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 328 毫秒
1.
在构建并成功表达抗CD3/抗CD20双特异性单链抗体(bscCD3×CD20)的基础上,对其在体外介导T淋巴细胞杀伤Ramous B淋巴瘤细胞的生物活性进行了分析。Annexin V/PI(AV/PI)染色和形态学观察及扫描电镜分析表明bscCD3×CD20介导的B淋巴瘤细胞体外裂解作用是通过先诱导靶细胞凋亡而继发坏死、裂解的方式实现的。非放射性细胞毒性分析表明bscCD3×CD20介导的T淋巴细胞杀伤活性随抗体浓度、反应时间和效靶比的升高而增加。在抗体浓度为5μg/mL、作用时间为24h、效靶比为10∶1时,杀伤活性最高可达87·3%。采用美国SuperArray人细胞凋亡芯片检测细胞杀伤起始阶段细胞凋亡相关基因的表达水平变化,许多凋亡相关基因的表达均发生了不同程度的上调或下调,其中ATM基因表达升高了187倍,p53基因升高了15倍,提示ATM-p53途径可能是bscCD3×CD20介导T细胞诱导B淋巴瘤细胞凋亡的主要途径。  相似文献   

2.
抗CD3/抗Pgp微型双功能抗体的构建和表达   总被引:15,自引:1,他引:14  
构建和表达抗CD3/抗Pgp微型双功能抗体,并测定该微型双功能抗体的生物学活性。 采用PCR和overlap PCR方法构建抗CD3/抗Pgp微型双功能抗体,并用双脱氧终止法测定DNA序列;采用亲和层析法纯化该产物,并用Western blot和分子排阻层析鉴定纯化产物;采用免疫荧光法、放射免疫分析法鉴定纯化产物与靶细胞的结合活性。DNA序列测定结果表明:抗CD3/抗Pgp微型双功能抗体已构建成功,表达可溶性产物的产量达2mg/L以上,纯化产物中二聚体的比例达90%,具有与Jurkat(CD3+)和K562/A02细胞(Pgp+)结合的活性,与抗CD3 ScFv及抗Pgp ScFv的亲合常数相当。成功地构建了抗CD3/抗Pgp微型双功能抗体,并获得高效表达,表达产物具有与相应二个靶抗原结合的活性。  相似文献   

3.
抗体靶向超抗原的抗肿瘤作用研究进展   总被引:1,自引:0,他引:1  
作为一种强大的T细胞激活剂,超抗原具有激活T细胞杀伤HLA-DR 肿瘤细胞的能力,但这种杀伤作用无特异性。抗体靶向超抗原通过抗肿瘤抗体与超抗原偶联,既具有超抗原活性,又具有肿瘤靶向性,从而可以选择性地结合到目标细胞,进行有效的特异性杀伤,因此,抗体靶向超抗原在肿瘤的生物治疗领域具有广泛的应用前景。  相似文献   

4.
[目的]研究旨在制备抗CD47小分子单链抗体,并分析其与抗原的结合能力和阻断作用的活性。[方法]采用DNA合成方法,将抗CD47的单克隆抗体B6H12的重链可变区(VH)和轻链可变区(VL),通过短肽(Gly4Ser)3连接形成B6H12单链抗体(B6H12-scFv),在大肠杆菌中可溶性表达并纯化B6H12-scFv。ELISA和Western Blot方法检测与人重组CD47的结合。检测EC9706和KYSE150两种食管癌细胞表面CD47的表达,细胞ELISA和流式细胞术分析B6H12-scFv与细胞表面CD47的结合。最后采用竞争ELISA分析B6H12-scFv对CD47与髓样细胞表面的信号调节蛋白α(SIRPα)结合的阻断能力。[结果]成功获得纯度达到90%以上的可溶性单链抗体。EC9706和KYSE150细胞均高表达CD47。细胞ELISA和流式检测B6H12-scFv浓度为50μg/m L可与EC9706表面CD47有较好结合。B6H12-scFv浓度为40μg/m L时也可以竞争性阻断CD47与SIRPα的结合。[结论]成功地构建了抗CD47单链抗体,浓度在20μg/m L具有EC9706的结合活性,也具有阻断作用。  相似文献   

5.
利用基因工程方法将鼠源性抗CD3抗体HIT3a的可变区和人源抗体(IgG)的完整的恒定区连接起来,构建全抗型抗CD3嵌合抗体,该型抗体具有较低的免疫源性可作为免疫抑制剂应用于器官移植,减少受体产生免疫排斥,提高移植器官的存活率。利用PCR方法从抗CD3 ScFv重组噬菌体表达载体pCANTAB 5E上扩增抗CD3抗体的轻链和重链可变区,将轻链和重链可变区组装到含有人抗体(IgG)恒定区的表达载体中,构建抗CD3嵌合抗体IgG的轻链和重链表达载体PKN100和PG1D105,并用脂质体法共转染CHO细胞。结果证明,抗CD3嵌合抗体的VL和VH与HIT3a抗体的VL和VH完全相符,ELISA和Western blot检测结果证实转染细胞的培养上清中含有抗CD3嵌合抗体IgG的表达,表达产物能与Jurkat细胞结合,并能竞争性抑制HIT3a抗体和Jurkat细胞结合活性,3H-TdR掺入实验表明, 抗CD3嵌合抗体与亲代抗体HIT3a一样,具有促进外周血单核细胞增殖的作用。我室构建的全抗型抗CD3嵌合抗体分子表达载体可在CHO细胞中稳定表达,表达产物有较好生物活性,具有潜在的临床应用价值。  相似文献   

6.
旨在研究4-1BBL/CD20融合蛋白增强抗CD3/抗CD20 diabody介导的特异性靶向杀伤活性。采用亲和层析法纯化本室构建的抗-CD3/抗-CD20 diabody和4-1BBL/CD20融合蛋白可溶性表达产物;采用calcein释放试验测定其介导的体外靶向杀伤活性;采用人B淋巴瘤细胞系Raji裸鼠移植瘤模型测定其介导的体内靶向杀伤活性。纯化4-1BBL/CD20融合蛋白在体外能增强抗-CD3/抗-CD20 diabody介导激活的T细胞杀伤Raji细胞;在人B淋巴瘤细胞系Raji裸鼠移植瘤模型联合人T淋巴细胞4-1BBL/CD20融合蛋白增强抗-CD3/抗-CD20 diabody高效抑制Raji细胞裸鼠移植瘤的生长,明显延长荷瘤裸鼠的生存时间。在体外和体内4-1BBL/CD20融合蛋白均能增强抗-CD3/抗-CD20 diabody介导激活的T细胞杀伤表达CD20抗原的肿增细胞,是一个有望用于B细胞恶性肿瘤临床治疗的特异性融合蛋白。  相似文献   

7.
BiTEs(bispecificTcellengagers)是一种以T细胞作为效应细胞的双特异性单链抗体 ,它具有两个抗原结合臂 ,可以同时和T细胞及靶细胞结合 ,并激活细胞毒性T细胞杀伤病变细胞。和其它双特异性抗体相比 ,BiTEs的分子柔韧性更好 ,能更好地促进CD3复合体和肿瘤靶标的连接 ,并且它不受T细胞受体和靶细胞上MHCⅠ类分子的约束 ,不需要共刺激分子的参与 ,是一种极具应用潜力的抗体形式。就BiTEs的结构、作用机理及其在肿瘤临床上的应用前景几个方面做一综述。  相似文献   

8.
设计并合成多个60bp左右的DNA小片段 ,经重叠延伸PCR扩增获得1640bp的抗CD3 抗CD2 0双特异性单链抗体完整基因片段 ,将其克隆入真核定点表达载体pcDNA5 FRT中 ,脂质体法转染Flp-InTM CHO细胞 ,获得稳定表达细胞株 ,目的蛋白在上清中的表达量约为 300μg/L。采用Ni_NTA柱对其进行了纯化 ,经SDS-PAGE蛋白电泳及Western-blot分析结果表明 ,含组氨酸标签的目的蛋白的分子量约为 70kD ,与预期结果一致。活细胞间接免疫荧光实验和玫瑰花环实验证明抗CD3抗CD20双特异性单链抗体具有与Ramous(CD20+)及Jurkat(CD3+)细胞特异性结合的活性。光学显微镜下可以观察到抗CD3 抗CD2 0双特异性单链抗体可以有效介导人外周血淋巴细胞裂解B淋巴瘤细胞Ramous。以上工作为进一步了解抗CD3 抗CD2 0双特异性单链抗体的体内体外生物学活性奠定了基础。  相似文献   

9.
目的:利用基因工程技术构建和表达抗人整合素αvβ3单链抗体(scFv)与人可溶性组织因子(sTF)的融合蛋白scFv-sTF。方法:用重叠延伸PCR技术扩增scFv基因,同时用组装PCR方法人工合成sTF基因,然后以酶切连接方式融合2个基因,并克隆至原核表达载体pQE80L中,构建表达质粒pQE80L-scFv-sTF,以重组子转化大肠杆菌M15诱导目的基因表达。结果:SDS-PAGE分析显示工程菌可以表达相对分子质量约55×103的融合蛋白scFv-sTF,Western印迹分析证实表达的目的蛋白具有6×His标签;经低剂量IPTG诱导和较低温度培养,scFv-sTF获得了可溶性表达;纯化回收目的蛋白,ELISA试验证实,该重组抗体分子具有良好的抗原结合活性。结论:构建并表达了抗人整合素αvβ3的单链抗体融合蛋白scFv-sTF,并验证了其抗原结合活性,初步证明它可以与抗原特异性结合,为进一步研究其抗肿瘤作用打下了基础。  相似文献   

10.
抗CD20嵌合抗体的表达与活性检测   总被引:1,自引:0,他引:1  
表达了基因重组抗CD20嵌合抗体并对其生物学活性进行了初步鉴定。设计合成轻、重链可变区序列;提取血液RNA,通过RT-PCR得到人κ、IgG1的轻、重链恒定区序列。运用重叠延伸PCR,连接可变区与恒定区,将轻、重链基因连接至pIRES双表达载体。将质粒以阳离子脂质体转染CHO细胞,ELISA挑选阳性克隆,共获得7株表达较高的克隆,表达量约为2mg/L。扩大培养阳性克隆anti-CD20-1B3,收获上清,以蛋白A进行亲和层析纯化表达蛋白。SDS-PAGE检测表明纯化纯度达到95%,蛋白相对分子量与理论值吻合。以CD20+细胞Raji、Daudi、Ramous检测,表明该抗体能与CD20抗原特异性结合,体外杀伤试验说明抗体能够杀伤CD20+淋巴瘤细胞。  相似文献   

11.
Liu J  Zhao Q  Zhao B  Cheng J  Wang X  Song L  Zhong Z  Lin Q  Huang H 《Biotechnology letters》2005,27(22):1821-1827
A combination of bi-specific antibodies (BsAb), anti-tumor×anti-CD3 and anti-tumor×anti-CD28, is effective in vitro and in vivo, whereas production of two kinds of bi-specific antibodies is labor intensive and administration is complicated. Accordingly, we previously developed a new model of single chain tri-specific antibody (scTsAb), sTRI, which linked both the CD3 and CD28 signals for T-cell activation in one molecule, and demonstrated its capacity for triggering T-cells to kill ovary tumor cells. To improve the pharmacokinetics further and decrease the immunogenicity of scTsAb, we have now generated a new format of scTsAb, TR3H, whose molecular size is smaller than sTRI. Here we describe the construction, purification and characterization of TR3H. TR3H scTsAb bound to effector cells and tumor target cells specifically and induced redirected lyses of ovary tumor cells through freshly isolated, unstimulated human peripheral blood lymphocytes (PBLs). This new format of scTsAb possesses properties that support its potential as a new tumor immunotherapeutic agent. Revisions requested August 2005; Revisions received 14 September 2005  相似文献   

12.
Anti-tumor associated antigen (TAA).CD3.CD28 trispecific antibody(TsAb) is able to provide two signals for fully and continuously activation of T lymphocytes and recruit them around tumor cells, presenting an attractive concept in tumor immunotherapy. Here, a new single chain trispecific antibody (scTsAb), named CEA-scTsAb, was constructed by fusion of anti-CEA (Carcinoma Embryonic Antigen) single chain antibody (scFv), anti-CD3 scFv and anti-CD28 VH, spaced by polypeptide interlinkers taken from the fragment of constant region (FC) of human IgG and human serum albumin (HSA). It was expressed in Escherichia coli at low temperature (30 degrees C) with up to 50% of the antibody being present in soluble form. After one step of DEAE anion chromatography, the soluble product was sufficiently pure for further in vitro activity assays. First, it was proved that CEA-scTsAb could recognize three antigens (CEA, CD28 and Jurkat cell membrane antigen) specifically and could distinguish antigen positive cells from antigen negative cells in vitro. Then fresh PBMC (peripheral blood mononuclear cells), without being pre-treated by co-stimulatory reagents, such as IL-2 or CD28 mAb, were used as effector cells to test their ability to mediate tumor specific cytolysis of CEA-positive tumor cells, SW1116. It was found by photomicrography that T lymphocytes were attracted to SW1116 cells in the presence of CEA-scTsAb, which resulted in effective cytolysis of tumor cells. As shown by MTT assay, the efficacy of tumor specific cytolysis mediated by CEA-scTsAb related to both the quantity and activation of PBMC. At an effector cells/target cells ratio (E/T) of 5, it was proved by dual-color FACS with propidium iodide (PI) and FITC-annexin V that both necrosis and apoptosis of tumor cells were causes of tumor specific cytolysis. In summary, a new single chain trispecific (CEA x CD3 x CD28) antibody was constructed and characterized carefully in this paper and was found to possess functions: (i) to activate T lymphocytes independently of additional co-stimulatory signal, (ii) to attract activated T lymphocytes around CEA-positive tumor cells, (iii) to attack CEA-positive tumor cells with recruited T lymphocytes. Because it recognizes a widely distributed tumor antigen (CEA), with moderate molecular weight (about 75 kDa) and a simple production procedure, and is able to mediate a high level of tumor specific cytolysis without any additional co-stimulating reagents, CEA-scTsAb is very promising for the task of immunotherapy in future.  相似文献   

13.
Targeted cancer therapy concepts often aim at the induction of adjuvant antitumor immunity or stimulation of tumor cell apoptosis. There is further evidence that combined application of immune stimulating and tumor apoptosis-inducing compounds elicits a synergistic antitumor effect. Here, we describe the development and characterization of bifunctional fusion proteins consisting of a single-chain variable fragment (scFv) domain derived from the CD40-specific monoclonal antibody G28-5 that is fused to the N-terminus of stabilized trimeric soluble variants of the death ligand TNF-related apoptosis-inducing ligand (TRAIL). As shown before by us and others for other cell surface antigen-targeted scFv-TRAIL fusion proteins, scFv:G28-TRAIL displayed an enhanced capacity to induce apoptosis upon CD40 binding. Studies with scFv:G28 fusion proteins of TRAIL mutants that discriminate between the two TRAIL death receptors, TRAILR1 and TRAILR2, further revealed that the CD40 binding-dependent mode of apoptosis induction of scFv:G28-TRAIL is operable with each of the two TRAIL death receptors. Binding of scFv:G28-TRAIL fusion proteins to CD40 not only result in enhanced TRAIL death receptor signaling but also in activation of the targeted CD40 molecule. In accordance with the latter, the scFv:G28-TRAIL fusion proteins triggered strong CD40-mediated maturation of dendritic cells. The CD40-targeted TRAIL fusion proteins described in this study therefore represent a novel type of bifunctional fusion proteins that couple stimulation of antigen presenting cells and apoptosis induction.  相似文献   

14.
Single-chain variable fragment (scFv) is a fusion protein of the variable regions of the heavy (VH) and light (VL) chains of immunoglobulin, connected with a short linker peptide of 10 to about 20 amino acids. In this study, the scFv of a monoclonal antibody against the third domain of human CD4 was cloned from OKT4 hybridoma cells using the phage display technique and produced in E. coli. The expression, production, and purification of anti-CD4 scFv were tested using SDS-PAGE and Western blot, and the specificity of anti-CD4 scFv was examined using ELISA. A 31 kDa recombinant anti-CD4 scFv was expressed and produced in bacteria, which was confirmed by SDS-PAGE and Western blot assays. Sequence analysis proved the ScFv structure of the construct. It was able to bind to CD4 in quality ELISA assay. The canonical structure of anti-CD4 scFv antibody was obtained using the SWISS_MODEL bioinformatics tool for comparing with the scFv general structure. To the best of our knowledge, this is the first report for generating scFv against human CD4 antigen. Engineered anti-CD4 scFv could be used in immunological studies, including fluorochrome conjugation, bispecific antibody production, bifunctional protein synthesis, and other genetic engineering manipulations. Since the binding site of our product is domain 3 (D3) of the CD4 molecule and different from the CD4 immunological main domain, including D1 and D2, further studies are needed to evaluate the anti-CD4 scFv potential for diagnostic and therapeutic applications.  相似文献   

15.
Bispecific antibodies (Bs-Abs) containing an anti-CD3 and an anti-TAA specificity can recruit T cells to the tumor for cancer immunotherapy. To be effective, efficient activation at the tumor site is a prerequisite. This can be achieved by triggering both the T-cell receptor and the co-stimulatory molecule CD28. We engineered two recombinant cross-interacting Bs-Abs (CriBs-Abs) by incorporating a peptide tag and its cognate single-chain variable fragment (scFv), respectively, into a pair of (tumor × CD3) and (tumor × CD28) binding Bs-Abs. A 30-fold lower concentration of the activating CriBs-Ab as compared to non interacting Bs-Ab was sufficient for strong T-cell activation in the presence of tumor cells. One thousand-fold higher concentrations of both CriBs-Abs were required for marginal T-cell activation (70-fold below maximal response) in the absence of tumor cells. An optimized stoichiometry (1 : 1000) of activating versus co-stimulating CriBs-Ab thus allowed low doses of activating CriBs-Ab to induce tumor-cell dependent T-cell activation when used in combination with high concentrations of the pre-targeted co-stimulating CriBs-Ab in vitro. This indicates a large window of operation in which only tumor cell dependent T-cell activation is induced and systemic tumor cell independent T-cell activation is avoided, while ensuring optimal activation with a low concentration of the activating CriBs-Ab, which has the highest potential to induce toxic effects in vivo.  相似文献   

16.
Persistent activation of T-lymphocytes requires two signals: one is initiated by T-cell receptor binding to antigenic peptide presented by MHC molecules. In addition, binding of the B7 family members CD80 or CD86 on professional antigen presenting cells to CD28 on T cells is considered to provide an important costimulatory signal. Activation without costimulation induces T-cell unresponsiveness or anergy. To selectively localize costimulatory activity to the surface of tumor cells and enhance activation of tumor-specific T cells, we have developed a novel molecular design for bispecific costimulatory proteins with antibody-like structure. Within a single polypeptide chain we have assembled the IgV-like, CD28-binding domain of human CD86 (CD86(111)) together with hinge, CH2 and CH3 domains of human IgG1, and the scFv(FRP5) antibody fragment which recognizes the ErbB2 (HER2) protooncogene present at high levels on the surface of many human tumor cells. Upon expression in the yeast Pichia pastoris, the resulting CD86(111)-IgG-scFv(FRP5) protein could be purified as a homodimeric, tetravalent molecule from culture supernatants using single-step affinity chromatography. Bispecific binding of the molecule to ErbB2 on the surface of tumor cells and to the B7 counter receptor CTLA-4 was demonstrated by FACS analysis. Potent costimulatory activity of chimeric CD86(111)-IgG-scFv(FRP5) was confirmed by its ability to stimulate the proliferation of primary human lymphocytes pre-activated by low concentrations of anti-CD3 antibody. Our results suggest that such multivalent soluble proteins which combine specific targeting to tumor cells with costimulatory activity may become useful tools to elicit and/or improve T-cell mediated, tumor-specific immune responses.  相似文献   

17.
Recombinant bispecific antibodies such as tandem scFv molecules (taFv), diabodies (Db), or single chain diabodies (scDb) have shown to be able to retarget T lymphocytes to tumor cells, leading to their destruction. However, therapeutic efficacy is hampered by a short serum half-life of these small molecules having molecule masses of 50-60 kDa. Thus, improvement of the pharmacokinetic properties of small bispecific antibody formats is required to enhance efficacy in vivo. In this study, we generated several recombinant bispecific antibody-albumin fusion proteins and analyzed these molecules for biological activity and pharmacokinetic properties. Three recombinant antibody formats were produced by fusing two different scFv molecules, bispecific scDb or taFv molecules, respectively, to human serum albumin (HSA). These constructs (scFv(2)-HSA, scDb-HSA, taFv-HSA), directed against the tumor antigen carcinoembryonic antigen (CEA) and the T cell receptor complex molecule CD3, retained full binding capacity to both antigens compared with unfused scFv, scDb, and taFv molecules. Tumor antigen-specific retargeting and activation of T cells as monitored by interleukin-2 release was observed for scDb, scDb-HSA, taFv-HSA, and to a lesser extent for scFv(2)-HSA. T cell activation could be further enhanced by a target cell-specific costimulatory signal provided by a B7-DbCEA fusion protein. Furthermore, we could demonstrate that fusion to serum albumin strongly increases circulation time of recombinant bispecific antibodies. In addition, our comparative study indicates that single chain diabody-albumin fusion proteins seem to be the most promising format for further studying cytotoxic activities in vitro and in vivo.  相似文献   

18.
目的:将已成功构建表达anti-CD20scFv/CD80/CD28/zeta转染人T淋巴细胞,体外观察该类细胞特异性清除CD20+原代慢性淋巴细胞白血病(CLL)细胞的能力,为肿瘤的过继免疫治疗提供新思路。方法:将本室成功构建的含anti-CD20scFv/IgGFc/CD80片段的PLNCX质粒,转染PA317包装细胞,挑取高滴度的包装细胞株收获逆转录病毒,用收获的病毒感染刺激分裂的人外周血T细胞,经G418筛选后与CD20+原代CLL细胞在体外共同培养,在显微镜下观察CD20+的原代CLL细胞生长状态,ELISA检测试剂盒检测T细胞分泌细胞因子的功能。结果:重组基因修饰的T细胞能在体外杀伤CD20+原代CLL细胞,而对CD20-细胞无杀伤作用;同时靶细胞为CD20+组上清液中IL-2(1301.00pg/ml)和IFN-γ(602.18pg/ml)水平与CD20-组相比明显升高。结论:嵌合锚定T细胞能够成功构建;该类T细胞在体外能特异性杀伤CD20+的原代CLL细胞。  相似文献   

19.
为构建能同时表达抗人CD28嵌合重链和嵌合轻链基因的双启动子杆状病毒转移载体,利用PCR法扩增出抗人CD28鼠源性单抗的可变区基因和人IgG1的恒定区基因,再采用一种不需要限制性核酸内切酶和连接酶的新方法——三引物PCR(TP-PCR)法将两者拼接后分别插入杆状病毒转移载体pAcUW3的php10启动子后,并通过酶切、电泳、PCR扩增和测序对重组体进行了鉴定。研究结果表明,TP-PCR法快速、方便、准确,无需设计外源的DNA序列,就能构建完好的融合表达基因。该转移载体的构建成功为嵌合抗体基因在昆虫细胞中的表达奠定了基础。  相似文献   

20.
G3(3) is a novel murine monoclonal antibody directed against the CD3 antigen of human T lymphocytes which could be used to analyze lymphoid malignancies. We have produced and characterized a recombinant colorimetric immunoconjugate with the antigen-binding specificity of antibody G3(3). A gene encoding a single-chain antibody variable fragment (scFv) was assembled using the original hybridoma cells as a source of antibody variable heavy (VH) and variable light (VL) chain genes. The chimeric gene was introduced into a prokaryotic expression vector in order to produce a soluble scFv fused to bacterial alkaline phosphatase. DNA sequencing and Western blotting analyses demonstrated the integrity of the soluble immunoconjugate recovered from induced recombinant bacteria. The scFv/AP protein was bifunctional and similar in immunoreactivity to the parent G3(3) antibody. Flow cytometry and immunostaining experiments confirmed that the activity of the scFv/AP protein compares favourably with that of the parent antibody. The scFv/AP conjugate was bound to CD3 antigen at the surface of T cells and was directly detected by its enzymatic activity. Thus this novel fusion protein has potential applications as an immunodiagnostic reagent.  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