chimeric antigen receptor (CAR)

嵌合抗原受体 (CAR)
  • 文章类型: Journal Article
    表观遗传机制涉及几种细胞功能,它们在免疫系统中的作用至关重要。组蛋白脱乙酰酶(HDAC)是调节和催化脱乙酰过程的一组重要酶。HDAC已被证明是改善免疫疗法功效的有益靶标。HDAC11是一种参与T细胞功能负调节的酶。这里,我们研究了在CAR-T细胞中使用RNA干扰下调HDAC11以改善针对前列腺癌的免疫治疗结果的潜力.我们设计并测试了靶向HDAC11的四种不同的短发夹RNA(shRNA)序列,以鉴定对后续分析最有效的一种。HDAC11缺陷型CAR-T细胞(shD-NKG2D-CAR-T)对前列腺癌细胞系表现出比野生型CAR-T细胞更好的细胞毒性。这种效应归因于增强的激活,脱粒,shD-NKG2D-CAR-T与前列腺癌细胞系共培养时的细胞因子释放能力。我们的研究结果表明,HDAC11干扰显着增强CAR-T细胞增殖,减少耗竭标志物PD-1和TIM3,促进T中枢记忆中医群体的形成。对潜在分子机制的进一步探索揭示了转录因子Eomes的表达增加,提供对CAR-T细胞分化调控的见解。最后,shD-NKG2D-CAR-T细胞提供了有效的肿瘤控制,导致与野生型对应物相比,荷瘤小鼠的体内存活率提高.目前的研究强调了HDAC11下调在改善CAR-T细胞治疗中的潜力。该研究将为进一步研究铺平道路,重点是理解和利用免疫治疗结果的表观遗传机制。
    Epigenetic mechanisms are involved in several cellular functions, and their role in the immune system is of prime importance. Histone deacetylases (HDACs) are an important set of enzymes that regulate and catalyze the deacetylation process. HDACs have been proven beneficial targets for improving the efficacy of immunotherapies. HDAC11 is an enzyme involved in the negative regulation of T cell functions. Here, we investigated the potential of HDAC11 downregulation using RNA interference in CAR-T cells to improve immunotherapeutic outcomes against prostate cancer. We designed and tested four distinct short hairpin RNA (shRNA) sequences targeting HDAC11 to identify the most effective one for subsequent analyses. HDAC11-deficient CAR-T cells (shD-NKG2D-CAR-T) displayed better cytotoxicity than wild-type CAR-T cells against prostate cancer cell lines. This effect was attributed to enhanced activation, degranulation, and cytokine release ability of shD-NKG2D-CAR-T when co-cultured with prostate cancer cell lines. Our findings reveal that HDAC11 interference significantly enhances CAR-T cell proliferation, diminishes exhaustion markers PD-1 and TIM3, and promotes the formation of T central memory TCM populations. Further exploration into the underlying molecular mechanisms reveals increased expression of transcription factor Eomes, providing insight into the regulation of CAR-T cell differentiation. Finally, the shD-NKG2D-CAR-T cells provided efficient tumor control leading to improved survival of tumor-bearing mice in vivo as compared to their wild-type counterparts. The current study highlights the potential of HDAC11 downregulation in improving CAR-T cell therapy. The study will pave the way for further investigations focused on understanding and exploiting epigenetic mechanisms for immunotherapeutic outcomes.
    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
    实体肿瘤微环境中T细胞有限的扩增能力和功能失活是体内使用肿瘤浸润淋巴细胞(TIL)的应用中面临的主要问题。我们试图确定携带PD-1-CD28增强受体和CD19CAR的TIL是否可以克服这一限制并介导肿瘤消退。首先,PD-1-CD28增强受体或CD19CAR修饰的NY-ESO-1-TCR-T细胞模拟TIL功能的抗肿瘤作用(以下简称“PD-1-CD28-TCR-T”或“CD19CAR-TCR-T”细胞,分别)在体外和荷瘤模型中使用NY-ESO-1过表达的肿瘤细胞系进行了测试。此外,在体内评估了S-TIL(通过用编码PD-1-CD28-T2A-CD19CAR的质粒转导修饰的TIL)的安全性和抗肿瘤能力.PD-1-CD28-TCR-T细胞显示出强大的抗肿瘤能力,该能力在体内不受PD-1/PD-L1信号传导的影响。用CD19+B细胞刺激的CD19CAR-TCR-T细胞在体外和体内都表现出强大的扩增和抗肿瘤能力。3例难治性实体瘤患者接受了S-TIL输注。没有观察到治疗相关的死亡率,而且没有患者出现严重的副作用。一名黑色素瘤患者获得了部分反应,2例结肠癌或肾癌患者在接受S-TIL治疗后病情长期稳定.据我们所知,这是第一项描述自体S-TIL过继转移以控制晚期癌症患者疾病的安全性和有效性的研究,这表明S-TIL可能是一种有前途的癌症替代疗法。
    The limited expansion ability and functional inactivation of T cells within the solid tumor microenvironment are major problems faced during in the application of using tumor-infiltrating lymphocytes (TILs) in vivo. We sought to determine whether TILs carrying a PD-1-CD28-enhanced receptor and CD19 CAR could overcome this limitation and mediate tumor regression. First, anti-tumor effects of PD-1-CD28-enhanced receptor or CD19 CAR modified NY-ESO-1-TCR-T cells to mimic the TILs function (hereafter \"PD-1-CD28-TCR-T\" or \"CD19 CAR-TCR-T\" cells, respectively) were tested using the NY-ESO-1 over-expressed tumor cell line in vitro and in a tumor-bearing model. Furthermore, the safety and anti-tumor ability of S-TILs (TILs modified through transduction with a plasmid encoding the PD-1-CD28-T2A-CD19 CAR) were evaluated in vivo. PD-1-CD28-TCR-T cells showed a formidable anti-tumor ability that was not subject to PD-1/PD-L1 signaling in vivo. CD19 CAR-TCR-T cells stimulated with CD19+ B cells exhibited powerful expansion and anti-tumor abilities both in vitro and in vivo. Three patients with refractory solid tumors received S-TILs infusion. No treatment-related mortality was observed, and none of the patients experienced serious side effects. One patient with melanoma achieved a partial response, and two patients with colon or kidney cancer achieved long-term stable disease following S-TILs therapy. To the best of our knowledge, this is the first study describing the safety and efficacy of the adoptive transfer of autologous S-TILs to control disease in patients with advanced cancers, suggesting that S-TILs may be a promising alternative therapy for cancer.
    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
    多发性骨髓瘤(MM),大多数患者的浆细胞异常增殖和单克隆免疫球蛋白重链或轻链的产生,传统上与生存不良有关,尽管自引入新型药物以来,所有年龄组的中位生存期均有所改善。随着新药和新治疗方案的发展,生存率显著提高,如嵌合抗原受体T细胞疗法(CAR-T),在MM治疗中显示出了希望并给出了新的希望。汽车现在被列为第一-,第二-,和第三代CAR,基于整合到其设计中的单价至三价共刺激分子的数量。这篇综述的范围相对狭窄,因为它主要是关于CAR-T疗法在MM中的临床应用的文献比较。因此,我们的目标是概述CAR-T细胞在治疗MM方面的新进展,因此,在这篇综述中,我们综述了CAR-T细胞在MM中的临床使用进展,试图为其在管理MM时的临床使用提供参考。
    Multiple myeloma (MM), marked by abnormal proliferation of plasma cells and production of monoclonal immunoglobulin heavy or light chains in the majority of patients, has traditionally been associated with poor survival, despite improvements achieved in median survival in all age groups since the introduction of novel agents. Survival has significantly improved with the development of new drugs and new treatment options, such as chimeric antigen receptor T-cell therapy (CAR-T), which have shown promise and given new hope in MM therapy. CARs are now classified as first-, second-, and third-generation CARs based on the number of monovalent to trivalent co-stimulatory molecules incorporated into their design. The scope of this review was relatively narrow because it was mainly about a comparison of the literature on the clinical application of CAR-T therapy in MM. Thus, our goal is to provide an overview of the new advances of CAR-T cells in the cure of MM, so in this review we looked at the progress of the clinical use of CAR-T cells in MM to try to provide a reference for their clinical use when managing MM.
    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:尽管抗CD19嵌合抗原受体(CAR)T细胞疗法被批准为复发性/难治性(R/R)B细胞淋巴瘤的非常有效的补救策略,在R/R胃肠道(GI)淋巴瘤的经验仍然不足。
    方法:我们总结了抗CD19CART细胞治疗12例R/RGI淋巴瘤患者的疗效和副作用。根据文献,将R/R胃肠道淋巴瘤患者分为具有不同特征的亚组:大体积/无大体积疾病,胃/胃肠道受累,胃肠/合并胃肠外病变,溃疡/肿块或结节类型,有/无消化道出血。
    结果:这12例患者的客观缓解率(ORR)为66.67%。无大病组ORR为83.33%,胃受累组80.00%,溃疡型组100.00%,无消化道出血组为80.00%。12例患者的CR率为33.33%。无大病组的CR为50.0%,胃受累组60.00%,溃疡型组80.00%。12例患者输液后6个月的PFS和OS分别为54.55%和58.33%,分别。6个月时的总生存期(OS)在无大体积疾病组中较高。其他组之间在6个月时的OS或无进展生存期(PFS)没有差异。胃肠道病变组的CAR-T细胞平均峰值和细胞因子释放综合征(CRS)分级较高。消化道出血组IFN-γ平均峰值和CRS分级较高。胃肠道病变组6例患者中有4例为肿瘤负荷高的患者。仅胃肠道受累的患者发生胃肠道出血的风险较高。
    结论:高肿瘤负荷的ORR和CR,胃肠道受累,肿块或结节类型和消化道出血组较低。消化道病变组和消化道出血组CRS分级较高。仅胃肠道受累的患者发生胃肠道出血的风险较高。
    BACKGROUND: Although anti-CD19 chimeric antigen receptor (CAR) T cell therapy was approved as a very effective salvage strategy in relapsed/refractory (R/R) B cell lymphoma, the experience in R/R gastrointestinal (GI) lymphoma is still insufficient.
    METHODS: We summarized the efficacy and side effects of anti-CD19 CAR T-cell therapy in 12 patients with R/R GI lymphoma. Based on literature, the R/R GI lymphoma patients were divided into subgroups with different characteristics: Bulky/No bulky disease, Gastric/Gastrointestinal involvement, Gastrointestinal/Combined extra-gastrointestinal lesions, Ulcer/Lumps or nodules type, With/without gastrointestinal bleeding.
    RESULTS: The objective response rate (ORR) was 66.67% in these 12 patients. The ORR was 83.33% in no bulky disease group, 80.00% in gastric involvement group, 100.00% in ulcer type group, and 80.00% in no gastrointestinal bleeding group. The CR rate was 33.33% in these 12 patients. The CR was 50.0% in no bulky disease group, 60.00% in gastric involvement group, and 80.00% in ulcer type group. The PFS and OS rate of the 12 patients at 6 months after infusion were 54.55% and 58.33%, respectively. The overall survival (OS) at 6 months was higher in no bulky disease group. There was no difference of the OS or the progression free survival (PFS) at 6 months between the other groups. The mean peak of CAR-T cells and Cytokine Release Syndrome (CRS) grade were higher in gastrointestinal lesions group. The mean peak of IFN-γ and CRS grade were higher in gastrointestinal bleeding group. Four out of six patients in group of gastrointestinal lesions group were patient with high tumor burden. Patients with gastrointestinal involvement only were at higher risk for gastrointestinal bleeding.
    CONCLUSIONS: The ORR and CR of high tumor load, gastrointestinal involvement, lumps or nodules type and gastrointestinal bleeding group were lower. The CRS grade was higher in gastrointestinal lesions group and in gastrointestinal bleeding group. Patients with gastrointestinal involvement only were at higher risk for gastrointestinal bleeding.
    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
    已经证明了CAR-NK细胞对表达CD19的血癌的临床功效,它们也显示出治疗实体瘤的潜力。然而,尚未研究CAR-NK细胞治疗人口腔舌鳞状细胞癌(OTSCC)的疗效.
    我们使用免疫组织化学和免疫荧光评估了人OTSCC组织和细胞系中的MUC1表达。我们构建了从多能干细胞(iPSC衍生的MUC1靶向CAR-NK细胞)表达靶向MUC1的CAR的NK细胞,并使用xCELLigence实时细胞分析系统和CCK8测定在体外评估了它们对OTSCC的有效性。并且通过在用MUC1靶向的CAR-NK细胞处理的BNDG小鼠中每天测量异种移植物生长来在体内进行。作为控制,我们使用iPSC衍生的NK细胞和无NK培养基,它们是无汽车和空白的,分别。
    MUC1表达在所有OTSCC患者的79.5%(66/83)和III期和IV期的72.7%(24/33)中检测到。在III和IV阶段MUC1阳性OTSCC中,63.6%(21/33)和48.5%(16/33)的患者MUC1阳性癌细胞率超过50%和80%,分别。iPSC来源的MUC1靶向CAR-NK细胞在体外对表达MUC1的OTSCC细胞表现出显著的细胞毒性,以时间和剂量依赖的方式,与iPSC来源的NK细胞和空白对照相比,对异种移植物生长均显示出明显的抑制作用。我们没有观察到体重减轻,BNDG小鼠的严重血液学毒性或NK细胞介导的死亡。
    MUC1靶向的CAR-NK细胞对人类OTSCC具有显著功效,他们有希望的治疗反应需要进一步的临床试验。
    The clinical efficacy of CAR-NK cells against CD19-expressing blood cancers has been demonstrated, and they have shown potential for treating solid tumors as well. However, the efficacy of CAR-NK cells for treating human oral tongue squamous cell carcinoma (OTSCC) has not been examined.
    We assessed MUC1 expression in human OTSCC tissue and a cell line using immunohistochemistry and immunofluorescence. We constructed NK cells that express CAR targeted to MUC1 from pluripotent stem cells (iPSC-derived MUC1-targeted CAR-NK cells) and evaluated their effectiveness against OTSCC in vitro using the xCELLigence Real-Time Cell Analysis system and CCK8 assay, and in vivo by measuring xenograft growth daily in BNDG mice treated with MUC1-targeted CAR-NK cells. As controls, we used iPSC-derived NK cells and NK-free media, which were CAR-free and blank, respectively.
    MUC1 expression was detected in 79.5% (66/83) of all OTSCC patients and 72.7% (24/33) of stage III and IV. In stage III and IV MUC1 positive OTSCC, 63.6% (21/33) and 48.5% (16/33) patients had a MUC1-positive cancer cell rate of more than 50% and 80%, respectively. The iPSC-derived MUC1-targeted CAR-NK cells exhibited significant cytotoxicity against MUC1-expressing OTSCC cells in vitro, in a time- and dose-dependent manner, and showed a significant inhibitory effect on xenograft growth compared to both the iPSC-derived NK cells and the blank controls. We observed no weight loss, severe hematological toxicity or NK cell-mediated death in the BNDG mice.
    The MUC1-targeted CAR-NK cells had significant efficacy against human OTSCC, and their promising therapeutic response warrants further clinical trials.
    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
    CAR-T细胞疗法,一种新颖的治疗方法,目前在癌症治疗领域备受关注,已成为许多研究的主题,并在血液恶性肿瘤的治疗中显示出巨大的潜力,如白血病和淋巴瘤。本研究旨在分析淋巴瘤领域发表的CAR-T细胞治疗相关文章的特点,探讨现有的热点和前沿。2013年至2022年发表的相关文章来自WebofScience核心合集。CiteSpace,VOSviewer,文献计量在线分析平台,MicrosoftExcel,使用R软件进行文献计量分析和可视化。与这项研究有关的出版物数量逐年增加,包括来自62个国家和地区的1023篇文章和760篇评论,2092个机构,1040日记帐,8727位作者美国,中国,德国是这一研究领域的主要出版国。排名前10位的机构都来自美国,影响因子最高的期刊是血液,出版物最多的作者是弗雷德里克·L·洛克,最有影响力的作家是卡尔·H·琼。前三个关键词是\"淋巴瘤,\"\"免疫疗法,\"和\"治疗。\“\”Maude(2014)\”是过去十年中引用最多和最强的突发性参考。这项研究提供了对淋巴瘤中CAR-T细胞疗法的全面文献计量分析,这可以帮助研究人员了解该领域当前的研究热点,探索潜在的研究方向,并确定未来的发展趋势。
    CAR-T cell therapy, a novel therapeutic approach that has attracted much attention in the field of cancer treatment at present, has become the subject of many studies and has shown great potential in the treatment of hematological malignancies, such as leukemia and lymphoma. This study aims to analyze the characteristics of articles published on CAR-T cell therapy in the lymphoma field and explore the existing hotspots and frontiers. The relevant articles published from 2013 to 2022 were retrieved from the Web of Science Core Collection. CiteSpace, VOSviewer, Bibliometric online analysis platform, Microsoft Excel, and R software were used for bibliometric analysis and visualization. The number of publications related to the research has been increasing year by year, including 1023 articles and 760 reviews from 62 countries and regions, 2092 institutions, 1040 journals, and 8727 authors. The United States, China, and Germany are the main publishing countries in this research field. The top 10 institutions are all from the United States, the journal with the highest impact factor is BLOOD, the author with the most publications is Frederick L Locke, and the most influential author is Carl H June. The top three keywords are \"Lymphoma,\" \"Immunotherapy,\" and \"Therapy.\" \"Maude (2014)\" is the most cited and strongest burstiness reference over the past decade. This study provides a comprehensive bibliometric analysis of CAR-T cell therapy in lymphoma, which can help researchers understand the current research hotspots in this field, explore potential research directions, and identify future development trends.
    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:卵巢癌(OC)是全球流行的高度侵袭性妇科恶性肿瘤。大多数OC病例通常在晚期诊断,这导致常规治疗后5年总生存率低于35%。此外,免疫检查点抑制剂疗法在治疗OC患者中显示出有限的疗效,由于T细胞浸润不足,CAR-T疗法也显示出适度的结果。因此,必须开发新的策略来增强OC肿瘤微环境中T细胞的持久性和运输。
    方法:在本研究中,我们基于嵌合抗原受体结构开发了一种新的卵巢癌过继性T细胞疗法.我们使用配体-受体结合基序来增强靶向CA125的治疗效果。由于间皮素可以高亲和力天然结合CA125,我们将间皮素的核心结合片段与4-1BB和CD3ζ信号片段连接在一起,以组装一种新型的CA125靶向嵌合受体(CR)。还构建了靶向源自4H11抗体的CA125的CAR结构。将CR-和CAR-编码RNA电穿孔到T细胞中以评估它们在体外和体内的抗肿瘤活性。
    结果:虽然CR-T或CAR-T细胞对两种卵巢癌细胞系表现出中等活性,与单独表达CR或CAR的T细胞相比,共表达CR和CAR的T细胞表现出优异的杀伤作用。此外,与卵巢肿瘤相互作用后,CR和CART细胞释放活化标志物和功能性细胞因子的能力显著增强。同样,CR和CAR共表达T细胞持续控制NSG小鼠移植卵巢癌肿瘤的生长,并显著延长肿瘤攻击小鼠的总生存期。转录组测序显示,与表达CR或CAR的T细胞相比,共表达CR和CAR的T细胞的存活和细胞毒性显著改变。
    结论:我们的研究结果表明,靶向CR和CAR的CA125可以协同杀死卵巢癌细胞,这表明在肿瘤中通过两个结合基序同时靶向CA125可能改善卵巢癌治疗的治疗结果。
    Ovarian cancer (OC) is a highly aggressive gynecological malignancy prevalent worldwide. Most OC cases are typically diagnosed at advanced stages, which has led to a 5-year overall survival rate of less than 35% following conventional treatment. Furthermore, immune checkpoint inhibitor therapy has shown limited efficacy in the treatment of patients with OC, and CAR-T therapy has also demonstrated modest results owing to inadequate T cell infiltration. Therefore, novel strategies must be developed to enhance T cell persistence and trafficking within the OC tumor microenvironment.
    In this study, we developed a novel adoptive T-cell therapy for ovarian cancer based on a chimeric antigen receptor structure. We used a ligand-receptor binding motif to enhance the therapeutic effect of targeting CA125. Since mesothelin can naturally bind to CA125 with high affinity, we concatenated the core-binding fragment of mesothelin with the 4-1BB and CD3ζ signal fragments to assemble a novel CA125-targeting chimeric receptor (CR). The CAR structure targeting CA125 derived from the 4H11 antibody was also constructed. CR- and CAR-encoding RNA were electroporated into T cells to evaluate their antitumor activity both in vitro and in vivo.
    While CR-T or CAR-T cells exhibited moderate activity against two ovarian cancer cell lines, T cells co-expressing CR and CAR exhibited a superior killing effect compared to T cells expressing either CR or CAR alone. Furthermore, upon interaction with ovarian tumors, the ability of CR and CAR T cells to release activation markers and functional cytokines increased significantly. Similarly, CR and CAR co-expressing T cells persistently controlled the growth of transplanted ovarian cancer tumors in NSG mice and significantly prolonged the overall survival of tumor-challenged mice. Transcriptome sequencing revealed that the survival and cytotoxicity of T cells co-expressing CR and CAR were significantly altered compared with those of T cells expressing either CR or CAR.
    Our findings demonstrate that CA125 targeting CR and CAR can synergistically kill ovarian cancer cells, indicating that CA125 targeting by the two binding motifs simultaneously in tumors may improve the therapeutic outcomes of ovarian 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
    CD19嵌合抗原受体(CAR)工程化NK细胞已用于治疗患有复发性和/或难治性B细胞恶性肿瘤的患者,并显示出令人鼓舞的结果和安全性。然而,NK细胞的持久性差仍然是CARNK细胞治疗的主要挑战。由IL-12,IL-15和IL-18诱导的记忆样NK细胞(MLNK)显示出对肿瘤再刺激的增强和延长的反应。使它们成为过继性细胞免疫疗法的有吸引力的候选者。这里,我们显示了使用逆转录病毒载体将CD19CAR有效且稳定的基因递送至记忆样NK细胞,其转导效率与常规NK细胞相当。表面分子的分析揭示了CAR工程记忆样NK细胞(CARMLNK)的独特表型,如CD94的表达增加和NKp30以及KIR2DL1的下调所证明的。与传统的CARNK细胞相比,CARMLNK细胞表现出显著增加的IFN-γ产生和脱粒响应CD19+靶细胞,导致对CD19+白血病细胞和淋巴瘤细胞的细胞毒活性增强。此外,IL-12/-15/-18诱导的记忆特性改善了CAR-MLNK细胞的体内持久性,并显着抑制了淋巴瘤移植瘤小鼠模型中的肿瘤生长,导致CD19+荷瘤小鼠的生存期延长。总之,我们的数据表明,CD19CAR工程记忆样NK细胞表现出优异的持久性和抗肿瘤活性对CD19+肿瘤,这可能是治疗复发或难治性B细胞恶性肿瘤的有吸引力的方法。
    CD19 chimeric antigen receptor (CAR) engineered NK cells have been used for treating patients with relapsed and/or refractory B cell malignancies and show encouraging outcomes and safety profile. However, the poor persistence of NK cells remains a major challenge for CAR NK cell therapy. Memory-like NK cells (MLNK) induced by IL-12, IL-15, and IL-18 have shown enhanced and prolonged responses to tumor re-stimulation, making them an attractive candidate for adoptive cellular immunotherapy. Here, we show efficient and stable gene delivery of CD19 CAR to memory-like NK cells using retroviral vectors with transduction efficiency comparable to those achieved with conventional NK cells. Analysis of surface molecules revealed a distinct phenotypic profile in CAR engineered memory-like NK cells (CAR MLNK), as evidenced by increased expression of CD94 and downregulation of NKp30 as well as KIR2DL1. Compared to conventional CAR NK cells, CAR MLNK cells exhibited significantly increased IFN-γ production and degranulation in response to CD19+ target cells, resulting in enhanced cytotoxic activity against CD19+ leukemia cells and lymphoma cells. Furthermore, memory properties induced by IL-12/-15/-18 improved the in vivo persistence of CAR MLNK cells and significantly suppressed tumor growth in a exnograft mouse model of lymphoma, leading to prolonged survival of CD19+ tumor-bearing mouse. Altogether, our data indicate that CD19 CAR engineered memory-like NK cells exhibited superior persistence and antitumor activity against CD19+ tumors, which might be an attractive approach for treating patient with relapse or refractory B cell malignancies.
    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
    尽管抗CD19嵌合抗原受体(CAR)T细胞治疗在复发/难治性(R/R)滤泡性淋巴瘤(FL)中取得了令人满意的结果,具有R/RFL和高危疾病特征的患者,以前的造血干细胞移植,庞大的疾病,2年内疾病进展(POD24)的完全反应(CR)较低。27例R/RFL患者,晚期疾病阶段,更高的肿瘤负担,在抗CD19CART细胞治疗之前接受过Bruton酪氨酸激酶(BTK)抑制剂或更高的先前治疗线,或接受BTK抑制剂作为联合治疗,包括在这项研究中。观察抗CD19CART细胞治疗中的临床反应和不良事件(AE)。所有接受BTK抑制剂联合抗CD19-CAR-T细胞治疗的R/RFL患者的疾病分期较晚,更高的肿瘤负担,与未接受BTK抑制剂联合治疗的患者相比,治疗线更高。然而,两组间临床反应无差异.POD24组的临床反应低于非POD24组;然而,FL和转化FL(tFL)组之间的临床反应没有差异,在滤泡性淋巴瘤国际预后指数(FLIPI)11-2和FLIPI13-5组之间,在FLIPI21-2和FLIPI23-5组之间。具有BTK抑制剂的CAR-T组的平均抗CD19CART细胞峰高于不具有BTK抑制剂的CAR-T组。同时,非POD24组患者比例较高,FL组,PR组2个月后达到CR。在有和没有BTK抑制剂的CAR-T组之间没有发现细胞因子分泌的差异。非POD24组较高,FLIPI13-5组,和FLIPI23-5组。在有或没有BTK抑制剂的CAR-T组之间以及在其他组之间没有发现细胞因子释放综合征和免疫效应细胞相关的神经毒性综合征等级的差异。不良预后因素,除POD24外,R/RFL患者对BTK抑制剂联合抗CD19CART细胞疗法的临床反应无影响。因此,BTK抑制剂联合抗CD19CAR-T治疗可能是R/RFL和高危因素患者的有效和安全的方法。试验注册:该研究已在http://www上注册。chictr.org.cn/index。aspx为ChiCTR-ONN-16009862和http://www。chictr.org.cn/index。aspx为ChiCTR1800019622。
    Although anti-CD19 chimeric antigen receptor (CAR) T cell therapy has achieved satisfactory results in relapsed/refractory (R/R) follicular lymphoma (FL), patients with R/R FL and high-risk disease characteristics, previous hematopoietic stem cell transplantation, bulky disease, and progression of disease within 2 years (POD24) had a low complete response (CR). Twenty-seven patients with R/R FL, later disease stages, higher tumor burden, or higher previous treatment lines who had received Bruton tyrosine kinase (BTK) inhibitors before anti-CD19 CAR T cell therapy, or received BTK inhibitors as combination therapy, were included in this study. The clinical response and adverse events (AEs) in anti-CD19 CAR T cell therapy were observed. All patients with R/R FL who received BTK inhibitors combined with anti-CD19-CAR T cell therapy had later disease stages, higher tumor burden, and higher treatment lines than those who did not receive BTK inhibitor combination therapy. However, no difference in the clinical response was found between the two groups. The clinical response in the POD24 group was lower than that in the non-POD24 group; however, no difference in the clinical response was found between the FL and transformed FL (tFL) groups, between the follicular lymphoma international prognostic index (FLIPI) 1 1-2 and FLIPI 1 3-5 groups, and between the FLIPI 2 1-2 and FLIPI 2 3-5 groups. The mean anti-CD19 CAR T cell peak was higher in the CAR-T group with BTK inhibitor than in the CAR-T group without BTK inhibitor. Meanwhile, a higher proportion of patients in the non-POD24 group, FL group, and PR group achieved CR after 2 months. No difference in cytokine secretion was found between the CAR-T group with and without BTK inhibitors. It was higher in the non-POD24 group, FLIPI 1 3-5 group, and FLIPI 2 3-5 group. No difference in cytokine release syndrome and immune effector cell-associated neurotoxic syndrome grades was found between the CAR-T groups with or without BTK inhibitors and between the other groups. Poor prognostic factors, other than POD24, did not affect the clinical response to BTK inhibitors in combination with anti-CD19 CAR T cell therapy in patients with R/R FL. Therefore, BTK inhibitors combined with anti-CD19 CAR-T therapy may be an effective and safe approach for patients with R/R FL and high-risk factors.Trial registration: The study was registered at http://www.chictr.org.cn/index.aspx as ChiCTR-ONN-16009862 and http://www.chictr.org.cn/index.aspx as ChiCTR1800019622.
    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
    Notch信号通路的失调,在物种中高度保守,可以驱动异常的表观遗传修饰,转录,和翻译。由Notch信号传导失调引起的缺陷基因调节通常影响控制肿瘤发生和肿瘤进展的网络。同时,Notch信号传导可以调节参与抗肿瘤或前肿瘤应答和肿瘤免疫原性的免疫细胞。对这些过程的全面了解可以帮助设计靶向Notch信号的新药。从而增强癌症免疫治疗的效果。这里,我们提供了关于Notch信号如何内在调节免疫细胞以及肿瘤细胞或基质细胞中Notch信号的改变如何外在调节肿瘤微环境(TME)中的免疫应答的最新和全面概述.我们还讨论了Notch信号在肠道微生物群介导的肿瘤免疫中的潜在作用。最后,我们提出了在癌症免疫治疗中靶向Notch信号的策略.这些包括溶瘤病毒疗法结合抑制Notch信号,负载有Notch信号调节子的纳米颗粒(NPs)特异性靶向肿瘤相关巨噬细胞(TAMs),以使它们的功能复极化并重塑TME,结合特异性和有效的Notch信号抑制剂或激活剂与免疫检查点阻断剂(ICBs)协同抗肿瘤治疗,并实施定制且有效的synNotch电路系统,以增强嵌合抗原受体(CAR)免疫细胞的安全性。总的来说,这篇综述旨在总结Notch信号内在和外在如何塑造免疫反应以改善免疫治疗。
    Dysregulation of the Notch signaling pathway, which is highly conserved across species, can drive aberrant epigenetic modification, transcription, and translation. Defective gene regulation caused by dysregulated Notch signaling often affects networks controlling oncogenesis and tumor progression. Meanwhile, Notch signaling can modulate immune cells involved in anti- or pro-tumor responses and tumor immunogenicity. A comprehensive understanding of these processes can help with designing new drugs that target Notch signaling, thereby enhancing the effects of cancer immunotherapy. Here, we provide an up-to-date and comprehensive overview of how Notch signaling intrinsically regulates immune cells and how alterations in Notch signaling in tumor cells or stromal cells extrinsically regulate immune responses in the tumor microenvironment (TME). We also discuss the potential role of Notch signaling in tumor immunity mediated by gut microbiota. Finally, we propose strategies for targeting Notch signaling in cancer immunotherapy. These include oncolytic virotherapy combined with inhibition of Notch signaling, nanoparticles (NPs) loaded with Notch signaling regulators to specifically target tumor-associated macrophages (TAMs) to repolarize their functions and remodel the TME, combining specific and efficient inhibitors or activators of Notch signaling with immune checkpoint blockers (ICBs) for synergistic anti-tumor therapy, and implementing a customized and effective synNotch circuit system to enhance safety of chimeric antigen receptor (CAR) immune cells. Collectively, this review aims to summarize how Notch signaling intrinsically and extrinsically shapes immune responses to improve immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号