Drug Discovery Today: Technologies最新文献

筛选
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
Drug Discovery Today: Technologies Pub Date : 2019-04-01 DOI: 10.1016/j.ddtec.2018.12.003
Cristina Mayor-Ruiz, Georg E. Winter
{"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
Drug Discovery Today: Technologies Pub Date : 2019-04-01 DOI: 10.1016/j.ddtec.2018.11.001
Aisha Yesbolatova , Yusuke Tominari , Masato T. Kanemaki
{"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
Drug Discovery Today: Technologies Pub Date : 2019-04-01 DOI: 10.1016/j.ddtec.2019.01.001
Yi Zhang, Christine Loh, Jesse Chen, Nello Mainolfi
{"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
Drug Discovery Today: Technologies Pub Date : 2019-04-01 DOI: 10.1016/j.ddtec.2018.12.001
Danette L. Daniels, Kristin M. Riching, Marjeta Urh
{"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
Drug Discovery Today: Technologies Pub Date : 2019-04-01 DOI: 10.1016/j.ddtec.2019.02.005
Gillian F. Watt , Paul Scott-Stevens , Lu Gaohua
{"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
Drug Discovery Today: Technologies Pub Date : 2019-04-01 DOI: 10.1016/j.ddtec.2019.01.002
Mariell Pettersson, Craig M. Crews
{"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
Drug Discovery Today: Technologies Pub Date : 2019-04-01 DOI: 10.1016/j.ddtec.2019.02.004
Philip P. Chamberlain, Brian E. Cathers
{"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
Drug Discovery Today: Technologies Pub Date : 2019-04-01 DOI: 10.1016/j.ddtec.2018.12.002
Mikihiko Naito, Nobumichi Ohoka, Norihito Shibata
{"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
Drug Discovery Today: Technologies Pub Date : 2019-04-01 DOI: 10.1016/j.ddtec.2019.04.001
Chao-Yie Yang, Chong Qin, Longchuan Bai, Shaomeng Wang
{"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
Drug Discovery Today: Technologies Pub Date : 2019-04-01 DOI: 10.1016/j.ddtec.2019.02.001
Paola Grandi, Marcus Bantscheff
{"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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信