CTCs

CTC
  • 文章类型: Journal Article
    背景:循环肿瘤细胞(CTC)在肿瘤转移中至关重要,然而它们在肾癌中的具体作用尚不清楚.
    方法:本研究利用生物信息学研究了C-C基序趋化因子配体5(CCL5)对肾癌细胞和CTC的致瘤影响,在体内,和体外实验。它还通过Lasso回归和Kaplan-Meier存活曲线评估肾癌患者CTC的预后价值。
    结果:生物信息学分析揭示了关注CTC和肿瘤细胞之间的细胞粘附和迁移的差异基因。CCL5在各种CTC中表现出高表达,与肾癌预后不良有关。在786-O-CTC中,CCL5增强恶性肿瘤,而在肾细胞癌细胞系CAKI-2和786-O中,它通过smad2/3促进上皮-间质转化(EMT),影响细胞特性。裸鼠模型提示CCL5增加CTC和增强EMT,增强肺转移。临床结果显示不同EMT型CTC的预后值不同,间充质CTC具有最高的价值。
    结论:总之,CCL5通过smad2/3促进肾癌细胞和CTC的EMT,增强恶性表型并促进肺转移。间充质型CTC相关因子可构建肾癌患者的风险模型,允许基于转移风险预测的个性化治疗。
    BACKGROUND: Circulating tumor cells (CTCs) are pivotal in tumor metastasis across cancers, yet their specific role in renal cancer remains unclear.
    METHODS: This study investigated C-C motif chemokine ligand 5 (CCL5)\'s tumorigenic impact on renal cancer cells and CTCs using bioinformatics, in vivo, and in vitro experiments. It also assessed renal cancer patients\' CTCs prognostic value through Lasso regression and Kaplan-Meier survival curves.
    RESULTS: Bioinformatics analysis revealed differential genes focusing on cellular adhesion and migration between CTCs and tumor cells. CCL5 exhibited high expression in various CTCs, correlating with poor prognosis in renal cancer. In 786-O-CTCs, CCL5 enhanced malignancy, while in renal cell carcinoma cell line CAKI-2 and 786-O, it promoted epithelial-mesenchymal transition (EMT) via smad2/3, influencing cellular characteristics. The nude mouse model suggested CCL5 increased CTCs and intensified EMT, enhancing lung metastasis. Clinical results shown varying prognostic values for different EMT-typed CTCs, with mesenchymal CTCs having the highest value.
    CONCLUSIONS: In summary, CCL5 promoted EMT in renal cancer cells and CTCs through smad2/3, enhancing the malignant phenotype and facilitating lung metastasis. Mesenchymal-type CTC-related factors can construct a risk model for renal cancer patients, allowing personalized treatment based on metastatic risk prediction.
    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
    肿瘤转移,癌症进展的顶点,在治疗努力中构成了巨大的挑战。循环肿瘤细胞(CTC),源自原发性肿瘤或其转移的弹性实体,通过展示显著的适应性,对这一过程做出了显著贡献。它们经受住了剪切应力,抵抗anoikis,逃避免疫监视,阻止化疗。这篇全面的综述旨在阐明CTC形成的复杂景观,转移机制,以及影响他们行为的无数因素。整合信号通路,例如整合素相关的信号,细胞自噬,上皮-间质转化,以及与血小板的相互作用,进行了详细的检查。此外,我们探索精密纳米医学设计的领域,特别强调anoikis-血小板界面。这种创新方法战略性地针对CTC生存机制,提供了有希望的途径,以前所未有的精度和效力对抗转移性癌症。该综述强调了基于血小板的纳米药物的合理设计在抑制CTC驱动的转移中不可或缺的作用。
    Tumor metastasis, the apex of cancer progression, poses a formidable challenge in therapeutic endeavors. Circulating tumor cells (CTCs), resilient entities originating from primary tumors or their metastases, significantly contribute to this process by demonstrating remarkable adaptability. They survive shear stress, resist anoikis, evade immune surveillance, and thwart chemotherapy. This comprehensive review aims to elucidate the intricate landscape of CTC formation, metastatic mechanisms, and the myriad factors influencing their behavior. Integral signaling pathways, such as integrin-related signaling, cellular autophagy, epithelial-mesenchymal transition, and interactions with platelets, are examined in detail. Furthermore, we explore the realm of precision nanomedicine design, with a specific emphasis on the anoikis‒platelet interface. This innovative approach strategically targets CTC survival mechanisms, offering promising avenues for combatting metastatic cancer with unprecedented precision and efficacy. The review underscores the indispensable role of the rational design of platelet-based nanomedicine in the pursuit of restraining CTC-driven metastasis.
    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
    在血液或其他生理液体中,“循环细胞和亚细胞生物颗粒”包括许多微观生物元素,如循环肿瘤细胞(CTC),无细胞DNA(cfDNA),外泌体,microRNAs,血小板,免疫细胞,和蛋白质是最知名和研究的。这些结构是医疗保健和医学研究中的关键生物标志物,用于早期发现癌症和其他疾病。使治疗能够在临床症状出现之前开始,并提高治疗效果。随着这些待检测的生物标志物的大小减少,它们在体液中的数量减少,检测材料,从视觉检查到先进的显微镜技术,开始变小,更敏感,更快,更有效,得益于纳米技术的发展.这篇综述首先定义了循环细胞和亚细胞生物颗粒及其生物,物理,和机械性能,其次关注它们的诊断重要性,包括他们作为生物标志物的最新应用,用来检测它们的生物传感器,目前必须克服的障碍,以及该领域的未来发展。随着技术的进步和生物分子途径的深入,诊断测试将变得更加敏感,具体,和彻底。最后,将循环细胞和生物颗粒的诊断应用的最新进展整合到临床实践中,对于精准医学和患者预后是有希望的。
    In the bloodstream or other physiological fluids, \"circulating cells and sub-cellular bio-particles\" include many microscopic biological elements such as circulating tumor cells (CTCs), cell-free DNA (cfDNA), exosomes, microRNAs, platelets, immune cells, and proteins are the most well-known and investigated. These structures are crucial biomarkers in healthcare and medical research for the early detection of cancer and other disorders, enabling treatment to commence before the onset of clinical symptoms and enhancing the efficacy of treatments. As the size of these biomarkers to be detected decreases and their numbers in body fluids diminishes, the detection materials, ranging from visual inspection to advanced microscopy techniques, begin to become smaller, more sensitive, faster, and more effective, thanks to developing nanotechnology. This review first defines the circulating cells and subcellular bio-particles with their biological, physical, and mechanical properties and second focuses on their diagnostic importance, including their most recent applications as biomarkers, the biosensors that are utilized to detect them, the present obstacles that must be surmounted, and prospective developments in the domain. As technology advances and biomolecular pathways are deepens, diagnostic tests will become more sensitive, specific, and thorough. Finally, integrating recent advances in the diagnostic use of circulating cells and bioparticles into clinical practice is promising for precision medicine and patient outcomes.
    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
    确定性横向位移(DLD)微流体装置基于由微柱阵列产生的流线工作。支柱的配置改变了这些装置的隔离效率。本文优化了用于分离可变形循环肿瘤细胞的DLD设备的性能。输入变量包括细胞直径(d),杨氏模量(Es${E}_s$),雷诺数(Re),和tanθ,其中θ是微柱的倾斜角。输出,这是系统的反应,是DLD。数值模拟结果采用响应面法对装置进行优化,导致将DLD估计为输入变量的函数的相关性的命题。证明了对细胞横向位移的最大和最小影响对应于Es${E}_s$和Re,分别。
    Deterministic lateral displacement (DLD) microfluidic devices work based on the streamlines created by an array of micro-posts. The configuration of pillars alters the isolation efficiency of these devices. The present paper optimizes the performance of a DLD device for isolating deformable circulating tumor cells. The input variables include cell diameter (d), Young\'s modulus ( E s ${E}_s$ ), Reynolds number (Re), and tan θ, where θ is the tilted angle of micro-posts. The output, which is the response of the system, is DLD. The numerical simulation results are employed to optimize the device using the response surface method, leading to the proposition of a correlation to estimate DLD as a function of input variables. It is demonstrated that the maximum and minimum impacts on cell lateral displacement correspond to E s ${E}_s$ and Re, respectively.
    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
    手术后早期预测或检测结直肠癌(CRC)复发的能力使医生能够应用适当的治疗计划和不同的随访策略来提高患者的生存率。总的来说,30-50%的CRC患者在根治性手术后经历癌症复发,但目前的监测工具在癌症复发的精确和早期检测方面存在局限性.循环肿瘤细胞(CTC)是从原发性肿瘤分离并进入血流的癌细胞。这些可以提供有关疾病状态的实时信息。CTC可能成为预测CRC复发的新标志物,更重要的是,决定额外的辅助化疗。在这次审查中,描述了CTC作为II期CRC治疗标志物的临床应用.然后讨论了CTC用于监测接受新辅助放化疗的晚期直肠癌患者的癌症复发的实用性。最后,探讨了CTC亚型和CTC结合临床病理因素在建立预测CRC复发的多标志物模型中的作用。
    The ability to predict or detect colorectal cancer (CRC) recurrence early after surgery enables physicians to apply appropriate treatment plans and different follow-up strategies to improve patient survival. Overall, 30-50% of CRC patients experience cancer recurrence after radical surgery, but current surveillance tools have limitations in the precise and early detection of cancer recurrence. Circulating tumor cells (CTCs) are cancer cells that detach from the primary tumor and enter the bloodstream. These can provide real-time information on disease status. CTCs might become novel markers for predicting CRC recurrence and, more importantly, for making decisions about additional adjuvant chemotherapy. In this review, the clinical application of CTCs as a therapeutic marker for stage II CRC is described. It then discusses the utility of CTCs for monitoring cancer recurrence in advanced rectal cancer patients who undergo neoadjuvant chemoradiotherapy. Finally, it discusses the roles of CTC subtypes and CTCs combined with clinicopathological factors in establishing a multimarker model for predicting CRC recurrence.
    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
    循环肿瘤细胞(CTC),源自原发肿瘤并携带遗传信息,对肿瘤转移的进程有显著的贡献。CTCs的分析和检测可用于评估肿瘤患者的预后和治疗反应,以及帮助研究肿瘤的转移机制和新药的开发。由于血液中的CTC非常罕见,有效地丰富CTC是一个具有挑战性的问题。在本文中,我们提供了近年来用于CTC的基于微流体的富集装置的全面概述。我们详细探讨了基于CTC的物理或生物学特性的富集方法;其中,物理性质包括尺寸等因素,密度,和介电性能,而生物学特性主要与CTC表面的肿瘤特异性标志物有关。此外,我们对单个CTC的富集方法进行了深入的描述,并说明了单个CTC对进行肿瘤分析的重要性。未来的研究将集中在提高分离效率等方面。降低成本,提高了检测的灵敏度和准确度。
    Circulating tumor cells (CTCs), derived from the primary tumor and carrying genetic information, contribute significantly to the process of tumor metastasis. The analysis and detection of CTCs can be used to assess the prognosis and treatment response in patients with tumors, as well as to help study the metastatic mechanisms of tumors and the development of new drugs. Since CTCs are very rare in the blood, it is a challenging problem to enrich CTCs efficiently. In this paper, we provide a comprehensive overview of microfluidics-based enrichment devices for CTCs in recent years. We explore in detail the methods of enrichment based on the physical or biological properties of CTCs; among them, physical properties cover factors such as size, density, and dielectric properties, while biological properties are mainly related to tumor-specific markers on the surface of CTCs. In addition, we provide an in-depth description of the methods for enrichment of single CTCs and illustrate the importance of single CTCs for performing tumor analyses. Future research will focus on aspects such as improving the separation efficiency, reducing costs, and increasing the detection sensitivity and accuracy.
    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
    循环肿瘤细胞(CTC)是一种从主要肿瘤扩散到血液的癌细胞,它们通常是可以从血液中分离出来的各种实体中最重要的。对于癌症的诊断,传统的活检通常是侵入性的和不可靠的,而液体活检,将受影响的物品与血液或淋巴液隔离,是一种侵入性较小且有效的诊断技术。微流体技术为进行液体活检提供了合适的通道,该技术用于通过基于物理和生物亲和力的技术在微流控芯片中提取CTC。这项工作在独特的微流体芯片中使用功能化的磁性纳米颗粒(MNPs),以使用具有高捕获率的混合(基于物理和生物亲和力的/引导磁性)捕获方法来收集CTC。因此,叶酸功能化的Fe3O4纳米粒子已用于捕获MCF-7(乳腺癌)CTC,在10µL/min的流速下捕获效率高达95%。此外,已经进行了研究来支持这一说法,包括模拟和仿生研究。
    Circulating tumor cells (CTCs) are a type of cancer cell that spreads from the main tumor to the bloodstream, and they are often the most important among the various entities that can be isolated from the blood. For the diagnosis of cancer, conventional biopsies are often invasive and unreliable, whereas a liquid biopsy, which isolates the affected item from blood or lymph fluid, is a less invasive and effective diagnostic technique. Microfluidic technologies offer a suitable channel for conducting liquid biopsies, and this technology is utilized to extract CTCs in a microfluidic chip by physical and bio-affinity-based techniques. This effort uses functionalized magnetic nanoparticles (MNPs) in a unique microfluidic chip to collect CTCs using a hybrid (physical and bio-affinity-based/guided magnetic) capturing approach with a high capture rate. Accordingly, folic acid-functionalized Fe3O4 nanoparticles have been used to capture MCF-7 (breast cancer) CTCs with capture efficiencies reaching up to 95% at a 10 µL/min flow rate. Moreover, studies have been conducted to support this claim, including simulation and biomimetic investigations.
    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
    尽管循环肿瘤细胞(CTC)在液体活检中已证明相当重要,它们的检测受到低浓度和复杂样品成分的限制。在这里,我们开发了一种同质的,简单,和靶向乳腺癌细胞的高灵敏度策略。该方法基于从全血中分离CTC的非免疫逐步离心预处理方法。通过适体与乳腺癌细胞的过表达的粘蛋白1(MUC1)和人表皮生长因子受体2(HER2)蛋白的特异性结合来实现精确定量。随后,在胆固醇堆积DNA机器上启动DNA酶裂解和平行催化发夹组装(CHA)反应,打开发夹结构T-Hg2+-T和C-Ag+-C,实现多个扩增。这通过释放的离子导致来自Hg2+特异性碳点(CD)和CdTe量子点(QD)的荧光信号减少。该策略证明了检测极限(LOD)为3个细胞/mL和线性范围为5-100个细胞/mL的检测性能。已经验证了42个临床样本,证实其与临床影像学的一致性,病理结果和叶酸受体(FR)-PCR试剂盒结果,表现出100%的特异性和80.6%的灵敏度。这些结果突出了我们的方法用于诊断和监测乳腺癌的有希望的适用性。
    Although circulating tumor cells (CTCs) have demonstrated considerable importance in liquid biopsy, their detection is limited by low concentrations and complex sample components. Herein, we developed a homogeneous, simple, and high-sensitivity strategy targeting breast cancer cells. This method was based on a non-immunological stepwise centrifugation preprocessing approach to isolate CTCs from whole blood. Precise quantification is achieved through the specific binding of aptamers to the overexpressed mucin 1 (MUC1) and human epidermal growth factor receptor 2 (HER2) proteins of breast cancer cells. Subsequently, DNAzyme cleavage and parallel catalytic hairpin assembly (CHA) reactions on the cholesterol-stacking DNA machine were initiated, which opened the hairpin structures T-Hg2+-T and C-Ag+-C, enabling multiple amplifications. This leads to the fluorescence signal reduction from Hg2+-specific carbon dots (CDs) and CdTe quantum dots (QDs) by released ions. This strategy demonstrated a detection performance with a limit of detection (LOD) of 3 cells/mL and a linear range of 5-100 cells/mL. 42 clinical samples have been validated, confirming their consistency with clinical imaging, pathology findings and the folate receptor (FR)-PCR kit results, exhibiting desirable specificity of 100% and sensitivity of 80.6%. These results highlight the promising applicability of our method for diagnosing and monitoring breast cancer.
    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
    这篇叙述性综述旨在全面概述上皮癌患者循环肿瘤细胞(CTC)分析的现状及其临床意义。这篇综述探讨了CTC检测方法的进步,它们的临床应用,以及未来的挑战。通过考察这一领域的重要研究成果,这篇综述为读者理解CTC分析的演变格局及其对临床实践的潜在影响奠定了坚实的基础.CTC的综合分析为肿瘤生物学提供了有价值的见解,治疗反应,微小残留病检测,和预后评估。此外,该综述强调了CTC作为个性化医疗和治疗疗效监测的非侵入性生物标志物的潜力.尽管反恐委员会的研究取得了进展,一些挑战,如标准化,验证,与常规临床实践的整合仍然存在。该综述最后讨论了CTC分析对改善患者预后和指导上皮癌治疗决策的未来方向和潜在影响。
    This narrative review aims to provide a comprehensive overview of the current state of circulating tumor cell (CTC) analysis and its clinical significance in patients with epithelial cancers. The review explores the advancements in CTC detection methods, their clinical applications, and the challenges that lie ahead. By examining the important research findings in this field, this review offers the reader a solid foundation to understand the evolving landscape of CTC analysis and its potential implications for clinical practice. The comprehensive analysis of CTCs provides valuable insights into tumor biology, treatment response, minimal residual disease detection, and prognostic evaluation. Furthermore, the review highlights the potential of CTCs as a non-invasive biomarker for personalized medicine and the monitoring of treatment efficacy. Despite the progress made in CTC research, several challenges such as standardization, validation, and integration into routine clinical practice remain. The review concludes by discussing future directions and the potential impact of CTC analysis on improving patient outcomes and guiding therapeutic decision-making in epithelial 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
    将肿瘤蛋白质标记物的检测与循环肿瘤细胞(CTC)的捕获相结合代表了用于早期肿瘤检测的超有前景的方法。然而,目前的方法还没有达到必要的低检测限和有效的捕获。这里,我们引入了一种新型的聚吡咯纳米触角传感平台,该平台具有海葵样结构,能够同时检测蛋白质生物标志物和捕获CTC.纳米触角的掺入显着增强了电极表面积,为抗体结合提供丰富的活性位点。这种增强允许以2.39和3.12pg/mL的检出限检测核基质蛋白22(NMP22)和膀胱肿瘤抗原(BTA),分别。此外,我们开发的传感平台有效捕获血液样品中的MCF-7细胞,检测限小于10个细胞/mL,归因于特异性识别抗体和纳米触角表面上的正电荷促进的协同多价结合。该传感平台展示了出色的检测能力和出色的捕获效率,提供一个简单的,准确,和有效的早期肿瘤检测策略。本文受版权保护。保留所有权利。
    Combining the detection of tumor protein markers with the capture of circulating tumor cells (CTCs) represents an ultra-promising approach for early tumor detection. However, current methodologies have not yet achieved the necessary low detection limits and efficient capture. Here, a novel polypyrrole nanotentacles sensing platform featuring anemone-like structures capable of simultaneously detecting protein biomarkers and capturing CTCs is introduced. The incorporation of nanotentacles significantly enhances the electrode surface area, providing abundant active sites for antibody binding. This enhancement allows detecting nucleus matrix protein22 and bladder tumor antigen with 2.39 and 3.12 pg mL-1 detection limit, respectively. Furthermore, the developed sensing platform effectively captures MCF-7 cells in blood samples with a detection limit of fewer than 10 cells mL-1, attributed to the synergistic multivalent binding facilitated by the specific recognition antibodies and the positive charge on the nanotentacles surface. This sensing platform demonstrates excellent detection capabilities and outstanding capture efficiency, offering a simple, accurate, and efficient strategy for early tumor detection.
    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