15/10/02说明:此前论坛服务器频繁出错,现已更换服务器。今后论坛继续数据库备份,不备份上传附件。

肝胆相照论坛

 

 

肝胆相照论坛 论坛 学术讨论& HBV English Adoptive T细胞治疗
查看: 1015|回复: 7
go

Adoptive T细胞治疗 [复制链接]

Rank: 8Rank: 8

现金
62111 元 
精华
26 
帖子
30441 
注册时间
2009-10-5 
最后登录
2022-12-28 

才高八斗

1
发表于 2013-6-5 09:21 |只看该作者 |倒序浏览 |打印
VIRAL HEPATITIS CARJACKING HBV
THERAPY

No cure exists for chronic hepatitis B.
Current antiviral agents effectively
suppress HBV replication, but cannot
eliminate the infection. Now, Ulrike Protzer
and colleagues have demonstrated in a
preclinical model of chronic hepatitis B
that adoptive T-cell therapy is effective
in controlling HBV infection.
A strong T-cell response eradicates HBV
during acute hepatitis B episodes, but
patients with chronic HBV infection lack this
immune response. Protzer and co-workers
therefore developed an immunotherapeutic
approach using the adoptive transfer of
genetically modified HBV-specific T cells.
In brief, CD8+ T cells were genetically
modified to express HBV-specific chimaeric
antigen receptors (CARs), which bind to
HBV surface antigen (HBsAg). These cells
were adoptively transferred by injection into
immune-competent HBV transgenic mice.
The most important findings from
this study were that the HBV-specific
T cells redirected to the liver of
transgenic mice, where they successfully
engrafted and expanded without prior
immunosuppression. So, the HBV-specific
CAR modification enabled recognition
of HBV envelope proteins expressed on
the surface of HBV-infected hepatocytes;
HBV-specific CARs also recognised
different HBV subtypes. The redirected
T cells rapidly and efficiently reduced HBV
replication when compared with controls
(mice that received CD8+ T cells induced
by vaccination or T cells expressing a CAR
with a nonfunctioning signalling domain
or a CAR that does not bind to HBV).
Of note, this approach seems to be
safe, as only transient liver damage was
observed and the animals displayed
no obvious signs of distress, such as
weight loss. Moreover, the high levels of
circulating viral antigens did not inactivate
the adoptively transferred T cells or induce
an uncontrolled immune response.
These findings provide “the proof-of-
concept for further translation of adoptive
T cell therapy for chronic hepatitis B into
the clinics,” write the authors. Indeed, the
researchers hope that this immunotherapy
can be developed for use in patients with
chronic hepatitis B of any HLA-type and in
patients with HBsAg-positive hepatocellular
carcinoma.  Katherine Smith Original article Krebs, K. et al. T cells expressing a chimeric
antigen receptor that binds hepatitis B virus envelop
proteins control virus replication in mice. Gastroenterology
doi:10.1053/j.gastro.2013.04.047 V

Rank: 8Rank: 8

现金
62111 元 
精华
26 
帖子
30441 
注册时间
2009-10-5 
最后登录
2022-12-28 

才高八斗

2
发表于 2013-6-5 09:24 |只看该作者
存在无法治愈的慢性乙型肝炎
目前的抗病毒药剂有效
抑制HBV的复制,但不能
消除感染。现在,乌尔里克Protzer
和他的同事已经证明,在一个
慢性乙型肝炎的临床前模型
过继性T细胞治疗是有效的
控制乙肝病毒感染。
一个强大的T细胞反应根除HBV
在急性B型肝炎发作,但
慢性HBV感染患者缺乏这种
免疫反应。 protzer和同事
因此研制出一种免疫治疗
接近使用的过继转移
转基因的HBV特异性的T细胞。
总之,CD8 + T细胞基因
修改表达HBV特异性嵌合
抗原受体(汽车),结合
乙肝病毒表面抗原(HBsAg)。这些细胞
继转移注入
免疫能力的HBV转基因小鼠。
最重要的发现
本研究中,HBV特异性
重定向到肝脏的T细胞
转基因小鼠,在那里他们成功
嫁接扩大,恕不另行通知。
免疫抑制。因此,HBV特异性
汽车改装启用识别
乙肝包膜蛋白表达
HBV感染的肝细胞的表面;
,HBV特异性汽车也承认
不同的乙肝病毒亚型。重定向
T细胞,迅速和有效地减少HBV
复制时,与对照组相比
(诱导CD8 + T细胞的老鼠接受
通过接种疫苗或T细胞表达一辆车
与非功能性的信号域
或一辆汽车,不绑定到HBV)。
值得注意的是,这种方法似乎是
安全,因为只有短暂的肝功能损害
观察所显示的动物
没有明显的迹象的困扰,如
减肥。此外,高含量的
循环病毒抗原没有灭活
继转移的T细胞或诱导
不受控制的免疫反应。
这些发现提供“证明
概念为进一步翻译收养
T细胞治疗慢性乙型肝炎
诊所,“写作者。事实上,
研究人员希望,这种免疫治疗
可被开发用于患者使用
慢性B型肝炎的任何HLA-型和
HBsAg阳性的肝癌患者
癌。凯瑟琳·史密斯原创文章克雷布斯,K.等。 T细胞表达的嵌合
抗原受体结合乙肝病毒的信封
蛋白质控制病毒复制的作用。胃肠病
DOI:10.1053/j.gastro.2013.04.047 V

Rank: 8Rank: 8

现金
62111 元 
精华
26 
帖子
30441 
注册时间
2009-10-5 
最后登录
2022-12-28 

才高八斗

3
发表于 2013-6-5 09:25 |只看该作者
T Cells Expressing a Chimeric Antigen Receptor That Binds Hepatitis B Virus Envelop Proteins Control Virus Replication in Mice.      Authors                  Krebs K, Böttinger N, Huang LR, Chmielewski M, Arzberger S, Gasteiger G, Jäger C, Schmitt E, Bohne F, Aichler M, Uckert W, Abken H, Heikenwalder M, Knolle P, Protzer U.
   
    Journal   

Gastroenterology. 2013 Apr 29. pii: S0016-5085(13)00684-7. doi: 10.1053/j.gastro.2013.04.047. [Epub ahead of print]

    Affiliation   

Institute of Virology, Technische Universität München / Helmholtz Zentrum München, Trogerstr. 30, 81675 München, Germany.

  
  Abstract

BACKGROUND & AIMS: Antiviral agents suppress hepatitis B virus (HBV) replication but do not clear the infection. A strong effector T-cell response is required to eradicate HBV, but this does not occur in patients with chronic infection. T cells might be directed toward virus-infected cells by expressing HBV-specific receptors, and thereby clear HBV and help to prevent development of liver cancer. In mice, we studied whether redirected T cells can engraft following adoptive transfer, without prior T-cell depletion, and whether the large amounts of circulating viral antigens inactivate the transferred T cells or lead to uncontrolled, immune-mediated damage.

METHODS: CD8(+) T cells were isolated from mice and stimulated using an optimized protocol. Chimeric antigen receptors (CARs) that bind HBV envelope proteins (S-CAR) and activate T cells were expressed on the surface of cells using retroviral vectors. S-CAR-expressing CD8(+) T cells, which carried the marker CD45.1, were injected into CD45.2(+) HBV transgenic mice. We compared these mice with mice that received CD8(+) T cells induced by vaccination, cells that express a CAR without a proper signaling domain, or cells that express a CAR that does not bind HBV proteins (controls).

RESULTS: CD8(+) T cells that expressed HBV-specific CARs recognized different HBV subtypes and were able to engraft and expand in immune-competent HBV transgenic mice. Following adoptive transfer, the S-CAR-expressing T cells localized to and functioned in the liver; they rapidly and efficiently controlled HBV replication, compared with controls, causing only transient liver damage. The large amount of circulating viral antigen did not impair or over-activate the S-CAR grafted T cells.

CONCLUSION: T cells with a CAR specific for HBV envelop proteins localize to the livers of mice to reduce HBV replication, causing only transient liver damage. This immune-cell therapy might be developed for patients with chronic hepatitis B, regardless of their HLA-type.

Copyright © 2013 AGA Institute. Published by Elsevier Inc. All rights reserved.
            PMID      23639914 [PubMed - as supplied by publisher]   
    Full text: Elsevier Science
  

Rank: 8Rank: 8

现金
62111 元 
精华
26 
帖子
30441 
注册时间
2009-10-5 
最后登录
2022-12-28 

才高八斗

4
发表于 2013-6-5 09:26 |只看该作者
T细胞表达嵌合抗原受体结合在小鼠体内的乙肝病毒外型蛋白控制病毒复制。
作者
Arzberger Chmielewski黄立人,ÑBöttinger,克雷布斯K,M,S,Gasteigerĝ,耶格尔Ç,施密特ÉBohne F中号Aichler,W Uckert,ħAbken,中号Heikenwalder,P Knolle,Protzer U。
刊物

胃肠病。 2013年4月29。 PII:S0016-5085(13)00684-7。 DOI:10.1053/j.gastro.2013.04.047。 [EPUB的提前打印]
联系

病毒学研究所,慕尼黑工业大学/亥姆霍兹慕尼黑中心,Trogerstr。 30,81675慕尼黑,德国。
抽象

背景与目的:抗病毒药物抑制B型肝炎病毒(HBV)复制,但不清除感染。需要一个强大的效应T细胞反应,以消除乙肝病毒,但这种情况不会发生在慢性感染患者。向病毒感染的细胞,T细胞可能通过表达HBV特异性受体,从而清除HBV和帮助,以防止发展为肝癌。在小鼠实验中,我们研究了重定向的T细胞是否可以嫁接过继转移后,恕不另行T细胞耗竭,以及是否大量的循环病毒抗原灭活T细胞转移或导致失控,免疫介导损伤。

从小鼠中分离的方法:CD8(+)T细胞刺激使用一个优化的协议。嵌合抗原受体(中亚)结合HBV的包膜蛋白(S-CAR),激活T细胞,使用逆转录病毒载体的细胞的表面上表达。 S-CAR表达CD8(+)T细胞,从而进行标记CD45.1,分别注入CD45.2(+),HBV转基因小鼠。我们比较了这些小鼠与老鼠接受CD8(+),通过接种疫苗,细胞表达一辆车,没有一个适当的信令域,或细胞诱导T细胞表达了车,不具约束力的HBV蛋白(对照组)。

结果:CD8(+)T细胞,表示HBV特异性汽车的认识不同的HBV亚型和得以嫁接和扩大免疫能力的HBV转基因小鼠。本地化的S-CAR表达的T细胞过继转移后,在肝脏中发挥作用,迅速和有效地控制HBV的复制,与对照组相比,造成唯一的一过性肝损伤。循环病毒抗原的大量不损害或过度激活的S-CAR接枝T细胞。

结论:HBV信封蛋白与CAR特定的T细胞本地化的小鼠肝脏,降低HBV的复制,只造成短暂的肝功能损害。此免疫细胞疗法可能会发展为慢性乙型肝炎患者,而不管他们的HLA型。
版权所有©2013 AGA研究所。由Elsevier公司出版保留所有权利。
治疗
23639914 [1  - 由出版商提供]
全文:Elsevier科学

Rank: 8Rank: 8

现金
62111 元 
精华
26 
帖子
30441 
注册时间
2009-10-5 
最后登录
2022-12-28 

才高八斗

5
发表于 2013-6-5 09:28 |只看该作者
[This abstract from the APASL might be the most fundamentally promising of all. Epitope specific T cells can be engineered in large scale and without dangerous genetic manipulations that will attack surface antigen producing liver cells and also produce interferon gamma upon contact with these cells, which could clear neighboring cells from cccDNA.
Studyforhope, Medhelp]

Engineering T Cell Immunity Targeting HBV-expressing Hepatocellular Carcinoma for Clinical Use

Sarene Koh1,2, Noriko Shimasaki3, Rossarin Suwanarsuk4, Laurent Renia4, Adam Gehring1, Matti Sallberg2, Dario Campana3, Antonio Bertoletti1
1Singapore Institute for Clinical Sciences (A*STAR), Singapore, Singapore, 2Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden, 3Department of Paediatrics, National University of Singapore, 4Singapore Immunology Network (A*STAR), Singapore, Singapore

Background/aims: The therapeutic efficacy of tumor-specific T-cell adoptive transfer for hepatocellular carcinoma (HCC) therapy needs to be evaluated before clinical translation. We previously showed that lymphocytes from patients with hepatitis B virus (HBV)-related HCC can be engineered to express HBV-specific T-cell receptors (TCR) through retroviral mediated gene transfer, and were capable of recognizing HCC cells expressing HBV-viral antigens from integrated HBV DNA. Due to potential safety issues that could limit the use of viral vectors in clinical application, we explored a safer and easier alternative strategy to redirect the specificity of T cells using mRNA electroporation.
Methods: Activated T cells were transfected with mRNA encoding HBV s183-191 TCR. HBV-specific TCR expression and function of electroporated cells were tested in vitro and in vivo in a xenograft tumor model produced by injecting HCC cell line constitutively expressing HBV surface antigen and luciferase into NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ mice. The kinetics of tumor development in vivo was measured by bioluminescence imaging.
Results: Approximately 90% of CD8 T cells expressed HBV TCR 24 h post-electroporation, 3 fold higher than what retroviral transduction routinely achieved. Electroporated T cells produced IFN-gamma, TNF-alpha and IL-2 after HBV-specific recognition, with a large contribution from transfected CD4 T cells. Cytokine production capacity, determined by mean fluorescence intensity, and cytotoxic activity was similar to that of retroviral transduced T cells. In vivo experiments showed electroporated T cells could suppress the growth of established tumors in mice.
Conclusions: Messenger RNA electroporation is a powerful tool to introduce TCR into human T lymphocytes with high efficiency. Electroporated cells are fully functional in vitro and in vivo. As stable integration of genes into host genome is avoided, this technology is particularly attractive for clinical translation since it can rapidly generate at low cost, large batches of antigen-specific T cells for immunotherapy.

Rank: 8Rank: 8

现金
62111 元 
精华
26 
帖子
30441 
注册时间
2009-10-5 
最后登录
2022-12-28 

才高八斗

6
发表于 2013-6-5 09:28 |只看该作者
这种抽象的APASL可能是最根本前途的。抗原表位的特异性T细胞可以设计大规模,没有危险的遗传操作,将攻击表面抗原肝细胞也产生γ干扰素接触后,这些细胞中,它可以清除cccDNA的相邻小区。


工程T细胞免疫靶向HBV表达的肝癌临床使用

2,子Shimasaki3,Rossarin Suwanarsuk4,洛朗Renia4,亚当Gehring1,马蒂Sallberg2,达里奥Campana3,Antonio Bertoletti1的Sarene Koh1
1Singapore临床科学研究所(A * STAR),新加坡,新加坡,2,瑞典斯德哥尔摩卡罗林斯卡医学院检验医学,儿科3,新加坡国立大学,4Singapore免疫网络(A * STAR),新加坡,新加坡

背景/目的:肿瘤特异性T细胞过继转移肝细胞癌(HCC)治疗的疗效需要进行评估的临床前翻译。我们以前发现,乙型肝炎病毒(HBV)感染的患者相关的肝癌的淋巴细胞,可以设计表达HBV特异性T细胞受体(TCR),通过逆转录病毒介导的基因转移,能够识别肝癌细胞表达HBV病毒抗原从整合型HBV DNA。由于潜在的安全问题,可能会限制使用的病毒载体在临床应用中,我们探索出了一条更安全,更容易的替代战略,以重定向的特异性T细胞mRNA的电。
方法:活化的T细胞转染与mRNA编码HBV S183-191的TCR。 HBV特异性T细胞受体的表达和功能的电穿孔的细胞在体外和体内进行了测试,所产生的组成型表达HBV表面抗原和荧光素酶到NOD.Cg Prkdcscid Il2rgtm1Wjl/SzJ小鼠肝癌细胞株注入在异种移植肿瘤模型。在体内肿瘤的发展的动力学测定生物发光成像。
结果:约90%的CD8 T细胞表达HBV TCR电后24小时,比逆转录病毒转导经常达到3倍。电穿孔的T细胞产生IFN-γ,TNF-α和IL-2的HBV特异性识别后,从转染的CD4 + T细胞具有大的贡献。细胞因子的产生能力,确定平均荧光强度,和细胞毒活性的逆转录病毒转导的T细胞相似。体内实验表明电穿孔的T细胞可以抑制小鼠建立肿瘤生长。
结论:信使RNA电穿孔是一个功能强大的工具引入TCR进入人体T淋巴细胞高效率。电穿孔的细胞在体外和体内功能齐全。稳定整合到宿主基因组的基因得以避免,这是特别有吸引力的技术,因为它可以迅速地以较低的成本,大批量进行免疫治疗的抗原特异性T细胞产生的临床翻译。

现金
1904 元 
精华
帖子
1665 
注册时间
2011-11-30 
最后登录
2023-5-5 
7
发表于 2013-6-6 15:42 |只看该作者
怎么一下子这么多研究乙肝的项目出现啊?难道是中国人钱多的原因?

现金
1904 元 
精华
帖子
1665 
注册时间
2011-11-30 
最后登录
2023-5-5 
8
发表于 2013-6-6 15:42 |只看该作者
怎么一下子这么多研究乙肝的项目出现啊?难道是中国人钱多的原因?
‹ 上一主题|下一主题
你需要登录后才可以回帖 登录 | 注册

肝胆相照论坛

GMT+8, 2024-5-12 17:14 , Processed in 0.014017 second(s), 11 queries , Gzip On.

Powered by Discuz! X1.5

© 2001-2010 Comsenz Inc.