Histone deacetylase inhibitor

组蛋白去乙酰化酶抑制剂
  • 文章类型: Journal Article
    在雌激素受体(ER)阳性乳腺癌的治疗中建立了抗雌激素疗法。然而,为了克服耐药性并改善治疗效果,需要新的策略,例如针对广泛认可的异常表观遗传学.本研究旨在探讨芳香化酶抑制剂依西美坦和组蛋白去乙酰化酶(HDAC)抑制剂和抗氧化剂α-硫辛酸在ER阳性乳腺癌细胞中的联合作用。首先,α-硫辛酸的对映体和外消旋混合物,研究了rac-二氢-硫辛酸对HDAC的抑制作用。我们发现HDAC抑制活性在1-3位数微摩尔范围内,优选HDAC6。Rac-二氢-硫辛酸比rac-α-硫辛酸略强。α-硫辛酸的抗增殖IC50值在3位数微摩尔范围内。值得注意的是,依西美坦和α-硫辛酸的组合在不同的孵育时间(24h至10d)和读数(MTT,活细胞荧光显微镜,半胱天冬酶激活)通过Chou-Talalay方法分析。α-硫辛酸增加线粒体融合和凋亡相关蛋白p21、APAF-1、BIM、FOXO1,并降低抗凋亡蛋白survivin的表达,BCL-2和c-myc。总之,联合使用依西美坦和α-硫辛酸是ER阳性乳腺癌的一种有前景的新型治疗选择.
    Anti-estrogenic therapy is established in the management of estrogen receptor (ER)-positive breast cancer. However, to overcome resistance and improve therapeutic outcome, novel strategies are needed such as targeting widely recognized aberrant epigenetics. The study aims to investigate the combination of the aromatase inhibitor exemestane and the histone deacetylase (HDAC) inhibitor and antioxidant α-lipoic acid in ER-positive breast cancer cells. First, the enantiomers and the racemic mixture of α-lipoic acid, and rac-dihydro-lipoic acid were investigated for HDAC inhibition. We found HDAC inhibitory activity in the 1-3-digit micromolar range with a preference for HDAC6. Rac-dihydro-lipoic acid is slightly more potent than rac-α-lipoic acid. The antiproliferative IC50 value of α-lipoic acid is in the 3-digit micromolar range. Notably, the combination of exemestane and α-lipoic acid resulted in synergistic behavior under various incubation times (24 h to 10 d) and readouts (MTT, live-cell fluorescence microscopy, caspase activation) analyzed by the Chou-Talalay method. α-lipoic acid increases mitochondrial fusion and the expression of apoptosis-related proteins p21, APAF-1, BIM, FOXO1, and decreases expression of anti-apoptotic proteins survivin, BCL-2, and c-myc. In conclusion, combining exemestane with α-lipoic acid is a promising novel treatment option for ER-positive 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
    免疫检查点抑制剂(CPIs)已在许多早期和晚期恶性肿瘤中广泛采用。已经提出组蛋白脱乙酰酶抑制剂(HDACis)和烷化剂(AAs)增强CPIs对肿瘤细胞的作用。我们进行了全面的文献综述,以探索CPIs之间的潜在协同活性。AAS,和HDACis。
    在PubMed中使用预定义的搜索字符串识别了临床和非临床研究,这些研究描述了接受CPIs和伴随或序贯(CPI前或后)AA或HDACis的癌症患者的预后。类似地进行关键肿瘤学大会的手动搜索。所有相关文章和摘要都是手动筛选相关性的,根据使用的特定抗癌剂(CPIs,AAS,或HDACis),肿瘤实体,以及治疗是否伴随或序贯。
    总的来说,针对一系列肿瘤类型的227项独特临床研究,实体瘤和血液恶性肿瘤,已确定。审查了关于I期和II期临床研究的109种出版物以及关于III期研究的41种出版物。最常见的肿瘤类型是黑色素瘤,三阴性乳腺癌,非小细胞肺癌,和霍奇金淋巴瘤.随机临床研究确定,所有这些都报告了CPI与AA的组合,与CPI或AA单药治疗相比,联合治疗组的结局更好。同样,CPIs和HDACis的联合治疗证明了有希望的活性。
    CPI与AA或HDACi的序贯或伴随给药可以改善患有多种肿瘤类型的患者的预后。有理由支持对CPI之间协同作用的潜力进行进一步调查,烷化剂和/或HDAC在非临床和临床设置。
    接受癌症治疗的人通常一次会接受一种以上的药物,和癌症药物联合治疗的概念经常被认为是改善患者预后的潜在机会。我们回顾了已发表的临床试验文献和实验室开展的工作,以探索将阻止癌细胞增殖的靶向药物(称为检查点抑制剂)与杀死癌细胞的传统化学疗法相结合是否可能是一种有用的方法。我们在出版物中看到了在化疗的同时使用检查点抑制剂的证据,或在化疗前或后立即给予。最重要的证据来自临床试验,其中直接将接受联合治疗的患者的结果与接受单一治疗的患者的结果进行比较。这些研究表明,与接受单一疗法的患者相比,接受癌症药物组合治疗的患者的预后更好。我们还发现了添加另一类抗癌药物的证据,称为组蛋白脱乙酰酶抑制剂,可能会使肿瘤对检查点抑制剂敏感。这些发现为检查烷化剂和/或组蛋白脱乙酰酶抑制剂与检查点抑制剂的组合提供了理论基础。
    UNASSIGNED: Immune checkpoint inhibitors (CPIs) have been widely adopted in a number of early and advanced malignancies. Histone deacetylase inhibitors (HDACis) and alkylating agents (AAs) have been suggested to potentiate the actions of CPIs on tumor cells. We conducted a comprehensive literature review to explore the potential synergistic activity between CPIs, AAs, and HDACis.
    UNASSIGNED: Clinical and non-clinical studies describing outcomes in patients with cancer receiving CPIs and either concomitant or sequential (pre- or post-CPI) AAs or HDACis were identified in PubMed using pre-defined search strings. Manual searches of key oncology congresses were similarly performed. All relevant articles and abstracts were manually screened for relevance, classified according to the specific anticancer agents used (CPIs, AAs, or HDACis), tumor entity, and whether treatment was concomitant or sequential.
    UNASSIGNED: Overall, 227 unique clinical studies across a range of tumor types, both solid tumors and hematological malignancies, were identified. One hundred and fifty-nine publications on Phase I and II clinical studies together with 41 publications on Phase III studies were examined. The most commonly investigated tumor types were melanoma, triple-negative breast cancer, non-small cell lung cancer, and Hodgkin lymphoma. The randomized clinical studies identified, all of which reported on the combination of a CPI with an AA, demonstrated superior outcomes in the combination arm compared with CPI or AA monotherapy. Similarly, combination therapy with CPIs and HDACis demonstrated promising activity.
    UNASSIGNED: Sequential or concomitant administration of a CPI with an AA or an HDACi may improve outcomes for patients with a range of tumor types. There is a rationale to support further investigation into the potential for synergy between CPIs, alkylating agents and/or HDACis in both the non-clinical and clinical settings.
    People being treated for cancer will often receive more than one drug at a time, and the concept of combining cancer drugs is frequently investigated as a potential opportunity to improve outcomes for patients. We reviewed the published literature for clinical trials and work undertaken in laboratories to explore whether combining targeted agents that stop cancer cells from multiplying (known as checkpoint inhibitors) with traditional chemotherapy that kills cancer cells could be a useful approach. We looked at evidence in publications where checkpoint inhibitors were used at the same time as chemotherapy, or given immediately before or after chemotherapy. The most important evidence came from clinical trials where outcomes for patients receiving combinations of treatment were directly compared with those from patients receiving a single treatment. These studies showed superior outcomes for patients who were treated with a combination of cancer drugs compared with patients receiving monotherapy. We also found evidence that adding another class of cancer drug, called histone deacetylase inhibitors, might sensitize tumors to checkpoint inhibitors. These findings provide a rationale for examining alkylating agents and/or histone deacetylase inhibitors combined with checkpoint 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
    组蛋白脱乙酰酶(HDAC)催化去除组蛋白和非组蛋白的乙酰化修饰,调节基因表达和其他细胞过程。HDAC抑制剂(HDACi),批准的抗癌剂,成为心脏病的潜在新疗法。在许多心脏疾病的临床前动物模型中观察到HDACi的心脏保护作用。已经开发了遗传小鼠模型来了解每种HDAC在心脏功能中的作用。一些发现是有争议的。这里,我们概述了HDACi和HDAC在生理或病理条件下如何影响心脏功能.我们专注于锌依赖性经典HDAC的体内研究,强调涉及心脏肥大的疾病,心肌梗死(MI),缺血再灌注(I/R)损伤,和心力衰竭。特别是,我们回顾了无偏组学研究如何帮助我们理解HDACi和HDAC心脏效应的潜在机制.
    Histone deacetylases (HDAC) catalyze the removal of acetylation modifications on histones and non-histone proteins, which regulates gene expression and other cellular processes. HDAC inhibitors (HDACi), approved anti-cancer agents, emerge as a potential new therapy for heart diseases. Cardioprotective effects of HDACi are observed in many preclinical animal models of heart diseases. Genetic mouse models have been developed to understand the role of each HDAC in cardiac functions. Some of the findings are controversial. Here, we provide an overview of how HDACi and HDAC impact cardiac functions under physiological or pathological conditions. We focus on in vivo studies of zinc-dependent classical HDACs, emphasizing disease conditions involving cardiac hypertrophy, myocardial infarction (MI), ischemic reperfusion (I/R) injury, and heart failure. In particular, we review how non-biased omics studies can help our understanding of the mechanisms underlying the cardiac effects of HDACi and HDAC.
    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
    胰腺导管腺癌(PDAC)是胰腺癌的最致命的形式,其特征是治疗抵抗和早期转移。导致存活率低。组蛋白去乙酰化酶(HDAC)抑制剂显示出治疗血液恶性肿瘤的潜力。在PDAC中,HDAC2的过表达与上皮-间质转化(EMT)有关,主要伴随着上皮标志物E-cadherin的下调和转移能力的增加。已知效应细胞因子转化生长因子-β(TGFβ)是PDAC中EMT的主要诱导因子,导致高转移和侵袭潜力。此外,PDAC中HDAC6的过表达与细胞凋亡减少有关。这里,我们已经证明,一种新型HDAC2/6抑制剂不仅显著增加了PANC-1细胞(5.5倍)和3DPDAC共培养球体(2.5倍)中E-cadherin的表达,而且能够逆转TGF-β诱导的E-cadherin表达下调.此外,我们的研究表明,与对照组相比,HDAC抑制剂介导的再分化导致肿瘤细胞侵袭的显著抑制约60%.特别是,我们已经证明HDAC抑制剂诱导细胞凋亡(2倍)和细胞周期停滞.总之,HDAC2/6抑制剂通过上调HDAC2阻断介导的E-cadherin来抑制侵袭,并通过HDAC6抑制诱导细胞周期停滞导致细胞凋亡。这些结果表明,HDAC2/6抑制剂可能代表了治疗PDAC肿瘤发生和转移的新治疗策略。
    Pancreatic ductal adenocarcinoma (PDAC) is the most lethal form of pancreatic cancer characterized by therapy resistance and early metastasis, resulting in a low survival rate. Histone deacetylase (HDAC) inhibitors showed potential for the treatment of hematological malignancies. In PDAC, the overexpression of HDAC 2 is associated with the epithelial-mesenchymal transition (EMT), principally accompanied by the downregulation of the epithelial marker E-cadherin and increased metastatic capacity. The effector cytokine transforming growth factor-β (TGF β) is known to be a major inducer of the EMT in PDAC, leading to high metastatic and invasive potential. In addition, the overexpression of HDAC 6 in PDAC is associated with reduced apoptosis. Here, we have demonstrated that a novel HDAC 2/6 inhibitor not only significantly increased E-cadherin expression in PANC-1 cells (5.5-fold) and in 3D PDAC co-culture spheroids (2.5-fold) but was also able to reverse the TGF-β-induced downregulation of E-cadherin expression. Moreover, our study indicates that the HDAC inhibitor mediated re-differentiation resulting in a significant inhibition of tumor cell invasion by approximately 60% compared to control. In particular, we have shown that the HDAC inhibitor induces both apoptosis (2-fold) and cell cycle arrest. In conclusion, the HDAC 2/6 inhibitor acts by suppressing invasion via upregulating E-cadherin mediated by HDAC 2 blockade and by inducing cell cycle arrest leading to apoptosis via HDAC 6 inhibition. These results suggest that the HDAC 2/6 inhibitor might represent a novel therapeutic strategy for the treatment of PDAC tumorigenesis and metastasis.
    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
    组蛋白脱乙酰酶(HDAC)在各种恶性肿瘤的病理生物学中起着关键的分子调节剂的作用,并作为治疗干预的可行靶标引起了人们的关注。已经开发了多种HDAC抑制剂(HDACis)来靶向HDAC。许多临床前研究最终证明了HDACis的抗肿瘤作用,无论是作为单一疗法还是联合治疗。在此基础上,研究人员进行了各种临床研究,以评估选择性和泛HDACis在临床环境中的潜力.在我们的工作中,我们广泛总结和组织了当前的临床试验,全面概述了目前靶向HDAC治疗的临床进展。此外,我们讨论了几个没有产生积极结果的临床试验,分析导致其缺乏预期治疗效果的因素。除了实验设计因素,毒理学副作用等问题,肿瘤异质性,意外的脱靶效应也导致了这些低于预期的结果。这些挑战自然成为HDACis应用的重大障碍。尽管面临这些挑战,我们相信,HDACi研究的进展和联合疗法的改进将为实体瘤治疗铺平道路或带来广阔而充满希望的未来.
    Histone deacetylase (HDAC) serves as a critical molecular regulator in the pathobiology of various malignancies and have garnered attention as a viable target for therapeutic intervention. A variety of HDAC inhibitors (HDACis) have been developed to target HDACs. Many preclinical studies have conclusively demonstrated the antitumor effects of HDACis, whether used as monotherapy or in combination treatments. On this basis, researchers have conducted various clinical studies to evaluate the potential of selective and pan-HDACis in clinical settings. In our work, we extensively summarized and organized current clinical trials, providing a comprehensive overview of the current clinical advancements in targeting HDAC therapy. Furthermore, we engaged in discussions about several clinical trials that did not yield positive outcomes, analyzing the factors that led to their lack of anticipated therapeutic effectiveness. Apart from the experimental design factors, issues such as toxicological side effects, tumor heterogeneity, and unexpected off-target effects also contributed to these less-than-expected results. These challenges have naturally become significant barriers to the application of HDACis. Despite these challenges, we believe that advancements in HDACi research and improvements in combination therapies will pave the way or lead to a broad and hopeful future in the treatment of solid tumors.
    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:神经性疼痛仍然是现代医学面临的巨大挑战。一线药物治疗表现出有限的疗效和不利的副作用,突出了对有效治疗药物的未满足需求。在过去的几十年中,将表观遗传概念转化为疼痛治疗的努力激增,并揭示了表观遗传学作为疼痛研究的有希望的途径。最近,组蛋白脱乙酰酶(HDAC)的异常活性已成为促进神经性疼痛发展和维持的关键机制。
    目的:在这篇综述中,我们强调了特定HDAC亚型在疼痛伤害感受中以细胞特异性方式的独特作用,并概述了最近的实验证据支持HDACi在神经性疼痛中的治疗潜力。
    方法:我们总结了HDAC在Pubmed神经病理性疼痛中的研究。
    结果:HDAC,广泛分布在背根神经节和脊髓的神经元和非神经元细胞中,通过组蛋白或非组蛋白的去乙酰化调节基因表达,并参与增加的神经元兴奋性和神经炎症,从而促进外周和中枢致敏。重要的是,在神经性疼痛的各种临床前模型中,使用HDAC靶向抑制剂(HDACi)对异常乙酰化的药理学操作显示出有希望的疼痛缓解特性.然而,其中许多表现出低特异性,可能诱导脱靶毒性,强调在疼痛管理中开发同工型选择性HDACi的必要性。
    结论:异常升高的HDAC通过表观遗传调节神经元和免疫细胞中关键基因的表达促进神经元兴奋性和神经炎症,在神经性疼痛的进展中有助于外周和中枢敏化,和HDACi显示出显著的疗效和缓解神经性疼痛的巨大潜力。
    Neuropathic pain remains a formidable challenge for modern medicine. The first-line pharmacological therapies exhibit limited efficacy and unfavorable side effect profiles, highlighting an unmet need for effective therapeutic medications. The past decades have witnessed an explosion in efforts to translate epigenetic concepts into pain therapy and shed light on epigenetics as a promising avenue for pain research. Recently, the aberrant activity of histone deacetylase (HDAC) has emerged as a key mechanism contributing to the development and maintenance of neuropathic pain.
    In this review, we highlight the distinctive role of specific HDAC subtypes in a cell-specific manner in pain nociception, and outline the recent experimental evidence supporting the therapeutic potential of HDACi in neuropathic pain.
    We have summarized studies of HDAC in neuropathic pain in Pubmed.
    HDACs, widely distributed in the neuronal and non-neuronal cells of the dorsal root ganglion and spinal cord, regulate gene expression by deacetylation of histone or non-histone proteins and involving in increased neuronal excitability and neuroinflammation, thus promoting peripheral and central sensitization. Importantly, pharmacological manipulation of aberrant acetylation using HDAC-targeted inhibitors (HDACi) has shown promising pain-relieving properties in various preclinical models of neuropathic pain. Yet, many of which exhibit low-specificity that may induce off-target toxicities, underscoring the necessity for the development of isoform-selective HDACi in pain management.
    Abnormally elevated HDACs promote neuronal excitability and neuroinflammation by epigenetically modulating pivotal gene expression in neuronal and immune cells, contributing to peripheral and central sensitization in the progression of neuropathic pain, and HDACi showed significant efficacy and great potential for alleviating neuropathic pain.
    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
    异常的表观遗传修饰是各种癌症发病机理的基本因素。因此,用小分子靶向这些畸变,如组蛋白去乙酰化酶(HDAC)抑制剂和DNA甲基转移酶(DNMT)抑制剂,提出了一种可行的癌症治疗策略。这项研究的目的是评估曲古抑菌素C(TSC)的抗癌功效,曲古抑菌素A的类似物,来自链霉菌属的发酵。CPCC203909。我们的研究表明,TSC显示出对人类肺癌和膀胱尿路上皮癌细胞系的有效活性,IC50值在低微摩尔范围内。此外,TSC诱导半胱天冬酶3/7介导的细胞凋亡,并将细胞周期阻滞在G2/M期。当与DNMT抑制剂地西他滨联合使用时,TSC表现出协同抗癌作用。此外,蛋白质分析阐明了酪氨酸激酶受体Axl表达的显著降低。值得注意的是,TSC浓度升高与转录因子叉头框类O1(FoxO1)的上调以及促凋亡蛋白Bim和p21的水平升高相关。总之,我们的研究结果表明TSC是一种有前景的具有HDAC抑制活性的抗癌药物.此外,我们的研究结果凸显了TSC与DNMT抑制剂联合用于癌症治疗的潜在效用.
    Aberrant epigenetic modifications are fundamental contributors to the pathogenesis of various cancers. Consequently, targeting these aberrations with small molecules, such as histone deacetylase (HDAC) inhibitors and DNA methyltransferase (DNMT) inhibitors, presents a viable strategy for cancer therapy. The objective of this study is to assess the anti-cancer efficacy of trichostatin C (TSC), an analogue of trichostatin A sourced from the fermentation of Streptomyces sp. CPCC 203909. Our investigations reveal that TSC demonstrates potent activity against both human lung cancer and urothelial bladder cancer cell lines, with IC50 values in the low micromolar range. Moreover, TSC induces apoptosis mediated by caspase 3/7 and arrests the cell cycle at the G2/M phase. When combined with the DNMT inhibitor decitabine, TSC exhibits a synergistic anti-cancer effect. Additionally, protein analysis elucidates a significant reduction in the expression of the tyrosine kinase receptor Axl. Notably, elevated concentrations of TSC correlate with the up-regulation of the transcription factor forkhead box class O1 (FoxO1) and increased levels of the proapoptotic proteins Bim and p21. In conclusion, our findings suggest TSC as a promising anti-cancer agent with HDAC inhibitory activity. Furthermore, our results highlight the potential utility of TSC in combination with DNMT inhibitors for cancer 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
    蛋白质错误折叠和错误定位是神经退行性疾病的常见主题,包括运动神经元疾病,肌萎缩侧索硬化(ALS)。维持蛋白质稳定是一个交叉的治疗目标,包括上调热休克蛋白(HSP)以增加伴侣能力。运动神经元对上调应激诱导的HSPA1A有很高的阈值,但组成性表达高水平的HSPA8。这项研究比较了这些HSP在培养的运动神经元中的表达,这些运动神经元表达与家族性ALS相关的三种变体:TDP-43G348C,FUSR521G或SOD1G93A。所有变体都是Hspa1a的不良诱导剂,Hspa8mRNA和蛋白水平降低,表明陪伴能力有多重妥协。为了促进HSP表达,用推定的HSP共诱导剂处理培养物,Arimoclomol,I类组蛋白去乙酰化酶(HDAC)抑制剂促进活性染色质转录,和组合。治疗有变化,通常对Hspa1a和Hspa8的表达有不同的影响,取决于所表达的ALS变体,mRNA分布(躯体和树突),和测量的毒性生物标志物(组蛋白乙酰化,维持核TDP-43和nBAF染色质重塑复合物组分Brg1,线粒体运输,FUS聚合)。总的来说,单独的HDAC抑制比阿利莫罗更有效。和FUS模型一样,在TDP-43模型中,Arimoclomol未能诱导HSPA1A或保留Hspa8mRNA,尽管保留了核TDP-43和Brg1,表明除HSP诱导外的神经保护特性。数据说明了针对ALS发病机理的多种生物标志物的药物机制的复杂性,以及HSPA8对躯体和树突中神经元蛋白稳定的重要性。
    Protein misfolding and mislocalization are common themes in neurodegenerative disorders, including motor neuron disease, and amyotrophic lateral sclerosis (ALS). Maintaining proteostasis is a crosscutting therapeutic target, including the upregulation of heat shock proteins (HSP) to increase chaperoning capacity. Motor neurons have a high threshold for upregulating stress-inducible HSPA1A, but constitutively express high levels of HSPA8. This study compared the expression of these HSPs in cultured motor neurons expressing three variants linked to familial ALS: TAR DNA binding protein 43 kDa (TDP-43)G348C, fused in sarcoma (FUS)R521G, or superoxide dismutase I (SOD1)G93A. All variants were poor inducers of Hspa1a, and reduced levels of Hspa8 mRNA and protein, indicating multiple compromises in chaperoning capacity. To promote HSP expression, cultures were treated with the putative HSP coinducer, arimoclomol, and class I histone deacetylase inhibitors, to promote active chromatin for transcription, and with the combination. Treatments had variable, often different effects on the expression of Hspa1a and Hspa8, depending on the ALS variant expressed, mRNA distribution (somata and dendrites), and biomarker of toxicity measured (histone acetylation, maintaining nuclear TDP-43 and the neuronal Brm/Brg-associated factor chromatin remodeling complex component Brg1, mitochondrial transport, FUS aggregation). Overall, histone deacetylase inhibition alone was effective on more measures than arimoclomol. As in the FUS model, arimoclomol failed to induce HSPA1A or preserve Hspa8 mRNA in the TDP-43 model, despite preserving nuclear TDP-43 and Brg1, indicating neuroprotective properties other than HSP induction. The data speak to the complexity of drug mechanisms against multiple biomarkers of ALS pathogenesis, as well as to the importance of HSPA8 for neuronal proteostasis in both somata and dendrites.
    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
    目前神经胶质瘤的诊断和治疗方法具有阻碍生存结果的局限性。我们建议波谱磁共振成像作为标准MRI的辅助手段来弥合这些差距。光谱MRI是一种体积MRI技术,能够基于其升高的胆碱(Cho)和降低的N-乙酰天冬氨酸(NAA)来识别肿瘤浸润。我们提出了Cho/NAA≥5x阈值的光谱成像的临床可翻译性,用于描绘诊断为非增强型神经胶质瘤的患者的活检目标。然后,我们描述了代谢物成像检测到的未治疗肿瘤与接受belinostat和放化疗的新诊断GBM患者的总生存期(OS)之间的关系.使用放疗前Cho/NAA≥2x与治疗的T1加权对比增强(T1w-CE)体积之间的中位数差异,将每个队列(对照组和belinostat)分为亚组。我们使用Kaplan-Meier估计器来计算每个亚组的中位OS。当Cho/NAA≥2x和T1w-CE体积之间的差异高于中位数时,中位OS为14.4个月,而当差异低于中位数时,中位OS为34.3个月。两个亚组的T1w-CE体积相似。我们发现,通过光谱学检测到的未治疗肿瘤体积较低的患者具有更好的生存结果。
    Current diagnostic and therapeutic approaches for gliomas have limitations hindering survival outcomes. We propose spectroscopic magnetic resonance imaging as an adjunct to standard MRI to bridge these gaps. Spectroscopic MRI is a volumetric MRI technique capable of identifying tumor infiltration based on its elevated choline (Cho) and decreased N-acetylaspartate (NAA). We present the clinical translatability of spectroscopic imaging with a Cho/NAA ≥ 5x threshold for delineating a biopsy target in a patient diagnosed with non-enhancing glioma. Then, we describe the relationship between the undertreated tumor detected with metabolite imaging and overall survival (OS) from a pilot study of newly diagnosed GBM patients treated with belinostat and chemoradiation. Each cohort (control and belinostat) were split into subgroups using the median difference between pre-radiotherapy Cho/NAA ≥ 2x and the treated T1-weighted contrast-enhanced (T1w-CE) volume. We used the Kaplan-Meier estimator to calculate median OS for each subgroup. The median OS was 14.4 months when the difference between Cho/NAA ≥ 2x and T1w-CE volumes was higher than the median compared with 34.3 months when this difference was lower than the median. The T1w-CE volumes were similar in both subgroups. We find that patients who had lower volumes of undertreated tumors detected via spectroscopy had better survival outcomes.
    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:三阴性乳腺癌(TNBC)是一种侵袭性乳腺癌亚型,预后不良。阿霉素是TNBC标准治疗的一部分,但化疗耐药仍然是一个重要的临床挑战。Bocodepsin(OKI-179)是一种小分子I类组蛋白脱乙酰酶(HDAC)抑制剂,可促进TNBC临床前模型中的细胞凋亡。本研究的目的是研究临床前TNBC模型中博考地辛和阿霉素的组合,并评估其对终末细胞命运的影响。包括细胞凋亡和衰老。
    方法:TNBC细胞系用阿霉素处理,CellTiter-Glo用于评估增殖并确定阿霉素敏感性。用OKI-005(体外版本的博科地辛)和阿霉素处理选择的细胞系,并评估其增殖情况。通过膜联蛋白V/PI测量的细胞凋亡,和细胞周期通过流式细胞术。免疫印迹用于评估细胞凋亡介质的变化,细胞周期停滞,和衰老。通过衰老相关的β-半乳糖苷酶测定来测量衰老。MDA-MB-231异种移植体内模型用bocodepsin处理,阿霉素,或组合并评估肿瘤生长的抑制作用。在CAL-51细胞系中进行p53的shRNA敲低,评估细胞凋亡和衰老对联合治疗的反应。
    结果:OKI-005和多柔比星在TNBC细胞系中产生协同抗增殖活性,无论p53突变状态如何。该组合导致细胞凋亡增加和衰老减少。在体内,与任一单一药物相比,该组合导致肿瘤生长抑制增加.p53的shRNA敲低导致阿霉素诱导的衰老增加,其在体外添加OKI-005时降低。
    结论:在多柔比星中添加博可地辛导致体外协同抗增殖活性,改善体内肿瘤生长抑制,和促进细胞凋亡,使其成为克服TNBC中阿霉素抗性的有希望的组合。Bocodepsin目前处于临床开发中,与其他HDAC抑制剂相比,具有良好的毒性特征,支持在TNBC患者中评估这种组合的可行性。
    Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype with a poor prognosis. Doxorubicin is part of standard curative therapy for TNBC, but chemotherapy resistance remains an important clinical challenge. Bocodepsin (OKI-179) is a small molecule class I histone deacetylase (HDAC) inhibitor that promotes apoptosis in TNBC preclinical models. The purpose of this study was to investigate the combination of bocodepsin and doxorubicin in preclinical TNBC models and evaluate the impact on terminal cell fate, including apoptosis and senescence.
    TNBC cell lines were treated with doxorubicin and CellTiter-Glo was used to assess proliferation and determine doxorubicin sensitivity. Select cell lines were treated with OKI-005 (in vitro version of bocodepsin) and doxorubicin and assessed for proliferation, apoptosis as measured by Annexin V/PI, and cell cycle by flow cytometry. Immunoblotting was used to assess changes in mediators of apoptosis, cell cycle arrest, and senescence. Senescence was measured by the senescence-associated β-galactosidase assay. An MDA-MB-231 xenograft in vivo model was treated with bocodepsin, doxorubicin, or the combination and assessed for inhibition of tumor growth. shRNA knockdown of p53 was performed in the CAL-51 cell line and proliferation, apoptosis and senescence were assessed in response to combination treatment.
    OKI-005 and doxorubicin resulted in synergistic antiproliferative activity in TNBC cells lines regardless of p53 mutation status. The combination led to increased apoptosis and decreased senescence. In vivo, the combination resulted in increased tumor growth inhibition compared to either single agent. shRNA knock-down of p53 led to increased doxorubicin-induced senescence that was decreased with the addition of OKI-005 in vitro.
    The addition of bocodepsin to doxorubicin resulted in synergistic antiproliferative activity in vitro, improved tumor growth inhibition in vivo, and promotion of apoptosis which makes this a promising combination to overcome doxorubicin resistance in TNBC. Bocodepsin is currently in clinical development and has a favorable toxicity profile compared to other HDAC inhibitors supporting the feasibility of evaluating this combination 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