Temozolomide

替莫唑胺
  • 文章类型: Journal Article
    胶质母细胞瘤(GBM)是一种侵袭性神经系统肿瘤,预后不良。虽然,手术,放射治疗,化疗是目前GBM患者的标准方案,这些患者的预后仍然较差。替莫唑胺(TMZ)作为GBM中的一线治疗剂,可以很容易地穿过血脑屏障以抑制肿瘤细胞增殖。然而,GBM患者TMZ耐药率较高。因为,出现TMZ耐药的GBM患者的治疗选择有限;需要阐明化疗耐药的分子机制以引入新的治疗靶点.microRNAs(miRNAs)通过调节药物代谢来调节化疗耐药,吸收,DNA修复,凋亡,和细胞周期。在这篇综述中,我们讨论了miRNAs在GBM细胞TMZ反应中的作用。据报道,miRNAs主要通过调节GBM细胞的信号通路和自噬来诱导TMZ敏感性。因此,miRNAs可作为GBM患者可靠的诊断/预后标志物。它们也可以用作治疗靶标以改善GBM细胞中的TMZ应答。
    Glioblastoma (GBM) is an aggressive nervous system tumor with a poor prognosis. Although, surgery, radiation therapy, and chemotherapy are the current standard protocol for GBM patients, there is still a poor prognosis in these patients. Temozolomide (TMZ) as a first-line therapeutic agent in GBM can easily cross from the blood-brain barrier to inhibit tumor cell proliferation. However, there is a high rate of TMZ resistance in GBM patients. Since, there are limited therapeutic choices for GBM patients who develop TMZ resistance; it is required to clarify the molecular mechanisms of chemo resistance to introduce the novel therapeutic targets. MicroRNAs (miRNAs) regulate chemo resistance through regulation of drug metabolism, absorption, DNA repair, apoptosis, and cell cycle. In the present review we discussed the role of miRNAs in TMZ response of GBM cells. It has been reported that miRNAs mainly induced TMZ sensitivity by regulation of signaling pathways and autophagy in GBM cells. Therefore, miRNAs can be used as the reliable diagnostic/prognostic markers in GBM patients. They can also be used as the therapeutic targets to improve the TMZ response in GBM 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
    胶质瘤癌症是全球男性和女性癌症相关死亡的主要原因。这种病症的传统化疗治疗经常导致疗效降低和显著的不良反应。这项研究使用pH敏感的药物递送沸石咪唑酯骨架(ZIF-8)为化疗药物替莫唑胺(TMZ)开发了一种新的药物递送系统。这些纳米平台显示出优异的生物相容性,并具有癌症治疗的潜力。利用ZIF提供的有利反应环境,“一锅法”用于制造和装载药物,导致一个良好的承载能力。TMZ@TA@ZIF-8NP对酸性环境表现出显著的反应,导致以受控释放结果为特征的增强的药物释放模式。TMZ@TA@ZIF-8NPs抑制U251胶质瘤细胞迁移和侵袭的作用,以及促进细胞凋亡,通过各种测试证实,包括MTT(3-(4,5)-二甲基硫代偶氮(-z-y1))测定,DAPI/PI双重染色,和细胞划痕分析。生化荧光染色实验表明TMZ@TA@ZIF-8NPs潜在地改善了ROS,减少MMP,并引发U251细胞凋亡。在用NP处理的U251细胞中,p53,Bax,Cyt-C,caspase-3、-8和-9表达显著增强,而Bcl-2表达减少。这些结果显示TMZ@TA@ZIF-8NP作为具有抗神经胶质瘤特性的治疗剂的潜力。总的来说,我们使用TMZ@TA@ZIF-8NP制造的pH响应性药物递送系统显示出巨大的癌症治疗潜力.该方法具有通过克服与基于TMZ的治疗相关的问题而对改善癌症治疗做出重大贡献的潜力。
    Glioma cancer is the primary cause of cancer-related fatalities globally for both men and women. Traditional chemotherapy treatments for this condition frequently result in reduced efficacy and significant adverse effects. This investigation developed a new drug delivery system for the chemotherapeutic drug temozolomide (TMZ) using pH-sensitive drug delivery zeolitic imidazolate frameworks (ZIF-8). These nanoplatforms demonstrate excellent biocompatibility and hold potential for cancer therapy. Utilizing the favorable reaction milieu offered by ZIFs, a \'one-pot method\' was employed for the fabrication and loading of drugs, leading to a good capacity for loading. TMZ@TA@ZIF-8 NPs exhibit a notable response to an acidic milieu, resulting in an enhanced drug release pattern characterized by a controlled release outcome. The effectiveness of TMZ@TA@ZIF-8 NPs in inhibiting the migration and invasion of U251 glioma cancer cells, as well as promoting apoptosis, was confirmed through various tests, including MTT (3-(4,5)-dimethylthiahiazo(-z-y1)) assay, DAPI/PI dual staining, and cell scratch assay. The biochemical fluorescent staining assays showed that TMZ@TA@ZIF-8 NPs potentially improved ROS, reduced MMP, and triggered apoptosis in U251 cells. In U251 cells treated with NPs, the p53, Bax, Cyt-C, caspase-3, -8, and -9 expressions were significantly enhanced, while Bcl-2 expression was diminished. These outcomes show the potential of TMZ@TA@ZIF-8 NPs as a therapeutic agent with anti-glioma properties. Overall, the pH-responsive drug delivery systems we fabricated using TMZ@TA@ZIF-8 NPs show great potential for cancer treatment. This approach has the potential to make significant contributions to the improvement of cancer therapy by overcoming the problems associated with TMZ-based treatments.
    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
    新诊断的弥漫性低度胶质瘤(DLGG)患者神经外科切除术后的治疗时机和选择仍存在争议。的确,这种治疗的效果必须与可能的副作用相平衡。这项研究评估了接受一线替莫唑胺治疗的DLGG患者的纵向力竭生活质量(QoL)和神经心理学评估的可行性。
    QoL,神经认知,和心理障碍进行前瞻性评估,直到疾病进展,使用测试,临床医生报告,和自我报告的问卷。主要终点是基线时(替莫唑胺开始前)对这一完整评估的参与和依从性,6个月和12个月的治疗,和治疗后6个月。还描述了QoL和神经心理学随时间的变化。
    29名符合条件的患者中有26名入组(参与率:89.7%,95%CI:72.6-97.8)。依从率为95.7%(95%CI:78.1-99.9;n=23,因为3名患者在治疗的前12个月内进展)。治疗后6个月,QoL和疲劳保持稳定(EORTCQLQC30和BN20,MFI-20);一些特定症状是短暂的。主观(FACT-Cog)和客观(神经认知测试的Z得分)神经认知结果均保持稳定或趋于改善。重度抑郁症患者(BDI-II)的百分比,焦虑(STAI-Y),或愤怒(STAXI-II)随着时间的推移是稳定的。
    这项前瞻性研究证明了对QoL进行详尽和纵向评估的可行性,神经认知,和心理障碍,接受化疗的DLGG患者具有很高的可接受性。一线替莫唑胺似乎对QoL和神经认知的短期影响有限。这些发现必须在长期和更大的队列中得到证实。
    UNASSIGNED: The treatment timing and choice after neurosurgical resection in patients with newly diagnosed diffuse low-grade glioma (DLGG) remain controversial. Indeed, the effect of such treatments must be balanced with the possible side effects. This study evaluated the feasibility of longitudinal exhaustive quality of life (QoL) and neuropsychological assessments in patients with DLGG receiving first-line temozolomide.
    UNASSIGNED: QoL, neurocognition, and psychological disorders were assessed prospectively until disease progression, using testing, clinician-reported, and self-reported questionnaires. The primary endpoint was the participation and adherence to this complete assessment at Baseline (before temozolomide initiation), months 6 and 12 of treatment, and month 6 post-treatment. The QoL and neuropsychological changes over time also were described.
    UNASSIGNED: Twenty-six of the twenty-nine eligible patients were enrolled (participation rate: 89.7%, 95% CI: 72.6-97.8). The adherence rate was 95.7% (95% CI: 78.1-99.9; n = 23 because 3 patients progressed in the first 12 months of treatment). Up to month 6 post-treatment, QoL and fatigue remained stable (EORTC QLQC30 and BN20, MFI-20); some specific symptoms were transitory. Both subjective (FACT-Cog) and objective (Z-scores of neurocognitive tests) neurocognitive outcomes remained stable or tended to improve. The percentage of patients with severe depression (BDI-II), anxiety (STAI-Y), or anger (STAXI-II) was stable over time.
    UNASSIGNED: This prospective study demonstrated the feasibility of an exhaustive and longitudinal evaluation of QoL, neurocognition, and psychological disorders, with high acceptability by patients with DLGG undergoing chemotherapy. First-line temozolomide seems to have limited short-term effects on QoL and neurocognition. These findings must be confirmed in the long term and in a larger cohort.
    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
    尽管在92%的TP53突变癌症中发生了突变,p53同工型的突变如何影响它们的活性仍然是未知的.因此,探索突变对p53同工型活性的影响是至关重要的,尽管在p53领域尚未探索的领域。在这篇文章中,我们首次报道了突变的Δ133p53α特异性途径,该途径增加了IL4I1和IDO1的表达并激活了AHR,促进肿瘤的机制。因此,而WTΔ133p53α减少细胞凋亡以促进DNA修复,突变体R273H也减少细胞凋亡,但不能保持基因组的稳定性,增加突变积累和肿瘤向更具侵袭性表型的风险。此外,使用2D和3D球体培养,我们显示WTΔ133p53α减少细胞增殖,EMT,和入侵,而突变体Δ133p53αR273H增强了所有三个过程,证实了其致癌潜力,并强烈暗示了类似的体内活性。重要的是,对细胞生长和侵袭的影响与突变体全长p53α无关,表明这些活性由突变体Δ133p53αR273H积极携带。此外,WT和突变型Δ133p53α均以衰老诱导剂依赖性方式(替莫唑胺或辐射)降低细胞衰老,因为它们调节不同的衰老相关靶基因。因此,WTΔ133p53α挽救了替莫唑胺诱导的衰老,但不是辐射诱导的衰老,而突变体Δ133p53αR273H挽救了辐射诱导的衰老,但不是替莫唑胺诱导的衰老。最后,我们确定IL4I1,IDO1和AHR在GBM中显著高于低级别胶质瘤.重要的是,LGG和IL4I1在GBM中的所有三个基因的高表达与较差的患者生存率显着相关,证实该途径在胶质母细胞瘤中的临床相关性。这些数据表明,与WTΔ133p53α相比,R273H突变将其活性重新定向为致癌作用,并激活致癌IL4I1/IDO1/AHR途径,通过结合特异性调节Δ133p53α表达的药物和IDO1/Il4I1/AHR抑制剂,成为GBM中潜在的预后标志物和治疗靶标。
    Despite being mutated in 92% of TP53 mutant cancers, how mutations on p53 isoforms affect their activities remain largely unknown. Therefore, exploring the effect of mutations on p53 isoforms activities is a critical, albeit unexplored area in the p53 field. In this article, we report for the first time a mutant Δ133p53α-specific pathway which increases IL4I1 and IDO1 expression and activates AHR, a tumor-promoting mechanism. Accordingly, while WT Δ133p53α reduces apoptosis to promote DNA repair, mutant R273H also reduces apoptosis but fails to maintain genomic stability, increasing the risks of accumulation of mutations and tumor\'s deriving towards a more aggressive phenotype. Furthermore, using 2D and 3D spheroids culture, we show that WT Δ133p53α reduces cell proliferation, EMT, and invasion, while the mutant Δ133p53α R273H enhances all three processes, confirming its oncogenic potential and strongly suggesting a similar in vivo activity. Importantly, the effects on cell growth and invasion are independent of mutant full-length p53α, indicating that these activities are actively carried by mutant Δ133p53α R273H. Furthermore, both WT and mutant Δ133p53α reduce cellular senescence in a senescence inducer-dependent manner (temozolomide or radiation) because they regulate different senescence-associated target genes. Hence, WT Δ133p53α rescues temozolomide-induced but not radiation-induced senescence, while mutant Δ133p53α R273H rescues radiation-induced but not temozolomide-induced senescence. Lastly, we determined that IL4I1, IDO1, and AHR are significantly higher in GBMs compared to low-grade gliomas. Importantly, high expression of all three genes in LGG and IL4I1 in GBM is significantly associated with poorer patients\' survival, confirming the clinical relevance of this pathway in glioblastomas. These data show that, compared to WT Δ133p53α, R273H mutation reorientates its activities toward carcinogenesis and activates the oncogenic IL4I1/IDO1/AHR pathway, a potential prognostic marker and therapeutic target in GBM by combining drugs specifically modulating Δ133p53α expression and IDO1/Il4I1/AHR 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: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
    多形性胶质母细胞瘤(GBM)是最常见和侵袭性的原发性脑肿瘤。尽管基于替莫唑胺(TMZ)的放化疗可改善GBM患者的总体生存率,它还增加了治疗后磁共振成像(MRI)评估肿瘤进展的假阳性频率.假性进展(PsP)是一种与治疗相关的反应,在MRI上,肿瘤部位或切除边缘的对比增强病变大小增加,影响肿瘤复发。在GBM患者的临床管理中,迫切需要准确可靠地预测GBM进展。临床资料分析表明,PsP患者的总体生存率和无进展生存率均较高。在这项研究中,我们旨在建立一个预后模型,以评估GBM患者接受标准治疗后的肿瘤进展潜能.我们应用字典学习方案从Wake数据集中获得具有PsP或真实肿瘤进展(TTP)的GBM患者的成像特征。基于这些射线照相特征,我们进行了放射基因组学分析,以鉴定显著相关的基因.这些显著相关的基因被用作构建2YS(2年生存率)逻辑回归模型的特征。根据从该模型得到的个体2YS评分将GBM患者分为低生存风险组和高生存风险组。我们使用独立的癌症基因组图谱计划(TCGA)数据集测试了我们的模型,发现2YS评分与患者的总生存期显着相关。我们使用了两组TCGA数据来训练和测试我们的模型。我们的结果表明,来自训练和测试TCGA数据集的基于2YS分数的分类结果与患者的总体生存率显着相关。我们还分析了其他临床因素(性别,年龄,KPS(Karnofsky性能状态),正常细胞比率),并发现这些因素与患者的生存无关或弱相关。总的来说,我们的研究证明了2YS模型在预测GBM患者接受标准治疗后的临床结局方面的有效性和稳健性.
    Glioblastoma multiforme (GBM)is the most common and aggressive primary brain tumor. Although temozolomide (TMZ)-based radiochemotherapy improves overall GBM patients\' survival, it also increases the frequency of false positive post-treatment magnetic resonance imaging (MRI) assessments for tumor progression. Pseudo-progression (PsP) is a treatment-related reaction with an increased contrast-enhancing lesion size at the tumor site or resection margins miming tumor recurrence on MRI. The accurate and reliable prognostication of GBM progression is urgently needed in the clinical management of GBM patients. Clinical data analysis indicates that the patients with PsP had superior overall and progression-free survival rates. In this study, we aimed to develop a prognostic model to evaluate the tumor progression potential of GBM patients following standard therapies. We applied a dictionary learning scheme to obtain imaging features of GBM patients with PsP or true tumor progression (TTP) from the Wake dataset. Based on these radiographic features, we conducted a radiogenomics analysis to identify the significantly associated genes. These significantly associated genes were used as features to construct a 2YS (2-year survival rate) logistic regression model. GBM patients were classified into low- and high-survival risk groups based on the individual 2YS scores derived from this model. We tested our model using an independent The Cancer Genome Atlas Program (TCGA) dataset and found that 2YS scores were significantly associated with the patient\'s overall survival. We used two cohorts of the TCGA data to train and test our model. Our results show that the 2YS scores-based classification results from the training and testing TCGA datasets were significantly associated with the overall survival of patients. We also analyzed the survival prediction ability of other clinical factors (gender, age, KPS (Karnofsky performance status), normal cell ratio) and found that these factors were unrelated or weakly correlated with patients\' survival. Overall, our studies have demonstrated the effectiveness and robustness of the 2YS model in predicting the clinical outcomes of GBM patients after standard 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
    多形性胶质母细胞瘤(GBM)是一种最具侵袭性的脑肿瘤,其特征是预后令人望而生畏,预期寿命徘徊在12-16个月左右。尽管进行了一个世纪的不懈研究,只有少数药物获得脑肿瘤治疗的批准,很大程度上是由于血脑屏障造成的强大屏障。目前的护理标准涉及多方面的方法结合手术,辐照,和化疗。然而,尽管采取了这些干预措施,但复发通常在数月内发生.药物递送至脑和克服治疗抗性的严峻挑战已成为脑肿瘤治疗中的焦点,并且被认为是克服肿瘤复发所必需的。近年来,一波有希望的先进治疗已经出现,提供了克服现有疗法局限性的希望。这篇综述旨在突出当前和持续发展阶段的尖端技术,为患者提供有价值的见解,以指导他们在脑肿瘤治疗中的选择。
    Glioblastoma multiforme (GBM) is one of the most aggressive forms of brain tumor, characterized by a daunting prognosis with a life expectancy hovering around 12-16 months. Despite a century of relentless research, only a select few drugs have received approval for brain tumor treatment, largely due to the formidable barrier posed by the blood-brain barrier. The current standard of care involves a multifaceted approach combining surgery, irradiation, and chemotherapy. However, recurrence often occurs within months despite these interventions. The formidable challenges of drug delivery to the brain and overcoming therapeutic resistance have become focal points in the treatment of brain tumors and are deemed essential to overcoming tumor recurrence. In recent years, a promising wave of advanced treatments has emerged, offering a glimpse of hope to overcome the limitations of existing therapies. This review aims to highlight cutting-edge technologies in the current and ongoing stages of development, providing patients with valuable insights to guide their choices in brain tumor 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
    胶质母细胞瘤(GBM)是一种致命的星形胶质细胞瘤,预后差,治疗耐药。重新使用潜在的FDA批准的药物,如抗精神病药,可以及时和具有成本效益的方式解决这些问题。流行病学研究表明,使用抗精神病药的精神分裂症患者的GBM发生率较低。因此,我们的目的是研究非典型抗精神病药物伊洛哌酮(ILO)单药和替莫唑胺(TMZ)联合治疗GBM的治疗潜力.该研究评估了国际劳工组织的生长抑制作用,TMZ,以及使用MTT测定在U-87MG和T-98G细胞系上的它们的组合(ILO+TMZ)。测定药物相互作用系数(CDI),并将具有协同作用的剂量用于后续实验,包括迁徙,入侵,和TUNEL检测。DRD2、β-catenin、Dvl2,Twist,和Slug通过RTq-PCR进行评估,而β-连环蛋白的表达也通过免疫细胞化学测定。ILO(p<0.05)和TMZ(p<0.01)在所有测试剂量下均显著抑制U-87MG细胞的生长。两种药物的60μM的组合显示与CDI<1的协同活性。在联合治疗的情况下,迁移和凋亡的抑制更为明显(p<0.001)。还发现在ILO和组合处理组中对侵入细胞的抑制是显著的(p<0.001)。ILO和联合治疗也显著下调DRD2的表达,而TMZ上调表达(p<0.001)。β-catenin的表达(p<0.001),Dvl2(p<0.001),Twist(p<0.001),和Slug(p<0.001)在所有治疗组中与载体对照相比也显著下调。数据表明,国际劳工组织具有很强的生长抑制活性,可能是由于其对DRD2和β-连环蛋白表达的影响,并且有可能针对GBM重新利用。
    Glioblastoma (GBM) is a fatal astrocytic glioma with poor prognosis and treatment resistance. Repurposing potential FDA-approved drugs like anti-psychotics can address the concerns in a timely and cost-effective manner. Epidemiological studies have shown that patients with schizophrenic using anti-psychotics have a low incidence of GBM. Therefore, we aimed to investigate the therapeutic potential of atypical anti-psychotic Iloperidone (ILO) alone and in combination with Temozolomide (TMZ) against GBM. The study assessed the growth inhibitory effect of ILO, TMZ, and their combination (ILO + TMZ) on U-87MG and T-98G cell lines using an MTT assay. The drug interaction coefficient (CDI) was determined, and doses with synergistic effects were used for subsequent experiments, including migratory, invasion, and TUNEL assays. The expressions of DRD2, β-catenin, Dvl2, Twist, and Slug were assessed by RTq-PCR, whereas the β-catenin protein expression was also determined by immunocytochemistry. ILO (p < 0.05) and TMZ (p < 0.01) significantly inhibited the growth of U-87MG cells at all tested doses. The combination of 60 µM of both drugs showed synergistic activity with CDI < 1. The inhibition of migration and apoptosis was more pronounced in the case of combination treatment (p < 0.001). Inhibition of the invading cells was also found to be significant in ILO- and combination-treated groups (p < 0.001). ILO and combination treatment also significantly downregulated the expression of DRD2, while TMZ upregulated the expression (p < 0.001). The expressions of β-catenin (p < 0.001), Dvl2 (p < 0.001), Twist (p < 0.001), and Slug (p < 0.001) were also significantly downregulated in all treatment groups as compared to the vehicle control. The data suggest that ILO possesses strong growth inhibitory activity, possibly due to its effect on DRD2 and β-catenin expression and has the potential to be repurposed against 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
    目的:胶质母细胞瘤是一种无法治愈的癌症,治疗选择有限,生存率低。替莫唑胺是用于胶质母细胞瘤治疗的标准市售小分子药物;因此,由于其脑迁移特性,我们旨在在市售的治疗脑疾病的药物中寻找新药,并发现了洛美利嗪,用于治疗偏头痛。
    方法:我们评估了洛美利嗪及其代谢物对U251胶质母细胞瘤细胞和替莫唑胺耐药细胞的作用,由O(6)-甲基鸟嘌呤-DNA甲基转移酶或P-糖蛋白的表达引起的T98G和GB-1,与替莫唑胺相比,并与之结合。使用坏死或凋亡抑制剂研究了作用机理。
    结果:洛美利嗪及其代谢产物(M6)比替莫唑胺抑制胶质母细胞瘤细胞增殖的效力和效力更大,包括对抗替莫唑胺抗性细胞。洛美利嗪和M6对胶质母细胞瘤的作用主要归因于抑制增殖,因为细胞死亡抑制剂无法挽救细胞,如坏死或凋亡抑制剂,尽管它们被坏死抑制素-1稍微拯救。此外,洛美利嗪和M6联合替莫唑胺在某些剂量下比单一治疗更有效地抑制U251和GB-1细胞的增殖。
    结论:洛美利嗪因其具有脑穿透性而没有严重副作用而被用于偏头痛的治疗;因此,它可能会被单独用于胶质母细胞瘤,包括替莫唑胺耐药的胶质母细胞瘤,或与替莫唑胺联合使用。
    OBJECTIVE: Glioblastoma is an incurable cancer with limited treatment options and a low survival rate. Temozolomide is the standard marketed small-molecule agent for glioblastoma therapy; therefore, we aimed to find new drugs among the marketed medicines for brain diseases because of their cerebral migratory property and found lomerizine, used for the treatment of migraine.
    METHODS: We evaluated the effect of lomerizine and its metabolites against U251 glioblastoma cells and temozolomide-resistant cells, T98G and GB-1, caused by the expression of O(6)-methylguanine-DNA methyltransferase or P-glycoprotein, compared with temozolomide, and combined with it. The mechanism of action was investigated using inhibitors of necrosis or apoptosis.
    RESULTS: Lomerizine and its metabolite (M6) inhibited the proliferation of glioblastoma cells with greater potency and efficacy than temozolomide, including against temozolomide-resistant cells. The effects of lomerizine and M6 on glioblastoma were mainly attributed to the inhibition of proliferation because cells were not rescued by cell death inhibitors, such as necrosis or apoptosis inhibitors, although they were slightly rescued by necrostatin-1. Additionally, lomerizine and M6 combined with temozolomide were more effective at inhibiting the proliferation of U251 and GB-1 cells at some doses than single treatments.
    CONCLUSIONS: Lomerizine has been used for migraine treatment because of its brain-penetrating properties without serious side-effects; thus, it might potentially be expected to be used alone for glioblastoma, including temozolomide-resistant glioblastoma, or in combination with temozolomide.
    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
    总结Goyal目的评估MGMT基因阳性与诊断为脑肿瘤的患者更好的生存相关世界卫生组织(WHO)III级和IV级材料和方法单研究所回顾性研究。共纳入80例患者,所有患者均接受了肿瘤全切除或次全切除手术,并通过RT-PCR对肿瘤组织进行MGMT基因检测.所有都接受了辅助辐射(60Gy/30份,5份/周)同时使用替莫唑胺(75mg/m2),随后进行12个周期的替莫唑胺辅助治疗(第1个周期为150mg/m2,随后为200mg/m2),并定期随访。结果共80例患者,75人接受了小计切除术,27人是WHOIII级与48世卫组织四级。5例接受全切除术1是WHOIII级与4世卫组织四级。与IV级相比,III级患者的5例患者的中位PFS和OS分别为9.0和20。其中PFS和OS中位数为8.8个月和17.8个月。在小计组的75名患者中,中位PFS和OS,分别,在III级组中分别为9.1和19.3,WHOIV级,中位PFS为8.8,OS为18.8。结论MGMT基因阳性是Ⅲ、Ⅳ级脑肿瘤的预后因素。
    Sumit GoyalObjective  To evaluate MGMT gene positivity is associated with better survival in patients diagnosed with brain tumor World Health Organization (WHO) grades III and IV Material and Methods  Single-institute restrospective study. A total of 80 patients were enrolled, all underwent surgery either total or subtotal excision of the tumor and MGMT gene testing on tumor tissue by RT-PCR. All received adjuvant radiation (60 Gy/30 fractions, 5 fractions/week) with concurrent temozolomide (75 mg/m 2 ), followed by 12 cycles of adjuvant temozolomide (150 mg/m 2 1st cycle followed by 200 mg/m 2 ) with regular follow-up. Results  A total of 80 patients, 75 underwent subtotal excision, 27 were WHO grade III vs. 48 WHO grade IV. Five underwent total excision 1 was WHO grade III vs. 4 WHO grade IV. The median PFS and OS in five patients in total excision in grade III patient was 9.0 and 20 compared with Grade IV, where the median PFS and OS was 8.8 and 17.8 months. Out of 75 patients in the subtotal group median PFS and OS, respectively, in Grade III group was 9.1 and 19.3 and, WHO grade IV with median PFS of 8.8 and OS of 18.8. Conclusion   MGMT gene positivity is a prognostic factor in grade III and IV brain tumor.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号