Cancer radiotherapy

癌症放射治疗
  • 文章类型: Journal Article
    放射治疗是一种公认的细胞毒性治疗局部实体癌,利用高能电离辐射摧毁癌细胞。然而,这种方法有几个限制,包括低辐射能量沉积,对周围正常细胞的严重损害,和高肿瘤对辐射的抵抗力。在各种放射治疗方法中,硼中子俘获疗法(BNCT)已成为提高恶性肿瘤治疗率和降低对周围正常组织的致死率的主要方法。但它仍然缺乏硼积累不足以及保留时间短,这限制了疗效。最近,已经开发了一系列可以选择性积累在癌细胞特定细胞器中的放射增敏剂,以精确靶向放射治疗,从而减少正常组织损伤的副作用,克服辐射抗性,提高放射敏感性。在这次审查中,我们主要关注基于纳米医学的癌症放射治疗领域,并讨论了细胞器靶向放射增敏剂,特别包括细胞核,线粒体,内质网和溶酶体。此外,特别介绍了BNCT中使用的细胞器靶向硼载体。通过展示细胞器靶向放射增敏的最新进展,我们希望为临床癌症治疗提供细胞器靶向放射增敏剂的设计。
    Radiotherapy is a well-established cytotoxic therapy for local solid cancers, utilizing high-energy ionizing radiation to destroy cancer cells. However, this method has several limitations, including low radiation energy deposition, severe damage to surrounding normal cells, and high tumor resistance to radiation. Among various radiotherapy methods, boron neutron capture therapy (BNCT) has emerged as a principal approach to improve the therapeutic ratio of malignancies and reduce lethality to surrounding normal tissue, but it remains deficient in terms of insufficient boron accumulation as well as short retention time, which limits the curative effect. Recently, a series of radiosensitizers that can selectively accumulate in specific organelles of cancer cells have been developed to precisely target radiotherapy, thereby reducing side effects of normal tissue damage, overcoming radioresistance, and improving radiosensitivity. In this review, we mainly focus on the field of nanomedicine-based cancer radiotherapy and discuss the organelle-targeted radiosensitizers, specifically including nucleus, mitochondria, endoplasmic reticulum and lysosomes. Furthermore, the organelle-targeted boron carriers used in BNCT are particularly presented. Through demonstrating recent developments in organelle-targeted radiosensitization, we hope to provide insight into the design of organelle-targeted radiosensitizers for clinical cancer 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
    N6-甲基腺苷(M6A)修饰是真核生物中RNA分子最常见的内部化学修饰。这种修饰可以影响mRNA代谢,调节RNA转录,核出口,拼接,降解,翻译,并显著影响生理和病理生物学的各个方面。放射治疗是最常见的肿瘤治疗方法。不同的内在细胞机制影响细胞对电离辐射(IR)的反应和癌症放疗的有效性。在这次审查中,我们总结并讨论了RNAM6A甲基化在细胞对放射诱导的DNA损伤的反应中的作用和机制以及在确定癌症放疗结局方面的最新进展。对放射生物学中RNAM6A甲基化的见解可能有助于改善癌症放射治疗的治疗策略和正常组织的放射防护。
    The N6-methyladenosine (M6A) modification is the most common internal chemical modification of RNA molecules in eukaryotes. This modification can affect mRNA metabolism, regulate RNA transcription, nuclear export, splicing, degradation, and translation, and significantly impact various aspects of physiology and pathobiology. Radiotherapy is the most common method of tumor treatment. Different intrinsic cellular mechanisms affect the response of cells to ionizing radiation (IR) and the effectiveness of cancer radiotherapy. In this review, we summarize and discuss recent advances in understanding the roles and mechanisms of RNA M6A methylation in cellular responses to radiation-induced DNA damage and in determining the outcomes of cancer radiotherapy. Insights into RNA M6A methylation in radiation biology may facilitate the improvement of therapeutic strategies for cancer radiotherapy and radioprotection of normal tissues.
    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(RT)引发免疫原性细胞死亡(ICD)。L-ASNase,催化天冬酰胺(Asn)的转化,从而耗尽它,用于治疗血癌。在以前的工作中,我们发现CRT3LP和CRT4LP,与钙网蛋白(CRT)特异性单体CRT3和CRT4缀合的PAS化L-ASN酶增加了ICD诱导化疗的功效。这里,我们评估了它们在接受RT治疗的荷瘤小鼠中的疗效。方法:通过计算机分子对接分析评估单体结合。通过共聚焦成像和流式细胞术评估ecto-CRT的表达和细胞定位。通过ELISA分析CRT3LP和CRT4LP在辐射(IR)诱导的肿瘤ICD中的抗肿瘤作用和作用。免疫组织化学,免疫分析方法。结果:分子对接分析表明CRT3和CRT4单体与CRT稳定结合。暴露于10GyIR以时间依赖性方式降低CT-26和MC-38肿瘤细胞的活力,直至72h,并增加ICD标记物ecto-CRT(暴露在细胞表面的CRT)和免疫检查点标记物PD-L1的表达,直到48小时。IR增强了CT-26和MC-38肿瘤细胞中CRT3LP和CRT4LP的细胞毒性,和增加活性氧(ROS)水平。在用6GyIR治疗的携带CT-26和MC-38皮下肿瘤的小鼠中,Rluc8缀合的CRT特异性单体(CRT3-Rluc8和CRT4-Rluc8)特异性靶向肿瘤组织,CRT3LP和CRT4LP增加了肿瘤组织中的总ROS水平,从而增强RT的抗肿瘤功效。这些小鼠的肿瘤组织显示成熟树突状细胞增多,CD4+T,和CD8+T细胞和促炎细胞因子(IFNγ和TNFα)和减少的调节性T细胞,肿瘤细胞增殖标志物(Ki67和CD31)表达下调。这些数据表明IR和CRT靶向L-ASN酶的组合激活并重新编程肿瘤微环境的免疫系统。与这些数据一致,免疫检查点抑制剂(抗PD-L1抗体)显著提高了联合IR和CRT靶向L-ASNase的疗效.结论:CRT特异性L-ASNases可用作RT治疗肿瘤的候选药物。与抗PD-L1抗体的联合治疗增加了它们的治疗功效。
    Radiotherapy (RT) triggers immunogenic cell death (ICD). L-ASNase, which catalyzes the conversion of asparagine (Asn), thereby depleting it, is used in the treatment of blood cancers. In previous work, we showed that CRT3LP and CRT4LP, PASylated L-ASNases conjugated to the calreticulin (CRT)-specific monobodies CRT3 and CRT4, increase the efficacy of ICD-inducing chemotherapy. Here, we assessed their efficacy in tumor-bearing mice treated with RT. Methods: Monobody binding was evaluated by in silico molecular docking analysis. The expression and cellular localization of ecto-CRT were assessed by confocal imaging and flow cytometry. The antitumor effect and the roles of CRT3LP and CRT4LP in irradiation (IR)-induced ICD in tumors were analyzed by ELISA, immunohistochemistry, and immune analysis methods. Results: Molecular docking analysis showed that CRT3 and CRT4 monobodies were stably bound to CRT. Exposure to 10 Gy IR decreased the viability of CT-26 and MC-38 tumor cells in a time-dependent manner until 72 h, and increased the expression of the ICD marker ecto-CRT (CRT exposed on the cell surface) and the immune checkpoint marker PD-L1 until 48 h. IR enhanced the cytotoxicity of CRT3LP and CRT4LP in CT-26 and MC-38 tumor cells, and increased reactive oxygen species (ROS) levels. In mice bearing CT-26 and MC-38 subcutaneous tumors treated with 6 Gy IR, Rluc8-conjugated CRT-specific monobodies (CRT3-Rluc8 and CRT4-Rluc8) specifically targeted tumor tissues, and CRT3LP and CRT4LP increased total ROS levels in tumor tissues, thereby enhancing the antitumor efficacy of RT. Tumor tissues from these mice showed increased mature dendritic, CD4+ T, and CD8+ T cells and pro-inflammatory cytokines (IFNγ and TNFα) and decreased regulatory T cells, and the expression of tumor cell proliferation markers (Ki67 and CD31) was downregulated. These data indicate that the combination of IR and CRT-targeting L-ASNases activated and reprogramed the immune system of the tumor microenvironment. Consistent with these data, an immune checkpoint inhibitor (anti-PD-L1 antibody) markedly increased the therapeutic efficacy of combined IR and CRT-targeting L-ASNases. Conclusion: CRT-specific L-ASNases are useful as additive drug candidates in tumors treated with RT, and combination treatment with anti-PD-L1 antibody increases their therapeutic efficacy.
    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
    细胞内转胞吞作用可以增强纳米药物对深部无血管肿瘤组织的渗透,但是可以改善胞吞作用的策略是有限的。在这项研究中,我们发现,真形细胞外基质(ECM)是一种屏障,可损害纳米颗粒的内吞作用及其在相邻细胞之间的运动,从而限制其在肿瘤中的活跃转胞吞作用。我们进一步证明了ECM关键成分的降解(即,胶原蛋白)有效增强纳米颗粒的细胞内胞吞作用。具体来说,胶原酶偶联的胞吞纳米颗粒(Col-TNP)可以解离成胶原酶和阳离子化的金纳米颗粒,以响应肿瘤酸度,这使得它们的ECM篡改能力和在肿瘤中的主动胞吞作用。ECM的断裂进一步增强了阳离子化纳米颗粒进入深部肿瘤组织的活性胞吞作用以及胰腺腺癌的放射增敏功效。我们的研究开辟了新的途径,以增强纳米药物的主动胞吞作用,用于治疗癌症和其他疾病。
    Intracellular transcytosis can enhance the penetration of nanomedicines to deep avascular tumor tissues, but strategies that can improve transcytosis are limited. In this study, we discovered that pyknomorphic extracellular matrix (ECM) is a shield that impairs endocytosis of nanoparticles and their movement between adjacent cells and thus limits their active transcytosis in tumors. We further showed that degradation of pivotal constituent of ECM (i.e., collagen) effectively enhances intracellular transcytosis of nanoparticles. Specifically, a collagenase conjugating transcytosis nanoparticle (Col-TNP) can dissociate into collagenase and cationized gold nanoparticles in response to tumor acidity, which enables their ECM tampering ability and active transcytosis in tumors. The breakage of ECM further enhances the active transcytosis of cationized nanoparticles into deep tumor tissues as well as radiosensitization efficacy of pancreatic adenocarcinoma. Our study opens up new paths to enhance the active transcytosis of nanomedicines for the treatment of cancers and other diseases.
    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(RT),作为临床肿瘤治疗的主要方法之一,已应用于大多数实体瘤的治疗。然而,肿瘤缺氧环境的抗辐射性和高剂量辐射引起的非特异性损伤损害了RT的效果。硫属元素铋(Bi2X3,X=S,Se)基纳米药物由于具有高的光电效应和优异的生物相容性,作为高效放射增敏剂受到了广泛的关注。更重要的是,特别设计的纳米复合材料可以有效缓解肿瘤组织的抗辐射能力。这里,第一次,我们系统地总结了Bi2X3纳米药物通过缓解缺氧肿瘤微环境来增强RT的最新进展。这些新兴的Bi2X3纳米药物主要包括三个方面,具有高效O2供应的Bi2X3纳米复合材料,非O2依赖性Bi2X3纳米复合材料RT增强剂,和Bi2X3纳米复合材料基光热增强放射增敏剂。这些Bi2X3纳米药物可以有效克服肿瘤缺氧微环境的RT耐药性,具有极高的治疗效果和临床应用前景。最后,提出了Bi2X3纳米材料在RT领域的挑战和前景。
    Radiotherapy (RT), as one of the main methods of clinical tumor treatment, has been applied to the treatment of most solid tumors. However, the effect of RT is compromised by the radiation resistance of tumor hypoxic environment and non-specific damage caused by high-dose radiation. Bismuth chalcogenides (Bi2X3, X = S, Se) based nanodrugs have attracted widespread attention as highly efficient radiosensitizers due to their high photoelectric effect and excellent biocompatibility. More importantly, specially designed nanocomposites can effectively alleviate the radiation resistance of tumor tissues. Here, for the first time, we systematically summarize the latest progresses of Bi2X3 nanodrugs to enhance RT by alleviating the hypoxic tumor microenvironment. These emerging Bi2X3 nanodrugs mainly include three aspects, which are Bi2X3 nanocomposites with high-efficient O2 supply, non-O2-dependent Bi2X3 nanocomposites RT enhancers, and Bi2X3 nanocomposites-based photothermal-enhanced radiosensitizers. These Bi2X3 nanodrugs can effectively overcome the RT resistance of tumor hypoxic microenvironment, and have extremely high therapeutic effects and clinical application prospects. Finally, we put forward the challenges and prospects of Bi2X3 nanomaterials in the field of RT.
    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(RT)是一种使用高剂量辐射杀死癌细胞和缩小肿瘤的癌症治疗方法。虽然在放射治疗方面取得了巨大的成功,增强对肿瘤组织的辐射损伤和减少对健康组织的副作用仍然是一个棘手的挑战。放射增敏剂是可以通过加速DNA损伤和间接产生自由基来增强对肿瘤细胞的杀伤作用的化学品或药剂。在大多数情况下,放射增敏剂对正常组织的影响较小。近年来,已经开发了几种策略来开发高效且低毒性的放射增敏剂。在这次审查中,我们首先总结了包括小分子在内的放射增敏剂的应用,大分子,和纳米材料,尤其是那些已经用于临床试验的。第二,综述了放射增敏剂的发展状况和提高放射增敏剂敏感性的可能机制。第三,介绍了肿瘤治疗中放射增敏剂临床转译面临的挑战和前景.
    Radiotherapy (RT) is a cancer treatment that uses high doses of radiation to kill cancer cells and shrink tumors. Although great success has been achieved on radiotherapy, there is still an intractable challenge to enhance radiation damage to tumor tissue and reduce side effects to healthy tissue. Radiosensitizers are chemicals or pharmaceutical agents that can enhance the killing effect on tumor cells by accelerating DNA damage and producing free radicals indirectly. In most cases, radiosensitizers have less effect on normal tissues. In recent years, several strategies have been exploited to develop radiosensitizers that are highly effective and have low toxicity. In this review, we first summarized the applications of radiosensitizers including small molecules, macromolecules, and nanomaterials, especially those that have been used in clinical trials. Second, the development states of radiosensitizers and the possible mechanisms to improve radiosensitizers sensibility are reviewed. Third, the challenges and prospects for clinical translation of radiosensitizers in oncotherapy are 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
    BACKGROUND: Greater than half of cancer patients experience radiation therapy, for both radical and palliative objectives. It is well known that researches on radiation response mechanisms are conducive to improve the efficacy of cancer radiotherapy. p21 was initially identified as a widespread inhibitor of cyclin-dependent kinases, transcriptionally modulated by p53 and a marker of cellular senescence. It was once considered that p21 acts as a tumour suppressor mainly to restrain cell cycle progression, thereby resulting in growth suppression. With the deepening researches on p21, p21 has been found to regulate radiation responses via participating in multiple cellular processes, including cell cycle arrest, apoptosis, DNA repair, senescence and autophagy. Hence, a comprehensive summary of the p21\'s functions in radiation response will provide a new perspective for radiotherapy against cancer.
    METHODS: We summarize the recent pertinent literature from various electronic databases, including PubMed and analyzed several datasets from Gene Expression Omnibus database. This review discusses how p21 influences the effect of cancer radiotherapy via involving in multiple signaling pathways and expounds the feasibility, barrier and risks of using p21 as a biomarker as well as a therapeutic target of radiotherapy.
    CONCLUSIONS: p21\'s complicated and important functions in cancer radiotherapy make it a promising therapeutic target. Besides, more thorough insights of p21 are needed to make it a safe therapeutic target.
    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
    The radiotherapy modulators used in clinic have disadvantages of high toxicity and low selectivity. For the first time, we used the in situ enzyme-instructed self-assembly (EISA) of a peptide derivative (Nap-GDFDFpYSV) to selectively enhance the sensitivity of cancer cells with high alkaline phosphatase (ALP) expression to ionizing radiation (IR). Compared with the in vitro pre-assembled control formed by the same molecule, assemblies formed by in situ EISA in cells greatly sensitized the ALP-high-expressing cancer cells to γ-rays, with a remarkable sensitizer enhancement ratio. Our results indicated that the enhancement was a result of fixing DNA damage, arresting cell cycles and inducing cell apoptosis. Interestingly, in vitro pre-formed assemblies mainly localized in the lysosomes after incubating with cells, while the assemblies formed via in situ EISA scattered in the cell cytosol. The accumulation of these molecules in cells could not be inhibited by endocytosis inhibitors. We believed that this molecule entered cancer cells by diffusion and then in situ self-assembled to form nanofibers under the catalysis of endogenous ALP. This study provides a successful example to utilize intracellular in situ EISA of small molecules to develop selective tumor radiosensitizers.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    The rapid development of nanotechnology offers a variety of potential therapeutic strategies for cancer treatment. High atomic element nanomaterials are often utilized as radiosensitizers due to their unique photoelectric decay characteristics. Among them, gold nanoparticles (GNPs) are one of the most widely investigated and are considered to be an ideal radiosensitizers for radiotherapy due to their high X-ray absorption and unique physicochemical properties. Over the last few decades, multi-disciplinary studies have focused on the design and optimization of GNPs to achieve greater dosing capability and higher therapeutic effects and highlight potential mechanisms for radiosensitization of GNPs. Although the radiosensitizing potential of GNPs has been widely recognized, its clinical translation still faces many challenges. This review analyses the different roles of GNPs as radiosensitizers in cancer radiotherapy and summarizes recent advances. In addition, the underlying mechanisms of GNP radiosensitization, including physical, chemical and biological mechanisms are discussed, which may provide new directions for the optimization and clinical transformation of next-generation GNPs.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

  • 文章类型: Journal Article
    在临床上经常遇到对放射治疗的抵抗,导致癌症患者预后不良。长链非编码RNA(lncRNA)在放射抗性的发展中起重要作用,因为它们在转录和转录后水平上都具有调节靶基因表达的功能。探索关键lncRNAs并阐明导致辐射抗性的机制对于开发逆转辐射抗性的有效策略至关重要。然而,这仍然具有挑战性。这里,肌动蛋白丝相关蛋白1反义RNA1(lncAFAP1-AS1)被认为是通过激活Wnt/β-catenin信号通路诱导三阴性乳腺癌(TNBC)放射抗性的关键因素。考虑到在辐射下产生高浓度的还原剂谷胱甘肽(GSH),还原响应性纳米颗粒(NP)平台被设计用于有效的lncAFAP1-AS1siRNA(siAFAP1-AS1)递送。siAFAP1-AS1与还原反应性NP的全身递送可以通过沉默lncAFAP1-AS1表达和清除细胞内GSH来协同逆转放射抗性,导致在异种移植和转移性TNBC肿瘤模型中的放疗效果显着增强。研究结果表明,lncAFAP1-AS1可用于预测TNBC放射治疗的结果,全身siAFAP1-AS1给药与放射治疗相结合可用于治疗复发的TNBC患者。
    Resistance to radiotherapy is frequently encountered in clinic, leading to poor prognosis of cancer patients. Long noncoding RNAs (lncRNAs) play important roles in the development of radioresistance due to their functions in regulating the expression of target genes at both transcriptional and posttranscriptional levels. Exploring key lncRNAs and elucidating the mechanisms contributing to radioresistance are crucial for the development of effective strategies to reverse radioresistance, which however remains challenging. Here, actin filament-associated protein 1 antisense RNA1 (lncAFAP1-AS1) is identified as a key factor in inducing radioresistance of triple-negative breast cancer (TNBC) via activating the Wnt/β-catenin signaling pathway. Considering the generation of a high concentration of reduction agent glutathione (GSH) under radiation, a reduction-responsive nanoparticle (NP) platform is engineered for effective lncAFAP1-AS1 siRNA (siAFAP1-AS1) delivery. Systemic delivery of siAFAP1-AS1 with the reduction-responsive NPs can synergistically reverse radioresistance by silencing lncAFAP1-AS1 expression and scavenging intracellular GSH, leading to a dramatically enhanced radiotherapy effect in both xenograft and metastatic TNBC tumor models. The findings indicate that lncAFAP1-AS1 can be used to predict the outcome of TNBC radiotherapy and combination of systemic siAFAP1-AS1 delivery with radiotherapy can be applied for the treatment of recurrent TNBC patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号