CRBN

CRBN
  • 文章类型: Journal Article
    乳腺癌是影响全球女性的最常见恶性肿瘤之一,强调迫切需要更有效和更具体的治疗方法。蛋白水解靶向嵌合体(PROTACs)已成为一种有希望的策略,通过选择性靶向癌蛋白进行降解来开发新的先导化合物。在这项研究中,我们设计的,合成并评估了基于CRBN的PROTAC,L055,其靶向CDK9。我们的发现表明,L055有效抑制增殖,诱导细胞周期停滞,并降低ERα阳性乳腺癌细胞的体外存活率。L055特异性结合CDK9,通过CRBN依赖性蛋白酶体途径促进其降解。此外,L055在裸小鼠中抑制源自T47D和MCF7细胞的类器官和肿瘤的生长。因此,L055代表ERα阳性乳腺癌和潜在其他恶性肿瘤的潜在新型治疗剂。
    Breast cancer is one of the most prevalent malignancies affecting women worldwide, underscoring the urgent need for more effective and specific treatments. Proteolysis-targeting chimeras (PROTACs) have emerged as a promising strategy to develop new lead compounds by selectively targeting oncoproteins for degradation. In this study, we designed, synthesized and evaluated a CRBN-based PROTAC, L055, which targets CDK9. Our findings demonstrate that L055 effectively inhibits the proliferation, induces cell cycle arrest, and decreases the survival of ERα-positive breast cancer cells in vitro. L055 specifically binds to CDK9, facilitating its degradation via the CRBN-dependent proteasomal pathway. Additionally, L055 suppressed the growth of organoids and tumors derived from T47D and MCF7 cells in nude mice. Thus, L055 represents a potential novel therapeutic agent for ERα-positive breast cancer and potentially other malignancies.
    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
    靶向蛋白质降解是通过劫持细胞内蛋白质清理机制选择性去除感兴趣的蛋白质。这个快速增长的领域目前在很大程度上依赖于使用E3连接酶Cereblon(CRBN)来靶向降解蛋白质,包括免疫调节药物(IMiDs)沙利度胺,来那度胺,和泊马度胺通过与CRBN作用的分子胶机制起作用。虽然CRBN募集可以导致特定目的蛋白的降解(例如功效),其他蛋白质(称为CRBN新底物)的降解也会发生。这些CRBN新底物中的一种或多种的降解被认为在兔和灵长类动物中观察到的沙利度胺相关的发育毒性中起重要作用。我们鉴定了一组25种与CRBN相关蛋白质稳态和/或胚胎/胎儿发育相关的感兴趣蛋白质。我们开发了一种针对这些蛋白质的靶向测定法,该测定法结合了肽免疫亲和富集和高分辨率质谱,并成功地将该测定法应用于服用三种IMiD的怀孕兔的兔胚胎样品。我们证实了先前报道的体内新底物如SALL4的减少,并提供了以前仅在体外检查的蛋白质的新底物变化的证据。虽然有许多蛋白质被所有三种IMiD类似地减少,没有化合物具有与另一种化合物完全相同的新底物降解谱。我们将我们的数据与以前的文献报道的IMiD诱导的降解和已知的发育生物学关联进行了比较。根据我们的观察,我们建议在开发测试系统中至少监测这些新底物的主要子集,以改善CRBN结合化合物特异性风险评估.我们分析的一个优势是它是可配置的,并且靶列表可以容易地调整以仅关注感兴趣的蛋白质的子集或随着发现关于CRBN生物学的额外信息而扩展以纳入新发现。
    Targeted protein degradation is the selective removal of a protein of interest through hijacking intracellular protein cleanup machinery. This rapidly growing field currently relies heavily on the use of the E3 ligase cereblon (CRBN) to target proteins for degradation, including the immunomodulatory drugs (IMiDs) thalidomide, lenalidomide, and pomalidomide which work through a molecular glue mechanism of action with CRBN. While CRBN recruitment can result in degradation of a specific protein of interest (e.g., efficacy), degradation of other proteins (called CRBN neosubstrates) also occurs. Degradation of one or more of these CRBN neosubstrates is believed to play an important role in thalidomide-related developmental toxicity observed in rabbits and primates. We identified a set of 25 proteins of interest associated with CRBN-related protein homeostasis and/or embryo/fetal development. We developed a targeted assay for these proteins combining peptide immunoaffinity enrichment and high-resolution mass spectrometry and successfully applied this assay to rabbit embryo samples from pregnant rabbits dosed with three IMiDs. We confirmed previously reported in vivo decreases in neosubstrates like SALL4, as well as provided evidence of neosubstrate changes for proteins only examined in vitro previously. While there were many proteins that were similarly decreased by all three IMiDs, no compound had the exact same neosubstrate degradation profile as another. We compared our data to previous literature reports of IMiD-induced degradation and known developmental biology associations. Based on our observations, we recommend monitoring at least a major subset of these neosubstrates in a developmental test system to improve CRBN-binding compound-specific risk assessment. A strength of our assay is that it is configurable, and the target list can be readily adapted to focus on only a subset of proteins of interest or expanded to incorporate new findings as additional information about CRBN biology is discovered.
    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
    GSPT1发挥着重要的生理功能,例如终止蛋白质翻译,在各种肿瘤中过表达。它是一个很有前途的抗肿瘤靶点,但也被认为是一种“不可食用”的蛋白质。最近的研究发现,一类小分子可以通过“分子胶”机制降解GSPT1,具有很强的抗肿瘤活性,有望成为血液系统恶性肿瘤的新疗法。目前可用的GSPT1降解剂主要来自免疫调节酰亚胺药物(IMiD)的支架,因此,具有新结构的更多活性化合物仍有待发现。在这项工作中,使用计算机辅助多轮虚拟筛选和生物测定,我们确定了一种非IMiD酰腙化合物,AN5782,可降低GPST1蛋白水平,明显抑制肿瘤细胞增殖。通过AN5782的亚结构搜索获得一些类似物。结构-活性关系分析揭示了这些化合物与CRBN-GSPT1之间可能的相互作用。进一步的生物学机制研究表明,AN5777通过泛素-蛋白酶体系统显著降低GSPT1,其有效细胞毒性是CRBN-和GSPT1依赖性的。此外,AN5777对U937和OCI-AML-2细胞表现出良好的抗增殖活性,并剂量依赖性地诱导G1期停滞和凋亡。这项工作中发现的结构可能是抗肿瘤药物开发的良好开端。
    GSPT1 plays crucial physiological functions, such as terminating protein translation, overexpressed in various tumors. It is a promising anti-tumor target, but is also considered as an \"undruggable\" protein. Recent studies have found that a class of small molecules can degrade GSPT1 through the \"molecular glue\" mechanism with strong antitumor activity, which is expected to become a new therapy for hematological malignancies. Currently available GSPT1 degraders are mostly derived from the scaffold of immunomodulatory imide drug (IMiD), thus more active compounds with novel structure remain to be found. In this work, using computer-assisted multi-round virtual screening and bioassay, we identified a non-IMiD acylhydrazone compound, AN5782, which can reduce the protein level of GPST1 and obviously inhibit the proliferation of tumor cells. Some analogs were obtained by a substructure search of AN5782. The structure-activity relationship analysis revealed possible interactions between these compounds and CRBN-GSPT1. Further biological mechanistic studies showed that AN5777 decreased GSPT1 remarkably through the ubiquitin-proteasome system, and its effective cytotoxicity was CRBN- and GSPT1-dependent. Furthermore, AN5777 displayed good antiproliferative activities against U937 and OCI-AML-2 cells, and dose-dependently induced G1 phase arrest and apoptosis. The structure found in this work could be good start for antitumor drug development.
    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
    自上次批准新型沙利度胺类似物以来的二十年中,许多有前途的候选药物已经出现了具有显著效力的靶向蛋白质降解剂。同样,PROTACs的出现抑制“不可药物的”蛋白质靶标加强了对具有改进的cereblon亲和力的新类似物的需求,靶标选择性和药物样特性。然而,沙利度胺及其批准的衍生物仍然受到几个缺点的困扰,如结构不稳定和溶解性差。在这里,我们对减轻这些缺点的策略进行了综述,并重点介绍了当代药物发现方法,这些方法产生了新型沙利度胺类似物,作为ceblon效应剂和/或抗癌剂具有增强的功效.
    In the two decades since a novel thalidomide analog was last approved, many promising drug candidates have emerged with remarkable potency as targeted protein degraders. Likewise, the advent of PROTACs for suppressing \'undruggable\' protein targets reinforces the need for new analogs with improved cereblon affinity, target selectivity and drug-like properties. However, thalidomide and its approved derivatives remain plagued by several shortcomings, such as structural instability and poor solubility. Herein, we present a review of strategies for mitigating these shortcomings and highlight contemporary drug discovery approaches that have generated novel thalidomide analogs with enhanced efficacy as cereblon effectors and/or anticancer agents.
    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(TPD)最近已成为一种令人兴奋的新药物形式。然而,在过去的二十年中,开发基于小分子的蛋白质降解剂的策略已经发展,现在已经建立了已经在临床使用的分子标签,以及嵌合分子,ProteesoldingTraftingChimeras(PROTACs),主要基于为两种E3连接酶CRBN和VHL开发的配体系统。人类E3连接酶家族的大尺寸表明,PROTACs可以通过靶向大量的E3连接酶来开发。其中一些具有限制的表达模式,具有设计疾病或组织特异性降解剂的潜力。的确,最近发表了许多新的E3配体,确认E3连接酶的可药用性。这篇综述总结了有关E3连接酶的最新数据,并强调了将这些分子开发成与已建立的降解剂系统相媲美的高效PROTACs所面临的挑战。
    Targeted protein degradation (TPD) has recently emerged as an exciting new drug modality. However, the strategy of developing small molecule-based protein degraders has evolved over the past two decades and has now established molecular tags that are already in clinical use, as well as chimeric molecules, PROteolysis TArgeting Chimeras (PROTACs), based mainly on ligand systems developed for the two E3 ligases CRBN and VHL. The large size of the human E3 ligase family suggests that PROTACs can be developed by targeting a large diversity of E3 ligases, some of which have restricted expression patterns with the potential to design disease- or tissue-specific degraders. Indeed, many new E3 ligands have been published recently, confirming the druggability of E3 ligases. This review summarises recent data on E3 ligases and highlights the challenges in developing these molecules into efficient PROTACs rivalling the established degrader systems.
    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
    分子胶是稳定蛋白质-蛋白质相互作用的小分子,启用新的分子药理学,如靶向蛋白质降解。它们提供优于蛋白水解靶向嵌合体(PROTACs)的优势,提出了与异双功能结构的大小和性质相关的挑战,但是胶水缺乏类似于PROTACs的合理设计原则。一个值得注意的例外是能够改变基于Cereblon(CRBN)的分子胶的结构并将其活性重定向到新的新底物蛋白。我们采取了一种专注的方法来修饰CRBN配体,5'-氨基来那度胺,通过并行化的硫(VI)-氟化物交换(SuFEx)转化,使用高通量化学多样化来改变其新底物特异性。我们使用这种方法合成了3,000多种5'-氨基来那度胺类似物,并使用表型筛选细胞活力筛选粗产物。鉴定数十种具有分化活性的类似物。我们表征了四种降解G-S相变1(GSPT1)蛋白的化合物,为基于SuFEx的CRBN分子胶的发现提供概念验证模型。
    Molecular glues are small molecules that stabilize protein-protein interactions, enabling new molecular pharmacologies, such as targeted protein degradation. They offer advantages over proteolysis targeting chimeras (PROTACs), which present challenges associated with the size and properties of heterobifunctional constructions, but glues lack the rational design principles analogous to PROTACs. One notable exception is the ability to alter the structure of Cereblon (CRBN)-based molecular glues and redirect their activity toward new neo-substrate proteins. We took a focused approach toward modifying the CRBN ligand, 5\'-amino lenalidomide, to alter its neo-substrate specificity using high-throughput chemical diversification by parallelized sulfur(VI)-fluoride exchange (SuFEx) transformations. We synthesized over 3,000 analogs of 5\'-amino lenalidomide using this approach and screened the crude products using a phenotypic screen for cell viability, identifying dozens of analogs with differentiated activity. We characterized four compounds that degrade G-to-S phase transition 1 (GSPT1) protein, providing a proof-of-concept model for SuFEx-based discovery of CRBN molecular glues.
    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
    CRBN是Cullin环E3泛素连接酶4(CRL4)复合物的底物受体。已经观察到CRBN可以被小分子利用以促进非天然CRL4底物的募集和泛素化。导致新底物通过泛素-蛋白酶体系统降解。这种现象,被称为分子胶诱导的蛋白质降解,与传统的小分子药物相比,已成为一种创新的治疗方法。分子胶的一个关键优势,与坚持Lipinski五法则的传统小分子药物相比,是它们在不需要靶蛋白上的特异性结合口袋的情况下操作的能力。这种独特的特性使分子胶能够与传统上难以处理的蛋白质靶标相互作用,如转录因子和支架蛋白。通过劫持泛素-蛋白酶体系统诱导这些先前难以捉摸的靶标降解的能力为治疗顽固性疾病提供了有希望的途径。然而,分子胶的合理设计仍然是一个巨大的挑战,由于对其机制和作用的理解有限。这篇综述概述了基于CRBN的分子胶领域的最新进展和突破,同时也探索了设计这些化合物的系统方法的前景。
    CRBN is a substrate receptor for the Cullin Ring E3 ubiquitin ligase 4 (CRL4) complex. It has been observed that CRBN can be exploited by small molecules to facilitate the recruitment and ubiquitination of non-natural CRL4 substrates, resulting in the degradation of neosubstrate through the ubiquitin-proteasome system. This phenomenon, known as molecular glue-induced protein degradation, has emerged as an innovative therapeutic approach in contrast to traditional small-molecule drugs. One key advantage of molecular glues, in comparison to conventional small-molecule drugs adhering to Lipinski\'s Rule of Five, is their ability to operate without the necessity for specific binding pockets on target proteins. This unique characteristic empowers molecular glues to interact with conventionally intractable protein targets, such as transcription factors and scaffold proteins. The ability to induce the degradation of these previously elusive targets by hijacking the ubiquitin-proteasome system presents a promising avenue for the treatment of recalcitrant diseases. Nevertheless, the rational design of molecular glues remains a formidable challenge due to the limited understanding of their mechanisms and actions. This review offers an overview of recent advances and breakthroughs in the field of CRBN-based molecular glues, while also exploring the prospects for a systematic approach to designing these compounds.
    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
    成人T细胞白血病/淋巴瘤(ATL)是与人T细胞白血病病毒1型(HTLV-1)感染相关的侵袭性T细胞瘤,预后极差。自2017年以来,来那度胺(LEN;第二代免疫调节药物[IMiD])已被用作ATL的额外治疗选择,但其作用机制尚未得到充分证明。最近的研究报告了对长期IMiD治疗的患者发生第二原发恶性肿瘤的担忧.我们在这项研究中的目的是阐明IMiD介导的抗ATL机制。将13个ATL相关细胞系分为LEN敏感或LEN抗性组。CRBN敲低(KD)导致抗性细胞中LEN功效和IKZF2-KD诱导的LEN功效的丧失。DNA微阵列分析显示LEN敏感性和LEN抗性ATL细胞系之间的LEN处理后明显的转录改变。对ATL细胞移植的严重联合免疫缺陷(SCID)小鼠口服LEN治疗也表明对肿瘤生长的明显抑制作用。最后,一种新型的cereblon调节剂(CELMoD),iberdomide(IBE),对ATL细胞表现出更广泛和更深入的生长抑制谱,有效的IKZF2降解,这在其他IMiD治疗中未观察到。基于这些发现,我们的研究强烈支持IBE对抗侵袭性和复发性ATL的新治疗优势.
    Adult T-cell leukemia/lymphoma (ATL) is an aggressive T-cell neoplasia associated with human T-cell leukemia virus type 1 (HTLV-1) infection and has an extremely poor prognosis. Lenalidomide (LEN; a second-generation immunomodulatory drug [IMiD]) has been employed as an additional therapeutic option for ATL since 2017, but its mechanism of action has not been fully proven, and recent studies reported emerging concerns about the development of second primary malignancies in patients treated with long-term IMiD therapy. Our purpose in this study was to elucidate the IMiD-mediated anti-ATL mechanisms. Thirteen ATL-related cell lines were divided into LEN-sensitive or LEN-resistant groups. CRBN knockdown (KD) led to a loss of LEN efficacy and IKZF2-KD-induced LEN efficacy in resistant cells. DNA microarray analysis demonstrated distinct transcriptional alteration after LEN treatment between LEN-sensitive and LEN-resistant ATL cell lines. Oral treatment of LEN for ATL cell-transplanted severe combined immunodeficiency (SCID) mice also indicated clear suppressive effects on tumor growth. Finally, a novel cereblon modulator (CELMoD), iberdomide (IBE), exhibited a broader and deeper spectrum of growth suppression to ATL cells with efficient IKZF2 degradation, which was not observed in other IMiD treatments. Based on these findings, our study strongly supports the novel therapeutic advantages of IBE against aggressive and relapsed ATL.
    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
    蛋白水解靶向嵌合体(PROTAC)技术通过直接靶向蛋白质并通过事件驱动的作用模式催化其降解而发挥作用,对多种疾病具有重要的临床应用前景。目前,最先进的PROTAC药物正在进行III期临床试验(NCT05654623).尽管PROTACs比传统的小分子抑制剂具有显著的优势,它们对正常细胞蛋白质的催化降解可能会引起毒副作用。因此,为了实现PROTACs的靶向释放并最大限度地减少不良反应,研究人员正在积极探索各种可控方案。在这次审查中,综合总结控制策略,为PROTAC技术的创新应用提供理论依据。
    Proteolysis-targeting chimaera (PROTAC) technology functions by directly targeting proteins and catalysing their degradation through an event-driven mode of action, a novel mechanism with significant clinical application prospects for various diseases. Currently, the most advanced PROTAC drug is undergoing phase III clinical trials (NCT05654623). Although PROTACs exhibit significant advantages over traditional small-molecule inhibitors, their catalytic degradation of normal cellular proteins can potentially cause toxic side effects. Therefore, to achieve targeted release of PROTACs and minimize adverse reactions, researchers are actively exploring diverse controllable PROTACs. In this review, we comprehensively summarize the control strategies to provide a theoretical basis for the innovative application of PROTAC technology.
    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
    沙利度胺,泊马度胺和来那度胺,统称为免疫调节酰亚胺药物(IMiDs),经常在蛋白水解靶向嵌合体(PROTACs)中用作cereblon(CRBN)E3连接酶募集配体。然而,它们的分子胶特性共同选择CRL4CRBN降解其非天然底物可能导致基于IMiD的PROTAC降解剂的不期望的脱靶效应。在这里,我们报道了一个小文库的有效和细胞可渗透的CRBN配体,通过基于结构的设计,对IMiD的众所周知的CRBN新衬底具有高选择性。它们被进一步用于构建含溴结构域的蛋白4(BRD4)降解物,这成功地耗尽了测试细胞中的BRD4。总的来说,我们报告了一系列功能化的CRBN招聘人员,这些招聘人员规避了传统IMiD的滥交,这项研究为许多其他治疗靶标的选择性CRBN招募PROTACs的开发提供了信息。
    Thalidomide, pomalidomide and lenalidomide, collectively referred to as immunomodulatory imide drugs (IMiDs), are frequently employed in proteolysis-targeting chimeras (PROTACs) as cereblon (CRBN) E3 ligase-recruiting ligands. However, their molecular glue properties that co-opt the CRL4CRBN to degrade its non-natural substrates may lead to undesired off-target effects for the IMiD-based PROTAC degraders. Herein, we reported a small library of potent and cell-permeable CRBN ligands, which exert high selectivity over the well-known CRBN neo-substrates of IMiDs by structure-based design. They were further utilized to construct bromodomain-containing protein 4 (BRD4) degraders, which successfully depleted BRD4 in the tested cells. Overall, we reported a series of functionalized CRBN recruiters that circumvent the promiscuity from traditional IMiDs, and this study is informative to the development of selective CRBN-recruiting PROTACs for many other therapeutic targets.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号