首页 > 最新文献

Drug Discovery Today: Technologies最新文献

英文 中文
Identification and characterization of cancer vulnerabilities via targeted protein degradation 通过靶向蛋白降解识别和表征癌症脆弱性
Q1 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2019-04-01 DOI: 10.1016/j.ddtec.2018.12.003
Cristina Mayor-Ruiz, Georg E. Winter

Target(ed) protein degradation (TPD) is a novel paradigm in drug discovery and a promising therapeutic strategy. TPD is based on small-molecules that catalyze the degradation of proteins by re-directing the ubiquitination activity of ubiquitin E3 ligases. Its unique molecular pharmacology enables robust, selective and fast elimination of proteins in cellular assays and in vivo. In addition to possible clinical applications, TPD is also emerging as an attractive alternative to traditional pharmacologic or genetic perturbation strategies. Directly acting degraders, as well as chemical-genetics derivatives offer unique opportunities in the pre-clinical identification, characterization and mechanistic validation of therapeutic targets.

靶蛋白降解(Target(ed) protein degradation, TPD)是一种新的药物发现模式,也是一种很有前景的治疗策略。TPD是基于通过重定向泛素E3连接酶的泛素化活性来催化蛋白质降解的小分子。其独特的分子药理学使其在细胞和体内检测中具有强大的选择性和快速消除蛋白质的能力。除了可能的临床应用之外,TPD也正在成为传统药理学或遗传扰动策略的一种有吸引力的替代方案。直接作用的降解剂以及化学遗传学衍生物为临床前鉴定、表征和治疗靶点的机制验证提供了独特的机会。
{"title":"Identification and characterization of cancer vulnerabilities via targeted protein degradation","authors":"Cristina Mayor-Ruiz,&nbsp;Georg E. Winter","doi":"10.1016/j.ddtec.2018.12.003","DOIUrl":"10.1016/j.ddtec.2018.12.003","url":null,"abstract":"<div><p><span>Target(ed) protein degradation<span><span> (TPD) is a novel paradigm in drug discovery<span> and a promising therapeutic strategy. TPD is based on small-molecules that catalyze the degradation of proteins by re-directing the ubiquitination activity of ubiquitin E3 </span></span>ligases<span>. Its unique molecular pharmacology enables robust, selective and fast elimination of proteins in cellular assays and </span></span></span><em>in vivo</em><span>. In addition to possible clinical applications, TPD is also emerging as an attractive alternative to traditional pharmacologic or genetic perturbation strategies. Directly acting degraders, as well as chemical-genetics derivatives offer unique opportunities in the pre-clinical identification, characterization and mechanistic validation of therapeutic targets.</span></p></div>","PeriodicalId":36012,"journal":{"name":"Drug Discovery Today: Technologies","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.ddtec.2018.12.003","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37336302","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 24
Ligand-induced genetic degradation as a tool for target validation 配体诱导的遗传降解作为靶标验证的工具
Q1 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2019-04-01 DOI: 10.1016/j.ddtec.2018.11.001
Aisha Yesbolatova , Yusuke Tominari , Masato T. Kanemaki

Targeted protein degraders, known as proteolysis targeting chimeras (PROTACs), are drawing more attention as next-generation drugs to target currently undruggable proteins. As drug discovery of functional degraders involves time- and cost-consuming laborious processes, we propose employing a ligand-induced genetic degradation system to validate candidate proteins before degrader development. Genetic degradation mimics degrader treatment by depleting a degron-fused protein in the presence of a defined ligand. All genetic systems use a combination of a degron and defined ligand that enables a protein of interest fused with the degron to be recruited to an E3 ubiquitin ligase for ubiquitylation and subsequent degradation by the proteasome. However, these events are based on different principles and have different features. We review the dTAG, HaloTag-based, auxin-inducible degron (AID), and destabilizing domain (DD) systems and discuss a strategy for degrader discovery against novel target proteins.

靶向蛋白质降解剂,即蛋白质水解靶向嵌合体(proteolysis targeting chimeras, PROTACs),作为靶向目前无法治疗的蛋白质的下一代药物,正受到越来越多的关注。由于功能降解物的药物发现涉及时间和成本的费力过程,我们建议在降解物开发之前采用配体诱导的遗传降解系统来验证候选蛋白质。遗传降解通过在确定配体存在的情况下耗尽降解融合蛋白来模拟降解处理。所有的遗传系统都使用degron和定义配体的组合,使与degron融合的感兴趣的蛋白质被招募到E3泛素连接酶上进行泛素化,随后被蛋白酶体降解。然而,这些事件基于不同的原则,具有不同的特点。我们回顾了dTAG、基于halotag、生长素诱导降解(AID)和不稳定结构域(DD)系统,并讨论了针对新靶蛋白的降解物发现策略。
{"title":"Ligand-induced genetic degradation as a tool for target validation","authors":"Aisha Yesbolatova ,&nbsp;Yusuke Tominari ,&nbsp;Masato T. Kanemaki","doi":"10.1016/j.ddtec.2018.11.001","DOIUrl":"10.1016/j.ddtec.2018.11.001","url":null,"abstract":"<div><p>Targeted protein degraders, known as proteolysis<span> targeting chimeras (PROTACs), are drawing more attention as next-generation drugs to target currently undruggable proteins. As drug discovery<span><span> of functional degraders involves time- and cost-consuming laborious processes, we propose employing a ligand-induced genetic degradation system to validate candidate proteins before degrader development. Genetic degradation mimics degrader treatment by depleting a degron-fused protein in the presence of a defined ligand. All genetic systems use a combination of a degron and defined ligand that enables a protein of interest fused with the degron to be recruited to an E3 </span>ubiquitin ligase<span><span> for ubiquitylation and subsequent degradation by the </span>proteasome. However, these events are based on different principles and have different features. We review the dTAG, HaloTag-based, auxin-inducible degron (AID), and destabilizing domain (DD) systems and discuss a strategy for degrader discovery against novel target proteins.</span></span></span></p></div>","PeriodicalId":36012,"journal":{"name":"Drug Discovery Today: Technologies","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.ddtec.2018.11.001","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37336303","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 23
Targeted protein degradation mechanisms 靶向蛋白降解机制
Q1 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2019-04-01 DOI: 10.1016/j.ddtec.2019.01.001
Yi Zhang, Christine Loh, Jesse Chen, Nello Mainolfi

Targeted protein degradation mediated by small molecule degraders represents an exciting new therapeutic opportunity to eliminate disease-causing proteins. These molecules recruit E3 ubiquitin ligases to the protein of interest and mediate its ubiquitination and subsequent proteolysis by the proteasome. Significant advancements have been made in the discovery and development of clinically relevant degraders. In this review we will focus on the recent progress in understanding ternary complex formation and structures, ubiquitination, and other critical factors that govern the efficiency of degraders both in vitro and in vivo. With deeper knowledges of these areas, the field is building guiding principles to reduce the level of empiricism and to identify therapeutically relevant degraders more rationally and efficiently.

由小分子降解物介导的靶向蛋白质降解代表了消除致病蛋白质的令人兴奋的新治疗机会。这些分子将E3泛素连接酶招募到感兴趣的蛋白质上,并介导其泛素化和随后由蛋白酶体进行的蛋白质水解。在临床相关降解物的发现和开发方面取得了重大进展。在这篇综述中,我们将集中在了解三元络合物的形成和结构,泛素化,以及其他控制降解剂在体外和体内效率的关键因素方面的最新进展。随着对这些领域的深入了解,该领域正在建立指导原则,以减少经验主义的水平,并更合理、更有效地识别与治疗相关的降解物。
{"title":"Targeted protein degradation mechanisms","authors":"Yi Zhang,&nbsp;Christine Loh,&nbsp;Jesse Chen,&nbsp;Nello Mainolfi","doi":"10.1016/j.ddtec.2019.01.001","DOIUrl":"10.1016/j.ddtec.2019.01.001","url":null,"abstract":"<div><p><span>Targeted protein degradation<span> mediated by small molecule<span> degraders represents an exciting new therapeutic opportunity to eliminate disease-causing proteins. These molecules recruit E3 ubiquitin ligases<span><span> to the protein of interest and mediate its ubiquitination and subsequent proteolysis by the </span>proteasome<span>. Significant advancements have been made in the discovery and development of clinically relevant degraders. In this review we will focus on the recent progress in understanding ternary complex formation and structures, ubiquitination, and other critical factors that govern the efficiency of degraders both </span></span></span></span></span><em>in vitro</em> and <em>in vivo</em>. With deeper knowledges of these areas, the field is building guiding principles to reduce the level of empiricism and to identify therapeutically relevant degraders more rationally and efficiently.</p></div>","PeriodicalId":36012,"journal":{"name":"Drug Discovery Today: Technologies","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.ddtec.2019.01.001","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37332834","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 30
Monitoring and deciphering protein degradation pathways inside cells 监测和破译细胞内的蛋白质降解途径
Q1 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2019-04-01 DOI: 10.1016/j.ddtec.2018.12.001
Danette L. Daniels, Kristin M. Riching, Marjeta Urh

A new series of therapeutic modalities resulting in degradation of target proteins, termed proteolysis targeting chimeras (PROTACs), hold significant therapeutic potential with possible prolonged pharmacodynamics, improved potency, and ability to target proteins previously thought of as “undruggable”. PROTACs are heterobifunctional small molecules consisting of a target binding handle bridged via a chemical linker to an E3 ligase handle which recruit the E3 ligase and ubiquitin machinery to target proteins, resulting in subsequent ubiquitination and degradation of the target. With the generation of small molecule PROTAC compound libraries for drug discovery, it becomes essential to have sensitive screening technologies to rapidly profile activity and have assays which can clearly inform on performance at the various cellular steps required for PROTAC-mediated degradation. For PROTAC compounds, this has been particularly challenging using either biochemical or cellular assay approaches. Biochemical assays are highly informative for the first part of the degradation process, including optimization of compound binding to targets and interrogation of target:PROTAC:E3 ligase ternary complex formation, but struggle with the remaining steps; recruitment of ternary complex into larger active E3 ligase complexes, ubiquitination, and proteasomal degradation. On the other hand, cellular assays are excellent at determining if the PROTAC successfully degrades the target in its relevant setting but struggle as early development PROTAC compounds are often poorly cell-permeable given their high molecular weight. Additionally, if degradation is not observed in a cellular assay, it is difficult to deconvolute the reason why or at which step there was failure. In this review we will highlight the current approaches along with recent advances to overcome the challenges faced for cellular PROTAC screening, which will enable and advance drug discovery of therapeutic degradation compounds.

一系列新的治疗方式导致靶蛋白降解,称为蛋白水解靶向嵌合体(proteolysis targeting chimeras, PROTACs),具有显著的治疗潜力,可能延长药效学,提高效力,并能够靶向以前认为“不可药物”的蛋白质。PROTACs是一种异功能小分子,由一个通过化学连接物连接到E3连接酶手柄的靶标结合手柄组成,该手柄将E3连接酶和泛素机制招募到靶标蛋白上,导致随后的泛素化和靶标降解。随着用于药物发现的小分子PROTAC化合物文库的产生,拥有敏感的筛选技术来快速分析活性和能够清楚地告知PROTAC介导的降解所需的各种细胞步骤的性能的分析变得至关重要。对于PROTAC化合物,使用生化或细胞测定方法尤其具有挑战性。生化分析对降解过程的第一部分具有很高的信息量,包括化合物与靶标结合的优化和靶标:PROTAC:E3连接酶三元复合物形成的询问,但在其余步骤中却很困难;三元复合物募集到更大的活性E3连接酶复合物,泛素化和蛋白酶体降解。另一方面,细胞分析在确定PROTAC是否成功降解相关环境中的靶标方面非常出色,但由于PROTAC化合物的高分子量,其早期开发通常具有较差的细胞渗透性。此外,如果在细胞测定中未观察到降解,则很难解卷积失败的原因或在哪一步失败。在这篇综述中,我们将重点介绍目前的方法以及最近的进展,以克服细胞PROTAC筛选面临的挑战,这将使和推进治疗性降解化合物的药物发现。
{"title":"Monitoring and deciphering protein degradation pathways inside cells","authors":"Danette L. Daniels,&nbsp;Kristin M. Riching,&nbsp;Marjeta Urh","doi":"10.1016/j.ddtec.2018.12.001","DOIUrl":"10.1016/j.ddtec.2018.12.001","url":null,"abstract":"<div><p>A new series of therapeutic modalities resulting in degradation of target proteins, termed proteolysis targeting chimeras (PROTACs), hold significant therapeutic potential with possible prolonged pharmacodynamics, improved potency, and ability to target proteins previously thought of as “undruggable”. PROTACs are heterobifunctional small molecules consisting of a target binding handle bridged via a chemical linker to an E3 ligase handle which recruit the E3 ligase and ubiquitin machinery to target proteins, resulting in subsequent ubiquitination and degradation of the target. With the generation of small molecule PROTAC compound libraries for drug discovery, it becomes essential to have sensitive screening technologies to rapidly profile activity and have assays which can clearly inform on performance at the various cellular steps required for PROTAC-mediated degradation. For PROTAC compounds, this has been particularly challenging using either biochemical or cellular assay approaches. Biochemical assays are highly informative for the first part of the degradation process, including optimization of compound binding to targets and interrogation of target:PROTAC:E3 ligase ternary complex formation, but struggle with the remaining steps; recruitment of ternary complex into larger active E3 ligase complexes, ubiquitination, and proteasomal degradation. On the other hand, cellular assays are excellent at determining if the PROTAC successfully degrades the target in its relevant setting but struggle as early development PROTAC compounds are often poorly cell-permeable given their high molecular weight. Additionally, if degradation is not observed in a cellular assay, it is difficult to deconvolute the reason why or at which step there was failure. In this review we will highlight the current approaches along with recent advances to overcome the challenges faced for cellular PROTAC screening, which will enable and advance drug discovery of therapeutic degradation compounds.</p></div>","PeriodicalId":36012,"journal":{"name":"Drug Discovery Today: Technologies","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.ddtec.2018.12.001","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37332835","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 41
Targeted protein degradation in vivo with Proteolysis Targeting Chimeras: Current status and future considerations 蛋白质水解靶向嵌合体在体内的靶向蛋白质降解:现状和未来的考虑
Q1 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2019-04-01 DOI: 10.1016/j.ddtec.2019.02.005
Gillian F. Watt , Paul Scott-Stevens , Lu Gaohua

Proteolysis Targeting Chimeras (PROTACs) are a rapidly expanding new therapeutic modality inducing selective protein degradation and offering the potential of a differentiated pharmacological profile across multiple therapeutic areas. As the repertoire of protein targets and E3 ligases available for incorporation into PROTACs continues to grow, understanding the drug- and system-dependent parameters for PROTACs will be critical for achieving tissue/cell specific pharmacology. The review discusses the current knowledge and future direction of in vivo PROTAC study evaluation. The importance of establishing the quantitative relationship between loss of protein target and biological function in vivo, coupled with building mechanistic PK/PD and ultimately PBPK/PD models, is emphasised with the aim to aid translation from preclinical to clinical space.

靶向嵌合体(Proteolysis Targeting Chimeras, PROTACs)是一种快速发展的新型治疗方式,可诱导选择性蛋白质降解,并在多个治疗领域提供差异化的药理学特征。随着可并入PROTACs的蛋白靶点和E3连接酶的不断增加,了解PROTACs的药物和系统依赖性参数对于实现组织/细胞特异性药理学至关重要。本文讨论了PROTAC体内研究评价的现状和未来发展方向。本文强调了在体内建立蛋白靶点损失与生物功能之间的定量关系,以及建立机制PK/PD和最终PBPK/PD模型的重要性,目的是帮助从临床前到临床空间的转化。
{"title":"Targeted protein degradation in vivo with Proteolysis Targeting Chimeras: Current status and future considerations","authors":"Gillian F. Watt ,&nbsp;Paul Scott-Stevens ,&nbsp;Lu Gaohua","doi":"10.1016/j.ddtec.2019.02.005","DOIUrl":"10.1016/j.ddtec.2019.02.005","url":null,"abstract":"<div><p><span>Proteolysis<span> Targeting Chimeras (PROTACs) are a rapidly expanding new therapeutic modality inducing selective protein degradation and offering the potential of a differentiated pharmacological profile across multiple therapeutic areas. As the repertoire of protein targets and E3 ligases available for incorporation into PROTACs continues to grow, understanding the drug- and system-dependent parameters for PROTACs will be critical for achieving tissue/cell specific pharmacology. The review discusses the current knowledge and future direction of </span></span><em>in vivo</em> PROTAC study evaluation. The importance of establishing the quantitative relationship between loss of protein target and biological function <em>in vivo</em>, coupled with building mechanistic PK/PD and ultimately PBPK/PD models, is emphasised with the aim to aid translation from preclinical to clinical space.</p></div>","PeriodicalId":36012,"journal":{"name":"Drug Discovery Today: Technologies","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.ddtec.2019.02.005","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37332836","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 55
PROteolysis TArgeting Chimeras (PROTACs) — Past, present and future 靶向嵌合体(PROTACs)的蛋白水解-过去,现在和未来
Q1 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2019-04-01 DOI: 10.1016/j.ddtec.2019.01.002
Mariell Pettersson, Craig M. Crews

The majority of currently used therapeutics are small molecule-based and utilize occupancy-driven pharmacology as the mode of action (MOA), in which the protein function is modulated via temporary inhibition. New modalities that operate using alternative MOAs are essential for tapping into the “undruggable” proteome. The PROteolysis Targeting Chimera (PROTAC) technology provides an attractive new approach that utilizes an event-driven MOA. Small molecule-based heterobifunctional PROTACs modulate protein target levels by hijacking the ubiquitin-proteasome system to induce degradation of the target. Here, we address important milestones in the development of the PROTAC technology, as well as emphasize key findings from this previous year and highlight future directions of this promising drug discovery modality.

目前使用的大多数治疗方法都是基于小分子的,并利用占位驱动的药理学作为作用模式(MOA),其中蛋白质功能通过暂时抑制来调节。使用替代moa的新模式对于利用“不可药物”的蛋白质组至关重要。PROteolysis Targeting Chimera (PROTAC)技术提供了一种有吸引力的新方法,利用事件驱动的MOA。基于小分子的异功能PROTACs通过劫持泛素-蛋白酶体系统来诱导靶标的降解来调节蛋白靶标水平。在这里,我们讨论了PROTAC技术发展中的重要里程碑,强调了去年的主要发现,并强调了这种有前途的药物发现模式的未来方向。
{"title":"PROteolysis TArgeting Chimeras (PROTACs) — Past, present and future","authors":"Mariell Pettersson,&nbsp;Craig M. Crews","doi":"10.1016/j.ddtec.2019.01.002","DOIUrl":"10.1016/j.ddtec.2019.01.002","url":null,"abstract":"<div><p>The majority of currently used therapeutics are small molecule-based and utilize occupancy-driven pharmacology as the mode of action (MOA), in which the protein function is modulated <em>via</em> temporary inhibition. New modalities that operate using alternative MOAs are essential for tapping into the “undruggable” proteome. The PROteolysis Targeting Chimera (PROTAC) technology provides an attractive new approach that utilizes an event-driven MOA. Small molecule-based heterobifunctional PROTACs modulate protein target levels by hijacking the ubiquitin-proteasome system to induce degradation of the target. Here, we address important milestones in the development of the PROTAC technology, as well as emphasize key findings from this previous year and highlight future directions of this promising drug discovery modality.</p></div>","PeriodicalId":36012,"journal":{"name":"Drug Discovery Today: Technologies","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.ddtec.2019.01.002","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37332830","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 376
Cereblon modulators: Low molecular weight inducers of protein degradation 小脑调节剂:蛋白质降解的低分子量诱导剂
Q1 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2019-04-01 DOI: 10.1016/j.ddtec.2019.02.004
Philip P. Chamberlain, Brian E. Cathers

Targeted protein degradation has become an exciting new paradigm in drug discovery with the potential to target new protein families for therapeutic intervention. In 2010, Hiroshi Handa and colleagues discovered that the drug thalidomide binds to the protein cereblon, a component of the CRL4CRBN E3 ubiquitin ligase. In contrast to the heterobifunctional small molecule degraders reported in the literature, thalidomide is of very low molecular weight (∼258Da) with molecular properties (solubility, metabolic stability, permeability etc) that readily support pharmaceutical dosing. It was subsequently shown that thalidomide and the analogues lenalidomide and pomalidomide are able to degrade the transcription factors Ikaros and Aiolos. CK1α and GSPT1 were subsequently identified as substrates for specific ligands, indicating that this molecular class could be tuned for selective protein degradation. Structural studies showed that the thalidomide analogues bind to a shallow hydrophobic pocket on the surface of cereblon, and scaffold a protein-protein interaction with target proteins. Target proteins do not need any affinity for the cereblon modulators, and as such undruggable, or even unligandable, proteins can be targeted for degradation. A similar mechanism of action was subsequently identified for the clinical molecule indisulam, indicating that low molecular weight degraders are not unique to cereblon. The groundbreaking work on cereblon represents a case study for the discovery and characterization of low molecular weight protein degraders for other ligases.

靶向蛋白降解已经成为药物发现的一个令人兴奋的新范式,有可能针对新的蛋白家族进行治疗干预。2010年,Hiroshi Handa和他的同事发现药物沙利度胺与小脑蛋白结合,小脑蛋白是CRL4CRBN E3泛素连接酶的一个组成部分。与文献中报道的异双功能小分子降解剂相比,沙利度胺的分子量非常低(~ 258Da),其分子特性(溶解度、代谢稳定性、渗透性等)很容易支持药物剂量。随后的研究表明,沙利度胺及其类似物来那度胺和泊马度胺能够降解转录因子Ikaros和Aiolos。CK1α和GSPT1随后被鉴定为特定配体的底物,表明该分子类可以被调整为选择性蛋白质降解。结构研究表明,沙利度胺类似物与小脑表面的浅疏水口袋结合,并支架与靶蛋白的蛋白质相互作用。靶蛋白不需要与小脑调节剂有任何亲和力,因此不可药物,甚至不可配体,蛋白质可以作为降解的靶标。类似的作用机制随后被确定为临床分子胰岛素,表明低分子量降解物并非小脑所独有。小脑的开创性工作代表了其他连接酶的低分子量蛋白质降解物的发现和表征的案例研究。
{"title":"Cereblon modulators: Low molecular weight inducers of protein degradation","authors":"Philip P. Chamberlain,&nbsp;Brian E. Cathers","doi":"10.1016/j.ddtec.2019.02.004","DOIUrl":"10.1016/j.ddtec.2019.02.004","url":null,"abstract":"<div><p><span>Targeted protein degradation<span> has become an exciting new paradigm in drug discovery<span><span> with the potential to target new protein families for therapeutic intervention. In 2010, Hiroshi Handa and colleagues discovered that the drug thalidomide binds to the protein </span>cereblon, a component of the CRL4</span></span></span><sup>CRBN</sup><span><span> E3 ubiquitin ligase<span><span><span>. In contrast to the heterobifunctional small molecule degraders reported in the literature, thalidomide is of very low molecular weight (∼258Da) with molecular properties (solubility, metabolic stability, permeability etc) that readily support pharmaceutical dosing. It was subsequently shown that thalidomide and the analogues </span>lenalidomide and </span>pomalidomide<span><span> are able to degrade the transcription factors Ikaros and Aiolos. CK1α and GSPT1 were subsequently identified as substrates for specific ligands, indicating that this molecular class could be tuned for selective protein degradation. Structural studies showed that the thalidomide analogues bind to a shallow hydrophobic pocket on the surface of cereblon, and scaffold a protein-protein interaction with target proteins. Target proteins do not need any affinity for the cereblon modulators, and as such undruggable, or even unligandable, proteins can be targeted for degradation. A similar mechanism of action was subsequently identified for the clinical molecule </span>indisulam, indicating that low molecular weight degraders are not unique to cereblon. The groundbreaking work on cereblon represents a case study for the discovery and characterization of low molecular weight protein degraders for other </span></span></span>ligases.</span></p></div>","PeriodicalId":36012,"journal":{"name":"Drug Discovery Today: Technologies","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.ddtec.2019.02.004","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37332829","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 58
SNIPERs—Hijacking IAP activity to induce protein degradation snipers -劫持IAP活性诱导蛋白质降解
Q1 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2019-04-01 DOI: 10.1016/j.ddtec.2018.12.002
Mikihiko Naito, Nobumichi Ohoka, Norihito Shibata

The induction of protein degradation by chimeric small molecules represented by proteolysis-targeting chimeras (PROTACs) is an emerging approach for novel drug development. We have developed a series of chimeric molecules termed specific and non-genetic inhibitor of apoptosis protein (IAP)-dependent protein erasers (SNIPERs) that recruit IAP ubiquitin ligases to effect targeted degradation. Unlike the chimeric molecules that recruit von Hippel–Lindau and cereblon ubiquitin ligases, SNIPERs induce simultaneous degradation of IAPs such as cIAP1 and XIAP along with the target proteins. Because cancer cells often overexpress IAPs—a mechanism involved in the resistance to cancer therapy—SNIPERs could be used to kill cancer cells efficiently.

以靶向蛋白水解嵌合体(proteolysis-targeting chimeras, PROTACs)为代表的嵌合小分子诱导蛋白质降解是一种新兴的新药开发方法。我们已经开发了一系列嵌合分子,称为特异性和非遗传性凋亡蛋白抑制剂(IAP)依赖蛋白擦除剂(SNIPERs),它们募集IAP泛素连接酶来实现靶向降解。与利用von Hippel-Lindau和小脑泛素连接酶的嵌合分子不同,SNIPERs诱导IAPs(如cIAP1和XIAP)与靶蛋白同时降解。由于癌细胞经常过度表达iaps(一种与癌症治疗抵抗有关的机制),snipers可以用来有效地杀死癌细胞。
{"title":"SNIPERs—Hijacking IAP activity to induce protein degradation","authors":"Mikihiko Naito,&nbsp;Nobumichi Ohoka,&nbsp;Norihito Shibata","doi":"10.1016/j.ddtec.2018.12.002","DOIUrl":"10.1016/j.ddtec.2018.12.002","url":null,"abstract":"<div><p>The induction of protein degradation by chimeric small molecules represented by proteolysis-targeting chimeras (PROTACs) is an emerging approach for novel drug development. We have developed a series of chimeric molecules termed specific and non-genetic inhibitor of apoptosis protein (IAP)-dependent protein erasers (SNIPERs) that recruit IAP ubiquitin ligases to effect targeted degradation. Unlike the chimeric molecules that recruit von Hippel–Lindau and cereblon ubiquitin ligases, SNIPERs induce simultaneous degradation of IAPs such as cIAP1 and XIAP along with the target proteins. Because cancer cells often overexpress IAPs—a mechanism involved in the resistance to cancer therapy—SNIPERs could be used to kill cancer cells efficiently.</p></div>","PeriodicalId":36012,"journal":{"name":"Drug Discovery Today: Technologies","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.ddtec.2018.12.002","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37332831","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 93
Small-molecule PROTAC degraders of the Bromodomain and Extra Terminal (BET) proteins — A review Bromodomain和Extra Terminal (BET)蛋白的小分子PROTAC降解物研究进展
Q1 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2019-04-01 DOI: 10.1016/j.ddtec.2019.04.001
Chao-Yie Yang, Chong Qin, Longchuan Bai, Shaomeng Wang

The PROteolysis TArgeting Chimeric (PROTAC) concept has provided an opportunity for the discovery and development of a completely new type of therapy involving induction of protein degradation. The BET proteins, comprised of BRD2, BRD3, BRD4 and the testis-specific BRDT protein, are epigenetic readers and master transcription coactivators. Extremely potent and efficacious small-molecule PROTAC degraders of the BET proteins, based on available, potent and selective BET inhibitors, have been reported. BET degraders differ from BET inhibitors in their cellular potency, phenotypic effects, pharmacokinetic properties and toxicity profiles. Herein, we provide a review of BET degraders and the differential outcome observed in the cellular and animal models for BET degraders in comparison to BET inhibitors.

PROteolysis TArgeting Chimeric (PROTAC)概念为发现和开发一种涉及诱导蛋白质降解的全新治疗方法提供了机会。BET蛋白由BRD2、BRD3、BRD4和睾丸特异性BRDT蛋白组成,是表观遗传读取器和主转录共激活因子。基于可用的、有效的和选择性的BET抑制剂,已经报道了非常有效和有效的BET蛋白的小分子PROTAC降解剂。BET降解剂与BET抑制剂在细胞效力、表型效应、药代动力学性质和毒性方面不同。在此,我们综述了BET降解物以及在细胞和动物模型中与BET抑制剂相比,BET降解物观察到的不同结果。
{"title":"Small-molecule PROTAC degraders of the Bromodomain and Extra Terminal (BET) proteins — A review","authors":"Chao-Yie Yang,&nbsp;Chong Qin,&nbsp;Longchuan Bai,&nbsp;Shaomeng Wang","doi":"10.1016/j.ddtec.2019.04.001","DOIUrl":"10.1016/j.ddtec.2019.04.001","url":null,"abstract":"<div><p>The <u>PRO</u>teolysis <u>TA</u>rgeting <u>C</u><span><span><span><span>himeric (PROTAC) concept has provided an opportunity for the discovery and development of a completely new type of therapy involving induction of protein degradation. The BET<span> proteins, comprised of BRD2, BRD3, </span></span>BRD4<span><span> and the testis-specific BRDT protein, are </span>epigenetic readers and master transcription coactivators. Extremely potent and efficacious small-molecule PROTAC degraders of the BET proteins, based on available, potent and selective BET inhibitors, have been reported. BET degraders differ from BET inhibitors in their cellular potency, phenotypic effects, </span></span>pharmacokinetic properties and toxicity profiles. Herein, we provide a review of BET degraders and the differential outcome observed in the cellular and </span>animal models for BET degraders in comparison to BET inhibitors.</span></p></div>","PeriodicalId":36012,"journal":{"name":"Drug Discovery Today: Technologies","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.ddtec.2019.04.001","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37332832","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 76
Advanced proteomics approaches to unravel protein homeostasis 先进的蛋白质组学方法揭示蛋白质稳态
Q1 Pharmacology, Toxicology and Pharmaceutics Pub Date : 2019-04-01 DOI: 10.1016/j.ddtec.2019.02.001
Paola Grandi, Marcus Bantscheff

Quantitative proteomics methods are instrumental in measuring the interplay between protein synthesis and protein degradation in cells and tissues in different conditions and substantially contribute to the understanding of control mechanisms for protein homeostasis. Proteomics and chemoproteomics approaches enable the characterization of small molecule modifiers of protein degradation for therapeutic applications. Here, we review recent developments and applications of mass spectrometry-based (chemo-)proteomics methods for the study of cellular homeostasis.

定量蛋白质组学方法有助于测量不同条件下细胞和组织中蛋白质合成和蛋白质降解之间的相互作用,并有助于理解蛋白质稳态的控制机制。蛋白质组学和化学蛋白质组学方法能够表征用于治疗应用的蛋白质降解的小分子修饰剂。在这里,我们回顾了基于质谱(化学)蛋白质组学方法在细胞稳态研究中的最新进展和应用。
{"title":"Advanced proteomics approaches to unravel protein homeostasis","authors":"Paola Grandi,&nbsp;Marcus Bantscheff","doi":"10.1016/j.ddtec.2019.02.001","DOIUrl":"10.1016/j.ddtec.2019.02.001","url":null,"abstract":"<div><p>Quantitative proteomics methods are instrumental in measuring the interplay between protein synthesis and protein degradation in cells and tissues in different conditions and substantially contribute to the understanding of control mechanisms for protein homeostasis. Proteomics and chemoproteomics approaches enable the characterization of small molecule modifiers of protein degradation for therapeutic applications. Here, we review recent developments and applications of mass spectrometry-based (chemo-)proteomics methods for the study of cellular homeostasis.</p></div>","PeriodicalId":36012,"journal":{"name":"Drug Discovery Today: Technologies","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1016/j.ddtec.2019.02.001","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"37336304","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 13
期刊
Drug Discovery Today: Technologies
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1