undruggable target

  • 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
    癌症治疗中的靶向治疗可以通过改变特定生物分子的组织暴露来提高体内功效并减少不良反应。然而,癌症中仍然有大量的靶蛋白仍然是不可用的,由于以下因素,包括(1)缺乏配体结合袋,(2)基于蛋白质-蛋白质相互作用(PPI)的功能,(3)蛋白家族成员之间具有高度特异性的保守活性位点,(4)三级对接结构的变异性。KRAS等不可用靶蛋白的现状,TP53,C-MYC,PTP,在这次审查中仔细介绍。一些新的技术和药物设计策略已经被用于克服这些不可药用的蛋白质,最经典和众所周知的技术是蛋白水解靶向嵌合体(PROTACs)。在这次审查中,包括靶向蛋白质降解在内的新药开发策略,针对PPI,瞄准内在无序的区域,以及靶向蛋白质-DNA结合被描述,我们还讨论了这些策略克服不可用药目标的潜力。此外,像Alpha-Fold这样的智能辅助技术可以帮助我们预测蛋白质结构,有利于药物开发。新靶点的发现和靶向药物的研发,尤其是那些不可摧毁的目标,仍然是一个巨大的挑战。新药开发战略,更好的提取过程,不破坏蛋白质-蛋白质相互作用,而更精确的人工智能技术可能会在克服这些无法对付的目标方面提供重大帮助。
    Targeted therapies in cancer treatment can improve in vivo efficacy and reduce adverse effects by altering the tissue exposure of specific biomolecules. However, there are still large number of target proteins in cancer are still undruggable, owing to the following factors including (1) lack of ligand-binding pockets, (2) function based on protein-protein interactions (PPIs), (3) the highly specific conserved active sites among protein family members, and (4) the variability of tertiary docking structures. The current status of undruggable targets proteins such as KRAS, TP53, C-MYC, PTP, are carefully introduced in this review. Some novel techniques and drug designing strategies have been applicated for overcoming these undruggable proteins, and the most classic and well-known technology is proteolysis targeting chimeras (PROTACs). In this review, the novel drug development strategies including targeting protein degradation, targeting PPI, targeting intrinsically disordered regions, as well as targeting protein-DNA binding are described, and we also discuss the potential of these strategies for overcoming the undruggable targets. Besides, intelligence-assisted technologies like Alpha-Fold help us a lot to predict the protein structure, which is beneficial for drug development. The discovery of new targets and the development of drugs targeting them, especially those undruggable targets, remain a huge challenge. New drug development strategies, better extraction processes that do not disrupt protein-protein interactions, and more precise artificial intelligence technologies may provide significant assistance in overcoming these undruggable targets.
    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
    MYC功能在癌症中的重要性是在1970年代后期发现的,当时确定了导致粒细胞白血病的禽逆转录病毒的序列。从那以后,40多年的不断研究强调了这种蛋白质在恶性转化中的重要性,尤其是血液病。的确,原癌基因的较高表达之间的一些最早的联系(如MYC),在伯基特淋巴瘤中进行了基因重排及其与癌症发展的关系,慢性粒细胞白血病和小鼠浆细胞瘤。多发性骨髓瘤(MM),特别是,是与MYC失调严格相关的浆细胞恶性肿瘤,这表明针对它的治疗策略将有益于治疗这种疾病。然而,瞄准MYC是-而且,不知何故,由于其独特的性质,仍然具有挑战性:缺乏明确的三维结构,核定位和缺乏可靶向的酶口袋。尽管有这些困难,然而,许多研究显示了直接或间接抑制MYC的潜在治疗效果.已经测试了不同的分子,事实上,在MM的背景下。在这次审查中,我们总结了不同化合物的现状,包括他们的临床试验结果,并建议继续努力确定,repurpose,重新设计或改进候选药物,将其与标准治疗相结合,以克服耐药性并更好地管理骨髓瘤治疗。
    The importance of MYC function in cancer was discovered in the late 1970s when the sequence of the avian retrovirus that causes myelocytic leukemia was identified. Since then, over 40 years of unceasing research have highlighted the significance of this protein in malignant transformation, especially in hematologic diseases. Indeed, some of the earliest connections among the higher expression of proto-oncogenes (such as MYC), genetic rearrangements and their relation to cancer development were made in Burkitt lymphoma, chronic myeloid leukemia and mouse plasmacytomas. Multiple myeloma (MM), in particular, is a plasma cell malignancy strictly associated with MYC deregulation, suggesting that therapeutic strategies against it would be beneficial in treating this disease. However, targeting MYC was - and, somehow, still is - challenging due to its unique properties: lack of defined three-dimensional structure, nuclear localization and absence of a targetable enzymatic pocket. Despite these difficulties, however, many studies have shown the potential therapeutic impact of direct or indirect MYC inhibition. Different molecules have been tested, in fact, in the context of MM. In this review, we summarize the current status of the different compounds, including the results of their clinical testing, and propose to continue with the efforts to identify, repurpose, redesign or improve drug candidates to combine them with standard of care therapies to overcome resistance and enable better management of myeloma treatment.
    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),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
    UNASSIGNED: Undruggable targets refer to clinically meaningful therapeutic targets that are \'difficult to drug\' or \'yet to be drugged\' via traditional approaches. Featuring characteristics of lacking defined ligand-binding pockets, non-catalytic protein-protein interaction functional modes and less-investigated 3D structures, these undruggable targets have been targeted with novel therapeutic entities developed with the progress of unconventional drug discovery approaches, such as targeted degradation molecules and display technologies.
    UNASSIGNED: This review first presents the concept of \'undruggable\' exemplified by RAS and other targets. Next, detailed strategies are illustrated in two aspects: innovation of therapeutic entities and development of unconventional drug discovery technologies. Finally, case studies covering typical undruggable targets (Bcl-2, p53, and RAS) are depicted to further demonstrate the feasibility of the strategies and entities above.
    UNASSIGNED: Targeting the undruggable expands the scope of therapeutically reachable targets. Consequently, it represents the drug discovery frontier. Biomedical studies are capable of dissecting disease mechanisms, thus broadening the list of undruggable targets. Encouraged by the recent approval of the KRAS inhibitor Sotorasib, we believe that merging multiple discovery approaches and exploiting various novel therapeutic entities would pave the way for dealing with more \'undruggable\' targets in the future.
    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
    Oncogenic RAS proteins, common oncogenic drivers in many human cancers, have been refractory to conventional small-molecule and macromolecule inhibitors due to their intracellular localization and the lack of druggable pockets. Here, we present a feasible strategy for designing RAS inhibitors that involves intracellular delivery of RAS-binding domain (RBD), a nanomolar-affinity specific ligand of RAS. Screening of 51 different combinations of RBD and cell-permeable peptides has identified Pen-cRaf-v1 as a cell-permeable pan-RAS inhibitor capable of targeting both G12C and non-G12C RAS mutants. Pen-cRaf-v1 crosses the cell membrane via endocytosis, competitively inhibits RAS-effector interaction, and thereby exerts anticancer activity against several KRAS-mutant cancer cell lines. Moreover, Pen-cRaf-v1 exhibits excellent activity comparable with a leading pan-RAS inhibitor (BI-2852), as well as high target specificity in transcriptome analysis and alanine mutation analysis. These findings demonstrate that specific inhibition of oncogenic RAS, and possibly treatment of RAS-mutant cancer, is feasible by intracellular delivery of RBD.
    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
    传统上,已经设计了针对人类疾病的基于小分子或抗体的疗法来抑制酶活性或竞争病理靶蛋白的配体结合位点。尽管它表现出了有效性,比如在癌症治疗中,这种方法通常受到反复出现的耐药性的限制。更重要的是,并非所有的分子靶标都是具有可药物作用的“热点”的酶或受体,它们可以被活性位点定向抑制剂直接占据。最近,创造了一个有希望的新范式,其中小分子化学物质利用泛素-蛋白酶体系统的天然蛋白质质量控制机制来特异性根除细胞中的致病蛋白质。这种“化学诱导的蛋白质降解”可能为靶向本质上不可药用的蛋白质提供了前所未有的机会,如结构支架和其他非酶分子,用于治疗目的。这篇综述的重点是调查开发E3指导的蛋白水解靶向嵌合体(PROTACs)和蛋白酶体上游或蛋白酶体上的去泛素化酶的小分子化学调节剂的最新进展。
    Traditionally, small-molecule or antibody-based therapies against human diseases have been designed to inhibit the enzymatic activity or compete for the ligand binding sites of pathological target proteins. Despite its demonstrated effectiveness, such as in cancer treatment, this approach is often limited by recurring drug resistance. More importantly, not all molecular targets are enzymes or receptors with druggable \'hot spots\' that can be directly occupied by active site-directed inhibitors. Recently, a promising new paradigm has been created, in which small-molecule chemicals harness the naturally occurring protein quality control machinery of the ubiquitin-proteasome system to specifically eradicate disease-causing proteins in cells. Such \'chemically induced protein degradation\' may provide unprecedented opportunities for targeting proteins that are inherently undruggable, such as structural scaffolds and other non-enzymatic molecules, for therapeutic purposes. This review focuses on surveying recent progress in developing E3-guided proteolysis-targeting chimeras (PROTACs) and small-molecule chemical modulators of deubiquitinating enzymes upstream of or on the proteasome.
    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