CAR-T cell therapy

CAR - T 细胞疗法
  • 文章类型: Journal Article
    临床试验中的新型治疗剂为对抗这种流行和破坏性疾病的方法提供了范式转变,癌症治疗领域正处于转化革命的边缘。尽管手术等久经考验的癌症治疗方法很重要,辐射,和化疗,这种疾病继续进化和适应,制造新的,更有效的方法是必要的。癌症治疗领域目前正在见证各种创新方法的出现。免疫疗法,包括检查点抑制剂,CAR-T细胞治疗,和癌症疫苗,利用宿主的免疫系统来选择性地靶向和根除恶性细胞,同时最大限度地减少对正常组织的伤害。激酶抑制剂和单克隆抗体等靶向药物的开发允许更有针对性和更少有害的治疗癌症的方法。在基因组学和分子谱分析的帮助下,“精准医学”为每个患者定制独特的基因组成疗法,以最大限度地提高治疗效果,同时最大限度地减少不必要的副作用。表观遗传疗法,代谢干预,无线电药物,越来越重视具有协同效应的联合治疗,进一步拓宽了治疗领域。多阶段临床试验对于确定这些新药的安全性和有效性至关重要。使患者能够获得新的治疗方法,同时也加深了对科学的理解。癌症治疗的未来充满了希望,人工智能和大数据的融合有可能彻底改变早期检测和预防。研究人员之间的合作,和医疗保健提供者,患者的积极参与仍然是正在进行的抗癌斗争的基石。总之,癌症治疗的动态和不断发展的格局为改善治疗结果提供了希望,强调以病人为中心,数据驱动,在我们共同努力建立一个没有癌症的世界时,采取道德基础的方法。
    Novel therapeutic agents in clinical trials offer a paradigm shift in the approach to battling this prevalent and destructive disease, and the area of cancer therapy is on the precipice of a trans formative revolution. Despite the importance of tried-and-true cancer treatments like surgery, radiation, and chemotherapy, the disease continues to evolve and adapt, making new, more potent methods necessary. The field of cancer therapy is currently witnessing the emergence of a wide range of innovative approaches. Immunotherapy, including checkpoint inhibitors, CAR-T cell treatment, and cancer vaccines, utilizes the host\'s immune system to selectively target and eradicate malignant cells while minimizing harm to normal tissue. The development of targeted medicines like kinase inhibitors and monoclonal antibodies has allowed for more targeted and less harmful approaches to treating cancer. With the help of genomics and molecular profiling, \"precision medicine\" customizes therapies to each patient\'s unique genetic makeup to maximize therapeutic efficacy while minimizing unwanted side effects. Epigenetic therapies, metabolic interventions, radio-pharmaceuticals, and an increasing emphasis on combination therapy with synergistic effects further broaden the therapeutic landscape. Multiple-stage clinical trials are essential for determining the safety and efficacy of these novel drugs, allowing patients to gain access to novel treatments while also furthering scientific understanding. The future of cancer therapy is rife with promise, as the integration of artificial intelligence and big data has the potential to revolutionize early detection and prevention. Collaboration among researchers, and healthcare providers, and the active involvement of patients remain the bedrock of the ongoing battle against cancer. In conclusion, the dynamic and evolving landscape of cancer therapy provides hope for improved treatment outcomes, emphasizing a patient-centered, data-driven, and ethically grounded approach as we collectively strive towards a cancer-free world.
    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T细胞(CAR-T细胞)疗法的出现彻底改变了癌症治疗,特别是血液系统恶性肿瘤。这篇评论讨论了CAR-T细胞疗法的发展,关注控制T细胞命运和分化的分子机制。转录和表观遗传因素在确定特异性中起着关键作用,有效性,以及CAR-T细胞疗法的持久性。了解这些机制对于提高疗效和减少与CAR-T细胞疗法相关的不良事件至关重要。释放这些方法的全部潜力。CAR-T细胞产品制造中的T细胞分化在临床结果中起着重要作用。CAR-T细胞疗法的临床疗效与记忆特征之间存在正相关,而与效应子功能或耗尽的特征呈负相关。CAR-T细胞产物的有效性可能受T细胞频率及其增殖能力的影响。这与早期T细胞分化密切相关。涉及不同T记忆细胞亚群的分化过程在抗原消除后启动,指示感染分辨率。在慢性感染或癌症中,T细胞可能会耗尽,以持续的抑制性受体表达为标志,细胞因子产生减少,增殖能力减弱.其他细胞亚群,如CD4+T细胞,先天样T淋巴细胞,NKT细胞,和脐带血来源的造血干细胞,在开发下一代基于CAR-T细胞的疗法方面提供独特的优势。未来的研究应该集中在优化T细胞增强方法和开发策略,以潜在治愈血液系统疾病和实体瘤患者。
    The emergence of chimeric antigen receptor T cell (CAR-T cell) therapy has revolutionized cancer treatment, particularly for hematologic malignancies. This commentary discusses developments in CAR-T cell therapy, focusing on the molecular mechanisms governing T cell fate and differentiation. Transcriptional and epigenetic factors play a pivotal role in determining the specificity, effectiveness, and durability of CAR-T cell therapy. Understanding these mechanisms is crucial to improve the efficacy and decrease the adverse events associated with CAR-T cell therapies, unlocking the full potential of these approaches. T cell differentiation in CAR-T cell product manufacturing plays an important role in clinical outcomes. A positive correlation exists between the clinical efficacy of CAR-T cell therapy and signatures of memory, whereas a negative correlation has been observed with signatures of effector function or exhaustion. The effectiveness of CAR-T cell products is likely influenced by T-cell frequency and by their ability to proliferate, which is closely linked to early T cell differentiation. The differentiation process involving distinct T memory cell subsets is initiated upon antigen elimination, indicating infection resolution. In chronic infections or cancer, T cells may undergo exhaustion, marked by continuous inhibitory receptor expression, decreased cytokine production, and diminished proliferative capacity. Other cell subsets, such as CD4+ T cells, innate-like T lymphocytes, NKT cells, and cord blood-derived hematopoietic stem cells, offer unique advantages in developing the next-generation CAR-T cell-based therapies. Future research should focus on optimizing T-cell-enhancing approaches and developing strategies to potentially cure patients with hematological diseases and solid tumors.
    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
    急性淋巴细胞白血病(ALL)是儿科人群中的主要癌症,尽管它在成人中的发生相对罕见。治疗前风险分层对预测预后至关重要。评估的重要因素包括患者年龄,诊断时的白细胞(WBC)计数,髓外参与,免疫表型,和细胞遗传学畸变。微小残留病(MRD),缓解后主要通过流式细胞术评估,在指导管理计划方面发挥着重要作用。在过去的十年里,所有成果都取得了重大进展。常规化疗显著降低了死亡率;然而,其密集的性质引起了人们对安全性的担忧,并导致出现了复发的耐药病例。因此,美国食品和药物管理局(FDA)已经批准了几种针对复发性/难治性ALL的新型治疗方法,因为它们具有良好的疗效。如改善完全缓解和生存率所示。这些治疗包括酪氨酸激酶抑制剂(TKIs),抗CD19单克隆抗体blinatumomab,抗CD22伊托珠单抗奥佐大霉素,抗CD20利妥昔单抗,和嵌合抗原受体(CAR)T细胞疗法。确定影响治疗决策的变量是基于异质患者特征定制治疗的迫切需要。在各种观察性研究和临床试验中确定的关键预测因素包括淋巴耗竭前疾病负担,复杂的遗传异常,MRD。此外,治疗后严重不良事件的发展可以通过预测模型来预测,考虑采取适当的预防措施。最终目的是通过有效的预测平台将精准医学的概念纳入ALL领域,以利于选择最合适的治疗方法。
    UNASSIGNED: Acute lymphoblastic leukemia (ALL) represents the predominant cancer in pediatric populations, though its occurrence in adults is relatively rare. Pre-treatment risk stratification is crucial for predicting prognosis. Important factors for assessment include patient age, white blood cell (WBC) count at diagnosis, extramedullary involvement, immunophenotype, and cytogenetic aberrations. Minimal residual disease (MRD), primarily assessed by flow cytometry following remission, plays a substantial role in guiding management plans. Over the past decade, significant advancements in ALL outcomes have been witnessed. Conventional chemotherapy has remarkably reduced mortality rates; however, its intensive nature raises safety concerns and has led to the emergence of treatment-resistant cases with recurrence of relapses. Consequently, The U.S. Food and Drug Administration (FDA) has approved several novel treatments for relapsed/refractory ALL due to their demonstrated efficacy, as indicated by improved complete remission and survival rates. These treatments include tyrosine kinase inhibitors (TKIs), the anti-CD19 monoclonal antibody blinatumomab, anti-CD22 inotuzumab ozogamicin, anti-CD20 rituximab, and chimeric antigen receptor (CAR) T-cell therapy. Identifying the variables that influence treatment decisions is a pressing necessity for tailoring therapy based on heterogeneous patient characteristics. Key predictive factors identified in various observational studies and clinical trials include prelymphodepletion disease burden, complex genetic abnormalities, and MRD. Furthermore, the development of serious adverse events following treatment could be anticipated through predictive models, allowing for appropriate prophylactic measures to be considered. The ultimate aim is to incorporate the concept of precision medicine in the field of ALL through valid prediction platform to facilitate the selection of the most suitable treatment approach.
    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
    在最近的III期试验中观察到的结果不一致,该试验评估了嵌合抗原受体T(CAR-T)细胞疗法作为复发性/难治性弥漫性大B患者的二线治疗标准(SOC)细胞淋巴瘤(R/RDLBCL)促使进行荟萃分析以评估CAR-T细胞疗法在这种情况下的有效性。
    进行随机效应荟萃分析以汇集效应估计值,以比较CAR-T细胞疗法和SOC。使用频率网络荟萃分析方法进行混合治疗比较。
    对三个865例患者进行的荟萃分析显示,无事件生存率显着改善(EFS:HR:0.51;95%CI:0.27-0.97;I2:92%),与SOC相比,CAR-T细胞疗法的无进展生存期(PFS:HR:0.47;95%CI:0.37-0.60;I2:0%)。尽管CAR-T细胞疗法有潜在的总生存期(OS)改善的信号,两组之间的差异无统计学意义(HR0.76;95%CI:0.56至1.03;I2:29%)。混合治疗比较显示,与tisa-cel相比,采用Liso-cel(HR:0.37;95%CI:0.22-0.61)和axi-cel(HR:0.42;95%CI:0.29-0.61)的EFS益处显着。
    CAR-T细胞疗法,作为二线治疗,与R/RDLBCL的SOC相比,似乎可有效实现更高的应答率和延迟疾病进展。
    UNASSIGNED: Inconsistent results observed in recent phase III trials assessing chimeric antigenic receptor T (CAR-T) cell therapy as a second-line treatment compared to standard of care (SOC) in patients with relapsed/refractory diffuse large B-cell lymphoma (R/R DLBCL) prompted a meta-analysis to assess the effectiveness of CAR-T cell therapy in this setting.
    UNASSIGNED: Random-effects meta-analysis was conducted to pool effect estimates for comparison between CAR-T cell therapy and SOC. Mixed treatment comparisons were made using a frequentist network meta-analysis approach.
    UNASSIGNED: Meta-analysis of three trials with 865 patients showed significant improvement in event-free survival (EFS: HR: 0.51; 95% CI: 0.27-0.97; I2: 92%), progression-free survival (PFS: HR: 0.47; 95% CI: 0.37-0.60; I2: 0%) with CAR-T cell therapy compared to SOC. Although there was a signal of potential overall survival (OS) improvement with CAR-T cell therapy, the difference was not statistically significant between the two groups (HR 0.76; 95% CI: 0.56 to 1.03; I2: 29%). Mixed treatment comparisons showed significant EFS benefit with liso-cel (HR: 0.37; 95% CI: 0.22-0.61) and axi-cel (HR: 0.42; 95% CI: 0.29-0.61) compared to tisa-cel.
    UNASSIGNED: CAR-T cell therapy, as a second-line treatment, appears to be effective in achieving higher response rates and delaying the disease progression compared to SOC in R/R DLBCL.
    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细胞疗法代表了最具创新性的免疫治疗方法之一。CAR-T细胞疗法在血液系统疾病中取得的令人鼓舞的结果为CAR工程化T细胞在不同类型的实体瘤中的应用铺平了道路。这种过继细胞疗法代表了通过识别肿瘤相关抗原(TAA)根除肿瘤的选择性和有效的方法。工程化CAR-T细胞与TAA的结合激发了几种细胞因子的释放,granzyme,和穿孔素,最终导致癌细胞消除和患者的免疫系统增强。在肿瘤块内有一个癌细胞亚群,被称为癌症干细胞(CSC),在耐药性中起着至关重要的作用,肿瘤进展,和转移。CAR-T细胞疗法确实已被用于靶向CSC特异性抗原作为肿瘤异质性破坏的有效策略。然而,CAR-T细胞疗法功效的障碍表现为CAR-T细胞在CSC生态位的敌对环境中的持久性差。对单一靶向抗原的抗性的发展,肿瘤和T细胞代谢的变化,以及严重不良反应的发生。CSC抗性由免疫抑制肿瘤微环境(TME)的存在证实,其中包括基质细胞,癌症相关成纤维细胞(CAFs),肿瘤相关巨噬细胞(TAMs),骨髓来源的抑制细胞(MDSCs),和免疫细胞。TME组分和CSC之间的关系抑制了CAR-T细胞疗法的功效。为了克服这一挑战,基于CAR-T细胞疗法联合化疗的双重策略可能对逃避免疫抑制性TME至关重要.这里,我们总结了针对CSC的CAR-T细胞疗法的挑战和局限性,特别强调TME和T细胞代谢需求的作用。
    Chimeric antigen receptor (CAR)-T cell therapy represents one of the most innovative immunotherapy approaches. The encouraging results achieved by CAR-T cell therapy in hematological disorders paved the way for the employment of CAR engineered T cells in different types of solid tumors. This adoptive cell therapy represents a selective and efficacious approach to eradicate tumors through the recognition of tumor-associated antigens (TAAs). Binding of engineered CAR-T cells to TAAs provokes the release of several cytokines, granzyme, and perforin that ultimately lead to cancer cells elimination and patient\'s immune system boosting. Within the tumor mass a subpopulation of cancer cells, known as cancer stem cells (CSCs), plays a crucial role in drug resistance, tumor progression, and metastasis. CAR-T cell therapy has indeed been exploited to target CSCs specific antigens as an effective strategy for tumor heterogeneity disruption. Nevertheless, a barrier to the efficacy of CAR-T cell-based therapy is represented by the poor persistence of CAR-T cells into the hostile milieu of the CSCs niche, the development of resistance to single targeting antigen, changes in tumor and T cell metabolism, and the onset of severe adverse effects. CSCs resistance is corroborated by the presence of an immunosuppressive tumor microenvironment (TME), which includes stromal cells, cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), myeloid-derived suppressor cells (MDSCs), and immune cells. The relationship between TME components and CSCs dampens the efficacy of CAR-T cell therapy. To overcome this challenge, the double strategy based on the use of CAR-T cell therapy in combination with chemotherapy could be crucial to evade immunosuppressive TME. Here, we summarize challenges and limitations of CAR-T cell therapy targeting CSCs, with particular emphasis on the role of TME and T cell metabolic demands.
    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
    癌症干细胞(CSC)代表了肿瘤中存在的最具侵袭性的亚群,保留了侵袭能力。转移潜力和高表达水平的药物外排泵负责治疗耐药。癌症仍然是一种无法治愈的疾病,因为标准方案的无效性使这个亚群免于死亡。CSC的选择性靶向仍然是癌症研究领域中未满足的需求。异常的表观遗传重编程促进CSC的启动和维持,能够逃脱免疫系统的防御。能够诱导这种茎样小子集的选择性抑制的有希望的治疗方法包括单独或与表观遗传化合物组合的免疫疗法。这些策略基于仅存在于CSC中而不存在于其他癌细胞或正常细胞中的表位和/或表观遗传改变的特异性表达。因此,在已知CSC特异性改变的患者中,利用CAR-T免疫疗法和表观遗传探针的联合治疗方法可能克服治疗无效的障碍,朝着更精准的医学方法发展.在这篇透视文章中,我们将为表观免疫疗法在富含CSC的肿瘤中的未来应用提供新的启示。连同其潜在的副作用,局限性和治疗耐药性的发展。
    Cancer stem cells (CSCs) represent the most aggressive subpopulation present in the tumor bulk retaining invasive capabilities, metastatic potential and high expression levels of drug efflux pumps responsible for therapy resistance. Cancer is still an incurable disease due to the inefficacy of standard regimens that spare this subpopulation. Selective targeting of CSCs is still an unmet need in cancer research field. Aberrant epigenetic reprogramming promotes the initiation and maintenance of CSCs, which are able to escape the immune system defense. Promising therapeutic approaches able to induce the selective inhibition of this stem-like small subset include immunotherapy alone or in combination with epigenetic compounds. These strategies are based on the specific expression of epitopes and/or epigenetic alterations present only in the CSC and not in the other cancer cells or normal cells. Thus, the combined approach utilizing CAR-T immunotherapy along with epigenetic probes may overcome the barriers of treatment ineffectiveness towards a more precision medicine approach in patients with known specific alterations of CSCs. In this perspective article we will shed new lights on the future applications of epi-immunotherapy in tumors enriched in CSCs, along with its potential side-effects, limitations and the development of therapy 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
    背景:CAR-T细胞疗法被批准用于治疗复发性/难治性(R/R)大B细胞淋巴瘤(LBCL)。然而,由于其毒性,老年患者可能不是这种疗法的候选人,缺乏候选人选择的标准。我们的目的是分析70岁及以上人群中CAR-T细胞疗法的疗效和毒性结果,与现实世界中年轻患者相比。
    方法:进行了一项多中心回顾性研究,包括R/R侵袭性LBCL患者,他们在西班牙GETH-TC/GELTAMO中心接受了商业CAR-T细胞治疗,使用tisagenlecleucel或axicabtageneciloleucel。2019年至2023年。
    结果:截至2023年8月,442名患有侵袭性LBCL的成年患者接受了CAR-T细胞疗法的单采术作为第三行或后续行,并获得了随访数据。412名输液病人中,71(17%)岁以上。基线特征,单采术的产品选择和特征(包括疾病状态,安阿伯舞台,R-IPI,庞大的疾病,LDH和ECOG)在组间具有可比性。从批准到输注和单采到输注的中位时间没有差异。在1个月和3个月时,两组之间的总体和完全缓解率没有差异。中位随访时间为12.2个月(范围1-44),两组之间的12个月无进展生存期(PFS)和总生存期(OS)具有可比性(<70岁时35.2%与≥70y的35.9%(p=p=0.938)和51.1%和52.1%(p=p=0.885),分别)。在单变量和多变量分析中,年龄≥70岁不影响PFS(HR0.98,p=0.941)和OS(HR0.97,p=0.890)。在<70y的82%患者中观察到细胞因子释放综合征(CRS),在≥70y的患者中观察到84.5%(p=0.408)。≥3级CRS在老年组中更常见(5%与15%,p=0.002)。在多变量分析中,年龄≥70岁与≥3级CRS风险增加相关(OR3.7,p=0.013).在总体神经毒性方面没有观察到差异(35%vs.42%,p=0.281)或≥3级(12%与17%,p=0.33)。感染患者的比例,两组在1个月内入住重症监护病房和非复发死亡率相似.
    结论:CAR-T细胞疗法在70岁以上患者中的疗效与在现实世界中年轻患者中的疗效相似。然而,年龄≥70岁是3~4级CRS的独立危险因素.在未来的研究中,应解决减少该人群毒性的其他策略的需求。
    BACKGROUND: CAR-T cell therapy is approved for the treatment of relapsed/refractory (R/R) large B cell lymphoma (LBCL). However, elderly patients might not be candidates for this therapy due to its toxicity, and criteria for candidate selection are lacking. Our aim was to analyze efficacy and toxicity results of CAR-T cell therapy in the population of 70 years and older as compared to those obtained in younger patients in the real-world setting.
    METHODS: A multicentric retrospective study was performed including patients with R/R aggressive LBCL who received commercial CAR-T cell therapy with either tisagenlecleucel or axicabtagene ciloleucel within Spanish GETH-TC/GELTAMO centers between 2019 and 2023.
    RESULTS: As of August 2023, 442 adult patients with aggressive LBCL underwent apheresis for CAR-T cell therapy as third or subsequent line and had follow-up data. Of 412 infused patients, 71 (17%) were 70 years or older. Baseline characteristics, product selection and characteristics at apheresis (including disease status, Ann Arbor stage, R-IPI, bulky disease, LDH and ECOG) were comparable between groups. Median time from both approval to infusion and apheresis to infusion did not differ. No differences were found between groups in overall and complete response rates at one and three months. With a median follow-up of 12.2 months (range 1-44), 12-month progression-free survival (PFS) and overall survival (OS) were comparable between groups (35.2% in <70y vs. 35.9% in ≥70y (p= p=0.938) and 51.1% and 52.1% (p= p=0.885), respectively). Age ≥70 years did not affect PFS (HR 0.98, p=0.941) and OS (HR 0.97, p=0.890) in the univariate and multivariate analysis. Cytokine release syndrome (CRS) was observed in 82% patients in <70y and 84.5% in ≥70y (p=0.408). Grade ≥3 CRS was more frequent in the older group (5% vs. 15%, p=0.002). In the multivariate analysis, age ≥70y was associated with an increased risk of grade ≥3 CRS (OR 3.7, p=0.013). No differences were observed in terms of overall neurotoxicity (35% vs. 42%, p=0.281) or grade ≥3 (12% vs. 17%, p=0.33). The proportion of patients with infections, admission to intensive care unit within the first month and non-relapse mortality were similar between both groups.
    CONCLUSIONS: CAR-T cell therapy in patients older than 70 years showed similar efficacy than that observed in younger patients in the real-world setting. However, age ≥70 years was an independent risk factor for grade 3-4 CRS. Need of additional strategies to reduce toxicity in this population should be addressed in future studies.
    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
    全球癌症死亡的主要原因归因于非小细胞肺癌(NSCLC)。需要持续关注改善这种疾病的诊断和治疗。在这次审查中,重点介绍了治疗NSCLC的最新突破和新趋势.诊断方法的重大进步,包括更好的成像技术和分子生物标志物的利用,正在讨论。这些进步大大增强了早期检测和个性化治疗计划。通过新的靶向治疗和免疫疗法,已经实现了患者预后的显着改善。为晚期非小细胞肺癌患者提供新的希望。这篇综述讨论了尽管最近取得了进展,但获得先进治疗的持续挑战及其相关成本。对新疗法的有希望的研究,如CAR-T细胞疗法和溶瘤病毒,这可能会进一步彻底改变NSCLC治疗,也突出了。这篇综述旨在告知和激励全球NSCLC患者继续努力改善预后。通过全面概述非小细胞肺癌治疗的现状和未来的可能性。
    The leading cause of cancer deaths worldwide is attributed to non-small cell lung cancer (NSCLC), necessitating a continual focus on improving the diagnosis and treatment of this disease. In this review, the latest breakthroughs and emerging trends in managing NSCLC are highlighted. Major advancements in diagnostic methods, including better imaging technologies and the utilization of molecular biomarkers, are discussed. These advancements have greatly enhanced early detection and personalized treatment plans. Significant improvements in patient outcomes have been achieved by new targeted therapies and immunotherapies, providing new hope for individuals with advanced NSCLC. This review discusses the persistent challenges in accessing advanced treatments and their associated costs despite recent progress. Promising research into new therapies, such as CAR-T cell therapy and oncolytic viruses, which could further revolutionize NSCLC treatment, is also highlighted. This review aims to inform and inspire continued efforts to improve outcomes for NSCLC patients globally, by offering a comprehensive overview of the current state of NSCLC treatment and future possibilities.
    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细胞疗法是最有效的免疫疗法之一。CAR-T细胞疗法在血液系统恶性肿瘤的治疗中取得了巨大的成功。然而,由于实体恶性肿瘤的特点,如目标效应,肿瘤外毒性,免疫抑制肿瘤微环境(TME),贩运不足,CAR-T细胞治疗实体肿瘤仍处于探索阶段。间皮素(MSLN)是在各种实体恶性肿瘤细胞的表面上表达的分子,其适合作为具有用于CAR-T细胞疗法的高MSLN表达的肿瘤细胞的靶标。本文简要介绍了CAR-T细胞治疗的发展以及MSLN的结构特征,并通过总结一些临床前实验和临床试验,特别总结了MSLN靶向CAR-T细胞的结构优化策略和增强MSLN靶向CAR-T细胞抗肿瘤功效的方法。以MSLN靶向CAR-T细胞疗法为例,本文总结了研究人员在CAR-T细胞治疗实体肿瘤方面所做的努力,并通过整合现有研究成果总结出可行的治疗方案。
    Chimeric antigen receptor (CAR)-T-cell therapy is one of the most effective immunotherapies. CAR-T-cell therapy has achieved great success in the treatment of hematological malignancies. However, due to the characteristics of solid malignant tumors, such as on-target effects, off-tumor toxicity, an immunosuppressive tumor microenvironment (TME), and insufficient trafficking, CAR-T-cell therapy for solid tumors is still in the exploration stage. Mesothelin (MSLN) is a molecule expressed on the surface of various solid malignant tumor cells that is suitable as a target of tumor cells with high MSLN expression for CAR-T-cell therapy. This paper briefly described the development of CAR-T cell therapy and the structural features of MSLN, and especially summarized the strategies of structure optimization of MSLN-targeting CAR-T-cells and the enhancement methods of MSLN-targeting CAR-T cell anti-tumor efficacy by summarizing some preclinical experiment and clinical trials. When considering MSLN-targeting CAR-T-cell therapy as an example, this paper summarizes the efforts made by researchers in CAR-T-cell therapy for solid tumors and summarizes feasible treatment plans by integrating the existing research results.
    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:针对胶质母细胞瘤(GBM)相关抗原如白细胞介素-13受体亚基α-2(IL-13Rα2)的嵌合抗原受体(CAR)-T细胞疗法迄今取得了有限的临床疗效,部分是由于以抑制分子如转化生长因子-β(TGF-β)为特征的免疫抑制肿瘤微环境(TME)。这项研究的目的是通过在TME中对抗TGF-β介导的免疫抑制来设计更有效的靶向GBM的CAR-T细胞。
    方法:我们设计了一个单链,靶向IL-13Rα2和TGF-β的双特异性CAR,对肿瘤特异性T细胞进行编程以将TGF-β从免疫抑制剂转化为免疫刺激剂。评估双特异性IL-13Rα2/TGF-βCAR-T细胞针对患者来源的GBM异种移植物和鼠神经胶质瘤的同基因模型的功效和安全性。
    结果:与常规IL-13Rα2CAR-T细胞治疗相比,IL-13Rα2/TGF-βCAR-T细胞治疗导致荷瘤脑中更大的T细胞浸润和减少抑制性骨髓细胞存在,导致患者来源的GBM异种移植物和小鼠神经胶质瘤同基因模型的存活率都得到改善。
    结论:我们的研究结果表明,通过重新编程肿瘤特异性T细胞对TGF-β的反应,双特异性IL-13Rα2/TGF-βCAR-T细胞抵抗并重塑免疫抑制性TME以驱动GBM中的有效抗肿瘤反应。
    BACKGROUND: Chimeric antigen receptor (CAR)-T cell therapies targeting glioblastoma (GBM)-associated antigens such as interleukin-13 receptor subunit alpha-2 (IL-13Rα2) have achieved limited clinical efficacy to date, in part due to an immunosuppressive tumor microenvironment (TME) characterized by inhibitory molecules such as transforming growth factor-beta (TGF-β). The aim of this study was to engineer more potent GBM-targeting CAR-T cells by countering TGF-β-mediated immune suppression in the TME.
    METHODS: We engineered a single-chain, bispecific CAR targeting IL-13Rα2 and TGF-β, which programs tumor-specific T cells to convert TGF-β from an immunosuppressant to an immunostimulant. Bispecific IL-13Rα2/TGF-β CAR-T cells were evaluated for efficacy and safety against both patient-derived GBM xenografts and syngeneic models of murine glioma.
    RESULTS: Treatment with IL-13Rα2/TGF-β CAR-T cells leads to greater T-cell infiltration and reduced suppressive myeloid cell presence in the tumor-bearing brain compared to treatment with conventional IL-13Rα2 CAR-T cells, resulting in improved survival in both patient-derived GBM xenografts and syngeneic models of murine glioma.
    CONCLUSIONS: Our findings demonstrate that by reprogramming tumor-specific T-cell responses to TGF-β, bispecific IL-13Rα2/TGF-β CAR-T cells resist and remodel the immunosuppressive TME to drive potent anti-tumor responses in GBM.
    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