CAR-T

CAR - T
  • 文章类型: Case Reports
    CAR-T细胞免疫疗法的发展显着提高了治疗多发性骨髓瘤的功效。目前,各种各样的目标,包括BCMA,CS1、CD38、FcRH5和GPRC5D,正在接受调查。尽管取得了这些重大进展,挑战如抗原逃逸,CAR-T细胞的有限持久性,肿瘤微环境的复杂性持续存在,导致治疗后复发。
    我们报告了一例复发性和难治性多发性骨髓瘤(RRMM)患者,该患者在多轮放疗和化疗后,在下肢肌肉出现了大量髓外浆细胞瘤。患者接受了针对BCMA和CS1的CAR-T细胞免疫疗法;然而,尽管经过治疗,肿瘤仍有进展。随后进行了髓外浆细胞瘤的手术切除。将肿瘤组织与邻近组织进行比较,肿瘤组织中MYBL2的表达增加,可能导致双重靶向CAR-T细胞治疗后髓外复发缺乏改善.
    在接受了多个周期的化疗和放疗的复发和难治性多发性骨髓瘤患者中,针对BCMA和CS1的双靶向CAR-T细胞治疗未能有效治疗髓外复发.MYBL2在多发性骨髓瘤中的表达升高与预后较差相关。
    UNASSIGNED: The development of CAR-T-cell immunotherapy has notably elevated the efficacy of treating multiple myeloma. Currently, a variety of targets, including BCMA, CS1, CD38, FcRH5, and GPRC5D, are being investigated. Despite these significant advancements, challenges such as antigen escape, limited persistence of CAR-T cells, and the intricate nature of the tumor microenvironment persist, leading to relapses following treatment.
    UNASSIGNED: We report the case of a patient with recurrent and refractory multiple myeloma (RRMM) who developed a substantial extramedullary plasmacytoma in the muscles of the lower limb following multiple rounds of radiotherapy and chemotherapy. The patient underwent CAR-T-cell immunotherapy targeting BCMA and CS1; however, the tumor progressed despite treatment. Surgical resection of the extramedullary plasmacytoma was subsequently performed. Upon comparison of the tumor tissue with the adjacent tissue, increased expression of MYBL2 was noted in the tumor tissue, potentially contributing to the lack of improvement in extramedullary relapse after dual-targeted CAR-T cell therapy.
    UNASSIGNED: In patients with recurrent and refractory multiple myeloma who underwent multiple cycles of chemotherapy and radiotherapy, dual-targeted CAR-T cell therapy aimed at BCMA and CS1 failed to effectively manage extramedullary relapse. Elevated expression of MYBL2 in multiple myeloma correlates with a poorer prognosis.
    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
    肝癌,最常见的表现为肝细胞癌(HCC),是世界上第六大最常见的癌症。在HCC中,免疫系统在肿瘤细胞的生长和增殖中起着至关重要的作用。肝癌通过肿瘤微环境实现免疫逃逸,这显著促进了这种癌症的发展。这里,本文就调节性T细胞(Tregs)在肿瘤微环境中的功能和作用,强调Tregs如何抑制和调节免疫和肿瘤细胞的功能,细胞因子,配体和受体,etc,从而促进肿瘤免疫逃逸。此外,讨论了CAR-T治疗HCC的机制,并阐述了CAR-T与Tregs的关系。
    Liver cancer, which most commonly manifests as hepatocellular carcinoma (HCC), is the sixth most common cancer in the world. In HCC, the immune system plays a crucial role in the growth and proliferation of tumor cells. HCC achieve immune escape through the tumor microenvironment, which significantly promotes the development of this cancer. Here, this article introduces and summarizes the functions and effects of regulatory T cells (Tregs) in the tumor microenvironment, highlighting how Tregs inhibit and regulate the functions of immune and tumor cells, cytokines, ligands and receptors, etc, thereby promoting tumor immune escape. In addition, it discusses the mechanism of CAR-T therapy for HCC and elaborate on the relationship between CAR-T and Tregs.
    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
    在过去的十年里,嵌合抗原受体(CAR)-T细胞疗法已成为一种有前途的免疫治疗方法,用于对抗癌症,在儿童和成人患者的复发性/难治性血液系统恶性肿瘤中证明了显着疗效。CAR-自然杀伤(CAR-NK)细胞通过提供几个明显的优势来补充CAR-T细胞疗法。CAR-NK细胞不需要HLA相容性并且表现出低安全性问题。此外,CAR-NK细胞有利于“现成的”疗法,与CAR-T细胞相比,提供了显著的物流优势。CAR-T和CAR-NK细胞在血液恶性肿瘤中都显示出一致且有希望的结果。然而,由于各种障碍,包括有限的肿瘤运输和浸润,它们对实体瘤的疗效仍然有限,以及免疫抑制肿瘤微环境。在这次审查中,我们讨论了CAR-T和CAR-NK细胞免疫疗法的最新进展和当前挑战,特别关注它们在实体瘤中应用的障碍。我们还深入分析了CAR-NK细胞与CAR-T细胞相比的优缺点,并强调了CAR-NK的CAR优化。最后,我们探索这些过继免疫疗法的未来前景,强调尖端生物技术工具在塑造下一代细胞免疫疗法方面的贡献越来越大。
    In the past decade, chimeric antigen receptor (CAR)-T cell therapy has emerged as a promising immunotherapeutic approach for combating cancers, demonstrating remarkable efficacy in relapsed/refractory hematological malignancies in both pediatric and adult patients. CAR-natural killer (CAR-NK) cell complements CAR-T cell therapy by offering several distinct advantages. CAR-NK cells do not require HLA compatibility and exhibit low safety concerns. Moreover, CAR-NK cells are conducive to \"off-the-shelf\" therapeutics, providing significant logistic advantages over CAR-T cells. Both CAR-T and CAR-NK cells have shown consistent and promising results in hematological malignancies. However, their efficacy against solid tumors remains limited due to various obstacles including limited tumor trafficking and infiltration, as well as an immuno-suppressive tumor microenvironment. In this review, we discuss the recent advances and current challenges of CAR-T and CAR-NK cell immunotherapies, with a specific focus on the obstacles to their application in solid tumors. We also analyze in depth the advantages and drawbacks of CAR-NK cells compared to CAR-T cells and highlight CAR-NK CAR optimization. Finally, we explore future perspectives of these adoptive immunotherapies, highlighting the increasing contribution of cutting-edge biotechnological tools in shaping the next generation of cellular immunotherapy.
    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
    这是一项名为CARTITUDE-4的3期临床试验的摘要。该试验比较了抗癌疗法西塔卡塔那autoleucel(或cilta-cel)与多发性骨髓瘤患者的标准疗法,一种影响特定类型血细胞的癌症,称为浆细胞。研究中的人已经接受过1到3种多发性骨髓瘤的治疗方法,包括一种叫做来那度胺的常见抗骨髓瘤治疗,但是他们的多发性骨髓瘤没有好转。
    这项研究的419名参与者中约有一半接受了cilta-cel,而另一半接受标准疗法,或常用于治疗多发性骨髓瘤的疗法。接受cilta-cel的参与者从他们的血液中收集了一种称为T细胞的免疫细胞,并进行了基因修饰以识别骨髓瘤细胞上发现的特定蛋白质。这些修饰的T细胞,其中包括西塔塞尔,然后注入血液。
    经过大约1年的研究,与标准治疗组(49%)相比,cilta-cel组(76%)更多的参与者还活着,他们的癌症没有恶化.两组中最常见的副作用是感染和低血细胞计数。细胞因子释放综合征(由免疫系统过度激活引起的潜在严重副作用)很常见,但大多是轻度的。神经毒性(包括免疫效应细胞相关神经毒性综合征,会引起头痛等症状,意识的改变,和记忆的困难,注意,说话,或理解他人)较不常见,在使用cilta-cel治疗的参与者中,有20.5%的参与者报告。
    在CARTITUDE-4中,与接受标准治疗的参与者相比,接受cilta-cel治疗的参与者在接受cilta-cel12个月后表现出改善,并且在疾病控制下存活。临床试验注册:NCT04181827(CARTITUDE-4)(ClinicalTrials.gov)。
    UNASSIGNED: This is a summary of a phase 3 clinical trial called CARTITUDE-4. This trial compared the anti-cancer therapy ciltacabtagene autoleucel (or cilta-cel) with standard therapies in people who have multiple myeloma, a cancer that affects specific kinds of blood cells called plasma cells. The people in the study had been treated with 1 to 3 previous treatments for multiple myeloma, including a common anti-myeloma treatment called lenalidomide, but their multiple myeloma did not get better.
    UNASSIGNED: About half of the 419 participants in this study received cilta-cel, while the other half received standard therapies, or therapies that are commonly used to treat multiple myeloma. Participants who received cilta-cel had a type of immune cell called T cells collected from their blood and genetically modified to recognize a specific protein found on myeloma cells. These modified T cells, which comprise cilta-cel, were then infused back into the bloodstream.
    UNASSIGNED: After approximately 1 year in the study, more participants were alive without their cancer getting worse in the cilta-cel group (76%) than in the standard therapies group (49%). The most common side effects in both groups were infections and low blood cell counts. Cytokine release syndrome (a potentially serious side effect caused by overactivation of the immune system) was common but mostly mild. Neurotoxicities (including immune effector cell-associated neurotoxicity syndrome, which can cause symptoms such as headaches, changes in consciousness, and difficulty with memory, attention, speaking, or understanding others) were less common and were reported in 20.5% of participants treated with cilta-cel.
    UNASSIGNED: In CARTITUDE-4, more participants treated with cilta-cel showed improvements and were alive with control of their disease 12 months after receiving cilta-cel compared with participants who received standard therapies.Clinical Trial Registration: NCT04181827 (CARTITUDE-4) (ClinicalTrials.gov).
    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: Letter
    暂无摘要。
    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
    CAR-T细胞疗法已成为血液恶性肿瘤的高效治疗方法。目前有六种商业CAR-T产品已被FDA批准用于治疗B-ALL等疾病,LBCL,MCL,FL,MM,和CLL/SLL。“真实世界”研究使我们能够评估普通人群的结果,以确定与原始试验中包括的人群相比的有效性和安全性。基于几个进行得很好的代表不同人群的“真实世界”研究,我们报告,导致这些疗法获得批准的原始试验的结果与实践中的结果相当.
    CAR-T cell therapy has established itself as a highly effective treatment for hematological malignancies. There are currently six commercial CAR-T products that have been FDA approved for diseases such as B-ALL, LBCL, MCL, FL, MM, and CLL/SLL. \"Real-world\" studies allow us to evaluate outcomes from the general population to determine their efficacy and safety compared to those who were included in the original trials. Based on several well conducted \"Real-world\" studies that represent diverse populations, we report that outcomes from the original trials that led to the approval of these therapies are comparable to those in practice.
    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细胞恶性肿瘤为CAR-T细胞疗法带来了几个独特的问题,这些问题不是CAR-T细胞对B细胞恶性肿瘤的重大关注。在生产CAR-T细胞时要考虑的一个普遍问题是“脱靶”肿瘤毒性。“当CAR-T细胞靶向的抗原也在正常细胞上表达时,就会发生这种情况。不仅仅是肿瘤细胞,导致CAR-T细胞损伤这些正常细胞。在T细胞肿瘤的CAR-T细胞疗法中,在T细胞上表达的抗原(如CD5、CD7等。)是目标,这导致了一个被称为“自相残杀”的问题,“CAR-T细胞互相杀死。其他问题包括T细胞再生和肿瘤细胞对CAR-T细胞产物的污染。然而,最近的几项临床试验显示,当使用基因组编辑技术通过敲除靶抗原或T细胞受体来克服这些问题时,CAR-T细胞疗法具有优异的效果.本文概述了这些挑战及其解决方案,并讨论了最近的临床试验结果。
    T cell malignancies pose several unique issues for CAR-T cell therapy that were not significant concerns with CAR-T cells for B-cell malignancies. A general problem to consider in the production of CAR-T cells is \"on target-off tumor toxicity.\" This occurs when the antigen targeted by the CAR-T cells is also expressed on normal cells, not just tumor cells, which causes CAR-T cells to damage these normal cells. In CAR-T cell therapy for T cell tumors, antigens expressed on T cells (such as CD5, CD7, etc.) are the targets, which leads to a problem known as \"fratricide,\" where CAR-T cells kill each other. Other issues include T cell aplasia and contamination of CAR-T cell products with tumor cells. However, several recent clinical trials have shown excellent outcomes for CAR-T cell therapy when genome editing technology is used to overcome these issues by knocking out target antigens or T cell receptors. This review article outlines these challenges and their solutions and discusses the results of recent clinical trials.
    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
    免疫疗法通过检查点阻断抗体和过继性T细胞转移等策略在癌症治疗中取得了重大进展。嵌合抗原受体T细胞(CAR-T)已成为组合这些策略并克服其局限性的有希望的方法。这篇综述探讨了CAR-T细胞作为癌症治疗的活药。CAR-T细胞是基因工程免疫细胞,旨在通过识别特定抗原来靶向和消除肿瘤细胞。该研究涉及对CAR-T细胞技术的全面文献综述,覆盖层结构优化,世代,制造工艺,和基因治疗策略。它检查了血液肿瘤和实体瘤中的CAR-T疗法,强调挑战,并提出基于自杀基因的机制来提高安全性。结果表明,CAR-T技术取得了重大进展,特别是在结构优化和生成。为了更广泛的临床应用,制造工艺已经改进。然而,一系列固有的挑战和副作用仍然需要解决。总之,CAR-T细胞对癌症治疗大有希望,但正在进行的研究对于提高肿瘤患者的疗效和安全性至关重要.提出的基于自杀基因的机制提供了一种潜在的解决方案,以减轻副作用,包括细胞因子释放综合征(CAR-T疗法最常见的毒副作用)和相关的神经毒性。
    Immunotherapy has made significant strides in cancer treatment with strategies like checkpoint blockade antibodies and adoptive T cell transfer. Chimeric antigen receptor T cells (CAR-T) have emerged as a promising approach to combine these strategies and overcome their limitations. This review explores CAR-T cells as a living drug for cancer treatment. CAR-T cells are genetically engineered immune cells designed to target and eliminate tumor cells by recognizing specific antigens. The study involves a comprehensive literature review on CAR-T cell technology, covering structure optimization, generations, manufacturing processes, and gene therapy strategies. It examines CAR-T therapy in haematologic cancers and solid tumors, highlighting challenges and proposing a suicide gene-based mechanism to enhance safety. The results show significant advancements in CAR-T technology, particularly in structure optimization and generation. The manufacturing process has improved for broader clinical application. However, a series of inherent challenges and side effects still need to be addressed. In conclusion, CAR-T cells hold great promise for cancer treatment, but ongoing research is crucial to improve efficacy and safety for oncology patients. The proposed suicide gene-based mechanism offers a potential solution to mitigate side effects including cytokine release syndrome (the most common toxic side effect of CAR-T therapy) and the associated neurotoxicity.
    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CRISPR/Cas9技术有望彻底改变具有潜在基因突变的疾病的治疗方法,它面临着一些限制临床进入的重大问题。它们包括低效率的体内全身递送和不期望的脱靶效应。这里,我们展示,通过用硫代磷酸酯DNA寡核苷酸(PS)修饰Cas9,可以有效地在体外和体内递送单一和双特异性CRISPR/Cas9/向导RNA(gRNA)二聚体,并减少脱靶效应。我们表明PS-Cas9/gRNA介导的基因敲除在体外和体内保留了嵌合抗原受体T细胞的活力和扩增。PS-Cas9/gRNA介导患者来源的肿瘤类器官和小鼠异种移植肿瘤的基因扰动,导致有效的肿瘤抗肿瘤作用。Further,HER2抗体-PS-Cas9/gRNA缀合物在体内选择性干扰HER2+卵巢癌异种移植物中的靶向基因。此外,我们创建了双特异性PS-Cas9与两个gRNA靶向相同基因的两个相邻序列,导致体外和体内有效的靶向基因破坏,并显着减少了意外的基因扰动。因此,细胞穿透性PS-Cas9/gRNA可以实现有效的全身递送和基因破坏的精确性。
    Although CRISPR-Cas9 technology is poised to revolutionize the treatment of diseases with underlying genetic mutations, it faces some significant issues limiting clinical entry. They include low-efficiency in vivo systemic delivery and undesired off-target effects. Here, we demonstrate, by modifying Cas9 with phosphorothioate-DNA oligos (PSs), that one can efficiently deliver single and bi-specific CRISPR-Cas9/guide RNA (gRNA) dimers in vitro and in vivo with reduced off-target effects. We show that PS-Cas9/gRNA-mediated gene knockout preserves chimeric antigen receptor T cell viability and expansion in vitro and in vivo. PS-Cas9/gRNA mediates gene perturbation in patient-derived tumor organoids and mouse xenograft tumors, leading to potent tumor antitumor effects. Further, HER2 antibody-PS-Cas9/gRNA conjugate selectively perturbs targeted genes in HER2+ ovarian cancer xenografts in vivo. Moreover, we created bi-specific PS-Cas9 with two gRNAs to target two adjacent sequences of the same gene, leading to efficient targeted gene disruption ex vivo and in vivo with markedly reduced unintended gene perturbation. Thus, the cell-penetrating PS-Cas9/gRNA can achieve efficient systemic delivery and precision in gene disruption.
    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细胞在血液病中显示出显著的功效。然而,CART疗法在实体瘤中的疗效有限,包括胶质母细胞瘤(GBM)。其中最重要的原因是免疫抑制的肿瘤微环境(TME),促进肿瘤生长并抑制免疫细胞以消除肿瘤细胞。人转化生长因子-β(TGF-β)在形成抑制性GBMTME和驱动抗GBM反应的抑制中起关键作用。为了减轻TGF-β介导的抑制活性,我们将显性阴性TGF-β受体II(dnTGFβRII)与我们之前的双顺反子CART-EGFR-IL13Rα2构建体结合,目前正在临床试验中进行评估,产生CART-EGFR-IL13Rα2-dnTGFβRII,我们正在开发用于临床应用的三模块结构。我们假设这种方法将更有效地颠覆GBM观察到的抵抗机制。我们的数据表明CART-EGFR-IL13Rα2-dnTGFβRII显着增强T细胞增殖,增强功能反应,提高旁观者细胞的适应性,特别是通过降低富含TGF-β的TME中的TGF-β浓度。此外,体内研究验证了dnTGFβRII与CAR合作在NSG小鼠模型中靶向和根除GBM的安全性和有效性.
    Chimeric antigen receptor (CAR) T cells have shown significant efficacy in hematological diseases. However, CAR T therapy has demonstrated limited efficacy in solid tumors, including glioblastoma (GBM). One of the most important reasons is the immunosuppressive tumor microenvironment (TME), which promotes tumor growth and suppresses immune cells used to eliminate tumor cells. The human transforming growth factor β (TGF-β) plays a crucial role in forming the suppressive GBM TME and driving the suppression of the anti-GBM response. To mitigate TGF-β-mediated suppressive activity, we combined a dominant-negative TGF-β receptor II (dnTGFβRII) with our previous bicistronic CART-EGFR-IL13Rα2 construct, currently being evaluated in a clinical trial, to generate CART-EGFR-IL13Rα2-dnTGFβRII, a tri-modular construct we are developing for clinical application. We hypothesized that this approach would more effectively subvert resistance mechanisms observed with GBM. Our data suggest that CART-EGFR-IL13Rα2-dnTGFβRII significantly augments T cell proliferation, enhances functional responses, and improves the fitness of bystander cells, particularly by decreasing the TGF-β concentration in a TGF-β-rich TME. In addition, in vivo studies validate the safety and efficacy of the dnTGFβRII cooperating with CARs in targeting and eradicating GBM in an NSG mouse model.
    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