201
|
Gao K, Oerlemans R, Groves MR. Theory and applications of differential scanning fluorimetry in early-stage drug discovery. Biophys Rev 2020; 12:85-104. [PMID: 32006251 PMCID: PMC7040159 DOI: 10.1007/s12551-020-00619-2] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 01/08/2020] [Indexed: 02/06/2023] Open
Abstract
Differential scanning fluorimetry (DSF) is an accessible, rapid, and economical biophysical technique that has seen many applications over the years, ranging from protein folding state detection to the identification of ligands that bind to the target protein. In this review, we discuss the theory, applications, and limitations of DSF, including the latest applications of DSF by ourselves and other researchers. We show that DSF is a powerful high-throughput tool in early drug discovery efforts. We place DSF in the context of other biophysical methods frequently used in drug discovery and highlight their benefits and downsides. We illustrate the uses of DSF in protein buffer optimization for stability, refolding, and crystallization purposes and provide several examples of each. We also show the use of DSF in a more downstream application, where it is used as an in vivo validation tool of ligand-target interaction in cell assays. Although DSF is a potent tool in buffer optimization and large chemical library screens when it comes to ligand-binding validation and optimization, orthogonal techniques are recommended as DSF is prone to false positives and negatives.
Collapse
Affiliation(s)
- Kai Gao
- Structure Biology in Drug Design, Drug Design Group XB20, Departments of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Rick Oerlemans
- Structure Biology in Drug Design, Drug Design Group XB20, Departments of Pharmacy, University of Groningen, Groningen, The Netherlands
| | - Matthew R Groves
- Structure Biology in Drug Design, Drug Design Group XB20, Departments of Pharmacy, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
202
|
Verma AK, Fatima K, Dudi RK, Tabassum M, Iqbal H, Kumar Y, Luqman S, Mondhe D, Chanda D, Khan F, Shanker K, Negi AS. Antiproliferative activity of diarylnaphthylpyrrolidine derivative via dual target inhibition. Eur J Med Chem 2020; 188:111986. [DOI: 10.1016/j.ejmech.2019.111986] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/05/2019] [Accepted: 12/17/2019] [Indexed: 11/27/2022]
|
203
|
Erlanson DA, de Esch IJP, Jahnke W, Johnson CN, Mortenson PN. Fragment-to-Lead Medicinal Chemistry Publications in 2018. J Med Chem 2020; 63:4430-4444. [PMID: 31913033 DOI: 10.1021/acs.jmedchem.9b01581] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
This Perspective, the fourth in an annual series, summarizes fragment-to-lead (F2L) success stories published during 2018. Topics such as target class, screening methods, physicochemical properties, and ligand efficiency are discussed for the 2018 examples as well as for the combined 111 F2L examples covering 2015-2018. While the overall properties of fragments and leads have remained constant, a number of new trends are noted, for example, broadening of target class coverage and application of FBDD to covalent inhibitors. Moreover, several studies make use of fragment hits that were previously described in the literature, illustrating that fragments are versatile starting points that can be optimized to structurally diverse leads. By focusing on success stories, the hope is that this Perspective will identify and inform best practices in fragment-based drug discovery.
Collapse
Affiliation(s)
- Daniel A Erlanson
- Frontier Medicines, 151 Oyster Point Boulevard, South San Francisco, California 94080, United States
| | - Iwan J P de Esch
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, The Netherlands
| | - Wolfgang Jahnke
- Novartis Institutes for Biomedical Research, Chemical Biology and Therapeutics, 4002 Basel, Switzerland
| | - Christopher N Johnson
- Astex Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge CB4 0QA, United Kingdom
| | - Paul N Mortenson
- Astex Pharmaceuticals, 436 Cambridge Science Park, Milton Road, Cambridge CB4 0QA, United Kingdom
| |
Collapse
|
204
|
Schiedel M, Moroglu M, Ascough DMH, Chamberlain AER, Kamps JJAG, Sekirnik AR, Conway SJ. Chemical Epigenetics: The Impact of Chemical and Chemical Biology Techniques on Bromodomain Target Validation. Angew Chem Int Ed Engl 2019; 58:17930-17952. [DOI: 10.1002/anie.201812164] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 01/08/2019] [Indexed: 12/22/2022]
Affiliation(s)
- Matthias Schiedel
- Department of ChemistryChemistry Research LaboratoryUniversity of Oxford Mansfield Road Oxford OX1 3TA UK
| | - Mustafa Moroglu
- Department of ChemistryChemistry Research LaboratoryUniversity of Oxford Mansfield Road Oxford OX1 3TA UK
| | - David M. H. Ascough
- Department of ChemistryChemistry Research LaboratoryUniversity of Oxford Mansfield Road Oxford OX1 3TA UK
| | - Anna E. R. Chamberlain
- Department of ChemistryChemistry Research LaboratoryUniversity of Oxford Mansfield Road Oxford OX1 3TA UK
| | - Jos J. A. G. Kamps
- Department of ChemistryChemistry Research LaboratoryUniversity of Oxford Mansfield Road Oxford OX1 3TA UK
| | - Angelina R. Sekirnik
- Department of ChemistryChemistry Research LaboratoryUniversity of Oxford Mansfield Road Oxford OX1 3TA UK
| | - Stuart J. Conway
- Department of ChemistryChemistry Research LaboratoryUniversity of Oxford Mansfield Road Oxford OX1 3TA UK
| |
Collapse
|
205
|
Herkt M, Batkai S, Thum T. Studying Interactions between 2'-O-Me-Modified Inhibitors and MicroRNAs Utilizing Microscale Thermophoresis. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:259-268. [PMID: 31581050 PMCID: PMC6796726 DOI: 10.1016/j.omtn.2019.08.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 08/14/2019] [Accepted: 08/14/2019] [Indexed: 11/29/2022]
Abstract
Besides the acquisition of pharmacokinetic parameters of antisense oligonucleotide microRNA (miRNA) inhibitors, such as measuring in vivo concentration, their pharmacodynamic characteristics are also of interest. An emerging and straightforward method for studying molecular interactions is microscale thermophoresis (MST). This technique makes it possible to study interactions between miRNAs and various oligonucleotide inhibitors, independent of the chemical modifications of the inhibitors or their respective target structure, with very little sample volume required compared to competitive techniques, such as surface plasmon resonance (SPR) and isothermal titration calorimetry (ITC). Interaction studies between these inhibitors and their respective target structures were performed, and they allowed the assessment of binding characteristics and parameters, such as EC50 for a number of these inhibitors, with little effort. Furthermore, MST could be utilized for obtaining kinetic binding data of the Argonaute-2 protein with a miRNA, which showed a possible RNA-induced silencing complex (RISC)-mediated turnover of inhibited miRNAs.
Collapse
Affiliation(s)
- Markus Herkt
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School (MHH), Hannover, Germany.
| | - Sandor Batkai
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School (MHH), Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School (MHH), Hannover, Germany.
| |
Collapse
|
206
|
Hu S, Zhao X, Zhang L. Computational Screening of Potential Inhibitors of β-N-Acetyl-D-Hesosaminidases Using Combined Core-Fragment Growth and Pharmacophore Restraints. Appl Biochem Biotechnol 2019; 189:1262-1273. [PMID: 31240546 DOI: 10.1007/s12010-019-03064-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 06/07/2019] [Indexed: 01/15/2023]
Abstract
As a type of β-N-acetyl-D-hexosaminidase enzyme purified from the Ostriniafurnacalis (Asian corn borer), OfHex1 has been previously reported to participate in chitin degradation, indicating that it may be an ideal target for designing new environmentally friendly pesticides. Besides, a natural product, TMG-chitotriomycin, has been found to be an effective inhibitor of OfHex1, and some studies have shown that the interactions between TMG unit and residues in - 1 subsite of OfHex1 are very conservative and important, inspiring us to design new inhibitors of β-N-acetyl-D-hexosaminidase with a new strategy. In the present study, the virtual screening of TMG unit as the core fragment was conducted using the combined computational methods, such as docking, molecular dynamics, pharmacophore model, and pesticide-likeness rule. Nine compounds with the binding free energy lower than TMG-β-(GlcNAc)2 were obtained. According to the decomposition energy and the interactions analysis, compounds 2, 3, 6 and 8, forming the hydrogen bonds with Val327 and Trp490, were considered as the possible lead structures for the further study. Our findings indicated that fragment-based lead discovery strategy might provide valuable insights into designing novel potential OfHex1 inhibitors.
Collapse
Affiliation(s)
- Song Hu
- Department of Applied Chemistry, College of Science, China Agricultural University, Beijing, 100193, China
| | - Xiao Zhao
- Department of Applied Chemistry, College of Science, China Agricultural University, Beijing, 100193, China
| | - Li Zhang
- Department of Applied Chemistry, College of Science, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
207
|
Kirsch P, Hartman AM, Hirsch AKH, Empting M. Concepts and Core Principles of Fragment-Based Drug Design. Molecules 2019; 24:molecules24234309. [PMID: 31779114 PMCID: PMC6930586 DOI: 10.3390/molecules24234309] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 11/11/2019] [Accepted: 11/20/2019] [Indexed: 02/06/2023] Open
Abstract
In this review, a general introduction to fragment-based drug design and the underlying concepts is given. General considerations and methodologies ranging from library selection/construction over biophysical screening and evaluation methods to in-depth hit qualification and subsequent optimization strategies are discussed. These principles can be generally applied to most classes of drug targets. The examples given for fragment growing, merging, and linking strategies at the end of the review are set in the fields of enzyme-inhibitor design and macromolecule–macromolecule interaction inhibition. Building upon the foundation of fragment-based drug discovery (FBDD) and its methodologies, we also highlight a few new trends in FBDD.
Collapse
Affiliation(s)
- Philine Kirsch
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research (HZI), Department of Drug Design and Optimization (DDOP), Campus E8.1, 66123 Saarbrücken, Germany; (P.K.); (A.M.H.); (A.K.H.H.)
- Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123 Saarbrücken, Germany
| | - Alwin M. Hartman
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research (HZI), Department of Drug Design and Optimization (DDOP), Campus E8.1, 66123 Saarbrücken, Germany; (P.K.); (A.M.H.); (A.K.H.H.)
- Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
- Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Anna K. H. Hirsch
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research (HZI), Department of Drug Design and Optimization (DDOP), Campus E8.1, 66123 Saarbrücken, Germany; (P.K.); (A.M.H.); (A.K.H.H.)
- Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
- Stratingh Institute for Chemistry, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Martin Empting
- Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS)-Helmholtz Centre for Infection Research (HZI), Department of Drug Design and Optimization (DDOP), Campus E8.1, 66123 Saarbrücken, Germany; (P.K.); (A.M.H.); (A.K.H.H.)
- Department of Pharmacy, Saarland University, Campus E8.1, 66123 Saarbrücken, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, 66123 Saarbrücken, Germany
- Correspondence: ; Tel.: +49-681-988-062-031
| |
Collapse
|
208
|
Ceballos J, Schwalfenberg M, Karageorgis G, Reckzeh ES, Sievers S, Ostermann C, Pahl A, Sellstedt M, Nowacki J, Carnero Corrales MA, Wilke J, Laraia L, Tschapalda K, Metz M, Sehr DA, Brand S, Winklhofer K, Janning P, Ziegler S, Waldmann H. Synthesis of Indomorphan Pseudo-Natural Product Inhibitors of Glucose Transporters GLUT-1 and -3. Angew Chem Int Ed Engl 2019; 58:17016-17025. [PMID: 31469221 PMCID: PMC6900016 DOI: 10.1002/anie.201909518] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/12/2019] [Indexed: 12/22/2022]
Abstract
Bioactive compound design based on natural product (NP) structure may be limited because of partial coverage of NP-like chemical space and biological target space. These limitations can be overcome by combining NP-centered strategies with fragment-based compound design through combination of NP-derived fragments to afford structurally unprecedented "pseudo-natural products" (pseudo-NPs). The design, synthesis, and biological evaluation of a collection of indomorphan pseudo-NPs that combine biosynthetically unrelated indole- and morphan-alkaloid fragments are described. Indomorphane derivative Glupin was identified as a potent inhibitor of glucose uptake by selectively targeting and upregulating glucose transporters GLUT-1 and GLUT-3. Glupin suppresses glycolysis, reduces the levels of glucose-derived metabolites, and attenuates the growth of various cancer cell lines. Our findings underscore the importance of dual GLUT-1 and GLUT-3 inhibition to efficiently suppress tumor cell growth and the cellular rescue mechanism, which counteracts glucose scarcity.
Collapse
Affiliation(s)
- Javier Ceballos
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
- Current address: Laboratory of Catalysis and Organic SynthesisEPFL SB ISIC LCSO, BCH 42211015LausanneSwitzerland
| | - Melanie Schwalfenberg
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
| | - George Karageorgis
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
- Current address: School of ChemistryUniversity of LeedsLeedsLS2 9JTUK
| | - Elena S. Reckzeh
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
- Faculty of Chemistry and Chemical BiologyTechnical University DortmundOtto-Hahn-Strasse 644227DortmundGermany
| | - Sonja Sievers
- Compound Management and Screening Center, DortmundOtto-Hahn-Strasse 1144227DortmundGermany
| | - Claude Ostermann
- Compound Management and Screening Center, DortmundOtto-Hahn-Strasse 1144227DortmundGermany
| | - Axel Pahl
- Compound Management and Screening Center, DortmundOtto-Hahn-Strasse 1144227DortmundGermany
| | - Magnus Sellstedt
- Department of ChemistryUmeå University901 87UmeåSweden
- Current address: Clinical Chemistry, Laboratory MedicineUniversity Hospital of Umeå901 85UmeåSweden
| | - Jessica Nowacki
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
| | - Marjorie A. Carnero Corrales
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
| | - Julian Wilke
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
- Faculty of Chemistry and Chemical BiologyTechnical University DortmundOtto-Hahn-Strasse 644227DortmundGermany
| | - Luca Laraia
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
- Current address: Department of ChemistryTechnical University of DenmarkKemitorvet, Bygning 2072800Kgs LyngbyDenmark
| | - Kirsten Tschapalda
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
| | - Malte Metz
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
| | - Dominik A. Sehr
- Department of Molecular Cell BiologyInstitute of Biochemistry and PathobiochemistryRuhr University Bochum44801BochumGermany
| | - Silke Brand
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
| | - Konstanze Winklhofer
- Department of Molecular Cell BiologyInstitute of Biochemistry and PathobiochemistryRuhr University Bochum44801BochumGermany
| | - Petra Janning
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
| | - Slava Ziegler
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
| | - Herbert Waldmann
- Department of Chemical BiologyMax-Planck-Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
- Faculty of Chemistry and Chemical BiologyTechnical University DortmundOtto-Hahn-Strasse 644227DortmundGermany
| |
Collapse
|
209
|
Ceballos J, Schwalfenberg M, Karageorgis G, Reckzeh ES, Sievers S, Ostermann C, Pahl A, Sellstedt M, Nowacki J, Carnero Corrales MA, Wilke J, Laraia L, Tschapalda K, Metz M, Sehr DA, Brand S, Winklhofer K, Janning P, Ziegler S, Waldmann H. Synthesis of Indomorphan Pseudo‐Natural Product Inhibitors of Glucose Transporters GLUT‐1 and ‐3. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201909518] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Javier Ceballos
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
- Current address: Laboratory of Catalysis and Organic SynthesisEPFL SB ISIC LCSO, BCH 4221 1015 Lausanne Switzerland
| | - Melanie Schwalfenberg
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - George Karageorgis
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
- Current address: School of ChemistryUniversity of Leeds Leeds LS2 9JT UK
| | - Elena S. Reckzeh
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
- Faculty of Chemistry and Chemical BiologyTechnical University Dortmund Otto-Hahn-Strasse 6 44227 Dortmund Germany
| | - Sonja Sievers
- Compound Management and Screening Center, Dortmund Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Claude Ostermann
- Compound Management and Screening Center, Dortmund Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Axel Pahl
- Compound Management and Screening Center, Dortmund Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Magnus Sellstedt
- Department of ChemistryUmeå University 901 87 Umeå Sweden
- Current address: Clinical Chemistry, Laboratory MedicineUniversity Hospital of Umeå 901 85 Umeå Sweden
| | - Jessica Nowacki
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Marjorie A. Carnero Corrales
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Julian Wilke
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
- Faculty of Chemistry and Chemical BiologyTechnical University Dortmund Otto-Hahn-Strasse 6 44227 Dortmund Germany
| | - Luca Laraia
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
- Current address: Department of ChemistryTechnical University of Denmark Kemitorvet, Bygning 207 2800 Kgs Lyngby Denmark
| | - Kirsten Tschapalda
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Malte Metz
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Dominik A. Sehr
- Department of Molecular Cell BiologyInstitute of Biochemistry and PathobiochemistryRuhr University Bochum 44801 Bochum Germany
| | - Silke Brand
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Konstanze Winklhofer
- Department of Molecular Cell BiologyInstitute of Biochemistry and PathobiochemistryRuhr University Bochum 44801 Bochum Germany
| | - Petra Janning
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Slava Ziegler
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
| | - Herbert Waldmann
- Department of Chemical BiologyMax-Planck-Institute of Molecular Physiology Otto-Hahn-Strasse 11 44227 Dortmund Germany
- Faculty of Chemistry and Chemical BiologyTechnical University Dortmund Otto-Hahn-Strasse 6 44227 Dortmund Germany
| |
Collapse
|
210
|
Exploring fragment-based target-specific ranking protocol with machine learning on cathepsin S. J Comput Aided Mol Des 2019; 33:1095-1105. [PMID: 31729618 DOI: 10.1007/s10822-019-00247-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 11/02/2019] [Indexed: 12/12/2022]
Abstract
Cathepsin S (CatS), a member of cysteine cathepsin proteases, has been well studied due to its significant role in many pathological processes, including arthritis, cancer and cardiovascular diseases. CatS inhibitors have been included in D3R-GC3 for both docking pose prediction and affinity ranking, and in D3R-GC4 for binding affinity ranking. The difficulties posed by CatS inhibitors in D3R mainly come from three aspects: large size, high flexibility and similar chemical structures. We have participated in GC4; our best submitted model, which employs a similarity-based alignment docking and Vina scoring protocol, yielded Kendall's τ of 0.23 for 459 binders in GC4. In our further explorations with machine learning, by curating a CatS specific training set, adopting a similarity-based constrained docking method as well as an arm-based fragmentation strategy which can describe large inhibitors in a locality-sensitive fashion, our best structure-based ranking protocol can achieve Kendall's τ of 0.52 for all binders in GC4. In this exploration process, we have demonstrated the importance of training data, docking approaches and fragmentation strategies in inhibitor-ranking protocol development with machine learning.
Collapse
|
211
|
Schiedel M, Moroglu M, Ascough DMH, Chamberlain AER, Kamps JJAG, Sekirnik AR, Conway SJ. Chemische Epigenetik: der Einfluss chemischer und chemo‐biologischer Techniken auf die Zielstruktur‐Validierung von Bromodomänen. Angew Chem Int Ed Engl 2019. [DOI: 10.1002/ange.201812164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Matthias Schiedel
- Department of ChemistryChemistry Research LaboratoryUniversity of Oxford Mansfield Road Oxford OX1 3TA Großbritannien
| | - Mustafa Moroglu
- Department of ChemistryChemistry Research LaboratoryUniversity of Oxford Mansfield Road Oxford OX1 3TA Großbritannien
| | - David M. H. Ascough
- Department of ChemistryChemistry Research LaboratoryUniversity of Oxford Mansfield Road Oxford OX1 3TA Großbritannien
| | - Anna E. R. Chamberlain
- Department of ChemistryChemistry Research LaboratoryUniversity of Oxford Mansfield Road Oxford OX1 3TA Großbritannien
| | - Jos J. A. G. Kamps
- Department of ChemistryChemistry Research LaboratoryUniversity of Oxford Mansfield Road Oxford OX1 3TA Großbritannien
| | - Angelina R. Sekirnik
- Department of ChemistryChemistry Research LaboratoryUniversity of Oxford Mansfield Road Oxford OX1 3TA Großbritannien
| | - Stuart J. Conway
- Department of ChemistryChemistry Research LaboratoryUniversity of Oxford Mansfield Road Oxford OX1 3TA Großbritannien
| |
Collapse
|
212
|
Xiong Y, Greschik H, Johansson C, Seifert L, Bacher J, Park KS, Babault N, Martini M, Fagan V, Li F, Chau I, Christott T, Dilworth D, Barsyte-Lovejoy D, Vedadi M, Arrowsmith CH, Brennan P, Fedorov O, Jung M, Farnie G, Liu J, Oppermann U, Schüle R, Jin J. Discovery of a Potent and Selective Fragment-like Inhibitor of Methyllysine Reader Protein Spindlin 1 (SPIN1). J Med Chem 2019; 62:8996-9007. [PMID: 31260300 DOI: 10.1021/acs.jmedchem.9b00522] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
By screening an epigenetic compound library, we identified that UNC0638, a highly potent inhibitor of the histone methyltransferases G9a and GLP, was a weak inhibitor of SPIN1 (spindlin 1), a methyllysine reader protein. Our optimization of this weak hit resulted in the discovery of a potent, selective, and cell-active SPIN1 inhibitor, compound 3 (MS31). Compound 3 potently inhibited binding of trimethyllysine-containing peptides to SPIN1, displayed high binding affinity, was highly selective for SPIN1 over other epigenetic readers and writers, directly engaged SPIN1 in cells, and was not toxic to nontumorigenic cells. The crystal structure of the SPIN1-compound 3 complex indicated that it selectively binds tudor domain II of SPIN1. We also designed a structurally similar but inactive compound 4 (MS31N) as a negative control. Our results have demonstrated for the first time that potent, selective, and cell-active fragment-like inhibitors can be generated by targeting a single tudor domain.
Collapse
Affiliation(s)
- Yan Xiong
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Holger Greschik
- Department of Urology and Center for Clinical Research , University Freiburg Medical Center , Freiburg 79106 , Germany
| | - Catrine Johansson
- Structural Genomics Consortium, Botnar Research Center, NIHR Oxford BRU , University of Oxford , Oxford OX37LD , U.K
| | - Ludwig Seifert
- Institute of Pharmaceutical Sciences , University of Freiburg , Freiburg 79104 , Germany
| | - Johannes Bacher
- Institute of Pharmaceutical Sciences , University of Freiburg , Freiburg 79104 , Germany
| | - Kwang-Su Park
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Nicolas Babault
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Michael Martini
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Vincent Fagan
- Structural Genomics Consortium & Target Discovery Institute , University of Oxford , Oxford OX37DQ and OX37FZ, U.K
| | | | | | - Thomas Christott
- Structural Genomics Consortium & Target Discovery Institute , University of Oxford , Oxford OX37DQ and OX37FZ, U.K
| | | | | | - Masoud Vedadi
- Department of Pharmacology and Toxicology , University of Toronto , Toronto , Ontario M5S 1A8 , Canada
| | | | - Paul Brennan
- Structural Genomics Consortium & Target Discovery Institute , University of Oxford , Oxford OX37DQ and OX37FZ, U.K
| | - Oleg Fedorov
- Structural Genomics Consortium & Target Discovery Institute , University of Oxford , Oxford OX37DQ and OX37FZ, U.K
| | - Manfred Jung
- Institute of Pharmaceutical Sciences , University of Freiburg , Freiburg 79104 , Germany
- German Cancer Research Centre (DKFZ) , Heidelberg 69120 , Germany
- German Cancer Consortium (DKTK) , Freiburg 79106 , Germany
| | - Gillian Farnie
- Structural Genomics Consortium & Target Discovery Institute , University of Oxford , Oxford OX37DQ and OX37FZ, U.K
| | - Jing Liu
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| | - Udo Oppermann
- Structural Genomics Consortium, Botnar Research Center, NIHR Oxford BRU , University of Oxford , Oxford OX37LD , U.K
| | - Roland Schüle
- Department of Urology and Center for Clinical Research , University Freiburg Medical Center , Freiburg 79106 , Germany
- BIOSS Centre of Biological Signalling Studies , University of Freiburg , Freiburg 79106 , Germany
- German Cancer Consortium (DKTK) , Freiburg 79106 , Germany
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute , Icahn School of Medicine at Mount Sinai , New York , New York 10029 , United States
| |
Collapse
|
213
|
Fuglestad B, Kerstetter NE, Bédard S, Wand AJ. Extending the Detection Limit in Fragment Screening of Proteins Using Reverse Micelle Encapsulation. ACS Chem Biol 2019; 14:2224-2232. [PMID: 31550881 DOI: 10.1021/acschembio.9b00537] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Detection of very weak (Kd > 10 mM) interactions of proteins with small molecules has been elusive. This is particularly important for fragment-based drug discovery, where it is suspected that the majority of potentially useful fragments will be invisible to current screening methodologies. We describe an NMR approach that permits detection of protein-fragment interactions in the very low affinity range and extends the current detection limit of ∼10 mM up to ∼200 mM and beyond. Reverse micelle encapsulation is leveraged to effectively reach very high fragment and protein concentrations, a principle that is validated by binding model fragments to E. coli dihydrofolate reductase. The method is illustrated by target-detected screening of a small polar fragment library against interleukin-1β, which lacks a known ligand-binding pocket. Evaluation of binding by titration and structural context allows for validation of observed hits using rigorous structural and statistical criteria. The 21 curated hit molecules represent a remarkable hit rate of nearly 10% of the library. Analysis shows that fragment binding involves residues comprising two-thirds of the protein's surface. Current fragment screening methods rely on detection of relatively tight binding to ligand binding pockets. The method presented here illustrates a potential to faithfully discover starting points for development of small molecules that bind to a desired region of the protein, even if the targeted region is defined by a relatively flat surface.
Collapse
Affiliation(s)
- Brian Fuglestad
- Johnson Research Foundation and Department of Biochemistry and Biophysics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United States
| | - Nicole E. Kerstetter
- Graduate Group in Biochemistry and Molecular Biophysics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Sabrina Bédard
- Johnson Research Foundation and Department of Biochemistry and Biophysics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United States
| | - A. Joshua Wand
- Johnson Research Foundation and Department of Biochemistry and Biophysics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United States
- Graduate Group in Biochemistry and Molecular Biophysics, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| |
Collapse
|
214
|
Modular click chemistry libraries for functional screens using a diazotizing reagent. Nature 2019; 574:86-89. [PMID: 31578481 DOI: 10.1038/s41586-019-1589-1] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 08/08/2019] [Indexed: 12/20/2022]
Abstract
Click chemistry is a concept in which modular synthesis is used to rapidly find new molecules with desirable properties1. Copper(I)-catalysed azide-alkyne cycloaddition (CuAAC) triazole annulation and sulfur(VI) fluoride exchange (SuFEx) catalysis are widely regarded as click reactions2-4, providing rapid access to their products in yields approaching 100% while being largely orthogonal to other reactions. However, in the case of CuAAC reactions, the availability of azide reagents is limited owing to their potential toxicity and the risk of explosion involved in their preparation. Here we report another reaction to add to the click reaction family: the formation of azides from primary amines, one of the most abundant functional groups5. The reaction uses just one equivalent of a simple diazotizing species, fluorosulfuryl azide6-11 (FSO2N3), and enables the preparation of over 1,200 azides on 96-well plates in a safe and practical manner. This reliable transformation is a powerful tool for the CuAAC triazole annulation, the most widely used click reaction at present. This method greatly expands the number of accessible azides and 1,2,3-triazoles and, given the ubiquity of the CuAAC reaction, it should find application in organic synthesis, medicinal chemistry, chemical biology and materials science.
Collapse
|
215
|
Development of a Fragment-Based Screening Assay for the Focal Adhesion Targeting Domain Using SPR and NMR. Molecules 2019; 24:molecules24183352. [PMID: 31540099 PMCID: PMC6766811 DOI: 10.3390/molecules24183352] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 09/11/2019] [Accepted: 09/13/2019] [Indexed: 12/19/2022] Open
Abstract
The Focal Adhesion Targeting (FAT) domain of Focal Adhesion Kinase (FAK) is a promising drug target since FAK is overexpressed in many malignancies and promotes cancer cell metastasis. The FAT domain serves as a scaffolding protein, and its interaction with the protein paxillin localizes FAK to focal adhesions. Various studies have highlighted the importance of FAT-paxillin binding in tumor growth, cell invasion, and metastasis. Targeting this interaction through high-throughput screening (HTS) provides a challenge due to the large and complex binding interface. In this report, we describe a novel approach to targeting FAT through fragment-based drug discovery (FBDD). We developed two fragment-based screening assays-a primary SPR assay and a secondary heteronuclear single quantum coherence nuclear magnetic resonance (HSQC-NMR) assay. For SPR, we designed an AviTag construct, optimized SPR buffer conditions, and created mutant controls. For NMR, resonance backbone assignments of the human FAT domain were obtained for the HSQC assay. A 189-compound fragment library from Enamine was screened through our primary SPR assay to demonstrate the feasibility of a FAT-FBDD pipeline, with 19 initial hit compounds. A final total of 11 validated hits were identified after secondary screening on NMR. This screening pipeline is the first FBDD screen of the FAT domain reported and represents a valid method for further drug discovery efforts on this difficult target.
Collapse
|
216
|
Shi Y, von Itzstein M. How Size Matters: Diversity for Fragment Library Design. Molecules 2019; 24:molecules24152838. [PMID: 31387220 PMCID: PMC6696339 DOI: 10.3390/molecules24152838] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 08/02/2019] [Accepted: 08/03/2019] [Indexed: 12/11/2022] Open
Abstract
Fragment-based drug discovery (FBDD) has become a major strategy to derive novel lead candidates for various therapeutic targets, as it promises efficient exploration of chemical space by employing fragment-sized (MW < 300) compounds. One of the first challenges in implementing a FBDD approach is the design of a fragment library, and more specifically, the choice of its size and individual members. A diverse set of fragments is required to maximize the chances of discovering novel hit compounds. However, the exact diversity of a certain collection of fragments remains underdefined, which hinders direct comparisons among different selections of fragments. Based on structural fingerprints, we herein introduced quantitative metrics for the structural diversity of fragment libraries. Structures of commercially available fragments were retrieved from the ZINC database, from which libraries with sizes ranging from 100 to 100,000 compounds were selected. The selected libraries were evaluated and compared quantitatively, resulting in interesting size-diversity relationships. Our results demonstrated that while library size does matter for its diversity, there exists an optimal size for structural diversity. It is also suggested that such quantitative measures can guide the design of diverse fragment libraries under different circumstances.
Collapse
Affiliation(s)
- Yun Shi
- Institute for Glycomics, Griffith University, Gold Coast Campus, Gold Coast, Queensland 4222, Australia.
| | - Mark von Itzstein
- Institute for Glycomics, Griffith University, Gold Coast Campus, Gold Coast, Queensland 4222, Australia.
| |
Collapse
|
217
|
Methyl N-(carbazolyl)acetyl-2-aminotetrahydrobenzothiophene-3-carboxylates as modulators of NMDA receptors. Russ Chem Bull 2019. [DOI: 10.1007/s11172-019-2602-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
218
|
Prasher P, Sharma M. Tailored therapeutics based on 1,2,3-1 H-triazoles: a mini review. MEDCHEMCOMM 2019; 10:1302-1328. [PMID: 31534652 PMCID: PMC6748286 DOI: 10.1039/c9md00218a] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 05/13/2019] [Indexed: 12/19/2022]
Abstract
Contemporary drug discovery approaches rely on library synthesis coupled with combinatorial methods and high-throughput screening to identify leads. However, due to the multitude of components involved, a majority of optimization techniques face persistent challenges related to the efficiency of synthetic processes and the purity of compound libraries. These methods have recently found an upgradation as fragment-based approaches for target-guided synthesis of lead molecules with active involvement of their biological target. The click chemistry approach serves as a promising tool for tailoring the therapeutically relevant biomolecules of interest, improving their bioavailability and bioactivity and redirecting them as efficacious drugs. 1,2,3-1H-Triazole nucleus, being a planar and biologically acceptable scaffold, plays a crucial role in the design of biomolecular mimetics and tailor-made molecules with therapeutic relevance. This versatile scaffold also forms an integral part of the current fragment-based approaches for drug design, kinetic target guided synthesis and bioorthogonal methodologies.
Collapse
Affiliation(s)
- Parteek Prasher
- UGC Sponsored Centre for Advanced Studies , Department of Chemistry , Guru Nanak Dev University , Amritsar 143005 , India . ;
- Department of Chemistry , University of Petroleum & Energy Studies , Dehradun 248007 , India
| | - Mousmee Sharma
- UGC Sponsored Centre for Advanced Studies , Department of Chemistry , Guru Nanak Dev University , Amritsar 143005 , India . ;
| |
Collapse
|
219
|
Boyd SR, Young DW. Max-imizing the Attenuation of Myc Using Small Molecules. Trends Pharmacol Sci 2019; 40:608-612. [PMID: 31280901 DOI: 10.1016/j.tips.2019.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 06/11/2019] [Indexed: 11/18/2022]
Abstract
It has been a widely held notion within the biomedical research community that the reliable modulation of transcription factors with small molecules would represent a holy grail, given their role in directly potentiating oncogenic programs. Among the transcription factors that have been held in highest regard is Myc, since its dysregulation is among the most recurrent events in human cancer. Despite intense efforts, the ability to identify compounds that bind directly to Myc, resulting in its functional inhibition, have been met with only moderate success. However, a new approach reported by Struntz et al. (Cell Chem. Biol., 2019) focuses on a different strategy of discovering molecules that bind to Myc's obligate partner Max. Using a small-molecule microarray screen, they report the identification of KI-MS2-008, a compound that results in the stabilization of Max homodimers and the attenuation of Myc. KI-MS2-008 suppresses cancer cell grown both in vitro and within in vivo models.
Collapse
Affiliation(s)
- Shelton R Boyd
- Center for Drug Discovery (CDD), Baylor College of Medicine, Houston, TX 77030, USA; Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Damian W Young
- Center for Drug Discovery (CDD), Baylor College of Medicine, Houston, TX 77030, USA; Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX 77030, USA; Therapeutic Innovations Center (THINC@BCM), Baylor College of Medicine, Houston, TX 77030, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
220
|
Mishin A, Gusach A, Luginina A, Marin E, Borshchevskiy V, Cherezov V. An outlook on using serial femtosecond crystallography in drug discovery. Expert Opin Drug Discov 2019; 14:933-945. [PMID: 31184514 DOI: 10.1080/17460441.2019.1626822] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Introduction: X-ray crystallography has made important contributions to modern drug development but its application to many important drug targets has been extremely challenging. The recent emergence of X-ray free electron lasers (XFELs) and advancements in serial femtosecond crystallography (SFX) have offered new opportunities to overcome limitations of traditional crystallography to accelerate the structure-based drug discovery (SBDD) process. Areas covered: In this review, the authors describe the general principles of X-ray generation and the main properties of XFEL beams, outline details of SFX data collection and processing, and summarize the progress in the development of associated instrumentation for sample delivery and X-ray detection. An overview of the SFX applications to various important drug targets such as membrane proteins is also provided. Expert opinion: While SFX has already made clear advancements toward the understanding of the structure and dynamics of several major drug targets, its robust application in SBDD still needs further developments of new high-throughput techniques for sample production, automation of crystal delivery and data collection, as well as for processing and storage of large amounts of data. The expansion of the available XFEL beamtime is a key to the success of SFX in SBDD.
Collapse
Affiliation(s)
- Alexey Mishin
- a Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology , Dolgoprudny , Russia
| | - Anastasiia Gusach
- a Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology , Dolgoprudny , Russia
| | - Aleksandra Luginina
- a Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology , Dolgoprudny , Russia
| | - Egor Marin
- a Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology , Dolgoprudny , Russia
| | - Valentin Borshchevskiy
- a Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology , Dolgoprudny , Russia
| | - Vadim Cherezov
- a Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology , Dolgoprudny , Russia.,b Bridge Institute, Departments of Chemistry and Biological Sciences, University of Southern California , Los Angeles , CA , USA
| |
Collapse
|
221
|
Derda R, Ng S. Genetically encoded fragment-based discovery. Curr Opin Chem Biol 2019; 50:128-137. [DOI: 10.1016/j.cbpa.2019.03.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/09/2019] [Accepted: 03/12/2019] [Indexed: 12/30/2022]
|
222
|
Sveiczer A, North AJP, Mateu N, Kidd SL, Sore HF, Spring DR. Spirocycles as Rigidified sp3-Rich Scaffolds for a Fragment Collection. Org Lett 2019; 21:4600-4604. [DOI: 10.1021/acs.orglett.9b01499] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Attila Sveiczer
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, U.K
| | - Andrew J. P. North
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, U.K
| | - Natalia Mateu
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, U.K
| | - Sarah L. Kidd
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, U.K
| | - Hannah F. Sore
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, U.K
| | - David R. Spring
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, U.K
| |
Collapse
|
223
|
De Oliveira Silva A, McQuade J, Szostak M. Recent Advances in the Synthesis and Reactivity of Isothiazoles. Adv Synth Catal 2019. [DOI: 10.1002/adsc.201900072] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
| | - James McQuade
- Department of Chemistry Rutgers University 73 Warren Street Newark NJ 07102 USA
| | - Michal Szostak
- College of Chemistry and Chemical Engineering and Key Laboratory of Auxiliary Chemistry and Technology for Chemical Industry Ministry of Education Shaanxi University of Science and Technology Xi'an 710021 People's Republic of China
- Department of Chemistry Rutgers University 73 Warren Street Newark NJ 07102 USA
| |
Collapse
|
224
|
Flodén NJ, Trowbridge A, Willcox D, Walton SM, Kim Y, Gaunt MJ. Streamlined Synthesis of C(sp3)-Rich N-Heterospirocycles Enabled by Visible-Light-Mediated Photocatalysis. J Am Chem Soc 2019; 141:8426-8430. [DOI: 10.1021/jacs.9b03372] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Nils J. Flodén
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| | - Aaron Trowbridge
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| | - Darren Willcox
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| | - Scarlett M. Walton
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
- Medicinal Chemistry, Oncology, IMED Biotech Unit, AstraZeneca, Cambridge CB4 0WG, U.K
| | - Yongjoon Kim
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| | - Matthew J. Gaunt
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge CB2 1EW, U.K
| |
Collapse
|
225
|
Tatum N, Duarte F, Kamerlin SCL, Pohl E. Relative Binding Energies Predict Crystallographic Binding Modes of Ethionamide Booster Lead Compounds. J Phys Chem Lett 2019; 10:2244-2249. [PMID: 30965004 PMCID: PMC6503467 DOI: 10.1021/acs.jpclett.9b00741] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 04/09/2019] [Indexed: 06/09/2023]
Abstract
Transcriptional repressor EthR from Mycobacterium tuberculosis is a valuable target for antibiotic booster drugs. We previously reported a virtual screening campaign to identify EthR inhibitors for development. Two ligand binding orientations were often proposed, though only the top scoring pose was utilized for filtering of the large data set. We obtained biophysically validated hits, some of which yielded complex crystal structures. In some cases, the crystallized binding mode and top scoring mode agree, while for others an alternate ligand binding orientation was found. In this contribution, we combine rigid docking, molecular dynamics simulations, and the linear interaction energy method to calculate binding free energies and derive relative binding energies for a number of EthR inhibitors in both modes. This strategy allowed us to correctly predict the most favorable orientation. Therefore, this widely applicable approach will be suitable to triage multiple binding modes within EthR and other potential drug targets with similar characteristics.
Collapse
Affiliation(s)
- Natalie
J. Tatum
- Department
of Chemistry, Durham University, South Road, Durham DH1 3LE, U.K.
| | - Fernanda Duarte
- Department
of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford OX1 3TA, U.K.
| | - Shina C. L. Kamerlin
- Department
of Chemistry - BMC, Uppsala University, BMC Box 576, S-751 23 Uppsala, Sweden
| | - Ehmke Pohl
- Department
of Chemistry, Durham University, South Road, Durham DH1 3LE, U.K.
- Department
of Biosciences, Durham University, Durham DH1 3LE, U.K.
- Biophysical
Sciences Institute, Durham University, Durham DH1 3LE, U.K.
| |
Collapse
|
226
|
Ni S, Padial NM, Kingston C, Vantourout JC, Schmitt DC, Edwards JT, Kruszyk MM, Merchant RR, Mykhailiuk PK, Sanchez BB, Yang S, Perry MA, Gallego GM, Mousseau JJ, Collins MR, Cherney RJ, Lebed PS, Chen JS, Qin T, Baran PS. A Radical Approach to Anionic Chemistry: Synthesis of Ketones, Alcohols, and Amines. J Am Chem Soc 2019; 141:6726-6739. [PMID: 30943023 DOI: 10.1021/jacs.9b02238] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Historically accessed through two-electron, anionic chemistry, ketones, alcohols, and amines are of foundational importance to the practice of organic synthesis. After placing this work in proper historical context, this Article reports the development, full scope, and a mechanistic picture for a strikingly different way of forging such functional groups. Thus, carboxylic acids, once converted to redox-active esters (RAEs), can be utilized as formally nucleophilic coupling partners with other carboxylic derivatives (to produce ketones), imines (to produce benzylic amines), or aldehydes (to produce alcohols). The reactions are uniformly mild, operationally simple, and, in the case of ketone synthesis, broad in scope (including several applications to the simplification of synthetic problems and to parallel synthesis). Finally, an extensive mechanistic study of the ketone synthesis is performed to trace the elementary steps of the catalytic cycle and provide the end-user with a clear and understandable rationale for the selectivity, role of additives, and underlying driving forces involved.
Collapse
Affiliation(s)
- Shengyang Ni
- Department of Chemistry , Scripps Research , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| | - Natalia M Padial
- Department of Chemistry , Scripps Research , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| | - Cian Kingston
- Department of Chemistry , Scripps Research , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| | - Julien C Vantourout
- Department of Chemistry , Scripps Research , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| | - Daniel C Schmitt
- Pfizer Medicinal Sciences , Eastern Point Road , Groton , Connecticut 06340 , United States
| | - Jacob T Edwards
- Department of Chemistry , Scripps Research , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| | - Monika M Kruszyk
- Department of Chemistry , Scripps Research , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| | - Rohan R Merchant
- Department of Chemistry , Scripps Research , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| | - Pavel K Mykhailiuk
- Department of Chemistry , Scripps Research , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States.,Enamine Ltd. , Chervonotkatska 78 , 02094 Kyiv , Ukraine.,Chemistry Department , Taras Shevchenko National University of Kyiv , Volodymyrska 64 , 01601 Kyiv , Ukraine
| | - Brittany B Sanchez
- Automated Synthesis Facility , Scripps Research , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| | - Shouliang Yang
- Department of Chemistry , La Jolla Laboratories , Pfizer 10770 Science Center Drive , San Diego , California 92121 , United States
| | - Matthew A Perry
- Pfizer Medicinal Sciences , Eastern Point Road , Groton , Connecticut 06340 , United States
| | - Gary M Gallego
- Department of Chemistry , La Jolla Laboratories , Pfizer 10770 Science Center Drive , San Diego , California 92121 , United States
| | - James J Mousseau
- Pfizer Medicinal Sciences , Eastern Point Road , Groton , Connecticut 06340 , United States
| | - Michael R Collins
- Department of Chemistry , La Jolla Laboratories , Pfizer 10770 Science Center Drive , San Diego , California 92121 , United States
| | - Robert J Cherney
- Research & Development , Bristol-Myers Squibb Company , Rt. 206 & Province Line Road , Princeton , New Jersey 08543 , United States
| | - Pavlo S Lebed
- Enamine Ltd. , Chervonotkatska 78 , 02094 Kyiv , Ukraine.,ChemBioCenter , Taras Shevchenko National University of Kyiv , Volodymyrska 64 , 01601 Kyiv , Ukraine
| | - Jason S Chen
- Automated Synthesis Facility , Scripps Research , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| | - Tian Qin
- Department of Chemistry , Scripps Research , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| | - Phil S Baran
- Department of Chemistry , Scripps Research , 10550 North Torrey Pines Road , La Jolla , California 92037 , United States
| |
Collapse
|
227
|
Szakács D, Kocsis A, Szász R, Gál P, Pál G. Novel MASP-2 inhibitors developed via directed evolution of human TFPI1 are potent lectin pathway inhibitors. J Biol Chem 2019; 294:8227-8237. [PMID: 30952698 PMCID: PMC6527154 DOI: 10.1074/jbc.ra119.008315] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 03/28/2019] [Indexed: 12/28/2022] Open
Abstract
The lectin pathway (LP) of the complement system is an important antimicrobial defense mechanism, but it also contributes significantly to ischemia reperfusion injury (IRI) associated with myocardial infarct, stroke, and several other clinical conditions. Mannan-binding lectin-associated serine proteinase 2 (MASP-2) is essential for LP activation, and therefore, it is a potential drug target. We have previously developed the first two generations of MASP-2 inhibitors by in vitro evolution of two unrelated canonical serine proteinase inhibitors. These inhibitors were selective LP inhibitors, but their nonhuman origin rendered them suboptimal lead molecules for drug development. Here, we present our third-generation MASP-2 inhibitors that were developed based on a human inhibitor scaffold. We subjected the second Kunitz domain of human tissue factor pathway inhibitor 1 (TFPI1 D2) to directed evolution using phage display to yield inhibitors against human and rat MASP-2. These novel TFPI1-based MASP-2 inhibitor (TFMI-2) variants are potent and selective LP inhibitors in both human and rat serum. Directed evolution of the first Kunitz domain of TFPI1 had already yielded the potent kallikrein inhibitor, Kalbitor® (ecallantide), which is an FDA-approved drug to treat acute attacks of hereditary angioedema. Like hereditary angioedema, acute IRI is also related to the uncontrolled activation of a specific plasma serine proteinase. Therefore, TFMI-2 variants are promising lead molecules for drug development against IRI.
Collapse
Affiliation(s)
- Dávid Szakács
- Department of Biochemistry, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/C, H-1117 Budapest
| | - Andrea Kocsis
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, H-1117 Budapest
| | - Róbert Szász
- Department of Hematology, Institute of Internal Medicine, University of Debrecen, Nagyerdei krt. 98, H-4032 Debrecen
| | - Péter Gál
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, H-1117 Budapest
| | - Gábor Pál
- Department of Biochemistry, ELTE Eötvös Loránd University, Pázmány Péter sétány 1/C, H-1117 Budapest; EvolVeritas Biotechnology Ltd., Somogyi Béla u. 17, H-6600 Szentes, Hungary.
| |
Collapse
|
228
|
Deaton DN, Do Y, Holt J, Jeune MR, Kramer HF, Larkin AL, Orband-Miller LA, Peckham GE, Poole C, Price DJ, Schaller LT, Shen Y, Shewchuk LM, Stewart EL, Stuart JD, Thomson SA, Ward P, Wilson JW, Xu T, Guss JH, Musetti C, Rendina AR, Affleck K, Anders D, Hancock AP, Hobbs H, Hodgson ST, Hutchinson J, Leveridge MV, Nicholls H, Smith IE, Somers DO, Sneddon HF, Uddin S, Cleasby A, Mortenson PN, Richardson C, Saxty G. The discovery of quinoline-3-carboxamides as hematopoietic prostaglandin D synthase (H-PGDS) inhibitors. Bioorg Med Chem 2019; 27:1456-1478. [DOI: 10.1016/j.bmc.2019.02.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/30/2019] [Accepted: 02/08/2019] [Indexed: 11/30/2022]
|
229
|
Giordanetto F, Jin C, Willmore L, Feher M, Shaw DE. Fragment Hits: What do They Look Like and How do They Bind? J Med Chem 2019; 62:3381-3394. [PMID: 30875465 PMCID: PMC6466478 DOI: 10.1021/acs.jmedchem.8b01855] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
![]()
A “fragment hit”, a
molecule of low molecular weight
that has been validated to bind to a target protein, can be an effective
chemical starting point for a drug discovery project. Our ability
to find and progress fragment hits could potentially be improved by
enhancing our understanding of their binding properties, which to
date has largely been based on tacit knowledge and reports from individual
projects. In the work reported here, we systematically analyzed the
molecular and binding properties of fragment hits using 489 published
protein–fragment complexes. We identified a number of notable
features that these hits tend to have in common, including preferences
in buried surface area upon binding, hydrogen bonding and other directional
interactions with the protein targets, structural topology, functional-group
occurrence, and degree of carbon saturation. In the future, taking
account of these preferences in designing and selecting fragments
to screen against protein targets may increase the chances of success
in fragment screening campaigns.
Collapse
Affiliation(s)
| | - Chentian Jin
- D. E. Shaw Research , New York , New York 10036 , United States
| | | | - Miklos Feher
- D. E. Shaw Research , New York , New York 10036 , United States
| | - David E Shaw
- D. E. Shaw Research , New York , New York 10036 , United States.,Department of Biochemistry and Molecular Biophysics , Columbia University , New York , New York 10032 , United States
| |
Collapse
|
230
|
Hilton MC, Zhang X, Boyle BT, Alegre-Requena JV, Paton RS, McNally A. Heterobiaryl synthesis by contractive C-C coupling via P(V) intermediates. Science 2019; 362:799-804. [PMID: 30442804 DOI: 10.1126/science.aas8961] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 07/03/2018] [Accepted: 09/25/2018] [Indexed: 12/19/2022]
Abstract
Heterobiaryls composed of pyridine and diazine rings are key components of pharmaceuticals and are often central to pharmacological function. We present an alternative approach to metal-catalyzed cross-coupling to make heterobiaryls using contractive phosphorus C-C couplings, also termed phosphorus ligand coupling reactions. The process starts by regioselective phosphorus substitution of the C-H bonds para to nitrogen in two successive heterocycles; ligand coupling is then triggered via acidic alcohol solutions to form the heterobiaryl bond. Mechanistic studies imply that ligand coupling is an asynchronous process involving migration of one heterocycle to the ipso position of the other around a central pentacoordinate P(V) atom. The strategy can be applied to complex drug-like molecules containing multiple reactive sites and polar functional groups, and also enables convergent coupling of drug fragments and late-stage heteroarylation of pharmaceuticals.
Collapse
Affiliation(s)
- Michael C Hilton
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA
| | - Xuan Zhang
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA
| | - Benjamin T Boyle
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA
| | | | - Robert S Paton
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA. .,Chemistry Research Laboratory, University of Oxford, Oxford OX1 3TA, UK
| | - Andrew McNally
- Department of Chemistry, Colorado State University, Fort Collins, CO 80523, USA.
| |
Collapse
|
231
|
Shamsiya A, Damodaran B. A Click Strategy for the Synthesis of Fluorescent Pyrimidinone‐Triazole Hybrids with CDK2 Selectivity in HeLa and A549 Cell Lines. ChemistrySelect 2019. [DOI: 10.1002/slct.201803748] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Aranhikkal Shamsiya
- Department of ChemistryUniversity of Calicut, Malappuram- 673635 Kerala India
| | - Bahulayan Damodaran
- Department of ChemistryUniversity of Calicut, Malappuram- 673635 Kerala India
| |
Collapse
|
232
|
Fumagalli F, de Melo SMG, Ribeiro CM, Solcia MC, Pavan FR, da Silva Emery F. Exploiting the furo[2,3-b]pyridine core against multidrug-resistant Mycobacterium tuberculosis. Bioorg Med Chem Lett 2019; 29:974-977. [PMID: 30803803 DOI: 10.1016/j.bmcl.2019.02.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/17/2019] [Accepted: 02/18/2019] [Indexed: 10/27/2022]
Abstract
Identification of new antibiotics suitable for the treatment of tuberculosis is required. In addition to selectivity, it is necessary to find new antibiotics that are effective when the tuberculous mycobacteria are resistant to the available therapies. The furo[2,3-b]pyridine core offers potential for this application. Herein, we have described the screening of our in-house library of furopyridines against Mycobacterium tuberculosis and identified a promising selective bioactive compound against different drug-resistant strains of this mycobacteria. The library of compounds was prepared by a CH amination reaction using mild and metal-free conditions, increasing the available information about the reactivity of furo[2,3-b]pyridine core through this reaction.
Collapse
Affiliation(s)
- Fernando Fumagalli
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (FCFRP-USP), Av. do Café, s/n° - Campus Universitário da USP, 14040-903 Ribeirão Preto, SP, Brazil
| | - Shaiani Maria Gil de Melo
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo (FCFRP-USP), Av. do Café, s/n° - Campus Universitário da USP, 14040-903 Ribeirão Preto, SP, Brazil
| | - Camila Maríngolo Ribeiro
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Rodovia Araraquara-Jaú, km01, s/n, Campos Ville, 14800-903 Araraquara, Brazil
| | - Mariana Cristina Solcia
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Rodovia Araraquara-Jaú, km01, s/n, Campos Ville, 14800-903 Araraquara, Brazil
| | - Fernando Rogério Pavan
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Rodovia Araraquara-Jaú, km01, s/n, Campos Ville, 14800-903 Araraquara, Brazil.
| | - Flavio da Silva Emery
- Department of Biological Sciences, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Rodovia Araraquara-Jaú, km01, s/n, Campos Ville, 14800-903 Araraquara, Brazil.
| |
Collapse
|
233
|
Robson-Tull J. Biophysical screening in fragment-based drug design: a brief overview. ACTA ACUST UNITED AC 2019. [DOI: 10.1093/biohorizons/hzy015] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Affiliation(s)
- Jacob Robson-Tull
- Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
234
|
Liang JW, Wang MY, Wang S, Li XY, Meng FH. Fragment-Based Structural Optimization of a Natural Product Itampolin A as a p38α Inhibitor for Lung Cancer. Mar Drugs 2019; 17:md17010053. [PMID: 30642059 PMCID: PMC6356581 DOI: 10.3390/md17010053] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 01/01/2019] [Accepted: 01/07/2019] [Indexed: 12/27/2022] Open
Abstract
Marine animals and plants provide abundant secondary metabolites with antitumor activity. Itampolin A is a brominated natural tyrosine secondary metabolite that is isolated from the sponge Iotrochota purpurea. Recently, we have achieved the first total synthesis of this brominated tyrosine secondary metabolite, which was found to be a potent p38α inhibitor exhibiting anticancer effects. A fragment-based drug design (FBDD) was carried out to optimize itampolin A. Forty-five brominated tyrosine derivatives were synthesized with interesting biological activities. Then, a QSAR study was carried out to explore the structural determinants responsible for the activity of brominated tyrosine skeleton p38α inhibitors. The lead compound was optimized by a FBDD method, then three series of brominated tyrosine derivatives were synthesized and evaluated for their inhibitory activities against p38α and tumor cells. Compound 6o (IC50 = 0.66 μM) exhibited significant antitumor activity against non-small cell lung A549 cells (A549). This also demonstrated the feasibility of the FBDD method of structural optimization.
Collapse
Affiliation(s)
- Jing-Wei Liang
- School of Pharmacy, China Medical University, Liaoning 110122, China.
| | - Ming-Yang Wang
- School of Pharmacy, China Medical University, Liaoning 110122, China.
| | - Shan Wang
- School of Pharmacy, China Medical University, Liaoning 110122, China.
| | - Xin-Yang Li
- School of Pharmacy, China Medical University, Liaoning 110122, China.
| | - Fan-Hao Meng
- School of Pharmacy, China Medical University, Liaoning 110122, China.
| |
Collapse
|
235
|
Kirienko DR, Kang D, Kirienko NV. Novel Pyoverdine Inhibitors Mitigate Pseudomonas aeruginosa Pathogenesis. Front Microbiol 2019; 9:3317. [PMID: 30687293 PMCID: PMC6333909 DOI: 10.3389/fmicb.2018.03317] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/20/2018] [Indexed: 11/13/2022] Open
Abstract
Pseudomonas aeruginosa is a clinically important pathogen that causes a variety of infections, including urinary, respiratory, and other soft-tissue infections, particularly in hospitalized patients with immune defects, cystic fibrosis, or significant burns. Antimicrobial resistance is a substantial problem in P. aeruginosa treatment due to the inherent insensitivity of the pathogen to a wide variety of antimicrobial drugs and its rapid acquisition of additional resistance mechanisms. One strategy to circumvent this problem is the use of anti-virulent compounds to disrupt pathogenesis without directly compromising bacterial growth. One of the principle regulatory mechanisms for P. aeruginosa’s virulence is the iron-scavenging siderophore pyoverdine, as it governs in-host acquisition of iron, promotes expression of multiple virulence factors, and is directly toxic. Some combination of these activities renders pyoverdine indispensable for pathogenesis in mammalian models. Here we report identification of a panel of novel small molecules that disrupt pyoverdine function. These molecules directly act on pyoverdine, rather than affecting its biosynthesis. The compounds reduce the pathogenic effect of pyoverdine and improve the survival of Caenorhabditis elegans when challenged with P. aeruginosa by disrupting only this single virulence factor. Finally, these compounds can synergize with conventional antimicrobials, forming a more effective treatment. These compounds may help to identify, or be modified to become, viable drug leads in their own right. Finally, they also serve as useful tool compounds to probe pyoverdine activity.
Collapse
Affiliation(s)
- Daniel R Kirienko
- Department of BioSciences, Rice University, Houston, TX, United States
| | - Donghoon Kang
- Department of BioSciences, Rice University, Houston, TX, United States
| | | |
Collapse
|
236
|
Schaduangrat N, Prachayasittikul V, Choomwattana S, Wongchitrat P, Phopin K, Suwanjang W, Malik AA, Vincent B, Nantasenamat C. Multidisciplinary approaches for targeting the secretase protein family as a therapeutic route for Alzheimer's disease. Med Res Rev 2019; 39:1730-1778. [PMID: 30628099 DOI: 10.1002/med.21563] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 11/21/2018] [Accepted: 12/24/2018] [Indexed: 12/27/2022]
Abstract
The continual increase of the aging population worldwide renders Alzheimer's disease (AD) a global prime concern. Several attempts have been focused on understanding the intricate complexity of the disease's development along with the on- andgoing search for novel therapeutic strategies. Incapability of existing AD drugs to effectively modulate the pathogenesis or to delay the progression of the disease leads to a shift in the paradigm of AD drug discovery. Efforts aimed at identifying AD drugs have mostly focused on the development of disease-modifying agents in which effects are believed to be long lasting. Of particular note, the secretase enzymes, a group of proteases responsible for the metabolism of the β-amyloid precursor protein (βAPP) and β-amyloid (Aβ) peptides production, have been underlined for their promising therapeutic potential. This review article attempts to comprehensively cover aspects related to the identification and use of drugs targeting the secretase enzymes. Particularly, the roles of secretases in the pathogenesis of AD and their therapeutic modulation are provided herein. Moreover, an overview of the drug development process and the contribution of computational (in silico) approaches for facilitating successful drug discovery are also highlighted along with examples of relevant computational works. Promising chemical scaffolds, inhibitors, and modulators against each class of secretases are also summarized herein. Additionally, multitarget secretase modulators are also taken into consideration in light of the current growing interest in the polypharmacology of complex diseases. Finally, challenging issues and future outlook relevant to the discovery of drugs targeting secretases are also discussed.
Collapse
Affiliation(s)
- Nalini Schaduangrat
- Faculty of Medical Technology, Center of Data Mining and Biomedical Informatics, Mahidol University, Bangkok, Thailand
| | - Veda Prachayasittikul
- Faculty of Medical Technology, Center of Data Mining and Biomedical Informatics, Mahidol University, Bangkok, Thailand
| | - Saowapak Choomwattana
- Faculty of Medical Technology, Center of Data Mining and Biomedical Informatics, Mahidol University, Bangkok, Thailand
| | - Prapimpun Wongchitrat
- Faculty of Medical Technology, Center for Research and Innovation, Mahidol University, Bangkok, Thailand
| | - Kamonrat Phopin
- Faculty of Medical Technology, Center for Research and Innovation, Mahidol University, Bangkok, Thailand
| | - Wilasinee Suwanjang
- Faculty of Medical Technology, Center for Research and Innovation, Mahidol University, Bangkok, Thailand
| | - Aijaz Ahmad Malik
- Faculty of Medical Technology, Center of Data Mining and Biomedical Informatics, Mahidol University, Bangkok, Thailand
| | - Bruno Vincent
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand.,Centre National de la Recherche Scientifique, Paris, France
| | - Chanin Nantasenamat
- Faculty of Medical Technology, Center of Data Mining and Biomedical Informatics, Mahidol University, Bangkok, Thailand
| |
Collapse
|
237
|
Lattice engineering enables definition of molecular features allowing for potent small-molecule inhibition of HIV-1 entry. Nat Commun 2019; 10:47. [PMID: 30604750 PMCID: PMC6318274 DOI: 10.1038/s41467-018-07851-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 11/20/2018] [Indexed: 12/12/2022] Open
Abstract
Diverse entry inhibitors targeting the gp120 subunit of the HIV-1 envelope (Env) trimer have been developed including BMS-626529, also called temsavir, a prodrug version of which is currently in phase III clinical trials. Here we report the characterization of a panel of small-molecule inhibitors including BMS-818251, which we show to be >10-fold more potent than temsavir on a cross-clade panel of 208-HIV-1 strains, as well as the engineering of a crystal lattice to enable structure determination of the interaction between these inhibitors and the HIV-1 Env trimer at higher resolution. By altering crystallization lattice chaperones, we identify a lattice with both improved diffraction and robust co-crystallization of HIV-1 Env trimers from different clades complexed to entry inhibitors with a range of binding affinities. The improved diffraction reveals BMS-818251 to utilize functional groups that interact with gp120 residues from the conserved β20-β21 hairpin to improve potency. Temsavir, a compound that inhibits HIV entry by binding envelope (Env), is currently in clinical development. Here, Lai et al. identify a more than 10-fold improved compound and, using lattice engineering, obtain crystal structures that give insights into improved inhibition between small molecules and Env.
Collapse
|
238
|
Panicker RC, Chattopadhaya S, Coyne AG, Srinivasan R. Allosteric Small-Molecule Serine/Threonine Kinase Inhibitors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1163:253-278. [PMID: 31707707 DOI: 10.1007/978-981-13-8719-7_11] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Deregulation of protein kinase activity has been linked to many diseases ranging from cancer to AIDS and neurodegenerative diseases. Not surprisingly, drugging the human kinome - the complete set of kinases encoded by the human genome - has been one of the major drug discovery pipelines. Majority of the approved clinical kinase inhibitors target the ATP binding site of kinases. However, the remarkable sequence and structural similarity of ATP binding pockets of kinases make selective inhibition of kinases a daunting task. To circumvent these issues, allosteric inhibitors that target sites other than the orthosteric ATP binding pocket have been developed. The structural diversity of the allosteric sites allows these inhibitors to have higher selectivity, lower toxicity and improved physiochemical properties and overcome drug resistance associated with the use of conventional kinase inhibitors. In this chapter, we will focus on the allosteric inhibitors of selected serine/threonine kinases, outline the benefits of using these inhibitors and discuss the challenges and future opportunities.
Collapse
Affiliation(s)
- Resmi C Panicker
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, People's Republic of China
| | | | - Anthony G Coyne
- University Chemical Laboratory, University of Cambridge, Cambridge, UK
| | - Rajavel Srinivasan
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, People's Republic of China.
| |
Collapse
|
239
|
Song K, Zhang J, Lu S. Progress in Allosteric Database. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1163:65-87. [PMID: 31707700 DOI: 10.1007/978-981-13-8719-7_4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
An allosteric mechanism refers to the biological regulation process wherein macromolecules propagate the effect of ligand binding at one site to a spatially distant orthosteric locus, thus affecting activity. The theory has remained a trending topic in biology research for over 50 years, since the understanding of allostery is fundamental for gleaning numerous biological processes and developing new drug therapies. In the past two decades, the allosteric paradigm has evolved into more descriptive models, with ever-expanding amounts of experimental data pertaining to newly identified allosteric molecules. The AlloSteric Database (ASD, accessible at http://mdl.shsmu.edu.cn/ASD ), which is a comprehensive knowledge repository, has provided the public with integrated information encompassing allosteric proteins, modulators, sites, pathways, and networks to investigate allostery since 2009. In this chapter, we introduce the history and usage of the ASD and give attention to specific applications that have benefited from the ASD.
Collapse
Affiliation(s)
- Kun Song
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Jian Zhang
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai, China
| | - Shaoyong Lu
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University, School of Medicine, Shanghai, China.
| |
Collapse
|
240
|
Evans GB, Schramm VL, Tyler PC. The transition to magic bullets - transition state analogue drug design. MEDCHEMCOMM 2018; 9:1983-1993. [PMID: 30627387 PMCID: PMC6295874 DOI: 10.1039/c8md00372f] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 08/24/2018] [Indexed: 12/17/2022]
Abstract
In the absence of industry partnerships, most academic groups lack the infrastructure to rationally design and build drugs via methods used in industry. Instead, academia needs to work smarter using mechanism-based design. Working smarter can mean the development of new drug discovery paradigms and then demonstrating their utility and reproducibility to industry. The collaboration between Vern Schramm's group at the Albert Einstein College of Medicine, USA and Peter Tyler at the Ferrier Research Institute at The Victoria University of Wellington, NZ has refined a drug discovery process called transition state analogue design. This process has been applied to several biomedically relevant nucleoside processing enzymes. In 2017, Mundesine®, conceived using transition state analogue design, received market approval for the treatment of peripheral T-cell lymphoma in Japan. This short review looks at a brief history of transition state analogue design, the fundamentals behind the development of this process, and the success of enzyme inhibitors produced using this drug design methodology.
Collapse
Affiliation(s)
- Gary B Evans
- The Ferrier Research Institute , Victoria University of Wellington , 69 Gracefield Rd , Lower Hutt , 5010 , New Zealand . ; Tel: +64 4 463 0048
- The Maurice Wilkins Centre for Molecular Biodiscovery , The University of Auckland , Auckland , New Zealand
| | - Vern L Schramm
- Department of Biochemistry , Albert Einstein College of Medicine , Bronx , NY 10461 , USA
| | - Peter C Tyler
- The Ferrier Research Institute , Victoria University of Wellington , 69 Gracefield Rd , Lower Hutt , 5010 , New Zealand . ; Tel: +64 4 463 0048
| |
Collapse
|
241
|
Mortenson PN, Erlanson DA, de Esch IJP, Jahnke W, Johnson CN. Fragment-to-Lead Medicinal Chemistry Publications in 2017. J Med Chem 2018; 62:3857-3872. [PMID: 30462504 DOI: 10.1021/acs.jmedchem.8b01472] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This Miniperspective is the third in a series reviewing fragment-to-lead publications from a given year. Following our reviews for 2015 and 2016, this Miniperspective provides tabulated summaries of relevant articles published in 2017 along with some general observations. In addition, we discuss insights obtained from analysis of the combined data set of 85 examples from all three years of publications.
Collapse
Affiliation(s)
- Paul N Mortenson
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road , Cambridge CB4 0QA , United Kingdom
| | - Daniel A Erlanson
- Carmot Therapeutics Inc. , 740 Heinz Avenue , Berkeley , California 94710 , United States
| | - Iwan J P de Esch
- Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS) , Vrije Universiteit Amsterdam , De Boelelaan 1108 , 1081 HZ , Amsterdam , The Netherlands
| | - Wolfgang Jahnke
- Chemical Biology and Therapeutics , Novartis Institutes for Biomedical Research , 4002 Basel , Switzerland
| | - Christopher N Johnson
- Astex Pharmaceuticals , 436 Cambridge Science Park, Milton Road , Cambridge CB4 0QA , United Kingdom
| |
Collapse
|
242
|
Silva DG, Emery FDS. Strategies towards expansion of chemical space of natural product-based compounds to enable drug discovery. BRAZ J PHARM SCI 2018. [DOI: 10.1590/s2175-97902018000001004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
243
|
Zhou Y, Li C, Peng J, Xie L, Meng L, Li Q, Zhang J, Li XD, Li X, Huang X, Li X. DNA-Encoded Dynamic Chemical Library and Its Applications in Ligand Discovery. J Am Chem Soc 2018; 140:15859-15867. [DOI: 10.1021/jacs.8b09277] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Yu Zhou
- Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, 2199 Lishui Road West, Shenzhen 518055, China
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong
| | - Chen Li
- Key Laboratory of Chemical Genomics, School of Chemical Biology and Biotechnology, Peking University Shenzhen Graduate School, 2199 Lishui Road West, Shenzhen 518055, China
| | - Jianzhao Peng
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong
- Department of Chemistry, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen 518055, China
| | - Liangxu Xie
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water
Bay, Kowloon, Hong Kong, Hong Kong
| | - Ling Meng
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong
| | - Qingrong Li
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong
- Department of Chemistry, Southern University of Science and Technology, 1088 Xueyuan Road, Shenzhen 518055, China
| | - Jianfu Zhang
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong
| | - Xiang David Li
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong
| | - Xin Li
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong
| | - Xuhui Huang
- Department of Chemistry, The Hong Kong University of Science and Technology, Clear Water
Bay, Kowloon, Hong Kong, Hong Kong
| | - Xiaoyu Li
- Department of Chemistry, The University of Hong Kong, Pokfulam Road, Hong Kong, Hong Kong
| |
Collapse
|
244
|
Ni S, Garrido-Castro AF, Merchant RR, de Gruyter JN, Schmitt DC, Mousseau JJ, Gallego GM, Yang S, Collins MR, Qiao JX, Yeung KS, Langley DR, Poss MA, Scola PM, Qin T, Baran PS. A General Amino Acid Synthesis Enabled by Innate Radical Cross-Coupling. Angew Chem Int Ed Engl 2018; 57:14560-14565. [PMID: 30212610 PMCID: PMC6352899 DOI: 10.1002/anie.201809310] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Indexed: 01/21/2023]
Abstract
The direct union of primary, secondary, and tertiary carboxylic acids with a chiral glyoxylate-derived sulfinimine provides rapid access into a variety of enantiomerically pure α-amino acids (>85 examples). Characterized by operational simplicity, this radical-based reaction enables the modular assembly of exotic α-amino acids, including both unprecedented structures and those of established industrial value. The described method performs well in high-throughput library synthesis, and has already been implemented in three distinct medicinal chemistry campaigns.
Collapse
Affiliation(s)
- Shengyang Ni
- Scripps Research, North Torrey Pines Road, La Jolla, CA, 92037, USA
| | | | - Rohan R Merchant
- Scripps Research, North Torrey Pines Road, La Jolla, CA, 92037, USA
| | | | - Daniel C Schmitt
- Pfizer Medicinal Sciences, Eastern Point Road, Groton, CT, 06340, USA
| | - James J Mousseau
- Pfizer Medicinal Sciences, Eastern Point Road, Groton, CT, 06340, USA
| | - Gary M Gallego
- Department of Chemistry, La Jolla Laboratories, Pfizer, 10770 Science Center Drive, San Diego, CA, 92121, USA
| | - Shouliang Yang
- Department of Chemistry, La Jolla Laboratories, Pfizer, 10770 Science Center Drive, San Diego, CA, 92121, USA
| | - Michael R Collins
- Department of Chemistry, La Jolla Laboratories, Pfizer, 10770 Science Center Drive, San Diego, CA, 92121, USA
| | - Jennifer X Qiao
- Department of Discovery Chemistry, Bristol-Myers Squibb Company, Research and Development, P.O. Box 4000, Princeton, NJ, 08543, USA
| | - Kap-Sun Yeung
- Department of Discovery Chemistry, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT, 06492, USA
| | - David R Langley
- Department of Discovery Chemistry, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT, 06492, USA
| | - Michael A Poss
- Department of Discovery Chemistry, Bristol-Myers Squibb Company, Research and Development, P.O. Box 4000, Princeton, NJ, 08543, USA
| | - Paul M Scola
- Department of Discovery Chemistry, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT, 06492, USA
| | - Tian Qin
- Scripps Research, North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Phil S Baran
- Scripps Research, North Torrey Pines Road, La Jolla, CA, 92037, USA
| |
Collapse
|
245
|
Moustakim M, Felce SL, Zaarour N, Farnie G, McCann FE, Brennan PE. Target Identification Using Chemical Probes. Methods Enzymol 2018; 610:27-58. [PMID: 30390803 DOI: 10.1016/bs.mie.2018.09.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Chemical probes are small molecules with potency and selectivity for a single or small number of protein targets. A good chemical probe engages its target intracellularly and is accompanied by a chemically similar, but inactive molecule to be used as a negative control in cellular phenotypic screening. The utility of these chemical probes is ultimately governed by how well they are developed and characterized. Chemical probes either as single entities, or in chemical probes sets are being increasingly used to interrogate the biological relevance of a target in a disease model. This chapter lays out the core properties of chemical probes, summarizes the seminal and emerging techniques used to demonstrate robust intracellular target engagement. Translation of target engagement assays to disease-relevant phenotypic assays using primary patient-derived cells and tissues is also reviewed. Two examples of epigenetic chemical probe discovery and utility are presented whereby target engagement pointed to novel disease associations elucidated from poorly understood protein targets. Finally, a number of examples are discussed whereby chemical probe sets, or "chemogenomic libraries" are used to illuminate new target-disease links which may represent future directions for chemical probe utility.
Collapse
Affiliation(s)
- Moses Moustakim
- Nuffield Department of Medicine, Structural Genomics Consortium, University of Oxford, Oxford, United Kingdom; Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, United Kingdom; Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Oxford, United Kingdom
| | - Suet Ling Felce
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom
| | - Nancy Zaarour
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom; Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Gillian Farnie
- Nuffield Department of Medicine, Structural Genomics Consortium, University of Oxford, Oxford, United Kingdom; Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom.
| | - Fiona E McCann
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, United Kingdom; Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom.
| | - Paul E Brennan
- Nuffield Department of Medicine, Structural Genomics Consortium, University of Oxford, Oxford, United Kingdom; Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, United Kingdom; Nuffield Department of Medicine, Alzheimer's Research UK Oxford Drug Discovery Institute, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
246
|
Kidd SL, Osberger TJ, Mateu N, Sore HF, Spring DR. Recent Applications of Diversity-Oriented Synthesis Toward Novel, 3-Dimensional Fragment Collections. Front Chem 2018; 6:460. [PMID: 30386766 PMCID: PMC6198038 DOI: 10.3389/fchem.2018.00460] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2018] [Accepted: 09/14/2018] [Indexed: 12/23/2022] Open
Abstract
Fragment-based drug discovery (FBDD) is a well-established approach for the discovery of novel medicines, illustrated by the approval of two FBBD-derived drugs. This methodology is based on the utilization of small "fragment" molecules (<300 Da) as starting points for drug discovery and optimization. Organic synthesis has been identified as a significant obstacle in FBDD, however, in particular owing to the lack of novel 3-dimensional (3D) fragment collections that feature useful synthetic vectors for modification of hit compounds. Diversity-oriented synthesis (DOS) is a synthetic strategy that aims to efficiently produce compound collections with high levels of structural diversity and three-dimensionality and is therefore well-suited for the construction of novel fragment collections. This Mini-Review highlights recent studies at the intersection of DOS and FBDD aiming to produce novel libraries of diverse, polycyclic, fragment-like compounds, and their application in fragment-based screening projects.
Collapse
Affiliation(s)
| | | | | | | | - David R. Spring
- Department of Chemistry, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
247
|
Ni S, Garrido-Castro AF, Merchant RR, de Gruyter JN, Schmitt DC, Mousseau JJ, Gallego GM, Yang S, Collins MR, Qiao JX, Yeung KS, Langley DR, Poss MA, Scola PM, Qin T, Baran PS. A General Amino Acid Synthesis Enabled by Innate Radical Cross-Coupling. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201809310] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Shengyang Ni
- Scripps Research; North Torrey Pines Road La Jolla CA 92037 USA
| | | | | | | | | | | | - Gary M. Gallego
- Department of Chemistry; La Jolla Laboratories; Pfizer; 10770 Science Center Drive San Diego CA 92121 USA
| | - Shouliang Yang
- Department of Chemistry; La Jolla Laboratories; Pfizer; 10770 Science Center Drive San Diego CA 92121 USA
| | - Michael R. Collins
- Department of Chemistry; La Jolla Laboratories; Pfizer; 10770 Science Center Drive San Diego CA 92121 USA
| | - Jennifer X. Qiao
- Department of Discovery Chemistry; Bristol-Myers Squibb Company; Research and Development; P.O. Box 4000 Princeton NJ 08543 USA
| | - Kap-Sun Yeung
- Department of Discovery Chemistry; Bristol-Myers Squibb Research and Development; 5 Research Parkway Wallingford CT 06492 USA
| | - David R. Langley
- Department of Discovery Chemistry; Bristol-Myers Squibb Research and Development; 5 Research Parkway Wallingford CT 06492 USA
| | - Michael A. Poss
- Department of Discovery Chemistry; Bristol-Myers Squibb Company; Research and Development; P.O. Box 4000 Princeton NJ 08543 USA
| | - Paul M. Scola
- Department of Discovery Chemistry; Bristol-Myers Squibb Research and Development; 5 Research Parkway Wallingford CT 06492 USA
| | - Tian Qin
- Scripps Research; North Torrey Pines Road La Jolla CA 92037 USA
| | - Phil S. Baran
- Scripps Research; North Torrey Pines Road La Jolla CA 92037 USA
| |
Collapse
|
248
|
Abstract
WaterLOGSY is a ligand-observed NMR method that is widely used for the studies of protein-small molecule interactions. The basis of waterLOGSY relies on the transfer of magnetization between water molecules, proteins, and small molecules via the nuclear Overhauser effect and chemical exchange. WaterLOGSY is used extensively for the screening of protein ligands, as it is a robust, relatively high-throughput, and reliable method to identify small molecules that bind proteins with a binding affinity (KD) in the μM to mM region. WaterLOGSY also enables the determination of KD via ligand titration, although careful optimization of the experimental setup is required to avoid overestimation of binding constants. Finally, waterLOGSY allows the water-accessible ligand protons of protein-bound ligands to be identified, thus providing structural information of the ligand binding orientation. In this chapter, we introduce and describe the waterLOGSY method, and provide a practical guide for ligand screening and KD determination. The use of waterLOGSY to study water accessibility is also discussed.
Collapse
Affiliation(s)
- Renjie Huang
- School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
| | - Ivanhoe K H Leung
- School of Chemical Sciences, The University of Auckland, Auckland, New Zealand.
| |
Collapse
|
249
|
Karageorgis G, Reckzeh ES, Ceballos J, Schwalfenberg M, Sievers S, Ostermann C, Pahl A, Ziegler S, Waldmann H. Chromopynones are pseudo natural product glucose uptake inhibitors targeting glucose transporters GLUT-1 and -3. Nat Chem 2018; 10:1103-1111. [DOI: 10.1038/s41557-018-0132-6] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 07/24/2018] [Indexed: 12/22/2022]
|
250
|
Srinivasan B, Tonddast-Navaei S, Roy A, Zhou H, Skolnick J. Chemical space of Escherichia coli dihydrofolate reductase inhibitors: New approaches for discovering novel drugs for old bugs. Med Res Rev 2018; 39:684-705. [PMID: 30192413 DOI: 10.1002/med.21538] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/16/2018] [Accepted: 08/09/2018] [Indexed: 12/15/2022]
Abstract
Escherichia coli Dihydrofolate reductase is an important enzyme that is essential for the survival of the Gram-negative microorganism. Inhibitors designed against this enzyme have demonstrated application as antibiotics. However, either because of poor bioavailability of the small-molecules resulting from their inability to cross the double membrane in Gram-negative bacteria or because the microorganism develops resistance to the antibiotics by mutating the DHFR target, discovery of new antibiotics against the enzyme is mandatory to overcome drug-resistance. This review summarizes the field of DHFR inhibition with special focus on recent efforts to effectively interface computational and experimental efforts to discover novel classes of inhibitors that target allosteric and active-sites in drug-resistant variants of EcDHFR.
Collapse
Affiliation(s)
- Bharath Srinivasan
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, Atlanta, Georgia
| | - Sam Tonddast-Navaei
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, Atlanta, Georgia
| | - Ambrish Roy
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, Atlanta, Georgia
| | - Hongyi Zhou
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, Atlanta, Georgia
| | - Jeffrey Skolnick
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|