CAR T-cells

CAR - T 细胞
  • 文章类型: Journal Article
    嵌合抗原受体(CAR)T细胞疗法在过去十年中被证明是癌症治疗的突破。对血液恶性肿瘤产生前所未有的效果。所有批准的CAR-T细胞产品,以及许多正在临床试验中评估的人,使用病毒载体将外源遗传物质部署到T细胞中产生。病毒载体在基因传递中具有长期的临床历史,因此进行了迭代优化,以提高其效率和安全性。尽管如此,它们半随机整合到宿主基因组中的能力使它们可能通过插入诱变和关键细胞基因的失调而致癌。CART细胞施用后的继发性癌症似乎是罕见的不良事件。然而,过去几年记录的几起案件引起了人们对这个问题的关注,到目前为止可能被低估了,考虑到相对较新的CART细胞疗法的部署。此外,在血液恶性肿瘤中获得的最初成功尚未在实体瘤中复制。现在很明显,需要进一步的增强才能使CAR-T细胞增加长期持久性,克服疲惫,应对免疫抑制肿瘤微环境。为了这个目标,各种基因组工程策略正在评估中,最依赖CRISPR/Cas9或其他基因编辑技术。这些方法很容易引入意想不到的,产物细胞中不可逆的基因组改变。在本文的第一部分,我们将讨论用于产生CAR-T细胞的病毒和非病毒方法,而在第二部分,我们将专注于基因编辑和非基因编辑T细胞工程,特别是在优势方面,局限性,和安全。最后,我们将批判性地分析不同的基因部署和基因组工程组合,描述具有卓越安全性的下一代CAR-T细胞生产策略。
    Chimeric antigen receptor (CAR) T-cell therapy has proven a breakthrough in cancer treatment in the last decade, giving unprecedented results against hematological malignancies. All approved CAR T-cell products, as well as many being assessed in clinical trials, are generated using viral vectors to deploy the exogenous genetic material into T-cells. Viral vectors have a long-standing clinical history in gene delivery, and thus underwent iterations of optimization to improve their efficiency and safety. Nonetheless, their capacity to integrate semi-randomly into the host genome makes them potentially oncogenic via insertional mutagenesis and dysregulation of key cellular genes. Secondary cancers following CAR T-cell administration appear to be a rare adverse event. However several cases documented in the last few years put the spotlight on this issue, which might have been underestimated so far, given the relatively recent deployment of CAR T-cell therapies. Furthermore, the initial successes obtained in hematological malignancies have not yet been replicated in solid tumors. It is now clear that further enhancements are needed to allow CAR T-cells to increase long-term persistence, overcome exhaustion and cope with the immunosuppressive tumor microenvironment. To this aim, a variety of genomic engineering strategies are under evaluation, most relying on CRISPR/Cas9 or other gene editing technologies. These approaches are liable to introduce unintended, irreversible genomic alterations in the product cells. In the first part of this review, we will discuss the viral and non-viral approaches used for the generation of CAR T-cells, whereas in the second part we will focus on gene editing and non-gene editing T-cell engineering, with particular regard to advantages, limitations, and safety. Finally, we will critically analyze the different gene deployment and genomic engineering combinations, delineating strategies with a superior safety profile for the production of next-generation CAR T-cell.
    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
    在这种情况下,与常规化疗相比,linatumomab和伊托单抗ozogamicin(INO)在复发性/难治性B细胞急性淋巴细胞白血病(ALL)中均具有活性,并改善了预后。一些前瞻性临床试验已经探索了这些药物在新诊断的成人B细胞ALL中的应用。在年轻人和老年人以及费城染色体(Ph)阳性和Ph阴性ALL中观察到有希望的结果。与单纯化疗相比,这些新方案可导致较高的深度可测量残留病(MRD)阴性率,并可提高生存率。减少对强化化疗和异基因造血干细胞移植(HSCT)的依赖。这篇综述讨论了将INO和/或blinatumomab整合到一线ALL方案中的新方法,包括无化疗方案在某些亚组中的潜在作用.还讨论了MRD监测的作用,包括这如何为整合同种异体HSCT或使用CD19CART细胞的研究性方法的决策提供信息。
    Blinatumomab and inotuzumab ozogamicin (INO) are both active in relapsed/refractory B-cell acute lymphoblastic leukemia (ALL) and improve outcomes compared with conventional chemotherapy in this setting. Several prospective clinical trials have explored the use of these agents in adults with newly diagnosed B-cell ALL, with promising outcomes observed in younger and older adults and in both Philadelphia chromosome (Ph)-positive and Ph-negative ALL. These novel regimens result in high rates of deep measurable residual disease (MRD) negativity and may improve survival compared with chemotherapy-only approaches, allowing for less reliance on intensive chemotherapy and allogeneic hematopoietic stem cell transplantation (HSCT). This review discusses novel approaches to integrating INO and/or blinatumomab into frontline ALL regimens, including the potential role of chemotherapy-free regimens in some subgroups. The role of MRD monitoring is also discussed, including how this can inform decisions for consolidative allogeneic HSCT or investigational approaches with CD19 CAR T-cells.
    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
    利妥昔单抗(RTX)加化疗(R-CHOP)作为淋巴瘤的一线治疗导致约40%的患者复发。因此,目前正在深入研究治疗侵袭性淋巴瘤的新方法.已经建立了几种RTX抗性(RR)细胞系作为替代模型来研究对R-CHOP的抗性。我们的研究表明,RR细胞的特征是CD37的主要下调,CD37是目前作为免疫疗法靶标的分子。使用CD20敲除(KO)细胞系,我们证明CD20和CD37形成复合物,并假设CD20的存在使细胞膜中的CD37稳定。因此,我们观察到,在RR和CD20KO细胞中,抗CD37单克隆抗体(mAb)的补体依赖性细胞毒性均减弱,在溶酶体抑制后可部分恢复.另一方面,与对照相比,抗CD37单克隆抗体在CD20KO细胞中的内在化率增加,提示抗体药物缀合物(ADC)的无阻碍疗效。重要的是,即使是CD37水平的大幅下调也不会妨碍CD37定向嵌合抗原受体(CAR)T细胞的功效.总之,我们在此提出了一种新的CD37调节机制,这对使用抗CD37免疫疗法具有进一步的意义.
    Rituximab (RTX) plus chemotherapy (R-CHOP) applied as a first-line therapy for lymphoma leads to a relapse in approximately 40% of the patients. Therefore, novel approaches to treat aggressive lymphomas are being intensively investigated. Several RTX-resistant (RR) cell lines have been established as surrogate models to study resistance to R-CHOP. Our study reveals that RR cells are characterized by a major downregulation of CD37, a molecule currently explored as a target for immunotherapy. Using CD20 knockout (KO) cell lines, we demonstrate that CD20 and CD37 form a complex, and hypothesize that the presence of CD20 stabilizes CD37 in the cell membrane. Consequently, we observe a diminished cytotoxicity of anti-CD37 monoclonal antibody (mAb) in complement-dependent cytotoxicity in both RR and CD20 KO cells that can be partially restored upon lysosome inhibition. On the other hand, the internalization rate of anti-CD37 mAb in CD20 KO cells is increased when compared to controls, suggesting unhampered efficacy of antibody drug conjugates (ADCs). Importantly, even a major downregulation in CD37 levels does not hamper the efficacy of CD37-directed chimeric antigen receptor (CAR) T cells. In summary, we present here a novel mechanism of CD37 regulation with further implications for the use of anti-CD37 immunotherapies.
    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细胞疗法在各种B细胞恶性肿瘤的管理方面取得了重大进展。然而,将其整合到慢性淋巴细胞白血病(CLL)治疗中一直具有挑战性,很大程度上归因于开发非常有效的无化学替代品。此外,CLL中的CART细胞反应不如其他B细胞淋巴瘤或白血病高。然而,对于对目前的CLL疗法有抵抗力的高风险疾病患者,治疗方案存在一个关键的空白,强调了对这些患者进行过继免疫治疗的紧迫性。CLL内降低的CART细胞功效可以追溯到诸如由于CLL固有的持续抗原刺激而导致的T细胞适应性受损的因素。耐药机制包括肿瘤相关因素,如抗原逃逸,CART细胞内在因素,如T细胞衰竭,和抑制性肿瘤微环境(TME)。对抗CAR-T细胞抗性的新策略包括同时施用增强CAR-T细胞耐力和功能的疗法。以及针对不同抗原的新型CAR-T细胞的工程。此外,“装甲”汽车T细胞的概念,用转基因调节剂来修饰CAR-T细胞功能和肿瘤环境,正在获得牵引力。除此之外,现成的发展,同种异体CART细胞和自然杀伤(NK)细胞为CLL患者的先天性T细胞缺陷提供了有希望的对策。在这次审查中,我们探讨了CAR-T细胞治疗在CLL中的作用,复杂的抗性机制挂毯,以及克服阻力的开创性方法。
    Chimeric antigen receptor (CAR) T-cell therapy has ushered in substantial advancements in the management of various B-cell malignancies. However, its integration into chronic lymphocytic leukemia (CLL) treatment has been challenging, attributed largely to the development of very effective chemo-free alternatives. Additionally, CAR T-cell responses in CLL have not been as high as in other B-cell lymphomas or leukemias. However, a critical void exists in therapeutic options for patients with high-risk diseases who are resistant to the current CLL therapies, underscoring the urgency for adoptive immunotherapies in these patients. The diminished CAR T-cell efficacy within CLL can be traced to factors such as compromised T-cell fitness due to persistent antigenic stimulation inherent to CLL. Resistance mechanisms encompass tumor-related factors like antigen escape, CAR T-cell-intrinsic factors like T-cell exhaustion, and a suppressive tumor microenvironment (TME). New strategies to combat CAR T-cell resistance include the concurrent administration of therapies that augment CAR T-cell endurance and function, as well as the engineering of novel CAR T-cells targeting different antigens. Moreover, the concept of \"armored\" CAR T-cells, armed with transgenic modulators to modify both CAR T-cell function and the tumor milieu, is gaining traction. Beyond this, the development of readily available, allogeneic CAR T-cells and natural killer (NK) cells presents a promising countermeasure to innate T-cell defects in CLL patients. In this review, we explore the role of CAR T-cell therapy in CLL, the intricate tapestry of resistance mechanisms, and the pioneering methods studied to overcome resistance.
    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
    经典霍奇金淋巴瘤(cHL)是一种高度可治愈的疾病,但约20%的患者在标准一线化疗方案后出现进展或复发.自体干细胞移植后的挽救方案代表了这些病例的历史治疗方法。在过去的十年里,随着人们对cHL生物学和肿瘤微环境在疾病过程中的作用的日益了解,在临床实践中引入了新的分子,改善复发/难治性患者的结局。抗CD30抗体-药物结合的本妥昔单抗vedotin和PD-1/PD-L1检查点抑制剂代表了当今化学难治性患者的治疗选择,和随机试验最近证明了它们在一线免疫-化疗联合模式中的有效性。几种能够调节患者T淋巴细胞和NK细胞活性的药物正在开发中,以及许多抗CD30嵌合抗原受体T细胞产物。正在研究多种肿瘤异常表观遗传机制作为抗肿瘤化合物的靶标,例如组蛋白脱乙酰酶抑制剂和低甲基化剂。此外,JAK2抑制与抗PD1阻断相结合揭示了cHL中潜在的互补治疗途径。在这次审查中,我们将总结cHL生物学的最新发现和临床上可用的新治疗方案,以及该领域有前途的未来前景。
    Classical Hodgkin Lymphoma (cHL) is a highly curable disease, but around 20% of patients experience progression or relapse after standard frontline chemotherapy regimens. Salvage regimens followed by autologous stem cell transplants represent the historical treatment approach for these cases. In the last decade, with the increasing understanding of cHL biology and tumor microenvironment role in disease course, novel molecules have been introduced in clinical practice, improving outcomes in the relapsed/refractory setting. The anti-CD30 antibody-drug conjugated brentuximab vedotin and PD-1/PD-L1 checkpoint inhibitors represent nowadays curative options for chemorefractory patients, and randomized trials recently demonstrated their efficacy in frontline immune-chemo-combined modalities. Several drugs able to modulate the patients\' T-lymphocytes and NK cell activity are under development, as well as many anti-CD30 chimeric antigen receptor T-cell products. Multiple tumor aberrant epigenetic mechanisms are being investigated as targets for antineoplastic compounds such as histone deacetylase inhibitors and hypomethylating agents. Moreover, JAK2 inhibition combined with anti-PD1 blockade revealed a potential complementary therapeutic pathway in cHL. In this review, we will summarize recent findings on cHL biology and novel treatment options clinically available, as well as promising future perspectives in the field.
    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细胞疗法已成为B细胞和浆细胞恶性肿瘤的有力治疗选择,许多患者受益于它的使用。迄今为止,六种CAR-T细胞产品已被FDA和EMA批准,还有更多正在临床试验中开发和研究。过继细胞转移的整个领域经历了令人难以置信的发展过程,我们现在正处于免疫疗法新时代的边缘,其影响将超越血液系统恶性肿瘤。感兴趣的领域是,例如,实体肿瘤学,自身免疫性疾病,传染病,和其他人。虽然到目前为止已经取得了很多成就,克服重大挑战和困难仍需付出巨大努力。我们目睹了知识的迅速扩张,由新的生物医学技术和CAR设计诱导。CAR-T细胞治疗的时代才刚刚开始,新产品将在未来拓宽治疗领域。本文综述了CAR-T细胞的临床应用,专注于批准的产品,强调它们的好处,但也表明局限性和挑战。
    Chimeric antigen receptor (CAR) T-cell therapy has become a powerful treatment option in B-cell and plasma cell malignancies, and many patients have benefited from its use. To date, six CAR T-cell products have been approved by the FDA and EMA, and many more are being developed and investigated in clinical trials. The whole field of adoptive cell transfer has experienced an unbelievable development process, and we are now at the edge of a new era of immune therapies that will have its impact beyond hematologic malignancies. Areas of interest are, e.g., solid oncology, autoimmune diseases, infectious diseases, and others. Although much has been achieved so far, there is still a huge effort needed to overcome significant challenges and difficulties. We are witnessing a rapid expansion of knowledge, induced by new biomedical technologies and CAR designs. The era of CAR T-cell therapy has just begun, and new products will widen the therapeutic landscape in the future. This review provides a comprehensive overview of the clinical applications of CAR T-cells, focusing on the approved products and emphasizing their benefits but also indicating limitations and challenges.
    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
    研究比较了复发性/难治性大B细胞淋巴瘤(LBCL)患者的嵌合抗原受体(CAR)T细胞疗法和挽救性化疗,但有必要进一步证明其相对有效性.
    我们的系统评价确定了比较axicabtageneciloleucel(axi-cel)疗效和安全性结果的研究,lisocabtagenemaraleucel(liso-cel)和tisagenlecleucel(tisa-cel)试验用于挽救LBCL患者的化疗队列,这些患者先前有≥2行治疗;扩展的证据网络包括间接比较CAR-T细胞治疗.我们使用贝叶斯分层建模进行了网络元分析。
    三项比较ZUMA-1(axi-cel)的研究,确定了在SCHOLAR-1队列中挽救化疗的TRANSCEND(liso-cel)和JULIET(tisa-cel)试验。Axi-cel(比值比[OR]:5.63;95%可信区间[CrI]:2.66-12.42)和liso-cel(OR:4.26;95%CrI:2.33-7.93)显示出与tisa-cel相比,总体反应率显着增加,但不是彼此。Axi-cel相对于liso-cel(风险比[HR]:0.54;95%CrI:0.37-0.79)和tisa-cel(HR:0.47;95%CrI:0.26-0.88)显示出总生存期的显着改善。axi-cel的≥3级神经系统事件发生率高于tisa-cel和liso-cel。
    我们强调了CAR-T细胞疗法在临床结果上的重要差异。与tisa-cel和liso-cel相比,Axi-cel表现出改善的总体生存率,与tisa-cel相比,axi-cel和liso-cel均显示出更高的应答率。
    UNASSIGNED: Studies have compared chimeric antigen receptor (CAR) T-cell therapies and salvage chemotherapy in relapsed/refractory large B-cell lymphoma (LBCL) patients, but further evidence of their relative effectiveness is warranted.
    UNASSIGNED: Our systematic review identified studies comparing efficacy and safety outcomes of axicabtagene ciloleucel (axi-cel), lisocabtagene maraleucel (liso-cel) and tisagenlecleucel (tisa-cel) trials to salvage chemotherapy cohorts in LBCL patients with ≥2 prior lines of treatment; and an extended evidence network included indirect comparisons comparing CAR T-cell therapies. We conducted network meta-analyzes using Bayesian hierarchical modeling.
    UNASSIGNED: Three studies comparing ZUMA-1 (axi-cel), TRANSCEND (liso-cel) and JULIET (tisa-cel) trials to salvage chemotherapy within the SCHOLAR-1 cohort were identified. Axi-cel (odds ratio [OR]:5.63; 95% credible interval [CrI]:2.66-12.42) and liso-cel (OR:4.26; 95%CrI:2.33-7.93) showed a significant increased overall response rate compared to tisa-cel, but not to one-another. Axi-cel demonstrated significant improvements in overall survival relative to liso-cel (hazard ratio [HR]:0.54; 95%CrI:0.37-0.79) and tisa-cel (HR:0.47; 95%CrI:0.26-0.88). Higher rates of grade ≥3 neurological events were observed with axi-cel than with tisa-cel and liso-cel.
    UNASSIGNED: We highlight important differences in clinical outcomes between CAR T-cell therapies. Axi-cel demonstrated improved overall survival compared to tisa-cel and liso-cel, and both axi-cel and liso-cel showed higher response rates compared to tisa-cel.
    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

       PDF(Pubmed)

  • 文章类型: Journal Article
    最近,嵌合抗原受体(CAR)T细胞的突破性免疫治疗策略已被引入血液肿瘤学.然而,将这种新的治疗方法应用于实体癌,必须在选择的肿瘤中确定合适的分子靶标。CEACAM家族蛋白参与一系列恶性肿瘤的进展,包括胰腺癌和乳腺癌,并为抗癌治疗提供有吸引力的目标。在这项工作中,我们使用一种新的CEACAM靶向的基于2A3单结构域抗体的嵌合抗原受体T细胞,在体外和动物模型中评估其抗肿瘤特性.最初,据报道,2A3抗体靶向CEACAM6分子;然而,我们的体外共孵育实验显示2A3-CAR-T细胞对CEACAM5和/或CEACAM6高人细胞系的活化和高细胞毒性,提示这种抗体的交叉反应性。此外,在BxPC-3异种移植物模型中体内测试的2A3-CART细胞在早期和晚期干预治疗方案中表现出对胰腺癌异种移植物的高疗效。我们的结果首次显示新的基于2A3sdAb的CART细胞对CEACAM5和CEACAM6分子的靶向增强。结果强烈支持2A3-CAR-T细胞作为针对CEACAM5/6过表达癌症的潜在治疗策略的进一步发展。
    Recently, a breakthrough immunotherapeutic strategy of chimeric antigen receptor (CAR) T-cells has been introduced to hematooncology. However, to apply this novel treatment in solid cancers, one must identify suitable molecular targets in the tumors of choice. CEACAM family proteins are involved in the progression of a range of malignancies, including pancreatic and breast cancers, and pose attractive targets for anticancer therapies. In this work, we used a new CEACAM-targeted 2A3 single-domain antibody-based chimeric antigen receptor T-cells to evaluate their antitumor properties in vitro and in animal models. Originally, 2A3 antibody was reported to target CEACAM6 molecule; however, our in vitro co-incubation experiments showed activation and high cytotoxicity of 2A3-CAR T-cells against CEACAM5 and/or CEACAM6 high human cell lines, suggesting cross-reactivity of this antibody. Moreover, 2A3-CAR T-cells tested in vivo in the BxPC-3 xenograft model demonstrated high efficacy against pancreatic cancer xenografts in both early and late intervention treatment regimens. Our results for the first time show an enhanced targeting toward CEACAM5 and CEACAM6 molecules by the new 2A3 sdAb-based CAR T-cells. The results strongly support the further development of 2A3-CAR T-cells as a potential treatment strategy against CEACAM5/6-overexpressing 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
    晚期肝细胞癌(HCC)患者有几种全身治疗选择。肝癌有许多已知的危险因素,虽然有些,比如丙型肝炎,现在可以治疗,其他人不是。例如,与代谢功能障碍相关的慢性肝病的发病率正在增加,并且没有特异性的治疗方法。潜在的肝脏疾病,耐药性,越来越多的无特异性生物标志物的治疗方案都是为每位患者选择最佳治疗方案的挑战.常规化疗几乎从未用于晚期疾病,而是用免疫疗法治疗,酪氨酸激酶抑制剂,和VEGF抑制剂。靶向各种受体的免疫检查点抑制剂已经或目前正在进行临床评估。正在进行的三药方案试验可能是晚期HCC治疗的未来。嵌合抗原受体修饰的T细胞的其他免疫调节方法,双特异性抗体,细胞因子诱导的杀伤细胞,自然杀伤细胞,疫苗正处于早期临床试验阶段。针对HCC的靶向治疗仍然有限,但代表了潜在的增长领域。随着我们从一线索拉非尼转向晚期肝癌,临床试验对照组应包括索拉非尼以外的标准治疗,这是一个更好的比较促进治疗。
    Patients with advanced hepatocellular carcinoma (HCC) have several systemic treatment options. There are many known risk factors for HCC, and although some, such as hepatitis C, are now treatable, others are not. For example, metabolic dysfunction-related chronic liver disease is increasing in incidence and has no specific treatment. Underlying liver disease, drug resistance, and an increasing number of treatment options without specific biomarkers are all challenges in selecting the best treatment for each patient. Conventional chemotherapy is almost never used for advanced-stage disease, which instead is treated with immunotherapy, tyrosine kinase inhibitors, and VEGF inhibitors. Immune checkpoint inhibitors targeting various receptors have been or are currently undergoing clinical evaluation. Ongoing trials with three-drug regimens may be the future of advanced-stage HCC treatment. Other immune-modulatory approaches of chimeric antigen receptor-modified T cells, bispecific antibodies, cytokine-induced killer cells, natural killer cells, and vaccines are in early-stage clinical trials. Targeted therapies remain limited for HCC but represent an area of potential growth. As we shift away from first-line sorafenib for advanced HCC, clinical trial control arms should comprise a standard treatment other than sorafenib, one that is a better comparator for advancing therapies.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号