target protein degradation

靶蛋白降解
  • 文章类型: Journal Article
    c-ros癌基因1(ROS1),一个致癌司机,已知过度激活时会诱导非小细胞肺癌(NSCLC),特别是通过融合蛋白的形成。传统的靶向治疗侧重于用ROS1抑制剂抑制ROS1活性以控制癌症进展。然而,一种涉及蛋白质降解物设计的新策略通过完全降解ROS1融合癌蛋白提供了一种更有效的方法,从而有效地阻断它们的激酶活性并增强抗肿瘤潜力。利用PROteasoly-Togeting嵌合体(PROTAC)技术,通过分子对接和合理设计,我们报告了第一个ROS1特定的PROTAC,SIAIS039.这种降解剂有效地靶向工程Ba/F3细胞和HCC78细胞中的多种ROS1融合癌蛋白(CD74-ROS1,SDC4-ROS1和SLC34A2-ROS1),证明了对ROS1融合驱动的癌细胞的抗肿瘤作用。它抑制细胞增殖,诱导细胞周期停滞,和细胞凋亡,并抑制克隆性。SIAIS039的抗肿瘤功效超过了两种已批准的药物,克唑替尼和恩替替尼,和顶级抑制剂的匹配,包括洛拉替尼和他列替尼。机理研究证实,039诱导的降解需要ROS1配体和E3泛素连接酶的参与,涉及蛋白酶体和泛素化。此外,039在小鼠异种移植模型中表现出优异的口服生物利用度,突出其临床应用潜力。总之,我们的研究通过靶向ROS1融合癌蛋白降解,为ROS1融合阳性NSCLC提供了一种有希望的新治疗策略,为进一步发展PROTAC奠定基础,并为ROS1融合阳性NSCLC患者提供希望。
    The c-ros oncogene 1 (ROS1), an oncogenic driver, is known to induce non-small cell lung cancer (NSCLC) when overactivated, particularly through the formation of fusion proteins. Traditional targeted therapies focus on inhibiting ROS1 activity with ROS 1 inhibitors to manage cancer progression. However, a new strategy involving the design of protein degraders offers a more potent approach by completely degrading ROS1 fusion oncoproteins, thereby effectively blocking their kinase activity and enhancing anti-tumour potential. Utilizing PROteolysis-TArgeting Chimera (PROTAC) technology and informed by molecular docking and rational design, we report the first ROS1-specific PROTAC, SIAIS039. This degrader effectively targets multiple ROS1 fusion oncoproteins (CD74-ROS1, SDC4-ROS1 and SLC34A2-ROS1) in engineered Ba/F3 cells and HCC78 cells, demonstrating anti-tumour effects against ROS1 fusion-driven cancer cells. It suppresses cell proliferation, induces cell cycle arrest, and apoptosis, and inhibits clonogenicity. The anti-tumour efficacy of SIAIS039 surpasses two approved drugs, crizotinib and entrectinib, and matches that of the top inhibitors, including lorlatinib and taletrectinib. Mechanistic studies confirm that the degradation induced by 039 requires the participation of ROS1 ligands and E3 ubiquitin ligases, and involves the proteasome and ubiquitination. In addition, 039 exhibited excellent oral bioavailability in a mouse xenograft model, highlighting its potential for clinical application. In conclusion, our study presents a promising and novel therapeutic strategy for ROS1 fusion-positive NSCLC by targeting ROS1 fusion oncoproteins for degradation, laying the foundation for the development of further PROTAC and offering hope for patients with ROS1 fusion-positive NSCLC.
    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
    PROTACs(蛋白水解靶向嵌合体)已成为一类开创性的化学工具,可通过利用泛素-蛋白酶体系统(UPS)促进靶蛋白的降解。然而,在实现细胞选择性蛋白质降解和体内应用方面,PROTACs在化学生物学研究和治疗中的有效利用面临着重大挑战。这篇综述文章旨在阐明Pro-PROTACs开发的最新进展,其表现出响应于外部刺激或疾病相关的内源性生化信号的受控蛋白质降解能力。本文探讨了用于调节PROTACs与E3泛素连接酶或靶蛋白之间相互作用的特定化学策略。这些策略能够对Pro-PROTACs的蛋白质降解潜力进行空间和时间控制。此外,这篇综述总结了最近关于使用可生物降解的纳米颗粒进行体内应用和靶向蛋白质降解的PROTACs递送的研究。这样的递送系统对于在体内实现有效和选择性的蛋白质降解具有巨大的前景。最后,本文对多功能PROTACs的未来设计及其细胞内传递机制提供了一个观点,特别关注实现细胞选择性蛋白质降解。
    PROTACs (Proteolysis-Targeting Chimeras) have emerged as a groundbreaking class of chemical tools that facilitate the degradation of target proteins by leveraging the ubiquitin-proteasome system (UPS). However, the effective utilization of PROTACs in chemical biology studies and therapeutics encounters significant challenges when it comes to achieving cell-selective protein degradation and in vivo applications. This review article aims to shed light on recent advancements in the development of Pro-PROTACs, which exhibit controlled protein degradation capabilities in response to external stimuli or disease-related endogenous biochemical signals. The article delves into the specific chemical strategies employed to regulate the interaction between PROTACs and E3 ubiquitin ligases or target proteins. These strategies enable spatial and temporal control over the protein degradation potential of Pro-PROTACs. Furthermore, the review summarizes recent investigations regarding the delivery of PROTACs using biodegradable nanoparticles for in vivo applications and targeted protein degradation. Such delivery systems hold great promise for enabling efficient and selective protein degradation in vivo. Lastly, the article provides a perspective on the future design of multifunctional PROTACs and their intracellular delivery mechanisms, with a particular focus on achieving cell-selective protein 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
    以小分子化学药物和生物制剂为代表的传统药物的开发和应用,尤其是抑制剂,已成为药物开发的主流。近年来,靶向蛋白降解(TPD)技术已成为利用细胞自我破坏机制去除特定疾病相关蛋白的最有前途的方法之一。基于泛素-蛋白酶体系统(UPS)和自噬-溶酶体途径(ALP),正在出现许多不同的TPD策略。包括但不限于蛋白水解靶向嵌合体(PROTAC),分子胶(MG),溶酶体靶向嵌合体(LYTAC),伴侣介导的自噬(CMA)靶向嵌合体,自噬靶向嵌合体(AUTAC),自噬体系链化合物(ATTEC),和自噬靶向嵌合体(AUTOTAC)。靶向降解技术的出现可以改变人类细胞中的大多数蛋白质靶标,从不可药用到可药用,大大拓展了代谢综合征等难治性疾病的治疗前景。这里,我们总结了主要TPD技术的最新进展,特别是在代谢综合征中,并期待为药物发现提供新的见解。
    The development and application of traditional drugs represented by small molecule chemical drugs and biological agents, especially inhibitors, have become the mainstream drug development. In recent years, targeted protein degradation (TPD) technology has become one of the most promising methods to remove specific disease-related proteins using cell self-destruction mechanisms. Many different TPD strategies are emerging based on the ubiquitin-proteasome system (UPS) and the autophagy-lysosomal pathway (ALP), including but not limited to proteolysis-targeting chimeras (PROTAC), molecular glues (MG), lysosome targeting chimeras (LYTAC), chaperone-mediated autophagy (CMA)-targeting chimeras, autophagy-targeting chimera (AUTAC), autophagosome-tethering compound (ATTEC), and autophagy-targeting chimera (AUTOTAC). The advent of targeted degradation technology can change most protein targets in human cells from undruggable to druggable, greatly expanding the therapeutic prospect of refractory diseases such as metabolic syndrome. Here, we summarize the latest progress of major TPD technologies, especially in metabolic syndrome and look forward to providing new insights for drug discovery.
    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
    慢性粒细胞白血病(CML)是造血系统的恶性疾病,其致病蛋白BCR-ABL,严重危及患者生命.作为靶向药物的里程碑,伊马替尼在治疗CML方面取得了巨大成功。然而,由于BCR-ABL激酶的多个突变,临床上经常发生伊马替尼不可避免的耐药.随后,针对BCR-ABL的第二代酪氨酸激酶抑制剂(TKIs)被开发用于解决伊马替尼耐药的突变体,除了T315I.迄今为止,已经开发了针对T315I的第三代TKIs,用于提高选择性和安全性.值得注意的是,第一种变构抑制剂已经上市,可以有效克服ATP结合位点的突变.同时,一些先进的技术,例如基于不同E3配体的蛋白水解靶向嵌合体(PROTAC),高度期望通过选择性降解目标蛋白质来克服耐药性。在这次审查中,本文就目前针对BCR-ABL的抑制剂和降解剂治疗CML的研究进展作一综述。
    Chronic myeloid leukemia (CML) is a malignant disease of the hematopoietic system with crucial pathogenic protein named BCR-ABL, which endangers the life of patients severely. As a milestone of targeted drug, Imatinib has achieved great success in the treatment of CML. Nevertheless, inevitable drug resistance of Imatinib has occurred frequently in clinical due to the several mutations in the BCR-ABL kinase. Subsequently, the second-generation of tyrosine kinase inhibitors (TKIs) against BCR-ABL was developed to address the mutants of Imatinib resistance, except T315I. To date, the third-generation of TKIs targeting T315I has been developed for improving the selectivity and safety. Notably, the first allosteric inhibitor has been in market which could overcome the mutations in ATP binding site effectively. Meanwhile, some advanced technology, such as proteolysis-targeting chimeras (PROTAC) based on different E3 ligand, are highly expected to overcome the drug resistance by selectively degrading the targeted proteins. In this review, we summarized the current research progress of inhibitors and degraders targeting BCR-ABL for the treatment of CML.
    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),由感兴趣的蛋白质(POI)的配体组成的双功能分子,E3连接酶配体和接头,已开发用于劫持泛素-蛋白酶体系统(UPS)以诱导不同的POIs降解。目前,第一个口服PROTACs(ARV-110和ARV-471)在前列腺癌和乳腺癌治疗的临床试验中显示出令人鼓舞的疗效,这为PROTAC研究的发展开辟了一条新途径。在这次审查中,重点对PROTACs的最新进展进行了详细的总结,阐明了PROTACs技术的优势。此外,讨论了PRTOAC的潜在挑战和前景。
    Proteolysis-targeting chimeras (PROTACs), bifunctional molecules consisting of a ligand of protein of interest (POI), an E3 ligase ligand and a linker, have been developed to hijack the ubiquitin-proteasome system (UPS) to induce different POIs degradation. Currently, the first oral PROTACs (ARV-110 and ARV-471) have shown encouraging efficacy in clinical trials of prostate and breast cancer treatment, which turns a new avenue for the development of PROTAC research. In this review, we focus on a detailed summary of the latest progress of PROTACs and elucidate the advantages of PROTACs technology. In addition, potential challenges and perspectives of PRTOACs are discussed.
    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)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)

公众号