels of transforming growth factor beta(1) (TGF-beta(1)) strongly predict progression in patients undergoing radical prostatectomy. J Clin Oncol ,2001,19:2856-2864
    [61] Anscher MS, Peters WP, Reisenbichler H, et al: Transforming growth factor beta as a predictor of liver and lung fibrosis after autologous bone marrow transplantation for advanced breast cancer. N Engl J Med ,1993,328:1592-1598
    [62] Li C, Gardy R, Seon BK, et al: Both high intratumoral microvessel density determined using CD105 antibody and elevated plasma levels of CD105 in colorectal cancer patients correlate with poor prognosis. Br J Cancer ,2003,88:1424-1431
    [63] Dales JP, Garcia S, Bonnier P, et al: CD105 expression is a marker of high metastatic risk and poor outcome in breast carcinomas:Correlations between immunohistochemical analysis and long-term follow-up in a series of 929 patients. Am J Clin Pathol ,2003,119:374-380
    [64] Park SH, Lee SR, Kim BC, et al: Transcriptional regulation of the transforming growth factor beta type II receptor gene by histone acetyltransferase and deacetylase is mediated by NF-Y in human breast cancer cells. J Biol Chem, 2002,277:5168-5174
    [65] Venkatasubbarao K, Ammanamanchi S, Brattain MG, et al: Reversion oftranscriptional repression of Sp1 by 5 aza-2’deoxycytidine restores TGF-beta type II receptor expression in the pancreatic cancer cell line MIA PaCa-2. Cancer Res ,2001,61:6239-6247
    [66] Stander M, Naumann U, Dumitrescu L, et al: Decorin gene transfer-mediated suppression of TGF-beta synthesis abrogates experimental malignant glioma growth in vivo. Gene Ther,1998, 5:1187-1194
    [67] Rowland-Goldsmith MA, Maruyama H, Matsuda K, et al: Soluble type II transforming growth factor-beta receptor attenuates expression of metastasis-associated genes and suppresses pancreatic cancer cell metastasis. Mol Cancer Ther ,2002,1:161-167
    [68] Lei X, Bandyopadhyay A, Le T, et al: Autocrine TGFbeta supports growth and survival of human breast cancer MDA-MB-231 cells. Oncogene 2002,21:7514-7523
    [69] Andres JL, Stanley K, Cheifetz S, et al: Membrane-anchored and soluble forms of betaglycan, a polymorphic proteoglycan that binds transforming growth factor-beta. J Cell Biol 1989,109: 3137-3145
    [70] Inman GJ, Nicolas FJ, Callahan JF, et al: SB-431542 is a potent and specific inhibitor of transforming growth factor-beta superfamily type I activin receptor-like kinase (ALK) receptors ALK4, ALK5, and ALK7. Mol Pharmacol, 2002,62:65- 74
    [71] Sawyer JS, Anderson BD, Beight DW, et al: Synthesis and activity of new aryl- and heteroaryl-substituted pyrazole inhibitors of the transforming growth factor-beta type I receptor kinase domain. J Med Chem ,2003,46:3953-3956
    [72] Yang YA, Dukhanina O, Tang B, et al:Lifetime exposure to a soluble TGF-beta antagonist protects mice against metastasis without adverse side effects. J Clin Invest ,2002,109:1607- 1615
    [73] Muraoka RS, Dumont N, Ritter CA, et al: Blockade of TGF-beta inhibits mammary tumor cell viability, migration, and metastases. J Clin Invest, 2002,109:1551-1559
    [74] Matsuno F, Haruta Y, Kondo M, et al: Induction of lasting complete regression of preformed distinct solid tumors by targeting the tumor vasculature using two new anti-endoglin monoclonal antibodies. Clin Cancer Res,1999 ,5:371- 382
    [75] Matthews E, Yang T, Janulis L, et al:Down-regulation of TGF-beta1 production restores immunogenicity in prostate cancer cells.Br J Cancer 2000,83:519-525
    [76] Gorelik L, Flavell RA: Immune-mediated eradication of tumors through the blockade of transforming growth factor-beta signaling in T cells. Nat Med ,2001,7:1118-1122
    [77] Mosier, D.E., Gulizia, R.J., Baird, S.M., et al.Transfer of a functional human immune system to mice with severe combined immunodeficiency.Nature ,1988,335: 256–259.
    [78] Berney, T., Molano, R.D., Pileggi, A., et al, Patterns of engraftment in different strains of immunodeficient mice reconstituted with human peripheral blood lymphocytes. Transplantation ,2001.72: 133–140.
    [79] Flanagan, S.P.‘Nude’, a new hairless gene with pleiotropic effects in the mouse. Genet. Res, 1966, 8:295–309
    [80] Rygaard, J., Povlsen, C.O., Heterotransplantation of a human malignant tumour to“Nude”mice. Acta Pathol. Microbiol. Scand, 1969, 77: 758–760.
    [81] Scher, I., Ahmed, A., Strong, D.M., et al. X-linked B-lymphocyte immune defect in CBA/HN mice. I. Studies of the function and composition of spleen cells. J. Exp. Med. , 1975, 141: 788–803.
    [82] Wortis, H.H., Burkly, L., Hughes, D.,et al. Lack of mature B cells in nudemice with X-linked immune deficiency. J. Exp. Med,1982,155:903–913.
    [83] Bosma, G.C., Custer, R.P., Bosma, M.J., A severe combined immunodeficiency mutation in the mouse. Nature ,1983,301:527–530.
    [84] Fulop, G.M., Phillips, R.A., The scid mutation in mice causes a general defect in DNA repair. Nature 1990,347:479–482.
    [85] Roder, J., Duwe, A.,. The beige mutation in the mouse selectively impairs natural killer cell function. Nature,1979 ,278: 451–453.
    [86] Mosier, D.E., Stell, K.L., Gulizia, R.J.,et al., Homozygous scid/scid;beige/beige mice have low levels of spontaneous or neonatal T cell-induced B cell generation. J. Exp. Med,1993,177:191–194.
    [87] Makino, S., Kunimoto, K., Muraoka, Y.et al,. Breeding of a non-obese, diabetic strain of mice. Jikken Dobutsu,1980 ,29: 1–13.
    [88] Prochazka, M., Gaskins, H.R., Shultz, L.D,et al. The nonobese diabetic scid mouse:model for spontaneous thymomagenesis associated with immunodeficiency. Proc. Natl. Acad. Sci. U.S.A. 1992,89:3290–3294.
    [89] Christianson, S.W., Greiner, D.L., Hesselton, R.A., et al, Enhanced human CD4+ T cell engraftment in beta2-microglobulin-deficient NOD-scid mice. J. Immunol,1997,158:3578–3586.
    [90] Rothenberg, B.E.,Voland, J.R.,. beta2 knockout mice develop parenchymal iron overload: a putative role for class I genes of the major histocompatibility complex in iron metabolism. Proc. Natl. Acad. Sci. U.S.A. 1996,93:1529–1534.
    [91] Ito, M., Hiramatsu, H., Kobayashi, K,et al.NOD/SCID/gamma(c)(null) mouse: an excellent recipient mouse model for engraftment of human cells. Blood ,2002,100:3175–3182.
    [92] Shultz, L.D., Lang, P.A., Christianson, S.W., et al. NOD/LtSz-Rag1null mice: an immunodeficient and radioresistant model for engraftment of human hematolymphoid cells, HIV infection, and adoptive transfer ofNOD mouse diabetogenic T cells. J. Immunol,2000,164: 2496–2507.
    [93] Shultz, L.D., Banuelos, S., Lyons, B., et al.NOD/LtSz- Rag1nullPfpnull mice: a new model system with increased levels of human peripheral leukocyte and hematopoietic stem-cell engraftment. Transplantation,2003, 76:1036–1042.
    [94] Westphal, C.H., Leder, P., Transposon-generated‘knock-out’and‘knock-in’gene-targeting constructs for use in mice. Curr. Biol, 1997,7: 530– 533.
    [95] Martino, G., Anastasi, J., Feng, J., et al. The fate of human peripheral blood lymphocytes after transplantation into SCID mice. Eur. J. Immunol,1993,23:1023–1028.
    [96] Alegre, M.L., Peterson, L.J., Jeyarajah, D.R., et al, Severe combined immunodeficient miceengrafted with human splenocytes have functional human T cells and reject human allografts. J. Immunol,1994,153: 2738–2749.
    [97] Sandhu, J., Shpitz, B., Gallinger, S.,et al, Human primary immune response in SCID mice engrafted with human peripheral blood lymphocytes. J. Immunol. 1994,152:3806–3813.
    [98] Tary-Lehmann, M., Lehmann, P.V., Schols, D.,et al. Anti-SCID mouse reactivity shapes the human CD4+ T cell repertoire in hu-PBL-SCID chimeras. J. Exp. Med, 1994,180: 1817–1827.
    [99] Hoffmann-Fezer, G., Gall, C., Zengerle, U., et al. Immunohistology and immunocytology of human T-cell chimerism and graft-versus-host disease in SCID mice. Blood, 1993,81: 3440–3448.
    [100] Huppes,W., De Geus, B., Zurcher, C., Van Bekkum, D.W.,. Acute human vs. mouse graft vs. host disease in normal and immunodeficient mice. Eur. J. Immunol,1992,22:197–206.
    [101] Wagar, E.J., Cromwell, M.A., Shultz, L.D., et al., Regulation of human cell engraftment and development of EBV-related lymphoproliferativedisorders in Hu-PBL-scid mice. J. Immunol,2000,165: 518–527.
    [102] Yacoub-Youssef, H., Marcheix, B., Calise, D., et al, Engraftment of human T B and NK cells in CB. 17 SCID/beige mice by transfer of human spleen cells. Transpl. Immunol, 2005, 15:157–164.
    [103] McCune, J.M., Namikawa, R., Kaneshima, H.,et al. The SCID-hu mouse: murine model for the analysis of human hematolymphoid differentiation and function. Science,1988, 241: 1632–1639.
    [104] Hiramatsu, H., Nishikomori, R., Heike, T., et al,. Complete reconstitution of human lymphocytes from cord blood CD34+ cells using the NOD/SCID/gammacnull mice model. Blood ,2003,102: 873–880.
    [105] Yeh, E.T., Zhang, S., Wu, H.D., et al. Transdifferentiation of human peripheral blood CD34+-enriched cell population into cardiomyocytes, endothelial cells, and smooth muscle cells in vivo. Circulation ,2003,108:2070–2073.
    [106] Ishikawa, F., Yasukawa, M., Yoshida, S., et al. Human cord blood- and bone-marrow-derived CD34+ cells regenerate gastrointestinal epithelial cells. FASEB J, 2004,18:1958–1960.
    [107] Fujino, H., Hiramatsu, H., Tsuchiya, A., et al.,. Human cord blood CD34+ cells develop into hepatocytes in the livers of NOD/SCID/gammacnull mice through cell fusion. FASEB J,2007, 21:3499-510
    [108] Libby, P, Pober, J.S. Chronic rejection. Immunity ,2001,14:387–397.
    [109] Lorber, M.I., Wilson, J.H., Robert, M.E., et al. Human allogeneic vascular rejection after arterial transplantation and peripheral lymphoid reconstitution in severe combined immunodeficient mice. Transplantation ,1999, 67: 897–903.
    [110] Grossman HB, Wedemeyer G, Ren L. Human renal carcinoma: Characterization of five new cell lines. J Surg Oncol 1985; 28: 237-44
    [111] Anglard P, Trahan E, Liu S et al. Molecular and cellular characterizationof human renal cell carcinoma cell lines. Cancer Res ,1992,52: 348-56
    [112] Gerharz CD, Ramp U, Olert J et al. Cytomorphological, cytogenetic, and molecular biological characterization of four new human renal carcinoma cell lines of the clear cell type. Virchows Archiv ,1994, 424: 403-9
    [113] An Z, Jiang P, Wang X, et al.Development of a high metastatic orthotopic model of human renal cell carcinoma in nude mice: benefits of fragment implantation compared to cell-suspension injection.Clin Exp Metastasis. 1999,17:265-270
    [114] Shin KH, Ku JL, Kim WH, et al.Establishment and characterization of seven human renal cell carcinoma cell lines. BJU Int, 2000 ;85:130-138.
    [115] Hegele A, Varga Z, von Knobloch R, et al. TGF-beta1 in patients with renal cell carcinoma. Urol Res, 2002,30:126-129.
    [116] Shibata S, Asano T, Noguchi A, et al. Peritoneal macrophages play an important role in eliminating human cells from severe combined immunodeficient mice transplanted with human peripheral blood lymphocytes. Immunology,1998,93:524-532
    [117] Zhang Q, Yang X, Pins M, et al.Adoptive transfer of tumor-reactive transforming growth factor-beta-insensitive CD8+ T cells: eradication of autologous mouse prostate cancer.Cancer Res,2005,65:1761-1769
    [118] Romero P, Zippelius A, Kurth I, et al.Four functionally distinct populations of human effector-memory CD8+ T lymphocytes. J Immunol, 2007,178:4112-4119
    [119] Tian F, Wang L, Qin W,et al. Vaccination with transforming growth factor-beta insensitive dendritic cells suppresses pulmonary metastases of renal carcinoma in mice. Cancer letters,2008,271:333-341
    [120] Zhang Q, Yang XJ, Kundu SD, et al. Blockade of transforming growth factor-{beta} signaling in tumor-reactive CD8(+) T cells activates the antitumor immune response cycle. Mol Cancer Ther,2006 ,5:1733-1743

© 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