Chimeric antigen receptor (CAR)-T cell

  • 文章类型: Journal Article
    靶抗原对于开发嵌合抗原受体(CAR)-T细胞至关重要,但是它们在卵巢癌中的应用是有限的。这项研究旨在确定潜在的基因作为卵巢癌的CAR-T细胞抗原候选物。对来自从GEO数据集获得的四个数据集的卵巢癌样品进行差异基因表达分析。功能注释,途径分析,蛋白质定位,使用各种数据集和工具进行基因表达分析。还进行了致癌性分析和网络分析。总的来说,在卵巢癌样本中鉴定了153个差异表达基因,与60个差异表达的基因表达的质膜蛋白质适合CAR-T细胞抗原。其中,预测21种血浆膜蛋白是卵巢癌的癌基因,九种蛋白质在网络中起着至关重要的作用。在卵巢癌的致癌途径中确定的关键基因包括MUC1,CXCR4,EPCAM,RACGAP1,UBE2C,PRAME,SORT1,JUP,和CLDN3,表明它们是卵巢癌CAR-T细胞治疗的推荐抗原。这项研究揭示了卵巢癌免疫治疗的潜在靶标。
    Target antigens are crucial for developing chimeric antigen receptor (CAR)-T cells, but their application to ovarian cancers is limited. This study aimed to identify potential genes as CAR-T-cell antigen candidates for ovarian cancers. A differential gene expression analysis was performed on ovarian cancer samples from four datasets obtained from the GEO datasets. Functional annotation, pathway analysis, protein localization, and gene expression analysis were conducted using various datasets and tools. An oncogenicity analysis and network analysis were also performed. In total, 153 differentially expressed genes were identified in ovarian cancer samples, with 60 differentially expressed genes expressing plasma membrane proteins suitable for CAR-T-cell antigens. Among them, 21 plasma membrane proteins were predicted to be oncogenes in ovarian cancers, with nine proteins playing crucial roles in the network. Key genes identified in the oncogenic pathways of ovarian cancers included MUC1, CXCR4, EPCAM, RACGAP1, UBE2C, PRAME, SORT1, JUP, and CLDN3, suggesting them as recommended antigens for CAR-T-cell therapy for ovarian cancers. This study sheds light on potential targets for immunotherapy in ovarian cancers.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    包含在嵌合抗原受体(CAR)分子中的CD28和4-1BB共刺激内域在促进CAR-T细胞的持续抗肿瘤活性中起关键作用。然而,与CAR-T细胞中CD28或4-1BB的异位和组成性展示相关的分子事件仅得到部分研究.在目前的研究中,我们证明,在不存在CAR强直信号的情况下,4-1BB掺入CAR导致细胞簇形成和细胞死亡,表现为细胞凋亡和坏死.机制研究表明,4-1BB以TRAF依赖性方式将A20隔离到细胞膜上,导致A20功能缺乏,进而导致NF-κB过度活跃。通过ICAM-1过表达的细胞聚集,和细胞死亡,包括通过RIPK1/RIPK3/MLKL途径的坏死。通过过表达A20或通过删除4-1BB拯救细胞簇形成和细胞死亡的TRAF结合基序从4-1BB释放A20而获得的遗传调制,并增强4-1BB共刺激的CAR-T细胞的抗肿瘤能力。
    CD28 and 4-1BB costimulatory endodomains included in chimeric antigen receptor (CAR) molecules play a critical role in promoting sustained antitumor activity of CAR-T cells. However, the molecular events associated with the ectopic and constitutive display of either CD28 or 4-1BB in CAR-T cells have been only partially explored. In the current study, we demonstrated that 4-1BB incorporated within the CAR leads to cell cluster formation and cell death in the forms of both apoptosis and necroptosis in the absence of CAR tonic signaling. Mechanistic studies illustrate that 4-1BB sequesters A20 to the cell membrane in a TRAF-dependent manner causing A20 functional deficiency that in turn leads to NF-κB hyperactivity, cell aggregation via ICAM-1 overexpression, and cell death including necroptosis via RIPK1/RIPK3/MLKL pathway. Genetic modulations obtained by either overexpressing A20 or releasing A20 from 4-1BB by deleting the TRAF-binding motifs of 4-1BB rescue cell cluster formation and cell death and enhance the antitumor ability of 4-1BB-costimulated CAR-T cells.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    心血管疾病(CVDs)是全球非传染性疾病死亡的主要原因。目前的心脏再生治疗具有局限性并且可能导致不良反应。因此,需要创新技术来解决这些缺点。信使RNA(mRNA)由于其在编码治疗性蛋白质和靶向“不可用”条件方面的多功能性而成为有前途的治疗剂。它提供低毒性,转染效率高,和控制蛋白质生产,没有基因组插入或诱变的风险。然而,mRNA面临的挑战,如免疫原性,不稳定性,细胞进入和内体逃逸的困难,阻碍其临床应用。为了克服这些障碍,脂质纳米颗粒(LNP),特别是在COVID-19疫苗中使用,具有巨大的潜力来提供用于CVD的mRNA疗法。这篇综述强调了mRNA-LNP治疗CVD的最新进展,包括心肌梗死(MI),心力衰竭(HF),和高胆固醇血症.此外,探索了LNP介导的mRNA递送用于CAR-T细胞治疗和CVDs中的CRISPR/Cas基因组编辑以及相关的临床试验。为了提高效率,安全,和mRNA-LNP的临床翻译,RNA结构设计中的人工智能(AGILE平台)等先进技术,并优化LNP配方。我们得出的结论是,促进mRNA-LNP的肝外递送和靶向器官嗜性的策略(SORT,资产,SMRT,和条形码LNP)在加速CVD治疗中mRNA-LNP的开发和翻译方面具有广阔的前景。
    Cardiovascular diseases (CVDs) are the leading cause of global mortality among non-communicable diseases. Current cardiac regeneration treatments have limitations and may lead to adverse reactions. Hence, innovative technologies are needed to address these shortcomings. Messenger RNA (mRNA) emerges as a promising therapeutic agent due to its versatility in encoding therapeutic proteins and targeting \"undruggable\" conditions. It offers low toxicity, high transfection efficiency, and controlled protein production without genome insertion or mutagenesis risk. However, mRNA faces challenges such as immunogenicity, instability, and difficulty in cellular entry and endosomal escape, hindering its clinical application. To overcome these hurdles, lipid nanoparticles (LNPs), notably used in COVID-19 vaccines, have a great potential to deliver mRNA therapeutics for CVDs. This review highlights recent progress in mRNA-LNP therapies for CVDs, including Myocardial Infarction (MI), Heart Failure (HF), and hypercholesterolemia. In addition, LNP-mediated mRNA delivery for CAR T-cell therapy and CRISPR/Cas genome editing in CVDs and the related clinical trials are explored. To enhance the efficiency, safety, and clinical translation of mRNA-LNPs, advanced technologies like artificial intelligence (AGILE platform) in RNA structure design, and optimization of LNP formulation could be integrated. We conclude that the strategies to facilitate the extra-hepatic delivery and targeted organ tropism of mRNA-LNPs (SORT, ASSET, SMRT, and barcoded LNPs) hold great prospects to accelerate the development and translation of mRNA-LNPs in CVD treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    背景:尽管现有治疗方法在管理自身免疫性风湿性疾病(ARD)方面取得了进展,许多患者仍然遇到挑战,如反应不足,难以维持缓解,和副作用。嵌合抗原受体(CAR)T细胞疗法,最初是为癌症开发的,现在已经成为难治性ARD病例的一个有希望的选择。
    方法:进行了文献检索,以撰写叙述性综述,探索当前的证据,潜在安全,局限性,潜在的修改,以及CAR-T细胞在ARD中的未来方向。
    结果:CAR-T细胞疗法已用于难治性ARDs患者,包括系统性红斑狼疮,抗合成酶综合征,和系统性硬化症,表现出显著的改善。值得注意的反应包括临床症状增强,血清自身抗体滴度降低,和疾病活动的持续缓解。使用动物和人类样品的临床前和体外研究也支持了CAR-T细胞对抗抗中性粒细胞胞质抗体相关血管炎和类风湿性关节炎的功效并阐述了其潜在机制。在谨慎监测不良事件的同时,如细胞因子释放综合征,是至关重要的,该疗法似乎具有很高的耐受性。然而,挑战依然存在,包括成本,由于潜在的CAR-T细胞耗尽,和制造复杂性,敦促开发创新解决方案,以进一步增强ARD中CAR-T细胞疗法的可及性。
    结论:CAR-T细胞疗法治疗难治性ARDs具有很高的有效性。虽然目前没有重大警告信号的报告,在治疗效果和安全性之间取得平衡对于采用CAR-T细胞疗法治疗ARD至关重要.此外,技术进步具有巨大的潜力,可以增强对患者的治疗效果,从而确保患者更安全、更有效的疾病控制。
    BACKGROUND: Despite advancements in managing autoimmune rheumatic diseases (ARDs) with existing treatments, many patients still encounter challenges such as inadequate responses, difficulty in maintaining remission, and side effects. Chimeric Antigen Receptor (CAR) T-cell therapy, originally developed for cancer, has now emerged as a promising option for cases of refractory ARDs.
    METHODS: A search of the literature was conducted to compose a narrative review exploring the current evidence, potential safety, limitations, potential modifications, and future directions of CAR-T cells in ARDs.
    RESULTS: CAR-T cell therapy has been administered to patients with refractory ARDs, including systemic lupus erythematosus, antisynthetase syndrome, and systemic sclerosis, demonstrating significant improvement. Notable responses include enhanced clinical symptoms, reduced serum autoantibody titers, and sustained remissions in disease activity. Preclinical and in vitro studies using both animal and human samples also support the efficacy and elaborate on potential mechanisms of CAR-T cells against antineutrophil cytoplasmic antibody-associated vasculitis and rheumatoid arthritis. While cautious monitoring of adverse events, such as cytokine release syndrome, is crucial, the therapy appears to be highly tolerable. Nevertheless, challenges persist, including cost, durability due to potential CAR-T cell exhaustion, and manufacturing complexities, urging the development of innovative solutions to further enhance CAR-T cell therapy accessibility in ARDs.
    CONCLUSIONS: CAR-T cell therapy for refractory ARDs has demonstrated high effectiveness. While no significant warning signs are currently reported, achieving a balance between therapeutic efficacy and safety is vital in adapting CAR-T cell therapy for ARDs. Moreover, there is significant potential for technological advancements to enhance the delivery of this treatment to patients, thereby ensuring safer and more effective disease control for patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    Besides traditional treatment strategies, including surgery, radiotherapy, and chemotherapy for lung cancer as the leading cause of cancer incidence and death, immunotherapy has also emerged as a new treatment strategy. The goal of immunotherapy is to stimulate the immune system responses against cancer, using various approaches such as therapeutic vaccines, monoclonal antibodies, immune checkpoint inhibitors, and T-cell therapy. Chimeric antigen receptor (CAR)-T cells, one of the most popular cancer immunotherapy approaches in the last decade, are genetically engineered T-cells to redirect patients\' immune responses to recognize and eliminate tumor-associated antigens (TAA)-expressing tumor cells. CAR-T cell therapy provides promising benefits in lung tumors. In this review, we summarize different immunotherapy approaches for lung cancer, the structure of CAR-T cells, currently undergoing CARs in clinical trials, and various TAAs are being investigated as potential targets in designing CAR-T cells for lung cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Editorial
    暂无摘要。
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    T细胞非霍奇金淋巴瘤(T-NHL)是一种罕见且异质的淋巴系统肿瘤。除了一些相对惰性的实体,T-NHL通常是侵略性的,治疗抗性,并与预后不良有关。对于复发或难治性疾病的患者,具有已证实的临床益处的选择相对较少。免疫疗法已成为治疗血液恶性肿瘤患者的有希望的治疗方法。肿瘤抗原的鉴定提供了大量潜在的靶标。因此,几种单克隆抗体(阿仑单抗,SGN-30,本妥昔单抗vedotin,和mogamulizumab),针对肿瘤抗原,已在T-NHL的不同亚型中进行了研究。除了靶向参与癌细胞生理学的抗原,抗体可以刺激免疫效应功能或抵消免疫抑制机制。针对CD30和免疫检查点抑制剂的嵌合抗原受体(CAR)-T细胞目前正在临床试验中进行研究。在这次审查中,我们总结了目前可用的T-NHL免疫治疗的临床证据,关注使用第一代单克隆抗体的临床试验结果,新的免疫治疗剂,免疫检查点抑制剂,和CAR-T细胞疗法。
    T cell non-Hodgkin lymphoma (T-NHL) is a rare and heterogeneous group of neoplasms of the lymphoid system. With the exception of a few relatively indolent entities, T-NHL is typically aggressive, treatment resistant, and associated with poor prognosis. Relatively few options with proven clinical benefit are available for patients with relapsed or refractory disease. Immunotherapy has emerged as a promising treatment for the management of patients with hematological malignancies. The identification of tumor antigens has provided a large number of potential targets. Therefore, several monoclonal antibodies (alemtuzumab, SGN-30, brentuximab vedotin, and mogamulizumab), directed against tumor antigens, have been investigated in different subtypes of T-NHL. In addition to targeting antigens involved in cancer cell physiology, antibodies can stimulate immune effector functions or counteract immunosuppressive mechanisms. Chimeric antigen receptor (CAR)-T cells directed against CD30 and immune checkpoint inhibitors are currently being investigated in clinical trials. In this review, we summarize the currently available clinical evidence for immunotherapy in T-NHL, focusing on the results of clinical trials using first generation monoclonal antibodies, new immunotherapeutic agents, immune checkpoint inhibitors, and CAR-T cell therapies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号