cell engineering

细胞工程
  • 文章类型: Journal Article
    一系列重组单克隆抗体(rMAb)已在治疗多种疾病中得到应用,跨越各种癌症和免疫系统疾病。中国仓鼠卵巢(CHO)细胞已成为生产这些rMAb的主要选择,因为它们的鲁棒性,易于转染,以及类似于人类细胞的翻译后修饰能力。可以进行瞬时转染和/或稳定表达以在CHO细胞中表达rMAb。为了增强CHO细胞中rMAb的产量,已经开发了多种方法,包含向量优化,中等配方,栽培参数,和细胞工程。这篇综述简洁地概述了这些方法,同时也解决了生产过程中遇到的挑战,如聚集和岩藻糖基化问题。
    A range of recombinant monoclonal antibodies (rMAbs) have found application in treating diverse diseases, spanning various cancers and immune system disorders. Chinese hamster ovary (CHO) cells have emerged as the predominant choice for producing these rMAbs due to their robustness, ease of transfection, and capacity for posttranslational modifications akin to those in human cells. Transient transfection and/or stable expression could be conducted to express rMAbs in CHO cells. To bolster the yield of rMAbs in CHO cells, a multitude of approaches have been developed, encompassing vector optimization, medium formulation, cultivation parameters, and cell engineering. This review succinctly outlines these methodologies when also addressing challenges encountered in the production process, such as issues with aggregation and fucosylation.
    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
    生命系统包含一个庞大的代谢反应网络,提供大量的酶和细胞作为化学过程的潜在生物催化剂。蛋白质和细胞生物催化剂的特性-高选择性,在环境友好条件下控制反应顺序和操作的能力提供了高效生产分子的方法,同时降低了工业化学的成本和环境影响。此外,生物催化提供了产生化学合成可能无法获得的化学结构和功能的机会。这里我们考虑酶的发展,生物合成途径和细胞工程,使其能够用于催化新的化学和超越。
    Living systems contain a vast network of metabolic reactions, providing a wealth of enzymes and cells as potential biocatalysts for chemical processes. The properties of protein and cell biocatalysts-high selectivity, the ability to control reaction sequence and operation in environmentally benign conditions-offer approaches to produce molecules at high efficiency while lowering the cost and environmental impact of industrial chemistry. Furthermore, biocatalysis offers the opportunity to generate chemical structures and functions that may be inaccessible to chemical synthesis. Here we consider developments in enzymes, biosynthetic pathways and cellular engineering that enable their use in catalysis for new chemistry and beyond.
    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
    工业中国仓鼠卵巢(CHO)细胞系所需的分泌水平可以挑战内质网(ER)稳态,错误折叠蛋白的积累引起的内质网压力可能是生物制造的瓶颈。未折叠的蛋白质反应(UPR)启动以恢复内质网应激的稳态,优化UPR可以提高CHO细胞产生治疗性蛋白。我们比较了分批补料生长,生产特点,和免疫球蛋白G1(IgG1)生产者对其亲本的转录组反应,非生产宿主细胞系。我们使用高通量RNA测序(RNASeq)和定量聚合酶链反应(qPCR)进行了差异基因表达分析,以研究补料分批培养过程中每个细胞系的ER应激反应。与宿主细胞系相比,IgG1生产者中的UPR被激活,我们对差异表达谱的分析表明,IgG1生产者中ATF6α靶mRNA的瞬时上调,建议UPRATF6部门的两个上游监管机构,ATF6β和WFS1是合理的工程目标。虽然ATF6β和WFS1都有负调控ATF6α的报道,这项研究表明,任一靶标的敲除在产生IgG1的CHO细胞系中引起不同的作用。ATF6β的稳定敲低降低了细胞生长而不降低滴度;然而,WFS1的敲低降低了滴度而不影响生长。通过qPCR测量的相对表达表明ATF6β和WFS1表达之间没有直接关系,但是一个池中WFS1的上调与ER伴侣mRNA的生长减少和上调相关。WFS1敲低对UPR激活和产物mRNA表达有负面影响,ATF6β的敲低特别是在补料分批中提高了UPR,从而提高了整体生产率。
    Secretion levels required of industrial Chinese hamster ovary (CHO) cell lines can challenge endoplasmic reticulum (ER) homeostasis, and ER stress caused by accumulation of misfolded proteins can be a bottleneck in biomanufacturing. The unfolded protein response (UPR) is initiated to restore homeostasis in response to ER stress, and optimization of the UPR can improve CHO cell production of therapeutic proteins. We compared the fed-batch growth, production characteristics, and transcriptomic response of an immunoglobulin G1 (IgG1) producer to its parental, non-producing host cell line. We conducted differential gene expression analysis using high throughput RNA sequencing (RNASeq) and quantitative polymerase chain reaction (qPCR) to study the ER stress response of each cell line during fed-batch culture. The UPR was activated in the IgG1 producer compared to the host cell line and our analysis of differential expression profiles indicated transient upregulation of ATF6α target mRNAs in the IgG1 producer, suggesting two upstream regulators of the ATF6 arm of the UPR, ATF6β and WFS1, are rational engineering targets. Although both ATF6β and WFS1 have been reported to negatively regulate ATF6α, this study shows knockdown of either target elicits different effects in an IgG1-producing CHO cell line. Stable knockdown of ATF6β decreased cell growth without decreasing titer; however, knockdown of WFS1 decreased titer without affecting growth. Relative expression measured by qPCR indicated no direct relationship between ATF6β and WFS1 expression, but upregulation of WFS1 in one pool was correlated with decreased growth and upregulation of ER chaperone mRNAs. While knockdown of WFS1 had negative impacts on UPR activation and product mRNA expression, knockdown of ATF6β improved the UPR specifically later in fed-batch leading to increased overall productivity.
    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
    外泌体表现出高生物利用度,生物稳定性,靶向特异性,低毒性,以及穿梭各种生物活性分子如蛋白质的低免疫原性,脂质,RNA,和DNA。天然外泌体,然而,产量有限,瞄准能力,和临床试验中的治疗效果。另一方面,工程外泌体已经证明了长期的循环,高稳定性,有针对性的交付,和有效的细胞内药物释放,引起了极大的关注。工程化的外泌体为开发下一代药物递送系统带来了新的见解,并在治疗应用中显示出巨大的潜力,比如肿瘤治疗,糖尿病管理,心血管疾病,组织再生和修复。在这次审查中,我们通过关注最新的细胞工程和外泌体工程策略,概述了与工程外泌体相关的最新进展。外泌体隔离方法,包括传统和新兴方法,系统比较了表征方法的进步。在工程外泌体的制备和应用方面进一步讨论了当前的挑战和未来的机遇。
    Exosomes exhibit high bioavailability, biological stability, targeted specificity, low toxicity, and low immunogenicity in shuttling various bioactive molecules such as proteins, lipids, RNA, and DNA. Natural exosomes, however, have limited production, targeting abilities, and therapeutic efficacy in clinical trials. On the other hand, engineered exosomes have demonstrated long-term circulation, high stability, targeted delivery, and efficient intracellular drug release, garnering significant attention. The engineered exosomes bring new insights into developing next-generation drug delivery systems and show enormous potential in therapeutic applications, such as tumor therapies, diabetes management, cardiovascular disease, and tissue regeneration and repair. In this review, we provide an overview of recent advancements associated with engineered exosomes by focusing on the state-of-the-art strategies for cell engineering and exosome engineering. Exosome isolation methods, including traditional and emerging approaches, are systematically compared along with advancements in characterization methods. Current challenges and future opportunities are further discussed in terms of the preparation and application of engineered exosomes.
    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
    在人类细胞中目击,细胞内通路和治疗货物运输,包括基因编辑工具(例如,CRISPR-Cas9和转座子),核酸(例如,DNA,mRNA和siRNA),肽,和蛋白质(例如,酶和抗体),被严格限制以确保健康的细胞功能和行为。该原理在用于离体免疫疗法的嵌合抗原受体(CAR)-T细胞的递送机制中示例。特别是,CAR-T细胞的临床成功通过治愈以前无法治愈的血癌建立了新的治疗标准.该方法涉及交付,通常通过使用电穿孔(EP)和慢病毒,治疗性CAR基因进入患者自己的T细胞,然后将其设计为表达靶向和对抗血液癌症的CAR。但关键的困难在于对这些细胞进行基因操纵,而不会造成不可逆转的损害或功能丧失,同时将制造的复杂性降至最低。安全问题,和成本,并确保最终CAR-T细胞产品的功效。纳米注射-使用纳米针(NN)进行细胞内递送的过程-是一种新兴的物理递送途径,可有效地协商许多细胞类型的质膜,包括原代人类T细胞。它以最小的扰动发生,侵入性,和毒性,在高空间和时间分辨率下具有高效率和吞吐量。纳米注射有望大大改善广泛的治疗性货物的递送,而对这些货物几乎没有或没有损害。纳米注射平台允许这些货物根据需要在细胞内空间中发挥作用。纳米注射平台的适应性现在在免疫调节方面带来了主要优势,机械传导,细胞状态采样(纳米活检),受控的细胞内询问,以及该帐户的主要焦点-细胞内递送及其在体外细胞工程中的应用。机械纳米注射通常对细胞膜施加直接的机械力,提供了一条直接的途径来改善NN的膜扰动以及随后将遗传货物运输到目标细胞类型(粘附或悬浮细胞)中。相比之下,通过将NN与电场耦合来控制电活性纳米注射,这是在纳米级激活电穿孔(EP)的新途径,可以显着降低施加给细胞的电压,从而最大程度地减少EP对细胞和货物的损伤。并克服了传统散装EP的许多局限性。纳米注射超越了单纯的技术;它是一种离体细胞工程的方法,提供了赋予细胞新的潜力,强大的功能,例如为未来的CAR-T细胞技术产生嵌合抗原受体(CAR)-T细胞。我们首先讨论神经网络器件的制造(第2节),然后深入研究纳米注射介导的细胞工程(第3节),纳米注射机制和接口方法(第4节),以及使用纳米注射产生功能性CAR-T细胞的新兴应用(第5节)。
    ConspectusIn human cells, intracellular access and therapeutic cargo transport, including gene-editing tools (e.g., CRISPR-Cas9 and transposons), nucleic acids (e.g., DNA, mRNA, and siRNA), peptides, and proteins (e.g., enzymes and antibodies), are tightly constrained to ensure healthy cell function and behavior. This principle is exemplified in the delivery mechanisms of chimeric antigen receptor (CAR)-T cells for ex-vivo immunotherapy. In particular, the clinical success of CAR-T cells has established a new standard of care by curing previously incurable blood cancers. The approach involves the delivery, typically via the use of electroporation (EP) and lentivirus, of therapeutic CAR genes into a patient\'s own T cells, which are then engineered to express CARs that target and combat their blood cancer. But the key difficulty lies in genetically manipulating these cells without causing irreversible damage or loss of function─all the while minimizing complexities of manufacturing, safety concerns, and costs, and ensuring the efficacy of the final CAR-T cell product.Nanoinjection─the process of intracellular delivery using nanoneedles (NNs)─is an emerging physical delivery route that efficiently negotiates the plasma membrane of many cell types, including primary human T cells. It occurs with minimal perturbation, invasiveness, and toxicity, with high efficiency and throughput at high spatial and temporal resolutions. Nanoinjection promises greatly improved delivery of a broad range of therapeutic cargos with little or no damage to those cargos. A nanoinjection platform allows these cargos to function in the intracellular space as desired. The adaptability of nanoinjection platforms is now bringing major advantages in immunomodulation, mechanotransduction, sampling of cell states (nanobiopsy), controlled intracellular interrogation, and the primary focus of this account─intracellular delivery and its applications in ex vivo cell engineering.Mechanical nanoinjection typically exerts direct mechanical force on the cell membrane, offering a straightforward route to improve membrane perturbation by the NNs and subsequent transport of genetic cargo into targeted cell type (adherent or suspension cells). By contrast, electroactive nanoinjection is controlled by coupling NNs with an electric field─a new route for activating electroporation (EP) at the nanoscale─allowing a dramatic reduction of the applied voltage to a cell and so minimizing post-EP damage to cells and cargo, and overcoming many of the limitations of conventional bulk EP. Nanoinjection transcends mere technique; it is an approach to cell engineering ex vivo, offering the potential to endow cells with new, powerful features such as generating chimeric antigen receptor (CAR)-T cells for future CAR-T cell technologies.We first discuss the manufacturing of NN devices (Section 2), then delve into nanoinjection-mediated cell engineering (Section 3), nanoinjection mechanisms and interfacing methodologies (Section 4), and emerging applications in using nanoinjection to create functional CAR-T cells (Section 5).
    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
    基于T细胞的癌症免疫疗法通常依赖于膜结合的细胞毒性增强剂,例如在自体αβT细胞中表达的嵌合抗原受体。这些方法受到合成构建体的强直信号传导和与制造相关的成本的限制。γδT细胞是细胞治疗的一种新兴替代方法,具有先天的抗肿瘤活性,强效的抗体依赖性细胞毒性,和最小的同种异体反应性。我们提出了一种围绕Vγ9Vδ2T细胞的先天特性构建的免疫治疗平台技术,利用这种细胞类型的特定特征,并提供可招募旁观者免疫的同种异体细胞疗法。我们改造了γδT细胞,以scFv-Fc融合蛋白和促有丝分裂IL-15Rα-IL-15融合蛋白(stIL15)的形式分泌合成的肿瘤靶向调理素。使用GD2作为模型抗原,我们表明,分泌GD2特异性调理素的Vγ9Vδ2T细胞(stIL15-OPS-γδT细胞)具有增强的细胞毒性并促进其他淋巴和骨髓细胞的旁观者活性。stIL-15的分泌消除了对外源性细胞因子补充的需要,并进一步介导了旁观者自然杀伤细胞的激活。与未修饰的γδT细胞相比,stIL15-OPS-γδT细胞表现出对皮下肿瘤的体内控制和血液中的持久性。此外,stIL15-OPS-γδT细胞在动物模型和体外对患者来源的骨肉瘤有效,添加唑来膦酸可以提高疗效。一起,数据确定stIL15-OPS-γδT细胞是一种候选的同种异体细胞治疗平台,它将直接细胞溶解与旁观者激活相结合以促进肿瘤控制.
    T cell-based cancer immunotherapy has typically relied on membrane-bound cytotoxicity enhancers such as chimeric antigen receptors expressed in autologous αβ T cells. These approaches are limited by tonic signaling of synthetic constructs and costs associated with manufacturing. γδ T cells are an emerging alternative for cellular therapy, having innate antitumor activity, potent antibody-dependent cellular cytotoxicity, and minimal alloreactivity. We present an immunotherapeutic platform technology built around the innate properties of the Vγ9Vδ2 T cell, harnessing specific characteristics of this cell type and offering an allocompatible cellular therapy that recruits bystander immunity. We engineered γδ T cells to secrete synthetic tumor-targeting opsonins in the form of an scFv-Fc fusion protein and a mitogenic IL-15Rα-IL-15 fusion protein (stIL15). Using GD2 as a model antigen, we show that GD2-specific opsonin-secreting Vγ9Vδ2 T cells (stIL15-OPS-γδ T cells) have enhanced cytotoxicity and promote bystander activity of other lymphoid and myeloid cells. Secretion of stIL-15 abrogated the need for exogenous cytokine supplementation and further mediated activation of bystander natural killer cells. Compared with unmodified γδ T cells, stIL15-OPS-γδ T cells exhibited superior in vivo control of subcutaneous tumors and persistence in the blood. Moreover, stIL15-OPS-γδ T cells were efficacious against patient-derived osteosarcomas in animal models and in vitro, where efficacy could be boosted with the addition of zoledronic acid. Together, the data identify stIL15-OPS-γδ T cells as a candidate allogeneic cell therapy platform combining direct cytolysis with bystander activation to promote tumor control.
    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
    中国仓鼠卵巢(CHO)细胞最广泛地用于产生治疗性抗体。在细胞系发育中,工程分泌过程,如折叠相关蛋白上调是构建具有高重组蛋白生产率的细胞系的有效方法。然而,关于内质网(ER)和高尔基体之间的重组蛋白转运的研究很少。在这项研究中,Sar1A,一种参与COPII囊泡形成的蛋白质,致力于通过增强COPII囊泡介导的抗体从ER到高尔基体的转运来提高抗体生产率,并阐明其对分泌过程的影响。将构建的Sar1A过表达CHO细胞系分批培养,其中它们显示出增加的特异性抗体产生率。通过使用翻译抑制剂的追踪测定研究了细胞内抗体的积累和细胞内抗体的特异性定位,并通过基于免疫荧光的成像分析进行了观察。结果表明,Sar1A过表达减少了细胞内抗体的积累,尤其是在急诊室。通过液相色谱-质谱和UPR相关基因表达评估分析了工程化抗体转运对抗体糖基化谱和未折叠蛋白反应(UPR)途径的影响。分别。Sar1A过表达降低了聚糖半乳糖基化并在分批培养结束时诱导了更强的UPR。Sar1A过表达通过改变CHO细胞的分泌过程来增强其抗体生产力。这种方法不仅有助于单克隆抗体的产生,还有助于需要通过COPII囊泡转运的其他治疗性蛋白质的产生。
    Chinese hamster ovary (CHO) cells are the most widely used for therapeutic antibody production. In cell line development, engineering secretion processes such as folding-related protein upregulation is an effective way of constructing cell lines with high recombinant protein productivity. However, there have been few studies on the transport of recombinant proteins between the endoplasmic reticulum (ER) and the Golgi apparatus. In this study, Sar1A, a protein involved in COPII vesicle formation, was focused on to improve antibody productivity by enhancing COPII vesicle-mediated antibody transport from the ER to the Golgi apparatus, and to clarify its effect on the secretion process. The constructed Sar1A-overexpressing CHO cell lines were batch-cultured, in which they showed an increased specific antibody production rate. The intracellular antibody accumulation and the specific localization of the intracellular antibodies were investigated by chase assay using a translation inhibitor and observed by immunofluorescence-based imaging analysis. The results showed that Sar1A overexpression reduced intracellular antibody accumulation, especially in the ER. The effects of the engineered antibody transport on the antibody\'s glycosylation profile and the unfolded protein response (UPR) pathway were analyzed by liquid chromatography-mass spectrometry and UPR-related gene expression evaluation, respectively. Sar1A overexpression lowered glycan galactosylation and induced a stronger UPR at the end of the batch culture. Sar1A overexpression enhanced the antibody productivity of CHO cells by modifying their secretion process. This approach could also contribute to the production of not only monoclonal antibodies but also other therapeutic proteins that require transport by COPII vesicles.
    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
    紫杉醇,一种从红豆杉树皮中提取的稀有二萜,以其抗癌活性而闻名,并作为治疗癌症的主要药物。由于树皮中紫杉醇的含量异常低,在紫杉醇的生产中使用了一种消耗红豆杉资源的半合成方法,which,然而,无法满足不断升级的临床需求。近年来,研究人员在生产紫杉醇的异源生物合成和代谢工程方面取得了重大进展。本文全面综述了紫杉醇生产的进展,包括化学合成,异源生物合成,和细胞工程。深入介绍了紫杉醇的生物合成途径和转录调控机制,旨在为紫杉醇生物合成的进一步研究提供有价值的参考。
    Paclitaxel, a rare diterpene extracted from the bark of Chinese yew (Taxus chinensis), is renowned for its anti-cancer activity and serves as a primary drug for treating cancers. Due to the exceptionally low content of paclitaxel in the bark, a semi-synthetic method that depletes Chinese yew resources is used in the production of paclitaxel, which, however, fails to meet the escalating clinical demand. In recent years, researchers have achieved significant progress in heterologous biosynthesis and metabolic engineering for the production of paclitaxel. This article comprehensively reviews the advancements in paclitaxel production, encompassing chemical synthesis, heterologous biosynthesis, and cell engineering. It provides an in-depth introduction to the biosynthetic pathway and transcriptional regulation mechanisms of paclitaxel, aiming to provide a valuable reference for further research on paclitaxel biosynthesis.
    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
    本书本章重点介绍了对癌症免疫治疗领域变革性创新的全面探索。CAR(嵌合抗原受体)T细胞疗法代表了通过重新编程患者免疫细胞以识别和破坏癌细胞来治疗癌症的开创性方法。本章强调了合成生物学在增强CAR-T细胞疗法的安全性和有效性方面的关键作用。它首先强调个性化医疗在癌症治疗中日益重要,强调从一刀切的方法到针对患者的解决方案的转变。合成生物学,多学科领域,在定制CAR-T细胞疗法方面发挥了重要作用,允许微调精度和最大限度地减少不必要的副作用。本章重点介绍了基因编辑的最新进展,合成基因回路,和分子工程,展示这些技术如何优化CAR-T细胞功能。总之,本书的这一章阐明了使用合成生物学开发CAR-T细胞疗法的显着进展,为癌症患者提供希望,并暗示未来高度个性化和有效的癌症治疗是常态。
    This book chapter highlights a comprehensive exploration of the transformative innovations in the field of cancer immunotherapy. CAR (Chimeric Antigen Receptor) T-cell therapy represents a groundbreaking approach to treat cancer by reprogramming a patient immune cells to recognize and destroy cancer cells. This chapter underscores the critical role of synthetic biology in enhancing the safety and effectiveness of CAR T-cell therapies. It begins by emphasizing the growing importance of personalized medicine in cancer treatment, emphasizing the shift from one-size-fits-all approaches to patient-specific solutions. Synthetic biology, a multidisciplinary field, has been instrumental in customizing CAR T-cell therapies, allowing for fine-tuned precision and minimizing unwanted side effects. The chapter highlights recent advances in gene editing, synthetic gene circuits, and molecular engineering, showcasing how these technologies are optimizing CAR T-cell function. In summary, this book chapter sheds light on the remarkable progress made in the development of CAR T-cell therapies using synthetic biology, providing hope for cancer patients and hinting at a future where highly personalized and effective cancer treatments are the norm.
    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