mTOR inhibitors

mTOR 抑制剂
  • 文章类型: Journal Article
    背景:PD-1/PD-L1抑制剂BMS-1166,抑制PD-L1与PD-1的结合,恢复T细胞功能,增强肿瘤免疫反应。然而,肿瘤抑制基因的突变或受损的细胞信号通路也可能导致细胞转化。在这项研究中,使用SW480和SW480R细胞系作为模型,阐明BMS-1166,BEZ235及其组合的治疗.方法:采用MTT法和集落形成法评价细胞增殖。伤口愈合测定用于评估细胞迁移。通过流式细胞术分析细胞周期和细胞凋亡。PI3K/Akt/mTOR和MAPK通路中关键激酶的磷酸化水平,PD-L1和与增殖相关的蛋白质水平,迁移,使用蛋白质印迹法评估细胞凋亡。结果:BEZ235增强BMS-1166介导的SW480和SW480R细胞增殖和迁移抑制作用,促进细胞凋亡。有趣的是,负调节剂PTEN的下调提高了PD-L1水平,它被Akt的抑制所废除。BMS-1166推广了PI3K,Akt,mTOR,和Erk磷酸化。然而,BEZ235与BMS-1166的组合抑制PI3K的表达,p-Akt,p-mTOR,通过抑制PD1与PD-L1的结合,与BMS-1166或BEZ235单次处理相比,SW480和SW480R细胞中的p-Erk。结论:PD-1与PD-L1结合并激活PI3K/mTOR和MAPK通路,这可能是CRC对BMS-1166获得性耐药的分子机制。两种药物联合抑制PI3K的磷酸化,Akt,和Erk在PI3K/mTOR和MAPK通路中,即,BEZ235通过阻断PI3K/mTOR通路和干扰MAPK通路的串扰增强BMS-1166治疗效果。因此,本研究结果为BMS-1166联合BEZ235治疗结直肠癌提供了理论依据。
    Background: BMS-1166, a PD-1/PD-L1 inhibitor, inhibits the binding of PD-L1 to PD-1, restores T cell function, and enhances tumor immune response. However, mutations in the tumor suppressor or impaired cellular signaling pathways may also lead to cellular transformation. In this study, the SW480 and SW480R cell lines were used as the model to elucidate the treatment with BMS-1166, BEZ235, and their combination. Methods: MTT and colony-formation assays were used to evaluate cell proliferation. Wound-healing assay was used to assess cell migration. Cell cycle and apoptosis were analyzed by flow cytometry. The phosphorylation level of the key kinases in the PI3K/Akt/mTOR and MAPK pathways, PD-L1, and the protein levels related to the proliferation, migration, and apoptosis were assessed using western blotting. Results: BEZ235 enhanced BMS-1166-mediated cell proliferation and migration inhibition in SW480 and SW480R cells and promoted apoptosis. Interestingly, the downregulation of the negative regulator PTEN raised the PD-L1 level, which was abolished by the inhibition of Akt. BMS-1166 promoted PI3K, Akt, mTOR, and Erk phosphorylation. However, the combination of BEZ235 with BMS-1166 suppressed the expression of PI3K, p-Akt, p-mTOR, and p-Erk in SW480 and SW480R cells compared to BMS-1166 or BEZ235 single treatment by inhibiting the binding of PD1 to PD-L1. Conclusions: PD-1 binds to PD-L1 and activates the PI3K/mTOR and MAPK pathways, which might be the molecular mechanism of acquired resistance of CRC to BMS-1166. The combination of the two drugs inhibited the phosphorylation of PI3K, Akt, and Erk in the PI3K/mTOR and MAPK pathway, i.e., BEZ235 enhanced the BMS-1166 treatment effect by blocking the PI3K/mTOR pathway and interfering with the crosstalk of the MAPK pathway. Therefore, these findings provide a theoretical basis for BMS-1166 combined with BEZ235 in the trial treatment of colorectal cancer.
    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:针对2019年冠状病毒病(COVID-19)的疫苗接种在淋巴管平滑肌瘤病(LAM)患者中的安全性和有效性尚不清楚。这项研究调查了COVID-19疫苗的犹豫,疫苗的安全性和有效性,LAM患者的COVID-19症状。
    结果:总计,181名LAM患者和143名健康个体回答了问卷。LAM患者的接种率为77.34%,15.7%的接种疫苗的LAM患者出现不良事件。接种疫苗降低了LAM患者发生厌食症的风险[OR:0.17,95%CI:(0.07,0.43)],肌痛[OR:0.34,95%CI:(0.13,0.84)],和不适应[OR:0.34,95%CI:(0.14,0.84)]。在LAM患者中,使用mTOR抑制剂可降低COVID-19期间出现症状的风险,包括疲劳[OR:0.18,95%CI:(0.03,0.95)],厌食症[OR:0.30,95%CI:(0.09,0.96)],和不适应[OR:0.20,95%CI:(0.06,0.67)]。
    结论:LAM人群的疫苗接种率低于一般人群,22.7%(41/181)的LAM患者对COVID-19疫苗有犹豫。然而,LAM队列中COVID-19疫苗接种的安全性与健康人群相当,和COVID-19疫苗接种降低了LAM患者COVID-19症状的发生率。此外,mTOR抑制剂似乎不能确定COVID-19期间LAM患者出现并发症的风险更大。
    BACKGROUND: The safety and efficacy of vaccination against coronavirus disease 2019 (COVID-19) in patients with lymphangioleiomyomatosis (LAM) is still unclear. This study investigates COVID-19 vaccine hesitancy, vaccine safety and efficacy, and COVID-19 symptoms in LAM patients.
    RESULTS: In total, 181 LAM patients and 143 healthy individuals responded to the questionnaire. The vaccination rate of LAM patients was 77.34%, and 15.7% of vaccinated LAM patients experienced adverse events. Vaccination decreased the risk of LAM patients developing anorexia [OR: 0.17, 95% CI: (0.07, 0.43)], myalgia [OR: 0.34, 95% CI: (0.13, 0.84)], and ageusia [OR: 0.34, 95% CI: (0.14, 0.84)]. In LAM patients, a use of mTOR inhibitors reduced the risk of developing symptoms during COVID-19, including fatigue [OR: 0.18, 95% CI: (0.03, 0.95)], anorexia [OR: 0.30, 95% CI: (0.09, 0.96)], and ageusia [OR: 0.20, 95% CI: (0.06, 0.67)].
    CONCLUSIONS: Vaccination rates in the LAM population were lower than those in the general population, as 22.7% (41/181) of LAM patients had hesitations regarding the COVID-19 vaccine. However, the safety of COVID-19 vaccination in the LAM cohort was comparable to the healthy population, and COVID-19 vaccination decreased the incidence of COVID-19 symptoms in LAM patients. In addition, mTOR inhibitors seem not to determine a greater risk of complications in patients with LAM during COVID-19.
    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
    免疫功能障碍是自发性流产(SM)背后的罪魁祸首。为了解决这个问题,免疫抑制剂已经成为一类新型的保胎药物,调节母体免疫系统对胚胎的耐受性。雷帕霉素(PubChemCID:5284616),双重用途的化合物,发挥免疫抑制剂的作用,并通过靶向mTOR通路触发自噬。它在治疗SM方面的功效最近引起了人们的重大研究兴趣。自噬,自我降解和再循环的细胞过程,在许多健康状况中起着举足轻重的作用。研究表明,自噬是子宫内膜蜕膜化的组成部分,滋养细胞入侵,以及健康怀孕期间蜕膜免疫细胞的正常功能。然而,在SM的情况下,在蜕膜基质细胞或母胎界面的免疫细胞中mTOR/自噬轴失调。体外和体内研究都强调了低剂量雷帕霉素在管理SM中的潜在益处。然而,考虑到mTOR在能量代谢中的关键作用,抑制它可能会损害怀孕。此外,虽然低剂量雷帕霉素被认为是安全的治疗复发性植入失败,由于数据不足,其潜在的致畸作用仍不确定。总之,雷帕霉素是治疗SM的一把双刃剑,平衡其对自噬和免疫调节的影响。需要进一步调查才能充分了解其含义。
    Immune dysfunction is a primary culprit behind spontaneous miscarriage (SM). To address this, immunosuppressive agents have emerged as a novel class of tocolytic drugs, modulating the maternal immune system\'s tolerance towards the embryo. Rapamycin (PubChem CID:5284616), a dual-purpose compound, functions as an immunosuppressive agent and triggers autophagy by targeting the mTOR pathway. Its efficacy in treating SM has garnered significant research interest in recent times. Autophagy, the cellular process of self-degradation and recycling, plays a pivotal role in numerous health conditions. Research indicates that autophagy is integral to endometrial decidualization, trophoblast invasion, and the proper functioning of decidual immune cells during a healthy pregnancy. Yet, in cases of SM, there is a dysregulation of the mTOR/autophagy axis in decidual stromal cells or immune cells at the maternal-fetal interface. Both in vitro and in vivo studies have highlighted the potential benefits of low-dose rapamycin in managing SM. However, given mTOR\'s critical role in energy metabolism, inhibiting it could potentially harm the pregnancy. Moreover, while low-dose rapamycin has been deemed safe for treating recurrent implant failure, its potential teratogenic effects remain uncertain due to insufficient data. In summary, rapamycin represents a double-edged sword in the treatment of SM, balancing its impact on autophagy and immune regulation. Further investigation is warranted to fully understand its implications.
    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
    食管癌是世界范围内常见的恶性肿瘤之一,以分子分类不明确和不同的临床结果为特征。PI3K/AKT/mTOR信号,人类恶性肿瘤中经常扰乱的失调途径之一,促进了针对该途径的各种抑制剂的开发,但许多ESCC患者对这些抑制剂表现出内在或适应性耐药.这里,我们旨在探讨ESCC患者对mTOR抑制剂不敏感的原因.我们通过确定各种ESCC细胞系各自的IC50值来评估对雷帕霉素的敏感性,并发现HMGA1水平低的细胞对雷帕霉素的耐受性更高。随后的实验支持了这一发现。通过转录组测序,我们鉴定了HMGA1的一个关键下游效应子FKBP12,并发现FKBP12对于HMGA1诱导的细胞对雷帕霉素的敏感性是必需的.HMGA1与ETS1相互作用,促进FKBP12的转录。最后,我们在体内实验中验证了这个调节轴,移植肿瘤中的HMGA1缺乏使它们对雷帕霉素具有抗性。因此,我们推测mTOR抑制剂治疗HMGA1或FKBP12水平降低的个体可能无效.相反,表现出升高的HMGA1或FKBP12水平的个体是mTOR抑制剂治疗的更合适的候选者。
    Esophageal carcinoma is amongst the prevalent malignancies worldwide, characterized by unclear molecular classifications and varying clinical outcomes. The PI3K/AKT/mTOR signaling, one of the frequently perturbed dysregulated pathways in human malignancies, has instigated the development of various inhibitory agents targeting this pathway, but many ESCC patients exhibit intrinsic or adaptive resistance to these inhibitors. Here, we aim to explore the reasons for the insensitivity of ESCC patients to mTOR inhibitors. We assessed the sensitivity to rapamycin in various ESCC cell lines by determining their respective IC50 values and found that cells with a low level of HMGA1 were more tolerant to rapamycin. Subsequent experiments have supported this finding. Through a transcriptome sequencing, we identified a crucial downstream effector of HMGA1, FKBP12, and found that FKBP12 was necessary for HMGA1-induced cell sensitivity to rapamycin. HMGA1 interacted with ETS1, and facilitated the transcription of FKBP12. Finally, we validated this regulatory axis in in vivo experiments, where HMGA1 deficiency in transplanted tumors rendered them resistance to rapamycin. Therefore, we speculate that mTOR inhibitor therapy for individuals exhibiting a reduced level of HMGA1 or FKBP12 may not work. Conversely, individuals exhibiting an elevated level of HMGA1 or FKBP12 are more suitable candidates for mTOR inhibitor 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
    PI3K/mTOR信号传导途径的异常激活与各种人类癌症有关。因此,靶向mTOR的抑制剂的开发引起了相当多的关注。在这项研究中,我们使用基于结构的药物设计策略来发现一种高效且激酶选择性的mTOR抑制剂24(PT-88),这表明mTOR抑制IC50值为1.2nM,对来自激酶谱分析筛选的另外195个激酶没有明显的抑制作用。PT-88显示对MCF-7细胞的选择性抑制(IC50:0.74μM),对正常细胞具有高生物安全性,其中涉及mTOR抑制诱导的自噬。在脂质体制剂中成功封装后,PT-88表现出良好的药代动力学和生物安全性,并在MCF-7皮下裸鼠模型中发挥了很大的抗肿瘤作用。我们的研究表明,使用基于结构的药物发现策略发现了高度选择性的mTOR抑制剂,并为未来的研究和开发提供了有希望的抗肿瘤候选物。
    The aberrant activation of the PI3K/mTOR signaling pathway is implicated in various human cancers. Thus, the development of inhibitors targeting mTOR has attracted considerable attention. In this study, we used a structure-based drug design strategy to discover a highly potent and kinase-selective mTOR inhibitor 24 (PT-88), which demonstrated an mTOR inhibitory IC50 value of 1.2 nM without obvious inhibition against another 195 kinases from the kinase profiling screening. PT-88 displayed selective inhibition against MCF-7 cells (IC50: 0.74 μM) with high biosafety against normal cells, in which autophagy induced by mTOR inhibition was implicated. After successful encapsulation in a lipodisc formulation, PT-88 demonstrated favorable pharmacokinetic and biosafety profiles and exerted a large antitumor effect in an MCF-7 subcutaneous bearing nude mice model. Our study shows the discovery of a highly selective mTOR inhibitor using a structure-based drug discovery strategy and provides a promising antitumor candidate for future study and development.
    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
    三阴性乳腺癌(TNBC)是一种高度异质性的缺乏雌激素受体(ER)的肿瘤,孕激素受体(PR),和人表皮生长因子受体2(HER2)的表达。它比其他亚型具有更高的侵袭性和转移性,有效的治疗策略有限,导致预后不良。磷酸肌醇3激酶(PI3K)/蛋白激酶B(AKT)/雷帕霉素的机制靶标(mTOR)信号通路在人类癌症中普遍过度激活,并有助于乳腺癌(BC)的生长,生存,扩散,和血管生成,这可能是一个有趣的治疗靶点。本文综述了PI3K/AKT/mTOR信号通路在TNBC中的激活机制,并讨论了其激活与TNBC各种亚型之间的关系。我们还报道了与该途径相关的激酶抑制剂治疗TNBC的最新临床研究。我们的综述讨论了这些抑制剂在临床应用中需要解决的问题。
    Triple-negative breast cancer (TNBC) is a highly heterogeneous tumor lacking estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) expression. It has higher aggressiveness and metastasis than other subtypes, with limited effective therapeutic strategies, leading to a poor prognosis. The phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mechanistic target of rapamycin (mTOR) signaling pathway is prevalently over-activated in human cancers and contributes to breast cancer (BC) growth, survival, proliferation, and angiogenesis, which could be an interesting therapeutic target. This review summarizes the PI3K/AKT/mTOR signaling pathway activation mechanism in TNBC and discusses the relationship between its activation and various TNBC subtypes. We also report the latest clinical studies on kinase inhibitors related to this pathway for treating TNBC. Our review discusses the issues that need to be addressed in the clinical application of these inhibitors.
    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:肝细胞癌(HCC)是一种非常普遍和致命的癌症类型,尽管药物治疗仍然是治疗的基石,治疗结果往往不能令人满意.哺乳动物雷帕霉素靶蛋白(mTOR)的药理学抑制与HCC的消退密切相关。
    方法:这里,我们共价结合AZD8055,一种有效的mTORC1/2阻断剂,与一组不饱和脂肪酸通过动态激活连接,使前药缀合物的水性自组装形成mTOR纳米阻滞剂。进行基于细胞的实验以评估纳米锁定剂对肝细胞癌(HCC)细胞的作用。建立原位和皮下HCC小鼠模型以检查其抗肿瘤活性。
    结果:在几种脂肪酸中,亚油酸共轭自组装纳米受体具有最佳的尺寸分布和优异的理化性质。与自由球员相比,在静脉内施用后,对PEG化AZD8055纳米阻滞剂(称为AZDNB)进行药代动力学优化。体内研究证实,AZDNB显着抑制皮下HCCLM3异种移植物中的肿瘤生长,肝癌-22和原位Hepa1-6肝肿瘤模型。引人注目的是,用AZDNB治疗,但不是自由球员,IFN-γ+CD8+T细胞和CD8+记忆T细胞的肿瘤内浸润增加,提示mTOR纳米受体在重塑肿瘤微环境中的潜在作用。总的来说,与脂肪酸的单一缀合将疏水性mTOR阻断剂转化为可系统注射的纳米药物,代表了提高基于mTOR抑制的癌症治疗的治疗指数的简便和可推广的策略。
    结论:与原始药物相比,本文提出的化学工程纳米阻滞剂对mTOR的抑制作用增强了对肿瘤的疗效,因此具有改善HCC患者生存结局的潜力。此外,这种来自小分子前药实体共组装的新纳米系统可以作为不同药物协同共给药的递送平台。
    背景:这项工作得到了国家自然科学基金(32171368,81721091)的支持,浙江省自然科学基金(LZ21H180001),济南市微生态生物医学实验室研究项目(JNL-2022039c和JNL-2022010B),传染病诊治国家重点实验室(zz202310),山东省自然科学基金(ZR2023ZD59).
    BACKGROUND: Hepatocellular carcinoma (HCC) is a highly prevalent and deadly type of cancer, and although pharmacotherapy remains the cornerstone of treatment, therapeutic outcomes are often unsatisfactory. Pharmacological inhibition of mammalian target of rapamycin (mTOR) has been closely associated with HCC regression.
    METHODS: Herein, we covalently conjugated AZD8055, a potent mTORC1/2 blocker, with a small panel of unsaturated fatty acids via a dynamically activating linkage to enable aqueous self-assembly of prodrug conjugates to form mTOR nanoblockers. Cell-based experiments were carried out to evaluate the effects of the nanoblocker against hepatocellular carcinoma (HCC) cells. The orthotopic and subcutaneous HCC mouse models were established to examine its antitumour activity.
    RESULTS: Among several fatty acids as promoieties, linoleic acid-conjugated self-assembling nanoblocker exhibited optimal size distribution and superior physiochemical properties. Compared with free agents, PEGylated AZD8055 nanoblocker (termed AZD NB) was pharmacokinetically optimized after intravenous administration. In vivo investigations confirmed that AZD NB significantly suppressed tumour outgrowth in subcutaneous HCCLM3 xenograft, Hepatoma-22, and orthotopic Hepa1-6 liver tumour models. Strikingly, treatment with AZD NB, but not free agent, increased intratumour infiltration of IFN-γ+CD8+ T cells and CD8+ memory T cells, suggesting a potential role of the mTOR nanoblocker to remodel the tumour microenvironment. Overall, a single conjugation with fatty acid transformed a hydrophobic mTOR blocker into a systemically injectable nanomedicine, representing a facile and generalizable strategy for improving the therapeutic index of mTOR inhibition-based cancer therapy.
    CONCLUSIONS: The mTOR inhibition by chemically engineered nanoblocker presented here had enhanced efficacy against tumours compared with the pristine drug and thus has the potential to improve the survival outcomes of patients with HCC. Additionally, this new nanosystem derived from co-assembling of small-molecule prodrug entities can serve as a delivery platform for the synergistic co-administration of distinct pharmaceutical agents.
    BACKGROUND: This work was supported by the National Natural Science Foundation of China (32171368,81721091), the Zhejiang Provincial Natural Science Foundation of China (LZ21H180001), the Jinan Provincial Laboratory Research Project of Microecological Biomedicine (JNL-2022039c and JNL-2022010B), State Key Laboratory for Diagnosis and Treatment of Infectious Diseases (zz202310), and Natural Science Foundation of Shandong Province (ZR2023ZD59).
    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
    雷帕霉素,在临床实践中建立的mTOR抑制剂,因其治疗功效而被广泛认可。Ridaforolimus,非前药rapalog,提供改善的水溶性,稳定性,和亲和力相比雷帕霉素。近年来,涉及ridaforolimus的临床试验激增。我们搜索了PubMed近十年来的ridaforolimus,并选择了ridaforolimus的临床试验,以总结ridaforolimus在临床试验中的研究进展。这些试验中的大多数探索了ridaforolimus在治疗各种肿瘤中的应用,包括子宫内膜癌,卵巢癌,前列腺癌,乳腺癌,肾细胞癌,和其他实体瘤。这些试验采用了不同的药物组合,掺入药物如博纳替尼,比卡鲁胺,dalotuzumab,MK-2206、MK-0752和紫杉烷。这些试验的结果揭示了ridaforolimus在疾病治疗中的多种潜在应用。我们的综述包括对信号通路的分析,作为单一治疗剂,它与其他药物的相容性,并评估不良事件(AE)。最后,我们建议进一步研究,以促进我们对ridaforolimus临床应用的理解。
    Rapamycin, an established mTOR inhibitor in clinical practice, is widely recognized for its therapeutic efficacy. Ridaforolimus, a non-prodrug rapalog, offers improved aqueous solubility, stability, and affinity compared to rapamycin. In recent years, there has been a surge in clinical trials involving ridaforolimus. We searched PubMed for ridaforolimus over the past decade and selected clinical trials of ridaforolimus to make a summary of the research progress of ridaforolimus in clinical trials. The majority of these trials explored the application of ridaforolimus in treating various tumors, including endometrial cancer, ovarian cancer, prostate cancer, breast cancer, renal cell carcinoma, and other solid tumors. These trials employed diverse drug combinations, incorporating agents such as ponatinib, bicalutamide, dalotuzumab, MK-2206, MK-0752, and taxanes. The outcomes of these trials unveiled the diverse potential applications of ridaforolimus in disease treatment. Our review encompassed analyses of signaling pathways, ridaforolimus as a single therapeutic agent, its compatibility in combination with other drugs, and an assessment of adverse events (AEs). We conclude by recommending further research to advance our understanding of ridaforolimus\'s clinical applications.
    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
    磷脂酰肌肽-3-激酶(PI3K)和哺乳动物雷帕霉素靶标(mTOR)最近已被鉴定为潜在的癌症靶标。在我们的工作中,设计了一个新的喹啉类似物家族,开发,并评价为PI3Kδ/mTOR的双重抑制剂。初步的生物活性分析导致发现了先导化合物5h和5e。化合物5h和5e表现出优异的抗肿瘤效力,对Ramos细胞的IC50为0.26µM和0.34µM,分别。重要的是,根据酶活性测定结果,化合物5h和5e被鉴定为PI3Kδ和mTOR的双重抑制剂,PI3Kδ的IC50值为0.042µM和0.056µM,mTOR的IC50值为0.059µM和0.073µM,分别。此外,与其他PI3K同工型相比,这些化合物对阻断PI3Kδ表现出优异的选择性(α,β,和γ),支持开发特异性靶向PI3Kδ/mTOR的抑制剂的概念。选择最有效的化合物5h用于另外的生物学测试。在0.5µM的低剂量下,一项蛋白质印迹研究证实了通过抑制PAM级联的抗癌作用,这反过来又降低了下游生物标志物pAkt(Ser473),pAkt(Thr308),和pRPS6(Ser235/236)。此外,与标准相比,它在膜联蛋白-V测定的早期(10.03倍)和晚期(17.95倍)阶段增加了细胞凋亡。此外,在ELISA试验中,化合物5h可显著增加p53、caspase-3、caspase-9的表达和Bax/BCl-2比值。基于这些结果,似乎5h可能激活内在的凋亡途径,进而引发细胞死亡。此外,抗癌作用可归因于抑制PI3Kδ/mTOR,如对接交互所示。最后,它证明了改善的体外代谢稳定性,并通过了计算机模拟ADMET/药物相似度测试。该概况推荐5小时用于动物癌症模型中的未来体内PK-PD和功效研究。
    Phosphatidylinositide-3-kinase (PI3K) and the mammalian target of rapamycin (mTOR) have recently been identified as potential cancer targets. In our work, a new family of quinoline analogues was designed, developed, and evaluated as dual inhibitors of PI3Kδ/mTOR. The preliminary biological activity analysis led to the discovery of the lead compounds 5h and 5e. Compounds 5h and 5e exhibited excellent anti-tumor potency with IC50 of 0.26 µM and 0.34 µM against Ramos cells, respectively. Importantly, based on the enzymatic activity assay results, compounds 5h and 5e were identified as dual inhibitors of PI3Kδ and mTOR, with IC50 values of 0.042 µM and 0.056 µM for PI3Kδ and 0.059 µM and 0.073 µM for mTOR, respectively. Furthermore, these compounds showed superior selectivity for blocking PI3Kδ compared to other PI3K isoforms (α, β, and γ), supporting the concept of developing inhibitors that specifically target PI3Kδ/mTOR. The most effective compound 5h was chosen for additional biological testing. At a low dose of 0.5 µM, a western blot investigation confirmed the anticancer effects by inhibiting the PAM cascade, which in turn reduced downstream biomarkers pAkt (Ser473), pAkt (Thr308), and pRPS6 (Ser235/236). Furthermore, it increased apoptosis at the early (10.03 times) and late (17.95 times) stages in the Annexin-V assay as compared to the standard. In addition, the expression of p53, caspase-3, caspase-9, and the Bax/BCl-2 ratio were all significantly increased by compound 5h in the ELISA assay. Based on these results, it appears that 5h may activate the intrinsic apoptosis pathway, which in turn triggers cell death. Furthermore, the anticancer effects could be attributed to the inhibition of PI3Kδ/mTOR, as shown by docking interactions. Lastly, it demonstrated improved in vitro metabolic stability and passed the in silico ADMET/drug-likeness test. This profile recommends 5h for future in vivo PK-PD and efficacy investigations in animal cancer models.
    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
    终末期鼻咽癌(NPC)的生存率不理想。化疗的益处有限和靶向药物的稀缺是鼻咽癌的主要挑战。迫切需要治疗晚期NPC的新方法。在这项研究中,我们探索了PI3K/mTOR双重抑制剂,PQR309在鼻咽癌中发挥了良好的抗肿瘤作用,并对吉西他滨的反应敏感。我们观察到,与正常鼻咽细胞系相比,14个NPC细胞系中的PI3K表达呈阳性且升高。PI3K水平较高的NPC患者预后较差。我们随后表明,PQR309单独有效地降低了生存能力,侵入性,小鼠异种移植模型中NPC细胞的迁移能力和肿瘤的发展,和剂量依赖性地诱导细胞凋亡。更重要的是,PQR309在体内和体外均显着增强了吉西他滨的抗NPC功能。机械上,PQR309通过增加caspase途径依赖性细胞凋亡使NPC对吉西他滨敏感,阻断GSK-3β和STAT3/HSP60信号,消融上皮-间质转换。因此,使用PQR309靶向PI3K/mTOR可能是促进NPC对吉西他滨反应的治疗选择,为靶向药物联合化疗治疗鼻咽癌提供理论基础。
    End-stage nasopharyngeal carcinoma (NPC) has unsatisfactory survival. The limited benefit of chemotherapy and the scarcity of targeted drugs are major challenges in NPC. New approaches to treat late-stage NPC are urgently required. In this study, we explored whether the dual PI3K/mTOR inhibitor, PQR309, exerted a favorable antineoplastic effect and sensitized the response to gemcitabine in NPC. We observed that PI3K expression was positive and elevated in 14 NPC cell lines compared with that in normal nasopharygeal cell lines. Patients with NPC with higher PI3K levels displayed poorer prognosis. We subsequently showed that PQR309 alone effectively decreased the viability, invasiveness, and migratory capability of NPC cells and neoplasm development in mice xenograft models, and dose-dependently induced apoptosis. More importantly, PQR309 remarkably strengthened the anti-NPC function of gemcitabine both in vivo and in vitro. Mechanistically, PQR309 sensitized NPC to gemcitabine by increasing caspase pathway-dependent apoptosis, blocking GSK-3β and STAT3/HSP60 signaling, and ablating epithelial-mesenchyme transition. Thus, targeting PI3K/mTOR using PQR309 might represent a treatment option to promote the response to gemcitabine in NPC, and provides a theoretical foundation for the study of targeted drugs combined with chemotherapy for NPC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号