Tumor-treating fields

肿瘤治疗领域
  • 文章类型: Journal Article
    根据世界卫生组织的数据,癌症仍然是致命疾病的最前沿。全球观察到癌症发病率和死亡率呈上升趋势,强调应继续努力开发检测和治疗方法。诊断路径通常从学习患者的病史开始;随后是基本的血液检查和成像测试,以指示癌症可能位于何处,以安排穿刺活检。迅速开始诊断至关重要,因为延迟的癌症检测需要更高的治疗和住院成本。因此,需要新的癌症检测方法,如液体活检,弹性成像,合成生物传感器,荧光成像,和反射共聚焦显微镜。常规治疗方法,虽然在临床实践中仍然很常见,存在许多局限性,并且不能令人满意。如今,临床研究的动态发展和更精确和有效的方法的发展,如溶瘤病毒疗法,基于外泌体的治疗,纳米技术,树突状细胞,嵌合抗原受体,免疫检查点抑制剂,基于天然产品的治疗,肿瘤治疗领域,和光动力疗法。本文比较了有关常规和现代癌症检测和治疗方法的可用数据,以促进对这一迅速发展的领域及其未来方向的理解。事实证明,现代方法并非没有缺点;仍然需要开发新的检测策略和治疗方法来提高灵敏度,特异性,安全,和功效。然而,已经采取了适当的路线,食品和药物管理局批准了一些现代方法。
    In accordance with the World Health Organization data, cancer remains at the forefront of fatal diseases. An upward trend in cancer incidence and mortality has been observed globally, emphasizing that efforts in developing detection and treatment methods should continue. The diagnostic path typically begins with learning the medical history of a patient; this is followed by basic blood tests and imaging tests to indicate where cancer may be located to schedule a needle biopsy. Prompt initiation of diagnosis is crucial since delayed cancer detection entails higher costs of treatment and hospitalization. Thus, there is a need for novel cancer detection methods such as liquid biopsy, elastography, synthetic biosensors, fluorescence imaging, and reflectance confocal microscopy. Conventional therapeutic methods, although still common in clinical practice, pose many limitations and are unsatisfactory. Nowadays, there is a dynamic advancement of clinical research and the development of more precise and effective methods such as oncolytic virotherapy, exosome-based therapy, nanotechnology, dendritic cells, chimeric antigen receptors, immune checkpoint inhibitors, natural product-based therapy, tumor-treating fields, and photodynamic therapy. The present paper compares available data on conventional and modern methods of cancer detection and therapy to facilitate an understanding of this rapidly advancing field and its future directions. As evidenced, modern methods are not without drawbacks; there is still a need to develop new detection strategies and therapeutic approaches to improve sensitivity, specificity, safety, and efficacy. Nevertheless, an appropriate route has been taken, as confirmed by the approval of some modern methods by the Food and Drug Administration.
    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
    肿瘤治疗场(TTFields)使用中频和低强度电场来抑制肿瘤细胞。然而,他们的机制仍然没有得到很好的理解。本文综述了它们在细胞和分子水平上的关键抗肿瘤机制。包括抑制增殖,死亡的诱导,迁徙的干扰,和免疫系统的激活。还总结了与其他癌症治疗相结合的多方面生物学效应。对其机制的深入了解将有助于开发更多潜在的抗肿瘤治疗方法。
    Tumor-Treating Fields (TTFields) use intermediate-frequency and low-intensity electric fields to inhibit tumor cells. However, their mechanisms are still not well understood. This article reviews their key antitumor mechanisms at the cellular and molecular levels, including inhibition of proliferation, induction of death, disturbance of migration, and activation of the immune system. The multifaceted biological effects in combination with other cancer treatments are also summarized. The deep insight into their mechanism will help develop more potential antitumor treatments.
    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: English Abstract
    Tumor-treating fields (TTFields) is a novel treatment modality for malignant solid tumors, often employing electric field simulations to analyze the distribution of electric fields on the tumor under different parameters of TTFields. Due to the present difficulties and high costs associated with reproducing or implementing the simulation model construction techniques, this study used readily available open-source software tools to construct a highly accurate, easily implementable finite element simulation model for TTFields. The accuracy of the model is at a level of 1 mm 3. Using this simulation model, the study carried out analyses of different factors, such as tissue electrical parameters and electrode configurations. The results show that factors influncing the distribution of the internal electric field of the tumor include changes in scalp and skull conductivity (with a maximum variation of 21.0% in the treatment field of the tumor), changes in tumor conductivity (with a maximum variation of 157.8% in the treatment field of the tumor), and different electrode positions and combinations (with a maximum variation of 74.2% in the treatment field of the tumor). In summary, the results of this study validate the feasibility and effectiveness of the proposed modeling method, which can provide an important reference for future simulation analyses of TTFields and clinical applications.
    肿瘤电场治疗是一种新的治疗恶性实体肿瘤的疗法,多采用电场仿真分析不同参数的肿瘤电场治疗在瘤体上产生的电场分布。但目前,由于仿真模型构建技术路线较难复现或实现成本较高,因此本研究采用常用的软件工具建立了易实现的高精度肿瘤电场治疗有限元仿真模型,模型精度在1 mm 3水平。本研究利用该仿真模型开展了不同组织电学参数、电极配置等因素分析,研究结果显示,对肿瘤内电场分布存在影响的因素包括:头皮、头骨电导率的变化(瘤体治疗场最高变化21.0%)、肿瘤电导率的变化(瘤体治疗场最高变化157.8%)和不同的电极位置及组合(瘤体治疗场最高变化74.2%)。综上,本文研究结果验证了所提建模方法的可行性与有效性,或可为今后的肿瘤电场治疗仿真分析和临床应用提供重要参考。.
    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
    肿瘤治疗领域(TTFields)目前是美国国家综合癌症中心针对新诊断的胶质母细胞瘤患者的1A类治疗建议。虽然已经部分阐明了TTFields的作用机制,实际的和标准化的指标是缺乏评估抗肿瘤剂量和治疗效果。本文概述并评估了估算大脑中TTFields分布和剂量测量的当前标准和方法,并强调了控制TTFields剂量测定的最重要原则。重点是临床实用性,以促进对这些原理的实际理解以及如何将其用于指导治疗。目前TTFields剂量之间存在相关性的证据,肿瘤生长,和临床结果将介绍和讨论。此外,我们将通过回顾TTField的剂量和热效应如何受到肿瘤位置和形态等因素的影响,提供对TTField治疗胶质母细胞瘤的计划和优化的观点和最新见解。瘤周水肿,电极阵列位置,治疗持续时间(依从性),数组“边缘效应”,\"电气占空比,和颅骨重塑手术.最后,就如何优化未来TTFields疗法的疗效提供了观点。
    Tumor-treating fields (TTFields) are currently a Category 1A treatment recommendation by the US National Comprehensive Cancer Center for patients with newly diagnosed glioblastoma. Although the mechanism of action of TTFields has been partly elucidated, tangible and standardized metrics are lacking to assess antitumor dose and effects of the treatment. This paper outlines and evaluates the current standards and methodologies in the estimation of the TTFields distribution and dose measurement in the brain and highlights the most important principles governing TTFields dosimetry. The focus is on clinical utility to facilitate a practical understanding of these principles and how they can be used to guide treatment. The current evidence for a correlation between TTFields dose, tumor growth, and clinical outcome will be presented and discussed. Furthermore, we will provide perspectives and updated insights into the planning and optimization of TTFields therapy for glioblastoma by reviewing how the dose and thermal effects of TTFields are affected by factors such as tumor location and morphology, peritumoral edema, electrode array position, treatment duration (compliance), array \"edge effect,\" electrical duty cycle, and skull-remodeling surgery. Finally, perspectives are provided on how to optimize the efficacy of future TTFields 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
    血脑屏障(BBB)对脑肿瘤的药物递送提出了重大挑战,大多数化学疗法对非恶性脑组织的渗透性有限,并且仅限制了原发性和转移性脑癌的进入。因此,由于药物不能有效地穿透血脑屏障,脑部化疗后的结局仍然不理想.已经提出了几种方法来打开BBB并在肿瘤中获得更高的药物浓度,根据目标肿瘤体积的大小选择最佳方法,选择的治疗剂,和个体患者特征。在这里,我们的目标是全面描述动脉内药物给药的渗透破坏,鞘内/脑室内给药,激光间质热疗,对流增强输送,和超声方法,包括高强度聚焦和低强度超声以及肿瘤治疗领域。我们解释了每种方法背后的科学概念,临床前/临床研究,优点和缺点,适应症,和潜在的改进途径。鉴于每种方法都有其局限性,BBB破坏的未来不太可能依赖于单一方法,而是依赖于联合方法的协同作用.渗透输注或高强度聚焦超声破坏血脑屏障,然后动脉内输送药物,是一种有希望的方法。为了获得最佳结果,必须对药物输送进行实时监测。
    The blood-brain barrier (BBB) poses a significant challenge to drug delivery for brain tumors, with most chemotherapeutics having limited permeability into non-malignant brain tissue and only restricted access to primary and metastatic brain cancers. Consequently, due to the drug\'s inability to effectively penetrate the BBB, outcomes following brain chemotherapy continue to be suboptimal. Several methods to open the BBB and obtain higher drug concentrations in tumors have been proposed, with the selection of the optimal method depending on the size of the targeted tumor volume, the chosen therapeutic agent, and individual patient characteristics. Herein, we aim to comprehensively describe osmotic disruption with intra-arterial drug administration, intrathecal/intraventricular administration, laser interstitial thermal therapy, convection-enhanced delivery, and ultrasound methods, including high-intensity focused and low-intensity ultrasound as well as tumor-treating fields. We explain the scientific concept behind each method, preclinical/clinical research, advantages and disadvantages, indications, and potential avenues for improvement. Given that each method has its limitations, it is unlikely that the future of BBB disruption will rely on a single method but rather on a synergistic effect of a combined approach. Disruption of the BBB with osmotic infusion or high-intensity focused ultrasound, followed by the intra-arterial delivery of drugs, is a promising approach. Real-time monitoring of drug delivery will be necessary for optimal 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: Case Reports
    我们介绍了一名31岁的女性患者,患有胸椎原发性多形性胶质母细胞瘤(GBM),大约在2020年中期诊断。她的症状是几个月前出现右脚感觉异常,从她的右下肢远端到近端进展为神经病变。几个月来,她出现腰胸痛,这是头颅放射。她的疼痛随着活动而恶化。胸椎MRI显示从T10-T11椎间盘水平到T12-L1椎间盘水平存在异常硬膜内增强的焦点,在脐带内产生大量水肿。她接受了全面的手术切除。根据组织病理学,患者患有WHOIV级脊髓GBM。患者接受辅助同步放疗和替莫唑胺化疗。她继续进行替莫唑胺的维护以及对脊柱的Novocure交变电场治疗的同情使用。她受到一个多专业团队的密切监视。放疗后32个月,她的病情稳定,没有进展的证据。她在步行和症状方面取得了显着改善。虽然GBM最常见的是颅内,原发性脊柱GBM相对罕见。尽管存在针对幕上GBM的既定治疗指南,脊柱GBM的治疗方案选择仍然存在争议,但通常反映用于颅内GBM的方案,包括手术,放射治疗,和化疗。交变电场治疗,也被称为肿瘤治疗场(TTFields),适用于颅内GBM的辅助治疗。虽然需要进一步研究脊髓GBM中的TTFields,TTField似乎是脊柱GBM的安全辅助治疗方法。仍需要进一步的研究旨在寻找脊柱GBM的改进治疗方法。
    We present a 31-year-old female patient with primary glioblastoma multiforme (GBM) of the thoracic spine, diagnosed in approximately mid-2020. Her symptoms began several months prior with right foot paresthesia, which progressed to neuropathy ascending from her distal to proximal right lower extremity. Over several months, she developed lumbo-thoracic throbbing pain, which was dermatomal radiating anteriorly. Her pain worsened with activity. A thoracic spine MRI showed a focus of abnormal intradural intramedullary enhancement present from the T10-T11 disc level to the T12-L1 disc level, producing a large amount of edema within the cord. She underwent a gross total surgical resection. The patient had WHO Grade IV spinal GBM per histopathology. The patient received adjuvant concurrent radiation therapy and temozolomide chemotherapy. She continues with maintenance temozolomide along with the compassionate use of Novocure alternating electrical field therapy for the spine. She is being monitored closely by a multi-specialty team. At 32 months post-radiation therapy, her disease is stable with no evidence of progression. She has made significant improvements in her ambulation and symptoms. While GBM is most commonly intracranial, primary spinal GBM is relatively rare. Although established treatment guidelines exist for supratentorial GBM, treatment protocol choices for spinal GBM remain controversial but typically mirror those used for intracranial GBM and include surgery, radiation therapy, and chemotherapy. Alternating electrical field therapy, also known as tumor-treating fields (TTFields), is indicated for adjuvant treatment of intracranial GBM. While further studies of TTFields in spinal GBM are needed, TTFields appear to be a safe adjunct treatment for spinal GBM. Further studies are still needed aimed at finding an improved treatment for spinal GBM.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • DOI:
    文章类型: Journal Article
    创新的方法已经产生了一种在4.9个月内治疗新诊断的GBM癌症患者的方法。在III期临床试验期间,中位总生存期(OS)改善,副作用最小。这种方法被称为肿瘤治疗场(TTFields)。这项研究的目的是确定TTFields治疗通过增强TTFields诱导的衰老来敏感GBM癌细胞的潜力。为了实现这一点,这项研究采用了包含几个要素的多方面方法,包括SA-β-gal染色的分析,流式细胞术,西方印迹,形态学评估,正电子发射断层扫描(PET)/计算机断层扫描(CT),免疫组织化学染色,和微量测定。在长达5天的时间里,表现出衰老特异性形态和SA-β-Gal活性阳性的细胞数量逐渐增加。这些发现表明p16,p21,p27和pRB是通过激活NF-κB诱导的TTField衰老的关键调节因子。结果表明,TTFields治疗通过独立于凋亡的机制有效促进了TTFields诱导的GBM细胞衰老。总之,这项研究强调了这种治疗方法作为解决常规GBM治疗相关局限性的可靠方案的可行性.
    Innovative approaches have given rise to a method for treating newly diagnosed GBM cancer patients within a span of 4.9 months, resulting in improved median overall survival (OS) and minimal side effects during the phase III clinical trial. This approach is referred to as Tumor Treating Fields (TTFields). The objective of this study is to ascertain the potential of TTFields treatment in sensitizing GBM cancer cells by enhancing TTFields-induced senescence. To achieve this, the research employed a multifaceted methodology that encompassed several elements, including the analysis of SA-β-gal staining, flow cytometry, Western blotting, morphology assessment, Positron Emission Tomography (PET)/Computed Tomography (CT), immunohistochemical staining, and microassay. Over a period of up to 5 days, the number of cells exhibiting senescence-specific morphology and positive SA-β-Gal activity progressively increased. These findings indicate that p16, p21, p27 and pRB are pivotal regulators of TTFields-induced senescence through NF-κB activation. The outcomes reveal that TTFields treatment effectively promotes TTFields-induced senescence in GBM cells through a mechanism independent of apoptosis. In conclusion, this research underscores the viability of this treatment approach as a reliable protocol to address the limitations associated with the conventional GBM treatment.
    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
    肿瘤内细胞间通讯的中断正在成为癌症定向治疗的一种新的潜在策略。肿瘤治疗场(TTFields)疗法是一种治疗方式,在过去十年中已出现在胶质母细胞瘤和恶性间皮瘤患者的积极临床应用中,基于使用低强度交变电场破坏正在进行有丝分裂的癌细胞中的微管的原理。需要鉴定这种治疗方法的其他细胞和分子效应,这些效应可以解释当将TTFields添加到标准全身性药物中时报告的总生存率增加。隧道纳米管(TNT)是细胞接触依赖性丝状肌动蛋白基细胞突起,可以在远距离连接两个或多个细胞。它们在癌症中上调,促进细胞生长,分化,在浸润性癌症表型的情况下,更具化学抗性的表型。为了确定TNTs是否为TTFields提供潜在的治疗靶点,我们将TTFields应用于恶性胸膜间皮瘤(MPM)细胞在体外形成TNTs。1.0V/cm的TTField显着抑制了双相亚型MPM中的TNT形成,但不是肉瘤样MPM,独立于对细胞数量的影响。通过测量经由完整的TNT的线粒体货物的细胞间转运来评估,TTField没有显著影响TNT的功能。我们进一步利用空间转录组学方法来使用MPM的动物模型在体内表征TTField诱导的分子谱变化。我们发现TTFields诱导免疫肿瘤学生物标志物的上调,同时下调与细胞过度增殖相关的通路,入侵,和其他致癌生长的关键调节剂。几个分子类别和途径与我们和其他人发现在癌细胞TNTs中差异表达的标记一致。具体包括MPM。我们在选择为空间基因组评估的感兴趣区域的致密基质肿瘤材料中可视化短TNT。将来自空间基因组学的这些感兴趣区域叠加在完整组织中成像的TNT簇的平面上是一种新方法,我们将其指定为隧道纳米管的空间剖面(SPOTT)。总之,这些结果将TNTs定位为针对TTFelds的癌症治疗策略的潜在治疗靶点.我们还确定了TTFelds在分子水平上重塑肿瘤微环境景观的能力,从而提出了一种潜在的新策略,用于将细胞水平的肿瘤从“冷”转变为“热”,以对免疫治疗药物产生潜在反应。
    Disruption of intercellular communication within tumors is emerging as a novel potential strategy for cancer-directed therapy. Tumor-Treating Fields (TTFields) therapy is a treatment modality that has itself emerged over the past decade in active clinical use for patients with glioblastoma and malignant mesothelioma, based on the principle of using low-intensity alternating electric fields to disrupt microtubules in cancer cells undergoing mitosis. There is a need to identify other cellular and molecular effects of this treatment approach that could explain reported increased overall survival when TTFields are added to standard systemic agents. Tunneling nanotube (TNTs) are cell-contact-dependent filamentous-actin-based cellular protrusions that can connect two or more cells at long-range. They are upregulated in cancer, facilitating cell growth, differentiation, and in the case of invasive cancer phenotypes, a more chemoresistant phenotype. To determine whether TNTs present a potential therapeutic target for TTFields, we applied TTFields to malignant pleural mesothelioma (MPM) cells forming TNTs in vitro. TTFields at 1.0 V/cm significantly suppressed TNT formation in biphasic subtype MPM, but not sarcomatoid MPM, independent of effects on cell number. TTFields did not significantly affect function of TNTs assessed by measuring intercellular transport of mitochondrial cargo via intact TNTs. We further leveraged a spatial transcriptomic approach to characterize TTFields-induced changes to molecular profiles in vivo using an animal model of MPM. We discovered TTFields induced upregulation of immuno-oncologic biomarkers with simultaneous downregulation of pathways associated with cell hyperproliferation, invasion, and other critical regulators of oncogenic growth. Several molecular classes and pathways coincide with markers that we and others have found to be differentially expressed in cancer cell TNTs, including MPM specifically. We visualized short TNTs in the dense stromatous tumor material selected as regions of interest for spatial genomic assessment. Superimposing these regions of interest from spatial genomics over the plane of TNT clusters imaged in intact tissue is a new method that we designate Spatial Profiling of Tunneling nanoTubes (SPOTT). In sum, these results position TNTs as potential therapeutic targets for TTFields-directed cancer treatment strategies. We also identified the ability of TTFields to remodel the tumor microenvironment landscape at the molecular level, thereby presenting a potential novel strategy for converting tumors at the cellular level from \'cold\' to \'hot\' for potential response to immunotherapeutic drugs.
    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:胶质母细胞瘤(GBM)是成人最常见的原发性恶性脑肿瘤。尽管付出了巨大的研究努力,GBM仍然是一种致命的疾病。根据国家癌症综合癌症网络(NCCN),新诊断为GBM的患者的标准护理治疗是最大程度的安全手术切除,然后进行同步放化疗和维持替莫唑胺(TMZ)以及辅助肿瘤治疗领域(TTF)。TTF是一种非药物干预,提供低强度,中频交变电场,通过破坏有丝分裂纺锤体来阻止细胞增殖。TTF已在一项大型临床试验中显示,当添加到放疗和化疗中时,可以改善患者的预后。SPARE试验(保留头皮的辐射,同时替莫唑胺和肿瘤治疗场)评估了在放疗和化疗中同时添加TTF。
    方法:本研究是对SPARE试验的探索性分析,旨在探讨常见GBM分子改变的预后意义,即MGMT,EGFR,TP53,PTEN和端粒酶逆转录酶(TERT),在接受TTF联合放疗和化疗的患者队列中。
    结果:如预期,在该队列中,MGMT启动子甲基化与改善的总生存期(OS)和无进展生存期(PFS)相关。此外,TERT启动子突变也与改善的OS和PFS相关。
    结论:利用GBM的分子特征以及推进TTF放化疗等治疗方法为改善GBM患者的精确肿瘤学和预后提供了新的机会。
    BACKGROUND: Glioblastoma (GBM) is the most common primary malignant brain tumor in adults. Despite enormous research efforts, GBM remains a deadly disease. The standard-of-care treatment for patients with newly diagnosed with GBM as per the National Cancer Comprehensive Cancer Network (NCCN) is maximal safe surgical resection followed by concurrent chemoradiation and maintenance temozolomide (TMZ) with adjuvant tumor treating fields (TTF). TTF is a non-pharmacological intervention that delivers low-intensity, intermediate frequency alternating electric fields that arrests cell proliferation by disrupting the mitotic spindle. TTF have been shown in a large clinical trial to improve patient outcomes when added to radiation and chemotherapy. The SPARE trail (Scalp-sparing radiation with concurrent temozolomide and tumor treating fields) evaluated adding TTF concomitantly to radiation and chemotherapy.
    METHODS: This study is an exploratory analysis of the SPARE trial looking at the prognostic significance of common GBM molecular alterations, namely MGMT, EGFR, TP53, PTEN and telomerase reverse transcriptase (TERT), in this cohort of patients treated with concomitant TTF with radiation and chemotherapy.
    RESULTS: As expected, MGMT promoter methylation was associated with improved overall survival (OS) and progression-free survival (PFS) in this cohort. In addition, TERT promoter mutation was associated with improved OS and PFS in this cohort as well.
    CONCLUSIONS: Leveraging the molecular characterization of GBM alongside advancing treatments such as chemoradiation with TTF presents a new opportunity to improve precision oncology and outcomes for GBM patients.
    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
    脂肪肉瘤(LPS)是一种罕见的软组织肉瘤,占成人所有肉瘤病例的20%。人LPS的有效治疗方案没有明确定义。肿瘤治疗领域(TTFields)是抗肿瘤治疗的新领域。TTField联合放化疗已被证明比TTField联合放疗或单独化疗更有效。本研究旨在评估TTField在抑制细胞增殖和活力方面的有效性,以用于LPS的抗癌治疗。本研究使用了TTFields(频率,150kHz;强度,1.0V/cm)处理两种LPS细胞系(94T778和SW872),并分析了抗肿瘤作用。根据锥虫蓝和MTT检测结果,TTField显着降低了LPS细胞系的活力和增殖,并在三维培养中形成了集落。根据Transwell室试验,TTFields处理还显著降低了LPS细胞的迁移。此外,如在Caspase-3活性测定中更高的caspase-3活化和活性氧(ROS)测定的结果所示,TTFields增加了细胞中ROS的形成并增加了凋亡细胞的比例。本研究还研究了TTField与多柔比星(DOX)组合对肿瘤细胞迁移能力的抑制作用。结果表明,TTFields处理协同诱导了ROS诱导的LPS癌细胞系凋亡,并抑制了其迁移行为。总之,本研究证明了TTField在提高LPS癌细胞敏感性方面的潜力,这可能为该联合治疗策略的未来临床试验奠定基础。
    Liposarcoma (LPS) is a rare type of soft tissue sarcoma that constitutes 20% of all sarcoma cases in adults. Effective therapeutic protocols for human LPS are not well-defined. Tumor-treating fields (TTFields) are a novel and upcoming field for antitumor therapy. TTFields combined with chemoradiotherapy have proven to be more effective than TTFields combined with radiotherapy or chemotherapy alone. The present study aimed to assess the effectiveness of TTFields in inhibiting cell proliferation and viability for the anticancer treatment of LPS. The present study used TTFields (frequency, 150 kHz; intensity, 1.0 V/cm) to treat two LPS cell lines (94T778 and SW872) and analyzed the antitumor effects. According to trypan blue and MTT assay results, TTFields markedly reduced the viability and proliferation of LPS cell lines along with the formation of colonies in three-dimensional culture. Based on the Transwell chamber assay, TTFields treatment also markedly reduced the migration of LPS cells. Furthermore, as shown by the higher activation of caspase-3 in the Caspase-3 activity assay and the results of the reactive oxygen species (ROS) assay, TTFields increased the formation of ROS in the cells and enhanced the proportion of apoptotic cells. The present study also investigated the inhibitory effect of TTFields in combination with doxorubicin (DOX) on the migratory capacity of tumor cells. The results demonstrated that TTFields treatment synergistically induced the ROS-induced apoptosis of LPS cancer cell lines and inhibited their migratory behavior. In conclusion, the present study demonstrated the potential of TTFields in improving the sensitivity of LPS cancer cells, which may lay the foundation for future clinical trials of this combination treatment strategy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号