Tim3

TIM3
  • 文章类型: Letter
    急性髓性白血病(AML)是一种侵袭性和遗传异质性疾病,临床预后较差。难治性AML很常见,复发仍然是一个重大挑战,归因于治疗抗性白血病干细胞(LSCs)的存在,具有自我更新和再生能力。靶向LSC目前是AML长期管理的最有希望的途径。同样,嵌合抗原受体(CAR)-自然杀伤(NK)细胞已成为CAR-T细胞的有希望的替代品,因为它们具有作为现成产品和更安全的临床特征的内在潜力。这里,我们介绍了一种带有TIM3scFv的第三代汽车,CD28,4-1BB,和CD3ζ(CAR-TIM3)进入人NK-92细胞,唯一FDA批准的用于临床试验的NK细胞系。由于其在LSC和正常造血干/祖细胞(HSPC)中的差异表达,选择TIM3作为靶抗原。已建立的CAR-TIM3NK-92细胞有效地靶向TIM3,并显示出对各种原始AML细胞的有效抗肿瘤活性,随后在体外引起白血病克隆生长的减少,而对HSPC的影响最小。CAR-TIM3NK-92细胞在体内显着降低白血病负担,并有趣地抑制AML细胞植入小鼠肝脏和骨髓。令人惊讶的是,我们发现CAR-TIM3NK-92细胞表达相对较低的表面TIM3,导致较低的自相残杀效应。由于TIM3和PD-1是涉及NK细胞功能障碍的免疫检查点,我们进一步测试并发现CAR-TIM3NK-92细胞有利于缓解NK细胞耗竭.我们的发现强调了CAR-TIM3NK细胞在TIM3+AML细胞免疫治疗中的潜在应用。
    Acute myeloid leukemia (AML) is an aggressive and genetically heterogeneous disease with poor clinical outcomes. Refractory AML is common, and relapse remains a major challenge, attributable to the presence of therapy-resistant leukemic stem cells (LSCs), which possess self-renewal and repopulating capability. Targeting LSCs is currently the most promising avenue for long-term management of AML. Likewise, chimeric antigen receptor (CAR)-natural killer (NK) cells have emerged as a promising alternative to CAR-T cells due to their intrinsic potential as off-the-shelf products and safer clinical profiles. Here, we introduced a third-generation CAR harboring TIM3 scFv, CD28, 4-1BB, and CD3ζ (CAR-TIM3) into human NK-92 cells, the only FDA-approved NK cell line for clinical trials. TIM3 was chosen as a target antigen owing to its differential expression in LSCs and normal hematopoietic stem/progenitor cells (HSPCs). The established CAR-TIM3 NK-92 cells effectively targeted TIM3 and displayed potent anti-tumor activity against various primitive AML cells, subsequently causing a reduction in leukemic clonogenic growth in vitro, while having minimal effects on HSPCs. CAR-TIM3 NK-92 cells significantly reduced leukemic burden in vivo and interestingly suppressed the engraftment of AML cells into the mouse liver and bone marrow. Surprisingly, we found that CAR-TIM3 NK-92 cells expressed relatively low surface TIM3, leading to a low fratricidal effect. As TIM3 and PD-1 are immune checkpoints involved in NK cell dysfunction, we further tested and found that CAR-TIM3 NK-92 cells are beneficial for alleviating NK cell exhaustion. Our findings highlight the potential application of CAR-TIM3 NK cells for cellular immunotherapy for TIM3+ AML.
    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
    背景:越来越多的证据表明免疫炎症是慢性阻塞性肺疾病(COPD)进展的关键因素。免疫检查点(IC)是调节免疫细胞功能激活和分化的关键目标,特别是与免疫炎症和T细胞活化和耗竭的调节有关。然而,ICs在COPD中的确切机制尚待了解.
    方法:从基因表达综合(GEO)获得COPD数据集,并使用GEO2R和Limma进行分析以鉴定差异表达基因。然后将LASSO回归应用于筛查与COPD密切相关的IC。最后,基于基因表达谱选择靶基因。基因本体论(GO),免疫浸润分析,和基因集富集分析(GSEA)用于评估IC基因(ICGs)与免疫细胞之间的关系。随后,烟草暴露的小鼠,抗Tim3治疗的小鼠,产生了HAVCR2基因敲除小鼠,流式细胞术用于确认结果。
    结果:通过GSE38974和LASSO回归分析,确定了五个ICG。随后使用GSE20257和GSE76925进行的验证证实了这些发现。基因表达谱强调HAVCR2与COPD具有最强的相关性。通过免疫浸润分析进一步调查,GO,GSEA提示COPD患者HAVCR2与CD8+T细胞之间存在联系。流式细胞术实验证明在暴露于烟草的小鼠的CD8+T细胞中Tim3高表达,促进Tc1和抑制Tc17,从而影响CD8+Tem激活和CD8+Tcm的形成,导致CD8+T细胞内的免疫失衡。
    结论:长期暴露于烟草会上调CD8+T细胞中的Tim3,触发其对Tc1/Tc17的调节作用。敲除HAVCR2进一步上调CD8+Tem的表达,同时抑制CD8+Tcm的表达,这表明Tim3在烟草暴露的情况下在CD8+T细胞的活化和分化中起作用。
    BACKGROUND: There is growing evidence indicating immune inflammation is a key factor in the progression of chronic obstructive pulmonary disease (COPD). Immune checkpoints (ICs) are crucial targets for modulating the functional activation and differentiation of immune cells, particularly in relation to immune inflammation and the regulation of T cell activation and exhaustion. However, the precise mechanisms of ICs in COPD remain understood.
    METHODS: COPD datasets were obtained from the Gene Expression Omnibus (GEO) and analyzed using GEO2R and Limma to identify differentially expressed genes. LASSO regression was then applied to screen ICs closely associated with COPD. Finally, target genes were selected based on gene expression profiles. Gene ontology (GO), immune infiltration analysis, and gene set enrichment analysis (GSEA) were utilized to assess the relationship between IC genes (ICGs) and immune cells. Subsequently, tobacco-exposed mice, anti-Tim3-treated mice, and HAVCR2-knockout mice were generated, with flow cytometry being used to confirm the results.
    RESULTS: Through the analysis of GSE38974 and LASSO regression, five ICGs were identified. Subsequent validation using GSE20257 and GSE76925 confirmed these findings. Gene expression profiling highlighted HAVCR2 as having the strongest correlation with COPD. Further investigation through immune infiltration analysis, GO, and GSEA indicated a link between HAVCR2 and CD8+ T cells in COPD. Flow cytometry experiments demonstrated high Tim3 expression in CD8+ T cells of mice exposed to tobacco, promoting Tc1 and inhibiting Tc17, thus affecting CD8+ Tem activation and CD8+ Tcm formation, leading to an immune imbalance within CD8+ T cells.
    CONCLUSIONS: Prolonged exposure to tobacco upregulates Tim3 in CD8+ T cells, triggering its regulatory effects on Tc1/Tc17. Knocking out HAVCR2 further upregulated the expression of CD8+ Tem while suppressing the expression of CD8+ Tcm, indicating that Tim3 plays a role in the activation and differentiation of CD8+ T cells in the context of tobacco exposure.
    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
    嵌合抗原受体(CAR)T细胞疗法已改变了血液恶性肿瘤的治疗方法。然而,其在实体瘤中的疗效受到免疫抑制性肿瘤微环境的限制,该环境损害了临床环境中的CART细胞抗肿瘤功能.为了克服这一挑战,研究人员研究了抑制特定免疫检查点受体的潜力,包括A2aR(腺苷A2受体)和Tim3(T细胞免疫球蛋白和含粘蛋白结构域的蛋白3),以增强CAR-T细胞功能。在这项研究中,我们在体外和体内评估了基因靶向Tim3和A2a受体对人间皮素特异性CAR-T细胞(MSLN-CAR)抗肿瘤功能的影响.
    使用标准细胞和分子技术产生第二代抗间皮素CART细胞。使用shRNA介导的基因沉默产生A2aR-敲低和/或Tim3-敲低抗间皮素-CART细胞。通过测量细胞因子的产生来评估CART细胞的抗肿瘤功能。扩散,通过与宫颈癌细胞(HeLa细胞系)共培养在体外具有细胞毒性。为了评估制造的CART细胞的体内抗肿瘤功效,在人宫颈癌异种移植模型中监测肿瘤生长和小鼠存活。
    体外实验表明,单独敲除A2aR或与Tim3联合使用可显著提高CAR-T细胞增殖,细胞因子产生,和以抗原特异性方式存在肿瘤细胞的细胞毒性。此外,在人性化的异种移植模型中,双敲低CART细胞和对照CART细胞均能有效控制肿瘤生长。然而,单个敲低CART细胞与小鼠存活率降低有关。
    这些发现强调了伴随基因靶向Tim3和A2a受体以增强CAR-T细胞疗法在实体瘤中的功效的潜力。然而,根据我们观察到的单敲除MSLN-CAR-T细胞治疗的小鼠存活率降低,应谨慎行事。强调需要仔细考虑功效。
    UNASSIGNED: Chimeric antigen receptor (CAR) T cell therapy has transformed the treatment of hematological malignancies. However, its efficacy in solid tumors is limited by the immunosuppressive tumor microenvironment that compromises CAR T cell antitumor function in clinical settings. To overcome this challenge, researchers have investigated the potential of inhibiting specific immune checkpoint receptors, including A2aR (Adenosine A2 Receptor) and Tim3 (T cell immunoglobulin and mucin domain-containing protein 3), to enhance CAR T cell function. In this study, we evaluated the impact of genetic targeting of Tim3 and A2a receptors on the antitumor function of human mesothelin-specific CAR T cells (MSLN-CAR) in vitro and in vivo.
    UNASSIGNED: Second-generation anti-mesothelin CAR T cells were produced using standard cellular and molecular techniques. A2aR-knockdown and/or Tim3- knockdown anti-mesothelin-CAR T cells were generated using shRNA-mediated gene silencing. The antitumor function of CAR T cells was evaluated by measuring cytokine production, proliferation, and cytotoxicity in vitro through coculture with cervical cancer cells (HeLa cell line). To evaluate in vivo antitumor efficacy of manufactured CAR T cells, tumor growth and mouse survival were monitored in a human cervical cancer xenograft model.
    UNASSIGNED: In vitro experiments demonstrated that knockdown of A2aR alone or in combination with Tim3 significantly improved CAR T cell proliferation, cytokine production, and cytotoxicity in presence of tumor cells in an antigen-specific manner. Furthermore, in the humanized xenograft model, both double knockdown CAR T cells and control CAR T cells could effectively control tumor growth. However, single knockdown CAR T cells were associated with reduced survival in mice.
    UNASSIGNED: These findings highlight the potential of concomitant genetic targeting of Tim3 and A2a receptors to augment the efficacy of CAR T cell therapy in solid tumors. Nevertheless, caution should be exercised in light of our observation of decreased survival in mice treated with single knockdown MSLN-CAR T cells, emphasizing the need for careful efficacy considerations.
    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
    免疫检查点抑制剂(ICIs)作为免疫疗法恢复T细胞功能和重新激活抗肿瘤免疫已显示出巨大的希望。美国食品和药物管理局(FDA)批准了第一个免疫检查点抑制剂,ipilimumab,2011年晚期黑色素瘤患者,显著提高生存率。随后,我们检测了其他免疫检查点靶向抗体.目前,七个ICIs,即ipilimumab(抗细胞毒性T淋巴细胞相关蛋白4[CTLA4]),pembrolizumab,纳武单抗(抗程序性细胞死亡蛋白1[PD-1]),阿替珠单抗,阿维鲁单抗,durvalumab,和cemiplimab(抗PD-L1),已被批准用于各种癌症类型。然而,靶向CTLA4或PD-1/程序性死亡-配体1(PD-L1)的抗体的疗效仍不理想.因此,正在进行的研究正在评估下一代ICI,如淋巴细胞活化基因-3(LAG3),T细胞免疫球蛋白和含粘蛋白结构域3(TIM3),和T细胞免疫球蛋白和ITIM结构域(TIGIT)。我们的综述提供了评估这些新型免疫检查点在癌症治疗中的临床试验的摘要。
    Immune checkpoint inhibitors (ICIs) have shown great promise as immunotherapy for restoring T cell function and reactivating anti-tumor immunity. The US Food and Drug Administration (FDA) approved the first immune checkpoint inhibitor, ipilimumab, in 2011 for advanced melanoma patients, leading to significant improvements in survival rates. Subsequently, other immune checkpoint-targeting antibodies were tested. Currently, seven ICIs, namely ipilimumab (anti-cytotoxic T lymphocyte-associated protein 4 [CTLA4]), pembrolizumab, nivolumab (anti-programmed cell death protein 1 [PD-1]), atezolizumab, avelumab, durvalumab, and cemiplimab (anti-PD-L1), have been approved for various cancer types. However, the efficacy of antibodies targeting CTLA4 or PD-1/programmed death-ligand 1 (PD-L1) remains suboptimal. Consequently, ongoing studies are evaluating the next generation of ICIs, such as lymphocyte activation gene-3 (LAG3), T cell immunoglobulin and mucin-domain containing 3 (TIM3), and T cell immunoglobulin and ITIM domain (TIGIT). Our review provides a summary of clinical trials evaluating these novel immune checkpoints in cancer treatment.
    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
    自然杀伤(NK)细胞在重型再生障碍性贫血(SAA)的免疫发病机制中发挥重要的免疫调节作用。我们先前的研究表明,SAA导致NK细胞上T细胞免疫球蛋白粘蛋白3(TIM3)表达降低。在这里,我们研究了表面受体的表达,SAA患者外周血TIM3+NK和TIM3-NK细胞的细胞毒性。
    用FCM检测外周血TIM3+NK和TIM3-NK细胞表面受体和胞浆蛋白的表达。mDC的功能,用FCM测定与TIM3+NK和TIM3-NK细胞共培养后K562细胞的凋亡率。Westren-blot用于检测TIM3+NK和TIM3-NK信号通路蛋白(AKT,P-AKT),并比较两组的功能活性。
    激活受体NKG2D和粒酶B较高,同时抑制受体NKG2A,在SAA患者中,与TIM3+NK细胞相比,CD158a和CD158b在TIM3-NK细胞上较低。在SAA,与TIM3-NK细胞孵育后,mDCs(髓样树突状细胞)上CD80和CD86的表达显着降低。在SAA中与TIM3-NK细胞孵育后,K562细胞的凋亡率(AR)显着增加。SAA中TIM3-NK细胞的信号通路蛋白AKT水平与TIM3+NK细胞相似,TIM3-NK细胞的P-AKT水平和P-AKT/AKT比值明显高于TIM3+NK细胞。
    因此,TIM3对NK细胞有抑制作用,参与SAA的免疫发病机制。TIM3的低表达有助于NK细胞活性的增强,这进而抑制SAA的免疫激活状态并改善疾病状态。我们的研究可能有助于基于TIM3-NK细胞输注治疗SAA的新治疗策略的开发。
    UNASSIGNED: Natural killer (NK) cells play important immunoregulatory roles in the immune pathogenesis of severe aplastic anemia (SAA). Our previous research showed that SAA caused a decrease in T cell immunoglobulin mucin-3 (TIM3) expression on NK cells. Here we investigated the expression of surface receptors, and the cytotoxicity of peripheral TIM3+ NK and TIM3- NK cells in patients with SAA.
    UNASSIGNED: The expressions of surface receptors and cytoplasmic protein of TIM3+ NK and TIM3- NK cells from peripheral blood were detected by FCM. The functions of mDCs, and apoptosis rate of K562 cells after co-culture with TIM3+ NK and TIM3- NK cells were maesured by FCM. Westren-blot was used to detect the changes of TIM3+ NK and TIM3- NK signaling pathway proteins (AKT, P-AKT) and compare the functional activity of the two groups.
    UNASSIGNED: Activating receptors NKG2D and Granzyme B were higher, while inhibiting receptors NKG2A, CD158a and CD158b were lower on TIM3- NK cells compared with TIM3+ NK cells in patients with SAA. In SAA, the expression of CD80 and CD86 on mDCs (Myeloid dendritic cells) was significantly decreased after incubation with TIM3- NK cells. The apoptosis rate (AR) of K562 cells was significantly increased after being incubated with TIM3- NK cells in SAA. The level of signal pathway protein AKT of TIM3- NK cells in SAA was similar to that of TIM3+ NK cells, and the levels of P-AKT and P-AKT/AKT ratio of TIM3- NK cells were significantly higher than those of TIM3+ NK cells.
    UNASSIGNED: Therefore, TIM3 exerts its inhibitory effect on NK cells and participates in the immune pathogenesis of SAA. Low expression of TIM3 contributes to the enhancement of NK cell activity which in turn inhibits the immune activation state of SAA and improves the disease state. Our research may aid the development of new therapeutic strategies based on TIM3-NK cells infusion for the treatment of SAA.
    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
    免疫检查点分子在启动过程中起着关键作用,regulation,和终止免疫反应。肿瘤细胞利用这些检查点来抑制免疫细胞功能,促进免疫逃避。临床干预通过阻止免疫检查点与其配体的结合来针对这种机制,从而恢复免疫细胞的抗肿瘤能力。值得注意的是,以免疫检查点抑制剂为中心的疗法,特别是PD-1/PD-L1和CTLA-4阻断抗体,已经证明了显著的临床前景。然而,相当一部分患者仍然遇到疗效欠佳并产生耐药性。近年来,研究新型免疫检查点分子如TIM3、LAG3、TIGIT、NKG2D,和CD47,以及它们各自的配体。控制免疫检查点分子的过程,从它们的合成到跨膜部署,与配体的相互作用,以及最终的退化,与翻译后修饰密切相关。这些修饰包括糖基化,磷酸化,泛素化,Neddylation,SUMOylation,棕榈酰化,和胞外域脱落。此讨论继续提供了几种新型免疫检查点及其配体的结构特征的简要概述。此外,它概述了由翻译后修饰控制的调节机制,提供他们在免疫检查点阻断的潜在临床应用的见解。
    Immune checkpoint molecules play a pivotal role in the initiation, regulation, and termination of immune responses. Tumor cells exploit these checkpoints to dampen immune cell function, facilitating immune evasion. Clinical interventions target this mechanism by obstructing the binding of immune checkpoints to their ligands, thereby restoring the anti-tumor capabilities of immune cells. Notably, therapies centered on immune checkpoint inhibitors, particularly PD-1/PD-L1 and CTLA-4 blocking antibodies, have demonstrated significant clinical promise. However, a considerable portion of patients still encounter suboptimal efficacy and develop resistance. Recent years have witnessed an exponential surge in preclinical and clinical trials investigating novel immune checkpoint molecules such as TIM3, LAG3, TIGIT, NKG2D, and CD47, along with their respective ligands. The processes governing immune checkpoint molecules, from their synthesis to transmembrane deployment, interaction with ligands, and eventual degradation, are intricately tied to post-translational modifications. These modifications encompass glycosylation, phosphorylation, ubiquitination, neddylation, SUMOylation, palmitoylation, and ectodomain shedding. This discussion proceeds to provide a concise overview of the structural characteristics of several novel immune checkpoints and their ligands. Additionally, it outlines the regulatory mechanisms governed by post-translational modifications, offering insights into their potential clinical applications in immune checkpoint blockade.
    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
    T细胞免疫球蛋白和粘蛋白结构域-3(TIM3;HAVCR2)是对T细胞应答施加负调节控制的跨膜蛋白。研究表明,癌症患者的肿瘤浸润淋巴细胞(TIL)中的TIM3表达上调。在这次调查中,通过杂交瘤技术产生了一系列靶向TIM3的单克隆抗体。其中,C23表现出良好的生物学特性。要启用特定绑定,我们通过N-溴代琥珀酰亚胺(NBS)介导的单克隆抗体C23标记开发了124I/125I-C23放射性示踪剂。使用293T-TIM3细胞系评估结合亲和力和特异性,其过度表达TIM3和亲本293T细胞。此外,在HEK293TIM3异种移植模型和4T1(小鼠乳腺癌细胞)和CT26(小鼠结肠癌细胞)的同种异体移植模型中检查了124I/125I-C23的生物分布和体内成像。在各模型小鼠中静脉内施用3.7-7.4MBq124I-C23后,以4、24、48、72和/或96小时的间隔进行微-PET/CT成像。此外,在解剖的肿瘤器官中进行TIM3表达的免疫组织化学(IHC)染色,以及对程序性死亡1(PD1)的相应表达的评估,肿瘤中的CD3和CD8。C23单克隆抗体(mAb)以23.28nM的解离常数与TIM3蛋白特异性结合。成功制备了124I-C23和125I-C23放射性示踪剂,标记产率为83.59±0.35%和92.35±0.20%,分别,放射化学纯度超过95.00%。稳定性结果表明,124I/125I-C23在磷酸盐缓冲液(PBS)和5%人血清白蛋白(HSA)中的放射化学纯度在96h后仍>80%。293T-TIM3细胞对125I-C23的摄取为2.80±0.12%,显著高于293T细胞(1.08±0.08%),在阻断组中,293T-TIM3细胞对125I-C23的摄取在60和120分钟被显著阻断。体内药代动力学分析显示,125I-C23的消除时间为14.62h,分布时间为0.4672h。Micro-PET/CT成像显示293T-TIM3模型中124I-C23探针摄取与阴性对照组和阻断组明显不同。在人性化小鼠模型中,124I-C23探针在4T1和CT26模型中具有明显的特异性摄取,在24h时在肿瘤组织中具有最大摄取(在4T1和CT26人源化TIM3小鼠肿瘤模型中SUVmax(最大标准化摄取值):0.59±0.01和0.76±0.02,分别)。来自这些小鼠模型的肿瘤组织的免疫组织化学显示出相当的TIM3表达。在表达TIM3的肿瘤组织中也观察到CD3和CD8细胞以及PD-1表达。TIM3靶向抗体C23显示出良好的亲和力和特异性。124I/125I-C23探针在体外和体内对TIM3具有明显的靶向特异性。我们的结果表明,124I/125I-C23是TIM3成像的有前途的示踪剂,可能在监测免疫检查点药物疗效方面具有巨大潜力。
    T cell immunoglobulin and mucin domain-3 (TIM3; HAVCR2) is a transmembrane protein that exerts negative regulatory control over T cell responses. Studies have demonstrated an upregulation of TIM3 expression in tumor-infiltrating lymphocytes (TILs) in cancer patients. In this investigation, a series of monoclonal antibodies targeting TIM3 were produced by hybridoma technology. Among them, C23 exhibited favorable biological properties. To enable specific binding, we developed a 124I/125I-C23 radio-tracer via N-bromosuccinimide (NBS)-mediated labeling of the monoclonal antibody C23. Binding affinity and specificity were assessed using the 293T-TIM3 cell line, which overexpresses TIM3, and the parent 293T cells. Furthermore, biodistribution and in vivo imaging of 124I/125I-C23 were examined in HEK293TIM3 xenograft models and allograft models of 4T1 (mouse breast cancer cells) and CT26 (mouse colon cancer cells). Micro-PET/CT imaging was conducted at intervals of 4, 24, 48, 72, and/or 96 h post intravenous administration of 3.7-7.4 MBq 124I-C23 in the respective model mice. Additionally, immunohistochemistry (IHC) staining of TIM3 expression in dissected tumor organs was performed, along with an assessment of the corresponding expression of Programmed Death 1 (PD1), CD3, and CD8 in the tumors. The C23 monoclonal antibody (mAb) specifically binds to TIM3 protein with a dissociation constant of 23.28 nM. The 124I-C23 and 125I-C23 radio-tracer were successfully prepared with a labeling yield of 83.59 ± 0.35% and 92.35 ± 0.20%, respectively, and over 95.00% radiochemical purity. Stability results indicated that the radiochemical purity of 124I/125I-C23 in phosphate-buffered saline (PBS) and 5% human serum albumin (HSA) was still >80% after 96 h. 125I-C23 uptake in 293T-TIM3 cells was 2.80 ± 0.12%, which was significantly higher than that in 293T cells (1.08 ± 0.08%), and 125I-C23 uptake by 293T-TIM3 cells was significantly blocked at 60 and 120 min in the blocking groups. Pharmacokinetics analysis in vivo revealed an elimination time of 14.62 h and a distribution time of 0.4672 h for 125I-C23. Micro-PET/CT imaging showed that the 124I-C23 probe uptake in the 293T-TIM3 model significantly differed from that of the negative control group and blocking group. In the humanized mouse model, the 124I-C23 probe had obvious specific uptake in the 4T1 and CT26 models and maximum uptake at 24 h in tumor tissues (SUVmax (the maximum standardized uptake value) in 4T1 and CT26 humanized TIM3 murine tumor models: 0.59 ± 0.01 and 0.76 ± 0.02, respectively). Immunohistochemistry of tumor tissues from these mouse models showed comparable TIM3 expression. CD3 and CD8 cells and PD-1 expression were also observed in TIM3-expressing tumor tissues. The TIM3-targeting antibody C23 showed good affinity and specificity. The 124I/125I-C23 probe has obvious targeting specificity for TIM3 in vitro and in vivo. Our results suggest that 124I/125I-C23 is a promising tracer for TIM3 imaging and may have great potential in monitoring immune checkpoint drug efficacy.
    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:探索ICB的预测性生物标志物和治疗策略已成为临床实践中的迫切需要。越来越多的证据表明,ARID1A缺乏可能在各种肿瘤的肿瘤环境中发挥关键作用,并可能用作免疫治疗结果的泛癌症生物标志物。本研究旨在探讨ARID1A缺陷在乙型肝炎病毒(HBV)相关肝细胞癌(HBV-HCC)中的免疫调节作用及其潜在的免疫治疗意义。
    方法:在目前的研究中,我们使用生物信息学方法和临床前实验进行了综合分析,以评估ARID1A对生物学行为的调节作用,和乙型肝炎病毒(HBV)相关肝细胞癌(HBV-HCC)的免疫景观。TCGA-LIHC共425例HBV相关肝细胞癌患者,AMC和CHCC-HBV队列纳入生物信息学分析。HBV-HCC标本和ARID1A缺乏细胞模型的免疫组织化学染色用于验证分析结果。
    结果:我们的结果表明,ARID1A缺乏促进了肿瘤的增殖和转移。更重要的是,ARID1A缺乏HBV-HCC与较高的TMB相关,提高免疫活性,和免疫检查点蛋白的上调表达,尤其是HBV-HCC中的TIM-3。Further,在ARID1A敲除的HBV阳性细胞系中,TIM-3的配体Galectin-9的表达升高。
    结论:总而言之,我们已经表明,ARID1A缺乏与HBV-HCC中更活跃的免疫特征和更高的免疫检查点表达相关。此外,本研究为探索ARID1A在对免疫治疗有反应的HBV-HCC患者中的预测作用的可能性提供了见解。
    BACKGROUND: Exploring predictive biomarkers and therapeutic strategies of ICBs has become an urgent need in clinical practice. Increasing evidence has shown that ARID1A deficiency might play a critical role in sculpting tumor environments in various tumors and might be used as pan-cancer biomarkers for immunotherapy outcomes. The current study aims to explored the immune-modulating role of ARID1A deficiency in Hepatitis B virus (HBV) related hepatocellular carcinoma (HBV-HCC) and its potential immunotherapeutic implications.
    METHODS: In the current study, we performed a comprehensive analysis using bioinformatics approaches and pre-clinical experiments to evaluate the ARID1A regulatory role on the biological behavior, and immune landscape of Hepatitis B virus (HBV) related hepatocellular carcinoma (HBV-HCC). A total of 425 HBV-related hepatocellular carcinoma patients from TCGA-LIHC, AMC and CHCC-HBV cohort were enrolled in bioinformatics analysis. Immunohistochemical staining of HBV-HCC specimens and ARID1A deficiency cellular models were used to validate the results of the analysis.
    RESULTS: Our results have shown that ARID1A deficiency promoted tumor proliferation and metastasis. More importantly, ARID1A deficiency in HBV-HCC was associated with the higher TMB, elevated immune activity, and up-regulated expression of immune checkpoint proteins, especially TIM-3 in HBV-HCC. Further, the expression of Galectin-9, which is the ligand of TIM-3, was elevated in the ARID1A knockout HBV positive cell line.
    CONCLUSIONS: To conclude, we have shown that the ARID1A deficiency was correlated with more active immune signatures and higher expression of immune checkpoints in HBV-HCC. Additionally, the present study provides insights to explore the possibility of the predictive role of ARID1A in HBV-HCC patients responsive to immunotherapy.
    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
    背景:急性髓性白血病(AML)是一种克隆性恶性疾病,起源于少量的白血病起始细胞或白血病干细胞(LSCs)-亚群也是复发/难治性AML的根本原因。嵌合抗原受体(CAR)-T细胞疗法已被证明在对抗某些血液系统恶性肿瘤方面是成功的。但是有几个障碍限制了它的广泛应用。CAR-自然杀伤(NK)细胞不携带诱导移植物抗宿主病(GvHD)的风险,经常与同种异体T细胞相关,从而克服耗时,自体细胞制造,并且比CAR-T细胞具有相对更安全的临床特征。本研究旨在从用第三代抗TIM3CAR工程化的克隆主诱导多能干细胞中产生靶向LSCs的抗TIM3CAR-NK细胞。
    方法:克隆母脐血NK来源的诱导多能干细胞(iPSC)系,MUSIi013-A,用作起始细胞,用于工程化带有TIM3scFv片段的抗TIM3CAR(克隆TSR-022),CD28,4-1BB,和CD3ζ信号(CAR-TIM3)。将建立的CAR-TIM3iPSC在无血清和无饲养细胞条件下进一步分化为功能性CAR-TIM3NK细胞,并测试其针对各种TIM3阳性AML细胞的抗肿瘤活性。
    结果:我们成功建立了CAR-TIM3iPSCs的单细胞克隆,通过基因组DNA测序以及抗体和抗原特异性检测验证。我们进行了彻底的iPSC表征以确认其保留的多能性和分化能力。建立的CAR-TIM3iPSC可以分化为CAR-TIM3NK样细胞,与来自亲本iPSC的野生型(WT)NK样细胞相比,进一步证明其对TIM3阳性AML细胞的抗肿瘤活性增强,对TIM3阴性细胞的影响最小。
    结论:用CAR工程化的iPSC,包括本文建立的单细胞克隆CAR-TIM3iPSC,是用于产生现成的CAR-NK细胞以及其他CAR免疫细胞的潜在替代细胞来源。功能性CAR-TIM3NK细胞在无血清和无饲养条件下分化的可行性支持良好生产规范(GMP)兼容方案可进一步建立用于未来临床应用。
    BACKGROUND: Acute myeloid leukemia (AML) is a clonal malignant disorder which originates from a small number of leukemia-initiating cells or leukemic stem cells (LSCs)-the subpopulation that is also the root cause of relapsed/refractory AML. Chimeric antigen receptor (CAR)-T cell therapy has proved successful at combating certain hematologic malignancies, but has several hurdles that limit its widespread applications. CAR-natural killer (NK) cells do not carry the risk of inducing graft-versus-host disease (GvHD) frequently associated with allogeneic T cells, thereby overcoming time-consuming, autologous cell manufacturing, and have relatively safer clinical profiles than CAR-T cells. The present study aimed to generate anti-TIM3 CAR-NK cells targeting LSCs from a clonal master induced pluripotent stem cells engineered with the third-generation anti-TIM3 CAR.
    METHODS: A clonal master umbilical cord blood NK-derived induced pluripotent stem cell (iPSC) line, MUSIi013-A, was used as a starting cells for engineering of an anti-TIM3 CAR harboring TIM3 scFv fragment (clone TSR-022), CD28, 4-1BB, and CD3ζ signaling (CAR-TIM3). The established CAR-TIM3 iPSCs were further differentiated under serum- and feeder-free conditions into functional CAR-TIM3 NK cells and tested for its anti-tumor activity against various TIM3-positive AML cells.
    RESULTS: We successfully established a single-cell clone of CAR-TIM3 iPSCs, as validated by genomic DNA sequencing as well as antibody and antigen-specific detection. We performed thorough iPSC characterization to confirm its retained pluripotency and differentiation capacity. The established CAR-TIM3 iPSCs can be differentiated into CAR-TIM3 NK-like cells, which were further proven to have enhanced anti-tumor activity against TIM3-positive AML cells with minimal effect on TIM3-negative cells when compared with wild-type (WT) NK-like cells from parental iPSCs.
    CONCLUSIONS: iPSCs engineered with CARs, including the established single-cell clone CAR-TIM3 iPSCs herein, are potential alternative cell source for generating off-the-shelf CAR-NK cells as well as other CAR-immune cells. The feasibility of differentiation of functional CAR-TIM3 NK cells under serum- and feeder-free conditions support that Good Manufacturing Practice (GMP)-compliant protocols can be further established for future clinical applications.
    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
    肝细胞癌(HCC)是世界上癌症相关死亡的第二常见原因。超过一半的肝癌患者存在晚期,而高度活跃的全身治疗对于改善预后至关重要.基于免疫检查点抑制剂(ICI)的疗法已成为晚期HCC的新型治疗选择。由于原发性耐药性或获得性耐药性,只有三分之一的患者使用基于ICI的疗法实现了客观反应。肝肿瘤微环境是天然免疫抑制的,和细胞信号传导途径中的特定突变允许肿瘤逃避免疫反应。接下来,肿瘤组织或循环肿瘤DNA的基因测序可以描述基于ICI的治疗的耐药机制,并为新型联合治疗提供理论基础.在这次审查中,我们讨论了导致批准基于ICI的晚期HCC治疗方案的关键临床试验的结果,并总结了正在进行的临床试验.我们回顾了ICIs的抗性机制,并讨论了如何基于肿瘤生物标志物和基因组改变的新兴研究来优化免疫疗法。
    Hepatocellular carcinoma (HCC) is the second most common cause of cancer-related deaths in the world. More than half of patients with HCC present with advanced stage, and highly active systemic therapies are crucial for improving outcomes. Immune checkpoint inhibitor (ICI)-based therapies have emerged as novel therapy options for advanced HCC. Only one third of patients achieve an objective response with ICI-based therapies due to primary resistance or acquired resistance. The liver tumor microenvironment is naturally immunosuppressive, and specific mutations in cell signaling pathways allow the tumor to evade the immune response. Next, gene sequencing of the tumor tissue or circulating tumor DNA may delineate resistance mechanisms to ICI-based therapy and provide a rationale for novel combination therapies. In this review, we discuss the results of key clinical trials that have led to approval of ICI-based therapy options in advanced HCC and summarize the ongoing clinical trials. We review resistance mechanisms to ICIs and discuss how immunotherapies may be optimized based on the emerging research of tumor biomarkers and genomic alterations.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号