The SGC beyond structural genomics: redefining the role of 3D structures by coupling genomic stratification with fragment-based discovery

The ongoing explosion in genomics data has long since outpaced the capacity of conventional biochemical methodology to verify the large number of hypotheses that emerge from the analysis of such data. In contrast, it is still a gold-standard for early phenotypic validation towards small-molecule drug discovery to use probe molecules (or tool compounds), notwithstanding the difficulty and cost of generating them. Rational structure-based approaches to ligand discovery have long promised the efficiencies needed to close this divergence; in practice, however, this promise remains largely unfulfilled, for a host of well-rehearsed reasons and despite the huge technical advances spearheaded by the structural genomics initiatives of the noughties. Therefore the current, fourth funding phase of the Structural Genomics Consortium (SGC), building on its extensive experience in structural biology of novel targets and design of protein inhibitors, seeks to redefine what it means to do structural biology for drug discovery. We developed the concept of a Target Enabling Package (TEP) that provides, through reagents, assays and data, the missing link between genetic disease linkage and the development of usefully potent compounds. There are multiple prongs to the ambition: rigorously assessing targets’ genetic disease linkages through crowdsourcing to a network of collaborating experts; establishing a systematic approach to generate the protocols and data that comprise each target’s TEP; developing new, X-ray-based fragment technologies for generating high quality chemical matter quickly and cheaply; and exploiting a stringently open access model to build multidisciplinary partnerships throughout academia and industry. By learning how to scale these approaches, the SGC aims to make structures finally serve genomics, as originally intended, and demonstrate how 3D structures systematically allow new modes of druggability to be discovered for whole classes of targets.

[1]  Edward W. Lowe,et al.  Computational Methods in Drug Discovery , 2014, Pharmacological Reviews.

[2]  J. Ng,et al.  Gentle, fast and effective crystal soaking by acoustic dispensing , 2016, bioRxiv.

[3]  John P. Overington,et al.  The druggable genome and support for target identification and validation in drug development , 2016, Science Translational Medicine.

[4]  Daniel Svozil,et al.  Probes &Drugs portal: an interactive, open data resource for chemical biology , 2017, Nature Methods.

[5]  Eddy Arnold,et al.  Advantages of crystallographic fragment screening: functional and mechanistic insights from a powerful platform for efficient drug discovery. , 2014, Progress in biophysics and molecular biology.

[6]  Nathan Brown,et al.  Fragment-based hit identification: thinking in 3D. , 2013, Drug discovery today.

[7]  Sebastian Kelm,et al.  A multi-crystal method for extracting obscured crystallographic states from conventionally uninterpretable electron density , 2017, Nature Communications.

[8]  Christopher W Murray,et al.  Opportunity Knocks: Organic Chemistry for Fragment-Based Drug Discovery (FBDD). , 2016, Angewandte Chemie.

[9]  Andrew D. Johnson,et al.  Large-scale genomic analyses link reproductive ageing to hypothalamic signaling, breast cancer susceptibility and BRCA1-mediated DNA repair , 2015, Nature Genetics.

[10]  D. Waugh,et al.  Escherichia coli maltose‐binding protein is uncommonly effective at promoting the solubility of polypeptides to which it is fused , 1999, Protein science : a publication of the Protein Society.

[11]  G. Franck Open access , 2012, Cell cycle.

[12]  Harren Jhoti,et al.  Twenty years on: the impact of fragments on drug discovery , 2016, Nature Reviews Drug Discovery.

[13]  Alexandre Varnek,et al.  Estimation of the size of drug-like chemical space based on GDB-17 data , 2013, Journal of Computer-Aided Molecular Design.

[14]  Jean-Louis Reymond,et al.  Enumeration of 166 Billion Organic Small Molecules in the Chemical Universe Database GDB-17 , 2012, J. Chem. Inf. Model..

[15]  U. H. Danielson,et al.  Biophysics in drug discovery: impact, challenges and opportunities , 2016, Nature Reviews Drug Discovery.

[16]  Patrick Collins,et al.  The XChemExplorer graphical workflow tool for routine or large-scale protein–ligand structure determination , 2017, Acta crystallographica. Section D, Structural biology.

[17]  Julian Blagg,et al.  Choose and Use Your Chemical Probe Wisely to Explore Cancer Biology , 2017, Cancer cell.

[18]  Paul E Brennan,et al.  Chemical probes and inhibitors of bromodomains outside the BET family† †The authors declare no competing interests. , 2016, MedChemComm.

[19]  Peter M Visscher,et al.  Large-scale genomics unveils the genetic architecture of psychiatric disorders , 2014, Nature Neuroscience.

[20]  N. Gray,et al.  Targeting cancer with small molecule kinase inhibitors , 2009, Nature Reviews Cancer.

[21]  John P. Overington,et al.  The promise and peril of chemical probes. , 2015, Nature chemical biology.

[22]  Stephen V Frye,et al.  The art of the chemical probe. , 2010, Nature chemical biology.

[23]  Brian D. Marsden,et al.  High-throughput production of human proteins for crystallization: The SGC experience , 2010, Journal of structural biology.

[24]  Frank von Delft,et al.  A poised fragment library enables rapid synthetic expansion yielding the first reported inhibitors of PHIP(2), an atypical bromodomain† †Electronic supplementary information (ESI) available. See DOI: 10.1039/c5sc03115j , 2015, Chemical science.

[25]  Randy J. Read,et al.  Overview of the CCP4 suite and current developments , 2011, Acta crystallographica. Section D, Biological crystallography.