High-power screening (HPS) empowered by DNA-encoded libraries

DNA 功率(物理) 计算生物学 心理学 计算机科学 遗传学 生物 物理 量子力学
作者
Yashoda Krishna Sunkari,Vijay Kumar Siripuram,Thu-Lan Nguyen,Marc Flajolet
出处
期刊:Trends in Pharmacological Sciences [Elsevier]
卷期号:43 (1): 4-15 被引量:37
标识
DOI:10.1016/j.tips.2021.10.008
摘要

We propose to group under the umbrella name HPS (high-power screening) technologies that screen an extremely large numbers [thousands to million times larger than HTS] (high-throuput screening) of chemical/biological compounds very effectively. The synergistic forces that differentiate HPS from HTS are the extra-large size of HPS libraries (combinatorial nature) and refinements for extremely efficient drug screening.HTS tests 1 million compounds in 1 million wells (50 000 compounds/week); HPS tests 1 billion compounds in a one tube (1 billion compounds/week).HPS offers novel screening options such as multiplex screening (parallel screening), thus allowing several screens to be performed at the same time (repeats, wild type vs mutants, different isoforms, competition with known compounds).Ex vivo screening is easily accessible with HPS.DNA-encoded libraries (DEL) technology offers new paths to druggable alternatives such as proteolysis targeting chimeras (PROTACs). The world is totally dependent on medications. As science progresses, new, better, and cheaper drugs are needed more than ever. The pharmaceutical industry has been predominantly dependent on high-throughput screening (HTS) for the past three decades. Considering that the discovery rate has been relatively constant, can one hope for a much-needed sudden trend uptick? DNA-encoded libraries (DELs) and similar technologies, that have several orders of magnitude more screening power than HTS, and that we propose to group together under the umbrella term of high-power screening (HPS), are very well positioned to do exactly that. HPS also offers novel screening options such as parallel screening, ex vivo and in vivo screening, as well as a new path to druggable alternatives such as proteolysis targeting chimeras (PROTACs). Altogether, HPS unlocks novel powerful drug discovery avenues. The world is totally dependent on medications. As science progresses, new, better, and cheaper drugs are needed more than ever. The pharmaceutical industry has been predominantly dependent on high-throughput screening (HTS) for the past three decades. Considering that the discovery rate has been relatively constant, can one hope for a much-needed sudden trend uptick? DNA-encoded libraries (DELs) and similar technologies, that have several orders of magnitude more screening power than HTS, and that we propose to group together under the umbrella term of high-power screening (HPS), are very well positioned to do exactly that. HPS also offers novel screening options such as parallel screening, ex vivo and in vivo screening, as well as a new path to druggable alternatives such as proteolysis targeting chimeras (PROTACs). Altogether, HPS unlocks novel powerful drug discovery avenues. Scientific progress and novel technologies are helping to uncover disease causes faster than ever before. Novel emergencies such as the advent of new pathogens (e.g., SARS-CoV2) are further pressuring the pharmaceutical world. To be even faster, cheaper, and more accessible to academia, a new type of power is urgently needed. High-throughput screening (HTS; see Glossary) has been extremely useful over the past three decades (Box 1) and remains at the center of most drug discovery campaigns. However, multiple unsolvable HTS hurdles persist and several technologies that have higher screening power than regular HTS are well positioned to tackle these limitations. These technologies, that have several orders of magnitude increased screening power than HTS, could be grouped together under the umbrella term of high-power screening (HPS). Those include, for example, all types of DNA-encoded library (DEL) technologies and combinatorial protein libraries (e.g., phage display). For reasons of space, we focus only on DEL technologies.Box 1Historical perspective of HTS and limitationsThe 1980s saw the birth of large-scale screening campaigns grouped under the code name of 'high-throughput screening' (HTS). The early evolution of HTS started in around 1984 in large pharmaceutical companies, and Pfizer was the uncontested leader. The catalyzer of the exponential growth of HTS was the birth of modern biochemistry and molecular biology that uncovered novel therapeutic targets (e.g., the h-Ras oncogene). Today, although the universal usefulness of HTS is obvious, its recognition did not take place without effort. It was with some skepticism, and under-preparedness, that the world welcomed HTS in the mid-1980s. In addition to the drug discovery process per se, the driving forces underlying HTS embraced, and revolutionized, diverse disciplines of biology (e.g., DNA sequencing, digital microscopy, protein crystallization). On the other hand, some of its limitations remained large obstacles. The technology has remained relatively cumbersome and rigid in some ways, limiting the number of compounds that can be tested (1 million compounds require 1 million wells) and the size/number of screening campaigns that can be accomplished. HTS libraries are typically smaller than 1 million compounds and are often over-used by the drug industry and academia. The target source, often E. coli, results in products that are often not optimal (e.g., folding, PTMs). Large amounts of protein targets and/or cells compatible with 105 wells are needed, necessitating costly steps, expensive equipment, and logistical requirements, as well as extensive infrastructure. The growing number of therapeutic targets combined with HTS limitations are driving the need for more screening power. This is how combinatorial chemistry erupted a few years after HTS; however, because of the lack of addressability, the application of combinatorial chemistry did not meet its expectations. As a result, the DNA-encoded library (DEL) technology was born, burgeoning from the combinatorial chemistry field. DEL technology, a concept proposed by Drs Richard Lerner and Sidney Brenner, was based on their proof of concept published in 1992 that aimed to solve 'addressability' by adding a unique DNA tag sequence to each drug-like molecule, thus allowing every molecule to be identified at the end of a screening campaign. DNA synthesis capabilities were very limited in the early 1990s, and large-scale sequencing had yet to be invented. The combination of a constantly decreasing cost of DNA synthesis and the emergence of next-generation DNA sequencing ('massively parallel sequencing'; this emerged in the 1990s and was first commercialized in 2005) led to the birth of DEL technology in the mid-2000s. If Pfizer has been associated with the birth of HTS, Drs Lerner and Brenner will certainly be associated with the birth of HPS, begetting it with their initial proof of principle in 1992, and being instrumental in its development since then; Dr Lerner remains at the forefront of the field. The 1980s saw the birth of large-scale screening campaigns grouped under the code name of 'high-throughput screening' (HTS). The early evolution of HTS started in around 1984 in large pharmaceutical companies, and Pfizer was the uncontested leader. The catalyzer of the exponential growth of HTS was the birth of modern biochemistry and molecular biology that uncovered novel therapeutic targets (e.g., the h-Ras oncogene). Today, although the universal usefulness of HTS is obvious, its recognition did not take place without effort. It was with some skepticism, and under-preparedness, that the world welcomed HTS in the mid-1980s. In addition to the drug discovery process per se, the driving forces underlying HTS embraced, and revolutionized, diverse disciplines of biology (e.g., DNA sequencing, digital microscopy, protein crystallization). On the other hand, some of its limitations remained large obstacles. The technology has remained relatively cumbersome and rigid in some ways, limiting the number of compounds that can be tested (1 million compounds require 1 million wells) and the size/number of screening campaigns that can be accomplished. HTS libraries are typically smaller than 1 million compounds and are often over-used by the drug industry and academia. The target source, often E. coli, results in products that are often not optimal (e.g., folding, PTMs). Large amounts of protein targets and/or cells compatible with 105 wells are needed, necessitating costly steps, expensive equipment, and logistical requirements, as well as extensive infrastructure. The growing number of therapeutic targets combined with HTS limitations are driving the need for more screening power. This is how combinatorial chemistry erupted a few years after HTS; however, because of the lack of addressability, the application of combinatorial chemistry did not meet its expectations. As a result, the DNA-encoded library (DEL) technology was born, burgeoning from the combinatorial chemistry field. DEL technology, a concept proposed by Drs Richard Lerner and Sidney Brenner, was based on their proof of concept published in 1992 that aimed to solve 'addressability' by adding a unique DNA tag sequence to each drug-like molecule, thus allowing every molecule to be identified at the end of a screening campaign. DNA synthesis capabilities were very limited in the early 1990s, and large-scale sequencing had yet to be invented. The combination of a constantly decreasing cost of DNA synthesis and the emergence of next-generation DNA sequencing ('massively parallel sequencing'; this emerged in the 1990s and was first commercialized in 2005) led to the birth of DEL technology in the mid-2000s. If Pfizer has been associated with the birth of HTS, Drs Lerner and Brenner will certainly be associated with the birth of HPS, begetting it with their initial proof of principle in 1992, and being instrumental in its development since then; Dr Lerner remains at the forefront of the field. HTS has some strong advantages and will continue to be highly valuable for the drug discovery industry. HTS libraries of compounds can be highly curated libraries (size, functionalities, chemical space). HTS includes functional screening, often in live cells, and library noise is often well established. However, HTS also has several limitations that HPS can address: (i) because of heavy and highly specialized automation, robot maintenance, and a large real-estate footprint, the high price of HTS reflects the cost of building a library [HTS, 1 billion dollars for 1 million compounds ($1000 per compound); compared to HPS, <1 million dollars for 1 billion compounds for DEL (<1 cent per DEL compound)] and for the cost of using it (HTS, 0.05 to 1 dollars per well; HPS, negligible on a per-compound basis); (ii) the relatively limited number of molecules that can be reasonably tested in a single screening campaign (typically a hundred thousand to a million), the inconveniency of introducing new compounds and pharmacophores into an existing library (need for relatively pure compounds), and the limited number of HTS compounds (a total of 16 million in all HTS libraries) indicate that the same compound libraries are used over and over; (iii) an overwhelmingly large fraction of HTS compounds are constrained by the Lipinski rule of five and are therefore not adapted to targeting larger areas such as protein–protein interactions; (iv) the format in multi-well plates necessitates large amounts of the target, thus increasing the cost and the constraints on target purification/origin [HTS requires Escherichia coli recombinants and a few micrograms per compound (500 mg per million compounds), whereas HPS requires only 25 micrograms comprising 1 billion compounds]; (v) local contamination of stock plates from reusing the same library is another significant drawback and requires compound/library maintenance (occasional liquid chromatography/mass spectrometry verification and well replenishment); (vi) a significant number of colored compounds and dyes interfere with various types of assays leading to higher false positive rates; and (vii) cellular assays represent a major improvement for HTS, but limitations persist and the constrained access and scalability of primary cultures and stem cells impedes their use for HTS. These limitations alone or in combination lead to overall low data quality. The two synergistic advantages of HPS that are new compared to HTS, and that differentiate between the two by several orders of magnitude, are the extra-large sizes of the libraries, reflecting their combinatorial nature and modifications that allow extremely efficient drug screening. For DEL technology, the DNA-tagging system allows very large numbers of compounds to coexist in a single tube and brings an entirely new set of screening prospects not seen before. The father of HPS, Dr Richard Lerner, was of primary and tremendous importance regarding phage display screening and perhaps even more for DEL technologies. He coinvented the DEL concept together with the late Dr Sidney Brenner about 30 years ago, long before DNA synthesis was mainstream and about 15 years before next-generation sequencing (NGS) was invented. The libraries built/screens performed and the therapeutic targets used are summarized in Table 1 and Figure 1A , respectively. Table 1 shows the great diversity in terms of library size, with the disappearance over the past few years of excessively large libraries (hundreds of billions and trillions), demonstrating some level of adaptability and evolution of the field. HPS is well positioned to significantly and positively shake up the drug discovery process and could do so in several ways that we review in the following sections.Table 1Studies published describing the construction of DNA-encoded libraries and DEL screening campaignsaAbbreviations: B, billion; BMS, Bristol-Myers Squibb; ETH, Eidgenössische Technische Hochschule; GSK, GlaxoSmithKline; M, million; N.S., not specified; T, trillion., bHPS library sizes: categories 2XS through to 4XL are defined in Figure 1; sizes are color-coded from extra-small to medium (black), large (green), to extra-large (orange and red). [1.Halpin D.R. Harbury P.B. DNA display II. Genetic manipulation of combinatorial chemistry libraries for small-molecule evolution.PLoS Biol. 2004; 2: 1022-1030Crossref Scopus (115) Google Scholar, 2.Gartner Z.J. et al.DNA-templated organic synthesis and selection of a library of macrocycles.Science. 2004; 305: 1601-1605Crossref PubMed Scopus (376) Google Scholar, 3.Melkko S. et al.Encoded self-assembling chemical libraries.Nat. Biotechnol. 2004; 22: 568-574Crossref PubMed Scopus (266) Google Scholar, 4.Melkko S. et al.Isolation of high-affinity trypsin inhibitors from a DNA-encoded chemical library.Angew. Chem. Int. Ed. Engl. 2007; 46: 4671-4674Crossref PubMed Scopus (86) Google Scholar, 5.Wrenn S.J. et al.Synthetic ligands discovered by in vitro selection.J. Am. Chem. Soc. 2007; 129: 13137-13143Crossref PubMed Scopus (126) Google Scholar, 6.Mannocci L. et al.High-throughput sequencing allows the identification of binding molecules isolated from DNA-encoded chemical libraries.Proc. Natl. Acad. Sci. U. S. A. 2008; 105: 17670-17675Crossref PubMed Scopus (159) Google Scholar, 7.Dumelin C.E. et al.A portable albumin binder from a DNA-encoded chemical library.Angew. Chem. Int. Ed. Engl. 2008; 47: 3196-3201Crossref PubMed Scopus (154) Google Scholar, 8.Scheuermann J. et al.DNA-encoded chemical libraries for the discovery of MMP-3 inhibitors.Bioconjug. Chem. 2008; 19: 778-785Crossref PubMed Scopus (70) Google Scholar, 9.Clark M.A. et al.Design, synthesis and selection of DNA-encoded small-molecule libraries.Nat. Chem. Biol. 2009; 5: 647-654Crossref PubMed Scopus (450) Google Scholar, 10.Hansen M.H. et al.A yoctoliter-scale DNA reactor for small-molecule evolution.J. Am. Chem. Soc. 2009; 131: 1322-1327Crossref PubMed Scopus (152) Google Scholar, 11.Buller F. et al.Discovery of TNF inhibitors from a DNA-encoded chemical library based on diels-alder cycloaddition.Chem. Biol. 2009; 16: 1075-1086Abstract Full Text Full Text PDF PubMed Scopus (87) Google Scholar, 12.Kleiner R.E. et al.In vitro selection of a DNA-templated small-molecule library reveals a class of macrocyclic kinase inhibitors.J. Am. Chem. Soc. 2010; 132: 11779-11791Crossref PubMed Scopus (122) Google Scholar, 13.Melkko S. et al.Isolation of a small-molecule inhibitor of the antiapoptotic protein Bcl-xL from a DNA-encoded chemical library.ChemMedChem. 2010; 5: 584-590Crossref PubMed Scopus (48) Google Scholar, 14.Mannocci L. et al.Isolation of potent and specific trypsin inhibitors from a DNA-encoded chemical library.Bioconjug. Chem. 2010; 21: 1836-1841Crossref PubMed Scopus (40) Google Scholar, 15.Buller F. et al.Selection of carbonic anhydrase IX inhibitors from one million DNA-encoded compounds.ACS Chem. Biol. 2011; 6: 336-344Crossref PubMed Scopus (113) Google Scholar, 16.Deng H. et al.Discovery of highly potent and selective small molecule ADAMTS-5 inhibitors that inhibit human cartilage degradation via encoded library technology (ELT).J. Med. Chem. 2012; 55: 7061-7079Crossref PubMed Scopus (114) Google Scholar, 17.Leimbacher M. et al.Discovery of small-molecule interleukin-2 inhibitors from a DNA-encoded chemical library.Chemistry. 2012; 18: 7729-7737Crossref PubMed Scopus (87) Google Scholar, 18.Disch J.S. et al.Discovery of thieno[3,2-d]pyrimidine-6-carboxamides as potent inhibitors of SIRT1, SIRT2, and SIRT3.J. Med. Chem. 2013; 56: 3666-3679Crossref PubMed Scopus (151) Google Scholar, 19.Podolin P.L. et al.In vitro and in vivo characterization of a novel soluble epoxide hydrolase inhibitor.Prostaglandins Other Lipid. Mediat. 2013; 104-105: 25-31Crossref PubMed Scopus (86) Google Scholar, 20.Thalji R.K. et al.Discovery of 1-(1,3,5-triazin-2-yl)piperidine-4-carboxamides as inhibitors of soluble epoxide hydrolase.Bioorg. Med. Chem. Lett. 2013; 23: 3584-3588Crossref PubMed Scopus (50) Google Scholar, 21.Encinas L. et al.Encoded library technology as a source of hits for the discovery and lead optimization of a potent and selective class of bactericidal direct inhibitors of Mycobacterium tuberculosis InhA.J. Med. Chem. 2014; 57: 1276-1288Crossref PubMed Scopus (96) Google Scholar, 22.Kollmann C.S. et al.Application of encoded library technology (ELT) to a protein–protein interaction target: discovery of a potent class of integrin lymphocyte function-associated antigen 1 (LFA-1) antagonists.Bioorg. Med. Chem. 2014; 22: 2353-2365Crossref PubMed Scopus (77) Google Scholar, 23.Franzini R.M. et al.Identification of structure–activity relationships from screening a structurally compact DNA-encoded chemical library.Angew Chem. Int. Ed. Engl. 2015; 54: 3927-3931Crossref PubMed Scopus (77) Google Scholar, 24.Daguer J.P. et al.DNA display of fragment pairs as a tool for the discovery of novel biologically active small molecules.Chem. Sci. 2015; 6: 739-744Crossref PubMed Google Scholar, 25.Wichert M. et al.Dual-display of small molecules enables the discovery of ligand pairs and facilitates affinity maturation.Nat. Chem. 2015; 7: 241-249Crossref PubMed Scopus (145) Google Scholar, 26.Yang H. et al.Discovery of a potent class of PI3Kalpha inhibitors with unique binding mode via encoded library technology (ELT).ACS Med. Chem. Lett. 2015; 6: 531-536Crossref PubMed Scopus (55) Google Scholar, 27.Arico-Muendel C. et al.Encoded library technology screening of hepatitis C virus NS4B yields a small-molecule compound series with in vitro replicon activity.Antimicrob. Agents Chemother. 2015; 59: 3450-3459Crossref PubMed Scopus (25) Google Scholar, 28.Seigal B.A. et al.The discovery of macrocyclic XIAP antagonists from a DNA-programmed chemistry library, and their optimization to give lead compounds with in vivo antitumor activity.J. Med. Chem. 2015; 58: 2855-2861Crossref PubMed Scopus (68) Google Scholar, 29.Wu Z. et al.Cell-based selection expands the utility of DNA-encoded small-molecule library technology to cell surface drug targets: identification of novel antagonists of the NK3 tachykinin receptor.ACS Comb. Sci. 2015; 17: 722-731Crossref PubMed Scopus (85) Google Scholar, 30.Litovchick A. et al.Encoded library synthesis using chemical ligation and the discovery of sEH inhibitors from a 334-million member library.Sci. Rep. 2015; 5: 10916Crossref PubMed Scopus (82) Google Scholar, 31.Deng H. et al.Discovery, SAR, and X-ray binding mode study of BCATm inhibitors from a novel DNA-encoded library.ACS Med. Chem. Lett. 2015; 6: 919-924Crossref PubMed Scopus (63) Google Scholar, 32.Ding Y. et al.Discovery of potent and selective inhibitors for ADAMTS-4 through DNA-encoded library technology (ELT).ACS Med. Chem. Lett. 2015; 6: 888-893Crossref PubMed Scopus (61) Google Scholar, 33.Samain F. et al.Tankyrase 1 inhibitors with drug-like properties identified by screening a DNA-encoded chemical library.J. Med. Chem. 2015; 58: 5143-5149Crossref PubMed Scopus (56) Google Scholar, 34.Harris P.A. et al.DNA-encoded library screening identifies benzo[b][1,4]oxazepin-4-ones as highly potent and monoselective receptor interacting protein 1 kinase inhibitors.J. Med. Chem. 2016; 59: 2163-2178Crossref PubMed Scopus (170) Google Scholar, 35.Petersen L.K. et al.Novel p38 alpha MAP kinase inhibitors identified from yoctoReactor DNA-encoded small molecule library.Medchemcomm. 2016; 7: 1332-1339Crossref Google Scholar, 36.Soutter H.H. et al.Discovery of cofactor-specific, bactericidal Mycobacterium tuberculosis InhA inhibitors using DNA-encoded library technology.Proc. Natl. Acad. Sci. U. S. A. 2016; 113: E7880-E7889Crossref PubMed Scopus (39) Google Scholar, 37.Belyanskaya S.L. et al.Discovering drugs with DNA-encoded library technology: from concept to clinic with an inhibitor of soluble epoxide hydrolase.Chembiochem. 2017; 18: 837-842Crossref PubMed Scopus (118) Google Scholar, 38.Cuozzo J.W. et al.Discovery of a potent BTK inhibitor with a novel binding mode by using parallel selections with a DNA-encoded chemical library.Chembiochem. 2017; 18: 864-871Crossref PubMed Scopus (49) Google Scholar, 39.Ahn S. et al.Allosteric 'beta-blocker' isolated from a DNA-encoded small molecule library.Proc. Natl. Acad. Sci. U. S. A. 2017; 114: 1708-1713Crossref PubMed Scopus (88) Google Scholar, 40.Fernandez-Montalvan A.E. et al.Isoform-selective ATAD2 chemical probe with novel chemical structure and unusual mode of action.ACS Chem. Biol. 2017; 12: 2730-2736Crossref PubMed Scopus (53) Google Scholar, 41.Chan A.I. et al.Discovery of a covalent kinase inhibitor from a DNA-encoded small-molecule library x protein library selection.J. Am. Chem. Soc. 2017; 139: 10192-10195Crossref PubMed Scopus (51) Google Scholar, 42.Mendes K.R. et al.High-throughput identification of DNA-encoded IgG ligands that distinguish active and latent Mycobacterium tuberculosis infections.ACS Chem. Biol. 2017; 12: 234-243Crossref PubMed Scopus (46) Google Scholar, 43.Zimmermann G. et al.A specific and covalent JNK-1 ligand selected from an encoded self-assembling chemical library.Chemistry. 2017; 23: 8152-8155Crossref PubMed Scopus (45) Google Scholar, 44.Zhu Z. et al.Design and application of a DNA-encoded macrocyclic peptide library.ACS Chem. Biol. 2018; 13: 53-59Crossref PubMed Scopus (63) Google Scholar, 45.Brown D.G. et al.Agonists and antagonists of protease-activated receptor 2 discovered within a DNA-encoded chemical library using mutational stabilization of the target.SLAS Discov. 2018; 23: 429-436PubMed Google Scholar, 46.Usanov D.L. et al.Second-generation DNA-templated macrocycle libraries for the discovery of bioactive small molecules.Nat. Chem. 2018; 10: 704-714Crossref PubMed Scopus (110) Google Scholar, 47.Ahn S. et al.Small-molecule positive allosteric modulators of the beta2-adrenoceptor isolated from DNA-encoded libraries.Mol. Pharmacol. 2018; 94: 850-861Crossref PubMed Scopus (40) Google Scholar, 48.Zhou Y. et al.DNA-encoded dynamic chemical library and its applications in ligand discovery.J. Am. Chem. Soc. 2018; 140: 15859-15867Crossref PubMed Scopus (61) Google Scholar, 49.Litovchick A. et al.Novel nucleic acid binding small molecules discovered using DNA-encoded chemistry.Molecules. 2019; 24: 2026Crossref Scopus (19) Google Scholar, 50.Li J.Y. et al.Palladium-catalyzed hydroxycarbonylation of (hetero)aryl halides for DNA-encoded chemical library synthesis.Bioconjug. Chem. 2019; 30: 2209-2215Crossref PubMed Scopus (20) Google Scholar, 51.Cochrane W.G. et al.Activity-based DNA-encoded library screening.ACS Comb. Sci. 2019; 21: 425-435Crossref PubMed Scopus (36) Google Scholar, 52.Shin M.H. et al.DNA-encoded combinatorial library of macrocyclic peptoids.Bioconjug. Chem. 2019; 30: 2931-2938Crossref PubMed Scopus (25) Google Scholar, 53.Yuen L.H. et al.A focused DNA-encoded chemical library for the discovery of inhibitors of NAD+-dependent enzymes.J. Am. Chem. Soc. 2019; 141: 5169-5181Crossref PubMed Scopus (63) Google Scholar, 54.Kim D. et al.Application of a substrate-mediated selection with c-Src tyrosine kinase to a DNA-encoded chemical library.Molecules. 2019; 24Crossref Scopus (7) Google Scholar, 55.Wang S. et al.Optimization of ligands using focused DNA-encoded libraries to develop a selective, cell-permeable CBX8 chromodomain inhibitor.ACS Chem. Biol. 2019; 15: 112-131Crossref PubMed Scopus (36) Google Scholar, 56.Hackler A. et al.Off-DNA DNA-encoded library affinity screening.ACS Comb. Sci. 2019; 22: 25-34Crossref PubMed Scopus (19) Google Scholar, 57.Valastyan J.S. et al.Discovery of PqsE thioesterase inhibitors for pseudomonas aeruginosa using DNA-encoded small molecule library screening.ACS Chem. Biol. 2020; 15: 446-456Crossref PubMed Scopus (18) Google Scholar, 58.Sannino A. et al.Critical evaluation of photo-cross-linking parameters for the implementation of efficient DNA-encoded chemical library selections.ACS Comb. Sci. 2020; 22: 204-212Crossref PubMed Scopus (21) Google Scholar, 59.Taylor D.M. et al.Identifying oxacillinase-48 carbapenemase inhibitors using DNA-encoded chemical libraries.ACS Infect. Dis. 2020; 6: 1214-1227Crossref PubMed Scopus (19) Google Scholar, 60.Su L. et al.Synthesis of multifunctional 2-aminobenzimidazoles on DNA via iodine-promoted cyclization.Org. Lett. 2020; 22: 1290-1294Crossref PubMed Scopus (18) Google Scholar, 61.Chen Y.C. et al.C–N coupling of DNA-conjugated (hetero)aryl bromides and chlorides for DNA-encoded chemical library synthesis.Bioconjug. Chem. 2020; 31: 770-780Crossref PubMed Scopus (28) Google Scholar, 62.Rectenwald J.M. et al.Design and construction of a focused DNA-encoded library for multivalent chromatin reader proteins.Molecules. 2020; 25: 979Crossref Scopus (7) Google Scholar, 63.Bassi G. et al.A single-stranded DNA-encoded chemical library based on a stereoisomeric scaffold enables ligand discovery by modular assembly of building blocks.Adv. Sci. (Weinh). 2020; 72001970Crossref PubMed Scopus (15) Google Scholar, 64.Kung P.P. et al.Characterization of specific N-alpha-acetyltransferase 50 (Naa50) inhibitors identified using a DNA encoded library.ACS Med. Chem. Lett. 2020; 11: 1175-1184Crossref PubMed Scopus (15) Google Scholar, 65.Dawadi S. et al.Discovery of potent thrombin inhibitors from a protease-focused DNA-encoded chemical library.Proc. Natl. Acad. Sci. U. S. A. 2020; 117: 16782-16789Crossref PubMed Scopus (27) Google Scholar, 66.McEnaney P. et al.Structural characterization of a peptoid-inspired conformationally constrained oligomer (PICCO) bound to streptavidin.Chem. Commun. (Camb.). 2020; 56: 10560-10563Crossref PubMed Google Scholar, 67.Cuozzo J.W. et al.Novel autotaxin inhibitor for the treatment of idiopathic pulmonary fibrosis: a clinical candidate discovered using DNA-encoded chemistry.J. Med. Chem. 2020; 63: 7840-7856Crossref PubMed Scopus (40) Google Scholar, 68.Kunig V.B.K. et al.TEAD–YAP interaction inhibitors and MDM2 binders from DNA-encoded indole-focused Ugi peptidomimetics.Angew Chem. Int. Ed. Engl. 2020; 59: 20338-20342Crossref PubMed Scopus (33) Google Scholar, 69.Favalli N. et al.Stereo- and regiodefined DNA-encoded chemical libraries enable efficient tumour-targeting applications.Nat. Chem. 2021; 13: 540-548Crossref PubMed Scopus (24) Google Scholar, 70.Deng Y. et al.Selection of DNA-encoded dynamic chemical libraries for direct inhibitor discovery.Angew Chem. Int. Ed. Engl. 2020; 59: 14965-14972Crossref PubMed Scopus (27) Google Scholar, 71.Catalano M. et al.Selective fragments for the CREBBP bromodomain identified from an encoded self-assembly chemical library.ChemMedChem. 2020; 15: 1752-1756Crossref PubMed Scopus (8) Google Scholar, 72.Veerman J.J.N. et al.Discovery of 2,4-1H-imidazole carboxamides as potent and selective TAK1 inhibitors.ACS Med. Chem. Lett. 2021; 12: 555-562Crossref PubMed Scopus (5) Google Scholar, 73.Onda Y. et al.A DNA-encoded chemical library based on peptide macrocycles.Chemistry. 2021; 27: 7160-7167Crossref PubMed Scopus (13) Google Scholar, 74.Wang X. et al.Diversified strategy for the synthesis of DNA-encoded oxindole libraries.Chem. Sci. 2021; 12: 2841-2847Crossref PubMed Google Scholar, 75.Ding Y. et al.Discovery of soluble epoxide hydrolase inhibitors through DNA-encoded library technology (ELT).Bioorg. Med. Chem. 2021; 41116216Crossref PubMed Scopus (7) Google Scholar, 76.Yang P. et al.Streamlined construction of peptide macrocycles via palladium-catalyzed intramolecular S-arylation in solution and on DNA.Chem. Sci. 2021; 12: 5804-5810Crossref PubMed Google Scholar, 77.Liang Q. et al.Selective discovery of GPCR ligands within DNA-encoded chemical libraries derived from natural products: a case study on antagonists of angiotensin ii type i receptor.J. Med. Chem. 2021; 64: 4196-4205Crossref PubMed Scopus (8) Google Scholar]a Abbreviations: B, billion; BMS, Bristol-Myers Squibb; ETH, Eidgenössische Technische Hochschule; GSK, GlaxoSmithKline; M, million; N.S., not specified; T, trillion.b HPS library sizes: categories 2XS through to 4XL are defined in Figure 1; sizes are color-coded from extra-small to mediu
最长约 10秒,即可获得该文献文件

科研通智能强力驱动
Strongly Powered by AbleSci AI
更新
大幅提高文件上传限制,最高150M (2024-4-1)

科研通是完全免费的文献互助平台,具备全网最快的应助速度,最高的求助完成率。 对每一个文献求助,科研通都将尽心尽力,给求助人一个满意的交代。
实时播报
Yuzuru_gyq完成签到 ,获得积分10
1秒前
老木虫发布了新的文献求助10
2秒前
2秒前
vrai发布了新的文献求助10
3秒前
本是个江湖散人完成签到,获得积分10
5秒前
6秒前
夏侯夏侯完成签到 ,获得积分10
7秒前
icaohao完成签到,获得积分10
8秒前
gzzzzz完成签到,获得积分10
9秒前
好玩和有趣完成签到,获得积分10
9秒前
9秒前
学呀学发布了新的文献求助10
10秒前
10秒前
vrai完成签到,获得积分10
10秒前
CC完成签到,获得积分10
12秒前
doctorw完成签到 ,获得积分10
12秒前
今后应助老木虫采纳,获得10
13秒前
冷冷完成签到 ,获得积分10
14秒前
自觉山柏发布了新的文献求助30
14秒前
lee1984612完成签到,获得积分10
15秒前
七月流子完成签到,获得积分10
15秒前
小马甲应助MaFY采纳,获得10
16秒前
东东呀发布了新的文献求助10
16秒前
忧心的碧完成签到,获得积分20
16秒前
满意的蜜蜂完成签到,获得积分10
18秒前
luanyi1011发布了新的文献求助20
19秒前
朝阳完成签到 ,获得积分10
19秒前
秋雪瑶应助Susan采纳,获得10
21秒前
徐昊完成签到,获得积分10
21秒前
Lucas应助自然棒棒糖采纳,获得10
23秒前
现代乌龟完成签到,获得积分10
23秒前
海森堡完成签到,获得积分10
25秒前
我是科研垃圾完成签到,获得积分10
25秒前
兔子先生完成签到 ,获得积分10
25秒前
SCINEXUS应助yue957采纳,获得10
25秒前
脑洞疼应助huihui采纳,获得10
26秒前
FashionBoy应助流光采纳,获得10
27秒前
可爱的函函应助jeronimo采纳,获得10
29秒前
30秒前
31秒前
高分求助中
Teaching Social and Emotional Learning in Physical Education 900
Plesiosaur extinction cycles; events that mark the beginning, middle and end of the Cretaceous 800
Recherches Ethnographiques sue les Yao dans la Chine du Sud 500
Two-sample Mendelian randomization analysis reveals causal relationships between blood lipids and venous thromboembolism 500
Chinese-English Translation Lexicon Version 3.0 500
Wisdom, Gods and Literature Studies in Assyriology in Honour of W. G. Lambert 400
薩提亞模式團體方案對青年情侶輔導效果之研究 400
热门求助领域 (近24小时)
化学 材料科学 医学 生物 有机化学 工程类 生物化学 纳米技术 物理 内科学 计算机科学 化学工程 复合材料 遗传学 基因 物理化学 催化作用 电极 光电子学 量子力学
热门帖子
关注 科研通微信公众号,转发送积分 2392033
求助须知:如何正确求助?哪些是违规求助? 2096714
关于积分的说明 5282358
捐赠科研通 1824242
什么是DOI,文献DOI怎么找? 909820
版权声明 559877
科研通“疑难数据库(出版商)”最低求助积分说明 486170