Adoptive cellular therapy

过继细胞疗法
  • 文章类型: Journal Article
    对于复发或难治性高危神经母细胞瘤(HRNB)患者,免疫疗法已成为一种有吸引力的选择。神经母细胞瘤(NB),由胚胎神经c细胞引起的交感神经系统癌症,在临床上是异质的,结果从孤立的腹部肿块自发消退到广泛转移的疾病,尽管进行了强化的多模式治疗,治愈率约为50%。风险组分层和分阶段适应治疗以实现最小毒性的治愈已经完成了重要的里程碑。靶向免疫治疗方法,包括单克隆抗体,疫苗,过继细胞疗法,他们的组合,将它们融入护理标准是有吸引力的治疗选择,尽管治疗挑战和毒性问题仍然存在。在这次审查中,我们提供了在临床转化框架内针对NB和肿瘤微环境(TME)的免疫方法的概述。我们提出了一种新的基于T细胞的治疗方法,该方法利用了肿瘤表面抗原的独特特性,例如神经节苷脂GD2,掺入了特异性单克隆抗体以及过继细胞治疗的最新进展。
    Immunotherapy has emerged as an attractive option for patients with relapsed or refractory high-risk neuroblastoma (HRNB). Neuroblastoma (NB), a sympathetic nervous system cancer arising from an embryonic neural crest cell, is heterogeneous clinically, with outcomes ranging from an isolated abdominal mass that spontaneously regresses to a widely metastatic disease with cure rates of about 50% despite intensive multimodal treatment. Risk group stratification and stage-adapted therapy to achieve cure with minimal toxicities have accomplished major milestones. Targeted immunotherapeutic approaches including monoclonal antibodies, vaccines, adoptive cellular therapies, their combinations, and their integration into standard of care are attractive therapeutic options, although curative challenges and toxicity concerns remain. In this review, we provide an overview of immune approaches to NB and the tumor microenvironment (TME) within the clinical translational framework. We propose a novel T cell-based therapeutic approach that leverages the unique properties of tumor surface antigens such as ganglioside GD2, incorporating specific monoclonal antibodies and recent advancements in adoptive cell therapy.
    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
    CART细胞需要优化才能在实体瘤患者中有效。有许多障碍影响他们成功的能力。一个障碍是坚持,为了达到最佳的抗肿瘤反应,输注的CART细胞必须移植并持续存在。这个独特的变量受到多种因素的影响-CAR-T细胞设计,使用淋巴清除方案,扩增方法产生T细胞产物,还有更多.此外,外部试剂可以用来增加CAR-T细胞,例如添加新的细胞因子,支持记忆形成的药物,或在离体扩增过程中或在CART细胞输注后的其他试剂,以支持它们在压迫性肿瘤微环境中。这篇综述强调了许多用于优化T细胞持久性的策略,以及改善输注细胞持久性的未来方向。
    CAR T cells require optimization to be effective in patients with solid tumors. There are many barriers affecting their ability to succeed. One barrier is persistence, as to achieve an optimal antitumor response, infused CAR T cells must engraft and persist. This singular variable is impacted by a multitude of factors-the CAR T cell design, lymphodepletion regimen used, expansion method to generate the T cell product, and more. Additionally, external agents can be utilized to augment CAR T cells, such as the addition of novel cytokines, pharmaceutical drugs that bolster memory formation, or other agents during either the ex vivo expansion process or after CAR T cell infusion to support them in the oppressive tumor microenvironment. This review highlights many strategies being used to optimize T cell persistence as well as future directions for improving the persistence of infused cells.
    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
    免疫治疗已成为肿瘤治疗的重要组成部分。它在各种癌症类型中的适用性令人印象深刻,以及它利用内生机制来达到预期目的。然而,脱靶或上靶-外肿瘤毒性,活动有限,在联合治疗中缺乏控制,尤其是实体瘤,当地积累低,其临床用途总体有限。通过递送系统部分地缓解了这些限制。基于脂质的纳米颗粒(NPs)由于具有良好的物理化学特性而成为革命性的载体,具有特定的应用和优势,特别适用于免疫治疗剂的递送。这篇综述的目的是强调免疫治疗所面临的挑战,以及基于脂质的NP是如何,并且可以进一步用于应对这些挑战。我们讨论了NPs在一系列领域的最新基础和临床应用,并详细讨论了免疫检查点抑制疗法的主要障碍。过继细胞疗法,和细胞因子疗法。我们强调了基于脂质的纳米系统如何通过两种递送来解决这些问题,直接调节免疫系统,或靶向免疫抑制肿瘤微环境。我们探索用于核酸递送的先进和新兴的脂质体和脂质纳米颗粒(LNP)系统,内在和外在刺激响应配方,和免疫疗法中基于脂质的仿生纳米系统。最后,我们讨论了与临床使用基于脂质的NP免疫疗法有关的关键挑战,建议近期未来的研究方向,以实现这些创新的基于脂质的纳米系统的潜力,因为它们成为必要增强免疫疗法疗效的关键垫脚石。
    Immunotherapy has become an important part of the oncotherapy arsenal. Its applicability in various cancer types is impressive, as well as its use of endogenous mechanisms to achieve desired ends. However, off-target or on-target-off-tumor toxicity, limited activity, lack of control in combination treatments and, especially for solid tumors, low local accumulation, have collectively limited clinical use thereof. These limitations are partially alleviated by delivery systems. Lipid-based nanoparticles (NPs) have emerged as revolutionary carriers due to favorable physicochemical characteristics, with specific applications and strengths particularly useful in immunotherapeutic agent delivery. The aim of this review is to highlight the challenges faced by immunotherapy and how lipid-based NPs have been, and may be further utilized to address such challenges. We discuss recent fundamental and clinical applications of NPs in a range of areas and provide a detailed discussion of the main obstacles in immune checkpoint inhibition therapies, adoptive cellular therapies, and cytokine therapies. We highlight how lipid-based nanosystems could address these through either delivery, direct modulation of the immune system, or targeting of the immunosuppressive tumor microenvironment. We explore advanced and emerging liposomal and lipid nanoparticle (LNP) systems for nucleic acid delivery, intrinsic and extrinsic stimulus-responsive formulations, and biomimetic lipid-based nanosystems in immunotherapy. Finally, we discuss the key challenges relating to the clinical use of lipid-based NP immunotherapies, suggesting future research directions for the near term to realize the potential of these innovative lipid-based nanosystems, as they become the crucial steppingstone towards the necessary enhancement of the efficacy of immunotherapy.
    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
    NKT细胞,独特的淋巴细胞桥接先天和适应性免疫,为控制炎症性疾病如哮喘提供了巨大的潜力。激活iNKT诱导增加的IFN-γ,TGF-β,IL-2和IL-10可能抑制过敏性哮喘。然而,它们的免疫调节作用,包括颗粒酶-穿孔素介导的细胞毒性,TIM-3和TRAIL的表达需要仔细考虑和有针对性的方法。尽管CAR-T细胞疗法在治疗某些癌症方面取得了显著成功,它的局限性需要探索替代方法。在这种情况下,CAR-NKT细胞成为克服这些挑战的一种有希望的方法,有可能实现更安全、更有效的免疫疗法。策略涉及靶向不同的IgE受体及其与CAR-NKT细胞的相互作用,潜在破坏变应原-肥大细胞/嗜碱性粒细胞相互作用并阻止炎性细胞因子释放。此外,靶向免疫检查点,如PDL-2,诱导型ICOS,FASL,CTLA-4和CD137或dectin-1治疗真菌性哮喘可进一步调节免疫反应。此外,人工智能和机器学习为基于NKT细胞的哮喘治疗带来了巨大的变革。AI可以优化CAR-NKT细胞功能,设计个性化的治疗策略,开启精准有效护理的未来.这篇综述讨论了增强CAR-NKT细胞功效和寿命的各种方法,以及它们在过敏性哮喘治疗中面临的挑战和机遇。
    NKT cells, unique lymphocytes bridging innate and adaptive immunity, offer significant potential for managing inflammatory disorders like asthma. Activating iNKT induces increasing IFN-γ, TGF-β, IL-2, and IL-10 potentially suppressing allergic asthma. However, their immunomodulatory effects, including granzyme-perforin-mediated cytotoxicity, and expression of TIM-3 and TRAIL warrant careful consideration and targeted approaches. Although CAR-T cell therapy has achieved remarkable success in treating certain cancers, its limitations necessitate exploring alternative approaches. In this context, CAR-NKT cells emerge as a promising approach for overcoming these challenges, potentially achieving safer and more effective immunotherapies. Strategies involve targeting distinct IgE-receptors and their interactions with CAR-NKT cells, potentially disrupting allergen-mast cell/basophil interactions and preventing inflammatory cytokine release. Additionally, targeting immune checkpoints like PDL-2, inducible ICOS, FASL, CTLA-4, and CD137 or dectin-1 for fungal asthma could further modulate immune responses. Furthermore, artificial intelligence and machine learning hold immense promise for revolutionizing NKT cell-based asthma therapy. AI can optimize CAR-NKT cell functionalities, design personalized treatment strategies, and unlock a future of precise and effective care. This review discusses various approaches to enhancing CAR-NKT cell efficacy and longevity, along with the challenges and opportunities they present in the treatment of allergic asthma.
    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细胞疗法已显示出改善血液恶性肿瘤患者预后的潜力。然而,由于严重的危及生命的毒性,如有限的抗肿瘤疗效,实现血癌的长期完全缓解仍然具有挑战性。抗原逃逸,贩运限制,和有限的肿瘤侵袭。此外,CAR-T细胞与其宿主肿瘤微环境之间的相互作用对CAR-T功能有显著影响。为了克服这些巨大的障碍,需要新的方法和方法来产生更强大的CAR-T细胞,这些细胞具有更大的抗肿瘤活性和更低的毒性.尽管CAR-T研究取得了进展,微生物抗性仍然是一个重大障碍。在这次审查中,我们讨论并描述了CAR-T结构的基础知识,世代,挑战,以及CAR-T细胞治疗中感染的潜在风险。
    Chimeric antigen receptor (CAR)-T cell therapy has shown potential in improving outcomes for individuals with hematological malignancies. However, achieving long-term full remission for blood cancer remains challenging due to severe life-threatening toxicities such as limited anti-tumor efficacy, antigen escape, trafficking restrictions, and limited tumor invasion. Furthermore, the interactions between CAR-T cells and their host tumor microenvironments have a significant impact on CAR-T function. To overcome these considerable hurdles, fresh methodologies and approaches are needed to produce more powerful CAR-T cells with greater anti-tumor activity and less toxicity. Despite advances in CAR-T research, microbial resistance remains a significant obstacle. In this review, we discuss and describe the basics of CAR-T structures, generations, challenges, and potential risks of infections in CAR-T cell therapy.
    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
    增强的细胞疗法已经成为继细胞疗法基础之后的新概念。这种治疗方式应用药物或生物技术来直接增强或遗传修饰细胞以增强过继性细胞疗法(ACT)的功效。增强免疫细胞杀伤能力的药物或生物技术包括免疫检查点抑制剂(ICIs)/抗体药物,小分子抑制剂,免疫调节因子,蛋白水解靶向嵌合体(PROTAC),溶瘤病毒(OV),等。首先,克服抑制性肿瘤微环境(TME)可以增强ACT的疗效,这可以通过阻断免疫检查点来实现。其次,细胞因子或细胞因子受体可以通过基因工程表达或直接添加到过继细胞中,以增强过继细胞向肿瘤细胞的迁移和浸润。此外,多抗原嵌合抗原受体(CAR)可用于增强肿瘤细胞相关抗原的特异性识别,和OVs也可以刺激抗原释放。除了将自杀基因插入过继细胞,PROTAC技术可用作免疫抑制因子的安全开关或降解剂,以增强过继细胞的安全性和有效性。本文全面总结了该机制,目前的情况,增强细胞疗法的临床应用,描述过继细胞疗法的潜在改进。
    Enhanced cellular therapy has emerged as a novel concept following the basis of cellular therapy. This treatment modality applied drugs or biotechnology to directly enhance or genetically modify cells to enhance the efficacy of adoptive cellular therapy (ACT). Drugs or biotechnology that enhance the killing ability of immune cells include immune checkpoint inhibitors (ICIs) / antibody drugs, small molecule inhibitors, immunomodulatory factors, proteolysis targeting chimera (PROTAC), oncolytic virus (OV), etc. Firstly, overcoming the inhibitory tumor microenvironment (TME) can enhance the efficacy of ACT, which can be achieved by blocking the immune checkpoint. Secondly, cytokines or cytokine receptors can be expressed by genetic engineering or added directly to adoptive cells to enhance the migration and infiltration of adoptive cells to tumor cells. Moreover, multi-antigen chimeric antigen receptors (CARs) can be designed to enhance the specific recognition of tumor cell-related antigens, and OVs can also stimulate antigen release. In addition to inserting suicide genes into adoptive cells, PROTAC technology can be used as a safety switch or degradation agent of immunosuppressive factors to enhance the safety and efficacy of adoptive cells. This article comprehensively summarizes the mechanism, current situation, and clinical application of enhanced cellular therapy, describing potential improvements to adoptive cellular therapy.
    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
    尽管免疫疗法取得了有希望的结果,由于从物理屏障到肿瘤外不良反应等几个因素,需要进一步考虑实验.令人惊讶的是过继性细胞免疫疗法,特别是树突状细胞和细胞因子诱导的杀伤(DC-CIK)治疗,在癌症疾病的治疗中远远没有得到重视。DC-CIK治疗癌症患者具有良好的效果,副作用低或无副作用,这一点不容忽视。更有趣的是,几乎所有DC-CIK临床试验都在中国进行,这突出了治疗策略的局限性,需要进行大规模的研究.迄今为止,建议考虑与化疗联合治疗,因为它已显示出有希望的结局和更高的疗效.在这篇文章中,总结了DC-CIK治疗癌症患者的疗效,强调缺乏规模空前的软性癌症实验.简而言之,DC-CIK治疗是一种安全有效的恶性和非恶性疾病治疗剂,可增强短期和长期疗效。
    Despite the promising results of immunotherapy, further experiments need to be considered because of several factors ranging from physical barriers to off-tumor adverse effects. It is surprising that adoptive cellular immunotherapy, particularly dendritic cell and cytokine-induced killer (DC-CIK) therapy, is far less emphasized in the treatment of cancer diseases. DC-CIK therapy in cancer patients presents auspicious results with low or no side effects, which should not be overlooked. More interestingly, almost all DC-CIK clinical trials are ongoing in China that highlight the limitations of therapeutic strategies and require large-scale research. To date, it is advisable to consider combination therapy with chemotherapy since it has shown promising outcomes with higher efficacy. In this article, the efficacy of DC-CIK therapy in patients with cancer is summarized by underscoring the lack of experiments on soft cancers on an unprecedented scale. In brief, DC-CIK therapy is a safe and effective therapeutic agent for malignant and nonmalignant diseases that enhances short-term and long-term effects.
    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
    免疫细胞疗法作为一种革命性的治疗方式,显著改变癌症护理。它是一种特殊形式的免疫疗法,利用活的免疫细胞作为治疗癌症的治疗试剂。与传统药物不同,细胞疗法被认为是“活的药物”,这些产品目前是定制的,需要先进的制造技术。尽管嵌合抗原受体(CAR)-T细胞疗法在血液系统恶性肿瘤的治疗方面在业界受到了极大的关注,它们治疗实体瘤的有效性通常受到限制,导致替代免疫细胞疗法的出现。肿瘤浸润淋巴细胞(TIL)细胞治疗,细胞因子诱导的杀伤(CIK)细胞治疗,树突状细胞(DC)疫苗,和DC/CIK细胞疗法旨在使用人体的自然防御机制来靶向和消除癌细胞,通常副作用或风险较小。另一方面,细胞疗法,如嵌合抗原受体T(CAR-T)细胞,T细胞受体(TCR)-T,嵌合抗原受体-自然杀伤(CAR-NK),或CAR-巨噬细胞(CAR-M)通常利用自体干细胞,同种异体或异种细胞,或转基因细胞,这需要更高水平的操纵,被认为是高风险的。这些高风险细胞疗法通常在肿瘤靶向和信号转导方面具有特殊的特征。引发新的抗肿瘤免疫反应。最近,抗肿瘤机制的基础和临床研究都取得了重大进展,细胞治疗产品设计,和技术创新。凭借快速的技术整合和高度创新的景观,未来重点发展方向已经出现。为了满足治疗癌症的细胞治疗技术进步的需求,本研究全面系统地研究了免疫细胞治疗的技术创新和临床进展。基于免疫细胞疗法的治疗机制和方法学特征,我们分析了与这些疗法相关的主要技术优势和临床转化风险.并对其应用前景进行了分析和预测,为相关企业提供必要的信息,为其研发方向选择做出明智的决策提供参考。
    Immune cell therapy as a revolutionary treatment modality, significantly transformed cancer care. It is a specialized form of immunotherapy that utilizes living immune cells as therapeutic reagents for the treatment of cancer. Unlike traditional drugs, cell therapies are considered \"living drugs,\" and these products are currently customized and require advanced manufacturing techniques. Although chimeric antigen receptor (CAR)-T cell therapies have received tremendous attention in the industry regarding the treatment of hematologic malignancies, their effectiveness in treating solid tumors is often restricted, leading to the emergence of alternative immune cell therapies. Tumor-infiltrating lymphocytes (TIL) cell therapy, cytokine-induced killer (CIK) cell therapy, dendritic cell (DC) vaccines, and DC/CIK cell therapy are designed to use the body\'s natural defense mechanisms to target and eliminate cancer cells, and usually have fewer side effects or risks. On the other hand, cell therapies, such as chimeric antigen receptor-T (CAR-T) cell, T cell receptor (TCR)-T, chimeric antigen receptor-natural killer (CAR-NK), or CAR-macrophages (CAR-M) typically utilize either autologous stem cells, allogeneic or xenogeneic cells, or genetically modified cells, which require higher levels of manipulation and are considered high risk. These high-risk cell therapies typically hold special characteristics in tumor targeting and signal transduction, triggering new anti-tumor immune responses. Recently, significant advances have been achieved in both basic and clinical researches on anti-tumor mechanisms, cell therapy product designs, and technological innovations. With swift technological integration and a high innovation landscape, key future development directions have emerged. To meet the demands of cell therapy technological advancements in treating cancer, we comprehensively and systematically investigate the technological innovation and clinical progress of immune cell therapies in this study. Based on the therapeutic mechanisms and methodological features of immune cell therapies, we analyzed the main technical advantages and clinical transformation risks associated with these therapies. We also analyzed and forecasted the application prospects, providing references for relevant enterprises with the necessary information to make informed decisions regarding their R&D direction selection.
    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
    卵巢癌通常免疫原性差,并且对免疫检查点抑制剂(ICI)疗法的反应令人失望。过继细胞疗法(ACT)提供了一种利用免疫系统的替代方法,特别是嵌合抗原受体T细胞(CAR-T)治疗血液恶性肿瘤的成功。到目前为止,ACT在实体器官恶性肿瘤的治疗中产生了适度的结果。这篇综述探讨了ACT作为卵巢癌当前护理标准的有效替代或额外治疗的可能性。我们将强调青蒿素的潜力,比如CAR-T,T细胞受体疗法(TCR-T),肿瘤浸润淋巴细胞(TIL)和基于细胞的疫苗,同时也讨论了他们的挑战。我们将介绍这些方法在免疫治疗“感冒”卵巢癌中的临床研究,并考虑未来研究的理由。
    Ovarian cancers are typically poorly immunogenic and have demonstrated disappointing responses to immune checkpoint inhibitor (ICI) therapy. Adoptive cellular therapy (ACT) offers an alternative method of harnessing the immune system that has shown promise, especially with the success of chimeric antigen receptor T-cell (CAR-T) therapy in haematologic malignancies. So far, ACT has led to modest results in the treatment of solid organ malignancies. This review explores the possibility of ACT as an effective alternative or additional treatment to current standards of care in ovarian cancer. We will highlight the potential of ACTs, such as CAR-T, T-cell receptor therapy (TCR-T), tumour-infiltrating lymphocytes (TILs) and cell-based vaccines, whilst also discussing their challenges. We will present clinical studies for these approaches in the treatment of immunologically \'cold\' ovarian cancer and consider the rationale for future research.
    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细胞疗法对血液系统恶性肿瘤患者产生了积极影响,但由于运输和肿瘤浸润不足,在实体瘤治疗中显示出有限的疗效。强烈的CAR-T相关毒性,和抗原逃逸。在这项工作中,我们开发并研究了一种可生物降解和生物相容性的聚合物环形螺旋颗粒(TSP),作为体内细胞培养箱和递送装置,可通过微创手术植入肿瘤附近,或通过使用活检针注射到实体瘤附近或实体瘤中.毫米大小的TSP的主要基质结构由明胶甲基丙烯酰胺(GelMA)和聚(乙二醇)二丙烯酸酯(PEGDA)的交联制成,降解速率从几天到几个月可调,提供合适的机械性能和共包封的药物和/或刺激化合物的持续释放。粒子的环形螺旋层,呈现高容量细胞负载的内部空隙体积和共同封装的小和大分子化合物的灵活性,具有单独操纵的释放时间表,充满胶原蛋白和悬浮的T细胞。TSP促进细胞增殖,激活,以及在肿瘤微环境中长期持续的迁移。在这项研究中,在临床前小鼠肿瘤模型中测试了从TSP释放的间皮素(MSLN)CAR-T细胞的功效.与全身注射和肿瘤内注射相比,使用TSP的MSLNCAR-T细胞的瘤周递送导致优异的抗肿瘤效果。由FDA批准的材料制成的TSP作为体内反应器,可以为CAR-T细胞有效地局部递送至实体瘤提供选择,以获得更高的功效和更低的毒性。微创给药程序。
    Chimeric antigen receptor (CAR) T cell therapy has resulted in positive effects on patients with hematologic malignancy but shows limited efficacy in solid tumor treatments due to insufficient trafficking and tumor infiltration, intensive CAR-T-related toxicities, and antigen escape. In this work, we developed and investigated a biodegradable and biocompatible polymeric toroidal-spiral particle (TSP) as a in vivo cell incubator and delivery device that can be implanted near tumor through a minimally invasive procedure or injected near or into solid tumors by using a biopsy needle. The main matrix structure of the millimeter-sized TSP is made from crosslinking of gelatin methacrylamine (GelMA) and poly (ethylene glycol) diacrylate (PEGDA) with a tunable degradation rate from a few days to months, providing appropriate mechanical properties and sustained release of co-encapsulated drugs and/or stimulation compounds. The toroidal-spiral layer of the particles, presenting an internal void volume for high-capacity cell loading and flexibility of co-encapsulating small and large molecular compounds with individually manipulated release schedules, is filled with collagen and suspended T cells. The TSPs promote cell proliferation, activation, and migration in the tumor micro-environment in a prolonged and sustained manner. In this study, the efficacy of mesothelin (MSLN) CAR-T cells released from the TSPs was tested in preclinical mouse tumor models. Compared to systemic and intratumoral injection, peritumoral delivery of MSLN CAR-T cells using the TSPs resulted in a superior antitumor effect. The TSPs made of FDA approved materials as an in vivo reactor may provide an option for efficiently local delivery of CAR-T cells to solid tumors for higher efficacy and lower toxicity, with a minimally invasive administration procedure.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号