AZD1775

AZD1775
  • 文章类型: Journal Article
    小细胞肺癌(SCLC)患者迫切需要更有效的治疗选择。SCLC细胞中G1检查点的频繁破坏产生对G2/M检查点的依赖性以维持基因组完整性。的确,在临床前模型中,抑制G2/M检查点激酶WEE1显示抑制SCLC生长的前景。然而,毒性和获得性耐药性限制了该策略的临床有效性。这里,在体外和体内使用CRISPR-Cas9基因敲除筛选,我们确定了影响SCLC细胞对WEE1激酶抑制剂AZD1775反应的多种因素,包括GCN2激酶及其信号通路的其他成员.AZD1775处理后GCN2的快速活化触发SCLC细胞中的应激反应。GCN2途径的药理学或遗传学激活增强了AZD1775对癌细胞的杀伤作用。因此,激活GCN2途径代表了增加SCLC中WEE1抑制剂疗效的有希望的策略.
    Patients with small-cell lung cancer (SCLC) are in dire need of more effective therapeutic options. Frequent disruption of the G1 checkpoint in SCLC cells creates a dependency on the G2/M checkpoint to maintain genomic integrity. Indeed, in pre-clinical models, inhibiting the G2/M checkpoint kinase WEE1 shows promise in inhibiting SCLC growth. However, toxicity and acquired resistance limit the clinical effectiveness of this strategy. Here, using CRISPR-Cas9 knockout screens in vitro and in vivo, we identified multiple factors influencing the response of SCLC cells to the WEE1 kinase inhibitor AZD1775, including the GCN2 kinase and other members of its signaling pathway. Rapid activation of GCN2 upon AZD1775 treatment triggers a stress response in SCLC cells. Pharmacological or genetic activation of the GCN2 pathway enhances cancer cell killing by AZD1775. Thus, activation of the GCN2 pathway represents a promising strategy to increase the efficacy of WEE1 inhibitors in SCLC.
    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:利用癌细胞脆弱性的靶向治疗有望改善患者预后并减少化疗的副作用。然而,精准治疗的疗效有限,部分原因是肿瘤细胞异质性。更好地理解药物作用如何与癌细胞状态多样性相关联对于确定可以预防疾病复发的有效组合疗法至关重要。
    结果:这里,我们描述了G2/M检查点抑制在急性淋巴细胞白血病(ALL)中的作用,并证明了WEE1靶向治疗对细胞命运决定调节回路的影响.我们发现KMT2A重排对ALL细胞增殖恢复的抑制作用最高。RS4的单细胞RNA-seq和ATAC-seq;11个带有KMT2A::AFF1的细胞,用WEE1抑制剂AZD1775处理,揭示细胞状态的多样化,部分细胞表现出与细胞凋亡和衰老相关的p53驱动过程的强烈激活,以及核心KMT2A-RUNX1-MYC监管网络的中断。在这种由WEE1抑制诱导的细胞状态多样化中,亚群过渡到药物耐受性细胞状态,其特征是转录因子的激活调节前B细胞命运,脂质代谢,和以可逆方式的前BCR信号传导。BCR信号抑制剂达沙替尼的序贯治疗,伊布替尼,或通过fatostatin或AZD2014干扰代谢通过诱导细胞死亡和抑制干性标志物有效抵消药物耐受性。
    结论:总的来说,我们的研究结果为与细胞周期和细胞命运调控相关的基因调控程序的紧密连接提供了新的见解,和顺序给药WEE1抑制剂与前BCR信号或代谢的低毒性抑制剂的基本原理。
    Targeted therapies exploiting vulnerabilities of cancer cells hold promise for improving patient outcome and reducing side-effects of chemotherapy. However, efficacy of precision therapies is limited in part because of tumor cell heterogeneity. A better mechanistic understanding of how drug effect is linked to cancer cell state diversity is crucial for identifying effective combination therapies that can prevent disease recurrence.
    Here, we characterize the effect of G2/M checkpoint inhibition in acute lymphoblastic leukemia (ALL) and demonstrate that WEE1 targeted therapy impinges on cell fate decision regulatory circuits. We find the highest inhibition of recovery of proliferation in ALL cells with KMT2A-rearrangements. Single-cell RNA-seq and ATAC-seq of RS4;11 cells harboring KMT2A::AFF1, treated with the WEE1 inhibitor AZD1775, reveal diversification of cell states, with a fraction of cells exhibiting strong activation of p53-driven processes linked to apoptosis and senescence, and disruption of a core KMT2A-RUNX1-MYC regulatory network. In this cell state diversification induced by WEE1 inhibition, a subpopulation transitions to a drug tolerant cell state characterized by activation of transcription factors regulating pre-B cell fate, lipid metabolism, and pre-BCR signaling in a reversible manner. Sequential treatment with BCR-signaling inhibitors dasatinib, ibrutinib, or perturbing metabolism by fatostatin or AZD2014 effectively counteracts drug tolerance by inducing cell death and repressing stemness markers.
    Collectively, our findings provide new insights into the tight connectivity of gene regulatory programs associated with cell cycle and cell fate regulation, and a rationale for sequential administration of WEE1 inhibitors with low toxicity inhibitors of pre-BCR signaling or metabolism.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Clinical Trial, Phase I
    目的:Adavosertib可能改变对细胞色素P450(CYP)家族酶底物的暴露。这项研究评估了其对CYP3A(咪达唑仑)探针底物混合物的药代动力学的影响,CYP2C19(奥美拉唑),和CYP1A2(咖啡因)。
    方法:第1期:局部晚期或转移性实体瘤患者接受“鸡尾酒”:咖啡因200毫克,奥美拉唑20毫克,和咪达唑仑2毫克(单剂量);第2期:在7至14天的冲洗后,患者在第1-3天(五剂)每天两次接受adavosertib225mg,在第三天喝鸡尾酒。在单独使用鸡尾酒或与adavosertib联合使用后,对探针底物及其各自的代谢物黄嘌呤进行24小时药代动力学采样,5-羟基奥美拉唑(5-HO),和1'-羟基咪唑安定(1'-HM)。在整个过程中对安全性进行了评估。
    结果:在33名患者中(中位年龄60.0岁,范围41-83)接收鸡尾酒,30人收到adavosertib。Adavosertib联合给药增加了咖啡因,奥美拉唑,咪达唑仑暴露量为49%,80%,和55%(AUC0-12),AUC0-t分别增加61%,98%,和55%。最大血浆药物浓度(Cmax)增加4%,46%,39%。Adavosertib联合给药使5-HO和1'-HM暴露增加了43%和54%(AUC0-12)和49%和58%(AUC0-t),分别;黄嘌呤暴露量不变。Adavosertib联合给药可将对黄嘌呤和5-HO的Cmax降低19%和7%;1'-HM的Cmax增加33%。收到adavosertib后,19例(63%)患者出现治疗相关不良事件(6例[20%]≥3级)。
    结论:Adavosertib(225mgbid)是CYP1A2、CYP2C19和CYP3A的弱抑制剂。
    结果:GOV:NCT03333824。
    Adavosertib may alter exposure to substrates of the cytochrome P450 (CYP) family of enzymes. This study assessed its effect on the pharmacokinetics of a cocktail of probe substrates for CYP3A (midazolam), CYP2C19 (omeprazole), and CYP1A2 (caffeine).
    Period 1: patients with locally advanced or metastatic solid tumors received \'cocktail\': caffeine 200 mg, omeprazole 20 mg, and midazolam 2 mg (single dose); period 2: after 7- to 14-day washout, patients received adavosertib 225 mg twice daily on days 1-3 (five doses), with cocktail on day 3. After cocktail alone or in combination with adavosertib administration, 24-h pharmacokinetic sampling occurred for probe substrates and their respective metabolites paraxanthine, 5-hydroxyomeprazole (5-HO), and 1\'-hydroxymidazolam (1\'-HM). Safety was assessed throughout.
    Of 33 patients (median age 60.0 years, range 41-83) receiving cocktail, 30 received adavosertib. Adavosertib co-administration increased caffeine, omeprazole, and midazolam exposure by 49%, 80%, and 55% (AUC0-12), respectively; AUC0-t increased by 61%, 98%, and 55%. Maximum plasma drug concentration (Cmax) increased by 4%, 46%, and 39%. Adavosertib co-administration increased 5-HO and 1\'-HM exposure by 43% and 54% (AUC0-12) and 49% and 58% (AUC0-t), respectively; paraxanthine exposure was unchanged. Adavosertib co-administration decreased Cmax for paraxanthine and 5-HO by 19% and 7%; Cmax increased by 33% for 1\'-HM. After receiving adavosertib, 19 (63%) patients had treatment-related adverse events (six [20%] grade ≥ 3).
    Adavosertib (225 mg bid) is a weak inhibitor of CYP1A2, CYP2C19, and CYP3A.
    GOV: NCT03333824.
    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
    目的:Adavosertib是一种小分子,Wee1激酶的ATP竞争性抑制剂。分子靶向肿瘤药物有可能增加心血管事件的风险,包括QT间期延长和相关心律失常。这项研究调查了adavosertib对晚期实体瘤患者QTc间期的影响。
    方法:符合条件的患者为≥18岁的晚期实体瘤,没有标准治疗。患者在第1-2天以12小时的间隔每天两次接受adavosertib225mg,在第3天接受一次。患者在给药前进行数字12导联心电图和药代动力学评估,并在给药期间进行时间匹配评估。使用预先指定的线性混合效应模型估计了最大血浆药物浓度(Cmax)与Fridericia的基线调整校正QT间期(QTcF)之间的关系。
    结果:21例患者接受了阿达沃司替。ΔQTcF的浓度-QT建模和对应于在第1天和第3天观察到的Cmax的几何平均值的90%置信区间的上限低于调节关注的阈值(不>10ms)。ΔQTcF(相对于基线)与adavosertib浓度之间没有显着关系(P=0.27)。药代动力学和不良事件(AE)概况与该剂量的先前研究一致。11例(52.4%)患者总共经历了17例治疗相关的不良事件,包括腹泻和恶心(均在6例[28.6%]患者中报告),呕吐(报告有两名[9.5%]病人),贫血,食欲下降,和便秘(均在一名[4.8%]患者中报告)。
    结论:Adavosertib对QTc延长没有临床重要影响。
    结果:GOV:NCT03333824。
    Adavosertib is a small-molecule, ATP-competitive inhibitor of Wee1 kinase. Molecularly targeted oncology agents have the potential to increase the risk of cardiovascular events, including prolongation of QT interval and associated cardiac arrhythmias. This study investigated the effect of adavosertib on the QTc interval in patients with advanced solid tumors.
    Eligible patients were ≥ 18 years of age with advanced solid tumors for which no standard therapy existed. Patients received adavosertib 225 mg twice daily on days 1-2 at 12-h intervals and once on day 3. Patients underwent digital 12-lead electrocardiogram and pharmacokinetic assessments pre-administration and time-matched assessments during the drug administration period. The relationship between maximum plasma drug concentration (Cmax) and baseline-adjusted corrected QT interval by Fridericia (QTcF) was estimated using a prespecified linear mixed-effects model.
    Twenty-one patients received adavosertib. Concentration-QT modeling of ΔQTcF and the upper limit of the 90% confidence interval corresponding to the geometric mean of Cmax observed on days 1 and 3 were below the threshold for regulatory concern (not > 10 ms). No significant relationship between ΔQTcF (vs baseline) and adavosertib concentration was identified (P = 0.27). Pharmacokinetics and the adverse event (AE) profile were consistent with previous studies at this dose. Eleven (52.4%) patients experienced 17 treatment-related AEs in total, including diarrhea and nausea (both reported in six [28.6%] patients), vomiting (reported in two [9.5%] patients), anemia, decreased appetite, and constipation (all reported in one [4.8%] patient).
    Adavosertib does not have a clinically important effect on QTc prolongation.
    GOV: NCT03333824.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Clinical Trial, Phase I
    Adavosertib选择性抑制Wee1,它调节S内和G2/M细胞周期检查点。这项研究调查了阿达沃塞替单药治疗的给药时间表,确定晚期实体瘤患者的最大耐受剂量(MTD)和推荐的II期剂量(RP2D)。患者接受口服adavosertibqd或按5/9时间表投标(治疗5天,9天休息)在14天周期内,或两个5/2时间表之一的qd(每周,或3周中的2周),为期21天。安全,功效,并进行了药代动力学分析。62名患者(女性,64.5%;年龄中位数,61.5年;最常见的原发性肿瘤:肺[24.2%],卵巢[21.0%])接受治疗(qd时间表,n=50;投标时间表,n=12),持续1.8个月(中位数)。达到最大adavosertib浓度的中位时间为2.2-4.1小时;平均半衰期为5-12小时。不良事件(AE)导致剂量减少,17人中有中断和中断(27.4%),25例(40.3%)和4例(6.5%)患者,分别。最常见的≥3级AE为贫血,中性粒细胞减少症(n=9,14.5%)和腹泻(n=8,12.9%)。7例(11.3%)患者经历了10例治疗相关的严重不良事件(肺炎n=2[3.2%],脱水n=2[3.2%],贫血n=1[1.6%],发热性中性粒细胞减少症n=1[1.6%],和血小板减少症n=1[1.6%])。总体客观缓解率为3.4%(2/58);疾病控制率为48.4%(30/62);中位无进展生存期为2.7个月。MTD为125mg(bid5/9)和300mg(qd5/9和5/2,共3周2周);RP2D为300mg(qd5/2,共3周2周)。安全性是可控的,可接受,通常与已知的安全特征一致。
    Adavosertib selectively inhibits Wee1, which regulates intra-S and G2/M cell-cycle checkpoints. This study investigated dosing schedules for adavosertib monotherapy, determining the maximum tolerated dose (MTD) and recommended Phase II dose (RP2D) in patients with advanced solid tumors.Patients received oral adavosertib qd or bid on a 5/9 schedule (5 days on treatment, 9 days off) in 14-day cycles, or qd on one of two 5/2 schedules (weekly, or for 2 of 3 weeks) in 21-day cycles. Safety, efficacy, and pharmacokinetic analyses were performed.Sixty-two patients (female, 64.5%; median age, 61.5 years; most common primary tumors: lung [24.2%], ovary [21.0%]) received treatment (qd schedules, n = 50; bid schedules, n = 12) for 1.8 months (median). Median time to maximum adavosertib concentration was 2.2-4.1 h; mean half-life was 5-12 h. Adverse events (AEs) caused dose reductions, interruptions and discontinuations in 17 (27.4%), 25 (40.3%) and 4 (6.5%) patients, respectively. Most common grade ≥ 3 AEs were anemia, neutropenia (each n = 9, 14.5%) and diarrhea (n = 8, 12.9%). Seven (11.3%) patients experienced 10 treatment-related serious AEs (pneumonia n = 2 [3.2%], dehydration n = 2 [3.2%], anemia n = 1 [1.6%], febrile neutropenia n = 1 [1.6%], and thrombocytopenia n = 1 [1.6%]). Overall objective response rate was 3.4% (2/58); disease control rate was 48.4% (30/62); median progression-free survival was 2.7 months.MTDs were 125 mg (bid 5/9) and 300 mg (qd 5/9 and 5/2 for 2 of 3 weeks); RP2D was 300 mg (qd 5/2 for 2 of 3 weeks). The safety profile was manageable, acceptable, and generally concordant with the known safety profile.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Clinical Trial, Phase II
    背景:adavosertib(AZD1775)对WEE1激酶的抑制增强了来自基因组不稳定性或化疗的复制应激。这项研究报告了联合伊立替康和阿达沃塞替尼的ADVL1312试验的小儿实体瘤2期结果。
    方法:小儿复发性神经母细胞瘤(B部分),髓母细胞瘤/中枢神经系统胚胎性肿瘤(C部分),或横纹肌肉瘤(D部分)每21天口服伊立替康和adavosertib治疗5天。如果在部分B和D中存在20个反应中的至少3个或在部分C中存在19个反应中的6个,则认为该组合是有效的。分析肿瘤组织的端粒和ATRX的选择性延长。提供了患者先前的肿瘤基因组分析。
    结果:20例神经母细胞瘤患者(B部分)的既往治疗方案中位数为3种,95%的患者既往有伊立替康病史。有三个客观反应(9、11和18个周期)符合方案定义的疗效终点。有客观反应的三名患者中有两名患有端粒交替延长的肿瘤。1例松果体母细胞瘤患者部分缓解(11个周期),但C部分和D部分不符合方案定义的疗效终点.在推荐的2期剂量下,该组合的耐受性良好,并且在第1周期或更长时间的ADVL1312的任何部分都没有剂量限制性毒性。
    结论:这是阿达沃司替布在儿科中的第一个2期临床试验,也是第一个使用伊立替康的临床试验。该组合可能具有足够的活性,可以考虑在神经母细胞瘤中进一步研究adavosertib。
    Inhibition of the WEE1 kinase by adavosertib (AZD1775) potentiates replicative stress from genomic instability or chemotherapy. This study reports the pediatric solid tumor phase 2 results of the ADVL1312 trial combining irinotecan and adavosertib.
    Pediatric patients with recurrent neuroblastoma (part B), medulloblastoma/central nervous system embryonal tumors (part C), or rhabdomyosarcoma (part D) were treated with irinotecan and adavosertib orally for 5 days every 21 days. The combination was considered effective if there were at least three of 20 responses in parts B and D or six of 19 responses in part C. Tumor tissue was analyzed for alternative lengthening of telomeres and ATRX. Patient\'s prior tumor genomic analyses were provided.
    The 20 patients with neuroblastoma (part B) had a median of three prior regimens and 95% had a history of prior irinotecan. There were three objective responses (9, 11, and 18 cycles) meeting the protocol defined efficacy end point. Two of the three patients with objective responses had tumors with alternative lengthening of telomeres. One patient with pineoblastoma had a partial response (11 cycles), but parts C and D did not meet the protocol defined efficacy end point. The combination was well tolerated and there were no dose limiting toxicities at cycle 1 or beyond in any parts of ADVL1312 at the recommended phase 2 dose.
    This is first phase 2 clinical trial of adavosertib in pediatrics and the first with irinotecan. The combination may be of sufficient activity to consider further study of adavosertib in neuroblastoma.
    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
    尤因肉瘤是一种侵袭性儿童癌症,其治疗选择仍然有限且有毒。迫切需要鉴定新的治疗策略。我们小组最近表明,尤因细胞依赖于S期激酶CDC7(DDK)来维持复制速率和细胞活力,并且DDK抑制导致CDK1磷酸化增加和有丝分裂进入的显着延迟。这里,我们扩展了先前的发现,并表明DDK抑制剂诱导的有丝分裂进入延迟取决于WEE1激酶。具体来说,由于存在复制不足的DNA,WEE1磷酸化CDK1并防止DDK抑制后有丝分裂进入,潜在限制DDK抑制的细胞毒性作用。为了克服这一点,我们将DDK的抑制与WEE1的抑制相结合,发现这导致过早有丝分裂进入的水平升高,有丝分裂灾难,和凋亡。重要的是,我们发现DDK和WEE1抑制剂在降低尤文氏肉瘤细胞的细胞活力方面显示出协同关系。有趣的是,这种组合的细胞毒性可以通过抑制CDK1或微管聚合来抑制,表明有丝分裂的进展需要引起细胞毒性作用。这是第一项显示利用DDK和WEE1的联合抑制来治疗癌症的潜力的研究。我们相信这将为尤文肉瘤以及其他对DDK抑制剂敏感的肿瘤类型的治疗提供潜在的治疗策略。
    Ewing sarcoma is an aggressive childhood cancer for which treatment options remain limited and toxic. There is an urgent need for the identification of novel therapeutic strategies. Our group has recently shown that Ewing cells rely on the S-phase kinase CDC7 (DDK) to maintain replication rates and cell viability and that DDK inhibition causes an increase in the phosphorylation of CDK1 and a significant delay in mitotic entry. Here, we expand on our previous findings and show that DDK inhibitor-induced mitotic entry delay is dependent upon WEE1 kinase. Specifically, WEE1 phosphorylates CDK1 and prevents mitotic entry upon DDK inhibition due to the presence of under-replicated DNA, potentially limiting the cytotoxic effects of DDK inhibition. To overcome this, we combined the inhibition of DDK with the inhibition of WEE1 and found that this results in elevated levels of premature mitotic entry, mitotic catastrophe, and apoptosis. Importantly, we have found that DDK and WEE1 inhibitors display a synergistic relationship with regards to reducing cell viability of Ewing sarcoma cells. Interestingly, the cytotoxic nature of this combination can be suppressed by the inhibition of CDK1 or microtubule polymerization, indicating that mitotic progression is required to elicit the cytotoxic effects. This is the first study to display the potential of utilizing the combined inhibition of DDK and WEE1 for the treatment of cancer. We believe this will offer a potential therapeutic strategy for the treatment of Ewing sarcoma as well as other tumor types that display sensitivity to DDK 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
    靶向G2/M检查点介体WEE1已被探索为卵巢癌的新治疗策略,但其功效和抗性的潜在机制仍有待理解。这里,已证明,在TP53突变(mtTP53)卵巢癌模型中,WEE1抑制剂AZD1775诱导内质网应激并激活未折叠蛋白反应(UPR)的蛋白激酶RNA样ER激酶(PERK)和肌醇必需的酶1α(IRE1α)分支。这通过NF-κB介导的衰老相关分泌表型来促进。经过AZD1775处理,激活的PERK通过C/EBP同源蛋白(CHOP)促进凋亡信号,而IRE1α诱导的XBP1剪接(XBP1s)通过抑制细胞凋亡维持细胞存活。这导致在卵巢癌的多种细胞系和临床前模型中组合AZD1775和IRE1α抑制剂MKC8866的令人鼓舞的协同抗肿瘤作用。一起来看,这些数据揭示了在响应AZD1775的mtTP53卵巢癌模型中UPR信号传导网络的重要双重作用,并提示IRE1α-XBP1s通路的抑制可能增强AZD1775在临床中的疗效.
    Targeting the G2/M checkpoint mediator WEE1 has been explored as a novel treatment strategy in ovarian cancer, but mechanisms underlying its efficacy and resistance remains to be understood. Here, it is demonstrated that the WEE1 inhibitor AZD1775 induces endoplasmic reticulum stress and activates the protein kinase RNA-like ER kinase (PERK) and inositol-required enzyme 1α (IRE1α) branches of the unfolded protein response (UPR) in TP53 mutant (mtTP53) ovarian cancer models. This is facilitated through NF-κB mediated senescence-associated secretory phenotype. Upon AZD1775 treatment, activated PERK promotes apoptotic signaling via C/EBP-homologous protein (CHOP), while IRE1α-induced splicing of XBP1 (XBP1s) maintains cell survival by repressing apoptosis. This leads to an encouraging synergistic antitumor effect of combining AZD1775 and an IRE1α inhibitor MKC8866 in multiple cell lines and preclinical models of ovarian cancers. Taken together, the data reveal an important dual role of the UPR signaling network in mtTP53 ovarian cancer models in response to AZD1775 and suggest that inhibition of the IRE1α-XBP1s pathway may enhance the efficacy of AZD1775 in the clinics.
    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
    我们研究了Wee1激酶在多种顺铂耐药的头颈部鳞状细胞癌(HNSCC)细胞系中的作用,并确定了Wee1抑制剂的疗效。AZD1775,或与顺铂联合使用,关于顺铂耐药的HNSCC抑制。
    通过蛋白质印迹分析评估细胞的磷酸化和总蛋白水平。通过MTS分析和流式细胞术检查细胞活力和凋亡,分别。
    五种顺铂耐药的HNSCC细胞类型中的Wee1激酶蛋白表达水平高于其亲本顺铂敏感伴侣中的表达水平。重要的是,Wee1敲低抑制细胞增殖并使细胞对顺铂处理再敏感。有趣的是,先前的研究还表明,Wee1抑制剂AZD1775与顺铂协同抑制顺铂敏感的HNSCC的细胞增殖。我们发现AZD1775抑制顺铂敏感和耐药HNSCC,IC50值相似,这表明AZD1775可以克服顺铂耐药的HNSCC。机械上,AZD1775和顺铂协同诱导DNA损伤和凋亡。
    Wee1抑制剂,AZD1775和顺铂协同抑制HNSCC的增殖和存活。
    We investigated the role of Wee1 kinase in cisplatin-resistant head and neck squamous cell carcinoma (HNSCC) in multiple cisplatin-resistant HNSCC cell lines and determined the efficacy of either Wee1 inhibitor, AZD1775 alone, or in combination with cisplatin, on cisplatin-resistant HNSCC inhibition.
    Phosphorylation and total protein levels of cells were assessed by Western blot analysis. Cell viability and apoptosis were examined by MTS assay and flow cytometry, respectively.
    Wee1 kinase protein expression levels in five cisplatin-resistant HNSCC cell types were higher than those in their parental cisplatin-sensitive partners. Importantly, Wee1 knockdown inhibited cell proliferation and re-sensitized cells to cisplatin treatment. Interestingly, previous studies have also shown that Wee1 inhibitor AZD1775 synergizes with cisplatin to suppress cell proliferation of cisplatin-sensitive HNSCC. We found that AZD1775 inhibited both cisplatin-sensitive and resistant HNSCC with similar IC50 values, which suggested that AZD1775 could overcome cisplatin resistance in cisplatin-resistant HNSCC. Mechanistically, AZD1775 and cisplatin cooperatively induced DNA damage and apoptosis.
    Wee1 inhibitor, AZD1775, and cisplatin coordinately suppressed proliferation and survival of HNSCC.
    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
    卵巢癌是最致命的妇科恶性肿瘤。复发和化疗耐药是导致预后不良的严峻挑战。HJURP是CENP-A的分子伴侣,这与多种肿瘤的侵袭性进展有关。然而,HJURP在卵巢癌中的功能尚未阐明。在我们的研究中,我们发现HJURP在卵巢癌中过度表达,HJURP的高表达与不良预后相关.HJURP敲低可以抑制增殖,转移并引诱G0/G1期卵巢癌细胞停滞。此外,下一代测序(NGS)揭示了WEE1通过沉默HJURP下调。进一步的机理研究表明,HJURP通过MYC调节WEE1,荧光素酶分析表明MYC是WEE1的转录因子。此外,我们研究了沉默HJURP通过MYC/WEE1轴增加卵巢癌细胞对顺铂的敏感性,HJURP参与顺铂损伤的DNA修复。更有趣的是,沉默HJURP可增强卵巢癌细胞对AZD1775的敏感性,提高顺铂联合AZD1775联合治疗的协同作用。总的来说,我们的数据显示,HJURP促进卵巢癌的肿瘤进展和化疗耐药,HJURP有可能成为卵巢癌顺铂和AZD1775联合治疗的新治疗靶点。
    Ovarian cancer is the most lethal gynecological malignancy. Recurrence and chemoresistance are tough challenges leading to poor prognosis. HJURP is a molecular chaperone of CENP-A, which is associated with aggressive progression in multiple tumors. However, the function of HJURP in ovarian cancer has not been elucidated. In our study, we found HJURP was over-expressed in ovarian cancer and high expression of HJURP was correlated to unfavorable prognosis. HJURP knockdown could inhibit proliferation, metastasis and induce G0/G1 stagnation of ovarian cancer cells. Besides, next-generation sequencing (NGS) unveiled that WEE1 was down-regulated by silencing HJURP. Further mechanistic research revealed that HJURP regulated WEE1 through MYC, and luciferase assay indicated that MYC was a transcription factor of WEE1. Additionally, we investigated that silencing HJURP increased sensitivity of ovarian cancer cells to cisplatin via MYC/WEE1 axis, and HJURP participated in DNA repair of cisplatin-induced damage. More interestingly, silencing HJURP could enhance sensitivity of ovarian cancer cells to AZD1775 and improve the synergistic effect of cisplatin plus AZD1775 combined therapy. Collectively, our data displays that HJURP promotes tumor progression and chemoresistance of ovarian cancer, and HJURP has potential to be a novel therapeutic target in the combined treatment with cisplatin and AZD1775 in ovarian cancer.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号