Drug Resistance, Neoplasm

抗药性, 肿瘤
  • 文章类型: Journal Article
    西妥昔单抗耐药一直是头颈部鳞状细胞癌(HNSCC)患者接受靶向治疗的主要挑战。然而,导致西妥昔单抗耐药的机制,特别是microRNA(miRNA)调控,尚不清楚。越来越多的证据表明,miRNA可能充当“核激活miRNA”,用于靶向与靶基因相关的启动子区域或增强子。这项研究阐明了HNSCC中西妥昔单抗抗性的新机制,涉及通过miR-451a对KDM7A转录的核激活。在这里,小RNA测序,定量实时聚合酶链反应(qRT-PCR)和荧光原位杂交(FISH)的结果提供了有说服力的证据,表明在西妥昔单抗治疗后miR-451a核富集.通过RNA纯化进行染色质分离,微阵列分析,和生物信息学分析显示,miR-451a与KDM7A中的增强子区相互作用,激活其表达并进一步促进西妥昔单抗抵抗。还已经证明,核miR-451a对KDM7A的激活是由西妥昔单抗治疗诱导的,并且是AGO2依赖性的。对87个HNSCC样品的Logistic回归分析表明miR-451a和KDM7A在西妥昔单抗抗性发展中的重要性。这些发现支持miR-451a和KDM7A作为西妥昔单抗抗性的有价值的生物标志物的潜力,并强调核激活miRNA的功能。
    Cetuximab resistance has been a major challenge for head and neck squamous cell carcinoma (HNSCC) patients receiving targeted therapy. However, the mechanism that causes cetuximab resistance, especially microRNA (miRNA) regulation, remains unclear. Growing evidence suggests that miRNAs may act as \"nuclear activating miRNAs\" for targeting promoter regions or enhancers related to target genes. This study elucidates a novel mechanism underlying cetuximab resistance in HNSCC involving the nuclear activation of KDM7A transcription via miR-451a. Herein, small RNA sequencing, quantitative real-time polymerase chain reaction (qRT‒PCR) and fluorescence in situ hybridization (FISH) results provided compelling evidence of miR-451a nuclear enrichment in response to cetuximab treatment. Chromatin isolation via RNA purification, microarray analysis, and bioinformatic analysis revealed that miR-451a interacts with an enhancer region in KDM7A, activating its expression and further facilitating cetuximab resistance. It has also been demonstrated that the activation of KDM7A by nuclear miR-451a is induced by cetuximab treatment and is AGO2 dependent. Logistic regression analyses of 87 HNSCC samples indicated the significance of miR-451a and KDM7A in the development of cetuximab resistance. These discoveries support the potential of miR-451a and KDM7A as valuable biomarkers for cetuximab resistance and emphasize the function of nuclear-activating miRNAs.
    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
    Ferroptosis,一种受调节的细胞死亡形式,与铁依赖性脂质过氧化密切相关。最近的证据强烈支持铁凋亡的诱导作为治疗对常规疗法有抗性的癌症的有希望的策略。铁凋亡调节的关键参与者是铁凋亡抑制蛋白1(FSP1),其通过促进辅酶Q10的抗氧化剂形式的产生来促进癌细胞抗性。值得注意的是,FSP1独立于谷胱甘肽(GSH)和谷胱甘肽过氧化物酶-4途径赋予铁死亡抗性。因此,靶向FSP1以削弱其对铁凋亡的抑制作用可能是治疗难治性癌症的可行策略。这篇综述旨在阐明铁死亡的分子机制,FSP1抑制铁凋亡的具体途径以及FSP1抑制剂对癌细胞的影响。
    Ferroptosis, a regulated form of cell death, is intricately linked to iron‑dependent lipid peroxidation. Recent evidence strongly supports the induction of ferroptosis as a promising strategy for treating cancers resistant to conventional therapies. A key player in ferroptosis regulation is ferroptosis suppressor protein 1 (FSP1), which promotes cancer cell resistance by promoting the production of the antioxidant form of coenzyme Q10. Of note, FSP1 confers resistance to ferroptosis independently of the glutathione (GSH) and glutathione peroxidase‑4 pathway. Therefore, targeting FSP1 to weaken its inhibition of ferroptosis may be a viable strategy for treating refractory cancer. This review aims to clarify the molecular mechanisms underlying ferroptosis, the specific pathway by which FSP1 suppresses ferroptosis and the effect of FSP1 inhibitors on cancer cells.
    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:L-茶氨酸,一种来自绿茶的非蛋白质氨基酸,被认为是抗癌候选人。然而,它在肿瘤化疗耐药发展中的作用尚不清楚,其分子机制亟待探索。
    方法:通过细胞计数试剂盒-8(CCK-8)试验验证L-茶氨酸对肺癌化疗耐药的影响,transwell分析,和体外肿瘤球体形成测定;通过聚合酶链反应(PCR)和蛋白质印迹法检测蛋白质的表达。使用RNA测序(RNA-seq)和生物信息学分析来鉴定L-茶氨酸诱导的差异表达基因。通过使用慢病毒介导的转染系统构建BMAL1敲低和过表达。
    结果:L-茶氨酸提高了对顺式二氨基二氯铂(DDP)的化学抗性,并抑制了DDP抗性肺癌细胞的干性,但不抑制非抗性肺癌细胞。RNA-seq分析的结果表明,STAT3/NOTCH1途径是参与L-茶氨酸改善DDP耐药肺癌化疗耐药的潜在显性信号。机械上,L-茶氨酸通过调控STAT3/NOTCH1/BMAL1信号传导诱导的干性标记物的表达以及抑制耐药相关基因的表达水平,阻碍DDP耐药肺癌细胞的迁移和干性激活。此外,L-茶氨酸和Stat3阻断联合协同改善DDP耐药肺癌的化疗耐药.
    结论:L-茶氨酸通过调节STAT3/NOTCH1/BMAL1信号通路改善化疗耐药,减少干性,抑制DDP耐药肺癌细胞的迁移。这一发现可能为克服癌症化学耐药性的治疗选择提供一些证据,包括肺癌.
    BACKGROUND: L-Theanine, a nonproteinogenic amino acid derived from green tea, is being recognized as an anti-cancer candidate. However, it\'s roles in the development of cancer chemoresistance is still unknown and the molecular mechanism is urgently to be explored.
    METHODS: The effects of L-Theanine on lung cancer chemoresistance were validated by Cell Counting Kit-8 (CCK-8) assay, transwell assay, and in vitro tumor spheroid formation assay; the expression of proteins was detected by using polymerase chain reaction (PCR) and western blotting. RNA-sequencing (RNA-seq) and bioinformatics analysis were used to identify differentially expressed genes induced by L-Theanine. BMAL1 knockdown and overexpression were constructed by using a lentivirus-mediated transfection system.
    RESULTS: L-Theanine improved the chemoresistance to cis-diamminedichloroplatinum (DDP) and inhibited stemness of DDP-resistant lung cancer cells but not non-resistant lung cancer cells. The results from RNA-seq analysis showed that STAT3/NOTCH1 pathway was a potential dominant signaling involved in L-Theanine improving the chemoresistance in DDP-resistant lung cancer. Mechanistically, L-Theanine impeded migration and stemness activation of DDP-resistant lung cancer cells via regulating the expression of STAT3/NOTCH1/BMAL1 signaling-induced stemness markers as well as inhibiting the expression levels of drug resistance-related genes. In addition, a combination of L-Theanine and Stat3 blockade synergistically improved the chemoresistance in DDP-resistant lung cancer.
    CONCLUSIONS: L-Theanine improves the chemoresistance by regulating STAT3/NOTCH1/BMAL1 signaling, reducing stemness, and inhibiting the migration of DDP-resistant lung cancer cells. The finding might provide some evidence for therapeutic options in overcoming the chemoresistance in cancers, including lung cancer.
    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:肺癌是癌症相关死亡的主要原因,是所有恶性肿瘤中发病率和死亡率最高的肿瘤之一。据报道,DEPDC1B的表达失调发生在各种肿瘤类型中。然而,这种改变在肺腺癌(LUAD)中的功能意义和潜在的分子机制尚不清楚.在这项研究中,我们研究了DEPDC1B在LUAD中的作用和临床意义。
    方法:在几个公开可用的数据集中系统评估了DEPDC1B在LUAD中的表达及其与预后的关系。DEPDC1B敲低对LUAD细胞增殖和运动的影响使用JULI阶段实时细胞历史记录,同时通过流式细胞术研究敲低对细胞周期的影响。此外,进行RNA测序(RNA-Seq)分析以鉴定由DEPDC1B调节的下游靶基因和途径。DEPDC1B的表达与免疫细胞浸润的相关性,免疫疗法抗性,还检查了化学抗性。此外,采用分子生物学方法探讨B-Myb对DEPDC1B表达的调控机制。
    结果:发现DEPDC1B在LUAD患者中上调,这与不良临床结局相关。敲除DEPDC1B抑制细胞生长,迁移和运动性,以及细胞周期进程。敲除还导致几个下游基因的下调,包括NID1、FN1和EGFR,以及多个关键途径的失活,如ERK和PI3K-AKT途径。对LUAD中肿瘤免疫环境的分析表明,高DEPDC1B表达与大量活化的CD4记忆T细胞有关,M0巨噬细胞,M1巨噬细胞,和CD8+T细胞。此外,这些肿瘤对免疫疗法的反应较差.化疗药物敏感性分析显示,DEPDC1B高表达的LUADs对长春瑞滨等一线化疗药物反应更敏感,顺铂,和依托泊苷。此外,机制研究表明,DEPDC1B是B-Myb的直接靶基因,并且其敲除减弱了B-Myb的增殖和运动作用。
    结论:总之,我们的研究结果表明,DEPDC1B是LUAD恶性进展过程中的关键调节因子.因此,DEPDC1B可能是LUAD诊断和治疗中一个有前途的预后标志物和治疗靶标。
    BACKGROUND: Lung cancer is the primary cause of cancer-related deaths, with one of the highest incidence and mortality rates of all malignant tumors. Dysregulated expression of DEPDC1B has been reported to occur in various tumor types. However, the functional implications of this alteration in lung adenocarcinoma (LUAD) and the underlying molecular mechanism remains unclear. In this study, we investigated the role and clinical significance of DEPDC1B in LUAD.
    METHODS: The expression of DEPDC1B in LUAD and its relationship with prognosis were systematically evaluated in several publically available datasets. The effects of DEPDC1B knockdown on the proliferation and motility of LUAD cells were assessed using the JULI Stage Real-time Cell History Recorder, while the effect of knockdown on the cell cycle was studied by flow cytometry. Furthermore, RNA-Sequencing (RNA-Seq) analysis was conducted to identify the downstream target genes and pathways regulated by DEPDC1B. Correlations between the expression of DEPDC1B and immune cell infiltration, immunotherapy resistance, and chemoresistance were also examined. Additionally, molecular biological methods were used to explore the regulatory mechanism of B-Myb on DEPDC1B expression.
    RESULTS: DEPDC1B was found to be upregulated in LUAD patients and this was associated with poor clinical outcomes. Knockdown of DEPDC1B inhibited cell growth, migration and motility, as well as cell cycle progression. Knockdown also resulted in the down-regulation of several downstream genes, including NID1, FN1, and EGFR, as well as the inactivation of multiple critical pathways, such as the ERK and PI3K-AKT pathways. Analysis of the tumor immuno-environment in LUAD revealed that high DEPDC1B expression was associated with an abundance of activated CD4+ memory T cells, M0 macrophages, M1 macrophages, and CD8+ T cells. Moreover, these tumors responded poorly to immunotherapy. Analysis of chemo-drug sensitivity showed that LUADs with high DEPDC1B expression were more responsive to frontline chemotherapeutic drugs such as Vinorelbine, Cisplatin, and Etoposide. Additionally, mechanistic investigations revealed that DEPDC1B is a direct target gene of B-Myb, and that its knockdown attenuated the proliferation and motility effects of B-Myb.
    CONCLUSIONS: In summary, our findings indicate that DEPDC1B is a critical regulator during the malignant progression of LUAD. DEPDC1B could therefore be a promising prognostic marker and therapeutic target in LUAD diagnosis and 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
    癌症临床化疗期间的多药耐药性(MDR)被认为是治疗效果的主要障碍。三磷酸腺苷结合盒(ABC)转运蛋白参与MDR机制显着降低了化学疗法的功效。这项研究调查了莫林的潜力,一种膳食生物类黄酮,克服KBChR-8-5MDR细胞中的秋水仙碱抗性。通过钙黄绿素-AM药物外排测定来测量桑林的P-gp抑制活性。蛋白质印迹分析用于评估在morin处理后的P-gp信使RNA和蛋白质表达。流式细胞术分析和吖啶橙/溴化乙锭荧光染色用于研究用桑色素和紫杉醇联合处理后对细胞凋亡和细胞周期停滞的诱导。此外,进行聚合酶链反应(PCR)阵列分析以研究与MDR相关的基因表达谱,用莫林治疗期间的细胞凋亡和细胞周期停滞,紫杉醇或其组合。Morin表现出与人P-gp的强结合相互作用。药物外排试验证实了这一点,其显示P-gp外排功能随着sorin浓度的增加而降低。此外,莫林和紫杉醇的组合增强了细胞凋亡的诱导和G2/M期细胞周期停滞。Morin处理显著下调MDR细胞ABCB1基因表达和P-gp膜表达。此外,PCR阵列基因表达分析表明,用桑色素和紫杉醇的组合处理上调了促凋亡和细胞周期停滞基因,同时下调了ABCB1基因和抗凋亡基因。因此,莫林可有效逆转KBChR-8-5耐药癌细胞中的紫杉醇耐药性,并得出结论,莫林可使KBChR8-5耐药癌细胞中的紫杉醇耐药性重新敏感。
    Multidrug resistance (MDR) during clinical chemotherapy for cancer has been considered a major obstacle to treatment efficacy. The involvement of adenosine triphosphate-binding cassette (ABC) transporters in the MDR mechanism significantly reduces the efficacy of chemotherapeutics. This study investigates the potential of morin, a dietary bioflavonoid, to overcome colchicine resistance in KBChR-8-5 MDR cells. The P-gp inhibitory activity by morin was measured by calcein-AM drug efflux assay. Western blot analysis was employed to evaluate P-gp messenger RNA and protein expressions following morin treatment. Flow cytometry analysis and acridine orange/ethidium bromide fluorescence staining were utilised to investigate the induction of apoptosis and cell cycle arrest upon treatment with morin and paclitaxel in combination. Additionally, polymerase chain reaction (PCR) array analysis was conducted to study the gene expression profiles related to MDR, apoptosis and cell cycle arrest during treatment with morin, paclitaxel or their combination. Morin exhibited a strong binding interaction with human P-gp. This was corroborated by drug efflux assays, which showed a reduction in P-gp efflux function with increasing morin concentration. Furthermore, morin and paclitaxel combination potentiated the induction of apoptosis and G2/M phase cell cycle arrest. Morin treatment significantly downregulated the gene expression of ABCB1 and P-gp membrane expressions in MDR cells. Additionally, PCR array gene expression analysis revealed that the combination treatment with morin and paclitaxel upregulated proapoptotic and cell cycle arrest genes while downregulating ABCB1 gene and antiapoptotic genes. Thus, morin effectively reversed paclitaxel resistance in KBChR-8-5 drug-resistant cancer cells and concluded that morin resensitized the paclitaxel resistance in KBChR8-5 drug-resistant cancer cells.
    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
    在非小细胞肺癌(NSCLC)中,药物耐受性已成为驱动靶向治疗(TT)抵抗的主要非遗传适应性过程之一。然而,控制这种适应性反应的分子事件的动力学和顺序仍然知之甚少。这里,我们将细胞周期动力学的实时监测和单细胞RNA测序结合在一组广泛的致癌成瘾中,如EGFR-,ALK-,BRAF和KRAS突变型非小细胞肺癌,用相应的TT治疗。我们确定了药物适应的共同路径,它总是涉及肺泡1型(AT1)分化和Rho相关蛋白激酶(ROCK)介导的细胞骨架重塑。我们还分离并描述了罕见的早期逃生者,它们代表了在治疗的最初几个小时内从AT1样群体中出现的最早的抗性起始细胞。表型药物筛选确定法尼基转移酶抑制剂(FTI)如替比法尼是在几种致癌成瘾模型中体外和体内预防TT复发的最有效药物。法尼基转移酶的遗传耗竭证实了这一点。这些发现为结合TT和FTI的治疗方法的发展铺平了道路,以有效预防癌基因成瘾的NSCLC患者的肿瘤复发。
    Drug-tolerance has emerged as one of the major non-genetic adaptive processes driving resistance to targeted therapy (TT) in non-small cell lung cancer (NSCLC). However, the kinetics and sequence of molecular events governing this adaptive response remain poorly understood. Here, we combine real-time monitoring of the cell-cycle dynamics and single-cell RNA sequencing in a broad panel of oncogenic addiction such as EGFR-, ALK-, BRAF- and KRAS-mutant NSCLC, treated with their corresponding TT. We identify a common path of drug adaptation, which invariably involves alveolar type 1 (AT1) differentiation and Rho-associated protein kinase (ROCK)-mediated cytoskeletal remodeling. We also isolate and characterize a rare population of early escapers, which represent the earliest resistance-initiating cells that emerge in the first hours of treatment from the AT1-like population. A phenotypic drug screen identify farnesyltransferase inhibitors (FTI) such as tipifarnib as the most effective drugs in preventing relapse to TT in vitro and in vivo in several models of oncogenic addiction, which is confirmed by genetic depletion of the farnesyltransferase. These findings pave the way for the development of treatments combining TT and FTI to effectively prevent tumor relapse in oncogene-addicted NSCLC patients.
    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
    据报道,表皮生长因子受体(EGFR)在大多数食管鳞状细胞癌(ESCC)患者中过度表达,但是抗EGFR治疗的生存获益有限.我们的临床前数据显示,阿法替尼在EGFR过表达的ESCC中具有良好的抗肿瘤活性。这个概念证明,II期试验评估了阿法替尼在EGFR过表达的转移性ESCC患者(n=41)中的疗效和安全性(NCT03940976).这项研究达到了主要终点,38例疗效可评估患者的客观缓解率(ORR)为39%,中位总生存期为7.8个月,具有可控的毒性。对治疗前肿瘤的转录组分析显示,神经营养受体酪氨酸激酶2(NTRK2)与阿法替尼敏感性呈负相关,并可能作为预测生物标志物。与EGFR表达无关。值得注意的是,敲低或抑制NTRK2致敏ESCC细胞对阿法替尼治疗。我们的研究提供了有关阿法替尼耐药的分子因素的新发现,并表明阿法替尼有可能成为转移性ESCC患者的重要治疗方法。
    Epidermal growth factor receptor (EGFR) is reportedly overexpressed in most esophageal squamous cell carcinoma (ESCC) patients, but anti-EGFR treatments offer limited survival benefits. Our preclinical data showed the promising antitumor activity of afatinib in EGFR-overexpressing ESCC. This proof-of-concept, phase II trial assessed the efficacy and safety of afatinib in pretreated metastatic ESCC patients (n = 41) with EGFR overexpression (NCT03940976). The study met its primary endpoint, with a confirmed objective response rate (ORR) of 39% in 38 efficacy-evaluable patients and a median overall survival of 7.8 months, with a manageable toxicity profile. Transcriptome analysis of pretreatment tumors revealed that neurotrophic receptor tyrosine kinase 2 (NTRK2) was negatively associated with afatinib sensitivity and might serve as a predictive biomarker, irrespective of EGFR expression. Notably, knocking down or inhibiting NTRK2 sensitized ESCC cells to afatinib treatment. Our study provides novel findings on the molecular factors underlying afatinib resistance and indicates that afatinib has the potential to become an important treatment for metastatic ESCC patients.
    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:治疗方案有限,铂耐药复发转移性(R/M)头颈部鳞状细胞癌(HNSCC)患者预后较差。这项研究评估了紫杉醇和异环磷酰胺(TI)方案在以铂类为基础的治疗后疾病进展的R/MHNSCC患者中的疗效和安全性。
    方法:在这项回顾性研究中,我们纳入了53例R/MHNSCC患者,他们接受了至少一个基于TI的治疗周期,铂金故障后,2020年2月至2023年8月。一些患者接受与免疫疗法和/或西妥昔单抗组合的TI方案。评估的关键指标包括客观反应率(ORR),疾病控制率,和无进展以及总生存期。
    结果:该研究观察到ORR为15.8%,疾病控制率为36.8%。整个队列的中位无进展生存期为3.3个月,中位总生存期为9.6个月.值得注意的是,TI与免疫疗法的组合产生了30.8%的更高的ORR,与单独使用TI的14.3%相比。最常见的1-2级不良事件是贫血(81%),体重减轻(68%)和高钠血症(55%)。
    结论:基于TI的方案在治疗R/MHNSCC方面表现出良好的疗效和安全性。当将其与免疫疗法相结合时,可以获得增强的结果。这项研究表明,基于TI的治疗可以作为该特定患者组的潜在挽救选择。
    BACKGROUND: Treatment options are limited, and the prognosis is poor for patients with platinum-resistant recurrent metastatic (R/M) head and neck squamous cell carcinoma (HNSCC). This study evaluated the efficacy and safety of a paclitaxel and ifosfamide (TI) regimen in patients with R/M HNSCC whose disease had progressed following platinum-based therapy.
    METHODS: In this retrospective study, we included 53 patients with R/M HNSCC who underwent at least one cycle of TI-based therapy, post platinum failure, between February 2020 and August 2023. Some patients received the TI regimen in combination with immunotherapy and/or cetuximab. Key metrics assessed included the objective response rate (ORR), disease control rate, and progression-free as well as overall survival.
    RESULTS: The study observed an ORR of 15.8% and a disease control rate of 36.8%. The median progression-free survival for the entire cohort was 3.3 months, and the median overall survival was 9.6 months. Notably, the combination of TI with immunotherapy yielded a higher ORR of 30.8%, compared to 14.3% with TI alone. The most prevalent grade 1-2 adverse events were anemia (81%), weight loss (68%) and hypernatremia (55%).
    CONCLUSIONS: The TI-based regimen demonstrated favorable efficacy and safety profile in treating R/M HNSCC. Enhanced outcomes may be attainable when combining it with immunotherapy. This study suggests that TI-based therapy could serve as a potential salvage option for this specific patient group.
    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:神经母细胞瘤是儿童中最常见的颅外实体瘤。复发或难治性神经母细胞瘤与不良预后相关。我们评估了伊立替康-替莫唑胺和达沙替尼-雷帕霉素(RIST)在复发或难治性神经母细胞瘤患者中的组合。
    方法:多中心,开放标签,随机化,控制,第2阶段,RIST-rNB-2011试验从德国和奥地利的40个儿科肿瘤中心招募.1-25岁高危复发患者(定义为所有IV期和MYCN扩增期复发,对治疗有反应后)或难治性(主要治疗期间的进行性疾病)神经母细胞瘤,Lansky和Karnofsky的表现至少达到50%,通过区组随机分配(1:1)至RIST(RIST组)或伊立替康-替莫唑胺(对照组),按MYCN状态分层。我们比较了RIST(口服雷帕霉素[第1天加载3mg/m2,第2-4天维持1mg/m2]和口服达沙替尼[每天2mg/kg]4天,休息3天,然后静脉注射伊立替康[每天50mg/m2]和口服替莫唑胺[每天150mg/m2]5天,休息2天;雷帕霉素-达沙替尼和伊立替康-替莫唑胺各一个疗程,为期8周,然后接受两个疗程的雷帕霉素-达沙替尼,然后接受一个疗程的伊立替康-替莫唑胺,持续12周),并单独使用伊立替康-替莫唑胺(与实验组相同的剂量)。在接受至少一个疗程的所有符合条件的患者中分析无进展生存期的主要终点。安全性人群由接受至少一个疗程的所有患者组成,并进行了至少一次基线后安全性评估。该试验在ClinicalTrials.gov注册,NCT01467986,并关闭到应计。
    结果:在2013年8月26日至2020年9月21日之间,129例患者被随机分配到RIST组(n=63)或对照组(n=66)。中位年龄为5·4岁(IQR3·7-8·1)。124例患者(78例[63%]男性和46例[37%]女性)被纳入疗效分析。在72个月的中位随访时间(IQR31-88),RIST组的中位无进展生存期为11个月(95%CI7-17),对照组为5个月(2-8)(风险比0·62,单侧90%CI0·81;p=0·019).RIST组MYCN扩增患者(n=48)的中位无进展生存期为6个月(95%CI4-24),对照组为2个月(2-5)(HR0·45[95%CI0·24-0·84],p=0·012);RIST组无MYCN扩增患者(n=76)的中位无进展生存期为14个月(95%CI9-7),而对照组为8个月(4-15)(HR0·84[95%CI0·51-1·38],p=0·49)。最常见的3级或更严重的不良事件是中性粒细胞减少症(接受RIST治疗的67例患者中有54[81%],而接受对照治疗的60例患者中有49例[82%])。血小板减少症(45[67%]vs41[68%]),贫血(39[58%]vs38[63%])。报告了9例严重的治疗相关不良事件(5例患者接受对照治疗,4例患者接受RIST治疗)。对照组和RIST组(多器官衰竭)无治疗相关死亡。
    结论:RIST-rNB-2011证明,多激酶抑制剂和mTOR抑制剂的通路定向节拍组合靶向MYCN扩增的复发性或难治性神经母细胞瘤可以改善无进展生存期和总生存期。这种独特的功效在MYCN中扩增,复发性神经母细胞瘤值得在一线进一步研究.
    背景:DeutscheKrebshilfe。
    BACKGROUND: Neuroblastoma is the most common extracranial solid tumour in children. Relapsed or refractory neuroblastoma is associated with a poor outcome. We assessed the combination of irinotecan-temozolomide and dasatinib-rapamycin (RIST) in patients with relapsed or refractory neuroblastoma.
    METHODS: The multicentre, open-label, randomised, controlled, phase 2, RIST-rNB-2011 trial recruited from 40 paediatric oncology centres in Germany and Austria. Patients aged 1-25 years with high-risk relapsed (defined as recurrence of all stage IV and MYCN amplification stages, after response to treatment) or refractory (progressive disease during primary treatment) neuroblastoma, with Lansky and Karnofsky performance status at least 50%, were assigned (1:1) to RIST (RIST group) or irinotecan-temozolomide (control group) by block randomisation, stratified by MYCN status. We compared RIST (oral rapamycin [loading 3 mg/m2 on day 1, maintenance 1 mg/m2 on days 2-4] and oral dasatinib [2 mg/kg per day] for 4 days with 3 days off, followed by intravenous irinotecan [50 mg/m2 per day] and oral temozolomide [150 mg/m2 per day] for 5 days with 2 days off; one course each of rapamycin-dasatinib and irinotecan-temozolomide for four cycles over 8 weeks, then two courses of rapamycin-dasatinib followed by one course of irinotecan-temozolomide for 12 weeks) with irinotecan-temozolomide alone (with identical dosing as experimental group). The primary endpoint of progression-free survival was analysed in all eligible patients who received at least one course of therapy. The safety population consisted of all patients who received at least one course of therapy and had at least one post-baseline safety assessment. This trial is registered at ClinicalTrials.gov, NCT01467986, and is closed to accrual.
    RESULTS: Between Aug 26, 2013, and Sept 21, 2020, 129 patients were randomly assigned to the RIST group (n=63) or control group (n=66). Median age was 5·4 years (IQR 3·7-8·1). 124 patients (78 [63%] male and 46 [37%] female) were included in the efficacy analysis. At a median follow-up of 72 months (IQR 31-88), the median progression-free survival was 11 months (95% CI 7-17) in the RIST group and 5 months (2-8) in the control group (hazard ratio 0·62, one-sided 90% CI 0·81; p=0·019). Median progression-free survival in patients with amplified MYCN (n=48) was 6 months (95% CI 4-24) in the RIST group versus 2 months (2-5) in the control group (HR 0·45 [95% CI 0·24-0·84], p=0·012); median progression-free survival in patients without amplified MYCN (n=76) was 14 months (95% CI 9-7) in the RIST group versus 8 months (4-15) in the control group (HR 0·84 [95% CI 0·51-1·38], p=0·49). The most common grade 3 or worse adverse events were neutropenia (54 [81%] of 67 patients given RIST vs 49 [82%] of 60 patients given control), thrombocytopenia (45 [67%] vs 41 [68%]), and anaemia (39 [58%] vs 38 [63%]). Nine serious treatment-related adverse events were reported (five patients given control and four patients given RIST). There were no treatment-related deaths in the control group and one in the RIST group (multiorgan failure).
    CONCLUSIONS: RIST-rNB-2011 demonstrated that targeting of MYCN-amplified relapsed or refractory neuroblastoma with a pathway-directed metronomic combination of a multkinase inhibitor and an mTOR inhibitor can improve progression-free survival and overall survival. This exclusive efficacy in MYCN-amplified, relapsed neuroblastoma warrants further investigation in the first-line setting.
    BACKGROUND: Deutsche Krebshilfe.
    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)患者接受传统化疗,比如紫杉烷类药物。一种这样的药物,紫杉醇(PTX),可以有效治疗TNBC;然而,许多肿瘤会产生耐药性,这可能会导致复发。为了改善患者的预后和生存率,迫切需要了解耐药性背后的机制。我们的实验室进行了新的观察,即在人TNBC细胞系MDA-MB-157中,使用shRNA降低了腺瘤性息肉病(APC)肿瘤抑制因子的表达。在缺乏APC的细胞中,PTX诱导的细胞凋亡减少,其通过切割的半胱天冬酶3和膜联蛋白/PI染色来测量。目前的研究表明,在另外两个TNBC细胞系中,CRISPR介导的APC敲除,MDA-MB-231和SUM159导致PTX抗性。此外,已经通过对BCL-2家族蛋白的分析研究了APC介导的PTX应答背后的细胞后果和分子机制.我们发现肿瘤起始细胞群体的显着增加和促生存家族成员Bcl-2的表达增加,这是其致癌行为众所周知的。ABT-199(维尼托克),是特异性靶向Bcl-2的BH3模拟物。ABT-199已被用作多种血液系统恶性肿瘤的单一或联合疗法,并已在多种亚型的乳腺癌中显示出希望。为了解决APC诱导的Bcl-2增加是PTX抵抗的原因的假设,我们联合治疗PTX和ABT-199。这种CRISPR介导的APC敲除MDA-MB-231细胞的联合治疗导致细胞凋亡的改变。提示Bcl-2抑制可恢复APC敲除乳腺癌细胞的PTX敏感性。我们的研究首次表明Bcl-2功能抑制可恢复APC突变乳腺癌细胞中的PTX敏感性。这些研究对于改善TNBC患者的治疗方案至关重要。
    Triple-negative breast cancer (TNBC) patients are treated with traditional chemotherapy, such as the taxane class of drugs. One such drug, paclitaxel (PTX), can be effective in treating TNBC; however, many tumors will develop drug resistance, which can lead to recurrence. In order to improve patient outcomes and survival, there lies a critical need to understand the mechanism behind drug resistance. Our lab made the novel observation that decreased expression of the Adenomatous Polyposis Coli (APC) tumor suppressor using shRNA caused PTX resistance in the human TNBC cell line MDA-MB-157. In cells lacking APC, induction of apoptosis by PTX was decreased, which was measured through cleaved caspase 3 and annexin/PI staining. The current study demonstrates that CRISPR-mediated APC knockout in two other TNBC lines, MDA-MB-231 and SUM159, leads to PTX resistance. In addition, the cellular consequences and molecular mechanisms behind APC-mediated PTX response have been investigated through analysis of the BCL-2 family of proteins. We found a significant increase in the tumor-initiating cell population and increased expression of the pro-survival family member Bcl-2, which is widely known for its oncogenic behavior. ABT-199 (Venetoclax), is a BH3 mimetic that specifically targets Bcl-2. ABT-199 has been used as a single or combination therapy in multiple hematologic malignancies and has shown promise in multiple subtypes of breast cancer. To address the hypothesis that APC-induced Bcl-2 increase is responsible for PTX resistance, we combined treatment of PTX and ABT-199. This combination treatment of CRISPR-mediated APC knockout MDA-MB-231 cells resulted in alterations in apoptosis, suggesting that Bcl-2 inhibition restores PTX sensitivity in APC knockout breast cancer cells. Our studies are the first to show that Bcl-2 functional inhibition restores PTX sensitivity in APC mutant breast cancer cells. These studies are critical to advance better treatment regimens in patients with TNBC.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号