Radiosensitization

放射增敏
  • 文章类型: Journal Article
    头颈部鳞状细胞癌(HNSCC)在全球范围内普遍存在,复发率高,存活率低,以及患者生活质量差。来自PAC-1的SM-1可以激活proaspase-3并诱导癌细胞凋亡以发挥抗肿瘤作用。然而,联合放疗后SM-1对HNSCC的抑制作用尚不清楚。本研究旨在探讨SM-1对HNSCC的体内外放射增敏作用。
    MTT方法用于检测SM-1对HNSCC细胞系(HONE1,HSC-2和CAL27)活力的影响。通过集落形成实验确定SM-1联合辐射对HONE1,HSC-2和CAL27细胞系存活指数的影响。采用流式细胞术观察SM-1与放疗联合应用对细胞凋亡和细胞周期的影响,进行蛋白质印迹实验以检测细胞凋亡和细胞周期相关蛋白的表达。最后,建立CAL27异种移植瘤模型,评价SM-1联合放射的体内抗肿瘤作用。
    体外,SM-1有效抑制HNSCC细胞系HONE1、HSC-2和CAL27细胞的活性,并在联合照射过程中协同表现出抗增殖活性。同时,SM-1对HNSCC的抗肿瘤作用高于Debio1143,细胞的放射敏感性大大提高。流式细胞术和westernblot分析表明,SM-1通过抑制CyclinB1和CDC2的表达,诱导头颈部鳞状细胞癌细胞G2/M期阻滞。此外,SM-1激活caspase-3活性并上调PARP1的裂解形式以诱导细胞凋亡。在体内,SM-1联合照射显示出良好的抗肿瘤效果。
    SM-1在体外和体内增强了HNSCC细胞的辐射敏感性,支持其作为放射增敏剂的潜力与放射治疗相结合的临床试验。
    UNASSIGNED: Head and neck squamous cell carcinoma (HNSCC) is globally prevalent with high recurrence, low survival rate, and poor quality of life for patients. Derived from PAC-1, SM-1 can activate procaspase-3 and induce apoptosis in cancer cells to exert anti-tumor effects. However, the inhibitory effect of SM-1 on HNSCC after combination with radiation are unclear. This study aims to investigate the radiosensitizing effect of SM-1 on HNSCC in vitro and in vivo.
    UNASSIGNED: MTT method was used to detect the effect of SM-1 on the viability of HNSCC cell lines (HONE1, HSC-2, and CAL27). The effects of SM-1 combined with radiation on the survival index of HONE1, HSC-2, and CAL27 cell lines were determined by colony formation assay. Flow cytometry was used to investigate the effects of SM-1 and radiation combination on cell apoptosis and cell cycle, and western blot experiments were performed to detect the expression of apoptosis and cell cycle-related proteins. Finally, a xenograft tumor model of CAL27 was established to evaluate the anti-tumor effect of SM-1 combined with radiation in vivo.
    UNASSIGNED: In vitro, SM-1 effectively inhibited the activity of HNSCC cell lines HONE1, HSC-2, and CAL27 cells, and synergistically showed anti-proliferation activity during combined irradiation. Meanwhile, anti-tumor effect of SM-1 on HNSCC was higher than that of Debio1143, and the radiosensitivity of cells was greatly increased. Flow cytometry and western blot analysis showed that SM-1 induced G2/M phase arrest of head and neck squamous cell carcinoma cells via inhibiting the expression of CyclinB1 and CDC2. Moreover, SM-1 activated caspase-3 activity and up-regulated the cleaved form of PARP1 to induce cell apoptosis. In vivo, SM-1 combined irradiation showed a good anti-tumor effect.
    UNASSIGNED: SM-1 enhances HNSCC cell radiation sensitivity in vitro and in vivo, supporting its potential as a radiosensitizer for clinical trials in combination with radiotherapy.
    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
    生脉健黄散(SMJHS)是一种传统的中药复方,据报道可以抑制鼻咽癌(NPC)的进展并增强放射敏感性。然而,SMJHS抗鼻咽癌的具体活性成分和调控机制,特别是在缺氧条件下,仍然不清楚。在这项研究中,Sprague-Dawley(SD)大鼠用生脉健黄散(SMJHS)灌胃,他们的血液是从腹主动脉采集的.UHPLC-Q-ExactiveorbitrapMS/MS用于鉴定SMJHS含药血清的代谢物谱。通过网络药理学和分子对接,构建了SMJHS靶向NPC中活性成分的分子网络。HIF-1α/VEGF通路处于关键位置。SMJHS对细胞增殖的影响,迁移,通过体外实验评估缺氧NPC细胞的放射敏感性。使用慢病毒建立稳定过表达HIF-1α的NPC细胞系,以研究SMJHS对缺氧NPC细胞中HIF-1α/VEGF信号传导的调节。通过结合网络药理分析,细胞生物功能验证,分子生化实验,我们的研究发现SMJHS对缺氧条件下培养的NPC细胞具有抗增殖作用,抑制其迁移并增加其放射敏感性。此外,SMJHS抑制HIF-1α和VEGFA的表达,显示作为改善NPC治疗的有效选择的潜力。
    Shengmai Jianghuang San (SMJHS) is a traditional Chinese herbal compound reported to inhibit Nasopharyngeal Carcinoma (NPC) progression and enhance radiosensitivity. However, the specific active ingredients and regulatory mechanisms of SMJHS against NPC, particularly under hypoxic conditions, remain unclear. In this study, Sprague-Dawley (SD) rats were gavaged with Shengmai Jianghuang San (SMJHS), and their blood was collected from the abdominal aorta. UHPLC-Q-Exactive orbitrap MS/MS was used to identify the metabolite profiles of SMJHS drug-containing serum. A molecular network of the active compositions in SMJHS targeting NPC was constructed through network pharmacology and molecular docking. The HIF-1α/VEGF pathway was in key positions. The effects of SMJHS on the proliferation, migration, and radiosensitivity of hypoxic NPC cells were assessed by in vitro experiments. NPC cell lines stably overexpressing HIF-1α were established using a lentivirus to investigate the regulation of HIF-1α/VEGF signaling in hypoxic NPC cells by SMJHS. Through a combination of network pharmacological analysis, cellular biofunctional validation, and molecular biochemical experiments, our study found that SMJHS had an anti-proliferative effect on NPC cells cultured under hypoxic conditions, inhibiting their migration and increasing their radiosensitivity. Additionally, SMJHS suppressed the expression of HIF-1α and VEGFA, exhibiting potential as an effective option for improving NPC treatment.
    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
    不可切除的直肠癌患者的治疗结果是复杂的,同步放化疗是主要的治疗选择。放射增敏剂可以增强局部瘤内缺氧的效果,有助于局部控制和症状缓解。本研究评估了无法切除的直肠癌患者使用过氧化氢联合放射治疗(RT)进行放射增敏的可行性和安全性。本研究共招募13例直肠癌患者。每周两次与RT组合进行放射增敏。将浸泡在3%过氧化氢溶液中的纱布插入肛门,确保与病变的牢固接触。总的来说,每周5天使用10MVX射线从四个方向以25-33个部分将45-65Gy递送至整个骨盆。在RT完成后1个月和6个月评估急性和晚期不良事件。治疗耐受性良好,没有发现急性3级或更严重的事件,没有病人出现直肠瘘,坏死,阻塞,穿孔,狭窄,溃疡或腹膜后出血。无明显的晚期不良事件,超过6个月,在分析结束时观察到。所有患者疼痛缓解,止血效果和肿瘤缩小。因此,对于无法手术的直肠肿瘤患者,在直肠中使用过氧化氢溶液浸泡的纱布可能是一个有希望的选择.本研究的局限性在于患者群体较少,观察时间相对较短。本研究在大学医院医学信息网络中心进行了回顾性注册(试验注册编号:R000061902)于2024年4月21日。
    The treatment outcomes of patients with unresectable rectal cancer are complex, and concurrent chemoradiation therapy is the main treatment option. Radiosensitizers can enhance the effect of localized intratumoral hypoxia, contributing to local control and symptomatic relief. The present study evaluated the feasibility and safety of radiosensitization using hydrogen peroxide combined with radiation therapy (RT) in patients with unresectable rectal cancer. A total of 13 patients with rectal cancer were recruited in the present study. Radiosensitization was performed twice weekly in combination with RT. Gauze soaked in 3% hydrogen peroxide solution was inserted into the anus, ensuring firm contact with the lesion. In total, 45-65 Gy was delivered in 25-33 fractions to the whole pelvis from four directions using 10 MV X-rays 5 days per week. Acute and late adverse events were evaluated 1 and 6 months after the completion of RT. Treatment was well tolerated, with no acute grade 3 or worse events noted, and no patient developed rectal fistula, necrosis, obstruction, perforation, stenosis, ulcer or retroperitoneal hemorrhage. No notable late adverse events, beyond 6 months, were observed at the end of the analysis. All patients experienced pain relief, hemostatic effects and tumor shrinkage. Therefore, the use of a hydrogen peroxide solution-soaked gauze in the rectum may be a promising option for patients with inoperable rectal tumors. The limitations of the present study are that the patient population was small and the observation time was relatively short. This study was retrospectively registered with the University Hospital Medical Information Network Center (trial registration no. R000061902) on April 21, 2024.
    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
    PRMT5是一种广泛表达的精氨酸甲基转移酶,可调节涉及肿瘤细胞增殖和存活的过程。在这里描述的研究中,我们调查了PRMT5是否为肿瘤放射增敏提供了靶点.使用siRNA敲除PRMT5增强了一组对应于通常用放射疗法治疗的肿瘤类型的细胞系的放射敏感性。为了将这些研究扩展到实验性治疗环境,使用PRMT5抑制剂LLY-283.肿瘤细胞系暴露于LLY-283会降低PRMT5活性并增强其放射敏感性。通过γH2AX病灶和中性彗星分析确定,放射敏感性的增加伴随着DNA双链断裂修复的抑制。对于正常的成纤维细胞系,虽然LLY-283降低了PRMT5活性,对他们的放射敏感性没有影响.对U251细胞的转录组分析表明,LLY-283处理降低了基因的表达并改变了参与DNA损伤反应的基因的mRNA剪接模式。然后使用皮下异种移植物来评估对LLY-283和辐射的体内反应。用LLY-283治疗小鼠降低了肿瘤PRMT5的活性,并显着增强了辐射诱导的生长延迟。这些结果表明PRMT5是放射增敏的肿瘤选择性靶标。
    PRMT5 is a widely expressed arginine methyltransferase that regulates processes involved in tumor cell proliferation and survival. In the study described here, we investigated whether PRMT5 provides a target for tumor radiosensitization. Knockdown of PRMT5 using siRNA enhanced the radiosensitivity of a panel of cell lines corresponding to tumor types typically treated with radiotherapy. To extend these studies to an experimental therapeutic setting, the PRMT5 inhibitor LLY-283 was used. Exposure of the tumor cell lines to LLY-283 decreased PRMT5 activity and enhanced their radiosensitivity. This increase in radiosensitivity was accompanied by an inhibition of DNA double-strand break repair as determined by γH2AX foci and neutral comet analyses. For a normal fibroblast cell line, although LLY-283 reduced PRMT5 activity, it had no effect on their radiosensitivity. Transcriptome analysis of U251 cells showed that LLY-283 treatment reduced the expression of genes and altered the mRNA splicing pattern of genes involved in the DNA damage response. Subcutaneous xenografts were then used to evaluate the in vivo response to LLY-283 and radiation. Treatment of mice with LLY-283 decreased tumor PRMT5 activity and significantly enhanced the radiation-induced growth delay. These results suggest that PRMT5 is a tumor selective target for radiosensitization.
    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
    胶质母细胞瘤(GB)患者的化疗治疗标准是放射治疗(RT)联合替莫唑胺(TMZ)。然而,在它推出以来的二十年里,这个所谓的Stupp协议揭示了主要的缺点,因为近一半的GBs具有内在的治疗抗病机制。其中主要是DNA修复蛋白O6-鸟嘌呤-DNA甲基转移酶(MGMT)的表达增加和DNA错配修复(MMR)中的细胞缺陷。患有这种肿瘤的患者接受的很少,如果有的话,受益于TMZ。我们正在开发一种新的分子,NEO212(与NEO100共轭的TMZ),有可能克服这些限制。
    我们使用原位植入GB细胞系或原代的小鼠模型,抗辐射人类GB干细胞,代表不同的治疗耐药机制。动物接受无或有RT的NEO212(或用于比较的TMZ)。记录总生存期,组织学研究量化了DNA损伤,凋亡,微血管密度,以及对骨髓的影响.
    在所有肿瘤模型中,在旨在模仿Stupp协议的时间表中,用NEO212代替TMZ实现了惊人的生存扩展,特别是在抗TMZ和抗RT模型。虽然NEO212表现出明显的辐射敏感性,DNA损伤,促凋亡,和肿瘤组织中的抗血管生成作用,它没有引起骨髓毒性。
    NEO212是标准Stupp方案中可能替代TMZ的候选药物。当与放射组合时,它有可能成为第一种显着延长对TMZ耐药的患者的总体生存期的化学治疗剂。
    UNASSIGNED: The chemotherapeutic standard of care for patients with glioblastoma (GB) is radiation therapy (RT) combined with temozolomide (TMZ). However, during the twenty years since its introduction, this so-called Stupp protocol has revealed major drawbacks, because nearly half of all GBs harbor intrinsic treatment resistance mechanisms. Prime among these are the increased expression of the DNA repair protein O6-guanine-DNA methyltransferase (MGMT) and cellular deficiency in DNA mismatch repair (MMR). Patients with such tumors receive very little, if any, benefit from TMZ. We are developing a novel molecule, NEO212 (TMZ conjugated to NEO100), that harbors the potential to overcome these limitations.
    UNASSIGNED: We used mouse models that were orthotopically implanted with GB cell lines or primary, radioresistant human GB stem cells, representing different treatment resistance mechanisms. Animals received NEO212 (or TMZ for comparison) without or with RT. Overall survival was recorded, and histology studies quantified DNA damage, apoptosis, microvessel density, and impact on bone marrow.
    UNASSIGNED: In all tumor models, replacing TMZ with NEO212 in a schedule designed to mimic the Stupp protocol achieved a strikingly superior extension of survival, especially in TMZ-resistant and RT-resistant models. While NEO212 displayed pronounced radiation-sensitizing, DNA-damaging, pro-apoptotic, and anti-angiogenic effects in tumor tissue, it did not cause bone marrow toxicity.
    UNASSIGNED: NEO212 is a candidate drug to potentially replace TMZ within the standard Stupp protocol. It has the potential to become the first chemotherapeutic agent to significantly extend overall survival in TMZ-resistant patients when combined with radiation.
    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:放射治疗(RT)是一种广泛使用的肿瘤治疗方法,而这种治疗方式的主要障碍是肿瘤细胞表现出的放射抗性。为了提高RT的有效性,科学家们已经探索了放射增敏方法,包括使用放射增敏剂和物理刺激。然而,几种方法表现出令人失望的结果,包括不良反应和疗效有限.一种更安全,更有效的放射增敏方法涉及低强度超声(LIUS),选择性靶向肿瘤组织并增强放射治疗的疗效。
    方法:本文总结了肿瘤放射抗性的原因,并探讨了LIUS潜在的放射增敏机制。此外,它涵盖了LIUS在放射增敏方面的不同应用策略,包括单独使用LIUS,超声靶向血管内微气泡破坏,超声介导的靶向放射增敏剂递送,和声动力疗法。最后,该综述介绍了在临床环境中采用LIUS-RT联合治疗的局限性和前景,强调需要将研究成果与实际应用联系起来。
    结论:LIUS采用具有成本效益的设备来促进肿瘤放射增敏,减少辐射暴露,提高患者的生活质量。这种功效归因于LIUS利用热量的能力,空化,和机械效应来克服肿瘤细胞对RT的抗性。多项实验分析强调了LIUS使用多种策略诱导肿瘤放射增敏的有效性。虽然初步研究显示出了有希望的结果,进行更全面的临床试验对于确认其在现实世界中的安全性和有效性至关重要。
    BACKGROUND: Radiotherapy (RT) is a widely utilized tumor treatment approach, while a significant obstacle in this treatment modality is the radioresistance exhibited by tumor cells. To enhance the effectiveness of RT, scientists have explored radiosensitization approaches, including the use of radiosensitizers and physical stimuli. Nevertheless, several approaches have exhibited disappointing results including adverse effects and limited efficacy. A safer and more effective method of radiosensitization involves low-intensity ultrasound (LIUS), which selectively targets tumor tissue and enhances the efficacy of radiation therapy.
    METHODS: This review summarized the tumor radioresistance reasons and explored LIUS potential radiosensitization mechanisms. Moreover, it covered diverse LIUS application strategies in radiosensitization, including the use of LIUS alone, ultrasound-targeted intravascular microbubble destruction, ultrasound-mediated targeted radiosensitizers delivery, and sonodynamic therapy. Lastly, the review presented the limitations and prospects of employing LIUS-RT combined therapy in clinical settings, emphasizing the need to connect research findings with practical applications.
    CONCLUSIONS: LIUS employs cost-effective equipment to foster tumor radiosensitization, curtail radiation exposure, and elevate the quality of life for patients. This efficacy is attributed to LIUS\'s ability to utilize thermal, cavitation, and mechanical effects to overcome tumor cell resistance to RT. Multiple experimental analyses have underscored the effectiveness of LIUS in inducing tumor radiosensitization using diverse strategies. While initial studies have shown promising results, conducting more comprehensive clinical trials is crucial to confirm its safety and effectiveness in real-world situations.
    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
    除了细胞毒性,COX-2酶的抑制剂已证明对癌症治疗具有重要的附加作用(例如肿瘤细胞的放射增敏作用和细胞抗增殖作用);然而,其他炎症相关酶5-LOX抑制剂的抑制与抗癌活性之间的关系仍未得到很好的理解。在我们的研究中,我们在3种癌细胞系(HCT116,HT-29和BxPC-3)和1种健康细胞系(MRC-5)上测试了我们组之前提出的13种COX-2和5-LOX抑制剂(1-13)的细胞毒性.化合物3、5、6和7显示出中等的细胞毒性,但对癌细胞系具有良好的选择性。IC50值在22.99-51.66µM(HCT116细胞系)的范围内,8.63-41.20µM(BxPC-3细胞系)和24.78-81.60µM(HT-29细胞系;化合物7>100µM)。与测试相比,市售COX-2和5-LOX抑制剂,细胞毒性和选择性均增加。在辐射处理中加入化合物6和7显示HT-29细胞系的细胞增殖最显著降低(p<0.001)。通过使用SW620细胞系的伤口愈合测定来测试最佳双COX-2和5-LOX抑制剂(化合物1、2、3和5)的抗迁移潜力。化合物1和3作为具有最有效作用的化合物(相对伤口闭合率为3.20%(24小时),化合物1的5.08%(48h)和3.86%(24h),7.68%(48h)的化合物3)。考虑到所有这些结果,化合物3是生物活性最佳的化合物,具有最佳的双重COX-2和5-LOX抑制活性,对测试的癌细胞系具有良好的选择性,显著的细胞抗迁移潜力和在治疗剂量缺乏毒性作用。
    Apart from cytotoxicity, inhibitors of the COX-2 enzyme have demonstrated additional effects important for cancer treatment (such as radiosensitization of tumor cells and cell antimigratory effects); however, the relationship between the inhibition of other inflammation-related enzyme 5-LOX inhibitors and anticancer activity is still not well understood. In our study, the cytotoxicity of thirteen COX-2 and 5-LOX inhibitors previously presented by our group (1-13) was tested on three cancer cell lines (HCT 116, HT-29 and BxPC-3) and one healthy cell line (MRC-5). Compounds 3, 5, 6 and 7 showed moderate cytotoxicity, but good selectivity towards cancer cell lines. IC50 values were in the range of 22.99-51.66 µM (HCT 116 cell line), 8.63-41.20 µM (BxPC-3 cell line) and 24.78-81.60 µM (HT-29 cell line; compound 7 > 100 µM). In comparison to tested, commercially available COX-2 and 5-LOX inhibitors, both cytotoxicity and selectivity were increased. The addition of compounds 6 and 7 to irradiation treatment showed the most significant decrease in cell proliferation of the HT-29 cell line (p < 0.001). The antimigratory potential of the best dual COX-2 and 5-LOX inhibitors (compounds 1, 2, 3 and 5) was tested by a wound-healing assay using the SW620 cell line. Compounds 1 and 3 were singled out as compounds with the most potent effect (relative wound closure was 3.20% (24 h), 5,08% (48 h) for compound 1 and 3.86% (24 h), 7.68% (48 h) for compound 3). Considering all these results, compound 3 stood out as the compound with the most optimal biological activity, with the best dual COX-2 and 5-LOX inhibitory activity, good selectivity towards tested cancer cell lines, significant cell antimigratory potential and a lack of toxic effects at therapeutic doses.
    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
    放射治疗广泛用于癌症治疗,但其临床应用受到放射抗性和无法靶向转移的限制。纳米级金属有机框架(MOF)已显示出有望作为高Z纳米放射增敏剂,以增强放射疗法并诱导肿瘤微环境的免疫刺激调节。我们假设MOFs可以提供小分子治疗剂以与放射疗法协同增强抗肿瘤功效。在这里,我们开发了一种强大的纳米放射增敏剂,GA-MOF,通过缀合STING激动剂,2\',3'-环磷酸鸟苷-磷酸腺苷(GA),关于MOFs的协同放射增敏和STING激活。GA-MOF通过形成富含免疫细胞的结节(人工类白细胞结构)并通过放射疗法将其转化为免疫刺激热点而表现出强大的抗癌功效。与免疫检查点阻断的进一步组合通过全身免疫激活抑制远端肿瘤。我们的工作不仅证明了GA-MOF的有效放射增敏作用,而且还提供了有关MOF分配的详细机制,免疫调节途径和长期免疫效应。
    Radiotherapy is widely used for cancer treatment, but its clinical utility is limited by radioresistance and its inability to target metastases. Nanoscale metal-organic frameworks (MOFs) have shown promise as high-Z nanoradiosensitizers to enhance radiotherapy and induce immunostimulatory regulation of the tumor microenvironment. We hypothesized that MOFs could deliver small-molecule therapeutics to synergize with radiotherapy for enhanced antitumor efficacy. Herein, we develop a robust nanoradiosensitizer, GA-MOF, by conjugating a STING agonist, 2\',3\'-cyclic guanosine monophosphate-adenosine monophosphate (GA), on MOFs for synergistic radiosensitization and STING activation. GA-MOF demonstrated strong anticancer efficacy by forming immune-cell-rich nodules (artificial leukocytoid structures) and transforming them into immunostimulatory hotspots with radiotherapy. Further combination with an immune checkpoint blockade suppressed distant tumors through systemic immune activation. Our work not only demonstrates the potent radiosensitization of GA-MOF, but also provides detailed mechanisms regarding MOF distribution, immune regulatory pathways and long-term immune effects.
    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
    在不能手术的非小细胞肺癌(NSCLC)患者的临床治疗中,辐射抵抗是不可避免的障碍。与放射增敏剂联合治疗可以提高放疗的疗效。以前,喹啉衍生物10E作为Nur77的新出口国在肝细胞癌中显示出优越的抗肿瘤活性。这里,我们旨在研究10E的放射增敏活性和作用机制。体外,A549和H460细胞用对照处理,电离辐射(IR),10E,和10E+IR。细胞活力,凋亡,并使用CCK-8和流式细胞术检测周期。使用蛋白质印迹和免疫荧光检查蛋白质表达和定位。建立肿瘤异种移植模型以评估10E的体内放射增敏作用。10E显着抑制细胞增殖并增加其放射敏感性,同时降低p-BCRA1,p-DNA-PKs的水平,53BP1参与DNA损伤修复途径,表明其放射增敏活性与抑制DNA损伤修复密切相关。A549细胞显示出低水平的Nur77和对IR的低反应,但10E处理的A549细胞显示出高水平的Nur77,表明Nur77是核心放射敏感性因子,10E恢复了Nur77的表达。Nur77和Ku80在10E处理的A549细胞中的核外共定位表明10E调节的Nur77核输出抑制DNA损伤修复途径并增加IR触发的凋亡。10E和IR的组合显著抑制肿瘤异种移植模型中的肿瘤生长。我们的发现表明10E作为放射增敏剂,将10E与放疗结合可能是NSCLC治疗的潜在策略。
    Radioresistance is an inevitable obstacle in the clinical treatment of inoperable patients with non-small cell lung cancer (NSCLC). Combining treatment with radiosensitizers may improve the efficacy of radiotherapy. Previously, the quinoline derivative 10E as new exporter of Nur77 has shown superior antitumor activity in hepatocellular carcinoma. Here, we aimed to investigate the radiosensitizing activity and acting mechanisms of 10E. In vitro, A549 and H460 cells were treated with control, ionizing radiation (IR), 10E, and 10E + IR. Cell viability, apoptosis, and cycle were examined using CCK-8 and flow cytometry assays. Protein expression and localization were examined using western blotting and immunofluorescence. Tumor xenograft models were established to evaluate the radiosensitizing effect of 10E in vivo. 10E significantly inhibited cell proliferation and increased their radiosensitivity while reducing level of p-BCRA1, p-DNA-PKs, and 53BP1 involved in the DNA damage repair pathway, indicating that its radiosensitizing activity is closely associated with repressing DNA damage repair. A549 cells showed low level of Nur77 and a low response to IR but 10E-treated A549 cells showed high level of Nur77 indicating that Nur77 is a core radiosensitivity factor and 10E restores the expression of Nur77. Nur77 and Ku80 extranuclear co-localization in the 10E-treated A549 cells suggested that 10E-modulated Nur77 nuclear exportation inhibits DNA damage repair pathways and increases IR-triggered apoptosis. The combination of 10E and IR significantly inhibits tumor growth in a tumor xenograft model. Our findings suggest that 10E acts as a radiosensitizer and that combining 10E with radiotherapy may be a potential strategy for NSCLC treatment.
    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
    背景:小儿型弥漫性高级别神经胶质瘤(pHGG)是儿童中最常见的恶性脑肿瘤,可以细分为多个实体。激活MET受体酪氨酸激酶的融合基因通常发生在婴儿型半球神经胶质瘤(IHG)中,也发生在其他pHGG中,并与破坏性的发病率和死亡率有关。
    方法:为了确定新的治疗方案,我们建立并表征了两种具有不同MET融合的新型原位小鼠模型。其中包括免疫能力,小鼠同种异体移植模型和患者来源的原位异种移植物(PDOX),来自MET融合IHG患者,该患者的常规治疗和卡博替尼靶向治疗失败。有了这些模型,我们分析了三种MET抑制剂的药效和药代动力学特性,卡马替尼,克唑替尼和卡博替尼,单独或联合放疗。
    结果:卡马替尼在两种模型中都显示出比卡博替尼或克唑替尼更好的脑药代动力学特性和更大的体外和体内功效。PDOX模型概括了患者经历的卡博替尼的不良疗效。相比之下,在两个互补小鼠模型中,卡马替尼与放疗联合治疗可延长生存期并诱导长期无进展生存期.卡马替尼治疗增加了辐射诱导的DNA双链断裂并延迟了其修复。
    结论:我们全面研究了MET抑制和放疗的组合作为MET驱动的pHGG的新治疗选择。我们开创性的临床前数据包包括药代动力学表征,临床结果的概述,多重补充体内研究的结果一致,以及对增加疗效的分子机制的见解。一起来看,我们证明了卡马替尼和放疗的突破性疗效,作为未来临床试验的一个非常有前景的概念.
    BACKGROUND: Pediatric-type diffuse high-grade glioma (pHGG) is the most frequent malignant brain tumor in children and can be subclassified into multiple entities. Fusion genes activating the MET receptor tyrosine kinase often occur in infant-type hemispheric glioma (IHG) but also in other pHGG and are associated with devastating morbidity and mortality.
    METHODS: To identify new treatment options, we established and characterized two novel orthotopic mouse models harboring distinct MET fusions. These included an immunocompetent, murine allograft model and patient-derived orthotopic xenografts (PDOX) from a MET-fusion IHG patient who failed conventional therapy and targeted therapy with cabozantinib. With these models, we analyzed the efficacy and pharmacokinetic properties of three MET inhibitors, capmatinib, crizotinib and cabozantinib, alone or combined with radiotherapy.
    RESULTS: Capmatinib showed superior brain pharmacokinetic properties and greater in vitro and in vivo efficacy than cabozantinib or crizotinib in both models. The PDOX models recapitulated the poor efficacy of cabozantinib experienced by the patient. In contrast, capmatinib extended survival and induced long-term progression-free survival when combined with radiotherapy in two complementary mouse models. Capmatinib treatment increased radiation-induced DNA double-strand breaks and delayed their repair.
    CONCLUSIONS: We comprehensively investigated the combination of MET inhibition and radiotherapy as a novel treatment option for MET-driven pHGG. Our seminal preclinical data package includes pharmacokinetic characterization, recapitulation of clinical outcomes, coinciding results from multiple complementing in vivo studies, and insights into molecular mechanism underlying increased efficacy. Taken together, we demonstrate the groundbreaking efficacy of capmatinib and radiation as a highly promising concept for future clinical trials.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号