Proteolysis-targeting chimera

蛋白水解靶向嵌合体
  • 文章类型: Journal Article
    PRC2的主要催化亚基EZH2的异常表达与许多癌症有关。包括白血病,乳房,还有前列腺.最近的研究强调了EZH2的非催化致癌功能,而EZH2催化抑制剂无法减弱。因此,已经探索了蛋白水解靶向嵌合体(PROTAC)降解剂作为抑制规范和非规范致癌活性的替代治疗方法。在这里,我们介绍MS8847,一部小说,高效的EZH2PROTAC降解剂,可招募E3连接酶vonHippel-Lindau(VHL)。MS8847降低EZH2浓度-,time-,和泛素-蛋白酶体系统(UPS)依赖性方式。值得注意的是,与先前发表的EZH2PROTAC降解剂相比,MS8847在MLL重排(MLL-r)急性髓细胞性白血病(AML)细胞中诱导优异的EZH2降解和抗增殖作用。此外,MS8847降解EZH2并抑制三阴性乳腺癌(TNBC)细胞系中的细胞生长,在3DTNBC体外模型中显示功效,并且具有适于体内功效研究的药代动力学(PK)概况。总的来说,MS8847是生物医学界研究EZH2的规范和非规范致癌功能的有价值的化学工具。
    Aberrant expression of EZH2, the main catalytic subunit of PRC2, has been implicated in numerous cancers, including leukemia, breast, and prostate. Recent studies have highlighted non-catalytic oncogenic functions of EZH2, which EZH2 catalytic inhibitors cannot attenuate. Therefore, proteolysis-targeting chimera (PROTAC) degraders have been explored as an alternative therapeutic approach to suppress both canonical and non-canonical oncogenic activity. Here we present MS8847, a novel, highly potent EZH2 PROTAC degrader that recruits the E3 ligase von Hippel-Lindau (VHL). MS8847 degrades EZH2 in a concentration-, time-, and ubiquitin-proteasome system (UPS)-dependent manner. Notably, MS8847 induces superior EZH2 degradation and anti-proliferative effects in MLL-rearranged (MLL-r) acute myeloid leukemia (AML) cells compared to previously published EZH2 PROTAC degraders. Moreover, MS8847 degrades EZH2 and inhibits cell growth in triple-negative breast cancer (TNBC) cell lines, displays efficacy in a 3D TNBC in vitro model, and has a pharmacokinetic (PK) profile suitable for in vivo efficacy studies. Overall, MS8847 is a valuable chemical tool for the biomedical community to investigate canonical and non-canonical oncogenic functions of EZH2.
    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
    像vemurafenib(VEM)这样的BRAF抑制剂(BRAFi)在突变的黑素瘤中提供初始消退,但快速发展抗性。负责对VEM产生抗性的分子途径最终收敛于致癌c-Myc的激活。我们确定了一种表观遗传学方法来抑制c-Myc表达并使BRAFi抗性黑色素瘤细胞重新敏感。ARV-825(ARV)被用作BRD4靶向的蛋白质溶解调整嵌合体,其选择性地降解BRD4以下调c-Myc。ARV在体外协同增强VEM的细胞毒性以克服其在黑色素瘤中的抗性。ARV和VEM负载的脂质纳米复合物(NANOVB)的开发显着改善了其口服给药的理化性质。最重要的是,在裸鼠中,口服NANOVB以41.07mm3/天的速率显著抑制肿瘤生长。NANOVB治疗导致延长的存活,50%的小鼠存活直到实验终点。组织病理学分析显示,体内肿瘤坏死明显,Ki-67和BRD4蛋白下调。NANOVB证明的有希望的体内抗肿瘤活性和延长的生存期表明其对BRAFi抗性黑色素瘤的临床转化潜力。
    BRAF inhibitors (BRAFi) like vemurafenib (VEM) provide initial regression in mutated melanoma but rapidly develop resistance. Molecular pathways responsible for development of resistance against VEM finally converge towards the activation of oncogenic c-Myc. We identified an epigenetic approach to inhibit the c-Myc expression and resensitize BRAFi-resistant melanoma cells. ARV-825 (ARV) was employed as a BRD4 targeted PROteolysis TArgeting Chimera that selectively degrades the BRD4 to downregulate c-Myc. ARV synergistically enhanced the cytotoxicity of VEM in vitro to overcome its resistance in melanoma. Development of ARV and VEM-loaded lipid nanocomplex (NANOVB) significantly improved their physicochemical properties for oral delivery. Most importantly, oral administration of NANOVB substantially inhibited tumor growth at rate of 41.07 mm3/day in nude athymic mice. NANOVB treatment resulted in prolonged survival with 50% of mice surviving until the experimental endpoint. Histopathological analysis revealed significant tumor necrosis and downregulation of Ki-67 and BRD4 protein in vivo. Promising in vivo antitumor activity and prolonged survival demonstrated by NANOVB signifies its clinical translational potential for BRAFi-resistant melanoma.
    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
    zeste同源物2(EZH2)和Bruton酪氨酸激酶(BTK)的增强子都是血液系统恶性肿瘤发生发展的关键因素。临床研究已经证明了各种EZH2抑制剂的潜力,靶向EZH2的甲基转移酶活性,用于治疗淋巴瘤。然而,尽管它们能够有效降低H3K27me3水平,这些抑制剂在阻断淋巴瘤细胞增殖方面显示出有限的功效。为了克服这一挑战,我们利用选择性EZH2降解剂MS1943采用疏水标记方法。在这项研究中,我们研究了两种药物的抑制作用,FDA批准的EZH2抑制剂Tazemetostat,目前正在进行临床试验,和新的药物MS1943,在伯基特淋巴瘤。此外,我们评估了这些药物与BTK抑制剂依鲁替尼联合使用的潜在协同作用.在这项研究中,我们评估了MS1943和Ibrutinib联合治疗对三种Burkitt淋巴瘤细胞系增殖的影响,即RPMI1788,拉莫斯,还有Daudi细胞.我们的结果表明,与Tazemetostat和Ibrutinib的组合相比,MS1943和Ibrutinib的组合在更大程度上显著抑制了细胞增殖。此外,我们研究了潜在的作用机制,发现MS1943和依鲁替尼的联合治疗导致miR29B介导的p53上调的凋亡调节剂PUMA,巴克斯,裂开的PARP,并在Burkitt淋巴瘤细胞中裂解caspase-3。这些发现凸显了这种创新治疗策略作为传统EZH2抑制剂替代品的潜力。为改善伯基特淋巴瘤的治疗结果提供了有希望的前景。
    Enhancer of zeste homolog 2 (EZH2) and Bruton\'s tyrosine kinase (BTK) are both key factors involved in the development and progression of hematological malignancies. Clinical studies have demonstrated the potential of various EZH2 inhibitors, which target the methyltransferase activity of EZH2, for the treatment of lymphomas. However, despite their ability to effectively reduce the H3K27me3 levels, these inhibitors have shown limited efficacy in blocking the proliferation of lymphoma cells. To overcome this challenge, we employed a hydrophobic tagging approach utilizing MS1943, a selective EZH2 degrader. In this study, we investigated the inhibitory effects of two drugs, the FDA-approved EZH2 inhibitor Tazemetostat, currently undergoing clinical trials, and the novel drug MS1943, on Burkitt\'s lymphoma. Furthermore, we assessed the potential synergistic effect of combining these drugs with the BTK inhibitor Ibrutinib. In this study, we evaluated the effects of combination therapy with MS1943 and Ibrutinib on the proliferation of three Burkitt\'s lymphoma cell lines, namely RPMI1788, Ramos, and Daudi cells. Our results demonstrated that the combination of MS1943 and Ibrutinib significantly suppressed cell proliferation to a greater extent compared to the combination of Tazemetostat and Ibrutinib. Additionally, we investigated the underlying mechanisms of action and found that the combination therapy of MS1943 and Ibrutinib led to the upregulation of miR29B-mediated p53-upregulated modulator of apoptosis PUMA, BAX, cleaved PARP, and cleaved caspase-3 in Burkitt\'s lymphoma cells. These findings highlight the potential of this innovative therapeutic strategy as an alternative to traditional EZH2 inhibitors, offering promising prospects for improving treatment outcomes in Burkitt\'s lymphoma.
    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
    雄激素受体(AR)是配体(雄激素)激活的转录因子和核受体(NR)超家族的成员。它是男性性激素功能所必需的。AR-FL(全长)具有NRs的结构域结构,反式激活所需的N端结构域(NTD),DNA结合域(DBD),核定位信号(NLS)和配体结合域(LBD)。悖论存在于内源性配体睾酮(T)和5α-二氢睾酮(DHT)对男性性发育有不同的影响,同时与相同的受体结合并实现转录特异性,即使雄激素反应元件(AREs)与孕酮相同,糖皮质激素和盐皮质激素受体。通过低温EM或X射线晶体学的AR-FL的高分辨率3维结构在很大程度上由于NTD的内在无序而仍然难以捉摸。AR功能受翻译后修饰的调节,导致大量的蛋白形式。多蛋白复合物中的这些蛋白形式与通过域间偶联驱动的共激活剂和共阻遏物的相互作用介导AR转录输出。AR是具有合成代谢或雄激素作用的选择性雄激素受体调节剂(SARMS)的药物靶标。用雄激素剥夺疗法或通过使用与LBD结合的AR拮抗剂来治疗前列腺癌。由于适应性AR上调和缺乏LBD并变成组成型活性的剪接变体的出现而发生药物抗性。双极T治疗和NTD拮抗剂可以克服这些抗性机制,分别。描述了AR信令中的这些最新进展。
    The Androgen Receptor (AR) is a ligand (androgen) activated transcription factor and a member of the nuclear receptor (NR) superfamily. It is required for male sex hormone function. AR-FL (full-length) has the domain structure of NRs, an N-terminal domain (NTD) required for transactivation, a DNA-binding domain (DBD), a nuclear localization signal (NLS) and a ligand-binding domain (LBD). Paradoxes exist in that endogenous ligands testosterone (T) and 5α-dihydrotestosterone (DHT) have differential effects on male sexual development while binding to the same receptor and transcriptional specificity is achieved even though the androgen response elements (AREs) are identical to those seen for the progesterone, glucocorticoid and mineralocorticoid receptors. A high resolution 3-dimensional structure of AR-FL by either cryo-EM or X-ray crystallography has remained elusive largely due to the intrinsic disorder of the NTD. AR function is regulated by post-translational modification leading to a large number of proteoforms. The interaction of these proteoforms in multiprotein complexes with co-activators and co-repressors driven by interdomain coupling mediates the AR transcriptional output. The AR is a drug target for selective androgen receptor modulators (SARMS) that either have anabolic or androgenic effects. Protstate cancer is treated with androgen deprivation therapy or by the use of AR antagonists that bind to the LBD. Drug resistance occurs due to adaptive AR upregulation and the appearance of splice variants that lack the LBD and become constitutively active. Bipolar T treatment and NTD-antagonists could surmount these resistance mechanisms, respectively. These recent advances in AR signaling are described.
    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
    通过蛋白水解靶向嵌合体(PROTAC)诱导蛋白质降解为科学研究和工业应用提供了巨大的机会。自2017年首次报道其诱导SIRT2降解的能力以来,组蛋白脱乙酰酶(HDAC)-PROTAC已得到广泛开发。迄今为止,十八个HDACs中的十个(HDACs1-8、HDAC10和SIRT2)已被HDAC-PROTACs成功靶向和降解。HDAC-PROTACs在许多方面超越了传统的HDAC抑制剂,如更高的选择性,更有效的抗增殖活性,以及破坏多功能蛋白质的非酶依赖性功能并克服耐药性的能力。合理设计HDAC-PROTAC是开发中的主要挑战,因为化学结构的微小变化会对降解的效率和选择性产生巨大影响。在未来,HDAC-PROTACs可以潜在地参与临床研究,并支持增加的体内数据量,药代动力学评估,和药理学研究。
    Inducing protein degradation by proteolysis targeting chimera (PROTAC) has provided great opportunities for scientific research and industrial applications. Histone deacetylase (HDAC)-PROTAC has been widely developed since the first report of its ability to induce the degradation of SIRT2 in 2017. To date, ten of the eighteen HDACs (HDACs 1-8, HDAC10, and SIRT2) have been successfully targeted and degraded by HDAC-PROTACs. HDAC-PROTACs surpass traditional HDAC inhibitors in many aspects, such as higher selectivity, more potent antiproliferative activity, and the ability to disrupt the enzyme-independent functions of a multifunctional protein and overcome drug resistance. Rationally designing HDAC-PROTACs is a main challenge in development because slight variations in chemical structure can lead to drastic effects on the efficiency and selectivity of the degradation. In the future, HDAC-PROTACs can potentially be involved in clinical research with the support of the increased amount of in vivo data, pharmacokinetic evaluation, and pharmacological studies.
    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
    靶向蛋白质降解是用于新药设计和治疗的新兴且快速发展的技术。随着一类有前途的药物分子的出现,异双功能蛋白水解靶向嵌合体(PROTACs),TPD已成为使用传统小分子抑制剂完全解决致病蛋白的强大工具。然而,传统的PROTACs逐渐暴露了口服生物利用度和药代动力学(PK)和吸收差的潜在缺点,分布,新陈代谢,排泄,和毒性(ADMET)特性,因为它们比常规小分子抑制剂具有更大的分子量和更复杂的结构。因此,PROTAC概念提出20年后,越来越多的科学家致力于开发新的TPD技术来克服其缺陷。已经基于“PROTAC”探索了几种新技术和手段,以靶向“不可药用的蛋白质”。这里,我们旨在全面总结和深入分析基于PROTAC靶向降解“不可药用”靶标的靶向蛋白降解的研究进展。为了阐明基于PROTACs的新兴和高效策略在治疗各种疾病,特别是在克服癌症耐药性方面的重要性,我们将关注分子结构,作用机制,设计理念,这些新兴方法的发展优势和挑战(例如,适体-PROTAC缀合物,抗体-蛋白质和叶酸-蛋白质)。
    Targeted Protein Degradation is an emerging and rapidly developing technique for designing and treating new drugs. With the emergence of a promising class of pharmaceutical molecules, Heterobifunctional Proteolysis-targeting chimeras (PROTACs), TPD has become a powerful tool to completely tackle pathogenic proteins with traditional small molecule inhibitors. However, the conventional PROTACs have gradually exposed potential disadvantages of poor oral bioavailability and pharmacokinetic (PK) and absorption, distribution, metabolism, excretion, and toxicity (ADMET) characteristics due to their larger molecular weight and more complex structure than the conventional small-molecule inhibitors. Therefore, 20 years after the concept of PROTAC was proposed, more and more scientists are committed to developing new TPD technology to overcome its defects. And several new technologies and means have been explored based on \"PROTAC\" to target \"undruggable proteins\". Here, we aim to comprehensively summarize and profoundly analyze the research progress of targeted protein degradation based on PROTAC targeting the degradation of \"undruggable\" targets. In order to clarify the significance of emerging and highly effective strategies based PROTACs in the treatment of various diseases especially in overcoming drug resistance in cancer, we will focus on the molecular structure, action mechanism, design concepts, development advantages and challenges of these emerging methods(e.g., aptamer-PROTAC conjugates, antibody-PROTACs and folate-PROTACs).
    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
    靶向蛋白质降解作为一种新颖的治疗策略和生物医学研究中的有用工具,最近获得了广泛的兴趣。靶向的蛋白质降解物通常是亚化学计量的,并且不需要对其靶标的强结合亲和力。启用对以前无法访问的目标的访问。蛋白水解靶向嵌合体(PROTAC)是一类靶向蛋白降解剂,其通过经由异双功能分子将靶蛋白募集至E3-连接酶复合物来促进降解。PROTAC的模块化特性允许其合理设计和系统优化。在这里,我们建议为研究人员开发PROTAC降解剂的资源和方法,这可能是该领域的新手。©2022Wiley期刊有限责任公司。
    Targeted protein degradation has recently gained widespread interest as both a novel therapeutic strategy and a useful tool in biomedical research. Targeted protein degraders are often sub-stoichiometric and do not require strong binding affinity for their targets, enabling access to previously inaccessible targets. Proteolysis-targeting chimeras (PROTACs) are one class of targeted protein degraders that promote degradation by recruiting a target protein to an E3-ligase complex via a heterobifunctional molecule. The modular nature of PROTACs allows for their rational design and systematic optimization. Here we suggest resources and methodologies for developing PROTAC degraders for researchers that may be new to the field. © 2022 Wiley Periodicals LLC.
    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
    雄激素受体(AR)是AR阳性三阴性乳腺癌(TNBC)的有希望的治疗靶标。然而,AR抑制剂的临床试验仅显示AR阳性TNBC的适度治疗效果,耐药性也是不可避免的。为了应对这些挑战,我们在此报道了AR靶向蛋白水解靶向嵌合体(AR-PROTAC)治疗AR阳性TNBC的用途。我们证明AR-PROTAC通过泛素-蛋白酶体途径在AR阳性TNBCBT549细胞中有效降解AR蛋白,半降解浓度为46.9nM。通过评估体外和体内的治疗效果,我们验证了AR-PROTAC优于恩扎鲁他胺,AR抑制剂。具体来说,100nM的AR-PROTAC使BT549细胞活力降低高达80%,在皮下BT549肿瘤小鼠模型中,当肿瘤内给药时,10mg/kg的AR-PRTOAC抑制肿瘤生长~60%。总的来说,这些结果首次证明PROTAC有望增强AR阳性TNBC的治疗效果.
    Androgen receptor (AR) is a promising therapeutic target for AR-positive triple-negative breast cancer (TNBC). However, clinical trials of AR inhibitors only reveal modest therapeutic efficacy for AR-positive TNBC, and drug resistance is also inevitable. To address these challenges, we herein report the use of an AR-targeting proteolysis targeting chimera (AR-PROTAC) to treat AR-positive TNBC. We demonstrated that AR-PROTAC potently degraded AR protein via the ubiquitin-proteasome pathway in AR-positive TNBC BT549 cells, with a half degradation concentration of ∼46.9 nM. By evaluating the therapeutic efficacies in vitro and in vivo, we validated that AR-PROTAC was superior to enzalutamide, an AR inhibitor. Specifically, AR-PROTAC at 100 nM reduced BT549 cell viability by up to ∼80%, and AR-PRTOAC at 10 mg/kg suppressed tumor growth by ∼60% when administrated intratumorally in subcutaneous BT549 tumor mice model. Overall, these results demonstrate for the first time that PROTAC holds promise to enhance the treatment of AR-positive TNBC.
    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
    ARV-110,一种新型的蛋白水解靶向嵌合体(PROTAC),据报道,在I期临床试验中,前列腺癌治疗显示出令人满意的安全性和耐受性。然而,缺乏生物样品中ARV-110测定的生物分析方法。在这项研究中,我们开发并验证了用于大鼠和小鼠血浆中ARV-110定量的LC-MS/MS方法,并将其应用于药代动力学研究。使用蛋白质沉淀法从血浆样品中提取ARV-110和泊马度胺(内标)。使用C18柱和在蒸馏水中的0.1%甲酸-在乙腈中的0.1%甲酸(30:70,v/v)的流动相进行样品分离。多反应监测用于量化ARV-110和泊马度胺,其离子跃迁分别为m/z813.4→452.2和273.8→201.0。所开发的方法在2-3000ng/mL的浓度范围内显示出良好的线性,具有可接受的准确性。精度,基体效应,过程效率,和恢复。ARV-110在大鼠和小鼠血浆中长期保存稳定,三个冻融循环,在自动进样器中,但在室温和37°C下不稳定。此外,通过大鼠的1期代谢消除ARV-110,鼠标,和人肝微粒体被证明是不可能的。所开发的方法在药代动力学研究中的应用表明,ARV-110在大鼠和小鼠中的口服生物利用度中等(23.83%和37.89%,分别)。这些药代动力学发现对于ARV-110和/或其他PROTACs的未来临床前和临床研究是有益的。
    ARV-110, a novel proteolysis-targeting chimera (PROTAC), has been reported to show satisfactory safety and tolerability for prostate cancer therapy in phase I clinical trials. However, there is a lack of bioanalytical assays for ARV-110 determination in biological samples. In this study, we developed and validated an LC-MS/MS method for the quantitation of ARV-110 in rat and mouse plasma and applied it to pharmacokinetic studies. ARV-110 and pomalidomide (internal standard) were extracted from the plasma samples using the protein precipitation method. Sample separation was performed using a C18 column and a mobile phase of 0.1% formic acid in distilled water-0.1% formic acid in acetonitrile (30:70, v/v). Multiple reaction monitoring was used to quantify ARV-110 and pomalidomide with ion transitions at m/z 813.4 → 452.2 and 273.8 → 201.0, respectively. The developed method showed good linearity in the concentration range of 2-3000 ng/mL with acceptable accuracy, precision, matrix effect, process efficiency, and recovery. ARV-110 was stable in rat and mouse plasma under long-term storage, three freeze-thaw cycles, and in an autosampler, but unstable at room temperature and 37 °C. Furthermore, the elimination of ARV-110 via phase 1 metabolism in rat, mouse, and human hepatic microsomes was shown to be unlikely. Application of the developed method to pharmacokinetic studies revealed that the oral bioavailability of ARV-110 in rats and mice was moderate (23.83% and 37.89%, respectively). These pharmacokinetic findings are beneficial for future preclinical and clinical studies of ARV-110 and/or other PROTACs.
    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
    溴结构域和末端外结构域(BET)蛋白由四个哺乳动物成员(BRD2,BRD3,BRD4和BRDT)组成,在炎症反应的转录调节中起关键作用。炎症失调是通过多种机制在各种中枢神经系统疾病中的关键病理过程。包括NF-κB和Nrf2通路,两个著名的炎症主调节器。更好地理解BET蛋白在调节炎症过程中的作用具有重要意义,因为它可以揭示新的治疗靶点,以减少与许多CNS疾病相关的神经炎症。在这篇小型评论中,我们首先概述了BET蛋白的结构特征,并总结了BET抑制的遗传和药理学方法,包括使用蛋白水解靶向嵌合体(PROTACs)的新策略。我们强调了体外和体内BET蛋白与NF-κB和Nrf2信号通路之间相互作用的证据。最后,我们总结了最近的研究表明,BET蛋白是各种CNS疾病中炎症和神经病理学的重要调节因子。
    Bromodomain and extra-terminal domain (BET) proteins consist of four mammalian members (BRD2, BRD3, BRD4, and BRDT), which play a pivotal role in the transcriptional regulation of the inflammatory response. Dysregulated inflammation is a key pathological process in various CNS disorders through multiple mechanisms, including NF-κB and Nrf2 pathways, two well-known master regulators of inflammation. A better mechanistic understanding of the BET proteins\' role in regulating the inflammatory process is of great significance since it could reveal novel therapeutic targets to reduce neuroinflammation associated with many CNS diseases. In this minireview, we first outline the structural features of BET proteins and summarize genetic and pharmacological approaches for BET inhibition, including novel strategies using proteolysis-targeting chimeras (PROTACs). We emphasize in vitro and in vivo evidence of the interplay between BET proteins and NF-κB and Nrf2 signaling pathways. Finally, we summarize recent studies showing that BET proteins are essential regulators of inflammation and neuropathology in various CNS diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号