1
|
Mammoli A, Bianconi E, Ruta L, Riccio A, Bigiotti C, Souma M, Carotti A, Rossini S, Suvieri C, Pallotta MT, Grohmann U, Camaioni E, Macchiarulo A. Critical Assessment of a Structure-Based Screening Campaign for IDO1 Inhibitors: Tips and Pitfalls. Int J Mol Sci 2022; 23:ijms23073981. [PMID: 35409342 PMCID: PMC8999677 DOI: 10.3390/ijms23073981] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 11/26/2022] Open
Abstract
Over the last two decades, indoleamine 2,3-dioxygenase 1 (IDO1) has attracted wide interest as a key player in immune regulation, fostering the design and development of small molecule inhibitors to restore immune response in tumor immunity. In this framework, biochemical, structural, and pharmacological studies have unveiled peculiar structural plasticity of IDO1, with different conformations and functional states that are coupled to fine regulation of its catalytic activity and non-enzymic functions. The large plasticity of IDO1 may affect its ligand recognition process, generating bias in structure-based drug design campaigns. In this work, we report a screening campaign of a fragment library of compounds, grounding on the use of three distinct conformations of IDO1 that recapitulate its structural plasticity to some extent. Results are instrumental to discuss tips and pitfalls that, due to the large plasticity of the enzyme, may influence the identification of novel and differentiated chemical scaffolds of IDO1 ligands in structure-based screening campaigns.
Collapse
Affiliation(s)
- Andrea Mammoli
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo n.1, 06123 Perugia, Italy; (A.M.); (E.B.); (L.R.); (A.R.); (C.B.); (M.S.); (A.C.); (E.C.)
| | - Elisa Bianconi
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo n.1, 06123 Perugia, Italy; (A.M.); (E.B.); (L.R.); (A.R.); (C.B.); (M.S.); (A.C.); (E.C.)
| | - Luana Ruta
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo n.1, 06123 Perugia, Italy; (A.M.); (E.B.); (L.R.); (A.R.); (C.B.); (M.S.); (A.C.); (E.C.)
| | - Alessandra Riccio
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo n.1, 06123 Perugia, Italy; (A.M.); (E.B.); (L.R.); (A.R.); (C.B.); (M.S.); (A.C.); (E.C.)
| | - Carlo Bigiotti
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo n.1, 06123 Perugia, Italy; (A.M.); (E.B.); (L.R.); (A.R.); (C.B.); (M.S.); (A.C.); (E.C.)
| | - Maria Souma
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo n.1, 06123 Perugia, Italy; (A.M.); (E.B.); (L.R.); (A.R.); (C.B.); (M.S.); (A.C.); (E.C.)
| | - Andrea Carotti
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo n.1, 06123 Perugia, Italy; (A.M.); (E.B.); (L.R.); (A.R.); (C.B.); (M.S.); (A.C.); (E.C.)
| | - Sofia Rossini
- Department of Medicine and Surgery, University of Perugia, P.le Gambuli, 06132 Perugia, Italy; (S.R.); (C.S.); (M.T.P.); (U.G.)
| | - Chiara Suvieri
- Department of Medicine and Surgery, University of Perugia, P.le Gambuli, 06132 Perugia, Italy; (S.R.); (C.S.); (M.T.P.); (U.G.)
| | - Maria Teresa Pallotta
- Department of Medicine and Surgery, University of Perugia, P.le Gambuli, 06132 Perugia, Italy; (S.R.); (C.S.); (M.T.P.); (U.G.)
| | - Ursula Grohmann
- Department of Medicine and Surgery, University of Perugia, P.le Gambuli, 06132 Perugia, Italy; (S.R.); (C.S.); (M.T.P.); (U.G.)
| | - Emidio Camaioni
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo n.1, 06123 Perugia, Italy; (A.M.); (E.B.); (L.R.); (A.R.); (C.B.); (M.S.); (A.C.); (E.C.)
| | - Antonio Macchiarulo
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo n.1, 06123 Perugia, Italy; (A.M.); (E.B.); (L.R.); (A.R.); (C.B.); (M.S.); (A.C.); (E.C.)
- Correspondence: ; Tel.: +39-(075)-585-5131
| |
Collapse
|
2
|
Khan MKA, Akhtar S. Novel drug design and bioinformatics: an introduction. PHYSICAL SCIENCES REVIEWS 2021. [DOI: 10.1515/psr-2018-0158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Abstract
In the current era of high-throughput technology, where enormous amounts of biological data are generated day by day via various sequencing projects, thereby the staggering volume of biological targets deciphered. The discovery of new chemical entities and bioisosteres of relatively low molecular weight has been gaining high momentum in the pharmacopoeia, and traditional combinatorial design wherein chemical structure is used as an initial template for enhancing efficacy pharmacokinetic selectivity properties. Once the compound is identified, it undergoes ADMET filtration to ensure whether it has toxic and mutagenic properties or not. If the compound has no toxicity and mutagenicity is either considered a potential lead molecule. Understanding the mechanism of lead molecules with various biological targets is imperative to advance related functions for drug discovery and development. Notwithstanding, a tedious and costly process, taking around 10–15 years and costing around $4 billion, cascaded approached of Bioinformatics and Computational biology viz., structure-based drug design (SBDD) and cognate ligand-based drug design (LBDD) respectively rely on the availability of 3D structure of target biomacromolecules and vice versa has made this process easy and approachable. SBDD encompasses homology modelling, ligand docking, fragment-based drug design and molecular dynamics, while LBDD deals with pharmacophore mapping, QSAR, and similarity search. All the computational methods discussed herein, whether for target identification or novel ligand discovery, continuously evolve and facilitate cost-effective and reliable outcomes in an era of overwhelming data.
Collapse
Affiliation(s)
- Mohammad Kalim Ahmad Khan
- Department of Bioengineering, Faculty of Engineering , Integral University , Lucknow , Uttar Pradesh , 226026 , India
| | - Salman Akhtar
- Department of Bioengineering, Faculty of Engineering , Integral University , Lucknow , Uttar Pradesh , 226026 , India
| |
Collapse
|
3
|
Wilson DM, Deacon AM, Duncton MAJ, Pellicena P, Georgiadis MM, Yeh AP, Arvai AS, Moiani D, Tainer JA, Das D. Fragment- and structure-based drug discovery for developing therapeutic agents targeting the DNA Damage Response. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2021; 163:130-142. [PMID: 33115610 PMCID: PMC8666131 DOI: 10.1016/j.pbiomolbio.2020.10.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 10/13/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022]
Abstract
Cancer will directly affect the lives of over one-third of the population. The DNA Damage Response (DDR) is an intricate system involving damage recognition, cell cycle regulation, DNA repair, and ultimately cell fate determination, playing a central role in cancer etiology and therapy. Two primary therapeutic approaches involving DDR targeting include: combinatorial treatments employing anticancer genotoxic agents; and synthetic lethality, exploiting a sporadic DDR defect as a mechanism for cancer-specific therapy. Whereas, many DDR proteins have proven "undruggable", Fragment- and Structure-Based Drug Discovery (FBDD, SBDD) have advanced therapeutic agent identification and development. FBDD has led to 4 (with ∼50 more drugs under preclinical and clinical development), while SBDD is estimated to have contributed to the development of >200, FDA-approved medicines. Protein X-ray crystallography-based fragment library screening, especially for elusive or "undruggable" targets, allows for simultaneous generation of hits plus details of protein-ligand interactions and binding sites (orthosteric or allosteric) that inform chemical tractability, downstream biology, and intellectual property. Using a novel high-throughput crystallography-based fragment library screening platform, we screened five diverse proteins, yielding hit rates of ∼2-8% and crystal structures from ∼1.8 to 3.2 Å. We consider current FBDD/SBDD methods and some exemplary results of efforts to design inhibitors against the DDR nucleases meiotic recombination 11 (MRE11, a.k.a., MRE11A), apurinic/apyrimidinic endonuclease 1 (APE1, a.k.a., APEX1), and flap endonuclease 1 (FEN1).
Collapse
Affiliation(s)
- David M Wilson
- Hasselt University, Biomedical Research Institute, Diepenbeek, Belgium; Boost Scientific, Heusden-Zolder, Belgium; XPose Therapeutics Inc., San Carlos, CA, USA
| | - Ashley M Deacon
- Accelero Biostructures Inc., San Francisco, CA, USA; XPose Therapeutics Inc., San Carlos, CA, USA
| | | | | | - Millie M Georgiadis
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA; XPose Therapeutics Inc., San Carlos, CA, USA
| | - Andrew P Yeh
- Accelero Biostructures Inc., San Francisco, CA, USA
| | - Andrew S Arvai
- Integrative Structural and Computational Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - Davide Moiani
- Department of Cancer Biology, MD Anderson Cancer Center, Houston, TX, USA; Department of Molecular and Cellular Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - John A Tainer
- Department of Cancer Biology, MD Anderson Cancer Center, Houston, TX, USA; Department of Molecular and Cellular Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Debanu Das
- Accelero Biostructures Inc., San Francisco, CA, USA; XPose Therapeutics Inc., San Carlos, CA, USA.
| |
Collapse
|
4
|
De Fusco C, Schimpl M, Börjesson U, Cheung T, Collie I, Evans L, Narasimhan P, Stubbs C, Vazquez-Chantada M, Wagner DJ, Grondine M, Sanders MG, Tentarelli S, Underwood E, Argyrou A, Smith JM, Lynch JT, Chiarparin E, Robb G, Bagal SK, Scott JS. Fragment-Based Design of a Potent MAT2a Inhibitor and in Vivo Evaluation in an MTAP Null Xenograft Model. J Med Chem 2021; 64:6814-6826. [PMID: 33900758 DOI: 10.1021/acs.jmedchem.1c00067] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
MAT2a is a methionine adenosyltransferase that synthesizes the essential metabolite S-adenosylmethionine (SAM) from methionine and ATP. Tumors bearing the co-deletion of p16 and MTAP genes have been shown to be sensitive to MAT2a inhibition, making it an attractive target for treatment of MTAP-deleted cancers. A fragment-based lead generation campaign identified weak but efficient hits binding in a known allosteric site. By use of structure-guided design and systematic SAR exploration, the hits were elaborated through a merging and growing strategy into an arylquinazolinone series of potent MAT2a inhibitors. The selected in vivo tool compound 28 reduced SAM-dependent methylation events in cells and inhibited proliferation of MTAP-null cells in vitro. In vivo studies showed that 28 was able to induce antitumor response in an MTAP knockout HCT116 xenograft model.
Collapse
Affiliation(s)
- Claudia De Fusco
- Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 0WG, United Kingdom
| | - Marianne Schimpl
- Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 0WG, United Kingdom
| | - Ulf Börjesson
- Discovery Sciences, R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Tony Cheung
- Oncology R&D, AstraZeneca, Boston, Massachusetts 02451, United States
| | - Iain Collie
- Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 0WG, United Kingdom
| | - Laura Evans
- Oncology R&D, AstraZeneca, Cambridge CB4 0WG, United Kingdom
| | | | | | | | - David J Wagner
- Oncology R&D, AstraZeneca, Boston, Massachusetts 02451, United States
| | - Michael Grondine
- Oncology R&D, AstraZeneca, Boston, Massachusetts 02451, United States
| | | | - Sharon Tentarelli
- Oncology R&D, AstraZeneca, Boston, Massachusetts 02451, United States
| | | | - Argyrides Argyrou
- Discovery Sciences, R&D, AstraZeneca, Cambridge CB4 0WG, United Kingdom
| | - James M Smith
- Oncology R&D, AstraZeneca, Cambridge CB4 0WG, United Kingdom
| | - James T Lynch
- Oncology R&D, AstraZeneca, Cambridge CB4 0WG, United Kingdom
| | | | - Graeme Robb
- Oncology R&D, AstraZeneca, Cambridge CB4 0WG, United Kingdom
| | - Sharan K Bagal
- Oncology R&D, AstraZeneca, Cambridge CB4 0WG, United Kingdom
| | - James S Scott
- Oncology R&D, AstraZeneca, Cambridge CB4 0WG, United Kingdom
| |
Collapse
|
5
|
Nuñez JR, Mcgrady M, Yesiltepe Y, Renslow RS, Metz TO. Chespa: Streamlining Expansive Chemical Space Evaluation of Molecular Sets. J Chem Inf Model 2020; 60:6251-6257. [PMID: 33283505 DOI: 10.1021/acs.jcim.0c00899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Thousands of chemical properties can be calculated for small molecules, which can be used to place the molecules within the context of a broader "chemical space." These definitions vary based on compounds of interest and the goals for the given chemical space definition. Here, we introduce a customizable Python module, chespa, built to easily assess different chemical space definitions through clustering of compounds in these spaces and visualizing trends of these clusters. To demonstrate this, chespa currently streamlines prediction of various molecular descriptors (predicted chemical properties, molecular substructures, AI-based chemical space, and chemical class ontology) in order to test six different chemical space definitions. Furthermore, we investigated how these varying definitions trend with mass spectrometry (MS)-based observability, that is, the ability of a molecule to be observed with MS (e.g., as a function of the molecule ionizability), using an example data set from the U.S. EPA's nontargeted analysis collaborative trial, where blinded samples had been analyzed previously, providing 1398 data points. Improved understanding of observability would offer many advantages in small-molecule identification, such as (i) a priori selection of experimental conditions based on suspected sample composition, (ii) the ability to reduce the number of candidate structures during compound identification by removing those less likely to ionize, and, in turn, (iii) a reduced false discovery rate and increased confidence in identifications. Factors controlling observability are not fully understood, making prediction of this property nontrivial and a prime candidate for chemical space analysis. Chespa is available at github.com/pnnl/chespa.
Collapse
Affiliation(s)
- Jamie R Nuñez
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, United States.,The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, Washington 99164, United States
| | - Monee Mcgrady
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| | - Yasemin Yesiltepe
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, United States.,The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, Washington 99164, United States
| | - Ryan S Renslow
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, United States.,The Gene and Linda Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, Washington 99164, United States
| | - Thomas O Metz
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington 99352, United States
| |
Collapse
|
6
|
Grygorenko OO, Volochnyuk DM, Ryabukhin SV, Judd DB. The Symbiotic Relationship Between Drug Discovery and Organic Chemistry. Chemistry 2019; 26:1196-1237. [PMID: 31429510 DOI: 10.1002/chem.201903232] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Revised: 08/19/2019] [Indexed: 12/20/2022]
Abstract
All pharmaceutical products contain organic molecules; the source may be a natural product or a fully synthetic molecule, or a combination of both. Thus, it follows that organic chemistry underpins both existing and upcoming pharmaceutical products. The reverse relationship has also affected organic synthesis, changing its landscape towards increasingly complex targets. This Review article sets out to give a concise appraisal of this symbiotic relationship between organic chemistry and drug discovery, along with a discussion of the design concepts and highlighting key milestones along the journey. In particular, criteria for a high-quality compound library design enabling efficient virtual navigation of chemical space, as well as rise and fall of concepts for its synthetic exploration (such as combinatorial chemistry; diversity-, biology-, lead-, or fragment-oriented syntheses; and DNA-encoded libraries) are critically surveyed.
Collapse
Affiliation(s)
- Oleksandr O Grygorenko
- Enamine Ltd., Chervonotkatska Street 78, Kiev, 02094, Ukraine.,Taras Shevchenko National University of Kiev, Volodymyrska Street 60, Kiev, 01601, Ukraine
| | - Dmitriy M Volochnyuk
- Enamine Ltd., Chervonotkatska Street 78, Kiev, 02094, Ukraine.,Taras Shevchenko National University of Kiev, Volodymyrska Street 60, Kiev, 01601, Ukraine.,Institute of Organic Chemistry, National Academy of Sciences of Ukraine, Murmanska Street 5, Kiev, 02660, Ukraine
| | - Sergey V Ryabukhin
- Enamine Ltd., Chervonotkatska Street 78, Kiev, 02094, Ukraine.,Taras Shevchenko National University of Kiev, Volodymyrska Street 60, Kiev, 01601, Ukraine
| | - Duncan B Judd
- Awridian Ltd., Stevenage Bioscience Catalyst, Gunnelswood Road, Stevenage, Herts, SG1 2FX, UK
| |
Collapse
|
7
|
Polshakov VI, Batuev EA, Mantsyzov AB. NMR screening and studies of target–ligand interactions. RUSSIAN CHEMICAL REVIEWS 2019. [DOI: 10.1070/rcr4836] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
8
|
Borysko P, Moroz YS, Vasylchenko OV, Hurmach VV, Starodubtseva A, Stefanishena N, Nesteruk K, Zozulya S, Kondratov IS, Grygorenko OO. Straightforward hit identification approach in fragment-based discovery of bromodomain-containing protein 4 (BRD4) inhibitors. Bioorg Med Chem 2018; 26:3399-3405. [DOI: 10.1016/j.bmc.2018.05.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 04/24/2018] [Accepted: 05/08/2018] [Indexed: 02/07/2023]
|
9
|
Abstract
Natural products (NPs) have been used as traditional medicines since antiquity. With more than 1060 estimated compounds with molecular weights less than 500 Da representing chemical space, NPs occupy a very small percentage; however, they are significantly overrepresented in biologically relevant chemical space. The classical approach concentrates on identifying one or more NPs with biological activity from a source organism. There is much more to be learned from NPs than we can discover this narrow view. In this review, we discuss ways to harness the global properties of NPs.
Collapse
Affiliation(s)
- Asmaa Boufridi
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, Queensland 4111, Australia; ,
| | - Ronald J Quinn
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, Queensland 4111, Australia; ,
| |
Collapse
|
10
|
Grove LE, Vajda S, Kozakov D. Computational Methods to Support Fragment-based Drug Discovery. FRAGMENT-BASED DRUG DISCOVERY LESSONS AND OUTLOOK 2016. [DOI: 10.1002/9783527683604.ch09] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
|
11
|
Davies TG, Jhoti H, Pathuri P, Williams G. Selecting the Right Targets for Fragment-Based Drug Discovery. FRAGMENT-BASED DRUG DISCOVERY LESSONS AND OUTLOOK 2016. [DOI: 10.1002/9783527683604.ch02] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
12
|
Wasko MJ, Pellegrene KA, Madura JD, Surratt CK. A Role for Fragment-Based Drug Design in Developing Novel Lead Compounds for Central Nervous System Targets. Front Neurol 2015; 6:197. [PMID: 26441817 PMCID: PMC4566055 DOI: 10.3389/fneur.2015.00197] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Accepted: 08/24/2015] [Indexed: 01/12/2023] Open
Abstract
Hundreds of millions of U.S. dollars are invested in the research and development of a single drug. Lead compound development is an area ripe for new design strategies. Therapeutic lead candidates have been traditionally found using high-throughput in vitro pharmacological screening, a costly method for assaying thousands of compounds. This approach has recently been augmented by virtual screening (VS), which employs computer models of the target protein to narrow the search for possible leads. A variant of VS is fragment-based drug design (FBDD), an emerging in silico lead discovery method that introduces low-molecular weight fragments, rather than intact compounds, into the binding pocket of the receptor model. These fragments serve as starting points for “growing” the lead candidate. Current efforts in virtual FBDD within central nervous system (CNS) targets are reviewed, as is a recent rule-based optimization strategy in which new molecules are generated within a 3D receptor-binding pocket using the fragment as a scaffold. This process not only places special emphasis on creating synthesizable molecules but also exposes computational questions worth addressing. Fragment-based methods provide a viable, relatively low-cost alternative for therapeutic lead discovery and optimization that can be applied to CNS targets to augment current design strategies.
Collapse
Affiliation(s)
- Michael J Wasko
- Mylan School of Pharmacy, Graduate School of Pharmaceutical Sciences, Duquesne University , Pittsburgh, PA , USA
| | - Kendy A Pellegrene
- Mylan School of Pharmacy, Graduate School of Pharmaceutical Sciences, Duquesne University , Pittsburgh, PA , USA
| | - Jeffry D Madura
- Department of Chemistry and Biochemistry, Center for Computational Sciences, Bayer School of Natural and Environmental Sciences, Duquesne University , Pittsburgh, PA , USA
| | - Christopher K Surratt
- Mylan School of Pharmacy, Graduate School of Pharmaceutical Sciences, Duquesne University , Pittsburgh, PA , USA
| |
Collapse
|
13
|
Teplitsky E, Joshi K, Ericson DL, Scalia A, Mullen JD, Sweet RM, Soares AS. High throughput screening using acoustic droplet ejection to combine protein crystals and chemical libraries on crystallization plates at high density. J Struct Biol 2015; 191:49-58. [PMID: 26027487 DOI: 10.1016/j.jsb.2015.05.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 05/21/2015] [Accepted: 05/27/2015] [Indexed: 11/30/2022]
Abstract
We describe a high throughput method for screening up to 1728 distinct chemicals with protein crystals on a single microplate. Acoustic droplet ejection (ADE) was used to co-position 2.5nL of protein, precipitant, and chemicals on a MiTeGen in situ-1 crystallization plate™ for screening by co-crystallization or soaking. ADE-transferred droplets follow a precise trajectory which allows all components to be transferred through small apertures in the microplate lid. The apertures were large enough for 2.5nL droplets to pass through them, but small enough so that they did not disrupt the internal environment created by the mother liquor. Using this system, thermolysin and trypsin crystals were efficiently screened for binding to a heavy-metal mini-library. Fluorescence and X-ray diffraction were used to confirm that each chemical in the heavy-metal library was correctly paired with the intended protein crystal. A fragment mini-library was screened to observe two known lysozyme ligands using both co-crystallization and soaking. A similar approach was used to identify multiple, novel thaumatin binding sites for ascorbic acid. This technology pushes towards a faster, automated, and more flexible strategy for high throughput screening of chemical libraries (such as fragment libraries) using as little as 2.5nL of each component.
Collapse
Affiliation(s)
- Ella Teplitsky
- Office of Educational Programs, Brookhaven National Laboratory, Upton, NY 11973-5000, USA; Department of Biochemistry and Cell Biology, Stony Brook University, NY 11794-5215, USA
| | - Karan Joshi
- Office of Educational Programs, Brookhaven National Laboratory, Upton, NY 11973-5000, USA; Department of Electronics and Electrical Communication Engineering, PEC University of Technology, Chandigarh, India
| | - Daniel L Ericson
- Office of Educational Programs, Brookhaven National Laboratory, Upton, NY 11973-5000, USA; Department of Biomedical Engineering, University at Buffalo, SUNY, 12 Capen Hall, Buffalo, NY 14260, USA
| | - Alexander Scalia
- Office of Educational Programs, Brookhaven National Laboratory, Upton, NY 11973-5000, USA; Department of Biological Sciences, 4400 Vestal Parkway East, Binghamton University, NY 13902, USA
| | - Jeffrey D Mullen
- Office of Educational Programs, Brookhaven National Laboratory, Upton, NY 11973-5000, USA; Physics Department, University of Oregon, Eugene, OR 97403-1274, USA
| | - Robert M Sweet
- Photon Sciences Directorate, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
| | - Alexei S Soares
- Photon Sciences Directorate, Brookhaven National Laboratory, Upton, NY 11973-5000, USA.
| |
Collapse
|
14
|
Istyastono EP, Kooistra AJ, Vischer HF, Kuijer M, Roumen L, Nijmeijer S, Smits RA, de Esch IJP, Leurs R, de Graaf C. Structure-based virtual screening for fragment-like ligands of the G protein-coupled histamine H4 receptor. MEDCHEMCOMM 2015. [DOI: 10.1039/c5md00022j] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Structure-based virtual screening using H1R- and β2R-based histamine H4R homology models identified 9 fragments with an affinity ranging from 0.14 to 6.3 μm for H4R.
Collapse
Affiliation(s)
- Enade P. Istyastono
- Division of Medicinal Chemistry
- Amsterdam Institute for Molecules, Medicines and Systems (AIMMS)
- Faculty of Exact Sciences
- VU University Amsterdam
- 1081 HV Amsterdam
| | - Albert J. Kooistra
- Division of Medicinal Chemistry
- Amsterdam Institute for Molecules, Medicines and Systems (AIMMS)
- Faculty of Exact Sciences
- VU University Amsterdam
- 1081 HV Amsterdam
| | - Henry F. Vischer
- Division of Medicinal Chemistry
- Amsterdam Institute for Molecules, Medicines and Systems (AIMMS)
- Faculty of Exact Sciences
- VU University Amsterdam
- 1081 HV Amsterdam
| | - Martien Kuijer
- Division of Medicinal Chemistry
- Amsterdam Institute for Molecules, Medicines and Systems (AIMMS)
- Faculty of Exact Sciences
- VU University Amsterdam
- 1081 HV Amsterdam
| | - Luc Roumen
- Division of Medicinal Chemistry
- Amsterdam Institute for Molecules, Medicines and Systems (AIMMS)
- Faculty of Exact Sciences
- VU University Amsterdam
- 1081 HV Amsterdam
| | - Saskia Nijmeijer
- Division of Medicinal Chemistry
- Amsterdam Institute for Molecules, Medicines and Systems (AIMMS)
- Faculty of Exact Sciences
- VU University Amsterdam
- 1081 HV Amsterdam
| | | | - Iwan J. P. de Esch
- Division of Medicinal Chemistry
- Amsterdam Institute for Molecules, Medicines and Systems (AIMMS)
- Faculty of Exact Sciences
- VU University Amsterdam
- 1081 HV Amsterdam
| | - Rob Leurs
- Division of Medicinal Chemistry
- Amsterdam Institute for Molecules, Medicines and Systems (AIMMS)
- Faculty of Exact Sciences
- VU University Amsterdam
- 1081 HV Amsterdam
| | - Chris de Graaf
- Division of Medicinal Chemistry
- Amsterdam Institute for Molecules, Medicines and Systems (AIMMS)
- Faculty of Exact Sciences
- VU University Amsterdam
- 1081 HV Amsterdam
| |
Collapse
|
15
|
|
16
|
Nirantar SR, Li X, Siau JW, Ghadessy FJ. Rapid screening of protein–protein interaction inhibitors using the protease exclusion assay. Biosens Bioelectron 2014; 56:250-7. [DOI: 10.1016/j.bios.2013.12.060] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 12/13/2013] [Accepted: 12/26/2013] [Indexed: 10/25/2022]
|
17
|
Yin X, Scalia A, Leroy L, Cuttitta CM, Polizzo GM, Ericson DL, Roessler CG, Campos O, Ma MY, Agarwal R, Jackimowicz R, Allaire M, Orville AM, Sweet RM, Soares AS. Hitting the target: fragment screening with acoustic in situ co-crystallization of proteins plus fragment libraries on pin-mounted data-collection micromeshes. ACTA CRYSTALLOGRAPHICA. SECTION D, BIOLOGICAL CRYSTALLOGRAPHY 2014; 70:1177-89. [PMID: 24816088 PMCID: PMC4014116 DOI: 10.1107/s1399004713034603] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Accepted: 12/24/2013] [Indexed: 11/17/2022]
Abstract
Acoustic droplet ejection (ADE) is a powerful technology that supports crystallographic applications such as growing, improving and manipulating protein crystals. A fragment-screening strategy is described that uses ADE to co-crystallize proteins with fragment libraries directly on MiTeGen MicroMeshes. Co-crystallization trials can be prepared rapidly and economically. The high speed of specimen preparation and the low consumption of fragment and protein allow the use of individual rather than pooled fragments. The Echo 550 liquid-handling instrument (Labcyte Inc., Sunnyvale, California, USA) generates droplets with accurate trajectories, which allows multiple co-crystallization experiments to be discretely positioned on a single data-collection micromesh. This accuracy also allows all components to be transferred through small apertures. Consequently, the crystallization tray is in equilibrium with the reservoir before, during and after the transfer of protein, precipitant and fragment to the micromesh on which crystallization will occur. This strict control of the specimen environment means that the crystallography experiments remain identical as the working volumes are decreased from the few microlitres level to the few nanolitres level. Using this system, lysozyme, thermolysin, trypsin and stachydrine demethylase crystals were co-crystallized with a small 33-compound mini-library to search for fragment hits. This technology pushes towards a much faster, more automated and more flexible strategy for structure-based drug discovery using as little as 2.5 nl of each major component.
Collapse
Affiliation(s)
- Xingyu Yin
- Office of Educational Programs, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
- Department of Biochemistry and Cell Biology, Stony Brook University, NY 11794-5215, USA
- Nanjing University, Nanjing, Jiangsu, People’s Republic of China
| | - Alexander Scalia
- Office of Educational Programs, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
- Department of Biological Sciences, Binghamton University, 4400 Vestal Parkway East, NY 13902, USA
| | - Ludmila Leroy
- Office of Educational Programs, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
- CAPES Foundation, Ministry of Education of Brazil, 70040-020 Brasilia-DF, Brazil
- Universidade Federal de Minas Gerais, 6627 Av. Antonio Carlos, 31270-901 Belo Horizonte-MG, Brazil
| | - Christina M. Cuttitta
- Office of Educational Programs, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
- Center for Developmental Neuroscience and Department of Biology, College of Staten Island, The City University of New York, 2800 Victory Boulevard, Staten Island, NY 10314, USA
| | - Gina M. Polizzo
- Office of Educational Programs, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
- St Joseph’s College, 155 West Roe Boulevard, East Patchogue, NY 11772, USA
| | - Daniel L. Ericson
- Office of Educational Programs, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
- Department of Biomedical Engineering, University at Buffalo, SUNY, 12 Capen Hall, Buffalo, NY 14260, USA
| | - Christian G. Roessler
- Photon Sciences Directorate, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
| | - Olven Campos
- Office of Educational Programs, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
- Department of Biological Science, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33414, USA
| | - Millie Y. Ma
- Office of Educational Programs, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
- Comsewogue High School, 565 Bicycle Path, Port Jefferson Station, NY 11776, USA
| | - Rakhi Agarwal
- Biosciences Department, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
| | - Rick Jackimowicz
- Photon Sciences Directorate, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
| | - Marc Allaire
- Photon Sciences Directorate, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
| | - Allen M. Orville
- Photon Sciences Directorate, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
- Biosciences Department, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
| | - Robert M. Sweet
- Photon Sciences Directorate, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
| | - Alexei S. Soares
- Photon Sciences Directorate, Brookhaven National Laboratory, Upton, NY 11973-5000, USA
| |
Collapse
|
18
|
Stornaiuolo M, De Kloe GE, Rucktooa P, Fish A, van Elk R, Edink ES, Bertrand D, Smit AB, de Esch IJP, Sixma TK. Assembly of a π-π stack of ligands in the binding site of an acetylcholine-binding protein. Nat Commun 2013; 4:1875. [PMID: 23695669 PMCID: PMC3674282 DOI: 10.1038/ncomms2900] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Accepted: 04/17/2013] [Indexed: 12/11/2022] Open
Abstract
Acetylcholine-binding protein is a water-soluble homologue of the extracellular ligand-binding domain of cys-loop receptors. It is used as a structurally accessible prototype for studying ligand binding to these pharmaceutically important pentameric ion channels, in particular to nicotinic acetylcholine receptors, due to conserved binding site residues present at the interface between two subunits. Here we report that an aromatic conjugated small molecule binds acetylcholine-binding protein in an ordered π-π stack of three identical molecules per binding site, two parallel and one antiparallel. Acetylcholine-binding protein stabilizes the assembly of the stack by aromatic contacts. Thanks to the plasticity of its ligand-binding site, acetylcholine-binding protein can accommodate the formation of aromatic stacks of different size by simple loop repositioning and minimal adjustment of the interactions. This type of supramolecular binding provides a novel paradigm in drug design.
Collapse
Affiliation(s)
- Mariano Stornaiuolo
- Division of Biochemistry and Center for Biomedical Genetics, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Doak BC, Morton CJ, Simpson JS, Scanlon MJ. Design and Evaluation of the Performance of an NMR Screening Fragment Library. Aust J Chem 2013. [DOI: 10.1071/ch13280] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The design of a suitable library is an essential prerequisite to establish a fragment-based screening capability. Several pharmaceutical companies have described their approaches to establishing fragment libraries; however there are few detailed reports of both design and analysis of performance for a fragment library maintained in an academic setting. Here we report our efforts towards the design of a fragment library for nuclear magnetic resonance spectroscopy-based screening, demonstrate the performance of the library through analysis of 14 screens, and present a comparison to previously reported fragment libraries.
Collapse
|
20
|
Francis CL, Kenny PW, Dolezal O, Saubern S, Kruger M, Savage GP, Peat TS, Ryan JH. Construction of the CSIRO Fragment Library. Aust J Chem 2013. [DOI: 10.1071/ch13325] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
A fundamental component of a successful fragment screening program is a productive fragment library, one that delivers hit fragments with potential for pharmaceutical development. A proprietary fragment library was developed by identifying and extracting subsets of CSIRO’s Compound Collection using two complimentary approaches. Over time, the use of surface plasmon resonance as a front-line screening tool has enabled identification and exclusion of problematic compounds and led to a more reliable fragment screening library.
Collapse
|
21
|
de Graaf C, Vischer HF, de Kloe GE, Kooistra AJ, Nijmeijer S, Kuijer M, Verheij MHP, England PJ, van Muijlwijk-Koezen JE, Leurs R, de Esch IJP. Small and colorful stones make beautiful mosaics: fragment-based chemogenomics. Drug Discov Today 2012; 18:323-30. [PMID: 23266367 DOI: 10.1016/j.drudis.2012.12.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2012] [Revised: 11/19/2012] [Accepted: 12/05/2012] [Indexed: 12/01/2022]
Abstract
Smaller stones with a wide variety of colors make a higher resolution mosaic. In much the same way, smaller chemical entities that are structurally diverse are better able to interrogate protein binding sites. This feature article describes the construction of a diverse fragment library and an analysis of the screening of six representative protein targets belonging to three diverse target classes (G protein-coupled receptors ADRB2, H1R, H3R, and H4R, the ligand-gated ion channel 5-HT3R, and the kinase PKA) using chemogenomics approaches. The integration of experimentally determined bioaffinity profiles across related and unrelated protein targets and chemogenomics analysis of fragment binding and protein structure allow the identification of: (i) unexpected similarities and differences in ligand binding properties, and (ii) subtle ligand affinity and selectivity cliffs. With a wealth of fragment screening data being generated in industry and academia, such approaches will contribute to a more detailed structural understanding of ligand-protein interactions.
Collapse
Affiliation(s)
- Chris de Graaf
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Nakagawa S, Bainbridge KA, Butcher K, Ellis D, Klute W, Ryckmans T. Application of Barluenga Boronic Coupling (BBC) to the Parallel Synthesis of Drug-like and Drug Fragment-like Molecules. ChemMedChem 2011; 7:233-6. [DOI: 10.1002/cmdc.201100339] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Revised: 10/20/2011] [Indexed: 11/07/2022]
|
23
|
Bottegoni G, Favia AD, Recanatini M, Cavalli A. The role of fragment-based and computational methods in polypharmacology. Drug Discov Today 2011; 17:23-34. [PMID: 21864710 DOI: 10.1016/j.drudis.2011.08.002] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Revised: 06/21/2011] [Accepted: 08/09/2011] [Indexed: 02/07/2023]
Abstract
Polypharmacology-based strategies are gaining increased attention as a novel approach to obtaining potentially innovative medicines for multifactorial diseases. However, some within the pharmaceutical community have resisted these strategies because they can be resource-hungry in the early stages of the drug discovery process. Here, we report on fragment-based and computational methods that might accelerate and optimize the discovery of multitarget drugs. In particular, we illustrate that fragment-based approaches can be particularly suited for polypharmacology, owing to the inherent promiscuous nature of fragments. In parallel, we explain how computer-assisted protocols can provide invaluable insights into how to unveil compounds theoretically able to bind to more than one protein. Furthermore, several pragmatic aspects related to the use of these approaches are covered, thus offering the reader practical insights on multitarget-oriented drug discovery projects.
Collapse
Affiliation(s)
- Giovanni Bottegoni
- Department of Drug Discovery and Development (D3), Istituto Italiano di Tecnologia, I-16163 Genoa, Italy
| | | | | | | |
Collapse
|
24
|
Blum LC, van Deursen R, Reymond JL. Visualisation and subsets of the chemical universe database GDB-13 for virtual screening. J Comput Aided Mol Des 2011; 25:637-47. [DOI: 10.1007/s10822-011-9436-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 05/13/2011] [Indexed: 10/18/2022]
|
25
|
Visualisation of the chemical space of fragments, lead-like and drug-like molecules in PubChem. J Comput Aided Mol Des 2011; 25:649-62. [DOI: 10.1007/s10822-011-9437-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2011] [Accepted: 05/13/2011] [Indexed: 10/18/2022]
|
26
|
Combining NMR and X-ray Crystallography in Fragment-Based Drug Discovery: Discovery of Highly Potent and Selective BACE-1 Inhibitors. Top Curr Chem (Cham) 2011; 317:83-114. [DOI: 10.1007/128_2011_183] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|