首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
Clinical trials with tumor antigen genetically modified dendritic cells   总被引:1,自引:0,他引:1  
Tumor antigen genetically modified dendritic cells (DC) have been extensively tested as cancer vaccine approaches in preclinical models. This testing has provided evidence of their ability to generate coordinated antitumor CD8+ cytotoxic T lymphocyte (CTL) and CD4+ T-helper cell responses. Their antitumor activity compared favorably to multiple other vaccination strategies in mice. This approach has been brought to patients within nine pilot clinical trials reported to date. These clinical trials have tested both RNA and DNA as means to introduce the foreign genetic material into the DC. Administration to human subjects has proven to be both feasible and safe. There is clear evidence of the ability to activate both CD8+ CTL and CD4+ T-helper cells, which has been the major scientific endpoint in most of these trials. However, antitumor activity has been marginal thus far. In conclusion, tumor antigen genetically modified DC are a feasible strategy to activate tumor-specific T cells in humans.  相似文献   

2.
Granulocyte macrophage colony-stimulating factor (GM-CSF) is a key cytokine for the generation and stimulation of dendritic cells (DCs), and it may also play a pivotal role in promoting the survival of DCs. In this study, the feasibility of creating a cancer vaccine using DCs adenovirally transduced with the carcinoembryonic antigen (CEA) gene and the GM-CSF gene was examined. In addition, the effect of the co-transduction of GM-CSF gene on the lifespan of these genetically modified DCs was determined. A cytotoxic assay using peripheral blood mononuclear cell (PBMC)-derived cytotoxic T lymphocytes (CTLs) was performed in a 4-h 51Cr release assay. The apoptosis of DCs was examined by TdT-mediated dUTP-FITC nick end labeling (TUNEL) assay. CEA-specific CTLs were generated from PBMCs stimulated with genetically modified DCs expressing CEA. The cytotoxicity of these CTLs was augmented by co-transduction of DCs with the GM-CSF gene. Co-transduction of the GM-CSF gene into DCs inhibited apoptosis of these DCs themselves via up-regulation of Bcl-x(L) expression, leading to the extension of the lifespan of these DCs. Furthermore, the transduction of the GM-CSF gene into DCs also suppressed the incidence of apoptosis of DCs induced by transforming growth factor-beta1 (TGFbeta-1). Immunotherapy using these genetically modified DCs may therefore be useful with several advantages as follows: i) adenoviral toxicity to DCs can be reduced; ii) the lifespan of vaccinated DCs can be prolonged; and iii) GM-CSF may protect DCs from apoptosis induced by tumor-derived TGFbeta-1 in the regional lymph nodes.  相似文献   

3.
This study was designed to determine whether the vaccination of genetically modified dendritic cells (DCs) simultaneously expressing carcinoembryonic antigen (CEA), granulocyte macrophage colony-stimulating factor (GM-CSF) and interleukin 12 (IL-12) can overcome the peripheral T-cell tolerance to CEA and thereby elicit a therapeutic response in CEA transgenic mice. CEA transgenic mice were immunized once by subcutaneous injection with DCs adenovirally transduced with CEA and T helper-type 1 cytokine genes. The cytotoxic activity of spleen cells against CEA-expressing tumors, MC38-CEA, in the mice immunized with DCs expressing CEA (DC-AxCACEA) was higher than that in those immunized with DCs-AxCALacZ (p < 0.0001), and was augmented by the cotransduction with the GM-CSF/IL-12 gene (p < 0.05). The vaccination with DC-AxCACEA/GM-CSF/IL-12 could elicit a more potent therapeutic immunity than the vaccination with DC-AxCACEA in subcutaneous tumor models (p < 0.0001), and 4 of 5 mice showed a complete eradication of the subcutaneous tumors in these vaccination groups. Even in a large tumor model, this vaccination therapy completely eliminated the subcutaneous tumors in all mice. This antitumor activity mostly vanished with the depletion of CD8(+) T cells and NK cells in vivo and was completely abrogated with the depletion of CD4(+) T cells. A histopathological examination showed no evidence of an autoimmune reaction. No other adverse effects were observed. This vaccination strategy resulted in the generation of highly efficient therapeutic immune responses against MC38-CEA in the absence of autoimmune responses and demonstrated no adverse effects, and may therefore be useful for future clinical applications as a cancer vaccine therapy.  相似文献   

4.
Wei H  Wang H  Lu B  Li B  Hou S  Qian W  Fan K  Dai J  Zhao J  Guo Y 《Cancer research》2008,68(10):3854-3862
Modest clinical outcomes of dendritic cell (DC) vaccine trials call for novel strategies. In this study, we have created a chimeric CD40 molecule that incorporates a single chain Fv (scFv) molecule specific for human ErbB2 antigen and fusing to the membrane spanning and cytosolic domains of murine CD40. After adenoviral transfer to bone marrow-derived DC, this chimeric receptor (CR) induced nuclear factor-kappaB (NF-kappaB)-dependent DC activation and effector function when cultured with immobilized ErbB2 protein or ErbB2-positive tumor cells in vitro. In vivo migration assays showed that approximately 40% injected CR-modified DC (scFv-CD40-DC) effectively migrated to ErbB2-positive tumors, where they were activated after ErbB2 antigen stimulation, and sequentially homed into the draining lymph nodes. In murine ErbB2-positive D2F2/E2 breast tumor (BALB/c) and EL4/E2 thymoma (C57BL/6) models, i.v. injection of 1 x 10(6) scFv-CD40-DC significantly inhibited tumor growth and cured established tumors. Importantly, the cured mice treated by injection of scFv-CD40-DC were effective in preventing both ErbB2-positive and parental ErbB2-negative tumor rechallenge. Analysis of the underlying mechanism revealed that i.v. infusion of scFv-CD40-DC elicited tumor-specific CTL responses, and the transfer of CTLs from scFv-CD40-DC-treated mice protected naive mice against a subsequent tumor challenge. These results support the concept that genetic modification of DC with tumor-associated antigen-specific CD40 chimeric receptor might be a useful strategy for treatment of human cancers.  相似文献   

5.
Dendritic cells (DCs) are powerful antigen-presenting cells that process antigens and present peptide epitopes in the context of the major histocompatibility complex molecules to generate immune responses. DCs are being studied as potential anticancer vaccines because of their ability to present antigens to naive T cells and to stimulate the expansion of antigen-specific T-cell populations. We investigated an antitumor vaccination using DCs modified by transfer of a nonsignaling neu oncogene, a homologue of human HER-2/neu, in a transgenic model of breast cancer. BALB-neuT mice develop breast cancers as a consequence of mammary gland-specific expression of an activated neu oncogene. We vaccinated BALB-neuT mice with bone marrow-derived DCs transduced with Ad.Neu, a recombinant adenovirus expressing a truncated neu oncoprotein. The vaccine stimulated the production of specific anti-neu antibodies, enhanced interferon-gamma expression by T cells, and prevented or delayed the onset of mammary carcinomas in the mice. Over 65% of vaccinated mice remained tumor free at 28 weeks of age, whereas all of the mice in the control groups developed tumors. When challenged with a neu-expressing breast cancer cell line, vaccinated tumor-free animals had delayed tumor growth compared with controls. The antitumor effect of the vaccine was specific for expression of neu. Studies showed that CD4+ T cells were required in order to generate antitumor immunity. Importantly, the effectiveness of the vaccine was not diminished by preexisting immunity to adenovirus, whereas the protection afforded by vaccination that used direct injection of Ad.Neu was markedly reduced in mice with anti-adenovirus antibody titers. DCs modified by recombinant adenoviruses expressing tumor-associated antigens may provide an effective antitumor vaccination strategy.  相似文献   

6.
7.
Interleukin (IL)-12 is a key factor for inducing cellular immune responses, which play a central role in the eradication of cancer. In the present study, in order to create a dendritic cell (DC)-based vaccine capable of positively skewing immune response toward a cellular immunity-dominant state, we analyzed immunological characteristics and vaccine efficacy of DCs cotransduced with melanoma-associated antigen (gp100) and IL-12 gene (gp100+IL12/DCs) by using RGD fiber-mutant adenovirus vector (AdRGD), which enables highly efficient gene transduction into DCs. gp100+IL12/DCs could simultaneously express cytoplasmic gp100 and secretory IL-12 at levels comparable to DCs transduced with each gene alone. In comparison with DCs transduced with gp100 alone (gp100/DCs), upregulation of major histocompatibility complex class I, CD40, and CD86 molecules on the cell surface and more potent T-cell-stimulating ability for proliferation and interferon-gamma secretion were observed as characteristic changes in gp100+IL12/DCs. In addition, administration of gp100+IL12/DCs, which were prepared by a relatively low dose of AdRGD-IL12, could induce more potent tumor-specific cellular immunity in the murine B16BL6 melanoma model than vaccination with gp100/DCs. However, antitumor effect and B16BL6-specific cytotoxic T-lymphocyte activity in mice vaccinated with gp100+IL12/DCs diminished with increasing AdRGD-IL12 dose during gene transduction, and paralleled the decrease in presentation levels via MHC class I molecules for antigen transduced with another AdRGD. Collectively, our results suggested that optimization of combined vector dose was required for development of a more efficacious DC-based vaccine for cancer immunotherapy, which relied on genetic engineering to simultaneously express tumor-associated antigen and IL-12.  相似文献   

8.
Immunotherapy with tumor-associated antigen-pulsed, ex vivo-generated dendritic cells (DCs) is a promising approach for the treatment of cancer that has shown efficacy in animal models and is now being tested in the clinic. The majority of studies performed to date make use of a single tumor-associated epitope. However, because of the high rate of mutation in tumor cells allowing for loss of expression of a single antigen, it is likely that use of multiple antigenic epitopes will induce a broader, longer-lasting, and effective tumor-specific immune response. Multiple vehicles for loading DCs with multiple antigenic epitopes are under investigation to determine the most effective method for vaccination, with many of these methods showing promise. These loading methods, as well as other critical considerations for making DC vaccination as efficacious as possible, are discussed in this article.  相似文献   

9.
A murine model of dendritic cell (DC)-based genetic immunization to a defined human melanoma antigen (Ag), MART-1/Melan-A (MART-1), was developed. The MART-1 gene was stably transfected into the nonimmunogenic mouse fibrosarcoma cell line NFSA that is syngeneic in C3Hf/Sem/Kam (C3H, H-2k) mice to generate the NFSA(MART1) cell line. In vivo protection from a lethal NFSA(MART1) tumor challenge could be generated by DCs transduced with a recombinant adenovirus (AdV) vector expressing MART-1 (AdVMART1). This model has the following characteristics: (a) immunological specificity and memory, (b) comparable protection for varying transduction multiplicities of infection, cell doses, and sites of DC inoculation but, interestingly, worse protection with increasing numbers of vaccinations, (c) the ability to treat small established tumors, (d) an absolute requirement for CD8 and CD4 T cells, (e) generation of MART-1-specific splenic cytotoxic T lymphocytes, and (f) up-regulation of both T helper type 1 and T helper type 2 cytokines. Genetically engineered DCs presenting defined tumor Ags represent an attractive method to generate effective immune responses.  相似文献   

10.
背景与目的:T细胞介导的特异性免疫效应在慢性粒细胞白血病的治疗中发挥着重要作用,而以树突细胞(dendriticcell,DC)为中心的免疫治疗是目前肿瘤生物免疫学治疗的热点之一。本研究以复制缺陷型重组腺病毒为载体,介导慢性粒细胞白血病bcr/abl基因片段转导DC制备疫苗,观察bcr/abl基因修饰DC疫苗诱导产生的特异性细胞毒性T淋巴细胞(cytotoxic T lymphocytes,CTLs)在体外对白血病K562细胞的杀伤效应。方法:应用RT—PCR法扩增慢性粒细胞白血病bcr/abl基因片段.构建复制缺陷型重组腺病毒质粒。并包装产生重组腺病毒。体外诱导培养外周血单个核细胞来源DC,观察DC分别经重组腺病毒转染及多肽负载后,诱导产生特异性的CTLs在体外对白血病K562细胞的杀伤效应。结果:成功构建了携带bcr/abl基因片段的复制缺陷型重组腺病毒表达载体,包装产生的重组腺病毒滴度高达2.0×10^10pfu/mL。体外转染DC效率达到50%~60%,成功制备了bcr/abl特异性DC疫苗。体外实验中,在40:1和20:1效靶比下,bcr/abl基因修饰DC疫苗对K562细胞的杀伤效应分别为(47.6±4.7)%和(47.5±1.6)%,多肽负载DC为(25.8±4.4)%和(24.6±6.3)%,空白DC为(5.7±1.3)%和(4.5±1.6)%,基因修饰DC疫苗诱导的杀伤效应明显高于多肽负载及空白组(P〈0.05)。结论:以重组腺病毒为载体介导bcr/abl基因片段转导DC制备的基因修饰DC疫苗,具有显著的诱导CTLs杀伤白血病K562细胞的作用。  相似文献   

11.
PURPOSE: Triggering of CD40 on antigen-presenting cells via its ligand CD154 is an important event in the initial phase of an immune response against cancer cells. In this study, we investigated the effects of adenoviral CD154 immunomodulatory gene therapy on the activation of human dendritic cells (DCs) in a well-defined in vitro system. EXPERIMENTAL DESIGN: Human bladder cancer cell lines and tumor cells from patients with renal cell carcinoma (RCC) were transduced with Ad-CD154 vectors or control vectors. Activation of human in vitro generated DCs after coculture with transduced tumor cells was analyzed. Therapeutic efficacy and cytotoxic T-lymphocyte (CTL) activity were assessed in a subcutaneous (s.c.) murine bladder cancer model. RESULTS: Human bladder cancer cell lines expressing CD154 showed a decreased growth rate, increased apoptosis, and modulated expression of molecules important for recognition by cytotoxic lymphocytes. Further, CD154-expressing allogeneic bladder tumor cell lines and autologous tumor cells from patients with renal cell cancer induced maturation of DCs and stimulated IFN-gamma production from lymphocytes cocultured with mature DCs. In vivo studies showed that CD154 gene therapy was highly effective in wild-type mice but only minimally effective in nude mice. Consequently, strong tumor-specific CTL activity was detected in mice vaccinated with tumor cells expressing CD154. CONCLUSIONS: Using tumor cell lines as well as patient-derived material, we could show that tumor cells expressing CD154 efficiently induce maturation and activation of DCs as well as activation of lymphocytes. Our murine in vivo studies demonstrate that lymphocytes contribute to the observed antitumor effect in a s.c. bladder tumor model. These studies should stimulate CD154 gene therapy approaches for the treatment of urologic malignancies.  相似文献   

12.
目的: 利用非小细胞肺癌(nonsmallcell lung cancer; NSCLC)患者的树突状细胞(dendritic cell,DC)负载自体肿瘤抗原制备DCCIK(cytokine induced killer cell)瘤苗,观察其在体外对患者自身肿瘤细胞的杀伤作用。方法:提取NSCLC患者外周血单个核细胞(peripheral blood mononuc  相似文献   

13.
The use of dendritic cells in cancer therapy   总被引:33,自引:0,他引:33  
Although the immune system evolved to protect the host from infection, what fires the popular imagination is its potential to recognise and destroy cancer. The immune system can generate potent cytotoxicity (eg transplant rejection), but can these mechanisms be harnessed for therapeutic benefit in patients with cancer? The discovery of an ever-increasing array of tumour antigens shows clearly that the targets exist. The challenge lies in generating a sufficiently potent response towards them. Central to the processes of antigen recognition, processing, and presentation to the immune system are dendritic cells. Understanding of the relation between these and the cellular immune response is crucial to elucidation of how to manipulate immune responses. The past 20 years have witnessed a dramatic expansion in this understanding and led to the first early-phase clinical trials of dendritic cells for the treatment of cancer. These studies have established the safety and feasibility of this approach and have produced encouraging evidence of therapeutic efficacy. This paper reviews the biology of dendritic cells and their use in clinical trials, as well as highlighting issues for future trial design.  相似文献   

14.
15.
Dendritic cells (DCs) are important cells in initiating an immune response. A generation of functional DCs has potential clinical use in treating cancer. However, the source of DCs and patient immunodeficiency with cancer have been hindrances in clinical therapy. We generated DCs from human umbilical cord blood mononuclear cells (UBMCs) with recombinant human granulocyte-macrophage colony stimulating factor, recombinant human interleukin-4, and recombinant human tumor necrosis factor-alpha. The mature DC-A549 lung cancer vaccine (AgL-DC) was prepared through loading A549 lysate, treating with lipopolysaccharide (LPS) and positive selecting with CD83 magnetic beads. AgL-DC can secrete interleukin (IL)-12 and IL-1. Further in vitro analysis showed that AgL-DC notably induced human UBMC lymphocyte proliferation (p < 0.01) by 3-(4,5-dimethylthiazol-z-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, increased the cytotoxic T-lymphocyte (CTL) activity of UBMC lymphocytes against A549 cells (p < 0.05, at effector cells:target cells ratios of 50:1 and 100:1) by lactate dehydrogenase (LDH) cytotoxic assay, and improved production of IL-6 and tumor necrosis factor-beta (p < 0.01, p < 0.05) by enzyme-linked immunosorbent assay. Subsequently, the reconstitute immunity model in severe combined immunodeficiencies (SCID) mice has been established using human UBMC transplantation, and similar trends to results of UBMC in vitro experiments have been shown in lymphocyte proliferation, CTL activity, and IL-6 and tumor necrosis factor-beta secretion levels in these models. AgL-DC also significantly (p < 0.01) increased the antitumor effect in vivo. The tumor infiltrating immunocytes were positively expressed human CD83 and CD3 molecules, and they were negatively expressed in tumor tissue treated with control. These results have demonstrated that umbilical cord DCs are a useful source of vaccine cells for augmenting CTL-mediated cytotoxicity and have potential usefulness in cellular therapy for human cancer in a new vaccination strategy.  相似文献   

16.
目的 观察经肺癌肿瘤可溶性抗原(TSA)和超抗原金黄色葡萄球菌肠毒素A(SEA)联合修饰致敏树突状细胞(DC)体外诱导抗肺癌的免疫效应。方法 3mol/L氯化钾提取法获得人肺癌细胞GLC 82的可溶性抗原;从人外周血单个核细胞(PBMC)中诱导扩增DC,并用流式细胞仪(FCM)检测表型;以肺癌TSA和SEA联合修饰致敏的DC、单纯肺癌抗原致敏的DC和未经抗原修饰的DC分别与同种异体外周血T淋巴细胞共同孵育,刺激T淋巴细胞活化增殖(作为效应细胞分别称为TSA-SEA-DCL、TSA-DCL、DCL),直接活细胞计数法观察增殖倍数;MTT法检测不同DC∶T淋巴细胞比例的效应细胞对靶细胞GLC-82的体外杀伤效应。结果 诱导出高表达CD1a、CD80、HLA-DR的DC,光镜下具有典型的DC特性;联合抗原修饰后的DC具有较强的免疫刺激活性,少量致敏DC即可强烈激发T细胞的增殖;TSA-SEA-DCL对靶细胞GLC-82的杀伤率明显高于TSA-DCL及DCL;联合抗原修饰的DC以1∶100与T淋巴细胞共孵后的杀伤肿瘤细胞效应最强。结论 经肺癌TSA和SEA联合修饰致敏的DC可强烈激发同种异体T淋巴细胞活化增殖;肺癌TSA联合超抗原SEA诱导的DC疫苗对肺癌细胞有高效杀伤作用,经肺癌TSA与超抗原SEA联合修饰DC的活性明显强于单用肺癌TSA。  相似文献   

17.
目的:探讨进展期胃癌根治术后化疗联合自体肿瘤细胞抗原致敏树突状细胞-细胞因子诱导的杀伤(autologous tumor antigen load dendritic cells-cytokin-induced killer,Ag-DC-CIK)细胞治疗的疗效和安全性.方法:收集2013年1月至2014年3月于荆州市中心医院胃肠外科诊断为进展期(Ⅱ期和Ⅲ期)胃癌的60例患者,均接受胃癌D2根治术,术后按照随机数字表法分为两组,单纯化疗组采用FOLFOX化疗方案,给予6个周期化疗;联合治疗组除给予上述化疗外,同时给予Ag-DC-CIK细胞进行治疗.随访期2年,观察两组患者2年OS和PFS、外周血T细胞亚群免疫学指标(CD3+、CD4+、CD8+、CD3+CD56+)、生活质量评分、化疗不良反应分级.结果:联合治疗组患者2年OS及PFS较单纯化疗组明显提高(均P<0.05).联合治疗组治疗前后外周血T细胞亚群水平无明显变化(P>0.05),而单纯化疗组治疗后外周血T细胞亚群水平明显降低(P<0.05),且明显低于联合治疗组(P<0.05);联合治疗组的综合生活质量评分明显高于单纯化疗组[(7.25±1.56) vs(5.54±1.27)分,P<0.05].联合治疗组不良反应发生率明显低于单纯化疗组(10.0% vs 20.0%,P<0.05).结论:Ag-DC-CIK细胞治疗联合化疗能显著提高胃癌术后患者的2年OS及PFS,保护机体的免疫功能,减少化疗的不良反应,改善其生活质量.  相似文献   

18.
19.
The close cooperation of both innate and acquired immunity is essential for the induction of truly effective antitumor immunity. We tested a strategy to enhance the cross-talk between NKT cells and conventional antigen-specific T cells with the use of alpha GalCer-loaded dendritic cells genetically engineered to express antigen plus chemokine, attracting both conventional T cells and NKT cells. DC genetically engineered to express a model antigen, OVA, along with SLC/CCL21 or monokine induced by IFN-gamma/CXCL9, had been generated using a method based on in vitro differentiation of DC from mouse ES cells. The ES-DC were loaded with alpha-GalCer and transferred to mice bearing MO4, an OVA-expressing melanoma, and their capacity to evoke antitumor immunity was evaluated. In vivo transfer of either OVA-expressing ES-DC, stimulating OVA-reactive T cells, or alpha-GalCer-loaded non-transfectant ES-DC, stimulating NKT cells, elicited a significant but limited degree of protection against the i.p. disseminated MO4. A more potent antitumor effect was observed when alpha-GalCer was loaded to ES-DC expressing OVA before in vivo transfer, and the effect was abrogated by the administration of anti-CD8, anti-NK1.1 or anti-asialo GM1 antibody. alpha-GalCer-loaded double transfectant ES-DC expressing SLC along with OVA induced the most potent antitumor immunity. Thus, alpha-GalCer-loaded ES-DC expressing tumor-associated antigen along with SLC can stimulate multiple subsets of effector cells to induce a potent therapeutic effect against peritoneally disseminated tumor cells. The present study suggests a novel way to use alpha-GalCer in immunotherapy for peritoneally  相似文献   

20.
目的 评价自体肿瘤冻融抗原致敏的自体树突状细胞(ADC)疫苗免疫治疗,对处于Ⅱ、ⅢA期雌、孕激素受体双阴性表达乳腺癌患者的疾病进展及生存情况的影响。方法利用自体肿瘤冻融抗原致敏的CD14+前体细胞产生树突状细胞,在细胞因子的作用下开始成熟,制备ADC疫苗。选取符合入组标准的63例患者,根据治疗方案分为实验组和对照组,实验组共接受4次皮下ADC回输。测定实验组疫苗接种前后IFN-γ+/CD8+T细胞含量及两组针对抗肿瘤裂解物发生的Ⅳ型变态反应情况,随访两组的疾病进展情况。结果 疫苗接种后提高了IFN-γ+/CD8+T细胞数量,与疫苗接种前比较差异有统计学意义[(23.4±4.1)% vs.(14.3±2.0)%, P<0.05];实验组18例(58.1%)为Ⅳ型变态反应阳性,而对照组均为阴性,差异有统计学意义(P<0.05)。实验组的3年无进展生存率高于对照组,差异有统计学意义(76.9% vs. 31.0%, P<0.05)。实验组未发生严重不良反应。结论ADC疫苗可能通过触发乳腺癌患者的免疫应答机制来治疗乳腺癌,具有较好的效果,并延长无进展生存期。  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号