targeted protein degradation (TPD)

  • 文章类型: Journal Article
    组蛋白去乙酰化酶6(HDAC6)在各种癌症的发生和发展中起着至关重要的作用。因为它的过度表达与肿瘤生长有关,入侵,迁移,生存,凋亡,和血管生成。因此,在过去的十年中,HDAC6已成为抗癌药物发现的有吸引力的靶标。然而,常规HDAC6抑制剂的开发因其有限的临床疗效而受到阻碍,获得性抵抗力,并且不能抑制HDAC6的非酶功能。为了克服这些挑战,新战略,如双重作用抑制剂,靶向蛋白质降解(TPD)技术(包括PROTACs,HyT),对增强HDAC6抑制剂的抗癌活性至关重要。在这次审查中,我们专注于HDAC6调制器的设计和开发的最新进展,包括同工型选择性HDAC6抑制剂,基于HDAC6的双靶点抑制剂,和靶向蛋白质降解剂(PROTACs,HyT),从合理设计的角度来看,药效学,药代动力学,和临床状态。最后,我们讨论了基于HDAC6的癌症治疗药物发现的挑战和未来方向.
    Histone deacetylase 6 (HDAC6) plays a crucial role in the initiation and progression of various cancers, as its overexpression is linked to tumor growth, invasion, migration, survival, apoptosis, and angiogenesis. Therefore, HDAC6 has emerged as an attractive target for anticancer drug discovery in the past decade. However, the development of conventional HDAC6 inhibitors has been hampered by their limited clinical efficacy, acquired resistance, and inability to inhibit non-enzymatic functions of HDAC6. To overcome these challenges, new strategies, such as dual-acting inhibitors, targeted protein degradation (TPD) technologies (including PROTACs, HyT), are essential to enhance the anticancer activity of HDAC6 inhibitors. In this review, we focus on the recent advances in the design and development of HDAC6 modulators, including isoform-selective HDAC6 inhibitors, HDAC6-based dual-target inhibitors, and targeted protein degraders (PROTACs, HyT), from the perspectives of rational design, pharmacodynamics, pharmacokinetics, and clinical status. Finally, we discuss the challenges and future directions for HDAC6-based drug discovery for cancer therapy.
    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
    SMARCA2和SMARCA4是SWI/SNF复合物的亚基,SWI/SNF复合物是染色质重塑复合物,是促进基因表达的关键表观遗传调节因子。SMARCA4功能缺失突变的肿瘤依赖于SMARCA2细胞存活,这种合成致死性是治疗癌症的潜在治疗策略。
    当前的评论集中在声称与SMARCA2的溴结构域位点结合的蛋白水解靶向嵌合体(PROTAC)降解物的专利申请中,并在2019年1月至2023年6月之间发布。共评估了9个不同申请人的29个申请。
    SMARCA2/4溴结构域抑制剂不会对癌症增殖产生预期效果;然而,公司已经将溴结构域结合剂转化为PROTACs来降解蛋白质,与SMARCA4相比,更喜欢SMARCA2。在缺乏SMARCA4的情况下,SMARCA2的选择性降解很可能是有效的,同时允许正常组织有足够的安全裕度。随着最近披露的几项专利申请,瞄准SMARCA2的兴趣应该继续,特别是现在在临床上使用PreludeTherapeutics的选择性SMARCA2PROTAC。临床试验的结果将影响选择性SMARCA2PROTACs开发的演变。
    UNASSIGNED: SMARCA2 and SMARCA4 are subunits of the SWI/SNF complex which is a chromatin remodeling complex and a key epigenetic regulator that facilitates gene expression. Tumors with loss of function mutations in SMARCA4 rely on SMARCA2 for cell survival and this synthetic lethality is a potential therapeutic strategy to treat cancer.
    UNASSIGNED: The current review focuses on patent applications that claim proteolysis-targeting chimeras (PROTAC) degraders that bind the bromodomain site of SMARCA2 and are published between January 2019-June 2023. A total of 29 applications from 9 different applicants were evaluated.
    UNASSIGNED: SMARCA2/4 bromodomain inhibitors do not lead to desired effects on cancer proliferation; however, companies have converted bromodomain binders into PROTACs to degrade the protein, with a preference for SMARCA2 over SMARCA4. Selective degradation of SMARCA2 is most likely required to be efficacious in the SMARCA4-deficient setting, while allowing for sufficient safety margin in normal tissues. With several patent applications disclosed recently, interest in targeting SMARCA2 should continue, especially with a selective SMARCA2 PROTAC now in the clinic from Prelude Therapeutics. The outcome of the clinical trials will influence the evolution of selective SMARCA2 PROTACs 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
    蛋白水解靶向嵌合体(PROTACs)是靶向目的蛋白(POI)以通过细胞泛素化机制降解的必需双功能分子。尽管在理解PROTACs的功能方面取得了重大进展,他们的治疗潜力在很大程度上仍未开发。由于高度灵活的成功,可扩展,和低成本的mRNA治疗,以及第一代肽PROTACs(p-PROTACs)的优点,我们首次提出了工程化mRNAPROTACs(m-PROTACs)策略。该设计结合了vonHippel-Lindau(VHL)募集肽编码mRNA和POI结合肽编码mRNA以形成m-PROTAC并促进细胞POI降解。然后,我们使用两个针对两种癌症相关蛋白的m-PROTACs进行概念验证实验,雌激素受体α和B细胞淋巴瘤-超大蛋白。我们的结果表明,m-PROTACs可以成功地降解不同细胞系中的POI,并且比传统的p-PROTACs更有效地抑制细胞增殖。此外,体内实验表明,m-PROTAC在4T1小鼠异种移植模型中导致明显的肿瘤消退。这一发现凸显了m-PROTAC作为靶向蛋白质降解疗法的有希望的方法的巨大潜力。
    Proteolysis-targeting chimeras (PROTACs) are essential bifunctional molecules that target proteins of interest (POIs) for degradation by cellular ubiquitination machinery. Despite significant progress made in understanding PROTACs\' functions, their therapeutic potential remains largely untapped. As a result of the success of highly flexible, scalable, and low-cost mRNA therapies, as well as the advantages of the first generation of peptide PROTACs (p-PROTACs), we present for the first time an engineering mRNA PROTACs (m-PROTACs) strategy. This design combines von Hippel-Lindau (VHL) recruiting peptide encoding mRNA and POI-binding peptide encoding mRNA to form m-PROTAC and promote cellular POI degradation. We then performed proof-of-concept experiments using two m-PROTACs targeting two cancer-related proteins, estrogen receptor alpha and B-cell lymphoma-extra large protein. Our results demonstrated that m-PROTACs could successfully degrade the POIs in different cell lines and more effectively inhibit cell proliferation than the traditional p-PROTACs. Moreover, the in vivo experiment demonstrated that m-PROTAC led to significant tumor regression in the 4T1 mouse xenograft model. This finding highlights the enormous potential of m-PROTAC as a promising approach for targeted protein degradation therapy.
    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
    组蛋白甲基转移酶(HMTs)在基因翻译后调控和多种生理过程中发挥重要作用。并与许多人类疾病有关,尤其是癌症。越来越多的证据表明,HMT可以作为癌症治疗的潜在治疗靶点。因此,在过去的十年中,HMTs抑制剂的开发一直受到越来越多的关注.然而,临床疗效不足等缺点,适度的选择性,和获得性抗性的倾向阻碍了常规HMT抑制剂的发展。新的技术和方法对于增强HMT抑制剂的抗癌活性至关重要。在这次审查中,我们首先回顾了几种重要的HMT的结构和生物学功能,如EZH2、G9a、PRMT5和DOT1L。阐述了这些HMTs与癌症的内在联系。接下来,我们主要关注包括双靶点抑制剂在内的HMT调节剂开发的最新进展,从合理设计等角度出发的靶向蛋白质降解剂和共价抑制剂,药效学,药代动力学,和临床状态。最后,我们还讨论了基于HMT的癌症治疗药物发现的挑战和未来方向.
    Histone methyltransferases (HMTs) play a critical role in gene post-translational regulation and diverse physiological processes, and are implicated in a plethora of human diseases, especially cancer. Increasing evidences demonstrate that HMTs may serve as a potential therapeutic target for cancer treatment. Thus, the development of HMTs inhibitor have been pursued with steadily increasing interest over the past decade. However, the disadvantages such as insufficient clinical efficacy, moderate selectivity, and propensity for acquired resistance have hindered the development of conventional HMT inhibitors. New technologies and methods are imperative to enhance the anticancer activity of HMT inhibitors. In this review, we first review the structure and biological functions of the several essential HMTs, such as EZH2, G9a, PRMT5, and DOT1L. The internal relationship between these HMTs and cancer is also expounded. Next, we mainly focus on the latest progress in the development of HMT modulators encompassing dual-target inhibitors, targeted protein degraders and covalent inhibitors from perspectives such as rational design, pharmacodynamics, pharmacokinetics, and clinical status. Lastly, we also discuss the challenges and future directions for HMT-based drug discovery for cancer therapy.
    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Arg/N-degron途径中,单个N末端(Nt)残基充当N-degrons,由含UBR盒的N-识别蛋白识别,可诱导底物泛素化和蛋白酶体降解。最近的研究发现了自噬Arg/N-degron通路,其中自噬受体p62/SQSTM1/Sequestosome-1充当N-识别素,将Nt-Arg和其他去稳定残基作为N-degrons结合。在绑定到Nt-Arg时,P62经历与其货物相关的自聚合,加速p62-货物复合物向自噬体的巨大自噬递送,导致溶酶体水解酶降解。这种自噬机制正在成为调节从蛋白质聚集体和亚细胞细胞器到入侵病原体的各种生物材料的溶酶体降解的重要途径。开发了生理N-degrons的化学模拟物,以在与神经变性和其他相关疾病相关的病理生理过程中发挥治疗作用。这里,我们描述了监测p62活性的方法,p62具有N-识别素和自噬受体的双重作用。该主题包括自聚合(用于货物冷凝),它与LC3在自噬膜上的相互作用(用于货物靶向),和溶酶体水解酶对p62货物复合物的降解。我们还讨论了在生物和病理生理过程中调节p62依赖性巨自噬的N-degrons小分子模拟物的开发和使用。
    In the Arg/N-degron pathway, single N-terminal (Nt) residues function as N-degrons recognized by UBR box-containing N-recognins that induce substrate ubiquitination and proteasomal degradation. Recent studies led to the discovery of the autophagic Arg/N-degron pathway, in which the autophagic receptor p62/SQSTM1/Sequestosome-1 acts as an N-recognin that binds the Nt-Arg and other destabilizing residues as N-degrons. Upon binding to Nt-Arg, p62 undergoes self-polymerization associated with its cargoes, accelerating the macroautophagic delivery of p62-cargo complexes to autophagosomes leading to degradation by lysosomal hydrolases. This autophagic mechanism is emerging as an important pathway that modulates the lysosomal degradation of various biomaterial ranging from protein aggregates and subcellular organelles to invading pathogens. Chemical mimics of the physiological N-degrons were developed to exert therapeutic efficacy in pathophysiological processes associated with neurodegeneration and other related diseases. Here, we describe the methods to monitor the activities of p62 in a dual role as an N-recognin and an autophagic receptor. The topic includes self-polymerization (for cargo condensation), its interaction with LC3 on autophagic membranes (for cargo targeting), and the degradation of p62-cargo complexes by lysosomal hydrolases. We also discuss the development and use of small molecule mimics of N-degrons that modulate p62-dependent macroautophagy in biological and pathophysiological processes.
    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
    蛋白质溶解-调整嵌合体(PROTACs)允许泛素-蛋白酶体系统(UPS)选择性降解感兴趣的蛋白质(POI)。有了这种独特的作用机制,针对断点簇区Abelson(BCR-ABL)酪氨酸激酶(TK,因为它们是治疗慢性粒细胞白血病(CML)的有前途的分子,主要的血液恶性肿瘤之一,这是由于BCR-ABL的组成型激活导致的不受控制的骨髓增殖。
    本综述总结了在Espacenet或世界知识产权组织等在线数据库中发布的有关促进BCR-ABL降解的PROTACs的专利/申请。专利将主要在化学结构方面进行描述,生化/药理活性,和潜在的临床应用。
    最近对PROTACs巨大潜力的发现导致产生了能够降解BCR-ABL的新化合物,用于治疗CML。虽然数量仍然减少,在发展的临床前阶段,一些化合物已经被证明可以克服常规BCR-ABL抑制剂带来的一些困难,例如著名的伊马替尼。因此,目前的一些PROTACs很有可能在未来几年进入未来的CML治疗.
    UNASSIGNED: PROteolysis-TArgeting Chimeras (PROTACs) allow the selective degradation of a protein of interest (POI) by the ubiquitin-proteasome system (UPS). With this unique mechanism of action, the research and development of PROTACs that target the Breakpoint Cluster Region Abelson (BCR-ABL) tyrosine kinase (TK) has been increasing dramatically, as they are promising molecules in the treatment of Chronic Myeloid Leukemia (CML), one of the main hematological malignancies, which results from an uncontrolled myeloproliferation due to the constitutive activation of BCR-ABL.
    UNASSIGNED: This review summarizes the patents/applications published in the online databases like Espacenet or World Intellectual Property Organization regarding PROTACs that promote BCR-ABL degradation. Patents will be described mostly in terms of chemical structure, biochemical/pharmacological activities, and potential clinical applications.
    UNASSIGNED: The recent discovery of the enormous potential of PROTACs led to the creation of new compounds capable of degrading BCR-ABL for the treatment of CML. Although still in reduced numbers, and in the pre-clinical phase of development, some compounds have already been shown to overcome some of the difficulties presented by conventional BCR-ABL inhibitors, such as the well-known imatinib. Therefore, it is very likely that some of the present PROTACs will enter future CML therapy in the coming years.
    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
    靶向蛋白降解(TPD)策略,例如蛋白水解靶向嵌合体(PROTACs)仅对细胞内蛋白降解起作用,因为它们涉及细胞内蛋白降解机制。近年来出现了几种用于细胞外和膜蛋白的TPD的新技术。尽管在细胞外和膜蛋白降解领域已经取得了一些进展,这些技术的应用仍处于起步阶段。在这次审查中,我们通过总结和回顾细胞外和膜蛋白(POI)降解的发现和障碍,调查了现有技术的治疗潜力。
    Targeted protein degradation (TPD) strategies, such as proteolysis-targeting chimeras (PROTACs) only work for intracellular protein degradation because they involve the intracellular protein degradation machinery. Several new technologies have emerged in recent years for TPD of extracellular and membrane proteins. Even though some progress has been demonstrated in the extracellular and membrane protein degradation field, the application of these technologies is still in its infancy. In this review, we survey the therapeutic potential of existing technologies by summarizing and reviewing discoveries and hurdles in extracellular and membrane protein-of-interest (POI) degradation.
    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
    在过去的二十年里,蛋白水解靶向嵌合体(PROTACs)在药物开发中具有革命性,使靶向蛋白降解(TPD)成为新兴的治疗方式。这些异双功能分子由三个单元组成:目的蛋白(POI)的配体,E3泛素连接酶的配体,和一个将两个图案绑在一起的接头。VonHippel-Lindau(VHL)是PROTACs开发中最广泛使用的E3连接酶之一,因为它在组织类型和特征明确的配体中普遍表达。接头组成和长度已被证明在确定POI-PROTAC-E3三元配合物的物理化学性质和空间取向中起重要作用,从而影响降解剂的生物活性。已经发表了许多文章和报告,展示了接头设计的药物化学方面。但很少有人关注将接头与E3连接酶配体连接的化学性质。在这次审查中,我们专注于目前在VHL招募PROTACs组装中采用的合成接头策略。我们的目标是涵盖一系列用于掺入不同长度的接头的基本化学物质,组成和功能。
    Over the last two decades, proteolysis targeting chimeras (PROTACs) have been revolutionary in drug development rendering targeted protein degradation (TPD) as an emerging therapeutic modality. These heterobifunctional molecules are comprised of three units: a ligand for the protein of interest (POI), a ligand for an E3 ubiquitin ligase, and a linker that tethers the two motifs together. Von Hippel-Lindau (VHL) is one of the most widely employed E3 ligases in PROTACs development due to its prevalent expression across tissue types and well-characterised ligands. Linker composition and length has proven to play an important role in determining the physicochemical properties and spatial orientation of the POI-PROTAC-E3 ternary complex, thus influencing the bioactivity of degraders. Numerous articles and reports have been published showcasing the medicinal chemistry aspects of the linker design, but few have focused on the chemistry around tethering linkers to E3 ligase ligands. In this review, we focus on the current synthetic linker strategies employed in the assembly of VHL-recruiting PROTACs. We aim to cover a range of fundamental chemistries used to incorporate linkers of varying length, composition and functionality.
    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
    蛋白水解靶向嵌合体(PROTACs)将感兴趣的蛋白质(POI)带入E3泛素连接酶的空间附近,促进POI泛素化和蛋白酶体降解。PROTACs依赖于内源性细胞机制来介导POI降解,因此,POI的亚细胞定位和对E3连接酶的获取可能会影响PROTAC的疗效.为了询问POI的亚细胞环境是否影响PROTAC介导的降解,我们表达了由不同定位信号组成的Halo或FKBP12F36V(dTAG)构建体,并测试了它们通过靶向Halo或dTAG的vonHippel-Lindau(VHL)-或cereblon(CRBN)-招募PROTACs降解的功效.POI定位于细胞核,细胞质,线粒体外膜,内质网,高尔基,过氧化物酶体或溶酶体。差异定位的Halo或FKBP12F36V蛋白使用相同的各自PROTACs表现出不同的降解水平,因此,表明POI的亚细胞环境可以影响PROTAC介导的POI降解的功效。
    Proteolysis-targeting chimeras (PROTACs) bring a protein of interest (POI) into spatial proximity of an E3 ubiquitin ligase, promoting POI ubiquitylation and proteasomal degradation. PROTACs rely on endogenous cellular machinery to mediate POI degradation, therefore the subcellular location of the POI and access to the E3 ligase being recruited potentially impacts PROTAC efficacy. To interrogate whether the subcellular context of the POI influences PROTAC-mediated degradation, we expressed either Halo or FKBP12F36V (dTAG) constructs consisting of varying localization signals and tested the efficacy of their degradation by von Hippel-Lindau (VHL)- or cereblon (CRBN)-recruiting PROTACs targeting either Halo or dTAG. POIs were localized to the nucleus, cytoplasm, outer mitochondrial membrane, endoplasmic reticulum, Golgi, peroxisome or lysosome. Differentially localized Halo or FKBP12F36V proteins displayed varying levels of degradation using the same respective PROTACs, suggesting therefore that the subcellular context of the POI can influence the efficacy of PROTAC-mediated POI degradation.
    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
    靶向蛋白质降解允许靶向不可药物的蛋白质用于治疗应用以及消除感兴趣的蛋白质用于研究目的。虽然已经开发了几种类型的利用蛋白酶体或溶酶体的降解剂,同时降解靶标和加速细胞自噬通量的技术仍然不可用。在这项研究中,我们开发了一种通用化学工具,通过该工具,将细胞内蛋白质靶向巨自噬以进行溶酶体降解。这个平台技术,被称为AUTOTAC(自动吞吐嵌合),使用由与自噬靶向配体(ATL)连接的靶结合配体(TBL)组成的双功能分子。通过TBL与靶标结合后,ATL结合其他休眠自噬受体SQSTM1/p62(螯合体1)的ZZ结构域,它激活与靶标相关的SQSTM1,并将它们隔离到寡聚物中以进行自噬靶向和溶酶体降解。AUTOTAC用于在体外和/或体内降解神经变性中的各种癌蛋白或聚集倾向蛋白。我们建议AUTOTAC作为研究工具和药物开发提供了选择性蛋白水解的平台。
    Targeted protein degradation allows targeting undruggable proteins for therapeutic applications as well as eliminating proteins of interest for research purposes. While several types of degraders that harness the proteasome or the lysosome have been developed, a technology that simultaneously degrades targets and accelerates cellular autophagic flux remains unavailable. In this study, we developed a general chemical tool by which given intracellular proteins are targeted to macroautophagy for lysosomal degradation. This platform technology, termed AUTOTAC (AUTOphagy-TArgeting Chimera), employs bifunctional molecules composed of target-binding ligands (TBLs) linked to autophagy-targeting ligands (ATLs). Upon binding to targets via the TBL, the ATL binds the ZZ domain of the otherwise dormant autophagy receptor SQSTM1/p62 (sequestosome 1), which activates SQSTM1 associated with targets and sequesters them into oligomeric species for autophagic targeting and lysosomal degradation. AUTOTACs were used to degrade various oncoproteins or aggregation-prone proteins in neurodegeneration both in vitro and/or in vivo. We suggest that AUTOTAC provides a platform for selective proteolysis as a research tool and in drug development.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号