CHK1

CHK1
  • 文章类型: Journal Article
    ATR-Chk1通路在细胞对DNA损伤和复制应激的反应中是必不可少的。而长链非编码RNA(lncRNAs)在调节该途径中的作用仍然未知。在这项研究中,我们鉴定了ATR和Chk1相互作用的lncRNA(ACIL,也称为LRRC75A-AS1或SNHG29),在DNA损伤时,ATR会促进Chk1的磷酸化。高ACIL水平与对DNA损伤剂的化学抗性和乳腺癌患者的不良预后相关。ACIL敲除在体外和体内使乳腺癌细胞对DNA损伤药物敏感。ACIL通过诱导细胞周期停滞保护癌细胞免受DNA损伤,稳定复制叉并抑制计划外的源发射,从而防范复制灾难并有助于DNA损伤修复。这些发现证明了lncRNA依赖性的激活ATR-Chk1通路的机制,并强调了利用ACIL作为化疗敏感性的预测生物标志物的潜力。以及靶向ACIL逆转乳腺癌化疗耐药。
    The ATR-Chk1 pathway is essential in cellular responses to DNA damage and replication stress, whereas the role of long noncoding RNAs (lncRNAs) in regulating this pathway remains largely unknown. In this study, we identify an ATR and Chk1 interacting lncRNA (ACIL, also known as LRRC75A-AS1 or SNHG29), which promotes the phosphorylation of Chk1 by ATR upon DNA damages. High ACIL levels are associated with chemoresistance to DNA damaging agents and poor outcome of breast cancer patients. ACIL knockdown sensitizes breast cancer cells to DNA damaging drugs in vitro and in vivo. ACIL protects cancer cells against DNA damages by inducing cell cycle arrest, stabilizing replication forks and inhibiting unscheduled origin firing, thereby guarding against replication catastrophe and contributing to DNA damage repair. These findings demonstrate a lncRNA-dependent mechanism of activating the ATR-Chk1 pathway and highlight the potential of utilizing ACIL as a predictive biomarker for chemotherapy sensitivity, as well as targeting ACIL to reverse chemoresistance in breast 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
    多药耐药性是使用基于顺铂(DDP)的辅助化疗和EGFR-酪氨酸激酶抑制剂(TKIs)等疗法治疗非小细胞肺癌(NSCLC)的实质性障碍。Aaptamine-7(AP-7),从Aaptosaaptos海绵中提取的苯并萘啶生物碱,已显示出广谱的抗肿瘤活性。然而,AP-7联合DDP在多药耐药NSCLC中的抗癌活性及其分子机制尚不清楚。我们的研究表明,AP-7可以增强DDP对多药耐药NSCLC细胞的生长抑制活性。AP-7显著破坏DDP诱导的细胞周期停滞并放大这些细胞中DDP诱导的DNA损伤效应。此外,AP-7和DDP的组合下调Chk1激活,中断DNA损伤修复依赖性Chk1/CDK1通路,并有助于克服多药耐药NSCLC细胞和吉非替尼耐药异种移植小鼠模型的耐药性和促进细胞凋亡。总之,AP-7似乎通过阻断Chk1信号通路增强DDP诱导的DNA损伤在多药耐药NSCLC中,从而增强生长抑制,在体外和体内。这些结果表明AP-7作为DDP增敏剂在治疗多药耐药NSCLC中的潜在用途。
    Multidrug resistance is a substantial obstacle in treating non-small cell lung cancer (NSCLC) with therapies like cisplatin (DDP)-based adjuvant chemotherapy and EGFR-tyrosine kinase inhibitors (TKIs). Aaptamine-7 (AP-7), a benzonaphthyridine alkaloid extracted from Aaptos aaptos sponge, has been shown to exhibit a broad spectrum of anti-tumor activity. However, the anti-cancer activity of AP-7 in combination with DDP and its molecular mechanisms in multidrug-resistant NSCLC are not yet clear. Our research indicates that AP-7 bolsters the growth inhibition activity of DDP on multidrug-resistant NSCLC cells. AP-7 notably disrupts DDP-induced cell cycle arrest and amplifies DDP-induced DNA damage effects in these cells. Furthermore, the combination of AP-7 and DDP downregulates Chk1 activation, interrupts the DNA damage repair-dependent Chk1/CDK1 pathway, and helps to overcome drug resistance and boost apoptosis in multidrug-resistant NSCLC cells and a gefitinib-resistant xenograft mice model. In summary, AP-7 appears to enhance DDP-induced DNA damage by impeding the Chk1 signaling pathway in multidrug-resistant NSCLC, thereby augmenting growth inhibition, both in vitro and in vivo. These results indicate the potential use of AP-7 as a DDP sensitizer in the treatment of multidrug-resistant NSCLC.
    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
    应激是促进阿尔茨海默病(AD)发病的重要启动因素。然而,应激导致AD样认知障碍的机制仍有待阐明。这里,我们证明,在应激激素促肾上腺皮质激素释放因子(CRF)处理的细胞和暴露于慢性束缚应激的小鼠的大脑中,DNA损伤增加。DNA损伤的积累驱动细胞周期检查点蛋白激酶1(Chk1)的激活,PP2A(CIP2A)癌性抑制剂的上调,tau过度磷酸化,Aβ生产过剩,最终导致突触损伤和认知缺陷。特异性抑制剂靶向Chk1的药物干预和维生素C的DNA损伤,在慢性应激动物模型中抑制DNA损伤-Chk1-CIP2A信号通路,反过来减弱AD样病变,突触损伤和认知缺陷。我们的研究揭示了应激诱导的AD样病变的新分子机制,并提供了针对该信号通路的有效预防和治疗策略。
    Stress is an important initiating factor in promoting Alzheimer\'s disease (AD) pathogenesis. However, the mechanism by which stress induces AD-like cognitive impairment remains to be clarified. Here, we demonstrate that DNA damage is increased in stress hormone Corticotropin-releasing factor (CRF)-treated cells and in brains of mice exposed to chronic restraint stress. Accumulation of DNA damage drives activation of cell cycle checkpoint protein kinase 1 (Chk1), upregulation of cancerous inhibitor of PP2A (CIP2A), tau hyperphosphorylation, and Aβ overproduction, eventually resulting in synaptic impairment and cognitive deficits. Pharmacological intervention targeting Chk1 by specific inhibitor and DNA damage by vitamin C, suppress DNA damage-Chk1-CIP2A signaling pathway in chronic stress animal model, which in turn attenuate AD-like pathologies, synaptic impairments and cognitive deficits. Our study uncovers a novel molecular mechanism of stress-induced AD-like pathologies and provides effective preventive and therapeutic strategies targeting this signaling pathway.
    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)

  • 文章类型: Journal Article
    尽管最近在诊断和治疗方面取得了进展,胰腺导管腺癌(PDAC),胰腺癌最常见的类型,仍然是最致命的癌症,五年生存率很低。迫切需要开发新的疗法来解决这个问题。在这项研究中,我们通过修饰肿瘤抑制miRNA开发了一种治疗策略,miR-15a和miR-194,与化疗吉西他滨(Gem)产生宝石修饰的模拟物,Gem-miR-15a和Gem-miR-194。在一组PDAC细胞系中,我们发现Gem-miR-15a和Gem-miR-194诱导细胞周期停滞和凋亡,并且这些模拟物是有效的抑制剂,其IC50值比它们的天然对应物或单独的Gem低几百倍。此外,我们发现Gem-miR-15a和Gem-miR-194通过下调Gem-miR-15a的几个关键靶标(包括WEE1,CHK1,BMI1和YAP1)的表达来保留miRNA功能,和FOXA1代表Gem-miR-194。我们还发现,与Gem相比,我们的Gem修饰的miRNA模拟物在患者来源的PDAC类器官中表现出增强的功效。此外,我们观察到Gem-miR-15a在体内显著抑制PDAC肿瘤生长,而未观察到任何明显的毒性迹象.总的来说,我们的结果证明了宝石修饰的miRNA作为PDAC治疗策略的治疗潜力.
    Despite the recent advancement in diagnosis and therapy, pancreatic ductal adenocarcinoma (PDAC), the most common type of pancreatic cancer, is still the most lethal cancer with a low five-year survival rate. There is an urgent need to develop new therapies to address this issue. In this study, we developed a treatment strategy by modifying tumor suppressor miRNAs, miR-15a and miR-194, with the chemotherapeutic gemcitabine (Gem) to create Gem-modified mimics, Gem-miR-15a and Gem-miR-194, respectively. In a panel of PDAC cell lines, we found that Gem-miR-15a and Gem-miR-194 induce cell-cycle arrest and apoptosis, and these mimics are potent inhibitors with IC50 values up to several hundred fold less than their native counterparts or Gem alone. Furthermore, we found that Gem-miR-15a and Gem-miR-194 retained miRNA function by downregulating the expression of several key targets including WEE1, CHK1, BMI1, and YAP1 for Gem-miR-15a, and FOXA1 for Gem-miR-194. We also found that our Gem-modified miRNA mimics exhibit an enhanced efficacy compared to Gem in patient-derived PDAC organoids. Furthermore, we observed that Gem-miR-15a significantly inhibits PDAC tumor growth in vivo without observing any noticeable signs of toxicity. Overall, our results demonstrate the therapeutic potential of Gem-modified miRNAs as a treatment strategy for PDAC.
    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2A(PP2A)是一种必需的丝氨酸/苏氨酸蛋白磷酸酶,它的功能障碍与癌症和神经退行性疾病的发生有关。PP2A充当三聚体全酶,其组成受PP2A催化亚基(PP2Ac)的甲酯化(甲基化)调节。蛋白磷酸酶甲基酯酶-1(PME-1)是唯一的PP2Ac甲基酯酶,在各种癌症和神经退行性疾病中观察到较高的PME-1表达。除了作为甲基酯酶,PME-1作为PP2A抑制蛋白,直接与PP2Ac结合并抑制其活性。PME-1的复杂功能通过靶向PME-1/PP2Ac轴来阻碍药物开发。本研究应用NanoBiT系统,基于生物发光的蛋白质相互作用测定,阐明调节未知PME-1/PP2Ac蛋白-蛋白相互作用(PPI)的分子机制。化合物筛选鉴定CHK1抑制剂抑制PME-1/PP2Ac缔合而不影响PP2Ac甲基化水平。CHK1直接磷酸化PP2Ac以促进PME-1缔合。磷酸质谱鉴定了PP2Ac上的多个磷酸位点,包括影响PME-1相互作用的Thr219。产生了抗磷酸-Thr219PP2Ac抗体,并显示CHK1调节细胞中该位点的磷酸化水平。相反,体外磷酸酶测定显示CHK1是PP2A的底物,和PME-1阻碍PP2A介导的CHK1去磷酸化。我们的数据为控制PME-1/PP2AcPPI的分子机制和PP2A之间的三联体关系提供了新的见解。PME-1和CHK1。
    Protein phosphatase 2A (PP2A) is an essential serine/threonine protein phosphatase, and its dysfunction is involved in the onset of cancer and neurodegenerative disorders. PP2A functions as a trimeric holoenzyme whose composition is regulated by the methyl-esterification (methylation) of the PP2A catalytic subunit (PP2Ac). Protein phosphatase methylesterase-1 (PME-1) is the sole PP2Ac methylesterase, and the higher PME-1 expression is observed in various cancer and neurodegenerative diseases. Apart from serving as a methylesterase, PME-1 acts as a PP2A inhibitory protein, binding directly to PP2Ac and suppressing its activity. The intricate function of PME-1 hinders drug development by targeting the PME-1/PP2Ac axis. This study applied the NanoBiT system, a bioluminescence-based protein interaction assay, to elucidate the molecular mechanism that modulates unknown PME-1/PP2Ac protein-protein interaction (PPI). Compound screening identified that the CHK1 inhibitors inhibited PME-1/PP2Ac association without affecting PP2Ac methylation levels. CHK1 directly phosphorylates PP2Ac to promote PME-1 association. Phospho-mass spectrometry identified multiple phospho-sites on PP2Ac, including the Thr219, that affect PME-1 interaction. An anti-phospho-Thr219 PP2Ac antibody was generated and showed that CHK1 regulates the phosphorylation levels of this site in cells. On the contrary, in vitro phosphatase assay showed that CHK1 is the substrate of PP2A, and PME-1 hindered PP2A-mediated dephosphorylation of CHK1. Our data provides novel insights into the molecular mechanisms governing the PME-1/PP2Ac PPI and the triad relationship between PP2A, PME-1, and CHK1.
    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PKD1或PKD2基因中具有种系突变的常染色体显性多囊肾病(ADPKD)中,无数的囊肿是从小管发育而来的,肾功能恶化。二次体细胞突变和肾小管上皮(RTE)细胞死亡是囊肿发生和疾病进展的重要特征。这里,我们使用已建立的RTE细胞系和具有疾病相关PKD1突变的原代ADPKD细胞来研究基因组不稳定性和DNA损伤反应.我们发现ADPKD细胞遭受严重的染色体断裂,非整倍体,对DNA损伤的敏感性提高,和延迟的检查点激活。人肾脏的免疫组织化学分析证实了培养细胞中的观察结果。DNA损伤传感器(ATM/ATR)被激活,但未定位在受损DNA的核位点,也未正确激活下游换能器(CHK1/CHK2)。ADPKD细胞也有转化的能力,当它们达到高饱和密度并在软琼脂中形成菌落时。我们的研究表明,缺陷的DNA损伤修复途径及其引起的体细胞突变从根本上导致了ADPKD的发病机理。获得的突变可以可选地赋予克隆扩增的细胞群体增殖优势或导致细胞凋亡。对ADPKD中异常DNA损伤反应的分子细节的进一步了解正在进行中,并有望用于靶向治疗。
    In autosomal dominant polycystic kidney disease (ADPKD) with germline mutations in a PKD1 or PKD2 gene, innumerable cysts develop from tubules, and renal function deteriorates. Second-hit somatic mutations and renal tubular epithelial (RTE) cell death are crucial features of cyst initiation and disease progression. Here, we use established RTE lines and primary ADPKD cells with disease-associated PKD1 mutations to investigate genomic instability and DNA damage responses. We found that ADPKD cells suffer severe chromosome breakage, aneuploidy, heightened susceptibility to DNA damage, and delayed checkpoint activation. Immunohistochemical analyses of human kidneys corroborated observations in cultured cells. DNA damage sensors (ATM/ATR) were activated but did not localize at nuclear sites of damaged DNA and did not properly activate downstream transducers (CHK1/CHK2). ADPKD cells also had the ability to transform, as they achieved high saturation density and formed colonies in soft agar. Our studies indicate that defective DNA damage repair pathways and the somatic mutagenesis they cause contribute fundamentally to the pathogenesis of ADPKD. Acquired mutations may alternatively confer proliferative advantages to the clonally expanded cell populations or lead to apoptosis. Further understanding of the molecular details of aberrant DNA damage responses in ADPKD is ongoing and holds promise for targeted therapies.
    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
    复制应激(RS)是癌细胞的特征状态,因为它们倾向于将复制的精确性交换为快速增殖和增加的基因组不稳定性。为了克服复制控制不当的后果,恶性细胞经常使其DNA损伤反应(DDR)途径(ATM-CHK2-p53途径)的部分失活,同时依赖于其他有助于保持复制叉稳定性的途径(ATR-CHK1)。这产生了对剩余DDR路径的依赖,DDR抑制剂的复制和合成致死性进一步不稳定,常见的致癌改变,如TP53,RB1,ATM的突变,MYC的扩增,CCNE1等。对RS的反应通常受到细胞周期协调的限制,转录和复制。抑制WEE1和PKMYT1激酶,防止计划外的有丝分裂进入,导致复制不足的网站的脆弱性。最近的证据还表明,抑制细胞周期蛋白依赖性激酶(CDKs),例如CDK4/6、CDK2、CDK8/19和CDK12/13可以通过破坏DNA修复和复制控制来促成RS。这里,我们回顾了RS在癌症中的主要原因以及主要的治疗靶点-ATR,CHK1、PARP及其抑制剂。
    Replication stress (RS) is a characteristic state of cancer cells as they tend to exchange precision of replication for fast proliferation and increased genomic instability. To overcome the consequences of improper replication control, malignant cells frequently inactivate parts of their DNA damage response (DDR) pathways (the ATM-CHK2-p53 pathway), while relying on other pathways which help to maintain replication fork stability (ATR-CHK1). This creates a dependency on the remaining DDR pathways, vulnerability to further destabilization of replication and synthetic lethality of DDR inhibitors with common oncogenic alterations such as mutations of TP53, RB1, ATM, amplifications of MYC, CCNE1 and others. The response to RS is normally limited by coordination of cell cycle, transcription and replication. Inhibition of WEE1 and PKMYT1 kinases, which prevent unscheduled mitosis entry, leads to fragility of under-replicated sites. Recent evidence also shows that inhibition of Cyclin-dependent kinases (CDKs), such as CDK4/6, CDK2, CDK8/19 and CDK12/13 can contribute to RS through disruption of DNA repair and replication control. Here, we review the main causes of RS in cancers as well as main therapeutic targets-ATR, CHK1, PARP and their 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
    多发性骨髓瘤(MM)是一种血液恶性肿瘤,其特征是恶性浆细胞的异常克隆增殖。尽管引入了具有显著改善临床结果的新型药物,大多数患者复发并产生耐药性。MM的特征在于基因组不稳定性和高水平的复制应激。为了应对复制和DNA损伤应激,MM细胞激活各种DNA损伤信号通路。在这项研究中,我们报道,在接受高剂量美法仑化疗或抗CD38免疫治疗的独立MM患者队列中,高CHK1和WEE1表达与不良结局相关.Chk1和Wee1的联合靶向显示对MM细胞的协同毒性,并与更高的DNA双链断裂诱导相关,如随后导致细胞凋亡的γH2AX阳性细胞百分比增加所证明的。Chk1/Wee1抑制剂组合的治疗兴趣在患者的原代MM细胞上得到验证。由于正常骨髓细胞没有受到显著影响,因此毒性是MM细胞的特异性。使用反卷积方法,具有高CHK1表达的MM患者表现出显著较低的NK细胞百分比,而具有高WEE1表达的患者在骨髓中表现出显著较高的调节性T细胞百分比。这些数据强调MM细胞通过Wee1和Chk1上调对复制应激的适应可以降低细胞固有的先天免疫应答的激活。我们的研究表明,在以CHK1和WEE1高表达为特征的高危MM患者中,Chk1和Wee1抑制剂的联合可能是一种有希望的治疗方法。
    Multiple myeloma (MM) is a hematological malignancy characterized by an abnormal clonal proliferation of malignant plasma cells. Despite the introduction of novel agents that have significantly improved clinical outcome, most patients relapse and develop drug resistance. MM is characterized by genomic instability and a high level of replicative stress. In response to replicative and DNA damage stress, MM cells activate various DNA damage signaling pathways. In this study, we reported that high CHK1 and WEE1 expression is associated with poor outcome in independent cohorts of MM patients treated with high dose melphalan chemotherapy or anti-CD38 immunotherapy. Combined targeting of Chk1 and Wee1 demonstrates synergistic toxicities on MM cells and was associated with higher DNA double-strand break induction, as evidenced by an increased percentage of γH2AX positive cells subsequently leading to apoptosis. The therapeutic interest of Chk1/Wee1 inhibitors\' combination was validated on primary MM cells of patients. The toxicity was specific of MM cells since normal bone marrow cells were not significantly affected. Using deconvolution approach, MM patients with high CHK1 expression exhibited a significant lower percentage of NK cells whereas patients with high WEE1 expression displayed a significant higher percentage of regulatory T cells in the bone marrow. These data emphasize that MM cell adaptation to replicative stress through Wee1 and Chk1 upregulation may decrease the activation of the cell-intrinsic innate immune response. Our study suggests that association of Chk1 and Wee1 inhibitors may represent a promising therapeutic approach in high-risk MM patients characterized by high CHK1 and WEE1 expression.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Preprint
    WEE1和CHEK1(CHK1)激酶是G2/M细胞周期检查点和DNA损伤应答途径的关键调节因子。WEE1抑制剂AZD1775和CHK1抑制剂SRA737正在进行各种癌症的临床试验,但没有在前列腺癌中检查过,特别是去势抵抗(CRPC)和神经内分泌前列腺癌(NEPC)。我们的数据表明在CRPC/NEPC细胞系和患者样品中WEE1和CHK1表达升高。AZD1775导致快速和有效的细胞杀伤,在不同的前列腺癌细胞系中具有相当的IC50,而SRA737显示时间依赖性进行性细胞杀伤,IC50差异为10至20倍。值得注意的是,它们的组合以浓度和时间依赖性方式协同降低了所有CRPC细胞系和肿瘤球体的活力。重要的是,在NEPC的转基因小鼠模型中,两种药物单独或联合抑制肿瘤生长,总生存率提高,并降低了远处转移的发生率,SRA737具有显着的单剂抗癌活性。机械上,SRA737通过阻断AZD1775诱导的前列腺癌细胞中CHK1的反馈激活与AZD1775协同作用,导致有丝分裂进入和DNA损伤积累增加。总之,这项临床前研究表明,CHK1抑制剂SRA737单独使用及其与AZD1775的联合使用为CRPC和NEPC提供了潜在的有效治疗方法.
    WEE1 and CHEK1 (CHK1) kinases are critical regulators of the G2/M cell cycle checkpoint and DNA damage response pathways. The WEE1 inhibitor AZD1775 and the CHK1 inhibitor SRA737 are in clinical trials for various cancers, but have not been examined in prostate cancer, particularly castration-resistant (CRPC) and neuroendocrine prostate cancers (NEPC). Our data demonstrated elevated WEE1 and CHK1 expressions in CRPC/NEPC cell lines and patient samples. AZD1775 resulted in rapid and potent cell killing with comparable IC50s across different prostate cancer cell lines, while SRA737 displayed time-dependent progressive cell killing with 10- to 20-fold differences in IC50s. Notably, their combination synergistically reduced the viability of all CRPC cell lines and tumor spheroids in a concentration- and time-dependent manner. Importantly, in a transgenic mouse model of NEPC, both agents alone or in combination suppressed tumor growth, improved overall survival, and reduced the incidence of distant metastases, with SRA737 exhibiting remarkable single agent anticancer activity. Mechanistically, SRA737 synergized with AZD1775 by blocking AZD1775-induced feedback activation of CHK1 in prostate cancer cells, resulting in increased mitotic entry and accumulation of DNA damage. In summary, this preclinical study shows that CHK1 inhibitor SRA737 alone and its combination with AZD1775 offer potential effective treatments for CRPC and NEPC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号