target protein degradation

靶蛋白降解
  • 文章类型: Journal Article
    本文探讨了调节蛋白质稳定性以实现靶蛋白降解的各种方法,这是研究生物过程和药物设计的一个重要方面。自利用N端规则引入热诱导的Degron细胞以来,已经过去了30年,以及使用泛素-蛋白酶体系统控制蛋白质稳定性的方法已经从学术界转移到了工业界。这篇综述涵盖了蛋白质稳定性控制方法,从早期到最近的进步,并讨论了该领域技术的发展。这篇综述还通过追踪从蛋白质稳定性控制方法开始到现在的发展,解决了蛋白质稳定性控制技术的挑战和未来方向。
    This review explores various methods for modulating protein stability to achieve target protein degradation, which is a crucial aspect in the study of biological processes and drug design. Thirty years have passed since the introduction of heat-inducible degron cells utilizing the N-end rule, and methods for controlling protein stability using the ubiquitin-proteasome system have moved from academia to industry. This review covers protein stability control methods, from the early days to recent advancements, and discusses the evolution of techniques in this field. This review also addresses the challenges and future directions of protein stability control techniques by tracing their development from the inception of protein stability control methods to the present day.
    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
    含有Src同源性2结构域的磷酸酶2(SHP2)由于其在肿瘤和免疫细胞中的多方面作用而成为癌症治疗的有吸引力的靶标。在这里,我们设计并合成了一系列新型的蛋白水解靶向嵌合体(PROTACs),使用SHP2变构抑制剂作为弹头,目的是在细胞内和体内实现SHP2降解。在这些分子中,化合物P9以浓度和时间依赖性方式诱导SHP2的有效降解(DC50=35.2±1.5nM)。机制研究表明,P9介导的SHP2降解需要E3连接酶的募集,并且是泛素化和蛋白酶体依赖性的。P9显示在许多癌细胞系中的抗肿瘤活性优于其亲本变构抑制剂。重要的是,在异种移植小鼠模型中,P9的施用导致几乎完全的肿瘤消退,作为肿瘤中SHP2强耗竭和磷酸化ERK1/2抑制的结果。因此,P9代表具有优异体内功效的第一SHP2PROTAC分子。预计P9不仅可以用作询问SHP2生物学的新化学工具,而且可以用作开发靶向SHP2的新疗法的起点。
    Src homology 2 domain-containing phosphatase 2 (SHP2) is an attractive target for cancer therapy due to its multifaceted roles in both tumor and immune cells. Herein, we designed and synthesized a novel series of proteolysis targeting chimeras (PROTACs) using a SHP2 allosteric inhibitor as warhead, with the goal of achieving SHP2 degradation both inside the cell and in vivo. Among these molecules, compound P9 induces efficient degradation of SHP2 (DC50 = 35.2 ± 1.5 nM) in a concentration- and time-dependent manner. Mechanistic investigation illustrates that the P9-mediated SHP2 degradation requires the recruitment of the E3 ligase and is ubiquitination- and proteasome-dependent. P9 shows improved anti-tumor activity in a number of cancer cell lines over its parent allosteric inhibitor. Importantly, administration of P9 leads to a nearly complete tumor regression in a xenograft mouse model, as a result of robust SHP2 depletion and suppression of phospho-ERK1/2 in the tumor. Hence, P9 represents the first SHP2 PROTAC molecule with excellent in vivo efficacy. It is anticipated that P9 could serve not only as a new chemical tool to interrogate SHP2 biology but also as a starting point for the development of novel therapeutics targeting SHP2.
    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KRAS这样的“不可用”目标在药物开发中尤其具有挑战性。我们设计了一种新颖的化学击倒策略,CANDDY(具有亲和力的化学击倒Nd降解动力学)技术,其使用与从蛋白酶体抑制剂修饰的降解标签(CANDDY标签)缀合的小分子(CANDDY分子)促进蛋白质降解。我们证明了CANDDY标签允许直接蛋白酶体靶标降解而不依赖于泛素化。我们合成了一种KRAS降解CANDDY分子,TUS-007,其在KRAS突变体(G12D和G12V)和野生型KRAS中诱导降解。我们证实了TUS-007在腹膜内给药的人结肠细胞皮下异种移植模型(KRASG12V)和口服给药的人胰腺细胞原位异种移植模型(KRASG12D)中的肿瘤抑制作用。因此,CANDDY技术有可能在治疗上靶向以前不可用的蛋白质,提供了一种更简单、更实用的药物靶向方法,并避免了E3酶与靶标匹配的困难。
    \"Undruggable\" targets such as KRAS are particularly challenging in the development of drugs. We devised a novel chemical knockdown strategy, CANDDY (Chemical knockdown with Affinity aNd Degradation DYnamics) technology, which promotes protein degradation using small molecules (CANDDY molecules) that are conjugated to a degradation tag (CANDDY tag) modified from proteasome inhibitors. We demonstrated that CANDDY tags allowed for direct proteasomal target degradation independent of ubiquitination. We synthesized a KRAS-degrading CANDDY molecule, TUS-007, which induced degradation in KRAS mutants (G12D and G12V) and wild-type KRAS. We confirmed the tumor suppression effect of TUS-007 in subcutaneous xenograft models of human colon cells (KRAS G12V) with intraperitoneal administrations and in orthotopic xenograft models of human pancreatic cells (KRAS G12D) with oral administrations. Thus, CANDDY technology has the potential to therapeutically target previously undruggable proteins, providing a simpler and more practical drug targeting approach and avoiding the difficulties in matchmaking between the E3 enzyme and the target.
    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(PROTACs)已被开发为靶向蛋白质降解的有效技术。每个PROTAC包含三个关键组件:感兴趣的蛋白质(POI)配体,E3连接酶配体,和一个链接。这些双功能分子可以劫持细胞内固有的泛素-蛋白酶体系统以降解不同的POI。与其他治疗策略相比有几个优点,近年来,PROTACs掀起了新的药物发现热潮。PRTOAC在全球范围内得到了广泛的探索,不仅在癌症疾病中而且在心血管疾病中都表现出色。脂肪肝,免疫性疾病,神经退行性疾病,和病毒感染。在这次审查中,我们旨在总结从2010年到2021年针对各种非癌蛋白的PROTACs的快速进展,并阐明PROTACs技术的优势。最后,还讨论了这一动态领域的潜在挑战。
    Proteolysis targeting chimeras (PROTACs) have been developed to be an effective technology for targeted protein degradation. Each PROTAC contains three key components: a protein-of-interest (POI) ligand, an E3 ligase ligand, and a linker. These bifunctional molecules can hijack the intracellular inherent ubiquitin-proteasome system to degrade different POIs. With several advantages over other therapeutic strategies, PROTACs have set off a new upsurge of drug discovery in recent years. PRTOACs have been extensively explored worldwide and have excelled not only in cancer diseases but also in cardiovascular diseases, fatty liver disease, immune diseases, neurodegenerative diseases, and viral infections. In this review, we aim to summarize the rapid progress from 2010 to 2021 in PROTACs targeting various non-oncoproteins and elucidate the advantages of PROTACs technology. Finally, the potential challenges of this dynamic field are also discussed.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    蛋白水解靶向嵌合体(PROTAC),劫持目标蛋白(POI)并通过泛素-蛋白酶体途径招募E3连接酶进行靶标降解,作为具有潜在临床应用价值的生物工具和药用分子,是一种新型的药物发现范式。目前,ARV-110是一种口服小分子PROTAC,旨在特异性靶向雄激素受体(AR),首先进入治疗转移性去势耐药前列腺癌的临床I期试验,这为PROTAC的发展开辟了一条新途径。我们在此提供对PROTAC靶向各种蛋白质的最新一年进展的详细总结,并阐明PROTAC技术的优势。最后,还讨论了这个充满活力的领域的潜在挑战。
    Proteolysis targeting chimera (PROTAC), hijacking protein of interest (POI) and recruiting E3 ligase for target degradation via the ubiquitin-proteasome pathway, is a novel drug discovery paradigm which has been widely used as biological tools and medicinal molecules with the potential of clinical application value. Currently, ARV-110, an orally small molecule PROTAC was designed to specifically target Androgen receptor (AR), firstly enters clinical phase I trials for the treatment of metastatic castration-resistant prostate cancer, which turns a new avenue for the development of PROTAC. We herein provide a detail summary on the latest one year progress of PROTAC target various proteins and elucidate the advantages of PROTAC technology. Finally, the potential challenges of this vibrant field are also discussed.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    Anaplastic lymphoma kinase (ALK) is a major target in treating non-small-cell lung cancer, and several ALK inhibitors have been developed to antagonize its kinase activity. However, patients treated with inhibitors ultimately develop drug resistance. Therefore, therapies with new mechanisms of action are needed. Proteolysis targeting chimeras (PROTACs) are molecules that comprise a ligand for binding a protein of interest (POI), a connecting linker and a ligand for recruiting E3 ligase, and cause degradation of the target POI. Here, the first multi-headed PROTAC, as a proof of concept, is developed as a gold nanoparticle (GNP)-based drug delivery system for delivering PROTACs to target ALK. Pegylated GNPs loaded with both ceritinib and pomalidomide molecules, termed Cer/Pom-PEG@GNPs, showed good stability in several media. The GNP conjugates potently decreased the levels of ALK fusion proteins in a dose- and time-dependent manner, and specifically inhibited the proliferation of NCI-H2228 cells. In comparison with small molecule PROTACs, the new multi-headed PROTAC promoted the formation of coacervates of POIs/multi-headed PROTAC/E3 ubiquitin ligases, and POI and E3 ubiquitin ligase interacted through multidirectional ligands and a flexible linker, thereby avoiding the need for complicated structure optimization of PROTACs. In conclusion, Cer/Pom-PEG@GNPs can degrade intracellular ALK fusion proteins with minor off-target toxicity and can be applied in patients resistant to ALK inhibitors. As a nano-based drug carrier, Cer/Pom-PEG@GNPs have the potential to enable prolonged circulation and specifically distribute drugs to tumor regions in vivo; thus, further investigation is warranted.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

  • 文章类型: Journal Article
    A new series of Proteolysis Targeting Chimeras (PROTACs) targeting Bruton\'s Tyrosine Kinase (BTK) was synthesized, with the goal of improving the pharmacokinetic properties of our previously reported PROTAC, MT802. We recently described the ability of MT802 to induce degradation of both wild-type and C481S mutant BTK in immortalized cells and patient-derived B-lymphocytes. However, the pharmacokinetic properties of MT802 were not suitable for further in vivo development. Therefore, we undertook a systematic medicinal chemistry campaign to overcome this issue and made a series of PROTACs with structural modifications to the linker and E3-recruiting ligand; more specifically, the new PROTACs were synthesized with different von Hippel-Lindau (VHL) and cereblon (CRBN) ligands while keeping the BTK ligand and linker length constant. This approach resulted in an equally potent PROTAC, SJF620, with a significantly better pharmacokinetic profile than MT802. This compound may hold promise for further in vivo exploration of BTK degradation.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Sci-hub)

       PDF(Pubmed)

公众号