Radiosensitizer

放射增敏剂
  • 文章类型: Journal Article
    胰腺导管腺癌(PDAC)具有低存活率和有限的治疗选择。同步放化疗被认为有利于改善肿瘤控制,但是肿瘤部位的低药物生物利用度和周围健康器官的低辐射耐受性极大地限制了其有效性。碘油,一种用于临床经动脉化疗栓塞的天然药物载体,由于其高Z元素碘组成,已显示出作为放射增敏剂的潜力。因此,本研究旨在重新利用碘油作为增敏剂,同时增强PDAC的化疗和放疗.为此,使用临床批准的表面活性剂设计负载吉西他滨的稳定碘油乳剂(IOE)。在体内水平,IOE显示出比现有的纳米放射增敏剂更好的放射治疗效果,并且比游离药物提高了药物的生物利用度,在同步放化疗的情况下,导致显著的肿瘤抑制和生存率的提高。这可能是由于持续的药物释放,均匀的空间分布,和IOE在实体PDAC肿瘤中的长期保留能力。此外,为了更好地了解载药IOE的作用机制,进行体外研究以揭示与ROS和DNA损伤相关的治疗途径。最后,综合毒性评估也证明了所制备的IOE具有良好的生物相容性和安全性。这项研究为同步放化疗提供了临床上可行的敏化剂,并具有临床上其他类型癌症治疗的潜力。
    Pancreatic ductal adenocarcinoma (PDAC) has a low survival rate and limited treatment options. Concurrent chemoradiotherapy is considered beneficial to improve tumor control, but the low drug bioavailability at tumor site and the low radiation tolerance of surrounding healthy organs greatly limits its effectiveness. Lipiodol, a natural drug carrier used in clinical transarterial chemoembolization, has shown potential as a radiosensitizer due to its high Z element iodine composition. Thus, this study aims to repurpose lipiodol as a sensitizer to simultaneously enhance chemo- and radiotherapy for PDAC. To this end, a stable lipiodol emulsion (IOE) loaded with gemcitabine is designed using clinically approved surfactants. At in vivo level, IOE demonstrates better radiotherapeutic effect than existing nanoradiosensitizers and enhanced drug bioavailability over free drug, leading to significant tumor inhibition and improved survival rates under concurrent chemo-radiotherapy. This may due to the sustained drug release, homogenous spatial distribution, and long-term retention ability of IOE in solid PDAC tumor. Furthermore, to better understand the functioning mechanism of drug-loaded IOE, in vitro study is conducted to reveal the ROS- and DNA damage-related therapeutic pathways. Lastly, a comprehensive toxicity assessment also proves the good biocompatibility and safety of as-prepared IOE. This study offers a clinically feasible sensitizer for simultaneous chemoradiotherapy and holds potential for other types of cancer treatment in clinics.
    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
    癌症,一种在不同社会中流行的疾病,在治疗研究中提出了重大挑战。研究表明,联合疗法是可以帮助有效治疗癌症的方法之一。化疗和放射治疗是主要的癌症治疗方法,在这个项目中,联合放化疗治疗,碳纳米管被用作肿瘤化疗的改良载体,以及制备用于局部放射治疗的辐射敏化剂的基质。在合成CNT-铂-姜黄素纳米颗粒(CNT-Pt-CUR)之后,进行了一系列分析以验证这些纳米颗粒的成功生产。技术,如透射电子显微镜(TEM),动态光散射(DLS),紫外-可见光谱,傅里叶变换红外光谱(FTIR),使用X射线衍射(XRD)。表征数据显示,在棒状CNT上具有8.5nm直径的球形Pt纳米颗粒形态,如通过TEM观察到的。此外,FTIR分析证实药物成功加载到纳米颗粒中,强调这种方法在癌症治疗中的潜力。然后,在正常细胞上使用溶血和(3(-4,5-二甲基噻唑-2-基)-2,5-二苯基四唑(MTT)测试来评估CNT-Pt-CUR纳米颗粒的生物相容性。它还探索了这些纳米颗粒在不同浓度下对癌细胞的抗癌功效,有和没有暴露于X射线。该研究证实了这些纳米颗粒的成功合成,并证明了它们对细胞活力的潜在影响。具体来说,当暴露于纳米粒子和X射线时,乳腺癌细胞表现出更高的毒性敏感性.进一步的分析表明,纳米粒子的毒性是剂量依赖性的,用CUR修饰碳纳米管(CNT)纳米颗粒的表面可显着降低血液毒性。有趣的是,纳米粒子的毒性在X射线的存在下显著放大,提示细胞内DNA损伤和活性氧(ROS)水平增加等机制。
    Cancer, a prevalent disease across various societies, presents a significant challenge in treatment research. Studies show that combination therapies are one of the methods that can help in the effective treatment of cancer. Chemotherapy and radiation therapy are among the main cancer treatments and in this project, for combined chemoradiotherapy treatment, carbon nanotubes were used as improved carriers of chemotherapy in tumors, as well as a substrate for the preparation of radiation sensitizers for local radiation therapy. Following the synthesis of CNT-Platinum-Curcumin nanoparticles (CNT-Pt-CUR), a series of analyses were conducted to verify the successful production of these nanoparticles. Techniques such as Transmission Electron Microscopy (TEM), Dynamic Light Scattering (DLS), UV-Vis spectroscopy, Fourier Transform Infrared Spectroscopy (FTIR), and X-Ray Diffraction (XRD) were employed. The characterization data revealed a spherical shape Pt nanoparticle morphology with an 8.5 nm diameter on rod-shape CNT, as observed through TEM. Furthermore, FTIR analysis confirmed the successful loaded of the drug into the nanoparticles, highlighting the potential of this approach in cancer treatment. Then, hemolysis and (3(-4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) tests on normal cells were used to assess the biocompatibility of CNT-Pt-CUR nanoparticles. It also explored the anticancer efficacy of these nanoparticles at varying concentrations against cancer cells, both with and without exposure to X-rays. The research confirmed the successful synthesis of these nanoparticles and demonstrated their potential impact on cell viability. Specifically, breast cancer cells exhibited heightened susceptibility to toxicity when exposed to nanoparticles and X-rays. Further analysis revealed that the toxicity of nanoparticles is dose-dependent, and modifying the surface of carbon nanotube (CNT) nanoparticles with CUR significantly reduced blood toxicity. Interestingly, nanoparticle toxicity was significantly amplified in the presence of X-rays, suggesting mechanisms such as DNA damage and increased reactive oxygen species (ROS) levels within 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
    放射治疗在癌症治疗中起着至关重要的作用。然而,肿瘤的缺氧微环境极大地限制了疗效,因此,开发一个简单的,高效,和安全的放射增敏剂逆转缺氧和改善放射治疗的疗效。受经典纳米药物Abraxane结构的启发,在这里,开发了通过生物矿化诱导的自组装制备的天然HSA修饰的CaO2纳米颗粒系统(CaO2-HSA)。CaO2-HSA会在肿瘤组织中积聚并分解产生氧气,改变肿瘤内的缺氧状况.同时,ROS和钙离子将导致钙超载并进一步引发免疫原性细胞死亡。值得注意的是,其增敏率(SER=3.47)远高于临床使用的甘氨双唑钠。此外,在原位口腔癌的动物模型中,CaO2-HSA能有效抑制肿瘤生长。凭借其高功效,容易准备,和不含重金属的生物安全,基于CaO2-HSA的放射增敏剂在口腔癌治疗中具有巨大潜力.
    Radiotherapy plays a vital role in cancer therapy. However, the hypoxic microenvironment of tumors greatly limits the effectiveness, thus it is crucial to develop a simple, efficient, and safe radiosensitizer to reverse hypoxia and ameliorate the efficacy of radiotherapy. Inspired by the structure of canonical nanodrug Abraxane, herein, a native HSA-modified CaO2 nanoparticle system (CaO2-HSA) prepared by biomineralization-induced self-assembly is developed. CaO2-HSA will accumulate in tumor tissue and decompose to produce oxygen, altering the hypoxic condition inside the tumor. Simultaneously, ROS and calcium ions will lead to calcium overload and further trigger immunogenic cell death. Notably, its sensitizing enhancement ratio (SER = 3.47) is much higher than that of sodium glycididazole used in the clinic. Furthermore, in animal models of in situ oral cancer, CaO2-HSA can effectively inhibit tumor growth. With its high efficacy, facile preparation, and heavy-metal free biosafety, the CaO2-HSA-based radiosensitizer holds enormous potential for oral cancer therapy.
    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
    放射增敏剂在增强放射治疗(RT)中起关键作用。RT中的挑战之一是纳米放射增敏剂的有限积累和激活抗肿瘤免疫的困难。在这里,使用智能策略实现纳米锰佐剂(MnAuNP-C&B)的原位聚集,以增强RT诱导的抗肿瘤免疫.聚合的MnAuNP-C&B系统克服了小尺寸纳米粒子容易流回血管并扩散到周围组织的缺点,它还延长了纳米锰在癌细胞和肿瘤中的保留时间。MnAuNP-C&B系统显著增强RT中的放射增敏效应。此外,MnAuNP-C&B的pH响应性拆解引发Mn2+的释放,进一步促进RT诱导的STING途径的激活,并引发强大的抗肿瘤免疫。总的来说,我们的研究提出了一个明智的策略,其中纳米锰的原位聚集通过放射增敏和激活抗肿瘤免疫有效抑制肿瘤生长。
    Radiosensitizers play a pivotal role in enhancing radiotherapy (RT). One of the challenges in RT is the limited accumulation of nanoradiosensitizers and the difficulty in activating antitumor immunity. Herein, a smart strategy was used to achieve in situ aggregation of nanomanganese adjuvants (MnAuNP-C&B) to enhance RT-induced antitumor immunity. The aggregated MnAuNP-C&B system overcomes the shortcomings of small-sized nanoparticles that easily flow back into blood vessels and diffuse into surrounding tissues, and it also prolongs the retention time of nanomanganese within cancer cells and tumors. The MnAuNP-C&B system significantly enhances the radiosensitization effect in RT. Additionally, the pH-responsive disassembly of MnAuNP-C&B triggers the release of Mn2+, further promoting RT-induced activation of the STING pathway and eliciting robust antitumor immunity. Overall, our study presents a smart strategy wherein in situ aggregation of nanomanganese effectively inhibits tumor growth through radiosensitization and the activation of antitumor immunity.
    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
    铜-半胱胺纳米颗粒(Cu-CyNP)已成为癌症治疗中有前途的放射增敏剂。本研究旨在探讨使用临床直线加速器的这些纳米颗粒和顺铂的联合治疗效果,以增强宫颈癌放化疗的疗效。在成功合成和表征Cu-CyNPs之后,这些纳米颗粒和顺铂在不同浓度下对HeLa癌细胞的细胞毒性作用进行了评估,单独和组合。此外,在6MV线性加速器下研究了这些药物的放射生物学作用。在浓度为25mg/L时,Cu-CyNP对HeLa癌细胞无明显的细胞毒性。然而,当结合这个浓度的2GyX射线照射时,纳米粒子显示出有效的放射增敏作用。值得注意的是,与单独辐射组相比,联合组(Cu-CyNPs顺铂辐射)的细胞活力和迁移率显着降低。此外,与单独放疗组相比,联合治疗诱导的细胞凋亡率显著更高.总的来说,Cu-CyNPs在X线照射下与顺铂联合时表现出显著的剂量依赖性协同增强的放射功效,并可能为改善常规放射治疗的治疗效果提供有希望的方法。
    Copper-Cysteamine nanoparticles (Cu-Cy NPs) have emerged as promising radiosensitizers in cancer treatment. This study aims to investigate the combined therapeutic effect of these nanoparticles and cisplatin using a clinical linear accelerator to enhance the efficacy of chemoradiation therapy for cervical cancer. Following successful synthesis and characterization of Cu-Cy NPs, the cytotoxicity effect of these nanoparticles and cisplatin in various concentrations was evaluated on HeLa cancer cells, individually and in combination. Additionally, the radiobiological effects of these agents were investigated under a 6MV linear accelerator. At a concentration of 25 mg/L, Cu-Cy NPs displayed no significant cytotoxicity toward HeLa cancer cells. However, when combined with 2Gy X-ray irradiation at this concentration, the nanoparticles demonstrated a potent radiosensitizing effect. Notably, cell viability and migration rate in the combination group (Cu-Cy NPs + cisplatin + radiation) were significantly reduced compared to the radiation-alone group. Additionally, the combination treatment induced a significantly higher rate of apoptosis compared to the radiation-alone group. Overall, Cu-Cy NPs exhibited a significant dose-dependent synergistic enhancement of radiation efficacy when combined with cisplatin under X-ray exposure, and may provide a promising approach to improve the therapeutic effect of conventional radiation therapy.
    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
    放射治疗是许多癌症患者治疗方案的重要组成部分。尽管最近的技术进步,以改善剂量输送技术,由于对周围健康组织不可避免的毒性,增强肿瘤控制所需的剂量递增是有限的。因此,肿瘤部位给药的局部增强可以提供改善治疗方式的必要手段。近年来,纳米技术的出现为提高放射治疗的疗效提供了独特的机会。高原子序数(Z)纳米粒子(NPs)的应用可以通过增加细胞对辐射的敏感性来增强放射治疗的效果。高-ZNP可以固有地充当放射增敏剂以及充当放射增敏剂的靶向递送载体。在这项工作中,高ZNP作为放射增敏剂的治疗益处,例如它们的肿瘤靶向能力和它们的致敏机制,正在讨论。将介绍支持其在放射治疗中应用的临床前数据以及其临床翻译的状况。
    Radiotherapy is an essential component of the treatment regimens for many cancer patients. Despite recent technological advancements to improve dose delivery techniques, the dose escalation required to enhance tumor control is limited due to the inevitable toxicity to the surrounding healthy tissue. Therefore, the local enhancement of dosing in tumor sites can provide the necessary means to improve the treatment modality. In recent years, the emergence of nanotechnology has facilitated a unique opportunity to increase the efficacy of radiotherapy treatment. The application of high-atomic-number (Z) nanoparticles (NPs) can augment the effects of radiotherapy by increasing the sensitivity of cells to radiation. High-Z NPs can inherently act as radiosensitizers as well as serve as targeted delivery vehicles for radiosensitizing agents. In this work, the therapeutic benefits of high-Z NPs as radiosensitizers, such as their tumor-targeting capabilities and their mechanisms of sensitization, are discussed. Preclinical data supporting their application in radiotherapy treatment as well as the status of their clinical translation will be presented.
    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
    放射治疗是癌症的主要治疗方法之一,但它会对正常组织造成损害并导致副作用。使用放射增敏剂可以增强癌细胞对放射的敏感性,从而减少所需的辐射量并最大程度地减少对健康组织的损害。硒化铋纳米颗粒(Bi2Se3NP)已被证明具有作为放射增敏剂的潜力。
    在这项研究中,我们在体外研究了Bi2Se3NP作为结肠癌细胞(HCT-116)放射增敏剂的潜力。用各种浓度的Bi2Se3NP处理细胞,然后暴露于电离辐射。使用MTT测定评估细胞的活力,并评估生存率。
    我们的结果表明,Bi2Se3NP以剂量依赖性方式显着增强了结肠癌细胞对电离辐射的敏感性。与单独的辐射相比,Bi2Se3NP和辐射的组合导致细胞活力和存活率的显著降低。
    Bi2Se3NP具有在结肠癌治疗中用作放射增敏剂的潜力。这项研究的结果表明,将Bi2Se3NPs与放疗相结合可能会增强放疗的有效性并降低与结肠癌相关的死亡率。需要进一步的研究来研究这种方法在体内的安全性和有效性。
    UNASSIGNED: Radiotherapy is one of the primary treatments for cancer, but it can cause damage to normal tissues and lead to side effects. The use of radiosensitizers can enhance the sensitivity of cancer cells to radiation, thereby reducing the amount of radiation required and minimizing damage to healthy tissues. Bismuth selenide nanoparticles (Bi2Se3 NPs) have been shown to have potential as radiosensitizers.
    UNASSIGNED: In this study, we investigated the potential of Bi2Se3 NPs as a radiosensitizer in colon cancer cells (HCT-116) in vitro. The cells were treated with various concentrations of Bi2Se3 NPs and then exposed to ionizing radiation. The viability of the cells was assessed using the MTT assay, and the survival rate was evaluated.
    UNASSIGNED: Our results showed that Bi2Se3 NPs significantly enhanced the sensitivity of colon cancer cells to ionizing radiation in a dose-dependent manner. The combination of Bi2Se3 NPs and radiation resulted in a significant decrease in cell viability and survival rate compared to radiation alone.
    UNASSIGNED: Bi2Se3 NPs have the potential to be used as a radiosensitizer in the treatment of colon cancer. The findings of this study suggest that combining Bi2Se3 NPs with radiation may enhance the effectiveness of radiotherapy and reduce the mortality rate associated with colon cancer. Further studies are needed to investigate the safety and efficacy of this approach in vivo.
    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
    放射疗法和光疗是常用的癌症治疗,其提供诸如副作用的低风险和靶向癌细胞同时保留健康组织的能力的优点。用于癌症治疗的有希望的策略包括使用纳米粒子(NP)与辐射和光热疗法相结合以靶向癌细胞并提高治疗功效。用于生物医学应用的金NP(AuNP)的合成传统上涉及有毒还原剂。在这里,我们利用多巴胺(DA)-缀合的藻酸盐(Alg)来轻松绿色地合成AuNP(Au@Alg-DANP)。Alg-DA共轭还原Au离子,同时稳定了所得的AuNPs,并防止聚集,导致颗粒具有窄的尺寸分布和改善的稳定性。当暴露于X射线时,可注射Au@Alg-DANPs显着促进4T1乳腺癌细胞中ROS的生成。此外,他们的给药在808nm的光激发下升高了温度,从而帮助更有效地消灭癌细胞。重要的是,在我们的Au@Alg-DANP中未检测到明显的细胞毒性。一起来看,我们的工作为获得可注射的无线电增强剂和光热活性纳米系统的组合提供了一个有希望的途径,用于进一步的潜在临床翻译。
    Radiation therapy and phototherapy are commonly used cancer treatments that offer advantages such as a low risk of adverse effects and the ability to target cancer cells while sparing healthy tissue. A promising strategy for cancer treatment involves using nanoparticles (NPs) in combination with radiation and photothermal therapy to target cancer cells and improve treatment efficacy. The synthesis of gold NPs (AuNPs) for use in biomedical applications has traditionally involved toxic reducing agents. Here we harnessed dopamine (DA)-conjugated alginate (Alg) for the facile and green synthesis of Au NPs (Au@Alg-DA NPs). Alg-DA conjugate reduced Au ions, simultaneously stabilized the resulting AuNPs, and prevented aggregation, resulting in particles with a narrow size distribution and improved stability. Injectable Au@Alg-DA NPs significantly promoted ROS generation in 4T1 breast cancer cells when exposed to X-rays. In addition, their administration raised the temperature under a light excitation of 808 nm, thus helping to destroy cancer cells more effectively. Importantly, no substantial cytotoxicity was detected in our Au@Alg-DA NPs. Taken together, our work provides a promising route to obtain an injectable combined radio enhancer and photothermally active nanosystem for further potential clinic translation.
    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
    目的:在肝细胞癌(HCC)治疗中,放射治疗(RT)是一种关键的方法,然而,抗辐射性的出现带来了巨大的挑战。本研究旨在探讨喹硫平与RT治疗HCC的潜在协同作用。
    方法:使用Hep3B异种移植小鼠模型,调查追踪肿瘤进展,安全参数,和分子机制。
    结果:研究结果表明,喹硫平与RT联用时具有协同的抗HCC作用,与对照组相比,延长了肿瘤生长时间和显着更高的生长抑制率。安全性评估表明病理变化很小,提示喹硫平在减轻RT诱导的肝肾功能改变方面的潜力。机械上,该组合抑制了转移和血管生成相关蛋白,同时通过靶向表皮生长因子受体(EGFR)介导的信号传导触发凋亡相关蛋白的激活。
    结论:强调喹硫平和RT联合用药的潜力,提供增强的抗肝癌疗效,安全档案,并将喹硫平定位为肝癌治疗的放射增敏剂。
    OBJECTIVE: In hepatocellular carcinoma (HCC) treatment, radiotherapy (RT) stands as a pivotal approach, yet the emergence of radioresistance poses a formidable challenge. This study aimed to explore the potential synergy between quetiapine and RT for HCC treatment.
    METHODS: A Hep3B xenograft mouse model was used, the investigation tracked tumor progression, safety parameters, and molecular mechanisms.
    RESULTS: The findings revealed a synergistic anti-HCC effect when quetiapine was coupled with RT that prolonged tumor growth time and a significantly higher growth inhibition rate compared to the control group. Safety assessments indicated minimal pathological changes, suggesting potential of quetiapine in mitigating RT-induced alterations in liver and kidney functions. Mechanistically, the combination suppressed metastasis and angiogenesis-related proteins, while triggering the activation of apoptosis-related proteins via targeting Epidermal growth factor receptor (EGFR)-mediated signaling.
    CONCLUSIONS: The potential of the quetiapine and RT combination is emphasized, offering enhanced anti-HCC efficacy, a safety profile, and positioning quetiapine as a radiosensitizer for HCC 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
    检查点激酶1(Chk1)是调节细胞周期进程的DNA损伤反应的关键介质,DNA损伤修复,和DNA复制。小分子Chk1抑制剂可使癌细胞对基因毒性剂敏感,并在以高水平复制应激为特征的癌症中显示出作为单一药物的临床前活性。然而,Chk1抑制剂敏感性的潜在遗传决定因素仍不清楚.尽管晚期结直肠癌的治疗选择有限,放射治疗是有效的。这里,我们报告说,暴露于一种新型的脒衍生物,K1586导致结直肠癌细胞增殖潜力的初始降低。细胞周期分析显示,由于Chk1不稳定,G2/M期的长度随K1586暴露而增加。暴露于K1586以时间和剂量依赖性方式增强了Chk1的降解,增加复制应激和使大肠癌细胞对辐射敏感。一起来看,结果表明,一种新型脒衍生物可能具有作为靶向Chk1的放疗增敏剂的潜力.
    Checkpoint kinase 1 (Chk1) is a key mediator of the DNA damage response that regulates cell cycle progression, DNA damage repair, and DNA replication. Small-molecule Chk1 inhibitors sensitize cancer cells to genotoxic agents and have shown preclinical activity as single agents in cancers characterized by high levels of replication stress. However, the underlying genetic determinants of Chk1-inhibitor sensitivity remain unclear. Although treatment options for advanced colorectal cancer are limited, radiotherapy is effective. Here, we report that exposure to a novel amidine derivative, K1586, leads to an initial reduction in the proliferative potential of colorectal cancer cells. Cell cycle analysis revealed that the length of the G2/M phase increased with K1586 exposure as a result of Chk1 instability. Exposure to K1586 enhanced the degradation of Chk1 in a time- and dose-dependent manner, increasing replication stress and sensitizing colorectal cancer cells to radiation. Taken together, the results suggest that a novel amidine derivative may have potential as a radiotherapy-sensitization agent that targets Chk1.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号