We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

The maturation of DNA encoded libraries: opportunities for new users

    Daniel Conole

    *Author for correspondence:

    E-mail Address: conoledaniel@gmail.com

    Department of Chemistry, Molecular Sciences Research Hub, Imperial College London, White City Campus, 80 Wood Lane, London, W12 0BZ, UK

    ,
    James H Hunter

    Cancer Research UK Drug Discovery Unit, Newcastle University Centre for Cancer, Chemistry, School of Natural & Environmental Sciences, Bedson Building, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK

    &
    Michael J Waring

    Cancer Research UK Drug Discovery Unit, Newcastle University Centre for Cancer, Chemistry, School of Natural & Environmental Sciences, Bedson Building, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK

    Published Online:https://doi.org/10.4155/fmc-2020-0285

    DNA-encoded combinatorial libraries (DECLs) represent an exciting new technology for high-throughput screening, significantly increasing its capacity and cost–effectiveness. Historically, DECLs have been the domain of specialized academic groups and industry; however, there has recently been a shift toward more drug discovery academic centers and institutes adopting this technology. Key to this development has been the simplification, characterization and standardization of various DECL subprotocols, such as library design, affinity screening and data analysis of hits. This review examines the feasibility of implementing DECL screening technology as a first-time user, particularly in academia, exploring the some important considerations for this, and outlines some applications of the technology that academia could contribute to the field.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Anzalone AV, Koblan LW, Liu DR. Genome editing with CRISPR–Cas nucleases, base editors, transposases and prime editors. Nat. Biotechnol. 38, 824–844 (2020).
    • 2. Callaway E. Molecular-imaging pioneers scoop Nobel. Nature. 550(7675), 167 (2017).
    • 3. Goodnow RA, Dumelin CE, Keefe AD. DNA-encoded chemistry: enabling the deeper sampling of chemical space. Nat. Rev. Drug Discov. 16, 131 (2017).
    • 4. Brenner S, Lerner RA. Encoded combinatorial chemistry. Proc. Natl. Acad. Sci. U. S. A. 89(12), 5381–5383 (1992).
    • 5. Thalji RK, McAtee JJ, Belyanskaya S et al. Discovery of 1-(1,3,5-triazin-2-yl)piperidine-4-carboxamides as inhibitors of soluble epoxide hydrolase. Bioorganic Med. Chem. Lett. 23(12), 3584–3588 (2013).
    • 6. Harris PA, King BW, Bandyopadhyay D 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. 59(5), 2163–2178 (2016).
    • 7. Belyanskaya SL, Ding Y, Callahan JF et al. Discovering drugs with DNA-encoded library technology: from concept to clinic with an inhibitor of soluble epoxide hydrolase. ChemBioChem. 18(9), 837–842 (2017). •• A powerful example of DECL technology leading to a clinical drug candidate.
    • 8. Yuen LH, Franzini RM. Achievements, challenges, and opportunities in DNA-encoded library research: an academic point of view. ChemBioChem. 18(9), 829–836 (2017).
    • 9. Bell C. Adopting new technology: barriers and breakthroughs. Pharm. Exec. 39(5), (2019). https://www.pharmexec.com/view/adopting-new-technology-barriers-and-breakthroughs
    • 10. Flood DT, Kingston C, Vantourout JC et al. DNA encoded libraries: a visitor's guide. 60, 268–280 (2020).
    • 11. Gerry CJ, Schreiber SL. Recent achievements and current trajectories of diversity-oriented synthesis. Curr. Opin. Chem. Biol. 56, 1–9 (2020).
    • 12. Song M, Hwang GT. DNA-encoded library screening as core platform technology in drug discovery: its synthetic method development and applications in DEL synthesis. J. Med. Chem. 63(13), 6578–6599 (2020).
    • 13. Wrenn SJ, Weisinger RM, Halpin DR, Harbury PB. Synthetic ligands discovered by in vitro selection. J. Am. Chem. Soc. 129(43), 13137–13143 (2007).
    • 14. Halpin DR, Harbury PB. DNA display II. Genetic manipulation of combinatorial chemistry libraries for small-molecule evolution. PLoS Biol. 2(7), (2004).
    • 15. Gartner ZJ, Tse BN, Grubina R et al. DNA-templated organic synthesis and selection of a library of macrocycles. Science (80-.). 305(5690), 1601–1605 (2004).
    • 16. Melkko S, Scheuermann J, Dumelin CE, Neri D. Encoded self-assembling chemical libraries. Nat. Biotechnol. 22(5), 568–574 (2004).
    • 17. Hansen MH, Blakskjaer P, Petersen LK et al. A yoctoliter-scale DNA reactor for small-molecule evolution. J. Am. Chem. Soc. 131(3), 1322–1327 (2009).
    • 18. Bassi G, Favalli N, Oehler S et al. Comparative evaluation of DNA-encoded chemical selections performed using DNA in single-stranded or double-stranded format. Biochem. Biophys. Res. Commun. (2020).
    • 19. Lipinski CA, Lombardo F, Dominy BW, Feeney PJ. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Deliv. Rev. 23, 3–25 (1997).
    • 20. Franzini RM, Randolph C. Chemical space of DNA-encoded libraries: Miniperspective. J. Med. Chem. 59(14), 6629–6644 (2016).
    • 21. Madsen D, Azevedo C, Micco I et al. Chapter Four – an overview of DNA-encoded libraries: a versatile tool for drug discovery. In: Progress in Medicinal Chemistry. Witty DRCox B (Eds). Elsevier, 181–249 (2020).
    • 22. Clark MA, Acharya RA, Arico-Muendel CC et al. Design, synthesis and selection of DNA-encoded small-molecule libraries. Nat. Chem. Biol. 5(9), 647–654 (2009).
    • 23. Decurtins W, Wichert M, Franzini RM et al. Automated screening for small organic ligands using DNA-encoded chemical libraries. Nat. Protoc. 11(4), 764–780 (2016). •• A recent detailed protocol of how to perform an affinity screen of a DECL, with specific details to run the quantification of the sequencing data also.
    • 24. Satz AL. Foundations of a DNA-encoded library (DEL). In: A Handbook for DNA-Encoded Chemistry. Goodnow RA Jr (Ed.). Wiley, 99–121 (2014).
    • 25. Leach AR, Hann MM. Molecular complexity and fragment-based drug discovery: ten years on. Curr. Opin. Chem. Biol. 15(4), 489–496 (2011).
    • 26. Hann MM, Leach AR, Harper G. Molecular complexity and its impact on the probability of finding leads for drug discovery. J. Chem. Inf. Comput. Sci. 41(3), 856–864 (2001).
    • 27. Muresan S, Sadowski J. Properties guiding drug- and lead-likeness. Mol. Drug Prop. Chapter 17, 439–461 (2007). https://onlinelibrary.wiley.com/doi/abs/10.1002/9783527621286.ch17
    • 28. Teague SJ, Davis AM, Leeson PD, Oprea T. The design of leadlike combinatorial libraries. Angew. Chemie Int. Ed. 38(24), 3743–3748 (1999).
    • 29. Satz AL. What do you get from DNA-encoded libraries? ACS Med. Chem. Lett. 8(5), 408–410 (2018).
    • 30. Harris PA, Berger SB, Jeong JU et al. Discovery of a first-in-class receptor interacting protein 1 (RIP1) kinase specific clinical candidate (GSK2982772) for the treatment of inflammatory diseases. J. Med. Chem. 60(4), 1247–1261 (2017).
    • 31. Cuozzo JW, Clark MA, Keefe AD et al. Novel autotaxin inhibitor for the treatment of idiopathic pulmonary fibrosis: a clinical candidate discovered using DNA-encoded chemistry. J. Med. Chem. 63(14), 7840–7856 (2020).
    • 32. Favalli N, Biendl S, Hartmann M et al. A DNA-encoded library of chemical compounds based on common scaffolding structures reveals the impact of ligand geometry on protein recognition. ChemMedChem. 13(13), 1303–1307 (2018).
    • 33. Kunig VBK, Potowski M, Akbarzadeh M et al. TEAD-YAP interaction inhibitors and MDM2 binders from DNA-encoded indole-focused Ugi-peptidomimetics. Angew. Chemie Int. Ed. 59, 20338–20342 (2020).
    • 34. Melkko S, Dumelin CE, Scheuermann J, Neri D. On the magnitude of the chelate effect for the recognition of proteins by pharmacophores scaffolded by self-assembling oligonucleotides. Chem. Biol. 13(2), 225–231 (2006).
    • 35. Wichert M, Krall N, Decurtins W et al. Dual-display of small molecules enables the discovery of ligand pairs and facilitates affinity maturation. Nat. Chem. 7(3), 241–249 (2015).
    • 36. Dawadi S, Simmons N, Miklossy G et al. Discovery of potent thrombin inhibitors from a protease-focused DNA-encoded chemical library. Proc. Natl Acad. Sci. USA 117(29), 16782–16789 (2020).
    • 37. Yuen LH, Dana S, Liu Y et al. A focused DNA-encoded chemical library for the discovery of inhibitors of NAD+-dependent enzymes. J. Am. Chem. Soc. 141(13), 5169–5181 (2019).
    • 38. Rectenwald JM, Guduru SKR, Dang Z et al. Design and construction of a focused DNA-encoded library for multivalent chromatin reader proteins. Molecules. 25(4), 1–12 (2020).
    • 39. Wang S, Denton KE, Hobbs KF et al. Optimization of ligands using focused DNA-encoded libraries to develop a selective, cell-permeable CBX8 chromodomain inhibitor. ACS Chem. Biol. 15(1), 112–131 (2020).
    • 40. Denton KE, Wang S, Gignac MC et al. Robustness of in vitro selection assays of DNA-encoded peptidomimetic ligands to CBX7 and CBX8. SLAS Discov. 23(5), 417–428 (2018).
    • 41. Catalano M, Moroglu M, Balbi P et al. Selective fragments for the CREBBP bromodomain identified from an encoded self-assembly chemical library. Chem Med Chem. doi: 10.1002/cmdc.202000528 (2020).
    • 42. Deng Y, Peng J, Xiong F et al. Selection of DNA-encoded dynamic chemical libraries for direct inhibitor discovery. Angew. Chemie – Int. Ed. 59, 14965–14972 (2020).
    • 43. Bosc D, Jakhlal J, Deprez B, Deprez-Poulain R. Kinetic target-guided synthesis in drug discovery and chemical biology: a comprehensive facts and figures survey. Future Med. Chem. 8(4), 381–404 (2016).
    • 44. Gerry CJ, Wawer MJ, Clemons PA, Schreiber SL. DNA barcoding a complete matrix of stereoisomeric small molecules. J. Am. Chem. Soc. 141(26), 10225–10235 (2019).
    • 45. Maetani M, Zoller J, Melillo B et al. Synthesis of a bicyclic azetidine with in vivo antimalarial activity enabled by stereospecific, directed C(sp3)-H arylation. J. Am. Chem. Soc. 139(32), 11300–11306 (2017).
    • 46. Drewry DH, Stanley Young S. Approaches to the design of combinatorial libraries. Chemom. Intell. Lab. Syst. 48(1), 1–20 (1999).
    • 47. Zhu H, Flanagan ME, Stanton RV. Designing DNA encoded libraries of diverse products in a focused property space. J. Chem. Inf. Model. 59(11), 4645–4653 (2019).
    • 48. Waldmann H, Valeur E, Guéret SM et al. New modalities for challenging targets in drug discovery. Angew. Chem. Int. Ed. Angew. Chem. 56, 10294–10323 (2016).
    • 49. Valeur E, Jimonet P. Lead generation: enabling a mode-of-action centric paradigm new modalities, technologies, and partnerships in probe and lead generation: enabling a mode-of-action centric paradigm. J. Med. Chem. 60(21), 9004–9029 (2018).
    • 50. Zhu Z. Shaginian A, Grady LSC et al. Cyclic peptide design. In: Methods in Molecular Biology 2001. Goetz G (Ed.). Humana Press, Springer Nature, NY, USA, 273–284 (2019).
    • 51. Usanov DL, Chan AI, Maianti JP, Liu DR. Second-generation DNA-templated macrocycle libraries for the discovery of bioactive small molecules. Nat. Chem. 10(7), 704–714 (2018).
    • 52. Shin MH, Lee KJ, Lim HS. DNA-encoded combinatorial library of macrocyclic peptoids. Bioconjug. Chem. 30(11), 2931–2938 (2019).
    • 53. Ma P, Xu H, Li J et al. Functionality-independent DNA encoding of complex natural products. Angew. Chemie – Int. Ed. 58(27), 9254–9261 (2019).
    • 54. Baell J, Walters MA. Chemistry: chemical con artists foil drug discovery. Nature 513(7519), 481–483 (2014).
    • 55. Xie J, Wang S, Ma P et al. Selection of small molecules that bind to and activate the insulin receptor from a DNA-encoded library of natural products. iScience 23(6), 101197 (2020).
    • 56. Paciaroni NG, Ndungu JM, Kodadek T. Solid-phase synthesis of DNA-encoded libraries: via an ‘aldehyde explosion’ strategy. Chem. Commun. 56(34), 4656–4659 (2020).
    • 57. McCarthy KA, Franklin GJ, Lancia DR et al. The Impact of variable selection coverage on detection of ligands from a DNA-encoded library screen. SLAS Discov. 25(5), 515–522 (2020).
    • 58. McGregor LM, Gorin DJ et al. Interaction-dependent PCR: identification of ligand-target pairs from libraries of ligands and libraries of targets in a single solution-phase experiment. J. Am. Chem. Soc. 132(44), 15522–15524 (2010).
    • 59. McGregor LM, Jain T, Liu DR. Identification of ligand-target pairs from combined libraries of small molecules and unpurified protein targets in cell lysates. J. Am. Chem. Soc. 136(8), 3264–3270 (2014).
    • 60. Zhao P, Chen Z, Li Y et al. Selection of DNA-encoded small molecule libraries against unmodified and non-immobilized protein targets. Angew. Chemie - Int. Ed. 53(38), 10056–10059 (2014).
    • 61. Hansen N, Christensen A, Larsen L et al. Method for making an enriched library. US10513700B2 (2013).
    • 62. Blakskjaer P, Heitner T, Hansen NJV. Fidelity by design: Yoctoreactor and binder trap enrichment for small-molecule DNA-encoded libraries and drug discovery. Curr. Opin. Chem. Biol. 26, 62–71 (2015).
    • 63. Petersen LK, Blakskjær P, Chaikuad A et al. Novel p38α MAP kinase inhibitors identified from yoctoReactor DNA-encoded small molecule library. Medchemcomm. 7(7), 1332–1339 (2016).
    • 64. Drabovich AP, Berezovski MV, Musheev MU, Krylov SN. Selection of smart small-molecule ligands: the proof of principle. Anal. Chem. 81(1), 490–494 (2009).
    • 65. Kochmann S, Le ATH, Hili R, Krylov SN. Predicting efficiency of NECEEM-based partitioning of protein binders from nonbinders in DNA-encoded libraries. Electrophoresis. 39(23), 2991–2996 (2018).
    • 66. Bao J, Krylova SM, Cherney LT et al. Predicting Electrophoretic Mobility of Protein-Ligand Complexes for Ligands from DNA-Encoded Libraries of Small Molecules. Anal. Chem. 88(10), 5498–5506 (2016).
    • 67. Ottl J, Leder L, Schaefer JV, Dumelin CE. Encoded library technologies as integrated lead finding platforms for drug discovery. Molecules 24(8), 1–22 (2019).
    • 68. MacConnell AB, McEnaney PJ, Cavett VJ, Paegel BM. DNA-encoded solid-phase synthesis: encoding language design and complex oligomer library synthesis. ACS Comb. Sci. 17(9), 518–534 (2015).
    • 69. MacConnell AB, Price AK, Paegel BM. An integrated microfluidic processor for DNA-encoded combinatorial library functional screening. ACS Comb. Sci. 19(3), 181–192 (2017).
    • 70. Price AK, MacConnell AB, Paegel BM. HvSABR: photochemical dose–response bead screening in droplets. Anal. Chem. 88(5), 2904–2911 (2016).
    • 71. Su MG, Weng JTY, Hsu JBK et al. Investigation and identification of functional post-translational modification sites associated with drug binding and protein-protein interactions. BMC Syst. Biol. 11(Suppl. 7), (2017).
    • 72. Wu Z, Graybill TL, Zeng X 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. 17(12), 722–731 (2015).
    • 73. Shi B, Deng Y, Zhao P, Li X. Selecting a DNA-encoded chemical library against non-immobilized proteins using a ‘ligate-cross-link-purify’ strategy. Bioconjug. Chem. 28(9), 2293–2301 (2017).
    • 74. Shi B, Deng Y, Li X. Polymerase-extension-based selection method for dna-encoded chemical libraries against nonimmobilized protein targets. ACS Comb. Sci. 21(5), 345–349 (2019).
    • 75. Denton KE, Krusemark CJ. Crosslinking of DNA-linked ligands to target proteins for enrichment from DNA-encoded libraries. Medchemcomm. 7(10), 2020–2027 (2016).
    • 76. Le ATH, Krylova SM, Kanoatov M et al. Ideal-filter capillary electrophoresis (IFCE) facilitates the one-step selection of aptamers. Angew. Chemie Int. Ed. 58(9), 2739–2743 (2019).
    • 77. Wang TY, Hu L, Krylov SN. Empirical predictor of conditions that support ideal-filter capillary electrophoresis. Electrophoresis. 41, 1225–1229 (2020).
    • 78. Kim D, Jetson RR, Krusemark CJ. A DNA-assisted immunoassay for enzyme activity: via a DNA-linked, activity-based probe. Chem. Commun. 53(68), 9474–9477 (2017). • The first example of DECL technology being used for identification of irreversible activity-based probes.
    • 79. Hackler AL, Fitzgerald FG, Dang VQ et al. Off-DNA DNA-encoded library affinity screening. ACS Comb. Sci. 22(1), 25–34 (2020).
    • 80. Cochrane WG, Malone ML, Dang VQ et al. Activity-based DNA-encoded library screening. ACS Comb. Sci. 21(5), 425–435 (2019).
    • 81. Cai B, Kim D, Akhand S et al. Selection of DNA-encoded libraries to protein targets within and on living cells. J. Am. Chem. Soc. 141(43), 17057–17061 (2019). •• The first example of DECL screening for intracellular targets, allowing target protein affinity to be measured in a more physiological context.
    • 82. Roy A, Kodadek T. DELs Inside Cells. Trends Pharmacol. Sci. 41(4), 225–227 (2020).
    • 83. Litovchick A, Tian X, Monteiro MI et al. Novel nucleic acid binding small molecules discovered using DNA-encoded chemistry. Molecules. 24(10), 1–14 (2019).
    • 84. Bentley DR, Balasubramanian S, Swerdlow HP et al. Accurate whole human genome sequencing using reversible terminator chemistry. Nature. 456(7218), 53–59 (2008).
    • 85. Buller F, Steiner M, Scheuermann J et al. High-throughput sequencing for the identification of binding molecules from DNA-encoded chemical libraries. Bioorganic Med. Chem. Lett. 20(14), 4188–4192 (2010).
    • 86. Bharagava RN, Purchase D, Saxena G, Mulla SI. Applications of metagenomics in microbial bioremediation of pollutants. In: Microbial Diversity in the Genomic Era Das SDash HR (Eds). Academic Press, Elsevier, London, UK, 459–477 (2019).
    • 87. Prati L, Bigatti M, Donckele EJ et al. On-DNA hit validation methodologies for ligands identified from DNA-encoded chemical libraries. Biochem. Biophys. Res. Commun. https://doi.org/10.1016/j.bbrc.2020.04.03 (2020).
    • 88. Satz AL, Hochstrasser R, Petersen AC. Analysis of current DNA encoded library screening data indicates higher false negative rates for numerically larger libraries. ACS Comb. Sci. 19(4), 234–238 (2017).
    • 89. Sannino A, Gabriele E, Bigatti M et al. Quantitative assessment of affinity selection performance by Using DNA-Encoded Chemical Libraries. ChemBioChem. 20(7), 955–962 (2019).
    • 90. Chen Q, Cheng X, Zhang L et al. Exploring the lower limit of individual DNA-encoded library molecules in selection. SLAS Discov. 25(5), 523–529 (2020).
    • 91. Bobers J, Škopić MK, Dinter R et al. Design of an automated reagent-dispensing system for reaction screening and validation with DNA-tagged substrates. ACS Comb. Sci. 22(3), 101–108 (2020).
    • 92. Hunter JH, Prendergast L, Valente LF et al. High fidelity Suzuki-Miyaura coupling for the synthesis of DNA encoded libraries enabled by micelle forming surfactants. Bioconjug. Chem. 31(1), 149–155 (2020).
    • 93. Faver JC, Riehle K, Lancia DR et al. Quantitative comparison of enrichment from DNA-encoded chemical library selections. ACS Comb. Sci. 21(2), 75–82 (2019).
    • 94. Kuai L, O'Keeffe T, Arico-Muendel C. Randomness in DNA encoded library selection data can be modeled for more reliable enrichment calculation. SLAS Discov. 23(5), 405–416 (2018).
    • 95. MacConnell AB, Paegel BM. Poisson statistics of combinatorial library sampling predict false discovery rates of screening. ACS Comb. Sci. 19(8), 524–532 (2017).
    • 96. Chen Q, Hall J, Foley TL et al. A method for estimating binding affinity from primary DEL selection data. Biochem. Biophys. Res. Commun. 533(2), 249–255 (2020).
    • 97. Hall J, Foley TL, Chen Q et al. A simple method for determining compound affinity and chemical yield from DNA-encoded library selections. Biochem. Biophys. Res. Commun. 527(1), 250–256 (2020).
    • 98. Komar P, Kalinić M. Denoising DNA encoded library screens with sparse learning. ACS Comb. Sci. 8, 410–421 (2020).
    • 99. Smith T, Heger A, Sudbery I. UMI-tools: modeling sequencing errors in Unique Molecular Identifiers to improve quantification accuracy. Genome Res. 27(3), 491–499 (2017).
    • 100. Amigo J, Rama-Garda R, Bello X et al. tagFinder: a novel tag analysis methodology that enables detection of molecules from DNA-encoded chemical libraries. SLAS Discov. 23(5), 397–404 (2018).
    • 101. McCloskey K, Sigel EA, Kearnes S et al. Machine learning on DNA-encoded libraries: a new paradigm for hit finding. 63(16), 8857–8866 (2020). • An application of machine learning the DECL to leverage their data to predict new hit scaffolds.
    • 102. Kollmann CS, Bai X, Tsai CH 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. Bioorganic Med. Chem. 22(7), 2353–2365 (2014).
    • 103. Johannes JW, Bates S, Beigie C et al. Structure based design of non-natural peptidic macrocyclic Mcl-1 inhibitors. ACS Med. Chem. Lett. 8(2), 239–244 (2017).
    • 104. Tavassoli A. SICLOPPS cyclic peptide libraries in drug discovery. Curr. Opin. Chem. Biol. 38, 30–35 (2017).
    • 105. Plexium company website. https://plexium.com/
    • 106. Schapira M, Calabrese MF, Bullock AN, Crews CM. Targeted protein degradation: expanding the toolbox. Nat. Rev. Drug Discov. 18(12), 949–963 (2019).
    • 107. Neklesa TK, Winkler JD, Crews CM. Targeted protein degradation by PROTACs. Pharmacol. Ther. 174, 138–144 (2017).
    • 108. Han X, Zhao L, Xiang W et al. Discovery of highly potent and efficient PROTAC degraders of androgen receptor (AR) by employing weak binding affinity VHL E3 ligase ligands. J. Med. Chem. 62(24), 11218–11231 (2019).
    • 109. Andersen JN, McRiner AJ, Fouser LA et al. Abstract 981: degradation of immuno-oncology targets via proprietary PROTAC platform integrating DNA-encoded library technology and rational drug design. Cancer Res. 79(Suppl. 13), 981–981 (2019).
    • 110. Annis DA, Nickbarg E, Yang X et al. Affinity selection-mass spectrometry screening techniques for small molecule drug discovery. Curr. Opin. Chem. Biol. 11(5), 518–526 (2007).
    • 111. Quartararo AJ, Gates ZP, Somsen BA et al. Ultra-large chemical libraries for the discovery of high-affinity peptide binders. Nat. Commun. 11, 3183 (2020).