CAR T cell therapy

CAR T 细胞疗法
  • 文章类型: Journal Article
    激活条件安全开关有可能逆转由工程细胞疗法引起的严重毒性,包括嵌合抗原受体(CAR)T细胞。功能惰性的,来自人表皮生长因子受体(EGFRt)的非免疫原性细胞表面标志物是一种有前途的安全开关,已用于多种临床构建体,可被西妥昔单抗靶向,临床上可用的单克隆抗体。然而,这种方法需要EGFRt的高的和持久的细胞表面表达,以确保抗体介导的工程细胞的清除是快速和完整的.在这里,我们表明将短的近膜序列掺入EGFRt多肽可增强其在T细胞表面的表达及其对抗体依赖性细胞毒性(ADCC)的敏感性。与EGFRt相比,将该优化变体(EGFRopt)并入双顺反子和三顺反子CAR设计中导致CART细胞的更快速的体内消除和它们的效应子活性的稳健终止。这些研究将EGFRopt确立为下一代基于细胞的疗法开发的卓越安全开关。
    Activation of a conditional safety switch has the potential to reverse serious toxicities arising from the administration of engineered cellular therapies, including chimeric antigen receptor (CAR) T cells. The functionally inert, non-immunogenic cell surface marker derived from human epidermal growth factor receptor (EGFRt) is a promising safety switch that has been used in multiple clinical constructs and can be targeted by cetuximab, a clinically available monoclonal antibody. However, this approach requires high and persistent cell surface expression of EGFRt to ensure that antibody-mediated depletion of engineered cells is rapid and complete. Here we show that incorporating a short juxtamembrane sequence into the EGFRt polypeptide enhances its expression on the surface of T cells and their susceptibility to antibody-dependent cellular cytotoxicity (ADCC). Incorporating this optimized variant (EGFRopt) into bicistronic and tricistronic CAR designs results in more rapid in vivo elimination of CAR T cells and robust termination of their effector activity compared to EGFRt. These studies establish EGFRopt as a superior safety switch for the development of next-generation cell-based therapeutics.
    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
    轻链(AL)淀粉样变性是一种浆细胞疾病,与多发性骨髓瘤(MM)的区别在于由于错误折叠蛋白的组织沉积引起的器官受累程度。AL淀粉样变性的治疗很大程度上是从为MM患者开发的治疗方法中借用的。高剂量化疗后自体干细胞移植(ASCT)历来与最佳结果相关。最近将daratumumab纳入前期治疗代表了一项重大进展,并改变了治疗模式。质疑ASCT的作用。非常活跃的新型免疫和细胞疗法的发展,特别是B细胞成熟抗原(BCMA)定向治疗,对于MM患者也具有同样的变革性,目前正在AL淀粉样变性患者中进行研究。这些包括嵌合抗原受体(CAR)T细胞,双特异性抗体,和抗体药物缀合物。虽然有限,复发性和难治性AL淀粉样变性患者的初步数据显示出有希望的结果,预计AL淀粉样变性的治疗前景将继续发展。特别注意安全,器官恢复的潜力,在评估新的治疗方法和/或治疗模式时,生活质量将是重要的。
    Light chain (AL) amyloidosis is a plasma cell disorder distinguished from multiple myeloma (MM) by the degree of organ involvement due to tissue deposition of misfolded proteins. Treatments for AL amyloidosis have largely been borrowed from those developed for patients with MM. High-dose chemotherapy followed by autologous stem cell transplant (ASCT) has historically been associated with the best outcomes. The recent incorporation of daratumumab into up front therapy represents a significant advance and has changed the treatment paradigm, calling into question the role of ASCT. The development of very active novel immune and cellular therapies, specifically B cell maturation antigen (BCMA)-directed therapies, has similarly been transformative for patients with MM and is now being studied in patients with AL amyloidosis. These include chimeric antigen receptor (CAR) T cells, bispecific antibodies, and antibody drug conjugates. Although limited, preliminary data in patients with relapsed and refractory AL amyloidosis are showing promising results, and it is expected that the treatment landscape for AL amyloidosis will continue to evolve. Particular attention to safety, potential for organ recovery, and quality of life will be important when evaluating new treatments and/or treatment paradigms.
    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
    多形性胶质母细胞瘤(GBM)的标准治疗包括手术和辅助放化疗。嵌合抗原受体(CAR)T细胞疗法已在GBM中证明了疾病修饰活性,并具有很大的前景。辐射,GBM的标准护理治疗,具有众所周知的免疫调节特性,可以克服免疫抑制肿瘤微环境(TME);然而,辐射剂量优化和与CART细胞治疗的整合还没有很好的定义。用滴定剂量的5、10和20Gray(Gy)的立体定向放射外科(SRS)治疗GBM的小鼠免疫功能模型,并使用Nanostring分析TME。根据肿瘤生长动力学和TME中的基因表达变化确定10Gy的调节剂量。我们证明了10Gy的调节剂量激活了TME中的先天和适应性免疫细胞。用10Gy与mCART细胞组合治疗的小鼠表现出增强的抗肿瘤活性和对IL13Rα2阳性肿瘤的再攻击的优异记忆应答。此外,10Gy加mCART细胞也通过一种机制保护免受IL13Rα2阴性肿瘤的侵害,在某种程度上,c-GAS-STING途径依赖性。一起,这些发现支持低剂量10Gy辐射联合mCART细胞作为GBM的治疗策略.
    Standard-of-care treatment for Glioblastoma Multiforme (GBM) is comprised of surgery and adjuvant chemoradiation. Chimeric Antigen Receptor (CAR) T cell therapy has demonstrated disease-modifying activity in GBM and holds great promise. Radiation, a standard-of-care treatment for GBM, has well-known immunomodulatory properties and may overcome the immunosuppressive tumor microenvironment (TME); however, radiation dose optimization and integration with CAR T cell therapy is not well defined. Murine immunocompetent models of GBM were treated with titrated doses of stereotactic radiosurgery (SRS) of 5, 10, and 20 Gray (Gy), and the TME was analyzed using Nanostring. A conditioning dose of 10 Gy was determined based on tumor growth kinetics and gene expression changes in the TME. We demonstrate that a conditioning dose of 10 Gy activates innate and adaptive immune cells in the TME. Mice treated with 10 Gy in combination with mCAR T cells demonstrated enhanced antitumor activity and superior memory responses to rechallenge with IL13Rα2-positive tumors. Furthermore, 10 Gy plus mCAR T cells also protected against IL13Rα2-negative tumors through a mechanism that was, in part, c-GAS-STING pathway-dependent. Together, these findings support combination conditioning with low-dose 10 Gy radiation in combination with mCAR T cells as a therapeutic strategy for GBM.
    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
    尽管有强化治疗,患有复发性或难治性实体瘤的儿科患者预后较差,需要新的治疗方法.免疫疗法提供了常规治疗选择的替代方案,但需要鉴定差异表达的抗原以将抗肿瘤活性导向疾病部位。B7-H3(CD276)是在一系列恶性肿瘤中表达并且在正常组织中具有有限表达的免疫调节蛋白。B7-H3在儿童实体瘤中高表达,包括骨肉瘤。横纹肌肉瘤,尤因肉瘤,肾母细胞瘤,神经母细胞瘤,和许多罕见的肿瘤。在本文中,我们回顾了针对小儿实体瘤的B7-H3靶向嵌合抗原受体(B7-H3-CAR)T细胞疗法,报告临床前发展战略和概述积极的儿科临床试验的景观。我们确定了CART细胞治疗实体肿瘤的成功挑战,包括定位和穿透实体肿瘤部位。逃避敌对的肿瘤微环境,支持T细胞扩增和持久性,并避免固有的肿瘤耐药性。我们强调了克服这些挑战并增强B7-H3-CART细胞效果的策略,包括先进的CART细胞设计和联合疗法的整合。
    Despite intensive therapies, pediatric patients with relapsed or refractory solid tumors have poor outcomes and need novel treatments. Immune therapies offer an alternative to conventional treatment options but require the identification of differentially expressed antigens to direct antitumor activity to sites of disease. B7-H3 (CD276) is an immune regulatory protein that is expressed in a range of malignancies and has limited expression in normal tissues. B7-H3 is highly expressed in pediatric solid tumors including osteosarcoma, rhabdomyosarcoma, Ewing sarcoma, Wilms tumor, neuroblastoma, and many rare tumors. In this article we review B7-H3-targeted chimeric antigen receptor (B7-H3-CAR) T cell therapies for pediatric solid tumors, reporting preclinical development strategies and outlining the landscape of active pediatric clinical trials. We identify challenges to the success of CAR T cell therapy for solid tumors including localizing to and penetrating solid tumor sites, evading the hostile tumor microenvironment, supporting T cell expansion and persistence, and avoiding intrinsic tumor resistance. We highlight strategies to overcome these challenges and enhance the effect of B7-H3-CAR T cells, including advanced CAR T cell design and incorporation of combination therapies.
    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
    使用治疗性免疫调节实现长期疾病控制是一个长期存在的概念,在血液恶性肿瘤中具有悠久的传统。除了异基因造血干细胞移植,继续为其他具有挑战性的诊断提供潜在的治愈性治疗,近年来,在免疫检查点阻断的白血病和淋巴瘤的免疫治疗方面取得了令人印象深刻的进展,双特异性单克隆抗体,和CART细胞疗法。尽管他们取得了成功,无反应,复发,免疫毒性仍然很常见,因此优先阐明潜在机制并确定预测性生物标志物.现在,越来越多的单细胞基因组工具提供了一个系统的免疫学观点,以前所未有的分辨率解决免疫疗法的分子和细胞机制。这里,我们回顾了利用这些技术进步来跟踪免疫反应的最新研究,免疫抗性的出现,和毒性。随着单细胞免疫监测工具的发展和变得更容易获得,我们期望它们广泛用于常规临床应用,以催化更精确的治疗指导个人免疫反应。
    Achieving long-term disease control using therapeutic immunomodulation is a long-standing concept with a strong tradition in blood malignancies. Besides allogeneic hematopoietic stem cell transplantation that continues to provide potentially curative treatment for otherwise challenging diagnoses, recent years have seen impressive progress in immunotherapies for leukemias and lymphomas with immune checkpoint blockade, bispecific monoclonal antibodies, and CAR T cell therapies. Despite their success, non-response, relapse, and immune toxicities remain frequent, thus prioritizing the elucidation of the underlying mechanisms and identifying predictive biomarkers. The increasing availability of single-cell genomic tools now provides a system\'s immunology view to resolve the molecular and cellular mechanisms of immunotherapies at unprecedented resolution. Here, we review recent studies that leverage these technological advancements for tracking immune responses, the emergence of immune resistance, and toxicities. As single-cell immune monitoring tools evolve and become more accessible, we expect their wide adoption for routine clinical applications to catalyze more precise therapeutic steering of personal immune responses.
    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
    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
    尽管最近的治疗进展,晚期肝细胞癌(HCC)的结局仍不令人满意,强调需要新的治疗方法。CRISPR(聚集定期间隔短回文重复)基因编辑技术提供了创新的治疗方法,涉及对癌细胞或过继性T细胞进行遗传操作以对抗HCC。这篇综述全面评估了CRISPR系统在HCC治疗中的应用。专注于体内靶向癌细胞以及嵌合抗原受体(CAR)T细胞和T细胞受体(TCR)工程化T细胞的开发。我们探索了基于CRISPR的癌症疗法与现有治疗方案之间的潜在协同作用。讨论正在进行的临床试验和CRISPR技术在通过先进的安全措施改善HCC治疗结果中的作用。总之,这篇综述提供了在HCC治疗中使用CRISPR技术的前景和当前挑战的见解,以改善患者预后和彻底改变HCC疗法的最终目标。
    Despite recent therapeutic advancements, outcomes for advanced hepatocellular carcinoma (HCC) remain unsatisfactory, highlighting the need for novel treatments. The CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) gene-editing technology offers innovative treatment approaches, involving genetic manipulation of either cancer cells or adoptive T cells to combat HCC. This review comprehensively assesses the applications of CRISPR systems in HCC treatment, focusing on in vivo targeting of cancer cells and the development of chimeric antigen receptor (CAR) T cells and T cell receptor (TCR)-engineered T cells. We explore potential synergies between CRISPR-based cancer therapeutics and existing treatment options, discussing ongoing clinical trials and the role of CRISPR technology in improving HCC treatment outcomes with advanced safety measures. In summary, this review provides insights into the promising prospects and current challenges of using CRISPR technology in HCC treatment, with the ultimate goal of improving patient outcomes and revolutionizing the landscape of HCC therapeutics.
    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
    嵌合抗原受体(CAR)T细胞疗法改变了复发性/难治性多发性骨髓瘤的治疗前景。目前的美国食品和药物管理局批准了CART细胞疗法idecabtagenevicleucel和ciltacabtageneautoleucel两种靶B细胞成熟抗原(BCMA),在恶性浆细胞表面表达。尽管大多数患者最初有很深的反应,抗BCMACART细胞治疗后复发很常见。对抗BCMACART细胞疗法的获得性抗性的研究正在进行中。同时,正在追求其他可行的抗原靶标,包括G蛋白偶联受体C类5组成员D(GPRC5D),信号淋巴细胞活化分子家族成员7(SLAMF7),和CD38等。针对这些抗原的CAR-T细胞,单独或与抗BCMA方法组合,当他们从临床前研究转向早期临床试验时,似乎非常有希望。这篇综述总结了BCMA以外的新型CART细胞靶标的当前数据,这些靶标有可能在不久的将来进入治疗领域。
    Chimeric antigen receptor (CAR) T cell therapy has transformed the treatment landscape of relapsed/refractory multiple myeloma. The current Food and Drug Administration approved CAR T cell therapies idecabtagene vicleucel and ciltacabtagene autoleucel both target B cell maturation antigen (BCMA), which is expressed on the surface of malignant plasma cells. Despite deep initial responses in most patients, relapse after anti-BCMA CAR T cell therapy is common. Investigations of acquired resistance to anti-BCMA CAR T cell therapy are underway. Meanwhile, other viable antigenic targets are being pursued, including G protein-coupled receptor class C group 5 member D (GPRC5D), signaling lymphocytic activation molecule family member 7 (SLAMF7), and CD38, among others. CAR T cells targeting these antigens, alone or in combination with anti-BCMA approaches, appear to be highly promising as they move from preclinical studies to early phase clinical trials. This review summarizes the current data with novel CAR T cell targets beyond BCMA that have the potential to enter the treatment landscape in the near future.
    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
    免疫抑制性肿瘤微环境(TME)减少了针对实体瘤的嵌合抗原受体(CAR)T细胞疗法。这里,制备CART细胞膜伪装纳米催化剂(ACSP@TCM)以增强CART细胞对实体瘤的治疗功效。ACSP@TCM是通过用CART细胞膜包封核/壳Au/Cu2-xSe和3-溴丙酮酸来制备的。结果表明,CART细胞膜伪装比同源肿瘤细胞膜伪装具有更好的靶向作用。ACSP@TCM具有吸引人的协同化学动力学/光热疗法(CDT/PTT)作用,可以诱导NALM6细胞的免疫原性细胞死亡(ICD)。此外,3-溴丙酮酸可以通过抑制糖酵解过程来抑制乳酸的流出,调节TME的酸度,并为CAR-T细胞的存活提供更有利的环境。此外,光声(PA)成像和计算机断层扫描(CT)成像性能可以指导ACSP@TCM介导的肿瘤治疗。结果表明,ACSP@TCM显着增强了CART细胞治疗对NALM6实体瘤的疗效,完全消除肿瘤。这项工作为实体肿瘤中的CAR-T细胞治疗提供了一种有效的肿瘤策略。
    The immunosuppressive tumor microenvironment (TME) reduces the chimeric antigen receptor (CAR) T-cell therapy against solid tumors. Here, a CAR T cell membrane-camouflaged nanocatalyst (ACSP@TCM) is prepared to augment CAR T cell therapy efficacy against solid tumors. ACSP@TCM is prepared by encapsulating core/shell Au/Cu2- xSe and 3-bromopyruvate with a CAR T cell membrane. It is demonstrated that the CAR T cell membrane camouflaging has much better-targeting effect than the homologous tumors cell membrane camouflaging. ACSP@TCM has an appealing synergistic chemodynamic/photothermal therapy (CDT/PTT) effect that can induce the immunogenic cell death (ICD) of NALM 6 cells. Moreover, 3-bromopyruvate can inhibit the efflux of lactic acid by inhibiting the glycolysis process, regulating the acidity of TME, and providing a more favorable environment for the survival of CAR T cells. In addition, the photoacoustic (PA) imaging and computed tomography (CT) imaging performance can guide the ACSP@TCM-mediated tumor therapy. The results demonstrated that the ACSP@TCM significantly enhanced the CAR T cell therapy efficacy against NALM 6 solid tumor mass, and completely eliminated tumors. This work provides an effective tumor strategy for CAR T cell therapy in solid tumors.
    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细胞疗法是一种有前途的治疗儿童复发性或难治性B细胞急性淋巴细胞白血病(R/RBALL)的方法。细胞因子释放综合征(CRS)是CART细胞治疗后的常见毒性,发烧通常是首发症状。在CART细胞治疗后区分CRS与感染可能是具有挑战性的。血浆微生物无细胞DNA(mcfDNA)是一种新颖的诊断工具,可对1000多种生物进行定性和定量评估。这项初步研究旨在表征在CART细胞治疗后的前2个月中患有R/RBALL的儿科患者的mcfDNA结果。
    Chimeric antigen receptor (CAR) T cell therapy is a promising treatment for pediatric patients with relapsed or refractory B cell acute lymphoblastic leukemia (R/R B ALL). Cytokine release syndrome (CRS) is a common toxicity after CAR T cell therapy and fever is often the first symptom. Differentiating CRS from infection after CAR T cell therapy can be challenging. Plasma microbial cell free DNA (mcfDNA) is a novel diagnostic tool which allows for qualitative and quantitative assessment of over 1000 organisms. This pilot study sought to characterize mcfDNA results in pediatric patients with R/R B ALL in the first 2 months after CAR T cell therapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号