Download Free Inducing Targeted Protein Degradation Book in PDF and EPUB Free Download. You can read online Inducing Targeted Protein Degradation and write the review.

This volume contains a collection of innovative techniques for studying targeted protein degradation. Chapters guide readers through heterobifunctional proteolysis-targeting chimeras (PROTACs) approaches, E3 ligase, E3 ligase-induced ubiquitylation, proteomic approaches, novel degrader molecules, molecular glue, and stabilize binding interaction between a target and E3 ubiquitin ligase. Written in the format of the highly successful Methods in Molecular Biology series, each chapter includes an introduction to the topic, lists necessary materials and reagents, includes tips on troubleshooting and known pitfalls, and step-by-step, readily reproducible protocols. Authoritative and cutting-edge, Targeted Protein Degradation: Methods and Protocols aims to ensure successful results in this emerging field of drug discovery.
Inducing Targeted Protein Degradation Enables drug developers in academia and industry to expand the range of accessible drug targets through induced protein degradation Since the breakthrough of the PROTAC technology in 2015, targeted protein degradation has revolutionized drug discovery, enabling pharma companies to develop completely novel therapeutics. Inducing Targeted Protein Degradation is a timely guide to navigating the complexities of the subject and understanding its practical application, with an eye on expanding the druggable space. In Inducing Targeted Protein Degradation, readers will find the most recent information on: Cellular mechanisms of targeted protein degradation and current approaches to utilize these mechanisms for drug discovery A comparison of different induced degradation approaches, including PROTAC, molecular glues, LYTACs and ATTECs as well as additional post translational modifications Drug development aspects such as DMPK optimization and criteria for the selection of clinical candidates A discussion of the potential of targeted degradation for expanding the druggable space Inducing Targeted Protein Degradation will serve as a practice-oriented reference on induced protein degradation for drug discovery professionals and for researchers employing chemical biology approaches.
Targeting protein degradation using small molecules is one of the most exciting small-molecule therapeutic strategies in decades and a rapidly growing area of research. In particular, the development of proteolysis targeting chimera (PROTACs) as potential drugs capable of recruiting target proteins to the cellular quality control machinery for elimination has opened new avenues to address traditionally ‘difficult to target’ proteins. This book provides a comprehensive overview from the leading academic and industrial experts on recent developments, scope and limitations in this dynamically growing research area; an ideal reference work for researchers in drug discovery and chemical biology as well as advanced students.
Proteolysis targeting chimeras (PROTACs) are heterobifunctional small molecules which induce targeted protein degradation by redirecting the ubiquitin-proteasome system. PROTACs simultaneously bind to both a protein of interest and an E3 ubiquitin ligase. The proximity of the target protein and the E3 ligase complex allows transfer of ubiquitin onto the target protein, after which the protein can be recognised and then degraded by the proteasome. The PROTAC mechanism-of-action offers a number of potential advantages over small molecule inhibition for the development of new medicines. Efficacy maybe achieved from low doses, extended duration of action is possible, arising from pharmacokinetic pharmacodynamic disconnects, and challenging targets may become tractable through the identification of suitable affinity binders. In this research, the development of PROTAC technology is explored. In order to expand the breadth of E3 ligases that are currently recruited using this approach, a promiscuous toolbox was established to prosecute new chemical matter for E3 ligases. In order to further elaborate the promiscuous toolbox already known for the degradation of kinases, investigation of a promiscuous bromodomain PROTAC was explored. After assessment of bromosporine derived PROTACs in multiple cell lines with two validated E3 ligases, it was deemed an unsuitable binder for the toolbox. The PROTACs were unable to induce potent nor promiscuous bromodomain degradation. As a result, a known BET bromodomain binder was selected for the promiscuous toolbox in addition to the known promiscuous kinase binder and a RIPK2 binder for new E3 ligase validation. With a promiscuous toolbox in hand, a new E3 ligase was evaluated. Indisulam, a small molecule "molecular glue", was found to bind to the E3 ligase DCAF15. Indisulam derived PROTACs were synthesised and evaluated with a range of linker lengths and multiple protein binders. These studies conclusively demonstrated that protein degradation was not achieved using these PROTACs. The indisulam derived binder was subsequently found not to be suitable for the PROTAC approach without further investigation to determine DCAF15 recruitment. Given the lack of degradation with the initial, empirically selected E3 ligases from the literature, a distinct E3 ligase agnostic approach to protein degradation was developed. A high throughput phenotypic screen was established using green fluorescent protein (GFP) as the protein of interest, where cellular fluorescence levels correlate with protein degradation. High throughput chemistry techniques were implemented and optimised to synthesise thousands of HaloCompounds in-situ by amide coupling. The compounds were tested directly in cells to find new chemical matter for the induction of protein degradation. This strategy allowed identification of several potential hits from a 3000-amine screen, with one high-confidence hit currently being further evaluated. For this effort the screen was optimised successfully and can potentially be employed for a target-agnostic high-throughput screening campaign of hundreds of thousands of compounds for new E3 ligases to employ in future protein degradation strategies.
Advances in anti-cancer chemotherapy over recent years have led to improved efficacy in curing or controlling many cancers. Some chemotherapy-related side-effects are well recognized and include: nausea, vomiting, bone marrow suppression, peripheral neuropathy, cardiac and skeletal muscle dysfunction and renal impairment. However, it is becoming clearer that some chemotherapy-related adverse effects may persist even in long term cancer survivors. Problems such as cognitive, cardiovascular and gastrointestinal dysfunction, and neuropathy may lead to substantial long term morbidity. Despite improvements in treatments to counteract acute chemotherapy-induced adverse effects, they are often incompletely effective. Furthermore, counter-measures for some acute side-effects and many potential longer term sequelae of anti-cancer chemotherapy have not been developed. Thus, new insights into prevalence and mechanisms of cancer chemotherapy-related side effects are needed and new approaches to improving tolerance and reduce sequelae of cancer chemotherapy are urgently needed. The present Research Topic focuses on adverse effects and sequelae of chemotherapy and strategies to counteract them.
Targeting protein degradation using small molecules is one of the most exciting small-molecule therapeutic strategies in decades and a rapidly growing area of research. In particular, the development of proteolysis targeting chimera (PROTACs) as potential drugs capable of recruiting target proteins to the cellular quality control machinery for elimination has opened new avenues to address traditionally 'difficult to target' proteins. This book provides a comprehensive overview from the leading academic and industrial experts on recent developments, scope and limitations in this dynamically growing research area; an ideal reference work for researchers in drug discovery and chemical biology as well as advanced students.
Filled with unique insights into current drugs that have made it to the marketplace In the fifth volume of Successful Drug Discovery, the inventors and primary developers of drugs that made it to the market tell the story of the drugs discovery and development. Case studies of drugs from different therapeutic fields reveal the all-too-often unpredictable path from the first drug candidate molecule to the successfully marketed drug. In addition, this new volume addresses overarching topics for drug discovery, such as drug discovery in academia, and discusses currently important classes of small molecule as well as biological drugs. Comprehensive in scope, the books nine chapters provide a representative cross-section of the present-day drug development effort. The authoritative fifth volume is filled with relevant data and chemical information, as well as the insight and experience of the best contemporary drug creators. This important volume: - Puts the focus on recently introduced drugs that have not yet made it into standard textbooks or general references - Contains information and insight that is new and often not even available from the primary literature - Reveals what it takes to successfully develop a drug molecule that has made it all the way to the market - Is endorsed and supported by the International Union of Pure and Applied Chemistry (IUPAC) Written for medicinal chemists, pharmaceutical chemists, organic chemists, Successful Drug Discovery, Volume Five reveals the most recent techniques used by drug innovators in the drug development process.
In this riveting medical detective story, Trent Stephens and Rock Brynner recount the history of thalidomide, from the epidemic of birth defects in the 1960's to the present day, as scientists work to create and test an alternative drug that captures thalidomide's curative properties without its cruel side effects. A parable about compassion-and the absence of it-Dark Remedy is a gripping account of thalidomide's extraordinary impact on the lives of individuals and nations over half a century.
This book summarizes state-of-the-art antiviral drug design and discovery approaches starting from natural products to de novo design, and provides a timely update on recently approved antiviral drugs and compounds in advanced clinical development. Special attention is paid to viral infections with a high impact on the world population or highly relevant from the public health perspective (HIV, hepatitis C, influenza virus, etc.). In these chapters, limitations associated with adverse effects and emergence of drug resistance are discussed in detail. In addition to classical antiviral strategies, chapters will be dedicated to discuss the non-classical drug development strategies to block viral infection, for instance, allosteric inhibitors, covalent antiviral agents, or antiviral compounds targeting protein–protein interactions. Finally, current prospects for producing broad-spectrum antiviral inhibitors will be also addressed. The book is distinctive in providing the most recent update in the rapidly evolving field of antiviral therapeutics. Authoritative reviews are written by international scientists well known for their contributions in their topics of research, which makes this book suitable for researchers not only within the antiviral research community but also attractive to a broad audience in the drug discovery field. This book covers molecular structures and biochemical mechanisms mediating the antiviral effects, while discussing various ligand design strategies, which include traditional medicinal chemistry, computational chemistry, and chemical biology approaches. The book provides a comprehensive review of antiviral drug discovery and development approaches, particularly focusing on current innovations and future trends.
Proteolysis targeting chimeras (Protacs) are heterobifunctional small molecules which induce targeted protein degradation by hijacking the natural intracellular quality control mechanism, the ubiquitin-proteasome system. Protacs simultaneously bind both a target protein and an E3 ubiquitin ligase, forming a ternary complex. The close proximity of the target protein and the E3 ligase allows transfer of the post-translational modifier ubiquitin onto the target protein, which allows the protein to be recognised and degraded into small peptidic fragments by the proteasome. The Protac approach offers several advantages over small-molecule inhibition alone as efficacy can be driven from low drug concentrations, extended duration of action can be achieved, and scaffolding functions of the target protein can be removed. In this thesis, the application of Protac technology towards a series of disease-relevant proteins is explored. In Chapters 2 and 3, Protacs targeting the kinase ActR2B and the thyroid hormone receptor were developed as potential treatments for sarcopenia and hyperthyroidism, respectively. However, in both cases, no target degradation was observed. Given the lack of degradation with the initial empirically selected targets, a distinct, non-selective approach to protein degradation was considered in Chapter 4. Protacs based on a highly promiscuous kinase inhibitor were designed and synthesised, then profiled using proteome-wide expression proteomics. This strategy allowed identification of several novel degradable targets, and also indicated proteins that may be more challenging to degrade. The opportunity for degradation selectivity in the absence of binding selectivity was also highlighted. Having identified Bruton's tyrosine kinase (BTK) as one of these degradable targets, selective BTK Protacs were then studied in Chapter 5. Protacs based on the covalent inhibitor Ibrutinib led to the surprising discovery that covalent inhibition prevents Protac-mediated degradation of BTK. Protacs developed from a selective, reversible BTK inhibitor allowed rapid interrogation of the kinase as a prototypical Protac target.