Fragment Merging Using a Graph Database Samples Different Catalogue Space than Similarity Search
- Stephanie WillsStephanie WillsDepartment of Statistics, University of Oxford, Oxford OX1 3LB, United KingdomCentre for Medicines Discovery, University of Oxford, Oxford OX3 7DQ, United KingdomMore by Stephanie Wills
- ,
- Ruben Sanchez-GarciaRuben Sanchez-GarciaDepartment of Statistics, University of Oxford, Oxford OX1 3LB, United KingdomCentre for Medicines Discovery, University of Oxford, Oxford OX3 7DQ, United KingdomMore by Ruben Sanchez-Garcia
- ,
- Tim DudgeonTim DudgeonInformatics Matters, Ltd., Perch Coworking, Franklins House, Bicester OX26 6JU, United KingdomMore by Tim Dudgeon
- ,
- Stephen D. RoughleyStephen D. RoughleyVernalis (R&D) Limited, Granta Park, Great Abington, Cambridge CB21 6GB, United KingdomMore by Stephen D. Roughley
- ,
- Andy MerrittAndy MerrittLifeArc, Lynton House, 7−12 Tavistock Square, London WC1H 9LT, United KingdomMore by Andy Merritt
- ,
- Roderick E. HubbardRoderick E. HubbardVernalis (R&D) Limited, Granta Park, Great Abington, Cambridge CB21 6GB, United KingdomMore by Roderick E. Hubbard
- ,
- James DavidsonJames DavidsonVernalis (R&D) Limited, Granta Park, Great Abington, Cambridge CB21 6GB, United KingdomMore by James Davidson
- ,
- Frank von DelftFrank von DelftCentre for Medicines Discovery, University of Oxford, Oxford OX3 7DQ, United KingdomDiamond Light Source, Didcot OX11 0DE, United KingdomResearch Complex at Harwell, Harwell Science and Innovation Campus, Didcot OX11 0FA, United KingdomDepartment of Biochemistry, University of Johannesburg, Auckland Park, Johannesburg 2006, South AfricaMore by Frank von Delft
- , and
- Charlotte M. Deane*Charlotte M. Deane*E-mail: [email protected]Department of Statistics, University of Oxford, Oxford OX1 3LB, United KingdomMore by Charlotte M. Deane
Abstract

Fragment merging is a promising approach to progressing fragments directly to on-scale potency: each designed compound incorporates the structural motifs of overlapping fragments in a way that ensures compounds recapitulate multiple high-quality interactions. Searching commercial catalogues provides one useful way to quickly and cheaply identify such merges and circumvents the challenge of synthetic accessibility, provided they can be readily identified. Here, we demonstrate that the Fragment Network, a graph database that provides a novel way to explore the chemical space surrounding fragment hits, is well-suited to this challenge. We use an iteration of the database containing >120 million catalogue compounds to find fragment merges for four crystallographic screening campaigns and contrast the results with a traditional fingerprint-based similarity search. The two approaches identify complementary sets of merges that recapitulate the observed fragment–protein interactions but lie in different regions of chemical space. We further show our methodology is an effective route to achieving on-scale potency by retrospective analyses for two different targets; in analyses of public COVID Moonshot and Mycobacterium tuberculosis EthR inhibitors, potential inhibitors with micromolar IC50 values were identified. This work demonstrates the use of the Fragment Network to increase the yield of fragment merges beyond that of a classical catalogue search.
This publication is licensed under
License Summary*
You are free to share (copy and redistribute) this article in any medium or format and to adapt (remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share (copy and redistribute) this article in any medium or format and to adapt (remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share (copy and redistribute) this article in any medium or format and to adapt (remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Introduction
Materials and Methods
Fragment Network Database
XChem Data Sets
Target | Type of binding site | Number of fragments | Number of pairs for merginga |
---|---|---|---|
DPP11 | Allosteric | 11 | 55 |
PARP14 | Active | 13 | 75 |
nsp13 | Active | 9 | 35 |
Mpro | Active | 19 | 134 |
Value reflects the number of pairs after removing fragment pairs that are highly similar.
Molecular Fingerprint Calculation
Computational Workflow: Querying
Figure 1

Figure 1. Pipeline for identifying fragment merges. Fragment hits from crystallographic fragment screens are used for finding fragment merges. All possible pairs of compounds are enumerated for merging (removing those with high similarity). Both the Fragment Network and similarity search are used to identify fragment merges. The Fragment Network enumerates all possible substructures of one of the fragments in the merge while the other fragment is regarded as the seed fragment. A series of optional hops are made away from the seed fragment (up to a maximum of two), after which an expansion is made by incorporating a substructure from the other fragment. The similarity search finds merges by calculating the Tversky (Tv) similarity against every compound in the database using the Morgan fingerprint (2048 bits and radius 2). The Tversky calculation uses α and β values of 0.7 and 0.3, respectively. All compounds with a mean similarity ≥0.4 are retained. The merges pass through a series of 2D and 3D filters, including pose generation with Fragmenstein, to result in scored poses.
Search by Fragment Network Query
Similarity Search
Computational Workflow: Filtering
Filtering Using Calculated Molecular Descriptors
Filtering Compounds That Resemble Expansions of One Fragment
Filtering Compounds Using Constrained Embedding
Filtering Compounds That Clash with the Protein Pocket
Filtering Compounds Following Pose Generation with Fragmenstein
Computational Workflow: Scoring and Analysis
Prediction of Protein–Ligand Interactions
Low-Dimensional Projections of Chemical Space
Retrospective Analysis Using Experimental Data
Mpro
EthR
Results
The Fragment Network Readily Identifies Pure Merges
1. | Complete overlap merges occur when most of the volume of one fragment overlaps with the other. | ||||
2. | Partial overlap by ring occurs when only a ring overlaps between fragment pairs. These are the “classical” merges described in the literature and are akin to the ligand overlap required for molecular hybridization. In these cases, the merging process is relatively straightforward as there is a clear hypothesis for the connectivity of the final molecule. | ||||
3. | Partial overlap without ring occurs when the overlap is some other substructure. In these cases, the connectivity of the final compound can be nonobvious. | ||||
4. | Nonoverlap of parent fragments requires identifying linking opportunities, which were found in the PARP14 and Mpro data sets. |
Number of hits | Search typea | Before filtering | After filtering | % filtered | Number of overlap | Complete overlap (%) | Partial overlap, ring (%) | Partial overlap, no ring (%) | No overlap (%) | |
---|---|---|---|---|---|---|---|---|---|---|
DPP11 | 11 | FN | 22,903 | 198 | 0.9 | 8 | 0.0 | 29.4 | 70.6 | NA |
SS | 85,919 | 271 | 0.3 | 4.0 | 32.0 | 64.0 | NA | |||
PARP14 | 13 | FN | 48,320 | 70 | 0.1 | 0 | 0.0 | 18.3 | 45.1 | 36.6 |
SS | 78,116 | 56 | 0.1 | 16.1 | 16.1 | 37.5 | 30.4 | |||
nsp13 | 9 | FN | 36,239 | 503 | 1.4 | 1 | 0.4 | 99.6 | 0.0 | NA |
SS | 40,102 | 530 | 1.3 | 1.6 | 97.4 | 1.0 | NA | |||
Mpro | 19 | FN | 109,012 | 952 | 0.9 | 4 | 2.5 | 6.8 | 49.6 | 41.2 |
SS | 169,424 | 918 | 0.5 | 71.0 | 7.1 | 12.5 | 9.4 |
FN, Fragment Network; SS, similarity search.
Figure 2

Figure 2. The Fragment Network identifies pure merges. (a) A fragment-merging opportunity for the main protease (Mpro) data set. Interactions are predicted using the protein–ligand interaction profiler (PLIP). Hydrogen bonds and π-stacking interactions are shown by cyan and magenta dotted lines, respectively. The PanDDA density is provided for the purple fragment in the Supporting Information (owing to the unusual conformation). (b) The linker-like merge (pose generated using Fragmenstein) joins substructures from partially overlapping fragments by a “linker-like” region, maintaining the hydrogen bond with THR-45; a change in orientation of the thiazole ring (with respect to the thiophene ring in the fragment) enables an additional π-stacking interaction with HIS-41. (c) A fragment-linking opportunity for Mpro. (d) The proposed compound maintains a hydrogen bond with PHE-140 and makes an additional bond with SER-144. It is worth noting that the linker group proposed by the Fragment Network is present in thioacetazone, an oral antibiotic. (e) The fragments and merge in a and b in 2D. (f) The fragments and merge in c and d in 2D.
Fragment Network and Similarity Searches Find Comparable Numbers of Compounds
Figure 3

Figure 3. The Fragment Network and similarity searches identify filtered compounds for different fragment pairs. The numbers of filtered compounds for each fragment pair found using the Fragment Network (blue) or similarity search (orange) are shown across targets (a) dipeptidyl peptidase 11 (DPP11), (b) poly(ADP-ribose) polymerase 14, (PARP14), (c) nonstructural protein 13 (nsp13), and (d) main protease (Mpro). Only pairs that resulted in filtered compounds are shown. Pairs are ordered from right to left according to the number of Fragment Network compounds found. The data show that each search technique was able to identify filtered compounds for pairs where the other technique identified none.
The Fragment Network and Similarity Search Identify Compounds from Distinct Areas of Chemical Space
Figure 4

Figure 4. Fragment Network and similarity search-derived compound sets populate different regions of chemical space. The chemical space occupied by the filtered compound sets is projected into two dimensions using the T-SNE algorithm across targets (a) dipeptidyl peptidase 11 (DPP11), (b) poly(ADP-ribose) polymerase 14 (PARP14), (c) nonstructural protein 13 (nsp13), and (d) main protease (Mpro). Fragment Network compounds are shown in blue, and similarity search compounds are shown in orange. The two compound sets are shown to occupy distinct areas of chemical space.
Fragment Network and Similarity Searches Identify Compounds That Form Interactions with Residues Not Reached by the Other Technique
Target | Fragment Network compounds (%) | Similarity search compounds (%) |
---|---|---|
DPP11 | 39.7 ± 5.0 | 60.3 ± 5.0 |
PARP14 | 40.3 ± 5.4 | 59.7 ± 5.4 |
nsp13 | 74.0 ± 2.8 | 26.0 ± 2.8 |
Mpro | 30.5 ± 4.3 | 69.5 ± 4.3 |
Mean and standard deviation are provided.
Efficiency of the Filtering Pipeline
The Fragment Network Has Efficiency Benefits over Similarity Search
Retrospective Analysis Using Experimental Data
Mpro
Figure 5

Figure 5. The Fragment Network identifies a known binder against Mpro. A Fragment Network search using (a) two fragment hits against the SARS-CoV-2 main protease (Mpro) identifies, (b) a known binder against Mpro (LON-WEI-b2874fec-25; RapidFire mass spectrometry (RF-MS) IC50 value of 59.6 μM) and similar compounds to known binders, (c) JAN-GHE-83b26c96–22 (fluorescence and RF-MS IC50 values of 96.9 μM and 24.5 μM), and (d) TRY-UNI-714a760b-18 (fluorescence and RF-MS IC50 values of 26.2 μM and 13.0 μM). Fragmenstein-predicted merge poses are shown in white, and crystal poses are in cyan. Interactions are predicted using the protein–ligand interaction profiler (PLIP), and key interaction residues are shown. Hydrogen bonds are shown in cyan, and π-stacking interactions are shown in magenta.
EthR
Figure 6

Figure 6. The Fragment Network identifies a known binder against EthR. A Fragment Network search using (a) two fragment hits against (which each bind in two different positions) Mycobacterium tuberculosis transcriptional repressor protein EthR identifies (b) a known binder (compound 4; IC50 value of >100 μM). (c) An alternative crystallographic arrangement of the equivalent fragments identifies (d) a similar compound to a known binder, compound 21 (IC50 value of 22 μM). Fragmenstein-predicted merge poses are shown in white, and crystal poses are in cyan. Hydrogen bonds are shown in cyan.
Discussion
Conclusions
Data Availability
The code to run the querying and filtering pipeline is publicly available through https://github.com/oxpig/fragment_network_merges. The code to generate the Fragment Network is available at https://github.com/InformaticsMatters/fragmentor. The query data retrieved from the database searches and the filtered compounds are available from https://zenodo.org/record/7957805. Access to the current snapshot of the database can be provided upon request.
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jcim.3c00276.
Figures and tables referenced in the text (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
S.W. is supported by the Engineering and Physical Sciences Research Council (EPSRC; Grant No. EP/S024093/1), Vernalis, and LifeArc. This work has been partially funded by Rosetrees Trust [M940]. The authors thank Alpha Lee for helpful discussion and Matteo Ferla for his help with Fragmenstein.
References
This article references 63 other publications.
- 1Lamoree, B.; Hubbard, R. Current perspectives in fragment-based lead discovery (FBLD). Essays Biochem 2017, 61, 453– 464, DOI: 10.1042/EBC20170028[Crossref], [PubMed], [CAS], Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1M7pt1Gmtw%253D%253D&md5=35bce3c5978329c6737ffd719135d363Current perspectives in fragment-based lead discovery (FBLD)Lamoree Bas; Hubbard Roderick E; Hubbard Roderick EEssays in biochemistry (2017), 61 (5), 453-464 ISSN:.It is over 20 years since the first fragment-based discovery projects were disclosed. The methods are now mature for most 'conventional' targets in drug discovery such as enzymes (kinases and proteases) but there has also been growing success on more challenging targets, such as disruption of protein-protein interactions. The main application is to identify tractable chemical startpoints that non-covalently modulate the activity of a biological molecule. In this essay, we overview current practice in the methods and discuss how they have had an impact in lead discovery - generating a large number of fragment-derived compounds that are in clinical trials and two medicines treating patients. In addition, we discuss some of the more recent applications of the methods in chemical biology - providing chemical tools to investigate biological molecules, mechanisms and systems.
- 2Davis, B. J.; Roughley, S. D. In Platform Technologies in Drug Discovery and Validation; Goodnow, R. A., Ed.; Annual Reports in Medicinal Chemistry series; Academic Press, 2017; Vol. 50; pp 371– 439.
- 3Hann, M. M.; Leach, A. R.; Harper, G. Molecular complexity and its impact on the probability of finding leads for drug discovery. J. Chem. Inf. Model. 2001, 41, 856– 864, DOI: 10.1021/ci000403i[ACS Full Text
], [CAS], Google Scholar
3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXivFynt7w%253D&md5=e89e76c8fb83fc5012369ae30f20e6b7Molecular Complexity and Its Impact on the Probability of Finding Leads for Drug DiscoveryHann, Michael M.; Leach, Andrew R.; Harper, GavinJournal of Chemical Information and Computer Sciences (2001), 41 (3), 856-864CODEN: JCISD8; ISSN:0095-2338. (American Chemical Society)Using a simple model of ligand-receptor interactions, the interactions between ligands and receptors of varying complexities are studied and the probabilities of binding calcd. It is obsd. that as the systems become more complex the chance of observing a useful interaction for a randomly chosen ligand falls dramatically. The implications of this for the design of combinatorial libraries is explored. A large set of drug leads and optimized compds. is profiled using several different properties relevant to mol. recognition. The changes obsd. for these properties during the drug optimization phase support the hypothesis that less complex mols. are more common starting points for the discovery of drugs. An extreme example of the use of simple mols. for directed screening against thrombin is provided. - 4Leach, A. R.; Hann, M. M. Molecular complexity and fragment-based drug discovery: ten years on. Curr. Opin. Chem. Biol. 2011, 15, 489– 496, DOI: 10.1016/j.cbpa.2011.05.008[Crossref], [PubMed], [CAS], Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXpvFCltrg%253D&md5=19c2d65de80a8bab1059468828da6ce2Molecular complexity and fragment-based drug discovery: ten years onLeach, Andrew R.; Hann, Michael M.Current Opinion in Chemical Biology (2011), 15 (4), 489-496CODEN: COCBF4; ISSN:1367-5931. (Elsevier B.V.)We review the concept of mol. complexity in the context of the very simple model of mol. interactions that we introduced over 10 years ago. A summary is presented of efforts to validate this simple model using screening data. The relationship between the complexity model and the problem of sampling chem. space is discussed, together with the relevance of these theor. concepts to fragment-based drug discovery.
- 5Keserű, G. M.; Erlanson, D. A.; Ferenczy, G. G.; Hann, M. M.; Murray, C. W.; Pickett, S. D. Design principles for fragment libraries: maximizing the value of learnings from pharma fragment-based drug discovery (FBDD) programs for use in academia. J. Med. Chem. 2016, 59, 8189– 8206, DOI: 10.1021/acs.jmedchem.6b00197[ACS Full Text
], [CAS], Google Scholar
5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XmvFSit7s%253D&md5=888fe2cf003c5564c2ec391cf54ea2acDesign Principles for Fragment Libraries: Maximizing the Value of Learnings from Pharma Fragment-Based Drug Discovery (FBDD) Programs for Use in AcademiaKeseru, Gyorgy M.; Erlanson, Daniel A.; Ferenczy, Gyorgy G.; Hann, Michael M.; Murray, Christopher W.; Pickett, Stephen D.Journal of Medicinal Chemistry (2016), 59 (18), 8189-8206CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Fragment-based drug discovery (FBDD) is well suited for discovering both drug leads and chem. probes of protein function; it can cover broad swaths of chem. space and allows the use of creative chem. FBDD is widely implemented for lead discovery in industry but is sometimes used less systematically in academia. Design principles and implementation approaches for fragment libraries are continually evolving, and the lack of up-to-date guidance may prevent more effective application of FBDD in academia. This Perspective explores many of the theor., practical, and strategic considerations that occur within FBDD programs, including the optimal size, complexity, physicochem. profile, and shape profile of fragments in FBDD libraries, as well as compd. storage, evaluation, and screening technologies. This compilation of industry experience in FBDD will hopefully be useful for those pursuing FBDD in academia. - 6Hall, R. J.; Mortenson, P. N.; Murray, C. W. Efficient exploration of chemical space by fragment-based screening. Prog. Biophys. Mol. Biol. 2014, 116, 82– 91, DOI: 10.1016/j.pbiomolbio.2014.09.007[Crossref], [PubMed], [CAS], Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhs1ymu7zO&md5=a63e451443207393b781d3d68b10bcd1Efficient exploration of chemical space by fragment-based screeningHall, Richard J.; Mortenson, Paul N.; Murray, Christopher W.Progress in Biophysics & Molecular Biology (2014), 116 (2-3), 82-91CODEN: PBIMAC; ISSN:0079-6107. (Elsevier Ltd.)Screening methods seek to sample a vast chem. space in order to identify starting points for further chem. optimization. Fragment based drug discovery exploits the superior sampling of chem. space that can be achieved when the mol. wt. is restricted. Here we show that com. available fragment space is still relatively poorly sampled and argue for highly sensitive screening methods to allow the detection of smaller fragments. We analyze the properties of our fragment library vs. the properties of X-ray hits derived from the library. We particularly consider properties related to the degree of planarity of the fragments.
- 7Murray, C. W.; Rees, D. C. The rise of fragment-based drug discovery. Nat. Chem. 2009, 1, 187– 192, DOI: 10.1038/nchem.217[Crossref], [PubMed], [CAS], Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXmtF2rs78%253D&md5=0bd1a40fd927cce9200f0fa296a7462fThe rise of fragment-based drug discoveryMurray, Christopher W.; Rees, David C.Nature Chemistry (2009), 1 (3), 187-192CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)A review. The search for new drugs is plagued by high attrition rates at all stages in research and development. Chemists have an opportunity to tackle this problem because attrition can be traced back, in part, to the quality of the chem. leads. Fragment-based drug discovery (FBDD) is a new approach, increasingly used in the pharmaceutical industry, for reducing attrition and providing leads for previously intractable biol. targets. FBDD identifies low-mol.-wt. ligands (∼150 Da) that bind to biol. important macromols. The three-dimensional exptl. binding mode of these fragments is detd. using X-ray crystallog. or NMR spectroscopy, and is used to facilitate their optimization into potent mols. with drug-like properties. Compared with high-throughput-screening, the fragment approach requires fewer compds. to be screened, and, despite the lower initial potency of the screening hits, offers more efficient and fruitful optimization campaigns. Here, we review the rise of FBDD, including its application to discovering clin. candidates against targets for which other chem. approaches have struggled.
- 8Roughley, S. D.; Hubbard, R. E. How well can fragments explore accessed chemical space? A case study from heat shock protein 90. J. Med. Chem. 2011, 54, 3989– 4005, DOI: 10.1021/jm200350g[ACS Full Text
], [CAS], Google Scholar
8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXmsFymurs%253D&md5=90c2c8028cb460c0c6fdc956e7daae28How Well Can Fragments Explore Accessed Chemical Space? A Case Study from Heat Shock Protein 90Roughley, Stephen D.; Hubbard, Roderick E.Journal of Medicinal Chemistry (2011), 54 (12), 3989-4005CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. This article summerizes the published literature on the generation of inhibitors and design of drug against the mol. chaperone, heat shock protein 90 (Hsp90), as potential therapeutics to treat cancer. - 9Collins, P. M.; Douangamath, A.; Talon, R.; Dias, A.; Brandao-Neto, J.; Krojer, T.; von Delft, F. Achieving a good crystal system for crystallographic X-ray fragment screening. Methods Enzymol 2018, 610, 251– 264, DOI: 10.1016/bs.mie.2018.09.027[Crossref], [PubMed], [CAS], Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsFKrsrvJ&md5=c122d61fb2371eff58429ce90e777c8eAchieving a good crystal system for crystallographic X-ray fragment screeningCollins, Patrick M.; Douangamath, Alice; Talon, Romain; Dias, Alexandre; Brandao-Neto, Jose; Krojer, Tobias; von Delft, FrankMethods in Enzymology (2018), 610 (Modern Approaches in Drug Discovery), 251-264CODEN: MENZAU; ISSN:0076-6879. (Elsevier Inc.)A review. The XChem facility at Diamond Light Source offers fragment screening by X-ray crystallog. as a general access user program. The main advantage of X-ray crystallog. as a primary fragment screen is that it yields directly the location and pose of the fragment hits, whether within pockets of interest or merely on surface sites: this is the key information for structure-based design and for enabling synthesis of follow up mols. Extensive streamlining of the screening expt. at XChem has engendered a very active user program that is generating large amts. of data: in 2017,36 academic and industry groups generated 35,000 datasets of uniquely soaked crystals. It has also generated a large no. of learnings concerning the main remaining bottleneck namely, obtaining a suitable crystal system that will support a successful fragment screen. Here we discuss the practicalities of generating screen-ready crystals that have useful electron d. maps, and how to ensure they will be successfully reproduced and usable at a facility outside the home lab.
- 10The COVID Moonshot Consortium. COVID Moonshot: open science discovery of SARS-CoV-2 main protease inhibitors by combining crowdsourcing, high-throughput experiments, computational simulations, and machine learning. bioRxiv Preprint , 2020. DOI: 10.1101/2020.10.29.339317
- 11Douangamath, A.; Powell, A.; Fearon, D.; Collins, P. M.; Talon, R.; Krojer, T.; Skyner, R.; Brandao-Neto, J.; Dunnett, L.; Dias, A.; Aimon, A.; Pearce, N. M.; Wild, C.; Gorrie-Stone, T.; von Delft, F. Achieving efficient fragment screening at XChem facility at Diamond Light Source. JoVE 2021, 171, e62414 DOI: 10.3791/62414
- 12Metz, A.; Wollenhaupt, J.; Glöckner, S.; Messini, N.; Huber, S.; Barthel, T.; Merabet, A.; Gerber, H.-D.; Heine, A.; Klebe, G.; Weiss, M. S. Frag4Lead: growing crystallographic fragment hits by catalog using fragment-guided template docking. Acta Crystallogr., Sect. D: Struct. Biol. 2021, 77, 1168– 1182, DOI: 10.1107/S2059798321008196[Crossref], [CAS], Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvFegt7%252FF&md5=391b172987e46364a4b877d2a34508fcFrag4Lead: growing crystallographic fragment hits by catalog using fragment-guided template dockingMetz, Alexander; Wollenhaupt, Jan; Gloeckner, Steffen; Messini, Niki; Huber, Simon; Barthel, Tatjana; Merabet, Ahmed; Gerber, Hans-Dieter; Heine, Andreas; Klebe, Gerhard; Weiss, Manfred S.Acta Crystallographica, Section D: Structural Biology (2021), 77 (9), 1168-1182CODEN: ACSDAD; ISSN:2059-7983. (International Union of Crystallography)In recent years, crystallog. fragment screening has matured into an almost routine expt. at several modern synchrotron sites. The hits of the screening expt., i.e. small mols. or fragments binding to the target protein, are revealed along with their 3D structural information. Therefore, they can serve as useful starting points for further structure-based hit-to-lead development. However, the progression of fragment hits to tool compds. or even leads is often hampered by a lack of chem. feasibility. As an attractive alternative, compd. analogs that embed the fragment hit structurally may be obtained from com. catalogs. Here, a workflow is reported based on filtering and assessing such potential follow-up compds. by template docking. This means that the crystallog. binding pose was integrated into the docking calcns. as a central starting parameter. Subsequently, the candidates are scored on their interactions within the binding pocket. In an initial proof-of-concept study using five starting fragments known to bind to the aspartic protease endothiapepsin, 28 follow-up compds. were selected using the designed workflow and their binding was assessed by crystallog. Ten of these compds. bound to the active site and five of them showed significantly increased affinity in isothermal titrn. calorimetry of up to single-digit micromolar affinity. Taken together, this strategy is capable of efficiently evolving the initial fragment hits without major synthesis efforts and with full control by X-ray crystallog.
- 13Müller, J.; Klein, R.; Tarkhanova, O.; Gryniukova, A.; Borysko, P.; Merkl, S.; Ruf, M.; Neumann, A.; Gastreich, M.; Moroz, Y. S.; Klebe, G.; Glinca, S. Magnet for the needle in haystack: “Crystal structure first” Fragment hits unlock active chemical matter using targeted exploration of vast chemical spaces. J. Med. Chem. 2022, 65, 15663– 15678, DOI: 10.1021/acs.jmedchem.2c00813[ACS Full Text
], [CAS], Google Scholar
13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XitlWrtrfI&md5=7d2ae978c7757c8f1b3083fa84c60f9eMagnet for the Needle in Haystack: "Crystal Structure First" Fragment Hits Unlock Active Chemical Matter Using Targeted Exploration of Vast Chemical SpacesMueller, Janis; Klein, Raphael; Tarkhanova, Olga; Gryniukova, Anastasiia; Borysko, Petro; Merkl, Stefan; Ruf, Moritz; Neumann, Alexander; Gastreich, Marcus; Moroz, Yurii S.; Klebe, Gerhard; Glinca, SergheiJournal of Medicinal Chemistry (2022), 65 (23), 15663-15678CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Fragment-based drug discovery (FBDD) has successfully led to approved therapeutics for challenging and "undruggable" targets. In the context of FBDD, we introduce a novel, multidisciplinary method to identify active mols. from purchasable chem. space. Starting from four small-mol. fragment complexes of protein kinase A (PKA), a template-based docking screen using Enamine's multibillion REAL Space was performed. A total of 93 mols. out of 106 selected compds. were successfully synthesized. Forty compds. were active in at least one validation assay with the most active follow-up having a 13,500-fold gain in affinity. Crystal structures for six of the most promising binders were rapidly obtained, verifying the binding mode. The overall success rate for this novel fragment-to-hit approach was 40%, accomplished in only 9 wk. The results challenge the established fragment prescreening paradigm since the std. industrial filters for fragment hit identification in a thermal shift assay would have missed the initial fragments. - 14Piticchio, S. G.; Martínez-Cartró, M.; Scaffidi, S.; Rachman, M.; Rodriguez-Arevalo, S.; Sanchez-Arfelis, A.; Escolano, C.; Picaud, S.; Krojer, T.; Filippakopoulos, P.; von Delft, F.; Galdeano, C.; Barril, X. Discovery of novel BRD4 ligand scaffolds by automated navigation of the fragment chemical space. J. Med. Chem. 2021, 64, 17887– 17900, DOI: 10.1021/acs.jmedchem.1c01108[ACS Full Text
], [CAS], Google Scholar
14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXislWrtbjE&md5=98aebd322c14abb1a7f56157e46013a6Discovery of Novel BRD4 Ligand Scaffolds by Automated Navigation of the Fragment Chemical SpacePiticchio, Serena G.; Martinez-Cartro, Miriam; Scaffidi, Salvatore; Rachman, Moira; Rodriguez-Arevalo, Sergio; Sanchez-Arfelis, Ainoa; Escolano, Carmen; Picaud, Sarah; Krojer, Tobias; Filippakopoulos, Panagis; von Delft, Frank; Galdeano, Carles; Barril, XavierJournal of Medicinal Chemistry (2021), 64 (24), 17887-17900CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Fragment-based drug discovery (FBDD) is a very effective hit identification method. However, the evolution of fragment hits into suitable leads remains challenging and largely artisanal. Fragment evolution is often scaffold-centric, meaning that its outcome depends crucially on the chem. structure of the starting fragment. Considering that fragment screening libraries cover only a small proportion of the corresponding chem. space, hits should be seen as probes highlighting privileged areas of the chem. space rather than actual starting points. We have developed an automated computational pipeline to mine the chem. space around any specific fragment hit, rapidly finding analogs that share a common interaction motif but are structurally novel and diverse. On a prospective application on the bromodomain-contg. protein 4 (BRD4), starting from a known fragment, the platform yields active mols. with nonobvious scaffold changes. The procedure is fast and inexpensive and has the potential to uncover many hidden opportunities in FBDD. - 15Miyake, Y.; Itoh, Y.; Hatanaka, A.; Suzuma, Y.; Suzuki, M.; Kodama, H.; Arai, Y.; Suzuki, T. Identification of novel lysine demethylase 5-selective inhibitors by inhibitor-based fragment merging strategy. Bioorg. Med. Chem. 2019, 27, 1119– 1129, DOI: 10.1016/j.bmc.2019.02.006[Crossref], [PubMed], [CAS], Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXivVWltbg%253D&md5=1d9473e11de97ee34b82b82b8d6ffb38Identification of novel lysine demethylase 5-selective inhibitors by inhibitor-based fragment merging strategyMiyake, Yuka; Itoh, Yukihiro; Hatanaka, Atsushi; Suzuma, Yoshinori; Suzuki, Miki; Kodama, Hidehiko; Arai, Yoshinobu; Suzuki, TakayoshiBioorganic & Medicinal Chemistry (2019), 27 (6), 1119-1129CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)Histone lysine demethylases (KDMs) have drawn much attention as targets of therapeutic agents. KDM5 proteins, which are Fe(II)/α-ketoglutarate-dependent demethylases, are assocd. with oncogenesis and drug resistance in cancer cells, and KDM5-selective inhibitors are expected to be anticancer drugs. However, few cell-active KDM5 inhibitors have been reported and there is an obvious need to discover more. In this study, we pursued the identification of highly potent and cell-active KDM5-selective inhibitors. Based on the reported KDM5 inhibitors, we designed several compds. by strategically merging two fragments for competitive inhibition with α-ketoglutarate and for KDM5-selective inhibition. Among them, compds. 10 and 13, which have a 3-cyano pyrazolo[1,5-a]pyrimidin-7-one scaffold, exhibited strong KDM5-inhibitory activity and significant KDM5 selectivity. In cellular assays using human lung cancer cell line A549, 10 and 13 increased the levels of trimethylated lysine 4 on histone H3, which is a specific substrate of KDM5s, and induced growth inhibition of A549 cells. These results should provide a basis for the development of cell-active KDM5 inhibitors to highlight the validity of our inhibitor-based fragment merging strategy.
- 16Ren, J.; Li, J.; Wang, Y.; Chen, W.; Shen, A.; Liu, H.; Chen, D.; Cao, D.; Li, Y.; Zhang, N.; Xu, Y.; Geng, M.; He, J.; Xiong, B.; Shen, J. Identification of a new series of potent diphenol HSP90 inhibitors by fragment merging and structure-based optimization. Bioorg. Med. Chem. Lett. 2014, 24, 2525– 2529, DOI: 10.1016/j.bmcl.2014.03.100[Crossref], [PubMed], [CAS], Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXmslahsrY%253D&md5=057e15a1982cf723de8c7436accf4fffIdentification of a new series of potent diphenol HSP90 inhibitors by fragment merging and structure-based optimizationRen, Jing; Li, Jian; Wang, Yueqin; Chen, Wuyan; Shen, Aijun; Liu, Hongchun; Chen, Danqi; Cao, Danyan; Li, Yanlian; Zhang, Naixia; Xu, Yechun; Geng, Meiyu; He, Jianhua; Xiong, Bing; Shen, JingkangBioorganic & Medicinal Chemistry Letters (2014), 24 (11), 2525-2529CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)Heat shock protein 90 (HSP90) is a mol. chaperone to fold and maintain the proper conformation of many signaling proteins, esp. some oncogenic proteins and mutated unstable proteins. Inhibition of HSP90 was recognized as an effective approach to simultaneously suppress several aberrant signaling pathways, and therefore it was considered as a novel target for cancer therapy. Here, by integrating several techniques including the fragment-based drug discovery method, fragment merging, computer aided inhibitor optimization, and structure-based drug design, the authors were able to identify a series of HSP90 inhibitors, e.g. I [R1 = H. HO2CCH2, NH2CH2CH2, MeCONHCH2CH2; r2 = H, Br, O2N, etc.; R3 = H, Br, Me, MeO]. Several compds. can inhibit HSP90 with IC50 about 20-40 nM, which is at least 200-fold more potent than initial fragments in the protein binding assay. These new HSP90 inhibitors not only explore interactions with an under-studied subpocket, also offer new chemotypes for the development of novel HSP90 inhibitors as anticancer drugs.
- 17Credille, C. V.; Chen, Y.; Cohen, S. M. Fragment-based identification of influenza endonuclease inhibitors. J. Med. Chem. 2016, 59, 6444– 6454, DOI: 10.1021/acs.jmedchem.6b00628[ACS Full Text
], [CAS], Google Scholar
17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XpsF2mtL8%253D&md5=a872083d373a846467c7f2eb5439bed4Fragment-Based Identification of Influenza Endonuclease InhibitorsCredille, Cy V.; Chen, Yao; Cohen, Seth M.Journal of Medicinal Chemistry (2016), 59 (13), 6444-6454CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The influenza virus is responsible for millions of cases of severe illness annually. Yearly variance in the effectiveness of vaccination, coupled with emerging drug resistance, necessitates the development of new drugs to treat influenza infections. One attractive target is the RNA-dependent RNA polymerase PA subunit. Herein we report the development of inhibitors of influenza PA endonuclease derived from lead compds. identified from a metal-binding pharmacophore (MBP) library screen. Pyromeconic acid and derivs. thereof were found to be potent inhibitors of endonuclease. Guided by modeling and previously reported structural data, several sublibraries of mols. were elaborated from the MBP hits. Structure-activity relationships were established, and more potent mols. were designed and synthesized using fragment growth and fragment merging strategies. This approach ultimately resulted in the development of a lead compd. with an IC50 value of 14 nM, which displayed an EC50 value of 2.1 μM against H1N1 influenza virus in MDCK cells. - 18Edink, E.; Rucktooa, P.; Retra, K.; Akdemir, A.; Nahar, T.; Zuiderveld, O.; van Elk, R.; Janssen, E.; van Nierop, P.; van Muijlwijk-Koezen, J.; Smit, A. B.; Sixma, T. K.; Leurs, R.; de Esch, I. J. P. Fragment growing induces conformational changes in acetylcholine-binding protein: a structural and thermodynamic analysis. J. Am. Chem. Soc. 2011, 133, 5363– 5371, DOI: 10.1021/ja110571r[ACS Full Text
], [CAS], Google Scholar
18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhvFeisrc%253D&md5=413798f2128e309545de77f8e995b565Fragment Growing Induces Conformational Changes in Acetylcholine-Binding Protein: A Structural and Thermodynamic AnalysisEdink, Ewald; Rucktooa, Prakash; Retra, Kim; Akdemir, Atilla; Nahar, Tariq; Zuiderveld, Obbe; van Elk, Rene; Janssen, Elwin; van Nierop, Pim; van Muijlwijk-Koezen, Jacqueline; Smit, August B.; Sixma, Titia K.; Leurs, Rob; de Esch, Iwan J. P.Journal of the American Chemical Society (2011), 133 (14), 5363-5371CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Optimization of fragment hits toward high-affinity lead compds. is a crucial aspect of fragment-based drug discovery (FBDD). In the current study, we have successfully optimized a fragment by growing into a ligand-inducible subpocket of the binding site of acetylcholine-binding protein (AChBP). This protein is a sol. homolog of the ligand binding domain (LBD) of Cys-loop receptors. The fragment optimization was monitored with X-ray structures of ligand complexes and systematic thermodn. analyses using surface plasmon resonance (SPR) biosensor anal. and isothermal titrn. calorimetry (ITC). Using site-directed mutagenesis and AChBP from different species, we find that specific changes in thermodn. binding profiles, are indicative of interactions with the ligand-inducible subpocket of AChBP. This study illustrates that thermodn. anal. provides valuable information on ligand binding modes and is complementary to affinity data when guiding rational structure- and fragment-based discovery approaches. - 19Hughes, S. J.; Millan, D. S.; Kilty, I. C.; Lewthwaite, R. A.; Mathias, J. P.; O’Reilly, M. A.; Pannifer, A.; Phelan, A.; Stühmeier, F.; Baldock, D. A.; Brown, D. G. Fragment based discovery of a novel and selective PI3 kinase inhibitor. Bioorg. Med. Chem. Lett. 2011, 21, 6586– 6590, DOI: 10.1016/j.bmcl.2011.07.117[Crossref], [PubMed], [CAS], Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXht12jtrjO&md5=be9322ee0268040ed1da843025df4cdeFragment based discovery of a novel and selective PI3 kinase inhibitorHughes, Samantha J.; Millan, David S.; Kilty, Iain C.; Lewthwaite, Russell A.; Mathias, John P.; O'Reilly, Mark A.; Pannifer, Andrew; Phelan, Anne; Stuehmeier, Frank; Baldock, Darren A.; Brown, David G.Bioorganic & Medicinal Chemistry Letters (2011), 21 (21), 6586-6590CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)We report the use of fragment screening and fragment based drug design to develop a PI3γ kinase fragment hit into a lead. Initial fragment hits were discovered by high concn. biochem. screening, followed by a round of virtual screening to identify addnl. ligand efficient fragments. These were developed into potent and ligand efficient lead compds. using structure guided fragment growing and merging strategies. This led to a potent, selective, and cell permeable PI3γ kinase inhibitor, 12 (I), with good metabolic stability that was useful as a preclin. tool compd.
- 20Brough, P. A.; Barril, X.; Borgognoni, J.; Chene, P.; Davies, N. G. M.; Davis, B.; Drysdale, M. J.; Dymock, B.; Eccles, S. A.; Garcia-Echeverria, C.; Fromont, C.; Hayes, A.; Hubbard, R. E.; Jordan, A. M.; Jensen, M. R.; Massey, A.; Merrett, A.; Padfield, A.; Parsons, R.; Radimerski, T.; Raynaud, F. I.; Robertson, A.; Roughley, S. D.; Schoepfer, J.; Simmonite, H.; Sharp, S. Y.; Surgenor, A.; Valenti, M.; Walls, S.; Webb, P.; Wood, M.; Workman, P.; Wright, L. Combining hit identification strategies: fragment-based and in silico approaches to orally active 2-aminothieno[2,3-d]pyrimidine inhibitors of the Hsp90 molecular chaperone. J. Med. Chem. 2009, 52, 4794– 4809, DOI: 10.1021/jm900357y[ACS Full Text
], [CAS], Google Scholar
20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXos1ehsbs%253D&md5=4d1988060b0f87dc776e1972ea14808eCombining Hit Identification Strategies: Fragment-Based and in Silico Approaches to Orally Active 2-Aminothieno[2,3-d]pyrimidine Inhibitors of the Hsp90 Molecular ChaperoneBrough, Paul A.; Barril, Xavier; Borgognoni, Jenifer; Chene, Patrick; Davies, Nicholas G. M.; Davis, Ben; Drysdale, Martin J.; Dymock, Brian; Eccles, Suzanne A.; Garcia-Echeverria, Carlos; Fromont, Christophe; Hayes, Angela; Hubbard, Roderick E.; Jordan, Allan M.; Jensen, Michael Rugaard; Massey, Andrew; Merrett, Angela; Padfield, Antony; Parsons, Rachel; Radimerski, Thomas; Raynaud, Florence I.; Robertson, Alan; Roughley, Stephen D.; Schoepfer, Joseph; Simmonite, Heather; Sharp, Swee Y.; Surgenor, Allan; Valenti, Melanie; Walls, Steven; Webb, Paul; Wood, Mike; Workman, Paul; Wright, LisaJournal of Medicinal Chemistry (2009), 52 (15), 4794-4809CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Inhibitors of the Hsp90 mol. chaperone are showing considerable promise as potential mol. therapeutic agents for the treatment of cancer. Here we describe novel 2-aminothieno[2,3-d]pyrimidine ATP competitive Hsp90 inhibitors, which were designed by combining structural elements of distinct low affinity hits generated from fragment-based and in silico screening exercises in concert with structural information from X-ray protein crystallog. Examples from this series have high affinity (IC50 = 50-100 nM) for Hsp90 as measured in a fluorescence polarization (FP) competitive binding assay and are active in human cancer cell lines where they inhibit cell proliferation and exhibit a characteristic profile of depletion of oncogenic proteins and concomitant elevation of Hsp72. Several compds. caused tumor growth regression at well tolerated doses when administered orally in a human BT474 human breast cancer xenograft model. - 21Nikiforov, P. O.; Surade, S.; Blaszczyk, M.; Delorme, V.; Brodin, P.; Baulard, A. R.; Blundell, T. L.; Abell, C. A fragment merging approach towards the development of small molecule inhibitors of Mycobacterium tuberculosis EthR for use as ethionamide boosters. Org. Biomol. Chem. 2016, 14, 2318– 2326, DOI: 10.1039/C5OB02630J[Crossref], [PubMed], [CAS], Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xoslajtg%253D%253D&md5=bbe5d5a96e650841b351f2fb9d221f22A fragment merging approach towards the development of small molecule inhibitors of Mycobacterium tuberculosis EthR for use as ethionamide boostersNikiforov, Petar O.; Surade, Sachin; Blaszczyk, Michal; Delorme, Vincent; Brodin, Priscille; Baulard, Alain R.; Blundell, Tom L.; Abell, ChrisOrganic & Biomolecular Chemistry (2016), 14 (7), 2318-2326CODEN: OBCRAK; ISSN:1477-0520. (Royal Society of Chemistry)With the ever-increasing instances of resistance to frontline TB drugs there is the need to develop novel strategies to fight the worldwide TB epidemic. Boosting the effect of the existing second-line antibiotic ethionamide by inhibiting the mycobacterial transcriptional repressor protein EthR is an attractive therapeutic strategy. Herein we report the use of a fragment based drug discovery approach for the structure-guided systematic merging of two fragment mols., each binding twice to the hydrophobic cavity of EthR from M. tuberculosis. These together fill the entire binding pocket of EthR. We elaborated these fragment hits and developed small mol. inhibitors which have a 100-fold improvement of potency in vitro over the initial fragments.
- 22Schade, M.; Merla, B.; Lesch, B.; Wagener, M.; Timmermanns, S.; Pletinckx, K.; Hertrampf, T. Highly selective sub-nanomolar cathepsin S inhibitors by merging fragment binders with nitrile inhibitors. J. Med. Chem. 2020, 63, 11801– 11808, DOI: 10.1021/acs.jmedchem.0c00949[ACS Full Text
], [CAS], Google Scholar
22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhslGqsrnI&md5=f7698389131c792e3bebb40123f6ec66Highly Selective Sub-Nanomolar Cathepsin S Inhibitors by Merging Fragment Binders with Nitrile InhibitorsSchade, Markus; Merla, Beatrix; Lesch, Bernhard; Wagener, Markus; Timmermanns, Simone; Pletinckx, Katrien; Hertrampf, TorstenJournal of Medicinal Chemistry (2020), 63 (20), 11801-11808CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Pharmacol. inhibition of cathepsin S (CatS) allows for a specific modulation of the adaptive immune system and many major diseases. Here, we used NMR fragment screening and crystal structure-aided merging to synthesize novel, highly selective CatS inhibitors with picomolar enzymic Ki values and nanomolar functional activity in human Raji cells. Noncovalent fragment hits revealed binding hotspots, while the covalent inhibitor structure-activity relationship enabled efficient potency optimization. - 23de Souza Neto, L. R.; Moreira-Filho, J. T.; Neves, B. J.; Maidana, R. L. B. R.; Guimarães, A. C. R.; Furnham, N.; Andrade, C. H.; Silva, F. P. In silico strategies to support fragment-to-lead optimization in drug discovery. Front. Chem. 2020, 8, 93, DOI: 10.3389/fchem.2020.00093[Crossref], [PubMed], [CAS], Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB387psFOgtg%253D%253D&md5=3fac2ef52a5f51e4e34826f08d5e28d5In silico Strategies to Support Fragment-to-Lead Optimization in Drug Discoveryde Souza Neto Lauro Ribeiro; Maidana Rocio Lucia Beatriz Riveros; Silva Floriano Paes Jr; Moreira-Filho Jose Teofilo; Neves Bruno Junior; Andrade Carolina Horta; Neves Bruno Junior; Maidana Rocio Lucia Beatriz Riveros; Guimaraes Ana Carolina Ramos; Furnham NicholasFrontiers in chemistry (2020), 8 (), 93 ISSN:2296-2646.Fragment-based drug (or lead) discovery (FBDD or FBLD) has developed in the last two decades to become a successful key technology in the pharmaceutical industry for early stage drug discovery and development. The FBDD strategy consists of screening low molecular weight compounds against macromolecular targets (usually proteins) of clinical relevance. These small molecular fragments can bind at one or more sites on the target and act as starting points for the development of lead compounds. In developing the fragments attractive features that can translate into compounds with favorable physical, pharmacokinetics and toxicity (ADMET-absorption, distribution, metabolism, excretion, and toxicity) properties can be integrated. Structure-enabled fragment screening campaigns use a combination of screening by a range of biophysical techniques, such as differential scanning fluorimetry, surface plasmon resonance, and thermophoresis, followed by structural characterization of fragment binding using NMR or X-ray crystallography. Structural characterization is also used in subsequent analysis for growing fragments of selected screening hits. The latest iteration of the FBDD workflow employs a high-throughput methodology of massively parallel screening by X-ray crystallography of individually soaked fragments. In this review we will outline the FBDD strategies and explore a variety of in silico approaches to support the follow-up fragment-to-lead optimization of either: growing, linking, and merging. These fragment expansion strategies include hot spot analysis, druggability prediction, SAR (structure-activity relationships) by catalog methods, application of machine learning/deep learning models for virtual screening and several de novo design methods for proposing synthesizable new compounds. Finally, we will highlight recent case studies in fragment-based drug discovery where in silico methods have successfully contributed to the development of lead compounds.
- 24Pierce, A. C.; Rao, G.; Bemis, G. W. BREED:generating novel inhibitors through hybridization of known ligands. Application to CDK2, P38, and HIV protease. J. Med. Chem. 2004, 47, 2768– 2775, DOI: 10.1021/jm030543u[ACS Full Text
], [CAS], Google Scholar
24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXjt1ajurk%253D&md5=e0bec9082fab05b694742498b3f41549BREED: Generating Novel Inhibitors through Hybridization of Known Ligands. Application to CDK2, P38, and HIV ProteasePierce, Albert C.; Rao, Govinda; Bemis, Guy W.Journal of Medicinal Chemistry (2004), 47 (11), 2768-2775CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)In this work we describe BREED, a method for the generation of novel inhibitors from structures of known ligands bound to a common target. The method is essentially an automation of the common medicinal chem. practice of joining fragments of two known ligands to generate a new inhibitor. The ligand-bound target structures are overlaid, all overlapping bonds in all pairs of ligands are found, and the fragments on each side of each matching bond are swapped to generate the new mols. Since the method is automated, it can be applied recursively to generate all possible combinations of known ligands. In an application of this method to HIV protease inhibitors and protein kinase inhibitors, hundreds of new mol. structures were generated. These included known inhibitor scaffolds not included in the initial set, entirely novel scaffolds, and novel substituents on known scaffolds. The method is fast, and since all of the ligand functional groups are known to bind the target in the precise position and orientation present in the novel ligand, the success rate of this method should be superior to more traditional de novo design techniques. In an era of increasingly high-throughput structural biol., such methods for high-throughput utilization of structural information will become increasingly valuable. - 25Lindert, S.; Durrant, J. D.; McCammon, J. A. LigMerge: a fast algorithm to generate models of novel potential ligands from sets of known binders. Chem. Biol. Drug Des. 2012, 80, 358– 365, DOI: 10.1111/j.1747-0285.2012.01414.x[Crossref], [PubMed], [CAS], Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtF2itbbF&md5=07cc659c48eccc967fc3b135182e9012LigMerge: a fast algorithm to generate models of novel potential ligands from sets of known bindersLindert, Steffen; Durrant, Jacob D.; McCammon, J. AndrewChemical Biology & Drug Design (2012), 80 (3), 358-365CODEN: CBDDAL; ISSN:1747-0277. (Wiley-Blackwell)One common practice in drug discovery is to optimize known or suspected ligands to improve binding affinity. In performing these optimizations, it is useful to look at as many known inhibitors as possible for guidance. Medicinal chemists often seek to improve potency by altering certain chem. moieties of known/endogenous ligands while retaining those crit. for binding. To our knowledge, no automated, ligand-based algorithm exists for systematically "swapping" the chem. moieties of known ligands to generate novel ligands with potentially improved potency. To address this need, we have created a novel algorithm called "LigMerge". LigMerge identifies the max. (largest) common substructure of two three-dimensional ligand models, superimposes these two substructures, and then systematically mixes and matches the distinct fragments attached to the common substructure at each common atom, thereby generating multiple compd. models related to the known inhibitors that can be evaluated using computer docking prior to synthesis and exptl. testing. To demonstrate the utility of LigMerge, we identify compds. predicted to inhibit peroxisome proliferator-activated receptor gamma, HIV reverse transcriptase, and dihydrofolate reductase with affinities higher than those of known ligands. We hope that LigMerge will be a helpful tool for the drug design community.
- 26Wang, H.; Pan, X.; Zhang, Y.; Wang, X.; Xiao, X.; Ji, C. MolHyb: a web server for structure-based drug design by molecular hybridization. J. Chem. Inf. Model. 2022, 62, 2916– 2922, DOI: 10.1021/acs.jcim.2c00443[ACS Full Text
], [CAS], Google Scholar
26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhsVyjsr3I&md5=f7682a620c9ca25bfbc1142b204e20faMolHyb: A Web Server for Structure-Based Drug Design by Molecular HybridizationWang, Hao; Pan, Xiaolin; Zhang, Yueqing; Wang, Xingyu; Xiao, Xudong; Ji, ChanggeJournal of Chemical Information and Modeling (2022), 62 (12), 2916-2922CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Mol. hybridization is a widely used ligand design method in drug discovery. In this study, we present MolHyb, a web server for structure-based ligand design by mol. hybridization. The input of MolHyb is a protein file and a seed compd. file. MolHyb tries to generate novel ligands through hybridizing the seed compd. with helper compds. that bind to the same protein target or similar proteins. To facilitate the job of getting helper compds., we compiled a modeled protein-ligand structure database as an extension to crystal structures in the PDB database by placing the bioactive compds. in ChEMBL into their corresponding 3D protein binding pocket properly. MolHyb works by searching for helper compds. from the protein-ligand structure database and migrating chem. moieties from helper compds. to the seed compd. efficiently. Hybridization is performed at both cyclic and acyclic bonds. The users can also input their own helper compds. to MolHyb. We hope that MolHyb will be a useful tool for rational drug design. MolHyb is freely available at http://molhyb.xundrug.cn/. - 27Li, Y.; Zhao, Y.; Liu, Z.; Wang, R. Automatic Tailoring and Transplanting: a practical method that makes virtual screening more useful. J. Chem. Inf. Model. 2011, 51, 1474– 1491, DOI: 10.1021/ci200036m[ACS Full Text
], [CAS], Google Scholar
27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXlvFKqsb0%253D&md5=3327b42fecda9f2af07a6864d05afaa4Automatic Tailoring and Transplanting: A Practical Method that Makes Virtual Screening More UsefulLi, Yan; Zhao, Yuan; Liu, Zhihai; Wang, RenxiaoJournal of Chemical Information and Modeling (2011), 51 (6), 1474-1491CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Docking-based virtual screening of large compd. libraries has been widely applied to lead discovery in structure-based drug design. However, subsequent lead optimizations often rely on other types of computational methods, such as de novo design methods. We have developed an automatic method, namely automatic tailoring and transplanting (AutoT&T), which can effectively utilize the outcomes of virtual screening in lead optimization. This method detects suitable fragments on virtual screening hits and then transplants them onto a lead compd. to generate new ligand mols. Binding affinities, synthetic feasibilities, and drug-likeness properties are considered in the selection of final designs. In this study, our AutoT&T program was tested on three different target proteins, including p38 MAP kinase, PPAR-α, and Mcl-1. In the first two cases, AutoT&T was able to produce mols. identical or similar to known inhibitors with better potency than the given lead compd. In the third case, we demonstrated how to apply AutoT&T to design novel ligand mols. from scratch. Compared to the solns. generated by other two de novo design methods, i.e., LUDI and EA-Inventor, the solns. generated by AutoT&T were structurally more diverse and more promising in terms of binding scores in all three cases. AutoT&T also completed the assigned jobs more efficiently than LUDI and EA-Inventor by several folds. Our AutoT&T method has certain tech. advantages over de novo design methods. Importantly, it expands the application of virtual screening from lead discovery to lead optimization and thus may serve as a valuable tool for many researchers. - 28Li, Y.; Zhao, Z.; Liu, Z.; Su, M.; Wang, R. AutoT&T v.2: an efficient and versatile tool for lead structure generation and optimization. J. Chem. Inf. Model. 2016, 56, 435– 453, DOI: 10.1021/acs.jcim.5b00691[ACS Full Text
], [CAS], Google Scholar
28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xht1KgurY%253D&md5=765cef8f54f2367d902682c547abaf52AutoT&T v.2: An Efficient and Versatile Tool for Lead Structure Generation and OptimizationLi, Yan; Zhao, Zhixiong; Liu, Zhihai; Su, Minyi; Wang, RenxiaoJournal of Chemical Information and Modeling (2016), 56 (2), 435-453CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)In structure-based drug design, automated de novo design methods are helpful tools for lead discovery as well as lead optimization. In a previous study the authors reported a new de novo design method, namely, Automatic Tailoring and Transplanting (AutoT&T). It overcomes some intrinsic problems in conventional fragment-based buildup methods. In this study, the authors describe an upgraded version, namely, AutoT&T2. Structural operations conducted by AutoT&T2 have been largely optimized by introducing several new algorithms. As a result, its overall speed in multiround optimization jobs has been improved by a few thousand fold. With this improvement, it is now practical to conduct structural crossover among multiple lead mols. using AutoT&T2. Three different test cases are described in this study that demonstrate the new features and versatile applications of AutoT&T2. The AutoT&T2 software suite is available to the public. Besides, a Web portal for running AutoT&T2 online is provided at http://www.sioc-ccbg.ac.cn/software/att2 for testing. - 29Nisius, B.; Rester, U. Fragment shuffling: an automated workflow for three-dimensional fragment-based ligand design. J. Chem. Inf. Model. 2009, 49, 1211– 1222, DOI: 10.1021/ci8004572[ACS Full Text
], [CAS], Google Scholar
29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXltlKls74%253D&md5=87aae00271c326a921cf38e7770405c8Fragment Shuffling: An Automated Workflow for Three-Dimensional Fragment-Based Ligand DesignNisius, Britta; Rester, UlrichJournal of Chemical Information and Modeling (2009), 49 (5), 1211-1222CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Fragment-based approaches display a promising alternative in lead discovery. Herein, we present the automated fragment shuffling workflow for the identification of novel lead compds. combining central elements from fragment-based lead identification and structure-based de novo design. Our method is based on sets of aligned 3D ligand structures binding to the same target or target family. The implementation comprises three different ligand fragmentation methods, a scoring scheme assigning individual scores to each fragment, and the incremental construction of novel ligands based on a greedy search algorithm guided by the calcd. fragment scores. The validation of our 3D ligand design workflow is presented on the basis of two pharmaceutically relevant drug targets. A retrospective study based on a selected protein kinase data set revealed that the fragment shuffling approach realizes extended results compared to the well-known BREED technique. Furthermore, we applied our approach in a prospective study for the design of novel non-peptidic thrombin inhibitors. The designed ligand structures in both studies demonstrate the potential of the fragment shuffling workflow. - 30Maass, P.; Schulz-Gasch, T.; Stahl, M.; Rarey, M. Recore: a fast and versatile method for scaffold hopping based on small molecule crystal structure conformations. J. Chem. Inf. Model. 2007, 47, 390– 399, DOI: 10.1021/ci060094h[ACS Full Text
], [CAS], Google Scholar
30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhvVWjtbk%253D&md5=e9d4e277da49f4ffe2c21472990c2387Recore: A Fast and Versatile Method for Scaffold Hopping Based on Small Molecule Crystal Structure ConformationsMaass, Patrick; Schulz-Gasch, Tanja; Stahl, Martin; Rarey, MatthiasJournal of Chemical Information and Modeling (2007), 47 (2), 390-399CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Replacing central elements of known active structures is a common procedure to enter new compd. classes. Different computational methods have already been developed to help with this task, varying in the description of possible replacements, the query input, and the similarity measure used. In this paper, a novel approach for scaffold replacement and a corresponding software tool, called Recore, is introduced. In contrast to prior methods, the authors main objective was to combine the following three properties in one tool: to avoid structures with strained conformations, to enable the exploration of large search spaces, and to allow interactive use through short response times. The authors introduce a new technique employing 3D fragments generated by combinatorial enumeration of cuts. It allows focusing on fragments suitable for scaffold replacement while retaining conformational information of the corresponding crystal structures. Based on this idea, the authors present an algorithm utilizing a geometric rank searching approach. Given a geometric arrangement of two or three exit vectors and addnl. pharmacophore features, the algorithm finds fragments fulfilling all these constraints ordered by increasing deviation from the query constraints. For the validation of the approach, three different drug design scenarios have been used. The results obtained show that the authors approach is able to propose new valid scaffold topologies. - 31Polishchuk, P. CReM: chemically reasonable mutations framework for structure generation. J. Cheminf. 2020, 12, 28, DOI: 10.1186/s13321-020-00431-w
- 32Lim, J.; Hwang, S.-Y.; Moon, S.; Kim, S.; Kim, W. Y. Scaffold-based molecular design with a graph generative model. Chem. Sci. 2020, 11, 1153– 1164, DOI: 10.1039/C9SC04503A[Crossref], [CAS], Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXit1Ortr%252FO&md5=be36a65abcce15f18b4c1e529bffd905Scaffold-based molecular design with a graph generative modelLim, Jaechang; Hwang, Sang-Yeon; Moon, Seokhyun; Kim, Seungsu; Kim, Woo YounChemical Science (2020), 11 (4), 1153-1164CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)Searching for new mols. in areas like drug discovery often starts from the core structures of known mols. Such a method has called for a strategy of designing deriv. compds. retaining a particular scaffold as a substructure. On this account, our present work proposes a graph generative model that targets its use in scaffold-based mol. design. Our model accepts a mol. scaffold as input and extends it by sequentially adding atoms and bonds. The generated mols. are then guaranteed to contain the scaffold with certainty, and their properties can be controlled by conditioning the generation process on desired properties. The learned rule of extending mols. can well generalize to arbitrary kinds of scaffolds, including those unseen during learning. In the conditional generation of mols., our model can simultaneously control multiple chem. properties despite the search space constrained by fixing the substructure. As a demonstration, we applied our model to designing inhibitors of the epidermal growth factor receptor and show that our model can employ a simple semi-supervised extension to broaden its applicability to situations where only a small amt. of data is available.
- 33Imrie, F.; Hadfield, T. E.; Bradley, A. R.; Deane, C. M. Deep generative design with 3D pharmacophoric constraints. Chem. Sci. 2021, 12, 14577– 14589, DOI: 10.1039/D1SC02436A[Crossref], [PubMed], [CAS], Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitlSrur%252FL&md5=1e51c4d1f28c6c8dff140c4c32d817f4Deep generative design with 3D pharmacophoric constraintsImrie, Fergus; Hadfield, Thomas E.; Bradley, Anthony R.; Deane, Charlotte M.Chemical Science (2021), 12 (43), 14577-14589CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)Generative models have increasingly been proposed as a soln. to the mol. design problem. However, it has proved challenging to control the design process or incorporate prior knowledge, limiting their practical use in drug discovery. In particular, generative methods have made limited use of three-dimensional (3D) structural information even though this is crit. to binding. This work describes a method to incorporate such information and demonstrates the benefit of doing so. We combine an existing graph-based deep generative model, DeLinker, with a convolutional neural network to utilize phys.-meaningful 3D representations of mols. and target pharmacophores. We apply our model, DEVELOP, to both linker and R-group design, demonstrating its suitability for both hit-to-lead and lead optimization. The 3D pharmacophoric information results in improved generation and allows greater control of the design process. In multiple large-scale evaluations, we show that including 3D pharmacophoric constraints results in substantial improvements in the quality of generated mols. On a challenging test set derived from PDBbind, our model improves the proportion of generated mols. with high 3D similarity to the original mol. by over 300%. In addn., DEVELOP recovers 10x more of the original mols. compared to the baseline DeLinker method. Our approach is general-purpose, readily modifiable to alternate 3D representations, and can be incorporated into other generative frameworks.
- 34Hadfield, T. E.; Imrie, F.; Merritt, A.; Birchall, K.; Deane, C. M. Incorporating target-specific pharmacophoric information into deep generative models for fragment elaboration. J. Chem. Inf. Model. 2022, 62, 2280– 2292, DOI: 10.1021/acs.jcim.1c01311[ACS Full Text
], [CAS], Google Scholar
34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhtFGru7rE&md5=c639b01f0c509bbd2e20308fa044bdb5Incorporating Target-Specific Pharmacophoric Information into Deep Generative Models for Fragment ElaborationHadfield, Thomas E.; Imrie, Fergus; Merritt, Andy; Birchall, Kristian; Deane, Charlotte M.Journal of Chemical Information and Modeling (2022), 62 (10), 2280-2292CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Despite recent interest in deep generative models for scaffold elaboration, their applicability to fragment-to-lead campaigns has so far been limited. This is primarily due to their inability to account for local protein structure or a user's design hypothesis. We propose a novel method for fragment elaboration, STRIFE, that overcomes these issues. STRIFE takes as input fragment hotspot maps (FHMs) extd. from a protein target and processes them to provide meaningful and interpretable structural information to its generative model, which in turn is able to rapidly generate elaborations with complementary pharmacophores to the protein. In a large-scale evaluation, STRIFE outperforms existing, structure-unaware, fragment elaboration methods in proposing highly ligand-efficient elaborations. In addn. to automatically extg. pharmacophoric information from a protein target's FHM, STRIFE optionally allows the user to specify their own design hypotheses. - 35Arús-Pous, J.; Patronov, A.; Bjerrum, E. J.; Tyrchan, C.; Reymond, J.-L.; Chen, H.; Engkvist, O. SMILES-based deep generative scaffold decorator for de novo drug design. J. Cheminf. 2020, 12, 38, DOI: 10.1186/s13321-020-00441-8[Crossref], [CAS], Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVyksLjK&md5=05a686a14f942874fd6f6ddbc85017a1SMILES-based deep generative scaffold decorator for de-novo drug designArus-Pous, Josep; Patronov, Atanas; Bjerrum, Esben Jannik; Tyrchan, Christian; Reymond, Jean-Louis; Chen, Hongming; Engkvist, OlaJournal of Cheminformatics (2020), 12 (1), 38CODEN: JCOHB3; ISSN:1758-2946. (SpringerOpen)Herein we report a new SMILES-based mol. generative architecture that generates mols. from scaffolds and can be trained from any arbitrary mol. set. This approach is possible thanks to a new mol. set pre-processing algorithm that exhaustively slices all possible combinations of acyclic bonds of every mol., combinatorically obtaining a large no. of scaffolds with their resp. decorations. Two examples showcasing the potential of the architecture in medicinal and synthetic chem. are described: First, models were trained with a training set obtained from a small set of Dopamine Receptor D2 active modulators and were able to meaningfully decorate a wide range of scaffolds and obtain mol. series predicted active on DRD2. Second, a larger set of drug-like mols. from ChEMBL was selectively sliced using synthetic chem. constraints. This filtering process allowed models trained with this dataset to selectively decorate diverse scaffolds with fragments that were generally predicted to be synthesizable and attachable to the scaffold using known synthetic approaches. In both cases, the models were already able to decorate mols. using specific knowledge without the need to add it with other techniques, such as reinforcement learning. We envision that this architecture will become a useful addn. to the already existent architectures for de novo mol. generation.
- 36Fialková, V.; Zhao, J.; Papadopoulos, K.; Engkvist, O.; Bjerrum, E. J.; Kogej, T.; Patronov, A. LibINVENT: reaction-based generative scaffold decoration for in silico library design. J. Chem. Inf. Model. 2022, 62, 2046– 2063, DOI: 10.1021/acs.jcim.1c00469[ACS Full Text
], [CAS], Google Scholar
36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvV2iu7vI&md5=dc7b0da044f6aa1c2554a43e0aa14672LibINVENT: Reaction-based Generative Scaffold Decoration for in Silico Library DesignFialkova, Vendy; Zhao, Jiaxi; Papadopoulos, Kostas; Engkvist, Ola; Bjerrum, Esben Jannik; Kogej, Thierry; Patronov, AtanasJournal of Chemical Information and Modeling (2022), 62 (9), 2046-2063CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Because of the strong relationship between the desired mol. activity and its structural core, the screening of focused, core-sharing chem. libraries is a key step in lead optimization. Despite the plethora of current research focused on in silico methods for mol. generation, to our knowledge, no tool capable of designing such libraries has been proposed. In this work, we present a novel tool for de novo drug design called LibINVENT. It is capable of rapidly proposing chem. libraries of compds. sharing the same core while maximizing a range of desirable properties. To further help the process of designing focused libraries, the user can list specific chem. reactions that can be used for the library creation. LibINVENT is therefore a flexible tool for generating virtual chem. libraries for lead optimization in a broad range of scenarios. Addnl., the shared core ensures that the compds. in the library are similar, possess desirable properties, and can also be synthesized under the same or similar conditions. The LibINVENT code is freely available in our public repository at https://github.com/MolecularAI/Lib-INVENT. The code necessary for data preprocessing is further available at: https://github.com/MolecularAI/Lib-INVENT-dataset. - 37Li, Y.; Hu, J.; Wang, Y.; Zhou, J.; Zhang, L.; Liu, Z. DeepScaffold: a comprehensive tool for scaffold-based de novo drug discovery using deep learning. J. Chem. Inf. Model. 2020, 60, 77– 91, DOI: 10.1021/acs.jcim.9b00727[ACS Full Text
], [CAS], Google Scholar
37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitlerurnN&md5=44ba7010e4824a37d97c66eed5bd1d6dDeepScaffold: A Comprehensive Tool for Scaffold-Based De Novo Drug Discovery Using Deep LearningLi, Yibo; Hu, Jianxing; Wang, Yanxing; Zhou, Jielong; Zhang, Liangren; Liu, ZhenmingJournal of Chemical Information and Modeling (2020), 60 (1), 77-91CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)The ultimate goal of drug design is to find novel compds. with desirable pharmacol. properties. Designing mols. retaining particular scaffolds as their core structures is an efficient way to obtain potential drug candidates. We propose a scaffold-based mol. generative model for drug discovery, which performs mol. generation based on a wide spectrum of scaffold definitions, including Bemis-Murcko scaffolds, cyclic skeletons, and scaffolds with specifications on side-chain properties. The model can generalize the learned chem. rules of adding atoms and bonds to a given scaffold. The generated compds. were evaluated by mol. docking in DRD2 targets, and the results demonstrated that this approach can be effectively applied to solve several drug design problems, including the generation of compds. contg. a given scaffold and de novo drug design of potential drug candidates with specific docking scores. - 38Yang, Y.; Zheng, S.; Su, S.; Zhao, C.; Xu, J.; Chen, H. SyntaLinker: automatic fragment linking with deep conditional transformer neural networks. Chem. Sci. 2020, 11, 8312– 8322, DOI: 10.1039/D0SC03126G[Crossref], [PubMed], [CAS], Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVagsrvK&md5=403cc7a09f96ecbe27afa3ce6bee51a2SyntaLinker: automatic fragment linking with deep conditional transformer neural networksYang, Yuyao; Zheng, Shuangjia; Su, Shimin; Zhao, Chao; Xu, Jun; Chen, HongmingChemical Science (2020), 11 (31), 8312-8322CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)Linking fragments to generate a focused compd. library for a specific drug target is one of the challenges in fragment-based drug design (FBDD). Hereby, we propose a new program named SyntaLinker, which is based on a syntactic pattern recognition approach using deep conditional transformer neural networks. This state-of-the-art transformer can link mol. fragments automatically by learning from the knowledge of structures in medicinal chem. databases (e.g.ChEMBL database). Conventionally, linking mol. fragments was viewed as connecting substructures that were predefined by empirical rules. In SyntaLinker, however, the rules of linking fragments can be learned implicitly from known chem. structures by recognizing syntactic patterns embedded in SMILES notations. With deep conditional transformer neural networks, SyntaLinker can generate mol. structures based on a given pair of fragments and addnl. restrictions. Case studies have demonstrated the advantages and usefulness of SyntaLinker in FBDD.
- 39Feng, Y.; Yang, Y.; Deng, W.; Chen, H.; Ran, T. SyntaLinker-Hybrid: a deep learning approach for target specific drug design. Artif. Intell. Life Sci. 2022, 2, 100035, DOI: 10.1016/j.ailsci.2022.100035
- 40Huang, Y.; Peng, X.; Ma, J.; Zhang, M. 3DLinker: an E(3) equivariant variational autoencoder for molecular linker design. arXiv Preprint , arXiv:2205.07309, 2022.Google ScholarThere is no corresponding record for this reference.
- 41Imrie, F.; Bradley, A. R.; van der Schaar, M.; Deane, C. M. Deep generative models for 3D linker design. J. Chem. Inf. Model. 2020, 60, 1983– 1995, DOI: 10.1021/acs.jcim.9b01120[ACS Full Text
], [CAS], Google Scholar
41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXltlenurs%253D&md5=430c929761c5020d1013538e028ba1bcDeep Generative Models for 3D Linker DesignImrie, Fergus; Bradley, Anthony R.; van der Schaar, Mihaela; Deane, Charlotte M.Journal of Chemical Information and Modeling (2020), 60 (4), 1983-1995CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Rational compd. design remains a challenging problem for both computational methods and medicinal chemists. Computational generative methods have begun to show promising results for the design problem. However, they have not yet used the power of three-dimensional (3D) structural information. We have developed a novel graph-based deep generative model that combines state-of-the-art machine learning techniques with structural knowledge. Our method ("DeLinker") takes two fragments or partial structures and designs a mol. incorporating both. The generation process is protein-context-dependent, utilizing the relative distance and orientation between the partial structures. This 3D information is vital to successful compd. design, and we demonstrate its impact on the generation process and the limitations of omitting such information. In a large-scale evaluation, DeLinker designed 60% more mols. with high 3D similarity to the original mol. than a database baseline. When considering the more relevant problem of longer linkers with at least five atoms, the outperformance increased to 200%. We demonstrate the effectiveness and applicability of this approach on a diverse range of design problems: fragment linking, scaffold hopping, and proteolysis targeting chimera (PROTAC) design. As far as we are aware, this is the first mol. generative model to incorporate 3D structural information directly in the design process. The code is available at https://github.com/oxpig/DeLinker. - 42Andrianov, G. V.; Gabriel Ong, W. J.; Serebriiskii, I.; Karanicolas, J. Efficient Hit-to-Lead Searching of Kinase Inhibitor Chemical Space via Computational Fragment Merging. J. Chem. Inf. Model. 2021, 61, 5967– 5987, DOI: 10.1021/acs.jcim.1c00630[ACS Full Text
], [CAS], Google Scholar
42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXisVGrs7zE&md5=d00fcbed94deb5955f19c3ee8d26af39Efficient Hit-to-Lead Searching of Kinase Inhibitor Chemical Space via Computational Fragment MergingAndrianov, Grigorii V.; Gabriel Ong, Wern Juin; Serebriiskii, Ilya; Karanicolas, JohnJournal of Chemical Information and Modeling (2021), 61 (12), 5967-5987CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)In early-stage drug discovery, the hit-to-lead optimization (or "hit expansion") stage entails starting from a newly identified active compd. and improving its potency or other properties. Traditionally, this process relies on synthesizing and evaluating a series of analogs to build up structure-activity relationships. Here, we describe a computational strategy focused on kinase inhibitors, intended to expedite the process of identifying analogs with improved potency. Our protocol begins from an inhibitor of the target kinase and generalizes the synthetic route used to access it. By searching for com. available replacements for the individual building blocks used to make the parent inhibitor, we compile an enumerated library of compds. that can be accessed using the same chem. transformations; these huge libraries can exceed many millions-or billions-of compds. Because the resulting libraries are much too large for explicit virtual screening, we instead consider alternate approaches to identify the top-scoring compds. We find that contributions from individual substituents are well described by a pairwise additivity approxn., provided that the corresponding fragments position their shared core in precisely the same way relative to the binding site. This key insight allows us to det. which fragments are suitable for merging into single new compds. and which are not. Further, the use of pairwise approxn. allows interaction energies to be assigned to each compd. in the library without the need for any further structure-based modeling: interaction energies instead can be reliably estd. from the energies of the component fragments, and the reduced computational requirements allow for flexible energy minimizations that allow the kinase to respond to each substitution. We demonstrate this protocol using libraries built from six representative kinase inhibitors drawn from the literature, which target five different kinases: CDK9, CHK1, CDK2, EGFRT790M, and ACK1. In each example, the enumerated library includes addnl. analogs reported by the original study to have activity, and these analogs are successfully prioritized within the library. We envision that the insights from this work can facilitate the rapid assembly and screening of increasingly large libraries for focused hit-to-lead optimization. To enable adoption of these methods and to encourage further analyses, we disseminate the computational tools needed to deploy this protocol. - 43Hall, R. J.; Murray, C. W.; Verdonk, M. L. The Fragment Network: a chemistry recommendation engine built using a graph database. J. Med. Chem. 2017, 60, 6440– 6450, DOI: 10.1021/acs.jmedchem.7b00809[ACS Full Text
], [CAS], Google Scholar
43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFKlsr3J&md5=79842771a068cdaabc2f9a841d791c1cThe Fragment Network: A Chemistry Recommendation Engine Built Using a Graph DatabaseHall, Richard J.; Murray, Christopher W.; Verdonk, Marcel L.Journal of Medicinal Chemistry (2017), 60 (14), 6440-6450CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The hit validation stage of a fragment-based drug discovery campaign involves probing the SAR around one or more fragment hits. This often requires a search for similar compds. in a corporate collection or from com. suppliers. The Fragment Network is a graph database that allows a user to efficiently search chem. space around a compd. of interest. The result set is chem. intuitive, naturally grouped by substitution pattern and meaningfully sorted according to the no. of observations of each transformation in medicinal chem. databases. This paper describes the algorithms used to construct and search the Fragment Network and provides examples of how it may be used in a drug discovery context. - 44Landrum, G. RDKit: A software suite for cheminformatics, computational chemistry, and predictive modeling. https://www.rdkit.org/ (accessed November 2022).Google ScholarThere is no corresponding record for this reference.
- 45Neo4j, Graph Modeling Guidelines. https://neo4j.com/developer/guide-data-modeling/ (accessed November 2022).Google ScholarThere is no corresponding record for this reference.
- 46Newman, J. A.; Douangamath, A.; Yadzani, S.; Yosaatmadja, Y.; Aimon, A.; Brandão-Neto, J.; Dunnett, L.; Gorrie-stone, T.; Skyner, R.; Fearon, D.; Schapira, M.; von Delft, F.; Gileadi, O. Structure, mechanism and crystallographic fragment screening of the SARS-CoV-2 NSP13 helicase. Nat. Commun. 2021, 12, 4848, DOI: 10.1038/s41467-021-25166-6[Crossref], [PubMed], [CAS], Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhslOns7%252FI&md5=3e643800631b8bb56328c4ddd97fb7a7Structure, mechanism and crystallographic fragment screening of the SARS-CoV-2 NSP13 helicaseNewman, Joseph A.; Douangamath, Alice; Yadzani, Setayesh; Yosaatmadja, Yuliana; Aimon, Antony; Brandao-Neto, Jose; Dunnett, Louise; Gorrie-stone, Tyler; Skyner, Rachael; Fearon, Daren; Schapira, Matthieu; von Delft, Frank; Gileadi, OpherNature Communications (2021), 12 (1), 4848CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Abstr.: There is currently a lack of effective drugs to treat people infected with SARS-CoV-2, the cause of the global COVID-19 pandemic. The SARS-CoV-2 Non-structural protein 13 (NSP13) has been identified as a target for anti-virals due to its high sequence conservation and essential role in viral replication. Structural anal. reveals two "druggable" pockets on NSP13 that are among the most conserved sites in the entire SARS-CoV-2 proteome. Here we present crystal structures of SARS-CoV-2 NSP13 solved in the APO form and in the presence of both phosphate and a non-hydrolysable ATP analog. Comparisons of these structures reveal details of conformational changes that provide insights into the helicase mechanism and possible modes of inhibition. To identify starting points for drug development we have performed a crystallog. fragment screen against NSP13. The screen reveals 65 fragment hits across 52 datasets opening the way to structure guided development of novel antiviral agents.
- 47Jin, Z.; Du, X.; Xu, Y.; Deng, Y.; Liu, M.; Zhao, Y.; Zhang, B.; Li, X.; Zhang, L.; Peng, C.; Duan, Y.; Yu, J.; Wang, L.; Yang, K.; Liu, F.; Jiang, R.; Yang, X.; You, T.; Liu, X.; Yang, X.; Bai, F.; Liu, H.; Liu, X.; Guddat, L. W.; Xu, W.; Xiao, G.; Qin, C.; Shi, Z.; Jiang, H.; Rao, Z.; Yang, H. Structure of Mpro from SARS-CoV-2 and discovery of its inhibitors. Nature 2020, 582, 289– 293, DOI: 10.1038/s41586-020-2223-y[Crossref], [PubMed], [CAS], Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVyhsrrO&md5=b84f350fe9ce1109485df6caf814ba82Structure of Mpro from SARS-CoV-2 and discovery of its inhibitorsJin, Zhenming; Du, Xiaoyu; Xu, Yechun; Deng, Yongqiang; Liu, Meiqin; Zhao, Yao; Zhang, Bing; Li, Xiaofeng; Zhang, Leike; Peng, Chao; Duan, Yinkai; Yu, Jing; Wang, Lin; Yang, Kailin; Liu, Fengjiang; Jiang, Rendi; Yang, Xinglou; You, Tian; Liu, Xiaoce; Yang, Xiuna; Bai, Fang; Liu, Hong; Liu, Xiang; Guddat, Luke W.; Xu, Wenqing; Xiao, Gengfu; Qin, Chengfeng; Shi, Zhengli; Jiang, Hualiang; Rao, Zihe; Yang, HaitaoNature (London, United Kingdom) (2020), 582 (7811), 289-293CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Abstr.: A new coronavirus, known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is the etiol. agent responsible for the 2019-2020 viral pneumonia outbreak of coronavirus disease 2019 (COVID-19). Currently, there are no targeted therapeutic agents for the treatment of this disease, and effective treatment options remain very limited. Here, we describe the results of a program that aimed to rapidly discover lead compds. for clin. use, by combining structure-assisted drug design, virtual drug screening and high-throughput screening. This program focused on identifying drug leads that target main protease (Mpro) of SARS-CoV-2: Mpro is a key enzyme of coronaviruses and has a pivotal role in mediating viral replication and transcription, making it an attractive drug target for SARS-CoV-2. We identified a mechanism-based inhibitor (N3) by computer-aided drug design, and then detd. the crystal structure of Mpro of SARS-CoV-2 in complex with this compd. Through a combination of structure-based virtual and high-throughput screening, we assayed more than 10,000 compds.-including approved drugs, drug candidates in clin. trials and other pharmacol. active compds.-as inhibitors of Mpro. Six of these compds. inhibited Mpro, showing half-maximal inhibitory concn. values that ranged from 0.67 to 21.4μM. One of these compds. (ebselen) also exhibited promising antiviral activity in cell-based assays. Our results demonstrate the efficacy of our screening strategy, which can lead to the rapid discovery of drug leads with clin. potential in response to new infectious diseases for which no specific drugs or vaccines are available.
- 48The COVID Moonshot Consortium. Achdout, H.; Aimon, A.; Bar-David, E.; Barr, H.; Ben-Shmuel, A.; Bennett, J.; Bilenko, V. A.; Bilenko, V. A.; Boby, M. L.; Borden, B.; Bowman, G. R.; Brun, J.; BVNBS, S.; Calmiano, M.; Carbery, A.; Carney, D.; Cattermole, E.; Chang, E.; Chernyshenko, E.; Chodera, J. D.; Clyde, A.; Coffland, J. E.; Cohen, G.; Cole, J.; Contini, A.; Cox, L.; Cvitkovic, M.; Dias, A.; Donckers, K.; Dotson, D. L.; Douangamath, A.; Duberstein, S.; Dudgeon, T.; Dunnett, L.; Eastman, P. K.; Erez, N.; Eyermann, C. J.; Fairhead, M.; Fate, G.; Fearon, D.; Fedorov, O.; Ferla, M.; Fernandes, R. S.; Ferrins, L.; Foster, R.; Foster, H.; Gabizon, R.; Garcia-Sastre, A.; Gawriljuk, V. O.; Gehrtz, P.; Gileadi, C.; Giroud, C.; Glass, W. G.; Glen, R.; Glinert, I.; Godoy, A. S.; Gorichko, M.; Gorrie-Stone, T.; Griffen, E. J.; Hart, S. H.; Heer, J.; Henry, M.; Hill, M.; Horrell, S.; Huliak, V. D.; Hurley, M. F.; Israely, T.; Jajack, A.; Jansen, J.; Jnoff, E.; Jochmans, D.; John, T.; Jonghe, S. D.; Kantsadi, A. L.; Kenny, P. W.; Kiappes, J. L.; Kinakh, S. O.; Koekemoer, L.; Kovar, B.; Krojer, T.; Lee, A.; Lefker, B. A.; Levy, H.; Logvinenko, I. G.; London, N.; Lukacik, P.; Macdonald, H. B.; MacLean, B.; Malla, T. R.; Matviiuk, T.; McCorkindale, W.; McGovern, B. L.; Melamed, S.; Melnykov, K. P.; Michurin, O.; Mikolajek, H.; Milne, B. F.; Morris, A.; Morris, G. M.; Morwitzer, M. J.; Moustakas, D.; Nakamura, A. M.; Neto, J. B.; Neyts, J.; Nguyen, L.; Noske, G. D.; Oleinikovas, V.; Oliva, G.; Overheul, G. J.; Owen, D.; Pai, R.; Pan, J.; Paran, N.; Perry, B.; Pingle, M.; Pinjari, J.; Politi, B.; Powell, A.; Psenak, V.; Puni, R.; Rangel, V. L.; Reddi, R. N.; Reid, S. P.; Resnick, E.; Ripka, E. G.; Robinson, M. C.; Robinson, R. P.; Rodriguez-Guerra, J.; Rosales, R.; Rufa, D.; Saar, K.; Saikatendu, K. S.; Schofield, C.; Shafeev, M.; Shaikh, A.; Shi, J.; Shurrush, K.; Singh, S.; Sittner, A.; Skyner, R.; Smalley, A.; Smeets, B.; Smilova, M. D.; Solmesky, L. J.; Spencer, J.; Strain-Damerell, C.; Swamy, V.; Tamir, H.; Tennant, R.; Thompson, W.; Thompson, A.; Tomasio, S.; Tsurupa, I. S.; Tumber, A.; Vakonakis, I.; van Rij, R. P.; Vangeel, L.; Varghese, F. S.; Vaschetto, M.; Vitner, E. B.; Voelz, V.; Volkamer, A.; von Delft, F.; von Delft, A.; Walsh, M.; Ward, W.; Weatherall, C.; Weiss, S.; White, K. M.; Wild, C. F.; Wittmann, M.; Wright, N.; Yahalom-Ronen, Y.; Zaidmann, D.; Zidane, H.; Zitzmann, N. Open science discovery of oral non-covalent SARS-CoV-2 main protease inhibitor therapeutics. bioRxiv Preprint , updated version, 2022. DOI: 10.1101/2020.10.29.339317
- 49Wahlberg, E.; Karlberg, T.; Kouznetsova, E.; Markova, N.; Macchiarulo, A.; Thorsell, A.-G.; Pol, E.; Frostell, s.; Ekblad, T.; Öncü, D.; Kull, B.; Robertson, G. M.; Pellicciari, R.; Schüler, H.; Weigelt, J. Family-wide chemical profiling and structural analysis of PARP and tankyrase inhibitors. Nat. Biotechnol. 2012, 30, 283– 288, DOI: 10.1038/nbt.2121[Crossref], [PubMed], [CAS], Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XisVKrsb4%253D&md5=71f17fd5b6bb602dd70c8fcc17df95f9Family-wide chemical profiling and structural analysis of PARP and tankyrase inhibitorsWahlberg, Elisabet; Karlberg, Tobias; Kouznetsova, Ekaterina; Markova, Natalia; Macchiarulo, Antonio; Thorsell, Ann-Gerd; Pol, Ewa; Frostell, Aasa; Ekblad, Torun; Oencue, Delal; Kull, Bjoern; Robertson, Graeme Michael; Pellicciari, Roberto; Schueler, Herwig; Weigelt, JohanNature Biotechnology (2012), 30 (3), 283-288CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)Inhibitors of poly-ADP-ribose polymerase (PARP) family proteins are currently in clin. trials as cancer therapeutics, yet the specificity of many of these compds. is unknown. Here we evaluated a series of 185 small-mol. inhibitors, including research reagents and compds. being tested clin., for the ability to bind to the catalytic domains of 13 of the 17 human PARP family members including the tankyrases, TNKS1 and TNKS2. Many of the best-known inhibitors, including TIQ-A, 6(5H)-phenanthridinone, olaparib, ABT-888 and rucaparib, bound to several PARP family members, suggesting that these mols. lack specificity and have promiscuous inhibitory activity. We also detd. X-ray crystal structures for five TNKS2 ligand complexes and four PARP14 ligand complexes. In addn. to showing that the majority of PARP inhibitors bind multiple targets, these results provide insight into the design of new inhibitors.
- 50Ohara-Nemoto, Y.; Shimoyama, Y.; Kimura, S.; Kon, A.; Haraga, H.; Ono, T.; Nemoto, T. K. Asp- and Glu-specific novel dipeptidyl peptidase 11 of Porphyromonas gingivalis ensures utilization of proteinaceous energy sources. J. Biol. Chem. 2011, 286, 38115– 38127, DOI: 10.1074/jbc.M111.278572[Crossref], [PubMed], [CAS], Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtlyktb%252FK&md5=419046327fe3bfdfb5619d722d9f9755Asp- and Glu-specific Novel Dipeptidyl Peptidase 11 of Porphyromonas gingivalis Ensures Utilization of Proteinaceous Energy SourcesOhara-Nemoto, Yuko; Shimoyama, Yu; Kimura, Shigenobu; Kon, Asako; Haraga, Hiroshi; Ono, Toshio; Nemoto, Takayuki K.Journal of Biological Chemistry (2011), 286 (44), 38115-38127, S38115/1-S38115/9CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Porphyromonas gingivalis and Porphyromonas endodontalis, asaccharolytic black-pigmented anaerobes, are predominant pathogens of human chronic and periapical periodontitis, resp. They incorporate di- and tripeptides from the environment as carbon and energy sources. In the present study we cloned a novel dipeptidyl peptidase (DPP) gene of P. endodontalis ATCC 35406, designated as DPP11. The DPP11 gene encoded 717 amino acids with a mol. mass of 81,090 Da and was present as a 75-kDa form with an N terminus of Asp22. A homol. search revealed the presence of a P. gingivalis ortholog, PGN0607, that has been categorized as an isoform of authentic DPP7. P. gingivalis DPP11 was exclusively cell-assocd. as a truncated 60-kDa form, and the gene ablation retarded cell growth. DPP11 specifically removed dipeptides from oligopeptides with the penultimate N-terminal Asp and Glu and has a P2-position preference to hydrophobic residues. Optimum pH was 7.0, and the kcat/Km value was higher for Asp than Glu. Those activities were lost by substitution of Ser652 in P. endodontalis and Ser655 in P. gingivalis DPP11 to Ala, and they were consistently decreased with increasing NaCl concn. Arg670 is a unique amino acid completely conserved in all DPP11 members distributed in the genera Porphyromonas, Bacteroides, and Parabacteroides, whereas this residue is converted to Gly in all authentic DPP7 members. Substitution anal. suggested that Arg670 interacts with an acidic residue of the substrate. Considered to preferentially utilize acidic amino acids, DPP11 ensures efficient degrdn. of oligopeptide substrates in these Gram-neg. anaerobic rods.
- 51Lipinski, C. A.; Lombardo, F.; Dominy, B. W.; Feeney, P. J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Delivery Rev. 1997, 23, 3– 25, DOI: 10.1016/S0169-409X(96)00423-1[Crossref], [CAS], Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXktlKlsQ%253D%253D&md5=405f70b0594d428f1275e1d56642cd3aExperimental and computational approaches to estimate solubility and permeability in drug discovery and development settingsLipinski, Christopher A.; Lombardo, Franco; Dominy, Beryl W.; Feeney, Paul J.Advanced Drug Delivery Reviews (1997), 23 (1-3), 3-25CODEN: ADDREP; ISSN:0169-409X. (Elsevier)A review with 50 refs. Exptl. and computational approaches to est. soly. and permeability in discovery and development settings are described. In the discovery setting 'the rule of 5' predicts that poor absorption or permeation is more likely when there are more than 5 H-bond donors, 10 H-bond acceptors, the mol. wt. (MWT) is >500 and the calcd. Log P (CLogP) is >5. Computational methodol. for the rule-based Moriguchi Log P (MLogP) calcn. is described. Turbidimetric soly. measurement is described and applied to known drugs. High throughput screening (HTS) leads tend to have higher MWT and Log P and lower turbidimetric soly. than leads in the pre-HTS era. In the development setting, soly. calcns. focus on exact value prediction and are difficult because of polymorphism. Recent work on linear free energy relationships and Log P approaches are critically reviewed. Useful predictions are possible in closely related analog series when coupled with exptl. thermodn. soly. measurements.
- 52Veber, D. F.; Johnson, S. R.; Cheng, H.-Y.; Smith, B. R.; Ward, K. W.; Kopple, K. D. Molecular properties that influence the oral bioavailability of drug candidates. J. Med. Chem. 2002, 45, 2615– 2623, DOI: 10.1021/jm020017n[ACS Full Text
], [CAS], Google Scholar
52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XjsFCmt7g%253D&md5=eaad26ed6a259de82ad65a8834fc397dMolecular Properties That Influence the Oral Bioavailability of Drug CandidatesVeber, Daniel F.; Johnson, Stephen R.; Cheng, Hung-Yuan; Smith, Brian R.; Ward, Keith W.; Kopple, Kenneth D.Journal of Medicinal Chemistry (2002), 45 (12), 2615-2623CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Oral bioavailability measurements in rats for over 1100 drug candidates studied at Smith-Kline Beecham Pharmaceuticals (now Glaxo Smith-Kline) have allowed us to analyze the relative importance of mol. properties considered to influence that drug property. Reduced mol. flexibility, as measured by the no. of rotatable bonds, and low polar surface area or total hydrogen bond count (sum of donors and acceptors) are found to be important predictors of good oral bioavailability, independent of mol. wt. That on av. both the no. of rotatable bonds and polar surface area or hydrogen bond count tend to increase with mol. wt. may in part explain the success of the mol. wt. parameter in predicting oral bioavailability. The commonly applied mol. wt. cutoff at 500 does not itself significantly sep. compds. with poor oral bioavailability from those with acceptable values in this extensive data set. Our observations suggest that compds. which meet only the 2 criteria of (1) 10 or fewer rotatable bonds and (2) polar surface area ≤140 Å2 (or 12 or fewer H-bond donors and acceptors) will have a high probability of good oral bioavailability in the rat. Data sets for the artificial membrane permeation rate and for clearance in the rat were also examd. Reduced polar surface area correlates better with increased permeation rate than does lipophilicity (C log P), and increased rotatable bond count has a neg. effect on the permeation rate. A threshold permeation rate is a prerequisite of oral bioavailability. The rotatable bond count does not correlate with the data examd. here for the in vivo clearance rate in the rat. - 53Gaulton, A.; Bellis, L. J.; Bento, A. P.; Chambers, J.; Davies, M.; Hersey, A.; Light, Y.; McGlinchey, S.; Michalovich, D.; Al-Lazikani, B.; Overington, J. P. ChEMBL: a large-scale bioactivity database for drug discovery. Nucleic Acids Res. 2012, 40, D1100– D1107, DOI: 10.1093/nar/gkr777[Crossref], [PubMed], [CAS], Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhs12htbjN&md5=aedf7793e1ca54b6a4fa272ea3ef7d0eChEMBL: a large-scale bioactivity database for drug discoveryGaulton, Anna; Bellis, Louisa J.; Bento, A. Patricia; Chambers, Jon; Davies, Mark; Hersey, Anne; Light, Yvonne; McGlinchey, Shaun; Michalovich, David; Al-Lazikani, Bissan; Overington, John P.Nucleic Acids Research (2012), 40 (D1), D1100-D1107CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)ChEMBL is an Open Data database contg. binding, functional and ADMET information for a large no. of drug-like bioactive compds. These data are manually abstracted from the primary published literature on a regular basis, then further curated and standardized to maximize their quality and utility across a wide range of chem. biol. and drug-discovery research problems. Currently, the database contains 5.4 million bioactivity measurements for more than 1 million compds. and 5200 protein targets. Access is available through a web-based interface, data downloads and web services at: https://www.ebi.ac.uk/chembldb.
- 54Ebejer, J.-P.; Morris, G. M.; Deane, C. M. Freely available conformer generation methods: how good are they?. J. Chem. Inf. Model. 2012, 52, 1146– 1158, DOI: 10.1021/ci2004658[ACS Full Text
], [CAS], Google Scholar
54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xlt1Snu7s%253D&md5=2990e7dcfc83d8c0f7671250c51aeb97Freely Available Conformer Generation Methods: How Good Are They?Ebejer, Jean-Paul; Morris, Garrett M.; Deane, Charlotte M.Journal of Chemical Information and Modeling (2012), 52 (5), 1146-1158CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)A review. Conformer generation has important implications in cheminformatics, particularly in computational drug discovery where the quality of conformer generation software may affect the outcome of a virtual screening exercise. We examine the performance of four freely available small mol. conformer generation tools (Balloon, Confab, Frog2, and RDKit) alongside a com. tool (MOE). The aim of this study is 3-fold: (i) to identify which tools most accurately reproduce exptl. detd. structures; (ii) to examine the diversity of the generated conformational set; and (iii) to benchmark the computational time expended. These aspects were tested using a set of 708 drug-like mols. assembled from the OMEGA validation set and the Astex Diverse Set. These mols. have varying physicochem. properties and at least one known X-ray crystal structure. We found that RDKit and Confab are statistically better than other methods at generating low rmsd conformers to the known structure. RDKit is particularly suited for less flexible mols. while Confab, with its systematic approach, is able to generate conformers which are geometrically closer to the exptl. detd. structure for mols. with a large no. of rotatable bonds (≥10). In our tests RDKit also resulted as the second fastest method after Frog2. In order to enhance the performance of RDKit, we developed a postprocessing algorithm to build a diverse and representative set of conformers which also contains a close conformer to the known structure. Our anal. indicates that, with postprocessing, RDKit is a valid free alternative to com., proprietary software. - 55Su, M.; Yang, Q.; Du, Y.; Feng, G.; Liu, Z.; Li, Y.; Wang, R. Comparative assessment of scoring functions: the CASF-2016 update. J. Chem. Inf. Model. 2019, 59, 895– 913, DOI: 10.1021/acs.jcim.8b00545[ACS Full Text
], [CAS], Google Scholar
55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitlWhtLjM&md5=573295cfd2298a2dee933027041c3d9cComparative Assessment of Scoring Functions: The CASF-2016 UpdateSu, Minyi; Yang, Qifan; Du, Yu; Feng, Guoqin; Liu, Zhihai; Li, Yan; Wang, RenxiaoJournal of Chemical Information and Modeling (2019), 59 (2), 895-913CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)In structure-based drug design, scoring functions are often employed to evaluate protein-ligand interactions. A variety of scoring functions have been developed so far, and thus, some objective benchmarks are desired for assessing their strength and weakness. The comparative assessment of scoring functions (CASF) benchmark developed by us provides an answer to this demand. CASF is designed as a "scoring benchmark", where the scoring process is decoupled from the docking process to depict the performance of scoring function more precisely. Here, we describe the latest update of this benchmark, i.e., CASF-2016. Each scoring function is still evaluated by four metrics, including "scoring power", "ranking power", "docking power", and "screening power". Nevertheless, the evaluation methods have been improved considerably in several aspects. A new test set is compiled, which consists of 285 protein-ligand complexes with high-quality crystal structures and reliable binding consts. A panel of 25 scoring functions are tested on CASF-2016 as a demonstration. Our results reveal that the performance of current scoring functions is more promising in terms of docking power than scoring, ranking, and screening power. Scoring power is somewhat correlated with ranking power, so are docking power and screening power. The results obtained on CASF-2016 may provide valuable guidance for the end users to make smart choices among available scoring functions. Moreover, CASF is created as an open-access benchmark so that other researchers can utilize it to test a wider range of scoring functions. The complete CASF-2016 benchmark will be released on the PDBbind-CN web server (http://www.pdbbind-cn.org/casf.asp/) once this article is published. - 56Ferla, M. Fragmenstein. https://github.com/matteoferla/Fragmenstein (accessed November 2022).Google ScholarThere is no corresponding record for this reference.
- 57Chaudhury, S.; Lyskov, S.; Gray, J. J. PyRosetta: a script-based interface for implementing molecular modeling algorithms using Rosetta. Bioinformatics 2010, 26, 689– 691, DOI: 10.1093/bioinformatics/btq007[Crossref], [PubMed], [CAS], Google Scholar57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXis1Wnt70%253D&md5=261b3b6b59e4987911556ea5314d9b19PyRosetta: a script-based interface for implementing molecular modeling algorithms using RosettaChaudhury, Sidhartha; Lyskov, Sergey; Gray, Jeffrey J.Bioinformatics (2010), 26 (5), 689-691CODEN: BOINFP; ISSN:1367-4803. (Oxford University Press)Summary: PyRosetta is a stand-alone Python-based implementation of the Rosetta mol. modeling package that allows users to write custom structure prediction and design algorithms using the major Rosetta sampling and scoring functions. PyRosetta contains Python bindings to libraries that define Rosetta functions including those for accessing and manipulating protein structure, calcg. energies and running Monte Carlo-based simulations. PyRosetta can be used in two ways: (i) interactively, using iPython and (ii) script-based, using Python scripting. Interactive mode contains a no. of help features and is ideal for beginners while script-mode is best suited for algorithm development. PyRosetta has similar computational performance to Rosetta, can be easily scaled up for cluster applications and has been implemented for algorithms demonstrating protein docking, protein folding, loop modeling and design. Availability: PyRosetta is a stand-alone package available at http://www.pyrosetta.org under the Rosetta license which is free for academic and non-profit users. A tutorial, user's manual and sample scripts demonstrating usage are also available on the web site.
- 58Adasme, M. F.; Linnemann, K. L.; Bolz, S. N.; Kaiser, F.; Salentin, S.; Haupt, V.; Schroeder, M. PLIP 2021: expanding the scope of the protein–ligand interaction profiler to DNA and RNA. Nucleic Acids Res. 2021, 49, W530– W534, DOI: 10.1093/nar/gkab294[Crossref], [PubMed], [CAS], Google Scholar58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvV2isLbL&md5=8f56ced733405aaa8f5325e3fa4b862cPLIP 2021 expanding scope of protein-ligand interaction profiler to DNA and RNAAdasme, Melissa F.; Linnemann, Katja L.; Bolz, Sarah Naomi; Kaiser, Florian; Salentin, Sebastian; Haupt, V. Joachim; Schroeder, MichaelNucleic Acids Research (2021), 49 (W1), W530-W534CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)With the growth of protein structure data, the anal. of mol. interactions between ligands and their target mols. is gaining importance. PLIP, the protein-ligand interaction profiler, detects and visualises these interactions and provides data in formats suitable for further processing. PLIP has proven very successful in applications ranging from the characterization of docking expts. to the assessment of novel ligand-protein complexes. Besides ligand-protein interactions, interactions with DNA and RNA play a vital role in many applications, such as drugs targeting DNA or RNA-binding proteins. To date, over 7% of all 3D structures in the Protein Data Bank include DNA or RNA. Therefore, we extended PLIP to encompass these important mols. We demonstrate the power of this extension with examples of a cancer drug binding to a DNA target, and an RNA-protein complex central to a neurol. disease.
- 59Carbery, A.; Skyner, R.; von Delft, F.; Deane, C. M. Fragment libraries designed to be functionally diverse recover protein binding information more efficiently than standard structurally diverse libraries. J. Med. Chem. 2022, 65, 11404– 11413, DOI: 10.1021/acs.jmedchem.2c01004[ACS Full Text
], [CAS], Google Scholar
59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XitFWktLnO&md5=20c210891befe38eb6ec658cba2a01b8Fragment Libraries Designed to Be Functionally Diverse Recover Protein Binding Information More Efficiently Than Standard Structurally Diverse LibrariesCarbery, Anna; Skyner, Rachael; von Delft, Frank; Deane, Charlotte M.Journal of Medicinal Chemistry (2022), 65 (16), 11404-11413CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Current fragment-based drug design relies on the efficient exploration of chem. space by using structurally diverse libraries of small fragments. However, structurally dissimilar compds. can exploit the same interactions and thus be functionally similar. Using three-dimensional structures of many fragments bound to multiple targets, we examd. if a better strategy for selecting fragments for screening libraries exists. We show that structurally diverse fragments can be described as functionally redundant, often making the same interactions. Ranking fragments by the no. of novel interactions they made, we show that functionally diverse selections of fragments substantially increase the amt. of information recovered for unseen targets compared to the amts. recovered by other methods of selection. Using these results, we design small functionally efficient libraries that can give significantly more information about new protein targets than similarly sized structurally diverse libraries. By covering more functional space, we can generate more diverse sets of drug leads. - 60Maaten, L. v. d.; Hinton, G. Visualizing data using t-SNE. J. Mach. Learn. Res. 2008, 9, 2579– 2605Google ScholarThere is no corresponding record for this reference.
- 61Pedregosa, F.; Varoquaux, G.; Gramfort, A.; Michel, V.; Thirion, B.; Grisel, O.; Blondel, M.; Prettenhofer, P.; Weiss, R.; Dubourg, V.; Vanderplas, J.; Passos, A.; Cournapeau, D.; Brucher, M.; Perrot, M.; Duchesnay, d. Scikit-learn: machine learning in Python. J. Mach. Learn. Res. 2011, 12, 2825– 2830Google ScholarThere is no corresponding record for this reference.
- 62PostEra, Mpro activity data. https://covid.postera.ai/covid/activity_data (accessed November 2022).Google ScholarThere is no corresponding record for this reference.
- 63Butina, D. Unsupervised data base clustering based on Daylight’s fingerprint and Tanimoto similarity: a fast and automated way to cluster small and large data sets. JCICS 1999, 39, 747– 750, DOI: 10.1021/ci9803381
Cited By
This article has not yet been cited by other publications.
Abstract
Figure 1
Figure 1. Pipeline for identifying fragment merges. Fragment hits from crystallographic fragment screens are used for finding fragment merges. All possible pairs of compounds are enumerated for merging (removing those with high similarity). Both the Fragment Network and similarity search are used to identify fragment merges. The Fragment Network enumerates all possible substructures of one of the fragments in the merge while the other fragment is regarded as the seed fragment. A series of optional hops are made away from the seed fragment (up to a maximum of two), after which an expansion is made by incorporating a substructure from the other fragment. The similarity search finds merges by calculating the Tversky (Tv) similarity against every compound in the database using the Morgan fingerprint (2048 bits and radius 2). The Tversky calculation uses α and β values of 0.7 and 0.3, respectively. All compounds with a mean similarity ≥0.4 are retained. The merges pass through a series of 2D and 3D filters, including pose generation with Fragmenstein, to result in scored poses.
Figure 2
Figure 2. The Fragment Network identifies pure merges. (a) A fragment-merging opportunity for the main protease (Mpro) data set. Interactions are predicted using the protein–ligand interaction profiler (PLIP). Hydrogen bonds and π-stacking interactions are shown by cyan and magenta dotted lines, respectively. The PanDDA density is provided for the purple fragment in the Supporting Information (owing to the unusual conformation). (b) The linker-like merge (pose generated using Fragmenstein) joins substructures from partially overlapping fragments by a “linker-like” region, maintaining the hydrogen bond with THR-45; a change in orientation of the thiazole ring (with respect to the thiophene ring in the fragment) enables an additional π-stacking interaction with HIS-41. (c) A fragment-linking opportunity for Mpro. (d) The proposed compound maintains a hydrogen bond with PHE-140 and makes an additional bond with SER-144. It is worth noting that the linker group proposed by the Fragment Network is present in thioacetazone, an oral antibiotic. (e) The fragments and merge in a and b in 2D. (f) The fragments and merge in c and d in 2D.
Figure 3
Figure 3. The Fragment Network and similarity searches identify filtered compounds for different fragment pairs. The numbers of filtered compounds for each fragment pair found using the Fragment Network (blue) or similarity search (orange) are shown across targets (a) dipeptidyl peptidase 11 (DPP11), (b) poly(ADP-ribose) polymerase 14, (PARP14), (c) nonstructural protein 13 (nsp13), and (d) main protease (Mpro). Only pairs that resulted in filtered compounds are shown. Pairs are ordered from right to left according to the number of Fragment Network compounds found. The data show that each search technique was able to identify filtered compounds for pairs where the other technique identified none.
Figure 4
Figure 4. Fragment Network and similarity search-derived compound sets populate different regions of chemical space. The chemical space occupied by the filtered compound sets is projected into two dimensions using the T-SNE algorithm across targets (a) dipeptidyl peptidase 11 (DPP11), (b) poly(ADP-ribose) polymerase 14 (PARP14), (c) nonstructural protein 13 (nsp13), and (d) main protease (Mpro). Fragment Network compounds are shown in blue, and similarity search compounds are shown in orange. The two compound sets are shown to occupy distinct areas of chemical space.
Figure 5
Figure 5. The Fragment Network identifies a known binder against Mpro. A Fragment Network search using (a) two fragment hits against the SARS-CoV-2 main protease (Mpro) identifies, (b) a known binder against Mpro (LON-WEI-b2874fec-25; RapidFire mass spectrometry (RF-MS) IC50 value of 59.6 μM) and similar compounds to known binders, (c) JAN-GHE-83b26c96–22 (fluorescence and RF-MS IC50 values of 96.9 μM and 24.5 μM), and (d) TRY-UNI-714a760b-18 (fluorescence and RF-MS IC50 values of 26.2 μM and 13.0 μM). Fragmenstein-predicted merge poses are shown in white, and crystal poses are in cyan. Interactions are predicted using the protein–ligand interaction profiler (PLIP), and key interaction residues are shown. Hydrogen bonds are shown in cyan, and π-stacking interactions are shown in magenta.
Figure 6
Figure 6. The Fragment Network identifies a known binder against EthR. A Fragment Network search using (a) two fragment hits against (which each bind in two different positions) Mycobacterium tuberculosis transcriptional repressor protein EthR identifies (b) a known binder (compound 4; IC50 value of >100 μM). (c) An alternative crystallographic arrangement of the equivalent fragments identifies (d) a similar compound to a known binder, compound 21 (IC50 value of 22 μM). Fragmenstein-predicted merge poses are shown in white, and crystal poses are in cyan. Hydrogen bonds are shown in cyan.
References
ARTICLE SECTIONSThis article references 63 other publications.
- 1Lamoree, B.; Hubbard, R. Current perspectives in fragment-based lead discovery (FBLD). Essays Biochem 2017, 61, 453– 464, DOI: 10.1042/EBC20170028[Crossref], [PubMed], [CAS], Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1M7pt1Gmtw%253D%253D&md5=35bce3c5978329c6737ffd719135d363Current perspectives in fragment-based lead discovery (FBLD)Lamoree Bas; Hubbard Roderick E; Hubbard Roderick EEssays in biochemistry (2017), 61 (5), 453-464 ISSN:.It is over 20 years since the first fragment-based discovery projects were disclosed. The methods are now mature for most 'conventional' targets in drug discovery such as enzymes (kinases and proteases) but there has also been growing success on more challenging targets, such as disruption of protein-protein interactions. The main application is to identify tractable chemical startpoints that non-covalently modulate the activity of a biological molecule. In this essay, we overview current practice in the methods and discuss how they have had an impact in lead discovery - generating a large number of fragment-derived compounds that are in clinical trials and two medicines treating patients. In addition, we discuss some of the more recent applications of the methods in chemical biology - providing chemical tools to investigate biological molecules, mechanisms and systems.
- 2Davis, B. J.; Roughley, S. D. In Platform Technologies in Drug Discovery and Validation; Goodnow, R. A., Ed.; Annual Reports in Medicinal Chemistry series; Academic Press, 2017; Vol. 50; pp 371– 439.
- 3Hann, M. M.; Leach, A. R.; Harper, G. Molecular complexity and its impact on the probability of finding leads for drug discovery. J. Chem. Inf. Model. 2001, 41, 856– 864, DOI: 10.1021/ci000403i[ACS Full Text
], [CAS], Google Scholar
3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXivFynt7w%253D&md5=e89e76c8fb83fc5012369ae30f20e6b7Molecular Complexity and Its Impact on the Probability of Finding Leads for Drug DiscoveryHann, Michael M.; Leach, Andrew R.; Harper, GavinJournal of Chemical Information and Computer Sciences (2001), 41 (3), 856-864CODEN: JCISD8; ISSN:0095-2338. (American Chemical Society)Using a simple model of ligand-receptor interactions, the interactions between ligands and receptors of varying complexities are studied and the probabilities of binding calcd. It is obsd. that as the systems become more complex the chance of observing a useful interaction for a randomly chosen ligand falls dramatically. The implications of this for the design of combinatorial libraries is explored. A large set of drug leads and optimized compds. is profiled using several different properties relevant to mol. recognition. The changes obsd. for these properties during the drug optimization phase support the hypothesis that less complex mols. are more common starting points for the discovery of drugs. An extreme example of the use of simple mols. for directed screening against thrombin is provided. - 4Leach, A. R.; Hann, M. M. Molecular complexity and fragment-based drug discovery: ten years on. Curr. Opin. Chem. Biol. 2011, 15, 489– 496, DOI: 10.1016/j.cbpa.2011.05.008[Crossref], [PubMed], [CAS], Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXpvFCltrg%253D&md5=19c2d65de80a8bab1059468828da6ce2Molecular complexity and fragment-based drug discovery: ten years onLeach, Andrew R.; Hann, Michael M.Current Opinion in Chemical Biology (2011), 15 (4), 489-496CODEN: COCBF4; ISSN:1367-5931. (Elsevier B.V.)We review the concept of mol. complexity in the context of the very simple model of mol. interactions that we introduced over 10 years ago. A summary is presented of efforts to validate this simple model using screening data. The relationship between the complexity model and the problem of sampling chem. space is discussed, together with the relevance of these theor. concepts to fragment-based drug discovery.
- 5Keserű, G. M.; Erlanson, D. A.; Ferenczy, G. G.; Hann, M. M.; Murray, C. W.; Pickett, S. D. Design principles for fragment libraries: maximizing the value of learnings from pharma fragment-based drug discovery (FBDD) programs for use in academia. J. Med. Chem. 2016, 59, 8189– 8206, DOI: 10.1021/acs.jmedchem.6b00197[ACS Full Text
], [CAS], Google Scholar
5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XmvFSit7s%253D&md5=888fe2cf003c5564c2ec391cf54ea2acDesign Principles for Fragment Libraries: Maximizing the Value of Learnings from Pharma Fragment-Based Drug Discovery (FBDD) Programs for Use in AcademiaKeseru, Gyorgy M.; Erlanson, Daniel A.; Ferenczy, Gyorgy G.; Hann, Michael M.; Murray, Christopher W.; Pickett, Stephen D.Journal of Medicinal Chemistry (2016), 59 (18), 8189-8206CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Fragment-based drug discovery (FBDD) is well suited for discovering both drug leads and chem. probes of protein function; it can cover broad swaths of chem. space and allows the use of creative chem. FBDD is widely implemented for lead discovery in industry but is sometimes used less systematically in academia. Design principles and implementation approaches for fragment libraries are continually evolving, and the lack of up-to-date guidance may prevent more effective application of FBDD in academia. This Perspective explores many of the theor., practical, and strategic considerations that occur within FBDD programs, including the optimal size, complexity, physicochem. profile, and shape profile of fragments in FBDD libraries, as well as compd. storage, evaluation, and screening technologies. This compilation of industry experience in FBDD will hopefully be useful for those pursuing FBDD in academia. - 6Hall, R. J.; Mortenson, P. N.; Murray, C. W. Efficient exploration of chemical space by fragment-based screening. Prog. Biophys. Mol. Biol. 2014, 116, 82– 91, DOI: 10.1016/j.pbiomolbio.2014.09.007[Crossref], [PubMed], [CAS], Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhs1ymu7zO&md5=a63e451443207393b781d3d68b10bcd1Efficient exploration of chemical space by fragment-based screeningHall, Richard J.; Mortenson, Paul N.; Murray, Christopher W.Progress in Biophysics & Molecular Biology (2014), 116 (2-3), 82-91CODEN: PBIMAC; ISSN:0079-6107. (Elsevier Ltd.)Screening methods seek to sample a vast chem. space in order to identify starting points for further chem. optimization. Fragment based drug discovery exploits the superior sampling of chem. space that can be achieved when the mol. wt. is restricted. Here we show that com. available fragment space is still relatively poorly sampled and argue for highly sensitive screening methods to allow the detection of smaller fragments. We analyze the properties of our fragment library vs. the properties of X-ray hits derived from the library. We particularly consider properties related to the degree of planarity of the fragments.
- 7Murray, C. W.; Rees, D. C. The rise of fragment-based drug discovery. Nat. Chem. 2009, 1, 187– 192, DOI: 10.1038/nchem.217[Crossref], [PubMed], [CAS], Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXmtF2rs78%253D&md5=0bd1a40fd927cce9200f0fa296a7462fThe rise of fragment-based drug discoveryMurray, Christopher W.; Rees, David C.Nature Chemistry (2009), 1 (3), 187-192CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)A review. The search for new drugs is plagued by high attrition rates at all stages in research and development. Chemists have an opportunity to tackle this problem because attrition can be traced back, in part, to the quality of the chem. leads. Fragment-based drug discovery (FBDD) is a new approach, increasingly used in the pharmaceutical industry, for reducing attrition and providing leads for previously intractable biol. targets. FBDD identifies low-mol.-wt. ligands (∼150 Da) that bind to biol. important macromols. The three-dimensional exptl. binding mode of these fragments is detd. using X-ray crystallog. or NMR spectroscopy, and is used to facilitate their optimization into potent mols. with drug-like properties. Compared with high-throughput-screening, the fragment approach requires fewer compds. to be screened, and, despite the lower initial potency of the screening hits, offers more efficient and fruitful optimization campaigns. Here, we review the rise of FBDD, including its application to discovering clin. candidates against targets for which other chem. approaches have struggled.
- 8Roughley, S. D.; Hubbard, R. E. How well can fragments explore accessed chemical space? A case study from heat shock protein 90. J. Med. Chem. 2011, 54, 3989– 4005, DOI: 10.1021/jm200350g[ACS Full Text
], [CAS], Google Scholar
8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXmsFymurs%253D&md5=90c2c8028cb460c0c6fdc956e7daae28How Well Can Fragments Explore Accessed Chemical Space? A Case Study from Heat Shock Protein 90Roughley, Stephen D.; Hubbard, Roderick E.Journal of Medicinal Chemistry (2011), 54 (12), 3989-4005CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A review. This article summerizes the published literature on the generation of inhibitors and design of drug against the mol. chaperone, heat shock protein 90 (Hsp90), as potential therapeutics to treat cancer. - 9Collins, P. M.; Douangamath, A.; Talon, R.; Dias, A.; Brandao-Neto, J.; Krojer, T.; von Delft, F. Achieving a good crystal system for crystallographic X-ray fragment screening. Methods Enzymol 2018, 610, 251– 264, DOI: 10.1016/bs.mie.2018.09.027[Crossref], [PubMed], [CAS], Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsFKrsrvJ&md5=c122d61fb2371eff58429ce90e777c8eAchieving a good crystal system for crystallographic X-ray fragment screeningCollins, Patrick M.; Douangamath, Alice; Talon, Romain; Dias, Alexandre; Brandao-Neto, Jose; Krojer, Tobias; von Delft, FrankMethods in Enzymology (2018), 610 (Modern Approaches in Drug Discovery), 251-264CODEN: MENZAU; ISSN:0076-6879. (Elsevier Inc.)A review. The XChem facility at Diamond Light Source offers fragment screening by X-ray crystallog. as a general access user program. The main advantage of X-ray crystallog. as a primary fragment screen is that it yields directly the location and pose of the fragment hits, whether within pockets of interest or merely on surface sites: this is the key information for structure-based design and for enabling synthesis of follow up mols. Extensive streamlining of the screening expt. at XChem has engendered a very active user program that is generating large amts. of data: in 2017,36 academic and industry groups generated 35,000 datasets of uniquely soaked crystals. It has also generated a large no. of learnings concerning the main remaining bottleneck namely, obtaining a suitable crystal system that will support a successful fragment screen. Here we discuss the practicalities of generating screen-ready crystals that have useful electron d. maps, and how to ensure they will be successfully reproduced and usable at a facility outside the home lab.
- 10The COVID Moonshot Consortium. COVID Moonshot: open science discovery of SARS-CoV-2 main protease inhibitors by combining crowdsourcing, high-throughput experiments, computational simulations, and machine learning. bioRxiv Preprint , 2020. DOI: 10.1101/2020.10.29.339317
- 11Douangamath, A.; Powell, A.; Fearon, D.; Collins, P. M.; Talon, R.; Krojer, T.; Skyner, R.; Brandao-Neto, J.; Dunnett, L.; Dias, A.; Aimon, A.; Pearce, N. M.; Wild, C.; Gorrie-Stone, T.; von Delft, F. Achieving efficient fragment screening at XChem facility at Diamond Light Source. JoVE 2021, 171, e62414 DOI: 10.3791/62414
- 12Metz, A.; Wollenhaupt, J.; Glöckner, S.; Messini, N.; Huber, S.; Barthel, T.; Merabet, A.; Gerber, H.-D.; Heine, A.; Klebe, G.; Weiss, M. S. Frag4Lead: growing crystallographic fragment hits by catalog using fragment-guided template docking. Acta Crystallogr., Sect. D: Struct. Biol. 2021, 77, 1168– 1182, DOI: 10.1107/S2059798321008196[Crossref], [CAS], Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvFegt7%252FF&md5=391b172987e46364a4b877d2a34508fcFrag4Lead: growing crystallographic fragment hits by catalog using fragment-guided template dockingMetz, Alexander; Wollenhaupt, Jan; Gloeckner, Steffen; Messini, Niki; Huber, Simon; Barthel, Tatjana; Merabet, Ahmed; Gerber, Hans-Dieter; Heine, Andreas; Klebe, Gerhard; Weiss, Manfred S.Acta Crystallographica, Section D: Structural Biology (2021), 77 (9), 1168-1182CODEN: ACSDAD; ISSN:2059-7983. (International Union of Crystallography)In recent years, crystallog. fragment screening has matured into an almost routine expt. at several modern synchrotron sites. The hits of the screening expt., i.e. small mols. or fragments binding to the target protein, are revealed along with their 3D structural information. Therefore, they can serve as useful starting points for further structure-based hit-to-lead development. However, the progression of fragment hits to tool compds. or even leads is often hampered by a lack of chem. feasibility. As an attractive alternative, compd. analogs that embed the fragment hit structurally may be obtained from com. catalogs. Here, a workflow is reported based on filtering and assessing such potential follow-up compds. by template docking. This means that the crystallog. binding pose was integrated into the docking calcns. as a central starting parameter. Subsequently, the candidates are scored on their interactions within the binding pocket. In an initial proof-of-concept study using five starting fragments known to bind to the aspartic protease endothiapepsin, 28 follow-up compds. were selected using the designed workflow and their binding was assessed by crystallog. Ten of these compds. bound to the active site and five of them showed significantly increased affinity in isothermal titrn. calorimetry of up to single-digit micromolar affinity. Taken together, this strategy is capable of efficiently evolving the initial fragment hits without major synthesis efforts and with full control by X-ray crystallog.
- 13Müller, J.; Klein, R.; Tarkhanova, O.; Gryniukova, A.; Borysko, P.; Merkl, S.; Ruf, M.; Neumann, A.; Gastreich, M.; Moroz, Y. S.; Klebe, G.; Glinca, S. Magnet for the needle in haystack: “Crystal structure first” Fragment hits unlock active chemical matter using targeted exploration of vast chemical spaces. J. Med. Chem. 2022, 65, 15663– 15678, DOI: 10.1021/acs.jmedchem.2c00813[ACS Full Text
], [CAS], Google Scholar
13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XitlWrtrfI&md5=7d2ae978c7757c8f1b3083fa84c60f9eMagnet for the Needle in Haystack: "Crystal Structure First" Fragment Hits Unlock Active Chemical Matter Using Targeted Exploration of Vast Chemical SpacesMueller, Janis; Klein, Raphael; Tarkhanova, Olga; Gryniukova, Anastasiia; Borysko, Petro; Merkl, Stefan; Ruf, Moritz; Neumann, Alexander; Gastreich, Marcus; Moroz, Yurii S.; Klebe, Gerhard; Glinca, SergheiJournal of Medicinal Chemistry (2022), 65 (23), 15663-15678CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Fragment-based drug discovery (FBDD) has successfully led to approved therapeutics for challenging and "undruggable" targets. In the context of FBDD, we introduce a novel, multidisciplinary method to identify active mols. from purchasable chem. space. Starting from four small-mol. fragment complexes of protein kinase A (PKA), a template-based docking screen using Enamine's multibillion REAL Space was performed. A total of 93 mols. out of 106 selected compds. were successfully synthesized. Forty compds. were active in at least one validation assay with the most active follow-up having a 13,500-fold gain in affinity. Crystal structures for six of the most promising binders were rapidly obtained, verifying the binding mode. The overall success rate for this novel fragment-to-hit approach was 40%, accomplished in only 9 wk. The results challenge the established fragment prescreening paradigm since the std. industrial filters for fragment hit identification in a thermal shift assay would have missed the initial fragments. - 14Piticchio, S. G.; Martínez-Cartró, M.; Scaffidi, S.; Rachman, M.; Rodriguez-Arevalo, S.; Sanchez-Arfelis, A.; Escolano, C.; Picaud, S.; Krojer, T.; Filippakopoulos, P.; von Delft, F.; Galdeano, C.; Barril, X. Discovery of novel BRD4 ligand scaffolds by automated navigation of the fragment chemical space. J. Med. Chem. 2021, 64, 17887– 17900, DOI: 10.1021/acs.jmedchem.1c01108[ACS Full Text
], [CAS], Google Scholar
14https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXislWrtbjE&md5=98aebd322c14abb1a7f56157e46013a6Discovery of Novel BRD4 Ligand Scaffolds by Automated Navigation of the Fragment Chemical SpacePiticchio, Serena G.; Martinez-Cartro, Miriam; Scaffidi, Salvatore; Rachman, Moira; Rodriguez-Arevalo, Sergio; Sanchez-Arfelis, Ainoa; Escolano, Carmen; Picaud, Sarah; Krojer, Tobias; Filippakopoulos, Panagis; von Delft, Frank; Galdeano, Carles; Barril, XavierJournal of Medicinal Chemistry (2021), 64 (24), 17887-17900CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Fragment-based drug discovery (FBDD) is a very effective hit identification method. However, the evolution of fragment hits into suitable leads remains challenging and largely artisanal. Fragment evolution is often scaffold-centric, meaning that its outcome depends crucially on the chem. structure of the starting fragment. Considering that fragment screening libraries cover only a small proportion of the corresponding chem. space, hits should be seen as probes highlighting privileged areas of the chem. space rather than actual starting points. We have developed an automated computational pipeline to mine the chem. space around any specific fragment hit, rapidly finding analogs that share a common interaction motif but are structurally novel and diverse. On a prospective application on the bromodomain-contg. protein 4 (BRD4), starting from a known fragment, the platform yields active mols. with nonobvious scaffold changes. The procedure is fast and inexpensive and has the potential to uncover many hidden opportunities in FBDD. - 15Miyake, Y.; Itoh, Y.; Hatanaka, A.; Suzuma, Y.; Suzuki, M.; Kodama, H.; Arai, Y.; Suzuki, T. Identification of novel lysine demethylase 5-selective inhibitors by inhibitor-based fragment merging strategy. Bioorg. Med. Chem. 2019, 27, 1119– 1129, DOI: 10.1016/j.bmc.2019.02.006[Crossref], [PubMed], [CAS], Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXivVWltbg%253D&md5=1d9473e11de97ee34b82b82b8d6ffb38Identification of novel lysine demethylase 5-selective inhibitors by inhibitor-based fragment merging strategyMiyake, Yuka; Itoh, Yukihiro; Hatanaka, Atsushi; Suzuma, Yoshinori; Suzuki, Miki; Kodama, Hidehiko; Arai, Yoshinobu; Suzuki, TakayoshiBioorganic & Medicinal Chemistry (2019), 27 (6), 1119-1129CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)Histone lysine demethylases (KDMs) have drawn much attention as targets of therapeutic agents. KDM5 proteins, which are Fe(II)/α-ketoglutarate-dependent demethylases, are assocd. with oncogenesis and drug resistance in cancer cells, and KDM5-selective inhibitors are expected to be anticancer drugs. However, few cell-active KDM5 inhibitors have been reported and there is an obvious need to discover more. In this study, we pursued the identification of highly potent and cell-active KDM5-selective inhibitors. Based on the reported KDM5 inhibitors, we designed several compds. by strategically merging two fragments for competitive inhibition with α-ketoglutarate and for KDM5-selective inhibition. Among them, compds. 10 and 13, which have a 3-cyano pyrazolo[1,5-a]pyrimidin-7-one scaffold, exhibited strong KDM5-inhibitory activity and significant KDM5 selectivity. In cellular assays using human lung cancer cell line A549, 10 and 13 increased the levels of trimethylated lysine 4 on histone H3, which is a specific substrate of KDM5s, and induced growth inhibition of A549 cells. These results should provide a basis for the development of cell-active KDM5 inhibitors to highlight the validity of our inhibitor-based fragment merging strategy.
- 16Ren, J.; Li, J.; Wang, Y.; Chen, W.; Shen, A.; Liu, H.; Chen, D.; Cao, D.; Li, Y.; Zhang, N.; Xu, Y.; Geng, M.; He, J.; Xiong, B.; Shen, J. Identification of a new series of potent diphenol HSP90 inhibitors by fragment merging and structure-based optimization. Bioorg. Med. Chem. Lett. 2014, 24, 2525– 2529, DOI: 10.1016/j.bmcl.2014.03.100[Crossref], [PubMed], [CAS], Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXmslahsrY%253D&md5=057e15a1982cf723de8c7436accf4fffIdentification of a new series of potent diphenol HSP90 inhibitors by fragment merging and structure-based optimizationRen, Jing; Li, Jian; Wang, Yueqin; Chen, Wuyan; Shen, Aijun; Liu, Hongchun; Chen, Danqi; Cao, Danyan; Li, Yanlian; Zhang, Naixia; Xu, Yechun; Geng, Meiyu; He, Jianhua; Xiong, Bing; Shen, JingkangBioorganic & Medicinal Chemistry Letters (2014), 24 (11), 2525-2529CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)Heat shock protein 90 (HSP90) is a mol. chaperone to fold and maintain the proper conformation of many signaling proteins, esp. some oncogenic proteins and mutated unstable proteins. Inhibition of HSP90 was recognized as an effective approach to simultaneously suppress several aberrant signaling pathways, and therefore it was considered as a novel target for cancer therapy. Here, by integrating several techniques including the fragment-based drug discovery method, fragment merging, computer aided inhibitor optimization, and structure-based drug design, the authors were able to identify a series of HSP90 inhibitors, e.g. I [R1 = H. HO2CCH2, NH2CH2CH2, MeCONHCH2CH2; r2 = H, Br, O2N, etc.; R3 = H, Br, Me, MeO]. Several compds. can inhibit HSP90 with IC50 about 20-40 nM, which is at least 200-fold more potent than initial fragments in the protein binding assay. These new HSP90 inhibitors not only explore interactions with an under-studied subpocket, also offer new chemotypes for the development of novel HSP90 inhibitors as anticancer drugs.
- 17Credille, C. V.; Chen, Y.; Cohen, S. M. Fragment-based identification of influenza endonuclease inhibitors. J. Med. Chem. 2016, 59, 6444– 6454, DOI: 10.1021/acs.jmedchem.6b00628[ACS Full Text
], [CAS], Google Scholar
17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XpsF2mtL8%253D&md5=a872083d373a846467c7f2eb5439bed4Fragment-Based Identification of Influenza Endonuclease InhibitorsCredille, Cy V.; Chen, Yao; Cohen, Seth M.Journal of Medicinal Chemistry (2016), 59 (13), 6444-6454CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The influenza virus is responsible for millions of cases of severe illness annually. Yearly variance in the effectiveness of vaccination, coupled with emerging drug resistance, necessitates the development of new drugs to treat influenza infections. One attractive target is the RNA-dependent RNA polymerase PA subunit. Herein we report the development of inhibitors of influenza PA endonuclease derived from lead compds. identified from a metal-binding pharmacophore (MBP) library screen. Pyromeconic acid and derivs. thereof were found to be potent inhibitors of endonuclease. Guided by modeling and previously reported structural data, several sublibraries of mols. were elaborated from the MBP hits. Structure-activity relationships were established, and more potent mols. were designed and synthesized using fragment growth and fragment merging strategies. This approach ultimately resulted in the development of a lead compd. with an IC50 value of 14 nM, which displayed an EC50 value of 2.1 μM against H1N1 influenza virus in MDCK cells. - 18Edink, E.; Rucktooa, P.; Retra, K.; Akdemir, A.; Nahar, T.; Zuiderveld, O.; van Elk, R.; Janssen, E.; van Nierop, P.; van Muijlwijk-Koezen, J.; Smit, A. B.; Sixma, T. K.; Leurs, R.; de Esch, I. J. P. Fragment growing induces conformational changes in acetylcholine-binding protein: a structural and thermodynamic analysis. J. Am. Chem. Soc. 2011, 133, 5363– 5371, DOI: 10.1021/ja110571r[ACS Full Text
], [CAS], Google Scholar
18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhvFeisrc%253D&md5=413798f2128e309545de77f8e995b565Fragment Growing Induces Conformational Changes in Acetylcholine-Binding Protein: A Structural and Thermodynamic AnalysisEdink, Ewald; Rucktooa, Prakash; Retra, Kim; Akdemir, Atilla; Nahar, Tariq; Zuiderveld, Obbe; van Elk, Rene; Janssen, Elwin; van Nierop, Pim; van Muijlwijk-Koezen, Jacqueline; Smit, August B.; Sixma, Titia K.; Leurs, Rob; de Esch, Iwan J. P.Journal of the American Chemical Society (2011), 133 (14), 5363-5371CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Optimization of fragment hits toward high-affinity lead compds. is a crucial aspect of fragment-based drug discovery (FBDD). In the current study, we have successfully optimized a fragment by growing into a ligand-inducible subpocket of the binding site of acetylcholine-binding protein (AChBP). This protein is a sol. homolog of the ligand binding domain (LBD) of Cys-loop receptors. The fragment optimization was monitored with X-ray structures of ligand complexes and systematic thermodn. analyses using surface plasmon resonance (SPR) biosensor anal. and isothermal titrn. calorimetry (ITC). Using site-directed mutagenesis and AChBP from different species, we find that specific changes in thermodn. binding profiles, are indicative of interactions with the ligand-inducible subpocket of AChBP. This study illustrates that thermodn. anal. provides valuable information on ligand binding modes and is complementary to affinity data when guiding rational structure- and fragment-based discovery approaches. - 19Hughes, S. J.; Millan, D. S.; Kilty, I. C.; Lewthwaite, R. A.; Mathias, J. P.; O’Reilly, M. A.; Pannifer, A.; Phelan, A.; Stühmeier, F.; Baldock, D. A.; Brown, D. G. Fragment based discovery of a novel and selective PI3 kinase inhibitor. Bioorg. Med. Chem. Lett. 2011, 21, 6586– 6590, DOI: 10.1016/j.bmcl.2011.07.117[Crossref], [PubMed], [CAS], Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXht12jtrjO&md5=be9322ee0268040ed1da843025df4cdeFragment based discovery of a novel and selective PI3 kinase inhibitorHughes, Samantha J.; Millan, David S.; Kilty, Iain C.; Lewthwaite, Russell A.; Mathias, John P.; O'Reilly, Mark A.; Pannifer, Andrew; Phelan, Anne; Stuehmeier, Frank; Baldock, Darren A.; Brown, David G.Bioorganic & Medicinal Chemistry Letters (2011), 21 (21), 6586-6590CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)We report the use of fragment screening and fragment based drug design to develop a PI3γ kinase fragment hit into a lead. Initial fragment hits were discovered by high concn. biochem. screening, followed by a round of virtual screening to identify addnl. ligand efficient fragments. These were developed into potent and ligand efficient lead compds. using structure guided fragment growing and merging strategies. This led to a potent, selective, and cell permeable PI3γ kinase inhibitor, 12 (I), with good metabolic stability that was useful as a preclin. tool compd.
- 20Brough, P. A.; Barril, X.; Borgognoni, J.; Chene, P.; Davies, N. G. M.; Davis, B.; Drysdale, M. J.; Dymock, B.; Eccles, S. A.; Garcia-Echeverria, C.; Fromont, C.; Hayes, A.; Hubbard, R. E.; Jordan, A. M.; Jensen, M. R.; Massey, A.; Merrett, A.; Padfield, A.; Parsons, R.; Radimerski, T.; Raynaud, F. I.; Robertson, A.; Roughley, S. D.; Schoepfer, J.; Simmonite, H.; Sharp, S. Y.; Surgenor, A.; Valenti, M.; Walls, S.; Webb, P.; Wood, M.; Workman, P.; Wright, L. Combining hit identification strategies: fragment-based and in silico approaches to orally active 2-aminothieno[2,3-d]pyrimidine inhibitors of the Hsp90 molecular chaperone. J. Med. Chem. 2009, 52, 4794– 4809, DOI: 10.1021/jm900357y[ACS Full Text
], [CAS], Google Scholar
20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXos1ehsbs%253D&md5=4d1988060b0f87dc776e1972ea14808eCombining Hit Identification Strategies: Fragment-Based and in Silico Approaches to Orally Active 2-Aminothieno[2,3-d]pyrimidine Inhibitors of the Hsp90 Molecular ChaperoneBrough, Paul A.; Barril, Xavier; Borgognoni, Jenifer; Chene, Patrick; Davies, Nicholas G. M.; Davis, Ben; Drysdale, Martin J.; Dymock, Brian; Eccles, Suzanne A.; Garcia-Echeverria, Carlos; Fromont, Christophe; Hayes, Angela; Hubbard, Roderick E.; Jordan, Allan M.; Jensen, Michael Rugaard; Massey, Andrew; Merrett, Angela; Padfield, Antony; Parsons, Rachel; Radimerski, Thomas; Raynaud, Florence I.; Robertson, Alan; Roughley, Stephen D.; Schoepfer, Joseph; Simmonite, Heather; Sharp, Swee Y.; Surgenor, Allan; Valenti, Melanie; Walls, Steven; Webb, Paul; Wood, Mike; Workman, Paul; Wright, LisaJournal of Medicinal Chemistry (2009), 52 (15), 4794-4809CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Inhibitors of the Hsp90 mol. chaperone are showing considerable promise as potential mol. therapeutic agents for the treatment of cancer. Here we describe novel 2-aminothieno[2,3-d]pyrimidine ATP competitive Hsp90 inhibitors, which were designed by combining structural elements of distinct low affinity hits generated from fragment-based and in silico screening exercises in concert with structural information from X-ray protein crystallog. Examples from this series have high affinity (IC50 = 50-100 nM) for Hsp90 as measured in a fluorescence polarization (FP) competitive binding assay and are active in human cancer cell lines where they inhibit cell proliferation and exhibit a characteristic profile of depletion of oncogenic proteins and concomitant elevation of Hsp72. Several compds. caused tumor growth regression at well tolerated doses when administered orally in a human BT474 human breast cancer xenograft model. - 21Nikiforov, P. O.; Surade, S.; Blaszczyk, M.; Delorme, V.; Brodin, P.; Baulard, A. R.; Blundell, T. L.; Abell, C. A fragment merging approach towards the development of small molecule inhibitors of Mycobacterium tuberculosis EthR for use as ethionamide boosters. Org. Biomol. Chem. 2016, 14, 2318– 2326, DOI: 10.1039/C5OB02630J[Crossref], [PubMed], [CAS], Google Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xoslajtg%253D%253D&md5=bbe5d5a96e650841b351f2fb9d221f22A fragment merging approach towards the development of small molecule inhibitors of Mycobacterium tuberculosis EthR for use as ethionamide boostersNikiforov, Petar O.; Surade, Sachin; Blaszczyk, Michal; Delorme, Vincent; Brodin, Priscille; Baulard, Alain R.; Blundell, Tom L.; Abell, ChrisOrganic & Biomolecular Chemistry (2016), 14 (7), 2318-2326CODEN: OBCRAK; ISSN:1477-0520. (Royal Society of Chemistry)With the ever-increasing instances of resistance to frontline TB drugs there is the need to develop novel strategies to fight the worldwide TB epidemic. Boosting the effect of the existing second-line antibiotic ethionamide by inhibiting the mycobacterial transcriptional repressor protein EthR is an attractive therapeutic strategy. Herein we report the use of a fragment based drug discovery approach for the structure-guided systematic merging of two fragment mols., each binding twice to the hydrophobic cavity of EthR from M. tuberculosis. These together fill the entire binding pocket of EthR. We elaborated these fragment hits and developed small mol. inhibitors which have a 100-fold improvement of potency in vitro over the initial fragments.
- 22Schade, M.; Merla, B.; Lesch, B.; Wagener, M.; Timmermanns, S.; Pletinckx, K.; Hertrampf, T. Highly selective sub-nanomolar cathepsin S inhibitors by merging fragment binders with nitrile inhibitors. J. Med. Chem. 2020, 63, 11801– 11808, DOI: 10.1021/acs.jmedchem.0c00949[ACS Full Text
], [CAS], Google Scholar
22https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhslGqsrnI&md5=f7698389131c792e3bebb40123f6ec66Highly Selective Sub-Nanomolar Cathepsin S Inhibitors by Merging Fragment Binders with Nitrile InhibitorsSchade, Markus; Merla, Beatrix; Lesch, Bernhard; Wagener, Markus; Timmermanns, Simone; Pletinckx, Katrien; Hertrampf, TorstenJournal of Medicinal Chemistry (2020), 63 (20), 11801-11808CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Pharmacol. inhibition of cathepsin S (CatS) allows for a specific modulation of the adaptive immune system and many major diseases. Here, we used NMR fragment screening and crystal structure-aided merging to synthesize novel, highly selective CatS inhibitors with picomolar enzymic Ki values and nanomolar functional activity in human Raji cells. Noncovalent fragment hits revealed binding hotspots, while the covalent inhibitor structure-activity relationship enabled efficient potency optimization. - 23de Souza Neto, L. R.; Moreira-Filho, J. T.; Neves, B. J.; Maidana, R. L. B. R.; Guimarães, A. C. R.; Furnham, N.; Andrade, C. H.; Silva, F. P. In silico strategies to support fragment-to-lead optimization in drug discovery. Front. Chem. 2020, 8, 93, DOI: 10.3389/fchem.2020.00093[Crossref], [PubMed], [CAS], Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB387psFOgtg%253D%253D&md5=3fac2ef52a5f51e4e34826f08d5e28d5In silico Strategies to Support Fragment-to-Lead Optimization in Drug Discoveryde Souza Neto Lauro Ribeiro; Maidana Rocio Lucia Beatriz Riveros; Silva Floriano Paes Jr; Moreira-Filho Jose Teofilo; Neves Bruno Junior; Andrade Carolina Horta; Neves Bruno Junior; Maidana Rocio Lucia Beatriz Riveros; Guimaraes Ana Carolina Ramos; Furnham NicholasFrontiers in chemistry (2020), 8 (), 93 ISSN:2296-2646.Fragment-based drug (or lead) discovery (FBDD or FBLD) has developed in the last two decades to become a successful key technology in the pharmaceutical industry for early stage drug discovery and development. The FBDD strategy consists of screening low molecular weight compounds against macromolecular targets (usually proteins) of clinical relevance. These small molecular fragments can bind at one or more sites on the target and act as starting points for the development of lead compounds. In developing the fragments attractive features that can translate into compounds with favorable physical, pharmacokinetics and toxicity (ADMET-absorption, distribution, metabolism, excretion, and toxicity) properties can be integrated. Structure-enabled fragment screening campaigns use a combination of screening by a range of biophysical techniques, such as differential scanning fluorimetry, surface plasmon resonance, and thermophoresis, followed by structural characterization of fragment binding using NMR or X-ray crystallography. Structural characterization is also used in subsequent analysis for growing fragments of selected screening hits. The latest iteration of the FBDD workflow employs a high-throughput methodology of massively parallel screening by X-ray crystallography of individually soaked fragments. In this review we will outline the FBDD strategies and explore a variety of in silico approaches to support the follow-up fragment-to-lead optimization of either: growing, linking, and merging. These fragment expansion strategies include hot spot analysis, druggability prediction, SAR (structure-activity relationships) by catalog methods, application of machine learning/deep learning models for virtual screening and several de novo design methods for proposing synthesizable new compounds. Finally, we will highlight recent case studies in fragment-based drug discovery where in silico methods have successfully contributed to the development of lead compounds.
- 24Pierce, A. C.; Rao, G.; Bemis, G. W. BREED:generating novel inhibitors through hybridization of known ligands. Application to CDK2, P38, and HIV protease. J. Med. Chem. 2004, 47, 2768– 2775, DOI: 10.1021/jm030543u[ACS Full Text
], [CAS], Google Scholar
24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXjt1ajurk%253D&md5=e0bec9082fab05b694742498b3f41549BREED: Generating Novel Inhibitors through Hybridization of Known Ligands. Application to CDK2, P38, and HIV ProteasePierce, Albert C.; Rao, Govinda; Bemis, Guy W.Journal of Medicinal Chemistry (2004), 47 (11), 2768-2775CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)In this work we describe BREED, a method for the generation of novel inhibitors from structures of known ligands bound to a common target. The method is essentially an automation of the common medicinal chem. practice of joining fragments of two known ligands to generate a new inhibitor. The ligand-bound target structures are overlaid, all overlapping bonds in all pairs of ligands are found, and the fragments on each side of each matching bond are swapped to generate the new mols. Since the method is automated, it can be applied recursively to generate all possible combinations of known ligands. In an application of this method to HIV protease inhibitors and protein kinase inhibitors, hundreds of new mol. structures were generated. These included known inhibitor scaffolds not included in the initial set, entirely novel scaffolds, and novel substituents on known scaffolds. The method is fast, and since all of the ligand functional groups are known to bind the target in the precise position and orientation present in the novel ligand, the success rate of this method should be superior to more traditional de novo design techniques. In an era of increasingly high-throughput structural biol., such methods for high-throughput utilization of structural information will become increasingly valuable. - 25Lindert, S.; Durrant, J. D.; McCammon, J. A. LigMerge: a fast algorithm to generate models of novel potential ligands from sets of known binders. Chem. Biol. Drug Des. 2012, 80, 358– 365, DOI: 10.1111/j.1747-0285.2012.01414.x[Crossref], [PubMed], [CAS], Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XhtF2itbbF&md5=07cc659c48eccc967fc3b135182e9012LigMerge: a fast algorithm to generate models of novel potential ligands from sets of known bindersLindert, Steffen; Durrant, Jacob D.; McCammon, J. AndrewChemical Biology & Drug Design (2012), 80 (3), 358-365CODEN: CBDDAL; ISSN:1747-0277. (Wiley-Blackwell)One common practice in drug discovery is to optimize known or suspected ligands to improve binding affinity. In performing these optimizations, it is useful to look at as many known inhibitors as possible for guidance. Medicinal chemists often seek to improve potency by altering certain chem. moieties of known/endogenous ligands while retaining those crit. for binding. To our knowledge, no automated, ligand-based algorithm exists for systematically "swapping" the chem. moieties of known ligands to generate novel ligands with potentially improved potency. To address this need, we have created a novel algorithm called "LigMerge". LigMerge identifies the max. (largest) common substructure of two three-dimensional ligand models, superimposes these two substructures, and then systematically mixes and matches the distinct fragments attached to the common substructure at each common atom, thereby generating multiple compd. models related to the known inhibitors that can be evaluated using computer docking prior to synthesis and exptl. testing. To demonstrate the utility of LigMerge, we identify compds. predicted to inhibit peroxisome proliferator-activated receptor gamma, HIV reverse transcriptase, and dihydrofolate reductase with affinities higher than those of known ligands. We hope that LigMerge will be a helpful tool for the drug design community.
- 26Wang, H.; Pan, X.; Zhang, Y.; Wang, X.; Xiao, X.; Ji, C. MolHyb: a web server for structure-based drug design by molecular hybridization. J. Chem. Inf. Model. 2022, 62, 2916– 2922, DOI: 10.1021/acs.jcim.2c00443[ACS Full Text
], [CAS], Google Scholar
26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhsVyjsr3I&md5=f7682a620c9ca25bfbc1142b204e20faMolHyb: A Web Server for Structure-Based Drug Design by Molecular HybridizationWang, Hao; Pan, Xiaolin; Zhang, Yueqing; Wang, Xingyu; Xiao, Xudong; Ji, ChanggeJournal of Chemical Information and Modeling (2022), 62 (12), 2916-2922CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Mol. hybridization is a widely used ligand design method in drug discovery. In this study, we present MolHyb, a web server for structure-based ligand design by mol. hybridization. The input of MolHyb is a protein file and a seed compd. file. MolHyb tries to generate novel ligands through hybridizing the seed compd. with helper compds. that bind to the same protein target or similar proteins. To facilitate the job of getting helper compds., we compiled a modeled protein-ligand structure database as an extension to crystal structures in the PDB database by placing the bioactive compds. in ChEMBL into their corresponding 3D protein binding pocket properly. MolHyb works by searching for helper compds. from the protein-ligand structure database and migrating chem. moieties from helper compds. to the seed compd. efficiently. Hybridization is performed at both cyclic and acyclic bonds. The users can also input their own helper compds. to MolHyb. We hope that MolHyb will be a useful tool for rational drug design. MolHyb is freely available at http://molhyb.xundrug.cn/. - 27Li, Y.; Zhao, Y.; Liu, Z.; Wang, R. Automatic Tailoring and Transplanting: a practical method that makes virtual screening more useful. J. Chem. Inf. Model. 2011, 51, 1474– 1491, DOI: 10.1021/ci200036m[ACS Full Text
], [CAS], Google Scholar
27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXlvFKqsb0%253D&md5=3327b42fecda9f2af07a6864d05afaa4Automatic Tailoring and Transplanting: A Practical Method that Makes Virtual Screening More UsefulLi, Yan; Zhao, Yuan; Liu, Zhihai; Wang, RenxiaoJournal of Chemical Information and Modeling (2011), 51 (6), 1474-1491CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Docking-based virtual screening of large compd. libraries has been widely applied to lead discovery in structure-based drug design. However, subsequent lead optimizations often rely on other types of computational methods, such as de novo design methods. We have developed an automatic method, namely automatic tailoring and transplanting (AutoT&T), which can effectively utilize the outcomes of virtual screening in lead optimization. This method detects suitable fragments on virtual screening hits and then transplants them onto a lead compd. to generate new ligand mols. Binding affinities, synthetic feasibilities, and drug-likeness properties are considered in the selection of final designs. In this study, our AutoT&T program was tested on three different target proteins, including p38 MAP kinase, PPAR-α, and Mcl-1. In the first two cases, AutoT&T was able to produce mols. identical or similar to known inhibitors with better potency than the given lead compd. In the third case, we demonstrated how to apply AutoT&T to design novel ligand mols. from scratch. Compared to the solns. generated by other two de novo design methods, i.e., LUDI and EA-Inventor, the solns. generated by AutoT&T were structurally more diverse and more promising in terms of binding scores in all three cases. AutoT&T also completed the assigned jobs more efficiently than LUDI and EA-Inventor by several folds. Our AutoT&T method has certain tech. advantages over de novo design methods. Importantly, it expands the application of virtual screening from lead discovery to lead optimization and thus may serve as a valuable tool for many researchers. - 28Li, Y.; Zhao, Z.; Liu, Z.; Su, M.; Wang, R. AutoT&T v.2: an efficient and versatile tool for lead structure generation and optimization. J. Chem. Inf. Model. 2016, 56, 435– 453, DOI: 10.1021/acs.jcim.5b00691[ACS Full Text
], [CAS], Google Scholar
28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28Xht1KgurY%253D&md5=765cef8f54f2367d902682c547abaf52AutoT&T v.2: An Efficient and Versatile Tool for Lead Structure Generation and OptimizationLi, Yan; Zhao, Zhixiong; Liu, Zhihai; Su, Minyi; Wang, RenxiaoJournal of Chemical Information and Modeling (2016), 56 (2), 435-453CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)In structure-based drug design, automated de novo design methods are helpful tools for lead discovery as well as lead optimization. In a previous study the authors reported a new de novo design method, namely, Automatic Tailoring and Transplanting (AutoT&T). It overcomes some intrinsic problems in conventional fragment-based buildup methods. In this study, the authors describe an upgraded version, namely, AutoT&T2. Structural operations conducted by AutoT&T2 have been largely optimized by introducing several new algorithms. As a result, its overall speed in multiround optimization jobs has been improved by a few thousand fold. With this improvement, it is now practical to conduct structural crossover among multiple lead mols. using AutoT&T2. Three different test cases are described in this study that demonstrate the new features and versatile applications of AutoT&T2. The AutoT&T2 software suite is available to the public. Besides, a Web portal for running AutoT&T2 online is provided at http://www.sioc-ccbg.ac.cn/software/att2 for testing. - 29Nisius, B.; Rester, U. Fragment shuffling: an automated workflow for three-dimensional fragment-based ligand design. J. Chem. Inf. Model. 2009, 49, 1211– 1222, DOI: 10.1021/ci8004572[ACS Full Text
], [CAS], Google Scholar
29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXltlKls74%253D&md5=87aae00271c326a921cf38e7770405c8Fragment Shuffling: An Automated Workflow for Three-Dimensional Fragment-Based Ligand DesignNisius, Britta; Rester, UlrichJournal of Chemical Information and Modeling (2009), 49 (5), 1211-1222CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Fragment-based approaches display a promising alternative in lead discovery. Herein, we present the automated fragment shuffling workflow for the identification of novel lead compds. combining central elements from fragment-based lead identification and structure-based de novo design. Our method is based on sets of aligned 3D ligand structures binding to the same target or target family. The implementation comprises three different ligand fragmentation methods, a scoring scheme assigning individual scores to each fragment, and the incremental construction of novel ligands based on a greedy search algorithm guided by the calcd. fragment scores. The validation of our 3D ligand design workflow is presented on the basis of two pharmaceutically relevant drug targets. A retrospective study based on a selected protein kinase data set revealed that the fragment shuffling approach realizes extended results compared to the well-known BREED technique. Furthermore, we applied our approach in a prospective study for the design of novel non-peptidic thrombin inhibitors. The designed ligand structures in both studies demonstrate the potential of the fragment shuffling workflow. - 30Maass, P.; Schulz-Gasch, T.; Stahl, M.; Rarey, M. Recore: a fast and versatile method for scaffold hopping based on small molecule crystal structure conformations. J. Chem. Inf. Model. 2007, 47, 390– 399, DOI: 10.1021/ci060094h[ACS Full Text
], [CAS], Google Scholar
30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXhvVWjtbk%253D&md5=e9d4e277da49f4ffe2c21472990c2387Recore: A Fast and Versatile Method for Scaffold Hopping Based on Small Molecule Crystal Structure ConformationsMaass, Patrick; Schulz-Gasch, Tanja; Stahl, Martin; Rarey, MatthiasJournal of Chemical Information and Modeling (2007), 47 (2), 390-399CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Replacing central elements of known active structures is a common procedure to enter new compd. classes. Different computational methods have already been developed to help with this task, varying in the description of possible replacements, the query input, and the similarity measure used. In this paper, a novel approach for scaffold replacement and a corresponding software tool, called Recore, is introduced. In contrast to prior methods, the authors main objective was to combine the following three properties in one tool: to avoid structures with strained conformations, to enable the exploration of large search spaces, and to allow interactive use through short response times. The authors introduce a new technique employing 3D fragments generated by combinatorial enumeration of cuts. It allows focusing on fragments suitable for scaffold replacement while retaining conformational information of the corresponding crystal structures. Based on this idea, the authors present an algorithm utilizing a geometric rank searching approach. Given a geometric arrangement of two or three exit vectors and addnl. pharmacophore features, the algorithm finds fragments fulfilling all these constraints ordered by increasing deviation from the query constraints. For the validation of the approach, three different drug design scenarios have been used. The results obtained show that the authors approach is able to propose new valid scaffold topologies. - 31Polishchuk, P. CReM: chemically reasonable mutations framework for structure generation. J. Cheminf. 2020, 12, 28, DOI: 10.1186/s13321-020-00431-w
- 32Lim, J.; Hwang, S.-Y.; Moon, S.; Kim, S.; Kim, W. Y. Scaffold-based molecular design with a graph generative model. Chem. Sci. 2020, 11, 1153– 1164, DOI: 10.1039/C9SC04503A[Crossref], [CAS], Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXit1Ortr%252FO&md5=be36a65abcce15f18b4c1e529bffd905Scaffold-based molecular design with a graph generative modelLim, Jaechang; Hwang, Sang-Yeon; Moon, Seokhyun; Kim, Seungsu; Kim, Woo YounChemical Science (2020), 11 (4), 1153-1164CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)Searching for new mols. in areas like drug discovery often starts from the core structures of known mols. Such a method has called for a strategy of designing deriv. compds. retaining a particular scaffold as a substructure. On this account, our present work proposes a graph generative model that targets its use in scaffold-based mol. design. Our model accepts a mol. scaffold as input and extends it by sequentially adding atoms and bonds. The generated mols. are then guaranteed to contain the scaffold with certainty, and their properties can be controlled by conditioning the generation process on desired properties. The learned rule of extending mols. can well generalize to arbitrary kinds of scaffolds, including those unseen during learning. In the conditional generation of mols., our model can simultaneously control multiple chem. properties despite the search space constrained by fixing the substructure. As a demonstration, we applied our model to designing inhibitors of the epidermal growth factor receptor and show that our model can employ a simple semi-supervised extension to broaden its applicability to situations where only a small amt. of data is available.
- 33Imrie, F.; Hadfield, T. E.; Bradley, A. R.; Deane, C. M. Deep generative design with 3D pharmacophoric constraints. Chem. Sci. 2021, 12, 14577– 14589, DOI: 10.1039/D1SC02436A[Crossref], [PubMed], [CAS], Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitlSrur%252FL&md5=1e51c4d1f28c6c8dff140c4c32d817f4Deep generative design with 3D pharmacophoric constraintsImrie, Fergus; Hadfield, Thomas E.; Bradley, Anthony R.; Deane, Charlotte M.Chemical Science (2021), 12 (43), 14577-14589CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)Generative models have increasingly been proposed as a soln. to the mol. design problem. However, it has proved challenging to control the design process or incorporate prior knowledge, limiting their practical use in drug discovery. In particular, generative methods have made limited use of three-dimensional (3D) structural information even though this is crit. to binding. This work describes a method to incorporate such information and demonstrates the benefit of doing so. We combine an existing graph-based deep generative model, DeLinker, with a convolutional neural network to utilize phys.-meaningful 3D representations of mols. and target pharmacophores. We apply our model, DEVELOP, to both linker and R-group design, demonstrating its suitability for both hit-to-lead and lead optimization. The 3D pharmacophoric information results in improved generation and allows greater control of the design process. In multiple large-scale evaluations, we show that including 3D pharmacophoric constraints results in substantial improvements in the quality of generated mols. On a challenging test set derived from PDBbind, our model improves the proportion of generated mols. with high 3D similarity to the original mol. by over 300%. In addn., DEVELOP recovers 10x more of the original mols. compared to the baseline DeLinker method. Our approach is general-purpose, readily modifiable to alternate 3D representations, and can be incorporated into other generative frameworks.
- 34Hadfield, T. E.; Imrie, F.; Merritt, A.; Birchall, K.; Deane, C. M. Incorporating target-specific pharmacophoric information into deep generative models for fragment elaboration. J. Chem. Inf. Model. 2022, 62, 2280– 2292, DOI: 10.1021/acs.jcim.1c01311[ACS Full Text
], [CAS], Google Scholar
34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhtFGru7rE&md5=c639b01f0c509bbd2e20308fa044bdb5Incorporating Target-Specific Pharmacophoric Information into Deep Generative Models for Fragment ElaborationHadfield, Thomas E.; Imrie, Fergus; Merritt, Andy; Birchall, Kristian; Deane, Charlotte M.Journal of Chemical Information and Modeling (2022), 62 (10), 2280-2292CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Despite recent interest in deep generative models for scaffold elaboration, their applicability to fragment-to-lead campaigns has so far been limited. This is primarily due to their inability to account for local protein structure or a user's design hypothesis. We propose a novel method for fragment elaboration, STRIFE, that overcomes these issues. STRIFE takes as input fragment hotspot maps (FHMs) extd. from a protein target and processes them to provide meaningful and interpretable structural information to its generative model, which in turn is able to rapidly generate elaborations with complementary pharmacophores to the protein. In a large-scale evaluation, STRIFE outperforms existing, structure-unaware, fragment elaboration methods in proposing highly ligand-efficient elaborations. In addn. to automatically extg. pharmacophoric information from a protein target's FHM, STRIFE optionally allows the user to specify their own design hypotheses. - 35Arús-Pous, J.; Patronov, A.; Bjerrum, E. J.; Tyrchan, C.; Reymond, J.-L.; Chen, H.; Engkvist, O. SMILES-based deep generative scaffold decorator for de novo drug design. J. Cheminf. 2020, 12, 38, DOI: 10.1186/s13321-020-00441-8[Crossref], [CAS], Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVyksLjK&md5=05a686a14f942874fd6f6ddbc85017a1SMILES-based deep generative scaffold decorator for de-novo drug designArus-Pous, Josep; Patronov, Atanas; Bjerrum, Esben Jannik; Tyrchan, Christian; Reymond, Jean-Louis; Chen, Hongming; Engkvist, OlaJournal of Cheminformatics (2020), 12 (1), 38CODEN: JCOHB3; ISSN:1758-2946. (SpringerOpen)Herein we report a new SMILES-based mol. generative architecture that generates mols. from scaffolds and can be trained from any arbitrary mol. set. This approach is possible thanks to a new mol. set pre-processing algorithm that exhaustively slices all possible combinations of acyclic bonds of every mol., combinatorically obtaining a large no. of scaffolds with their resp. decorations. Two examples showcasing the potential of the architecture in medicinal and synthetic chem. are described: First, models were trained with a training set obtained from a small set of Dopamine Receptor D2 active modulators and were able to meaningfully decorate a wide range of scaffolds and obtain mol. series predicted active on DRD2. Second, a larger set of drug-like mols. from ChEMBL was selectively sliced using synthetic chem. constraints. This filtering process allowed models trained with this dataset to selectively decorate diverse scaffolds with fragments that were generally predicted to be synthesizable and attachable to the scaffold using known synthetic approaches. In both cases, the models were already able to decorate mols. using specific knowledge without the need to add it with other techniques, such as reinforcement learning. We envision that this architecture will become a useful addn. to the already existent architectures for de novo mol. generation.
- 36Fialková, V.; Zhao, J.; Papadopoulos, K.; Engkvist, O.; Bjerrum, E. J.; Kogej, T.; Patronov, A. LibINVENT: reaction-based generative scaffold decoration for in silico library design. J. Chem. Inf. Model. 2022, 62, 2046– 2063, DOI: 10.1021/acs.jcim.1c00469[ACS Full Text
], [CAS], Google Scholar
36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvV2iu7vI&md5=dc7b0da044f6aa1c2554a43e0aa14672LibINVENT: Reaction-based Generative Scaffold Decoration for in Silico Library DesignFialkova, Vendy; Zhao, Jiaxi; Papadopoulos, Kostas; Engkvist, Ola; Bjerrum, Esben Jannik; Kogej, Thierry; Patronov, AtanasJournal of Chemical Information and Modeling (2022), 62 (9), 2046-2063CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Because of the strong relationship between the desired mol. activity and its structural core, the screening of focused, core-sharing chem. libraries is a key step in lead optimization. Despite the plethora of current research focused on in silico methods for mol. generation, to our knowledge, no tool capable of designing such libraries has been proposed. In this work, we present a novel tool for de novo drug design called LibINVENT. It is capable of rapidly proposing chem. libraries of compds. sharing the same core while maximizing a range of desirable properties. To further help the process of designing focused libraries, the user can list specific chem. reactions that can be used for the library creation. LibINVENT is therefore a flexible tool for generating virtual chem. libraries for lead optimization in a broad range of scenarios. Addnl., the shared core ensures that the compds. in the library are similar, possess desirable properties, and can also be synthesized under the same or similar conditions. The LibINVENT code is freely available in our public repository at https://github.com/MolecularAI/Lib-INVENT. The code necessary for data preprocessing is further available at: https://github.com/MolecularAI/Lib-INVENT-dataset. - 37Li, Y.; Hu, J.; Wang, Y.; Zhou, J.; Zhang, L.; Liu, Z. DeepScaffold: a comprehensive tool for scaffold-based de novo drug discovery using deep learning. J. Chem. Inf. Model. 2020, 60, 77– 91, DOI: 10.1021/acs.jcim.9b00727[ACS Full Text
], [CAS], Google Scholar
37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXitlerurnN&md5=44ba7010e4824a37d97c66eed5bd1d6dDeepScaffold: A Comprehensive Tool for Scaffold-Based De Novo Drug Discovery Using Deep LearningLi, Yibo; Hu, Jianxing; Wang, Yanxing; Zhou, Jielong; Zhang, Liangren; Liu, ZhenmingJournal of Chemical Information and Modeling (2020), 60 (1), 77-91CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)The ultimate goal of drug design is to find novel compds. with desirable pharmacol. properties. Designing mols. retaining particular scaffolds as their core structures is an efficient way to obtain potential drug candidates. We propose a scaffold-based mol. generative model for drug discovery, which performs mol. generation based on a wide spectrum of scaffold definitions, including Bemis-Murcko scaffolds, cyclic skeletons, and scaffolds with specifications on side-chain properties. The model can generalize the learned chem. rules of adding atoms and bonds to a given scaffold. The generated compds. were evaluated by mol. docking in DRD2 targets, and the results demonstrated that this approach can be effectively applied to solve several drug design problems, including the generation of compds. contg. a given scaffold and de novo drug design of potential drug candidates with specific docking scores. - 38Yang, Y.; Zheng, S.; Su, S.; Zhao, C.; Xu, J.; Chen, H. SyntaLinker: automatic fragment linking with deep conditional transformer neural networks. Chem. Sci. 2020, 11, 8312– 8322, DOI: 10.1039/D0SC03126G[Crossref], [PubMed], [CAS], Google Scholar38https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhsVagsrvK&md5=403cc7a09f96ecbe27afa3ce6bee51a2SyntaLinker: automatic fragment linking with deep conditional transformer neural networksYang, Yuyao; Zheng, Shuangjia; Su, Shimin; Zhao, Chao; Xu, Jun; Chen, HongmingChemical Science (2020), 11 (31), 8312-8322CODEN: CSHCCN; ISSN:2041-6520. (Royal Society of Chemistry)Linking fragments to generate a focused compd. library for a specific drug target is one of the challenges in fragment-based drug design (FBDD). Hereby, we propose a new program named SyntaLinker, which is based on a syntactic pattern recognition approach using deep conditional transformer neural networks. This state-of-the-art transformer can link mol. fragments automatically by learning from the knowledge of structures in medicinal chem. databases (e.g.ChEMBL database). Conventionally, linking mol. fragments was viewed as connecting substructures that were predefined by empirical rules. In SyntaLinker, however, the rules of linking fragments can be learned implicitly from known chem. structures by recognizing syntactic patterns embedded in SMILES notations. With deep conditional transformer neural networks, SyntaLinker can generate mol. structures based on a given pair of fragments and addnl. restrictions. Case studies have demonstrated the advantages and usefulness of SyntaLinker in FBDD.
- 39Feng, Y.; Yang, Y.; Deng, W.; Chen, H.; Ran, T. SyntaLinker-Hybrid: a deep learning approach for target specific drug design. Artif. Intell. Life Sci. 2022, 2, 100035, DOI: 10.1016/j.ailsci.2022.100035
- 40Huang, Y.; Peng, X.; Ma, J.; Zhang, M. 3DLinker: an E(3) equivariant variational autoencoder for molecular linker design. arXiv Preprint , arXiv:2205.07309, 2022.Google ScholarThere is no corresponding record for this reference.
- 41Imrie, F.; Bradley, A. R.; van der Schaar, M.; Deane, C. M. Deep generative models for 3D linker design. J. Chem. Inf. Model. 2020, 60, 1983– 1995, DOI: 10.1021/acs.jcim.9b01120[ACS Full Text
], [CAS], Google Scholar
41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXltlenurs%253D&md5=430c929761c5020d1013538e028ba1bcDeep Generative Models for 3D Linker DesignImrie, Fergus; Bradley, Anthony R.; van der Schaar, Mihaela; Deane, Charlotte M.Journal of Chemical Information and Modeling (2020), 60 (4), 1983-1995CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Rational compd. design remains a challenging problem for both computational methods and medicinal chemists. Computational generative methods have begun to show promising results for the design problem. However, they have not yet used the power of three-dimensional (3D) structural information. We have developed a novel graph-based deep generative model that combines state-of-the-art machine learning techniques with structural knowledge. Our method ("DeLinker") takes two fragments or partial structures and designs a mol. incorporating both. The generation process is protein-context-dependent, utilizing the relative distance and orientation between the partial structures. This 3D information is vital to successful compd. design, and we demonstrate its impact on the generation process and the limitations of omitting such information. In a large-scale evaluation, DeLinker designed 60% more mols. with high 3D similarity to the original mol. than a database baseline. When considering the more relevant problem of longer linkers with at least five atoms, the outperformance increased to 200%. We demonstrate the effectiveness and applicability of this approach on a diverse range of design problems: fragment linking, scaffold hopping, and proteolysis targeting chimera (PROTAC) design. As far as we are aware, this is the first mol. generative model to incorporate 3D structural information directly in the design process. The code is available at https://github.com/oxpig/DeLinker. - 42Andrianov, G. V.; Gabriel Ong, W. J.; Serebriiskii, I.; Karanicolas, J. Efficient Hit-to-Lead Searching of Kinase Inhibitor Chemical Space via Computational Fragment Merging. J. Chem. Inf. Model. 2021, 61, 5967– 5987, DOI: 10.1021/acs.jcim.1c00630[ACS Full Text
], [CAS], Google Scholar
42https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXisVGrs7zE&md5=d00fcbed94deb5955f19c3ee8d26af39Efficient Hit-to-Lead Searching of Kinase Inhibitor Chemical Space via Computational Fragment MergingAndrianov, Grigorii V.; Gabriel Ong, Wern Juin; Serebriiskii, Ilya; Karanicolas, JohnJournal of Chemical Information and Modeling (2021), 61 (12), 5967-5987CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)In early-stage drug discovery, the hit-to-lead optimization (or "hit expansion") stage entails starting from a newly identified active compd. and improving its potency or other properties. Traditionally, this process relies on synthesizing and evaluating a series of analogs to build up structure-activity relationships. Here, we describe a computational strategy focused on kinase inhibitors, intended to expedite the process of identifying analogs with improved potency. Our protocol begins from an inhibitor of the target kinase and generalizes the synthetic route used to access it. By searching for com. available replacements for the individual building blocks used to make the parent inhibitor, we compile an enumerated library of compds. that can be accessed using the same chem. transformations; these huge libraries can exceed many millions-or billions-of compds. Because the resulting libraries are much too large for explicit virtual screening, we instead consider alternate approaches to identify the top-scoring compds. We find that contributions from individual substituents are well described by a pairwise additivity approxn., provided that the corresponding fragments position their shared core in precisely the same way relative to the binding site. This key insight allows us to det. which fragments are suitable for merging into single new compds. and which are not. Further, the use of pairwise approxn. allows interaction energies to be assigned to each compd. in the library without the need for any further structure-based modeling: interaction energies instead can be reliably estd. from the energies of the component fragments, and the reduced computational requirements allow for flexible energy minimizations that allow the kinase to respond to each substitution. We demonstrate this protocol using libraries built from six representative kinase inhibitors drawn from the literature, which target five different kinases: CDK9, CHK1, CDK2, EGFRT790M, and ACK1. In each example, the enumerated library includes addnl. analogs reported by the original study to have activity, and these analogs are successfully prioritized within the library. We envision that the insights from this work can facilitate the rapid assembly and screening of increasingly large libraries for focused hit-to-lead optimization. To enable adoption of these methods and to encourage further analyses, we disseminate the computational tools needed to deploy this protocol. - 43Hall, R. J.; Murray, C. W.; Verdonk, M. L. The Fragment Network: a chemistry recommendation engine built using a graph database. J. Med. Chem. 2017, 60, 6440– 6450, DOI: 10.1021/acs.jmedchem.7b00809[ACS Full Text
], [CAS], Google Scholar
43https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhtFKlsr3J&md5=79842771a068cdaabc2f9a841d791c1cThe Fragment Network: A Chemistry Recommendation Engine Built Using a Graph DatabaseHall, Richard J.; Murray, Christopher W.; Verdonk, Marcel L.Journal of Medicinal Chemistry (2017), 60 (14), 6440-6450CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The hit validation stage of a fragment-based drug discovery campaign involves probing the SAR around one or more fragment hits. This often requires a search for similar compds. in a corporate collection or from com. suppliers. The Fragment Network is a graph database that allows a user to efficiently search chem. space around a compd. of interest. The result set is chem. intuitive, naturally grouped by substitution pattern and meaningfully sorted according to the no. of observations of each transformation in medicinal chem. databases. This paper describes the algorithms used to construct and search the Fragment Network and provides examples of how it may be used in a drug discovery context. - 44Landrum, G. RDKit: A software suite for cheminformatics, computational chemistry, and predictive modeling. https://www.rdkit.org/ (accessed November 2022).Google ScholarThere is no corresponding record for this reference.
- 45Neo4j, Graph Modeling Guidelines. https://neo4j.com/developer/guide-data-modeling/ (accessed November 2022).Google ScholarThere is no corresponding record for this reference.
- 46Newman, J. A.; Douangamath, A.; Yadzani, S.; Yosaatmadja, Y.; Aimon, A.; Brandão-Neto, J.; Dunnett, L.; Gorrie-stone, T.; Skyner, R.; Fearon, D.; Schapira, M.; von Delft, F.; Gileadi, O. Structure, mechanism and crystallographic fragment screening of the SARS-CoV-2 NSP13 helicase. Nat. Commun. 2021, 12, 4848, DOI: 10.1038/s41467-021-25166-6[Crossref], [PubMed], [CAS], Google Scholar46https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhslOns7%252FI&md5=3e643800631b8bb56328c4ddd97fb7a7Structure, mechanism and crystallographic fragment screening of the SARS-CoV-2 NSP13 helicaseNewman, Joseph A.; Douangamath, Alice; Yadzani, Setayesh; Yosaatmadja, Yuliana; Aimon, Antony; Brandao-Neto, Jose; Dunnett, Louise; Gorrie-stone, Tyler; Skyner, Rachael; Fearon, Daren; Schapira, Matthieu; von Delft, Frank; Gileadi, OpherNature Communications (2021), 12 (1), 4848CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Abstr.: There is currently a lack of effective drugs to treat people infected with SARS-CoV-2, the cause of the global COVID-19 pandemic. The SARS-CoV-2 Non-structural protein 13 (NSP13) has been identified as a target for anti-virals due to its high sequence conservation and essential role in viral replication. Structural anal. reveals two "druggable" pockets on NSP13 that are among the most conserved sites in the entire SARS-CoV-2 proteome. Here we present crystal structures of SARS-CoV-2 NSP13 solved in the APO form and in the presence of both phosphate and a non-hydrolysable ATP analog. Comparisons of these structures reveal details of conformational changes that provide insights into the helicase mechanism and possible modes of inhibition. To identify starting points for drug development we have performed a crystallog. fragment screen against NSP13. The screen reveals 65 fragment hits across 52 datasets opening the way to structure guided development of novel antiviral agents.
- 47Jin, Z.; Du, X.; Xu, Y.; Deng, Y.; Liu, M.; Zhao, Y.; Zhang, B.; Li, X.; Zhang, L.; Peng, C.; Duan, Y.; Yu, J.; Wang, L.; Yang, K.; Liu, F.; Jiang, R.; Yang, X.; You, T.; Liu, X.; Yang, X.; Bai, F.; Liu, H.; Liu, X.; Guddat, L. W.; Xu, W.; Xiao, G.; Qin, C.; Shi, Z.; Jiang, H.; Rao, Z.; Yang, H. Structure of Mpro from SARS-CoV-2 and discovery of its inhibitors. Nature 2020, 582, 289– 293, DOI: 10.1038/s41586-020-2223-y[Crossref], [PubMed], [CAS], Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtVyhsrrO&md5=b84f350fe9ce1109485df6caf814ba82Structure of Mpro from SARS-CoV-2 and discovery of its inhibitorsJin, Zhenming; Du, Xiaoyu; Xu, Yechun; Deng, Yongqiang; Liu, Meiqin; Zhao, Yao; Zhang, Bing; Li, Xiaofeng; Zhang, Leike; Peng, Chao; Duan, Yinkai; Yu, Jing; Wang, Lin; Yang, Kailin; Liu, Fengjiang; Jiang, Rendi; Yang, Xinglou; You, Tian; Liu, Xiaoce; Yang, Xiuna; Bai, Fang; Liu, Hong; Liu, Xiang; Guddat, Luke W.; Xu, Wenqing; Xiao, Gengfu; Qin, Chengfeng; Shi, Zhengli; Jiang, Hualiang; Rao, Zihe; Yang, HaitaoNature (London, United Kingdom) (2020), 582 (7811), 289-293CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Abstr.: A new coronavirus, known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is the etiol. agent responsible for the 2019-2020 viral pneumonia outbreak of coronavirus disease 2019 (COVID-19). Currently, there are no targeted therapeutic agents for the treatment of this disease, and effective treatment options remain very limited. Here, we describe the results of a program that aimed to rapidly discover lead compds. for clin. use, by combining structure-assisted drug design, virtual drug screening and high-throughput screening. This program focused on identifying drug leads that target main protease (Mpro) of SARS-CoV-2: Mpro is a key enzyme of coronaviruses and has a pivotal role in mediating viral replication and transcription, making it an attractive drug target for SARS-CoV-2. We identified a mechanism-based inhibitor (N3) by computer-aided drug design, and then detd. the crystal structure of Mpro of SARS-CoV-2 in complex with this compd. Through a combination of structure-based virtual and high-throughput screening, we assayed more than 10,000 compds.-including approved drugs, drug candidates in clin. trials and other pharmacol. active compds.-as inhibitors of Mpro. Six of these compds. inhibited Mpro, showing half-maximal inhibitory concn. values that ranged from 0.67 to 21.4μM. One of these compds. (ebselen) also exhibited promising antiviral activity in cell-based assays. Our results demonstrate the efficacy of our screening strategy, which can lead to the rapid discovery of drug leads with clin. potential in response to new infectious diseases for which no specific drugs or vaccines are available.
- 48The COVID Moonshot Consortium. Achdout, H.; Aimon, A.; Bar-David, E.; Barr, H.; Ben-Shmuel, A.; Bennett, J.; Bilenko, V. A.; Bilenko, V. A.; Boby, M. L.; Borden, B.; Bowman, G. R.; Brun, J.; BVNBS, S.; Calmiano, M.; Carbery, A.; Carney, D.; Cattermole, E.; Chang, E.; Chernyshenko, E.; Chodera, J. D.; Clyde, A.; Coffland, J. E.; Cohen, G.; Cole, J.; Contini, A.; Cox, L.; Cvitkovic, M.; Dias, A.; Donckers, K.; Dotson, D. L.; Douangamath, A.; Duberstein, S.; Dudgeon, T.; Dunnett, L.; Eastman, P. K.; Erez, N.; Eyermann, C. J.; Fairhead, M.; Fate, G.; Fearon, D.; Fedorov, O.; Ferla, M.; Fernandes, R. S.; Ferrins, L.; Foster, R.; Foster, H.; Gabizon, R.; Garcia-Sastre, A.; Gawriljuk, V. O.; Gehrtz, P.; Gileadi, C.; Giroud, C.; Glass, W. G.; Glen, R.; Glinert, I.; Godoy, A. S.; Gorichko, M.; Gorrie-Stone, T.; Griffen, E. J.; Hart, S. H.; Heer, J.; Henry, M.; Hill, M.; Horrell, S.; Huliak, V. D.; Hurley, M. F.; Israely, T.; Jajack, A.; Jansen, J.; Jnoff, E.; Jochmans, D.; John, T.; Jonghe, S. D.; Kantsadi, A. L.; Kenny, P. W.; Kiappes, J. L.; Kinakh, S. O.; Koekemoer, L.; Kovar, B.; Krojer, T.; Lee, A.; Lefker, B. A.; Levy, H.; Logvinenko, I. G.; London, N.; Lukacik, P.; Macdonald, H. B.; MacLean, B.; Malla, T. R.; Matviiuk, T.; McCorkindale, W.; McGovern, B. L.; Melamed, S.; Melnykov, K. P.; Michurin, O.; Mikolajek, H.; Milne, B. F.; Morris, A.; Morris, G. M.; Morwitzer, M. J.; Moustakas, D.; Nakamura, A. M.; Neto, J. B.; Neyts, J.; Nguyen, L.; Noske, G. D.; Oleinikovas, V.; Oliva, G.; Overheul, G. J.; Owen, D.; Pai, R.; Pan, J.; Paran, N.; Perry, B.; Pingle, M.; Pinjari, J.; Politi, B.; Powell, A.; Psenak, V.; Puni, R.; Rangel, V. L.; Reddi, R. N.; Reid, S. P.; Resnick, E.; Ripka, E. G.; Robinson, M. C.; Robinson, R. P.; Rodriguez-Guerra, J.; Rosales, R.; Rufa, D.; Saar, K.; Saikatendu, K. S.; Schofield, C.; Shafeev, M.; Shaikh, A.; Shi, J.; Shurrush, K.; Singh, S.; Sittner, A.; Skyner, R.; Smalley, A.; Smeets, B.; Smilova, M. D.; Solmesky, L. J.; Spencer, J.; Strain-Damerell, C.; Swamy, V.; Tamir, H.; Tennant, R.; Thompson, W.; Thompson, A.; Tomasio, S.; Tsurupa, I. S.; Tumber, A.; Vakonakis, I.; van Rij, R. P.; Vangeel, L.; Varghese, F. S.; Vaschetto, M.; Vitner, E. B.; Voelz, V.; Volkamer, A.; von Delft, F.; von Delft, A.; Walsh, M.; Ward, W.; Weatherall, C.; Weiss, S.; White, K. M.; Wild, C. F.; Wittmann, M.; Wright, N.; Yahalom-Ronen, Y.; Zaidmann, D.; Zidane, H.; Zitzmann, N. Open science discovery of oral non-covalent SARS-CoV-2 main protease inhibitor therapeutics. bioRxiv Preprint , updated version, 2022. DOI: 10.1101/2020.10.29.339317
- 49Wahlberg, E.; Karlberg, T.; Kouznetsova, E.; Markova, N.; Macchiarulo, A.; Thorsell, A.-G.; Pol, E.; Frostell, s.; Ekblad, T.; Öncü, D.; Kull, B.; Robertson, G. M.; Pellicciari, R.; Schüler, H.; Weigelt, J. Family-wide chemical profiling and structural analysis of PARP and tankyrase inhibitors. Nat. Biotechnol. 2012, 30, 283– 288, DOI: 10.1038/nbt.2121[Crossref], [PubMed], [CAS], Google Scholar49https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XisVKrsb4%253D&md5=71f17fd5b6bb602dd70c8fcc17df95f9Family-wide chemical profiling and structural analysis of PARP and tankyrase inhibitorsWahlberg, Elisabet; Karlberg, Tobias; Kouznetsova, Ekaterina; Markova, Natalia; Macchiarulo, Antonio; Thorsell, Ann-Gerd; Pol, Ewa; Frostell, Aasa; Ekblad, Torun; Oencue, Delal; Kull, Bjoern; Robertson, Graeme Michael; Pellicciari, Roberto; Schueler, Herwig; Weigelt, JohanNature Biotechnology (2012), 30 (3), 283-288CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)Inhibitors of poly-ADP-ribose polymerase (PARP) family proteins are currently in clin. trials as cancer therapeutics, yet the specificity of many of these compds. is unknown. Here we evaluated a series of 185 small-mol. inhibitors, including research reagents and compds. being tested clin., for the ability to bind to the catalytic domains of 13 of the 17 human PARP family members including the tankyrases, TNKS1 and TNKS2. Many of the best-known inhibitors, including TIQ-A, 6(5H)-phenanthridinone, olaparib, ABT-888 and rucaparib, bound to several PARP family members, suggesting that these mols. lack specificity and have promiscuous inhibitory activity. We also detd. X-ray crystal structures for five TNKS2 ligand complexes and four PARP14 ligand complexes. In addn. to showing that the majority of PARP inhibitors bind multiple targets, these results provide insight into the design of new inhibitors.
- 50Ohara-Nemoto, Y.; Shimoyama, Y.; Kimura, S.; Kon, A.; Haraga, H.; Ono, T.; Nemoto, T. K. Asp- and Glu-specific novel dipeptidyl peptidase 11 of Porphyromonas gingivalis ensures utilization of proteinaceous energy sources. J. Biol. Chem. 2011, 286, 38115– 38127, DOI: 10.1074/jbc.M111.278572[Crossref], [PubMed], [CAS], Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtlyktb%252FK&md5=419046327fe3bfdfb5619d722d9f9755Asp- and Glu-specific Novel Dipeptidyl Peptidase 11 of Porphyromonas gingivalis Ensures Utilization of Proteinaceous Energy SourcesOhara-Nemoto, Yuko; Shimoyama, Yu; Kimura, Shigenobu; Kon, Asako; Haraga, Hiroshi; Ono, Toshio; Nemoto, Takayuki K.Journal of Biological Chemistry (2011), 286 (44), 38115-38127, S38115/1-S38115/9CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Porphyromonas gingivalis and Porphyromonas endodontalis, asaccharolytic black-pigmented anaerobes, are predominant pathogens of human chronic and periapical periodontitis, resp. They incorporate di- and tripeptides from the environment as carbon and energy sources. In the present study we cloned a novel dipeptidyl peptidase (DPP) gene of P. endodontalis ATCC 35406, designated as DPP11. The DPP11 gene encoded 717 amino acids with a mol. mass of 81,090 Da and was present as a 75-kDa form with an N terminus of Asp22. A homol. search revealed the presence of a P. gingivalis ortholog, PGN0607, that has been categorized as an isoform of authentic DPP7. P. gingivalis DPP11 was exclusively cell-assocd. as a truncated 60-kDa form, and the gene ablation retarded cell growth. DPP11 specifically removed dipeptides from oligopeptides with the penultimate N-terminal Asp and Glu and has a P2-position preference to hydrophobic residues. Optimum pH was 7.0, and the kcat/Km value was higher for Asp than Glu. Those activities were lost by substitution of Ser652 in P. endodontalis and Ser655 in P. gingivalis DPP11 to Ala, and they were consistently decreased with increasing NaCl concn. Arg670 is a unique amino acid completely conserved in all DPP11 members distributed in the genera Porphyromonas, Bacteroides, and Parabacteroides, whereas this residue is converted to Gly in all authentic DPP7 members. Substitution anal. suggested that Arg670 interacts with an acidic residue of the substrate. Considered to preferentially utilize acidic amino acids, DPP11 ensures efficient degrdn. of oligopeptide substrates in these Gram-neg. anaerobic rods.
- 51Lipinski, C. A.; Lombardo, F.; Dominy, B. W.; Feeney, P. J. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Delivery Rev. 1997, 23, 3– 25, DOI: 10.1016/S0169-409X(96)00423-1[Crossref], [CAS], Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2sXktlKlsQ%253D%253D&md5=405f70b0594d428f1275e1d56642cd3aExperimental and computational approaches to estimate solubility and permeability in drug discovery and development settingsLipinski, Christopher A.; Lombardo, Franco; Dominy, Beryl W.; Feeney, Paul J.Advanced Drug Delivery Reviews (1997), 23 (1-3), 3-25CODEN: ADDREP; ISSN:0169-409X. (Elsevier)A review with 50 refs. Exptl. and computational approaches to est. soly. and permeability in discovery and development settings are described. In the discovery setting 'the rule of 5' predicts that poor absorption or permeation is more likely when there are more than 5 H-bond donors, 10 H-bond acceptors, the mol. wt. (MWT) is >500 and the calcd. Log P (CLogP) is >5. Computational methodol. for the rule-based Moriguchi Log P (MLogP) calcn. is described. Turbidimetric soly. measurement is described and applied to known drugs. High throughput screening (HTS) leads tend to have higher MWT and Log P and lower turbidimetric soly. than leads in the pre-HTS era. In the development setting, soly. calcns. focus on exact value prediction and are difficult because of polymorphism. Recent work on linear free energy relationships and Log P approaches are critically reviewed. Useful predictions are possible in closely related analog series when coupled with exptl. thermodn. soly. measurements.
- 52Veber, D. F.; Johnson, S. R.; Cheng, H.-Y.; Smith, B. R.; Ward, K. W.; Kopple, K. D. Molecular properties that influence the oral bioavailability of drug candidates. J. Med. Chem. 2002, 45, 2615– 2623, DOI: 10.1021/jm020017n[ACS Full Text
], [CAS], Google Scholar
52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XjsFCmt7g%253D&md5=eaad26ed6a259de82ad65a8834fc397dMolecular Properties That Influence the Oral Bioavailability of Drug CandidatesVeber, Daniel F.; Johnson, Stephen R.; Cheng, Hung-Yuan; Smith, Brian R.; Ward, Keith W.; Kopple, Kenneth D.Journal of Medicinal Chemistry (2002), 45 (12), 2615-2623CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Oral bioavailability measurements in rats for over 1100 drug candidates studied at Smith-Kline Beecham Pharmaceuticals (now Glaxo Smith-Kline) have allowed us to analyze the relative importance of mol. properties considered to influence that drug property. Reduced mol. flexibility, as measured by the no. of rotatable bonds, and low polar surface area or total hydrogen bond count (sum of donors and acceptors) are found to be important predictors of good oral bioavailability, independent of mol. wt. That on av. both the no. of rotatable bonds and polar surface area or hydrogen bond count tend to increase with mol. wt. may in part explain the success of the mol. wt. parameter in predicting oral bioavailability. The commonly applied mol. wt. cutoff at 500 does not itself significantly sep. compds. with poor oral bioavailability from those with acceptable values in this extensive data set. Our observations suggest that compds. which meet only the 2 criteria of (1) 10 or fewer rotatable bonds and (2) polar surface area ≤140 Å2 (or 12 or fewer H-bond donors and acceptors) will have a high probability of good oral bioavailability in the rat. Data sets for the artificial membrane permeation rate and for clearance in the rat were also examd. Reduced polar surface area correlates better with increased permeation rate than does lipophilicity (C log P), and increased rotatable bond count has a neg. effect on the permeation rate. A threshold permeation rate is a prerequisite of oral bioavailability. The rotatable bond count does not correlate with the data examd. here for the in vivo clearance rate in the rat. - 53Gaulton, A.; Bellis, L. J.; Bento, A. P.; Chambers, J.; Davies, M.; Hersey, A.; Light, Y.; McGlinchey, S.; Michalovich, D.; Al-Lazikani, B.; Overington, J. P. ChEMBL: a large-scale bioactivity database for drug discovery. Nucleic Acids Res. 2012, 40, D1100– D1107, DOI: 10.1093/nar/gkr777[Crossref], [PubMed], [CAS], Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhs12htbjN&md5=aedf7793e1ca54b6a4fa272ea3ef7d0eChEMBL: a large-scale bioactivity database for drug discoveryGaulton, Anna; Bellis, Louisa J.; Bento, A. Patricia; Chambers, Jon; Davies, Mark; Hersey, Anne; Light, Yvonne; McGlinchey, Shaun; Michalovich, David; Al-Lazikani, Bissan; Overington, John P.Nucleic Acids Research (2012), 40 (D1), D1100-D1107CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)ChEMBL is an Open Data database contg. binding, functional and ADMET information for a large no. of drug-like bioactive compds. These data are manually abstracted from the primary published literature on a regular basis, then further curated and standardized to maximize their quality and utility across a wide range of chem. biol. and drug-discovery research problems. Currently, the database contains 5.4 million bioactivity measurements for more than 1 million compds. and 5200 protein targets. Access is available through a web-based interface, data downloads and web services at: https://www.ebi.ac.uk/chembldb.
- 54Ebejer, J.-P.; Morris, G. M.; Deane, C. M. Freely available conformer generation methods: how good are they?. J. Chem. Inf. Model. 2012, 52, 1146– 1158, DOI: 10.1021/ci2004658[ACS Full Text
], [CAS], Google Scholar
54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xlt1Snu7s%253D&md5=2990e7dcfc83d8c0f7671250c51aeb97Freely Available Conformer Generation Methods: How Good Are They?Ebejer, Jean-Paul; Morris, Garrett M.; Deane, Charlotte M.Journal of Chemical Information and Modeling (2012), 52 (5), 1146-1158CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)A review. Conformer generation has important implications in cheminformatics, particularly in computational drug discovery where the quality of conformer generation software may affect the outcome of a virtual screening exercise. We examine the performance of four freely available small mol. conformer generation tools (Balloon, Confab, Frog2, and RDKit) alongside a com. tool (MOE). The aim of this study is 3-fold: (i) to identify which tools most accurately reproduce exptl. detd. structures; (ii) to examine the diversity of the generated conformational set; and (iii) to benchmark the computational time expended. These aspects were tested using a set of 708 drug-like mols. assembled from the OMEGA validation set and the Astex Diverse Set. These mols. have varying physicochem. properties and at least one known X-ray crystal structure. We found that RDKit and Confab are statistically better than other methods at generating low rmsd conformers to the known structure. RDKit is particularly suited for less flexible mols. while Confab, with its systematic approach, is able to generate conformers which are geometrically closer to the exptl. detd. structure for mols. with a large no. of rotatable bonds (≥10). In our tests RDKit also resulted as the second fastest method after Frog2. In order to enhance the performance of RDKit, we developed a postprocessing algorithm to build a diverse and representative set of conformers which also contains a close conformer to the known structure. Our anal. indicates that, with postprocessing, RDKit is a valid free alternative to com., proprietary software. - 55Su, M.; Yang, Q.; Du, Y.; Feng, G.; Liu, Z.; Li, Y.; Wang, R. Comparative assessment of scoring functions: the CASF-2016 update. J. Chem. Inf. Model. 2019, 59, 895– 913, DOI: 10.1021/acs.jcim.8b00545[ACS Full Text
], [CAS], Google Scholar
55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitlWhtLjM&md5=573295cfd2298a2dee933027041c3d9cComparative Assessment of Scoring Functions: The CASF-2016 UpdateSu, Minyi; Yang, Qifan; Du, Yu; Feng, Guoqin; Liu, Zhihai; Li, Yan; Wang, RenxiaoJournal of Chemical Information and Modeling (2019), 59 (2), 895-913CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)In structure-based drug design, scoring functions are often employed to evaluate protein-ligand interactions. A variety of scoring functions have been developed so far, and thus, some objective benchmarks are desired for assessing their strength and weakness. The comparative assessment of scoring functions (CASF) benchmark developed by us provides an answer to this demand. CASF is designed as a "scoring benchmark", where the scoring process is decoupled from the docking process to depict the performance of scoring function more precisely. Here, we describe the latest update of this benchmark, i.e., CASF-2016. Each scoring function is still evaluated by four metrics, including "scoring power", "ranking power", "docking power", and "screening power". Nevertheless, the evaluation methods have been improved considerably in several aspects. A new test set is compiled, which consists of 285 protein-ligand complexes with high-quality crystal structures and reliable binding consts. A panel of 25 scoring functions are tested on CASF-2016 as a demonstration. Our results reveal that the performance of current scoring functions is more promising in terms of docking power than scoring, ranking, and screening power. Scoring power is somewhat correlated with ranking power, so are docking power and screening power. The results obtained on CASF-2016 may provide valuable guidance for the end users to make smart choices among available scoring functions. Moreover, CASF is created as an open-access benchmark so that other researchers can utilize it to test a wider range of scoring functions. The complete CASF-2016 benchmark will be released on the PDBbind-CN web server (http://www.pdbbind-cn.org/casf.asp/) once this article is published. - 56Ferla, M. Fragmenstein. https://github.com/matteoferla/Fragmenstein (accessed November 2022).Google ScholarThere is no corresponding record for this reference.
- 57Chaudhury, S.; Lyskov, S.; Gray, J. J. PyRosetta: a script-based interface for implementing molecular modeling algorithms using Rosetta. Bioinformatics 2010, 26, 689– 691, DOI: 10.1093/bioinformatics/btq007[Crossref], [PubMed], [CAS], Google Scholar57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXis1Wnt70%253D&md5=261b3b6b59e4987911556ea5314d9b19PyRosetta: a script-based interface for implementing molecular modeling algorithms using RosettaChaudhury, Sidhartha; Lyskov, Sergey; Gray, Jeffrey J.Bioinformatics (2010), 26 (5), 689-691CODEN: BOINFP; ISSN:1367-4803. (Oxford University Press)Summary: PyRosetta is a stand-alone Python-based implementation of the Rosetta mol. modeling package that allows users to write custom structure prediction and design algorithms using the major Rosetta sampling and scoring functions. PyRosetta contains Python bindings to libraries that define Rosetta functions including those for accessing and manipulating protein structure, calcg. energies and running Monte Carlo-based simulations. PyRosetta can be used in two ways: (i) interactively, using iPython and (ii) script-based, using Python scripting. Interactive mode contains a no. of help features and is ideal for beginners while script-mode is best suited for algorithm development. PyRosetta has similar computational performance to Rosetta, can be easily scaled up for cluster applications and has been implemented for algorithms demonstrating protein docking, protein folding, loop modeling and design. Availability: PyRosetta is a stand-alone package available at http://www.pyrosetta.org under the Rosetta license which is free for academic and non-profit users. A tutorial, user's manual and sample scripts demonstrating usage are also available on the web site.
- 58Adasme, M. F.; Linnemann, K. L.; Bolz, S. N.; Kaiser, F.; Salentin, S.; Haupt, V.; Schroeder, M. PLIP 2021: expanding the scope of the protein–ligand interaction profiler to DNA and RNA. Nucleic Acids Res. 2021, 49, W530– W534, DOI: 10.1093/nar/gkab294[Crossref], [PubMed], [CAS], Google Scholar58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvV2isLbL&md5=8f56ced733405aaa8f5325e3fa4b862cPLIP 2021 expanding scope of protein-ligand interaction profiler to DNA and RNAAdasme, Melissa F.; Linnemann, Katja L.; Bolz, Sarah Naomi; Kaiser, Florian; Salentin, Sebastian; Haupt, V. Joachim; Schroeder, MichaelNucleic Acids Research (2021), 49 (W1), W530-W534CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)With the growth of protein structure data, the anal. of mol. interactions between ligands and their target mols. is gaining importance. PLIP, the protein-ligand interaction profiler, detects and visualises these interactions and provides data in formats suitable for further processing. PLIP has proven very successful in applications ranging from the characterization of docking expts. to the assessment of novel ligand-protein complexes. Besides ligand-protein interactions, interactions with DNA and RNA play a vital role in many applications, such as drugs targeting DNA or RNA-binding proteins. To date, over 7% of all 3D structures in the Protein Data Bank include DNA or RNA. Therefore, we extended PLIP to encompass these important mols. We demonstrate the power of this extension with examples of a cancer drug binding to a DNA target, and an RNA-protein complex central to a neurol. disease.
- 59Carbery, A.; Skyner, R.; von Delft, F.; Deane, C. M. Fragment libraries designed to be functionally diverse recover protein binding information more efficiently than standard structurally diverse libraries. J. Med. Chem. 2022, 65, 11404– 11413, DOI: 10.1021/acs.jmedchem.2c01004[ACS Full Text
], [CAS], Google Scholar
59https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XitFWktLnO&md5=20c210891befe38eb6ec658cba2a01b8Fragment Libraries Designed to Be Functionally Diverse Recover Protein Binding Information More Efficiently Than Standard Structurally Diverse LibrariesCarbery, Anna; Skyner, Rachael; von Delft, Frank; Deane, Charlotte M.Journal of Medicinal Chemistry (2022), 65 (16), 11404-11413CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Current fragment-based drug design relies on the efficient exploration of chem. space by using structurally diverse libraries of small fragments. However, structurally dissimilar compds. can exploit the same interactions and thus be functionally similar. Using three-dimensional structures of many fragments bound to multiple targets, we examd. if a better strategy for selecting fragments for screening libraries exists. We show that structurally diverse fragments can be described as functionally redundant, often making the same interactions. Ranking fragments by the no. of novel interactions they made, we show that functionally diverse selections of fragments substantially increase the amt. of information recovered for unseen targets compared to the amts. recovered by other methods of selection. Using these results, we design small functionally efficient libraries that can give significantly more information about new protein targets than similarly sized structurally diverse libraries. By covering more functional space, we can generate more diverse sets of drug leads. - 60Maaten, L. v. d.; Hinton, G. Visualizing data using t-SNE. J. Mach. Learn. Res. 2008, 9, 2579– 2605Google ScholarThere is no corresponding record for this reference.
- 61Pedregosa, F.; Varoquaux, G.; Gramfort, A.; Michel, V.; Thirion, B.; Grisel, O.; Blondel, M.; Prettenhofer, P.; Weiss, R.; Dubourg, V.; Vanderplas, J.; Passos, A.; Cournapeau, D.; Brucher, M.; Perrot, M.; Duchesnay, d. Scikit-learn: machine learning in Python. J. Mach. Learn. Res. 2011, 12, 2825– 2830Google ScholarThere is no corresponding record for this reference.
- 62PostEra, Mpro activity data. https://covid.postera.ai/covid/activity_data (accessed November 2022).Google ScholarThere is no corresponding record for this reference.
- 63Butina, D. Unsupervised data base clustering based on Daylight’s fingerprint and Tanimoto similarity: a fast and automated way to cluster small and large data sets. JCICS 1999, 39, 747– 750, DOI: 10.1021/ci9803381
Supporting Information
Supporting Information
ARTICLE SECTIONSThe Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acs.jcim.3c00276.
Figures and tables referenced in the text (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.