DNA-Activated Protein Kinase

DNA 活化蛋白激酶
  • 文章类型: Journal Article
    DNA-PKcs是参与DNA修复和反应途径的关键蛋白质靶标,其异常活性与各种癌症的发生和进展密切相关。在这项研究中,我们采用了基于深度学习的筛选和基于分子动力学(MD)模拟的管道,确定八个候选DNA-PKcs目标。随后的实验揭示了三种小分子(5025-0002、M769-1095和V008-1080)对DNA-PKcs介导的细胞增殖的有效抑制。这些分子表现出抗癌活性,IC50(抑制浓度为50%)值为152.6μM,30.71μM,和74.84μM,分别。值得注意的是,V008-1080增强由CRISPR/Cas9介导的同源定向修复(HDR),同时抑制非同源末端连接(NHEJ)效率。对结构-活性关系的进一步研究揭示了这些小分子与DNA-PKcs之间的结合位点和关键相互作用。这是DeepBindGCN_RG在实际药物筛选任务中的初次运用,一种新型DNA-PKcs抑制剂的成功发现证明了其作为筛选流程中核心成分的有效性。此外,这项研究为探索新的抗癌疗法和通过靶向DNA-PKcs推进基因编辑技术的发展提供了重要的见解。
    DNA-PKcs is a crucial protein target involved in DNA repair and response pathways, with its abnormal activity closely associated with the occurrence and progression of various cancers. In this study, we employed a deep learning-based screening and molecular dynamics (MD) simulation-based pipeline, identifying eight candidates for DNA-PKcs targets. Subsequent experiments revealed the effective inhibition of DNA-PKcs-mediated cell proliferation by three small molecules (5025-0002, M769-1095, and V008-1080). These molecules exhibited anticancer activity with IC50 (inhibitory concentration at 50%) values of 152.6 μM, 30.71 μM, and 74.84 μM, respectively. Notably, V008-1080 enhanced homology-directed repair (HDR) mediated by CRISPR/Cas9 while inhibiting non-homologous end joining (NHEJ) efficiency. Further investigations into the structure-activity relationships unveiled the binding sites and critical interactions between these small molecules and DNA-PKcs. This is the first application of DeepBindGCN_RG in a real drug screening task, and the successful discovery of a novel DNA-PKcs inhibitor demonstrates its efficiency as a core component in the screening pipeline. Moreover, this study provides important insights for exploring novel anticancer therapeutics and advancing the development of gene editing techniques by targeting DNA-PKcs.
    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
    部署DNA损伤反应(DDR)对抗各种形式的DNA损伤,确保基因组稳定性。癌细胞对基因组不稳定性的倾向提供了通过抑制DDR途径选择性杀死癌细胞的治疗机会。DNA依赖性蛋白激酶(DNA-PK),核丝氨酸/苏氨酸激酶,对于DNA双链断裂(DSB)修复中的非同源末端连接(NHEJ)途径至关重要。因此,靶向DNA-PK是一种有前途的癌症治疗策略.本文综述了DNA-PK及其相关大蛋白的结构,以及DNA-PK抑制剂的发展过程,以及其临床应用的最新进展。我们强调我们对基于不同支架的DNA-PK抑制剂的开发过程和结构-活性关系(SARs)的分析。我们希望这篇综述将为未来寻求开发新的DNA-PK抑制剂的研究人员提供实用信息。
    The deployment of DNA damage response (DDR) combats various forms of DNA damage, ensuring genomic stability. Cancer cells\' propensity for genomic instability offers therapeutic opportunities to selectively kill cancer cells by suppressing the DDR pathway. DNA-dependent protein kinase (DNA-PK), a nuclear serine/threonine kinase, is crucial for the non-homologous end joining (NHEJ) pathway in the repair of DNA double-strand breaks (DSBs). Therefore, targeting DNA-PK is a promising cancer treatment strategy. This review elaborates on the structures of DNA-PK and its related large protein, as well as the development process of DNA-PK inhibitors, and recent advancements in their clinical application. We emphasize our analysis of the development process and structure-activity relationships (SARs) of DNA-PK inhibitors based on different scaffolds. We hope this review will provide practical information for researchers seeking to develop novel DNA-PK inhibitors in the future.
    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:靶向DNA损伤修复因子,如DNA依赖性蛋白激酶催化亚基(DNA-PKcs),可能提供有效治疗多发性骨髓瘤(MM)的机会。结合DNA损伤诱导剂,该策略已被证明可以通过升高的细胞溶质DNA水平激活cGAS-STING途径来部分改善化疗。然而,由于cGAS主要被细胞核中的染色质隔离,目前尚不清楚cGAS如何在DNA损伤后从染色质中释放并易位到细胞质中,导致cGAS-STING激活。
    方法:我们通过进行质谱和机制研究,检查了DNA-PKcs抑制对cGAS-STING介导的MM化学敏感性的作用。
    结果:这里,我们发现DNA-PKcs抑制通过激活cGAS-STING信号增强DNA损伤诱导剂阿霉素诱导的抗MM作用。MM细胞中的cGAS-STING激活部分通过IRF3-NOXA-BAK轴引起细胞死亡,并诱导巨噬细胞的M1极化。此外,这种激活不是由经典的非同源末端连接(c-NHEJ)缺陷引起的。相反,多柔比星诱导的DNA损伤,抑制DNA-PKcs促进cGAS从细胞质染色质片段释放,并增加细胞溶质cGAS和DNA的量,激活cGAS-STING。
    结论:抑制DNA-PKcs可以通过去螯合cGAS来改善多柔比星治疗MM的疗效。
    BACKGROUND: Targeting DNA damage repair factors, such as DNA-dependent protein kinase catalytic subunit (DNA-PKcs), may offer an opportunity for effective treatment of multiple myeloma (MM). In combination with DNA damage-inducing agents, this strategy has been shown to improve chemotherapies partially via activation of cGAS-STING pathway by an elevated level of cytosolic DNA. However, as cGAS is primarily sequestered by chromatin in the nucleus, it remains unclear how cGAS is released from chromatin and translocated into the cytoplasm upon DNA damage, leading to cGAS-STING activation.
    METHODS: We examined the role of DNA-PKcs inhibition on cGAS-STING-mediated MM chemosensitivity by performing mass spectrometry and mechanism study.
    RESULTS: Here, we found DNA-PKcs inhibition potentiated DNA damage-inducing agent doxorubicin-induced anti-MM effect by activating cGAS-STING signaling. The cGAS-STING activation in MM cells caused cell death partly via IRF3-NOXA-BAK axis and induced M1 polarization of macrophages. Moreover, this activation was not caused by defective classical non-homologous end joining (c-NHEJ). Instead, upon DNA damage induced by doxorubicin, inhibition of DNA-PKcs promoted cGAS release from cytoplasmic chromatin fragments and increased the amount of cytosolic cGAS and DNA, activating cGAS-STING.
    CONCLUSIONS: Inhibition of DNA-PKcs could improve the efficacy of doxorubicin in treatment of MM by de-sequestrating cGAS in damaged chromatin.
    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
    驱动神经胶质瘤进展的因素仍然知之甚少。这里,表观遗传调节因子TRIM24被确定为神经胶质瘤进展的驱动因素,其中TRIM24过表达促进HRasV12间变性星形细胞瘤(AA)进展为上皮样GBM(Ep-GBM)样肿瘤。TRIM24与HRasV12的共转染还诱导具有肿瘤蛋白p53基因(TP53)敲低的人神经干细胞(hNSC)的Ep-GBM样转化。此外,TRIM24在临床Ep-GBM标本中高表达。使用单细胞RNA测序(scRNA-Seq),作者表明TRIM24过表达影响肿瘤内异质性和肿瘤微环境.机械上,HRasV12激活RNA输出(PHAX)的磷酸化衔接子,并上调U3小核仁RNA(U3snoRNA)以招募Ku依赖性DNA依赖性蛋白激酶催化亚基(DNA-PKcs)。过表达的TRIM24也被PHAX招募到U3snoRNA,从而促进TRIM24在S767/768残基处的DNA-PKcs磷酸化。磷酸化TRIM24诱导表观基因组和转录因子网络重编程并促进Ep-GBM样转化。用小分子抑制剂NU7441靶向DNA-PKcs与替莫唑胺协同作用以降低Ep-GBM致瘤性并延长动物存活。这些发现为Ep-GBM样转化的表观遗传调控提供了新的见解,并为Ep-GBM患者提出了潜在的治疗策略。
    The factors driving glioma progression remain poorly understood. Here, the epigenetic regulator TRIM24 is identified as a driver of glioma progression, where TRIM24 overexpression promotes HRasV12 anaplastic astrocytoma (AA) progression into epithelioid GBM (Ep-GBM)-like tumors. Co-transfection of TRIM24 with HRasV12 also induces Ep-GBM-like transformation of human neural stem cells (hNSCs) with tumor protein p53 gene (TP53) knockdown. Furthermore, TRIM24 is highly expressed in clinical Ep-GBM specimens. Using single-cell RNA-sequencing (scRNA-Seq), the authors show that TRIM24 overexpression impacts both intratumoral heterogeneity and the tumor microenvironment. Mechanically, HRasV12 activates phosphorylated adaptor for RNA export (PHAX) and upregulates U3 small nucleolar RNAs (U3 snoRNAs) to recruit Ku-dependent DNA-dependent protein kinase catalytic subunit (DNA-PKcs). Overexpressed TRIM24 is also recruited by PHAX to U3 snoRNAs, thereby facilitating DNA-PKcs phosphorylation of TRIM24 at S767/768 residues. Phosphorylated TRIM24 induces epigenome and transcription factor network reprogramming and promotes Ep-GBM-like transformation. Targeting DNA-PKcs with the small molecule inhibitor NU7441 synergizes with temozolomide to reduce Ep-GBM tumorigenicity and prolong animal survival. These findings provide new insights into the epigenetic regulation of Ep-GBM-like transformation and suggest a potential therapeutic strategy for patients with Ep-GBM.
    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:放射性肺纤维化(RIPF)是一种慢性,进步,放疗后发展的不可逆的肺间质疾病。尽管先前的一些研究集中在肺上皮细胞中上皮-间质转化(EMT)的机制上,参与这一过程的基本因素仍然知之甚少。当细胞遭受辐射诱导的损伤时,DNA依赖性蛋白激酶催化亚基(DNA-PKcs)表现出很强的修复能力;DNA-PKcs在RIPF期间是否调节EMT尚不清楚。
    目的:探讨DNA-PKcs在RIPF中的作用及分子机制,为利用DNA-PKcs靶向药物预防RIPF提供重要的理论依据。
    方法:通过Cas9/sgRNA技术产生DNA-PKcs敲除(DPK-/-)小鼠,并以20Gy剂量进行全胸部电离辐射(IR)。在整个胸部IR之前,小鼠胃内给药DNA-PKcs靶向药物VND3207.在IR后1和5个月收集肺组织。
    结果:肺纤维化(PF)患者中DNA-PKcs的表达较低。DNA-PKcs缺乏通过促进肺上皮细胞的EMT而显著加剧RIPF。机械上,shRNA或抑制剂NU7441的DNA-PKcs缺失维持了Twist1的蛋白质稳定性。此外,AKT1介导DNA-PKcs与Twist1的相互作用。胰岛素样生长因子-1(IGF-1)阻断了DNA-PKcs缺失引起的Twist1高表达和EMT相关变化,AKT1激动剂。辐射防护药物VND3207通过刺激DNA-PKcs的激酶活性来预防IR诱导的EMT并减轻小鼠的RIPF。
    结论:我们的研究阐明了DNA-PKcs在RIPF中的关键作用和机制,并表明它可能是预防RIPF的潜在靶标。
    BACKGROUND: Radiation-induced pulmonary fibrosis (RIPF) is a chronic, progressive, irreversible lung interstitial disease that develops after radiotherapy. Although several previous studies have focused on the mechanism of epithelial-mesenchymal transition (EMT) in lung epithelial cells, the essential factors involved in this process remain poorly understood. The DNA-dependent protein kinase catalytic subunit (DNA-PKcs) exhibits strong repair capacity when cells undergo radiation-induced damage; whether DNA-PKcs regulates EMT during RIPF remains unclear.
    OBJECTIVE: To investigate the role and molecular mechanism of DNA-PKcs in RIPF and provide an important theoretical basis for utilising DNA-PKcs-targeted drugs for preventing RIPF.
    METHODS: DNA-PKcs knockout (DPK-/-) mice were generated via the Cas9/sgRNA technique and subjected to whole chest ionizing radiation (IR) at a 20 Gy dose. Before whole chest IR, the mice were intragastrically administered the DNA-PKcs-targeted drug VND3207. Lung tissues were collected at 1 and 5 months after IR.
    RESULTS: The expression of DNA-PKcs is low in pulmonary fibrosis (PF) patients. DNA-PKcs deficiency significantly exacerbated RIPF by promoting EMT in lung epithelial cells. Mechanistically, DNA-PKcs deletion by shRNA or inhibitor NU7441 maintained the protein stability of Twist1. Furthermore, AKT1 mediated the interaction between DNA-PKcs and Twist1. High Twist1 expression and EMT-associated changes caused by DNA-PKcs deletion were blocked by insulin-like growth factor-1 (IGF-1), an AKT1 agonist. The radioprotective drug VND3207 prevented IR-induced EMT and alleviated RIPF in mice by stimulating the kinase activity of DNA-PKcs.
    CONCLUSIONS: Our study clarified the critical role and mechanism of DNA-PKcs in RIPF and showed that it could be a potential target for preventing RIPF.
    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
    尽管DNA-PK抑制剂(DNA-PK-i)已用于癌症治疗的临床试验,DNA-PK-i在肿瘤细胞抑制中的生物标志物和作用机制尚不清楚.这里,我们观察到,低剂量的DNA-PK-i和PARP抑制剂(PARP-i)可合成抑制BRCA缺陷型肿瘤细胞,而不诱导DNA双链断裂(DSB).相反,我们发现一部分DNA-PK位于核仁内部,我们没有观察到明显的DSB。此外,Ku蛋白识别pre-rRNA,其促进DNA-PKcs自磷酸化而不依赖于DNA损伤。核糖体蛋白也被DNA-PK磷酸化,调节前rRNA生物发生。此外,DNA-PK-i与PARP-i一起作用以抑制前rRNA生物发生和肿瘤细胞生长。总的来说,我们的研究揭示了DNA-PK-i在肿瘤抑制中的DNA损伤修复非依赖性作用.
    Although DNA-PK inhibitors (DNA-PK-i) have been applied in clinical trials for cancer treatment, the biomarkers and mechanism of action of DNA-PK-i in tumor cell suppression remain unclear. Here, we observed that a low dose of DNA-PK-i and PARP inhibitor (PARP-i) synthetically suppresses BRCA-deficient tumor cells without inducing DNA double-strand breaks (DSBs). Instead, we found that a fraction of DNA-PK localized inside of nucleoli, where we did not observe obvious DSBs. Moreover, the Ku proteins recognize pre-rRNA that facilitates DNA-PKcs autophosphorylation independent of DNA damage. Ribosomal proteins are also phosphorylated by DNA-PK, which regulates pre-rRNA biogenesis. In addition, DNA-PK-i acts together with PARP-i to suppress pre-rRNA biogenesis and tumor cell growth. Collectively, our studies reveal a DNA damage repair-independent role of DNA-PK-i in tumor suppression.
    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
    作为激活STING-TBK1信号级联的主要DNA传感器,cGAS主要存在于细胞质中。最近的许多报道表明,cGAS在细胞核中也起着关键作用。我们以前的工作首次证明了cGAS在DNA损伤发生后易位到细胞核并抑制同源重组(HR),DNA双链断裂(DSB)修复的两条主要途径之一。然而,核cGAS是否调节另一个DSB修复途径,非同源末端连接(NHEJ),可以进一步分为不易出错的规范NHEJ(c-NHEJ)和更诱变的替代NHEJ(alt-NHEJ)亚途径,没有被定性。这里,我们证明cGAS使两个NHEJ亚途径的平衡向c-NHEJ倾斜。机械上,cGAS-Ku80复合物增强了DNA-PKcs和去泛素酶USP7之间的相互作用,以提高DNA-PKcs蛋白的稳定性,从而促进c-NHEJ。相比之下,cGAS-Ku80复合物通过直接结合Polθ的启动子来抑制alt-NHEJ,从而抑制其转录。一起,这些发现揭示了核cGAS在调节DSB修复中的新功能,表明cGAS在细胞核中的存在对维持基因组完整性也很重要。
    As the major DNA sensor that activates the STING-TBK1 signaling cascade, cGAS is mainly present in the cytosol. A number of recent reports have indicated that cGAS also plays critical roles in the nucleus. Our previous work demonstrated for the first time that cGAS is translocated to the nucleus upon the occurrence of DNA damage and inhibits homologous recombination (HR), one of the two major pathways of DNA double strand break (DSB) repair. However, whether nuclear cGAS regulates the other DSB repair pathway, nonhomologous end joining (NHEJ), which can be further divided into the less error-prone canonical NHEJ (c-NHEJ) and more mutagenic alternative NHEJ (alt-NHEJ) subpathways, has not been characterized. Here, we demonstrated that cGAS tipped the balance of the two NHEJ subpathways toward c-NHEJ. Mechanistically, the cGAS-Ku80 complex enhanced the interaction between DNA-PKcs and the deubiquitinase USP7 to improve DNA-PKcs protein stability, thereby promoting c-NHEJ. In contrast, the cGAS-Ku80 complex suppressed alt-NHEJ by directly binding to the promoter of Polθ to suppress its transcription. Together, these findings reveal a novel function of nuclear cGAS in regulating DSB repair, suggesting that the presence of cGAS in the nucleus is also important in the maintenance of genome integrity.
    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
    蛋白磷酸酶1催化亚基γ(PPP1CC)促进DNA修复和肿瘤的发生发展,然而,其潜在机制仍不清楚。本研究探讨了PPP1CC参与DNA修复的分子机制及其潜在的临床意义。PPP1CC的高表达与鼻咽癌(NPC)患者的放射抵抗和不良预后显着相关。机理研究表明,PPP1CC通过促进DNA-PK全酶的形成与Ku70/Ku80异二聚体结合并激活DNA-PKcs,增强了非同源末端连接(NHEJ)介导的DNA修复并导致了放射抗性。重要的是,含有BRCA1-BRCA2的复合物亚基3(BRCC3)与PPP1CC相互作用,通过去除Lys234处的K48连接的聚泛素链以防止PPP1CC降解来增强其稳定性。因此,BRCC3帮助过表达的PPP1CC维持其高蛋白质水平,从而维持DNA修复能力和放射抗性的提高。我们的研究确定了PPP1CC促进NHEJ介导的DNA修复和放射抗性的分子机制,提示BRCC3-PPP1CC-Ku70轴是提高放疗疗效的潜在治疗靶点。
    Protein phosphatase 1 catalytic subunit gamma (PPP1CC) promotes DNA repair and tumor development and progression, however, its underlying mechanisms remain unclear. This study investigated the molecular mechanism of PPP1CC\'s involvement in DNA repair and the potential clinical implications. High expression of PPP1CC was significantly correlated with radioresistance and poor prognosis in human nasopharyngeal carcinoma (NPC) patients. The mechanistic study revealed that PPP1CC bound to Ku70/Ku80 heterodimers and activated DNA-PKcs by promoting DNA-PK holoenzyme formation, which enhanced nonhomologous end junction (NHEJ) -mediated DNA repair and led to radioresistance. Importantly, BRCA1-BRCA2-containing complex subunit 3 (BRCC3) interacted with PPP1CC to enhance its stability by removing the K48-linked polyubiquitin chain at Lys234 to prevent PPP1CC degradation. Therefore, BRCC3 helped the overexpressed PPP1CC to maintain its high protein level, thereby sustaining the elevation of DNA repair capacity and radioresistance. Our study identified the molecular mechanism by which PPP1CC promotes NHEJ-mediated DNA repair and radioresistance, suggesting that the BRCC3-PPP1CC-Ku70 axis is a potential therapeutic target to improve the efficacy of radiotherapy.
    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
    大多数癌症患者接受放射治疗作为其治疗方案的一部分,无论是使用外部束治疗还是局部递送的放射性同位素。虽然通常有效,一些肿瘤的放疗控制不充分,放疗对癌症幸存者有显著的短期和长期毒性.近年来,已经获得了对通过放射或其他癌症疗法引入的DNA断裂的细胞应答中所涉及的分子机制的见解,并且操纵这些应答以增强肿瘤细胞杀伤或降低正常组织毒性的方法是非常感兴趣的。这里,我们报告了XRD-0394的鉴定和初步表征,这是一种有效和特异性的两种DNA损伤反应激酶的双重抑制剂,ATM和DNA-PKcs。这种口服生物可利用的分子在体外和体内治疗性电离辐射的背景下显示出显著增强的肿瘤细胞杀伤作用。XRD-0394还增强了拓扑异构酶I抑制剂在体外的有效性。此外,在缺乏BRCA1/2的细胞中,XRD-0394显示出单一试剂活性和与PARP抑制剂组合的协同作用。使用XRD-0394与RT组合的Ia期临床试验(NCT05002140)已经完成。这些结果为XRD-0394与RT结合的未来临床试验提供了理论基础。PARP抑制剂和拓扑异构酶I抑制剂的靶向递送。
    A majority of patients with cancer receive radiotherapy as part of their treatment regimens whether using external beam therapy or locally-delivered radioisotopes. While often effective, some tumors are inadequately controlled with radiation and radiotherapy has significant short-term and long-term toxicities for cancer survivors. Insights into molecular mechanisms involved in cellular responses to DNA breaks introduced by radiation or other cancer therapies have been gained in recent years and approaches to manipulate these responses to enhance tumor cell killing or reduce normal tissue toxicity are of great interest. Here, we report the identification and initial characterization of XRD-0394, a potent and specific dual inhibitor of two DNA damage response kinases, ATM and DNA-PKcs. This orally bioavailable molecule demonstrates significantly enhanced tumor cell kill in the setting of therapeutic ionizing irradiation in vitro and in vivo. XRD-0394 also potentiates the effectiveness of topoisomerase I inhibitors in vitro. In addition, in cells lacking BRCA1/2 XRD-0394 shows single-agent activity and synergy in combination with PARP inhibitors. A phase Ia clinical trial (NCT05002140) with XRD-0394 in combination with radiotherapy has completed. These results provide a rationale for future clinical trials with XRD-0394 in combination with radiotherapy, PARP inhibitors, and targeted delivery of topoisomerase I inhibitors.
    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
    在这项工作中,合理设计了一系列新的杂三环DNA-PK抑制剂,合成,并评估其生物活性。在DNA-PK生化检测中,大多数化合物显示出有效的酶活性,IC50值在0.11和71.5nM之间。其中,SK10表现出最有效的DNA-PK抑制活性(IC50=0.11nM)。作用机制的研究表明,SK10可以降低γH2A。当与多柔比星组合时,X表达水平和显示针对Jurkat细胞的最佳协同抗增殖活性(IC50=25nM)。重要的是,在CT26和B16-F10荷瘤小鼠模型中,SK10与化疗药物阿霉素的联合治疗,PD-L1抗体,和SWS1(一种有效的PD-L1小分子抑制剂)表现出优异的协同抗癌作用和潜在的免疫调节作用。此外,SK10具有良好的体内药代动力学特性[例如,口服生物利用度(F)=31.8%]。一起来看,SK10代表具有抗肿瘤免疫作用和有利的药代动力学的新型杂三环DNA-PK抑制剂。
    In this work, a novel series of heterotricyclic DNA-PK inhibitors were rationally designed, synthesized, and assessed for their biological activity. In the DNA-PK biochemical assay, most compounds displayed potent enzymatic activity, with IC50 values between 0.11 and 71.5 nM. Among them, SK10 exhibited the most potent DNA-PK-inhibitory activity (IC50 = 0.11 nM). Studies of the mechanism of action indicated that SK10 could lower γH2A.X expression levels and demonstrate optimal synergistic antiproliferative activity against Jurkat cells (IC50 = 25 nM) when combined with doxorubicin. Importantly, in CT26 and B16-F10 tumor-bearing mouse models, the combination therapies of SK10 with chemotherapeutic drug doxorubicin, a PD-L1 antibody, and SWS1 (a potent PD-L1 small-molecule inhibitor) demonstrated superior synergistic anticancer and potential immunomodulatory effects. Furthermore, SK10 possessed favorable in vivo pharmacokinetic properties [e.g., oral bioavailability (F) = 31.8%]. Taken together, SK10 represents a novel heterotricyclic DNA-PK inhibitor with antitumor immune effects and favorable pharmacokinetics.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号