1
|
Wang TY, Rukundo JL, Mao Z, Krylov SN. Maximizing the Accuracy of Equilibrium Dissociation Constants for Affinity Complexes: From Theory to Practical Recommendations. ACS Chem Biol 2024; 19:1852-1867. [PMID: 39121869 DOI: 10.1021/acschembio.4c00259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2024]
Abstract
The equilibrium dissociation constant (Kd) is a major characteristic of affinity complexes and one of the most frequently determined physicochemical parameters. Despite its significance, the values of Kd obtained for the same complex under similar conditions often exhibit considerable discrepancies and sometimes vary by orders of magnitude. These inconsistencies highlight the susceptibility of Kd determination to large systematic errors, even when random errors are small. It is imperative to both minimize and quantitatively assess the systematic errors inherent in Kd determination. Traditionally, Kd values are determined through nonlinear regression of binding isotherms. This analysis utilizes three variables: concentrations of two reactants and a fraction R of unbound limiting reactant. The systematic errors in Kd arise directly from systematic errors in these variables. Therefore, to maximize the accuracy of Kd, this study thoroughly analyzes the sources of systematic errors within the three variables, including (i) non-additive signals to calculate R, (ii) mis-calibrated experimental instruments, (iii) inaccurate calibration parameters, (iv) insufficient incubation time, (v) unsaturated binding isotherm, (vi) impurities in the reactants, and (vii) solute adsorption onto surfaces. Through this analysis, we illustrate how each source contributes to inaccuracies in the determination of Kd and propose strategies to minimize these contributions. Additionally, we introduce a method for quantitatively assessing the confidence intervals of systematic errors in concentrations, a crucial step toward quantitatively evaluating the accuracy of Kd. While presenting original findings, this paper also reiterates the fundamentals of Kd determination, hence guiding researchers across all proficiency levels. By shedding light on the sources of systematic errors and offering strategies for their mitigation, our work will help researchers enhance the accuracy of Kd determination, thereby making binding studies more reliable and the conclusions drawn from such studies more robust.
Collapse
Affiliation(s)
- Tong Ye Wang
- Department of Chemistry, York University, Toronto, Ontario M3J 1P3, Canada
- Centre for Research on Biomolecular Interactions, York University, Toronto, Ontario M3J 1P3, Canada
| | - Jean-Luc Rukundo
- Department of Chemistry, York University, Toronto, Ontario M3J 1P3, Canada
- Centre for Research on Biomolecular Interactions, York University, Toronto, Ontario M3J 1P3, Canada
| | - Zhiyuan Mao
- Department of Chemistry, York University, Toronto, Ontario M3J 1P3, Canada
- Centre for Research on Biomolecular Interactions, York University, Toronto, Ontario M3J 1P3, Canada
| | - Sergey N Krylov
- Department of Chemistry, York University, Toronto, Ontario M3J 1P3, Canada
- Centre for Research on Biomolecular Interactions, York University, Toronto, Ontario M3J 1P3, Canada
| |
Collapse
|
2
|
Brindani N, Vuong LM, La Serra MA, Salvador N, Menichetti A, Acquistapace IM, Ortega JA, Veronesi M, Bertozzi SM, Summa M, Girotto S, Bertorelli R, Armirotti A, Ganesan AK, De Vivo M. Discovery of CDC42 Inhibitors with a Favorable Pharmacokinetic Profile and Anticancer In Vivo Efficacy. J Med Chem 2024; 67:10401-10424. [PMID: 38866385 PMCID: PMC11215724 DOI: 10.1021/acs.jmedchem.4c00855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/07/2024] [Accepted: 05/13/2024] [Indexed: 06/14/2024]
Abstract
We previously reported trisubstituted pyrimidine lead compounds, namely, ARN22089 and ARN25062, which block the interaction between CDC42 with its specific downstream effector, a PAK protein. This interaction is crucial for the progression of multiple tumor types. Such inhibitors showed anticancer efficacy in vivo. Here, we describe a second class of CDC42 inhibitors with favorable drug-like properties. Out of the 25 compounds here reported, compound 15 (ARN25499) stands out as the best lead compound with an improved pharmacokinetic profile, increased bioavailability, and efficacy in an in vivo PDX tumor mouse model. Our results indicate that these CDC42 inhibitors represent a promising chemical class toward the discovery of anticancer drugs, with ARN25499 as an additional lead candidate for preclinical development.
Collapse
Affiliation(s)
- Nicoletta Brindani
- Molecular
Modeling and Drug Discovery Lab, Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Linh M. Vuong
- Department
of Dermatology, University of California, Irvine, California 92697, United States
| | - Maria Antonietta La Serra
- Molecular
Modeling and Drug Discovery Lab, Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Noel Salvador
- Department
of Dermatology, University of California, Irvine, California 92697, United States
| | - Andrea Menichetti
- Molecular
Modeling and Drug Discovery Lab, Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Isabella Maria Acquistapace
- Molecular
Modeling and Drug Discovery Lab, Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Jose Antonio Ortega
- Molecular
Modeling and Drug Discovery Lab, Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Marina Veronesi
- Structural
Biophysics Facility, Istituto Italiano di
Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Sine Mandrup Bertozzi
- Analytical
Chemistry Facility, Istituto Italiano di
Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Maria Summa
- Translational
Pharmacology Facility, Istituto Italiano
di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Stefania Girotto
- Structural
Biophysics Facility, Istituto Italiano di
Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Rosalia Bertorelli
- Translational
Pharmacology Facility, Istituto Italiano
di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Andrea Armirotti
- Analytical
Chemistry Facility, Istituto Italiano di
Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Anand K. Ganesan
- Department
of Dermatology, University of California, Irvine, California 92697, United States
| | - Marco De Vivo
- Molecular
Modeling and Drug Discovery Lab, Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| |
Collapse
|
3
|
Frere GA, Hasabnis A, Francisco CB, Suleiman M, Alimowska O, Rahmatullah R, Gould J, Su CYC, Voznyy O, Gunning PT, Basso EA, Prosser RS. Next-Generation Tags for Fluorine Nuclear Magnetic Resonance: Designing Amplification of Chemical Shift Sensitivity. J Am Chem Soc 2024; 146:3052-3064. [PMID: 38279916 DOI: 10.1021/jacs.3c09730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2024]
Abstract
Fluorine NMR is a highly sensitive technique for delineating the conformational states of biomolecules and has shown great utility in drug screening and in understanding protein function. Current fluorinated protein tags leverage the intrinsic chemical shift sensitivity of the 19F nucleus to detect subtle changes in protein conformation and topology. This chemical shift sensitivity can be amplified by embedding the fluorine or trifluoromethyl reporter within a pyridone. Due to their polarizability and rapid tautomerization, pyridones exhibit a greater range of electron delocalization and correspondingly greater 19F NMR chemical shift dispersion. To assess the chemical shift sensitivity of these tautomeric probes to the local environment, 19F NMR spectra of all possible monofluorinated and trifluoromethyl-tagged versions of 2-pyridone were recorded in methanol/water mixtures ranging from 100% methanol to 100% water. 4-Fluoro-2-pyridone and 6-(trifluoromethyl)-2-pyridone (6-TFP) displayed the greatest sensitivity of the monofluorinated and trifluoromethylated pyridones, exceeding that of known conventional CF3 reporters. To evaluate the utility of tautomeric pyridone tags for 19F NMR of biomolecules, the alpha subunit of the stimulatory G protein (Gsα) and human serum albumin (HSA) were each labeled with a thiol-reactive derivative of 6-TFP and the spectra were recorded as a function of various adjuvants and drugs. The tautomeric tag outperformed the conventional tag, 2-bromo-N-(4-(trifluoromethyl)phenyl)acetamide through the improved resolution of several functional states.
Collapse
Affiliation(s)
- Geordon A Frere
- Department of Chemistry, University of Toronto, CPS, UTM, 3359 Mississauga Rd, Mississauga, ON L5L 1C6, Canada
| | - Advait Hasabnis
- Department of Chemistry, University of Toronto, CPS, UTM, 3359 Mississauga Rd, Mississauga, ON L5L 1C6, Canada
| | - Camila B Francisco
- Department of Chemistry, University of Toronto, CPS, UTM, 3359 Mississauga Rd, Mississauga, ON L5L 1C6, Canada
- Department of Chemistry, State University of Maringá, 5790, Maringá 87020-900, Brazil
| | - Motasem Suleiman
- Department of Chemistry, University of Toronto, CPS, UTM, 3359 Mississauga Rd, Mississauga, ON L5L 1C6, Canada
| | - Olga Alimowska
- Department of Chemistry, University of Toronto, CPS, UTM, 3359 Mississauga Rd, Mississauga, ON L5L 1C6, Canada
| | - Rima Rahmatullah
- Department of Chemistry, University of Toronto, CPS, UTM, 3359 Mississauga Rd, Mississauga, ON L5L 1C6, Canada
| | - Jerome Gould
- Department of Chemistry, University of Toronto, CPS, UTM, 3359 Mississauga Rd, Mississauga, ON L5L 1C6, Canada
| | - Celia Yi-Chia Su
- Department of Chemistry, University of Toronto, CPS, UTM, 3359 Mississauga Rd, Mississauga, ON L5L 1C6, Canada
| | - Oleksandr Voznyy
- Department of Chemistry, University of Toronto, CPS, UTM, 3359 Mississauga Rd, Mississauga, ON L5L 1C6, Canada
| | - Patrick T Gunning
- Department of Chemistry, University of Toronto, CPS, UTM, 3359 Mississauga Rd, Mississauga, ON L5L 1C6, Canada
| | - Ernani A Basso
- Department of Chemistry, State University of Maringá, 5790, Maringá 87020-900, Brazil
| | - Robert S Prosser
- Department of Chemistry, University of Toronto, CPS, UTM, 3359 Mississauga Rd, Mississauga, ON L5L 1C6, Canada
| |
Collapse
|
4
|
Myers SH, Poppi L, Rinaldi F, Veronesi M, Ciamarone A, Previtali V, Bagnolini G, Schipani F, Ortega Martínez JA, Girotto S, Di Stefano G, Farabegoli F, Walsh N, De Franco F, Roberti M, Cavalli A. An 19F NMR fragment-based approach for the discovery and development of BRCA2-RAD51 inhibitors to pursuit synthetic lethality in combination with PARP inhibition in pancreatic cancer. Eur J Med Chem 2024; 265:116114. [PMID: 38194775 DOI: 10.1016/j.ejmech.2023.116114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/29/2023] [Accepted: 12/30/2023] [Indexed: 01/11/2024]
Abstract
The BRCA2-RAD51 interaction remains an intriguing target for cancer drug discovery due to its vital role in DNA damage repair mechanisms, which cancer cells become particularly reliant on. Moreover, RAD51 has many synthetically lethal partners, including PARP1-2, which can be exploited to induce synthetic lethality in cancer. In this study, we established a 19F-NMR-fragment based approach to identify RAD51 binders, leading to two initial hits. A subsequent SAR program identified 46 as a low micromolar inhibitor of the BRCA2-RAD51 interaction. 46 was tested in different pancreatic cancer cell lines, to evaluate its ability to inhibit the homologous recombination DNA repair pathway, mediated by BRCA2-RAD51 and trigger synthetic lethality in combination with the PARP inhibitor talazoparib, through the induction of apoptosis. Moreover, we further analyzed the 46/talazoparib combination in 3D pancreatic cancer models. Overall, 46 showed its potential as a tool to evaluate the RAD51/PARP1-2 synthetic lethality mechanism, along with providing a prospect for further inhibitors development.
Collapse
Affiliation(s)
- Samuel H Myers
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, 16163, Genoa, Italy
| | - Laura Poppi
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Francesco Rinaldi
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, 16163, Genoa, Italy; Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Marina Veronesi
- Structural Biophysics Facility, Istituto Italiano di Tecnologia, 16163, Genoa, Italy; D3 PharmaChemistry, Istituto Italiano di Tecnologia, 16163, Genoa, Italy
| | - Andrea Ciamarone
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, 16163, Genoa, Italy
| | - Viola Previtali
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, 16163, Genoa, Italy
| | - Greta Bagnolini
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Fabrizio Schipani
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, 16163, Genoa, Italy
| | | | - Stefania Girotto
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, 16163, Genoa, Italy; Structural Biophysics Facility, Istituto Italiano di Tecnologia, 16163, Genoa, Italy
| | - Giuseppina Di Stefano
- Department of Surgical and Medical Sciences, University of Bologna, 40126, Bologna, Italy
| | - Fulvia Farabegoli
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Naomi Walsh
- School of Biotechnology, Dublin City University, D09 NR58, Dublin, Ireland
| | | | - Marinella Roberti
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy.
| | - Andrea Cavalli
- Computational and Chemical Biology, Istituto Italiano di Tecnologia, 16163, Genoa, Italy; Swiss Federal Institute of Technology Lausanne (EPFL), Switzerland
| |
Collapse
|
5
|
Soffer A, Viswas SJ, Alon S, Rozenberg N, Peled A, Piro D, Vilenchik D, Akabayov B. MolOptimizer: A Molecular Optimization Toolkit for Fragment-Based Drug Design. Molecules 2024; 29:276. [PMID: 38202859 PMCID: PMC10780997 DOI: 10.3390/molecules29010276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/22/2023] [Accepted: 01/01/2024] [Indexed: 01/12/2024] Open
Abstract
MolOptimizer is a user-friendly computational toolkit designed to streamline the hit-to-lead optimization process in drug discovery. MolOptimizer extracts features and trains machine learning models using a user-provided, labeled, and small-molecule dataset to accurately predict the binding values of new small molecules that share similar scaffolds with the target in focus. Hosted on the Azure web-based server, MolOptimizer emerges as a vital resource, accelerating the discovery and development of novel drug candidates with improved binding properties.
Collapse
Affiliation(s)
- Adam Soffer
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Data Science Research Centre, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Samuel Joshua Viswas
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Data Science Research Centre, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Shahar Alon
- Department of Software Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Nofar Rozenberg
- Department of Software Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Amit Peled
- Department of Software Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Daniel Piro
- Department of Software Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Dan Vilenchik
- School of Computer and Electrical Engineering, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Barak Akabayov
- Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Data Science Research Centre, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| |
Collapse
|
6
|
Larda S, Ayotte Y, Denk MM, Coote P, Heffron G, Bendahan D, Shahout F, Girard N, Iddir M, Bouchard P, Bilodeau F, Woo S, Farmer LJ, LaPlante SR. Robust Strategy for Hit-to-Lead Discovery: NMR for SAR. J Med Chem 2023; 66:13416-13427. [PMID: 37732695 PMCID: PMC10578354 DOI: 10.1021/acs.jmedchem.3c00656] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Indexed: 09/22/2023]
Abstract
Establishing robust structure-activity relationships (SARs) is key to successful drug discovery campaigns, yet it often remains elusive due to screening and hit validation artifacts (false positives and false negatives), which frequently result in unproductive downstream expenditures of time and resources. To address this issue, we developed an integrative biophysics-driven strategy that expedites hit-to-lead discovery, mitigates false positives/negatives and common hit validation errors, and provides a robust approach to obtaining accurate binding and affinity measurements. The advantage of this method is that it vastly improves the clarity and reproducibility for affinity-driven SAR by monitoring and eliminating confounding factors. We demonstrate the ease at which high-quality micromolar binders can be generated from the initial millimolar fragment screening hits against an "undruggable" protein target, HRas.
Collapse
Affiliation(s)
| | - Yann Ayotte
- NMX
Research and Solutions Inc., Laval H7V 5B7, Canada
- INRS
− Centre Armand-Frappier Santé Biotechnologie, Laval H7V 1B7, Canada
| | - Maria M. Denk
- NMX
Research and Solutions Inc., Laval H7V 5B7, Canada
- INRS
− Centre Armand-Frappier Santé Biotechnologie, Laval H7V 1B7, Canada
| | - Paul Coote
- NMX
Research and Solutions Inc., Laval H7V 5B7, Canada
- Harvard
Medical School, Boston, Massachusetts 02115, United States
- Dana-Farber
Cancer Institute, Boston, Massachusetts 02215, United States
| | - Gregory Heffron
- NMX
Research and Solutions Inc., Laval H7V 5B7, Canada
- Harvard
Medical School, Boston, Massachusetts 02115, United States
| | - David Bendahan
- INRS
− Centre Armand-Frappier Santé Biotechnologie, Laval H7V 1B7, Canada
| | - Fatma Shahout
- INRS
− Centre Armand-Frappier Santé Biotechnologie, Laval H7V 1B7, Canada
| | | | - Mustapha Iddir
- INRS
− Centre Armand-Frappier Santé Biotechnologie, Laval H7V 1B7, Canada
| | | | | | - Simon Woo
- NMX
Research and Solutions Inc., Laval H7V 5B7, Canada
- INRS
− Centre Armand-Frappier Santé Biotechnologie, Laval H7V 1B7, Canada
| | - Luc J. Farmer
- NMX
Research and Solutions Inc., Laval H7V 5B7, Canada
- INRS
− Centre Armand-Frappier Santé Biotechnologie, Laval H7V 1B7, Canada
| | - Steven R. LaPlante
- NMX
Research and Solutions Inc., Laval H7V 5B7, Canada
- INRS
− Centre Armand-Frappier Santé Biotechnologie, Laval H7V 1B7, Canada
- Harvard
Medical School, Boston, Massachusetts 02115, United States
| |
Collapse
|
7
|
Abstract
Although fragment-based drug discovery (FBDD) has been successfully implemented and well-explored for protein targets, its feasibility for RNA targets is emerging. Despite the challenges associated with the selective targeting of RNA, efforts to integrate known methods of RNA binder discovery with fragment-based approaches have been fruitful, as a few bioactive ligands have been identified. Here, we review various fragment-based approaches implemented for RNA targets and provide insights into experimental design and outcomes to guide future work in the area. Indeed, investigations surrounding the molecular recognition of RNA by fragments address rather important questions such as the limits of molecular weight that confer selective binding and the physicochemical properties favorable for RNA binding and bioactivity.
Collapse
Affiliation(s)
- Blessy M. Suresh
- UF Scripps Biomedical Research & The Scripps Research Institute, Department of Chemistry, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Amirhossein Taghavi
- UF Scripps Biomedical Research & The Scripps Research Institute, Department of Chemistry, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Jessica L. Childs-Disney
- UF Scripps Biomedical Research & The Scripps Research Institute, Department of Chemistry, 130 Scripps Way, Jupiter, Florida 33458, United States
| | - Matthew D. Disney
- UF Scripps Biomedical Research & The Scripps Research Institute, Department of Chemistry, 130 Scripps Way, Jupiter, Florida 33458, United States
| |
Collapse
|
8
|
Bührmann M, Kallepu S, Warmuth JD, Wiese JN, Ehrt C, Vatheuer H, Hiller W, Seitz C, Levy L, Czodrowski P, Sievers S, Müller MP, Rauh D. Fragtory: Pharmacophore-Focused Design, Synthesis, and Evaluation of an sp 3-Enriched Fragment Library. J Med Chem 2023; 66:6297-6314. [PMID: 37130057 DOI: 10.1021/acs.jmedchem.3c00187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Fragment-based drug discovery has played an important role in medicinal chemistry and pharmaceutical research. Despite numerous demonstrated successes, the limited diversity and overrepresentation of planar, sp2-rich structures in commercial libraries often hamper the full potential of this approach. Hence, the thorough design of screening libraries inevitably determines the probability for meaningful hits and subsequent structural elaboration. Against this background, we present the generation of an exclusive fragment library based on iterative entry nomination by a specifically designed computational workflow: "Fragtory". Following a pharmacophore diversity-driven approach, we used Fragtory in an interdisciplinary academic setting to guide both tailored synthesis efforts and the implementation of in-house compounds to build a curated 288-member library of sp3-enriched fragments. Subsequent NMR screens against a model protein and hit validation by protein crystallography led to the identification of structurally novel ligands that were further characterized by isothermal titration calorimetry, demonstrating the applicability of our experimental approach.
Collapse
Affiliation(s)
- Mike Bührmann
- Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse 4a, Dortmund 44227, Germany
- Drug Discovery Hub Dortmund (DDHD) am Zentrum für integrierte Wirkstoffforschung (ZIW), Dortmund 44227, Germany
| | - Shivakrishna Kallepu
- Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse 4a, Dortmund 44227, Germany
| | - Jonas D Warmuth
- Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse 4a, Dortmund 44227, Germany
| | - Jan N Wiese
- Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse 4a, Dortmund 44227, Germany
| | - Christiane Ehrt
- Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse 4a, Dortmund 44227, Germany
| | - Helge Vatheuer
- Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse 4a, Dortmund 44227, Germany
| | - Wolf Hiller
- Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse 4a, Dortmund 44227, Germany
| | - Carina Seitz
- Max Planck Institute of Molecular Physiology, Compound Management and Screening Center (COMAS), Otto-Hahn-Strasse 11/15, Dortmund 44227, Germany
| | - Laura Levy
- Drug Discovery Hub Dortmund (DDHD) am Zentrum für integrierte Wirkstoffforschung (ZIW), Dortmund 44227, Germany
- Taros Chemicals GmbH & Co. KG, Emil-Figge-Strasse 76a, Dortmund 44227, Germany
| | - Paul Czodrowski
- Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse 4a, Dortmund 44227, Germany
| | - Sonja Sievers
- Drug Discovery Hub Dortmund (DDHD) am Zentrum für integrierte Wirkstoffforschung (ZIW), Dortmund 44227, Germany
- Max Planck Institute of Molecular Physiology, Compound Management and Screening Center (COMAS), Otto-Hahn-Strasse 11/15, Dortmund 44227, Germany
| | - Matthias P Müller
- Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse 4a, Dortmund 44227, Germany
| | - Daniel Rauh
- Department of Chemistry and Chemical Biology, TU Dortmund University, Otto-Hahn-Strasse 4a, Dortmund 44227, Germany
- Drug Discovery Hub Dortmund (DDHD) am Zentrum für integrierte Wirkstoffforschung (ZIW), Dortmund 44227, Germany
| |
Collapse
|
9
|
Brindani N, Vuong LM, Acquistapace IM, La Serra MA, Ortega JA, Veronesi M, Bertozzi SM, Summa M, Girotto S, Bertorelli R, Armirotti A, Ganesan AK, De Vivo M. Design, Synthesis, In Vitro and In Vivo Characterization of CDC42 GTPase Interaction Inhibitors for the Treatment of Cancer. J Med Chem 2023; 66:5981-6001. [PMID: 37026468 PMCID: PMC10150367 DOI: 10.1021/acs.jmedchem.3c00276] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Indexed: 04/08/2023]
Abstract
CDC42 GTPases (RHOJ, CDC42, and RHOQ) are overexpressed in multiple tumor types and activate pathways critical for tumor growth, angiogenesis, and metastasis. Recently, we reported the discovery of a novel lead compound, ARN22089, which blocks the interaction of CDC42 GTPases with specific downstream effectors. ARN22089 blocks tumor growth in BRAF mutant mouse melanoma models and patient-derived xenografts (PDXs) in vivo. ARN22089 also inhibits tumor angiogenesis in three-dimensional vascularized microtumor models in vitro. Notably, ARN22089 belongs to a novel class of trisubstituted pyrimidines. Based on these results, we describe an extensive structure-activity relationship of ∼30 compounds centered on ARN22089. We discovered and optimized two novel inhibitors (27, ARN25062, and 28, ARN24928), which are optimal back-up/follow-up leads with favorable drug-like properties and in vivo efficacy in PDX tumors. These findings further demonstrate the potential of this class of CDC42/RHOJ inhibitors for cancer treatment, with lead candidates ready for advanced preclinical studies.
Collapse
Affiliation(s)
- Nicoletta Brindani
- Molecular
Modeling and Drug Discovery Lab, Istituto
Italiano di Tecnologia, Via Morego 30, Genova 16163, Italy
| | - Linh M. Vuong
- Department
of Dermatology, University of California, Irvine, California 92697, United States
| | - Isabella Maria Acquistapace
- Molecular
Modeling and Drug Discovery Lab, Istituto
Italiano di Tecnologia, Via Morego 30, Genova 16163, Italy
| | - Maria Antonietta La Serra
- Molecular
Modeling and Drug Discovery Lab, Istituto
Italiano di Tecnologia, Via Morego 30, Genova 16163, Italy
| | - José Antonio Ortega
- Molecular
Modeling and Drug Discovery Lab, Istituto
Italiano di Tecnologia, Via Morego 30, Genova 16163, Italy
| | - Marina Veronesi
- Structural
Biophysics Facility, Istituto Italiano di
Tecnologia, Via Morego
30, Genova 16163, Italy
| | - Sine Mandrup Bertozzi
- Analytical
Chemistry Facility, Istituto Italiano di
Tecnologia, Via Morego
30, Genova 16163, Italy
| | - Maria Summa
- Translational
Pharmacology Facility, Istituto Italiano
di Tecnologia, Via Morego
30, Genova 16163, Italy
| | - Stefania Girotto
- Structural
Biophysics Facility, Istituto Italiano di
Tecnologia, Via Morego
30, Genova 16163, Italy
| | - Rosalia Bertorelli
- Translational
Pharmacology Facility, Istituto Italiano
di Tecnologia, Via Morego
30, Genova 16163, Italy
| | - Andrea Armirotti
- Analytical
Chemistry Facility, Istituto Italiano di
Tecnologia, Via Morego
30, Genova 16163, Italy
| | - Anand K. Ganesan
- Department
of Dermatology, University of California, Irvine, California 92697, United States
| | - Marco De Vivo
- Molecular
Modeling and Drug Discovery Lab, Istituto
Italiano di Tecnologia, Via Morego 30, Genova 16163, Italy
| |
Collapse
|
10
|
Chang Y, Hawkins BA, Du JJ, Groundwater PW, Hibbs DE, Lai F. A Guide to In Silico Drug Design. Pharmaceutics 2022; 15:pharmaceutics15010049. [PMID: 36678678 PMCID: PMC9867171 DOI: 10.3390/pharmaceutics15010049] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/16/2022] [Accepted: 12/17/2022] [Indexed: 12/28/2022] Open
Abstract
The drug discovery process is a rocky path that is full of challenges, with the result that very few candidates progress from hit compound to a commercially available product, often due to factors, such as poor binding affinity, off-target effects, or physicochemical properties, such as solubility or stability. This process is further complicated by high research and development costs and time requirements. It is thus important to optimise every step of the process in order to maximise the chances of success. As a result of the recent advancements in computer power and technology, computer-aided drug design (CADD) has become an integral part of modern drug discovery to guide and accelerate the process. In this review, we present an overview of the important CADD methods and applications, such as in silico structure prediction, refinement, modelling and target validation, that are commonly used in this area.
Collapse
Affiliation(s)
- Yiqun Chang
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Bryson A. Hawkins
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Jonathan J. Du
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Paul W. Groundwater
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - David E. Hibbs
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Felcia Lai
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2006, Australia
- Correspondence:
| |
Collapse
|
11
|
Moinul M, Khatun S, Amin SA, Jha T, Gayen S. Recent trends in fragment-based anticancer drug design strategies against different targets: A mini-review. Biochem Pharmacol 2022; 206:115301. [DOI: 10.1016/j.bcp.2022.115301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/07/2022] [Accepted: 10/10/2022] [Indexed: 11/02/2022]
|
12
|
McAulay K, Bilsland A, Bon M. Reactivity of Covalent Fragments and Their Role in Fragment Based Drug Discovery. Pharmaceuticals (Basel) 2022; 15:1366. [PMID: 36355538 PMCID: PMC9694498 DOI: 10.3390/ph15111366] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/30/2022] [Accepted: 11/04/2022] [Indexed: 09/27/2023] Open
Abstract
Fragment based drug discovery has long been used for the identification of new ligands and interest in targeted covalent inhibitors has continued to grow in recent years, with high profile drugs such as osimertinib and sotorasib gaining FDA approval. It is therefore unsurprising that covalent fragment-based approaches have become popular and have recently led to the identification of novel targets and binding sites, as well as ligands for targets previously thought to be 'undruggable'. Understanding the properties of such covalent fragments is important, and characterizing and/or predicting reactivity can be highly useful. This review aims to discuss the requirements for an electrophilic fragment library and the importance of differing warhead reactivity. Successful case studies from the world of drug discovery are then be examined.
Collapse
Affiliation(s)
- Kirsten McAulay
- Cancer Research Horizons—Therapeutic Innovation, Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
- Centre for Targeted Protein Degradation, University of Dundee, Nethergate, Dundee DD1 4HN, UK
| | - Alan Bilsland
- Cancer Research Horizons—Therapeutic Innovation, Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Marta Bon
- Cancer Research Horizons—Therapeutic Innovation, Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
- Exscientia, The Schrödinger Building, Oxford Science Park, Oxford OX4 4GE, UK
| |
Collapse
|
13
|
Bagnolini G, Balboni B, Schipani F, Gioia D, Veronesi M, De Franco F, Kaya C, Jumde RP, Ortega JA, Girotto S, Hirsch AKH, Roberti M, Cavalli A. Identification of RAD51–BRCA2 Inhibitors Using N-Acylhydrazone-Based Dynamic Combinatorial Chemistry. ACS Med Chem Lett 2022; 13:1262-1269. [PMID: 35978685 PMCID: PMC9377020 DOI: 10.1021/acsmedchemlett.2c00063] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 07/22/2022] [Indexed: 11/29/2022] Open
Abstract
![]()
RAD51 is an ATP-dependent recombinase, recruited by BRCA2
to mediate
DNA double-strand breaks repair through homologous recombination and
represents an attractive cancer drug target. Herein, we applied for
the first-time protein-templated dynamic combinatorial chemistry on
RAD51 as a hit identification strategy. Upon design of N-acylhydrazone-based dynamic combinatorial libraries, RAD51 showed
a clear templating effect, amplifying 19 N-acylhydrazones.
Screening against the RAD51–BRCA2 protein–protein interaction
via ELISA assay afforded 10 inhibitors in the micromolar range. Further 19F NMR experiments revealed that 7 could bind
RAD51 and be displaced by BRC4, suggesting an interaction in the same
binding pocket of BRCA2. These results proved not only that ptDCC
could be successfully applied on full-length oligomeric RAD51, but
also that it could address the need of alternative strategies toward
the identification of small-molecule PPI inhibitors.
Collapse
Affiliation(s)
- Greta Bagnolini
- Computational & Chemical Biology (CCB), Istituto Italiano di Tecnologia (IIT), 16163 Genova, Italy
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, 40126 Bologna, Italy
- Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany
| | - Beatrice Balboni
- Computational & Chemical Biology (CCB), Istituto Italiano di Tecnologia (IIT), 16163 Genova, Italy
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, 40126 Bologna, Italy
| | - Fabrizio Schipani
- Computational & Chemical Biology (CCB), Istituto Italiano di Tecnologia (IIT), 16163 Genova, Italy
| | - Dario Gioia
- Computational & Chemical Biology (CCB), Istituto Italiano di Tecnologia (IIT), 16163 Genova, Italy
| | - Marina Veronesi
- Structural Biophysics and Translational Pharmacology, Istituto Italiano di Tecnologia (IIT), 16163 Genova, Italy
- D3-PharmaChemistry, Istituto Italiano di Tecnologia (IIT), 16163 Genova, Italy
| | | | - Cansu Kaya
- Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Ravindra P. Jumde
- Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany
| | - Jose Antonio Ortega
- Computational & Chemical Biology (CCB), Istituto Italiano di Tecnologia (IIT), 16163 Genova, Italy
| | - Stefania Girotto
- Computational & Chemical Biology (CCB), Istituto Italiano di Tecnologia (IIT), 16163 Genova, Italy
| | - Anna K. H. Hirsch
- Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), 66123 Saarbrücken, Germany
- Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany
| | - Marinella Roberti
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, 40126 Bologna, Italy
| | - Andrea Cavalli
- Computational & Chemical Biology (CCB), Istituto Italiano di Tecnologia (IIT), 16163 Genova, Italy
- Department of Pharmacy and Biotechnology (FaBiT), University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
14
|
Rüdisser SH, Goldberg N, Ebert MO, Kovacs H, Gossert AD. Efficient affinity ranking of fluorinated ligands by 19F NMR: CSAR and FastCSAR. JOURNAL OF BIOMOLECULAR NMR 2020; 74:579-594. [PMID: 32556806 DOI: 10.1007/s10858-020-00325-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 06/02/2020] [Indexed: 06/11/2023]
Abstract
Fluorine NMR has recently gained high popularity in drug discovery as it allows efficient and sensitive screening of large numbers of ligands. However, the positive hits found in screening must subsequently be ranked according to their affinity in order to prioritize them for follow-up chemistry. Unfortunately, the primary read-out from the screening experiments, namely the increased relaxation rate upon binding, is not proportional to the affinity of the ligand, as it is polluted by effects such as exchange broadening. Here we present the method CSAR (Chemical Shift-anisotropy-based Affinity Ranking) for reliable ranking of fluorinated ligands by NMR, without the need of isotope labeled protein, titrations or setting up a reporter format. Our strategy is to produce relaxation data that is directly proportional to the binding affinity. This is achieved by removing all other contributions to relaxation as follows: (i) exchange effects are efficiently suppressed by using high power spin lock pulses, (ii) dipolar relaxation effects are approximately subtracted by measuring at two different magnetic fields and (iii) differences in chemical shift anisotropy are normalized using calculated values. A similar ranking can be obtained with the simplified approach FastCSAR that relies on a measurement of a single relaxation experiment at high field (preferably > 600 MHz). An affinity ranking obtained in this simple way will enable prioritizing ligands and thus improve the efficiency of fragment-based drug design.
Collapse
Affiliation(s)
- Simon H Rüdisser
- Institute for Molecular Biology and Biophysics, ETH Zürich, 8093, Zürich, Switzerland
- Biomolecular NMR Spectroscopy Platform, ETH Zürich, 8093, Zürich, Switzerland
| | - Nils Goldberg
- Institute for Molecular Biology and Biophysics, ETH Zürich, 8093, Zürich, Switzerland
- Biomolecular NMR Spectroscopy Platform, ETH Zürich, 8093, Zürich, Switzerland
| | - Marc-Olivier Ebert
- Laboratorium für Organische Chemie, ETH Zürich, 8093, Zürich, Switzerland
| | | | - Alvar D Gossert
- Institute for Molecular Biology and Biophysics, ETH Zürich, 8093, Zürich, Switzerland.
- Biomolecular NMR Spectroscopy Platform, ETH Zürich, 8093, Zürich, Switzerland.
| |
Collapse
|
15
|
Competition NMR for Detection of Hit/Lead Inhibitors of Protein-Protein Interactions. Molecules 2020; 25:molecules25133017. [PMID: 32630327 PMCID: PMC7412237 DOI: 10.3390/molecules25133017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 06/28/2020] [Accepted: 06/29/2020] [Indexed: 12/19/2022] Open
Abstract
Screening for small-molecule fragments that can lead to potent inhibitors of protein–protein interactions (PPIs) is often a laborious step as the fragments cannot dissociate the targeted PPI due to their low μM–mM affinities. Here, we describe an NMR competition assay called w-AIDA-NMR (weak-antagonist induced dissociation assay-NMR), which is sensitive to weak μM–mM ligand–protein interactions and which can be used in initial fragment screening campaigns. By introducing point mutations in the complex’s protein that is not targeted by the inhibitor, we lower the effective affinity of the complex, allowing for short fragments to dissociate the complex. We illustrate the method with the compounds that block the Mdm2/X-p53 and PD-1/PD-L1 oncogenic interactions. Targeting the PD-/PD-L1 PPI has profoundly advanced the treatment of different types of cancers.
Collapse
|
16
|
Brancaccio D, Di Maro S, Cerofolini L, Giuntini S, Fragai M, Luchinat C, Tomassi S, Limatola A, Russomanno P, Merlino F, Novellino E, Carotenuto A. HOPPI-NMR: Hot-Peptide-Based Screening Assay for Inhibitors of Protein-Protein Interactions by NMR. ACS Med Chem Lett 2020; 11:1047-1053. [PMID: 32435424 DOI: 10.1021/acsmedchemlett.9b00620] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/20/2020] [Indexed: 12/23/2022] Open
Abstract
Protein-protein interactions (PPIs) contribute to the onset and/or progression of several diseases, especially cancer, and this discovery has paved the way for considering disruption of the PPIs as an attractive anti-tumor strategy. In this regard, simple and efficient biophysical methods for detecting the interaction of the inhibitors with the protein counterpart are still in high demand. Herein, we describe a convenient NMR method for the screening of putative PPI inhibitors based on the use of "hot peptides" (HOPPI-NMR). As a case study, HOPPI-NMR was successful applied to the well-known p53/MDM2 system. Our outcomes highlight the main advantages of the method, including the use of a small amount of unlabeled proteins, the minimization of the risk of protein aggregation, and the ability to identify weak binders. The last leaves open the possibility for application of HOPPI-NMR in tandem with fragment-based drug discovery as a valid strategy for the identification of novel chemotypes acting as PPI inhibitors.
Collapse
Affiliation(s)
- Diego Brancaccio
- Department of Pharmacy, University of Naples “Federico II”, 80131 Naples, Italy
| | - Salvatore Di Maro
- DISTABIF, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy
| | | | | | - Marco Fragai
- CERM, University of Florence, 50019 Sesto Fiorentino, Italy
| | | | - Stefano Tomassi
- Department of Pharmacy, University of Naples “Federico II”, 80131 Naples, Italy
| | - Antonio Limatola
- Department of Biology, Stanford University, Stanford, California 94305-5430, United States
| | - Pasquale Russomanno
- Department of Pharmacy, University of Naples “Federico II”, 80131 Naples, Italy
| | - Francesco Merlino
- Department of Pharmacy, University of Naples “Federico II”, 80131 Naples, Italy
| | - Ettore Novellino
- Department of Pharmacy, University of Naples “Federico II”, 80131 Naples, Italy
| | - Alfonso Carotenuto
- Department of Pharmacy, University of Naples “Federico II”, 80131 Naples, Italy
| |
Collapse
|
17
|
Coyle J, Walser R. Applied Biophysical Methods in Fragment-Based Drug Discovery. SLAS DISCOVERY 2020; 25:471-490. [PMID: 32345095 DOI: 10.1177/2472555220916168] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Fragment-based drug discovery (FBDD) has come of age in the last decade with the FDA approval of four fragment-derived drugs. Biophysical methods are at the heart of hit discovery and validation in FBDD campaigns. The three most commonly used methods, thermal shift, surface plasmon resonance, and nuclear magnetic resonance, can be daunting for the novice user. We aim here to provide the nonexpert user of these methods with a summary of problems and challenges that might be faced, but also highlight the potential gains that each method can contribute to an FBDD project. While our view on FBDD is slightly biased toward enabling structure-guided drug discovery, most of the points we address in this review are also valid for non-structure-focused FBDD.
Collapse
Affiliation(s)
- Joe Coyle
- Astex Pharmaceuticals, Cambridge, UK
| | | |
Collapse
|
18
|
Keeley A, Petri L, Ábrányi-Balogh P, Keserű GM. Covalent fragment libraries in drug discovery. Drug Discov Today 2020; 25:983-996. [PMID: 32298798 DOI: 10.1016/j.drudis.2020.03.016] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 03/07/2020] [Accepted: 03/23/2020] [Indexed: 12/20/2022]
Abstract
Targeted covalent inhibitors and chemical probes have become integral parts of drug discovery approaches. Given the advantages of fragment-based drug discovery, screening electrophilic fragments emerged as a promising alternative to discover and validate novel targets and to generate viable chemical starting points even for targets that are barely tractable. In this review, we present recent principles and considerations in the design of electrophilic fragment libraries from the selection of the appropriate covalent warhead through the design of the covalent fragment to the compilation of the library. We then summarize recent screening methodologies of covalent fragments against surrogate models, proteins, and the whole proteome, or living cells. Finally, we highlight recent drug discovery applications of covalent fragment libraries.
Collapse
Affiliation(s)
- Aaron Keeley
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
| | - László Petri
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
| | - Péter Ábrányi-Balogh
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar tudósok krt. 2, H-1117 Budapest, Hungary
| | - György M Keserű
- Medicinal Chemistry Research Group, Research Centre for Natural Sciences, Magyar tudósok krt. 2, H-1117 Budapest, Hungary.
| |
Collapse
|
19
|
Ermakova EA, Danilova AG, Khairutdinov BI. Interaction of ceftriaxone and rutin with human serum albumin. WaterLOGSY-NMR and molecular docking study. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2019.127444] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
20
|
Almawi AW, Langlois-Lemay L, Boulton S, Rodríguez González J, Melacini G, D'Amours D, Guarné A. Distinct surfaces on Cdc5/PLK Polo-box domain orchestrate combinatorial substrate recognition during cell division. Sci Rep 2020; 10:3379. [PMID: 32099015 PMCID: PMC7042354 DOI: 10.1038/s41598-020-60344-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 02/10/2020] [Indexed: 11/19/2022] Open
Abstract
Polo-like kinases (Plks) are key cell cycle regulators. They contain a kinase domain followed by a polo-box domain that recognizes phosphorylated substrates and enhances their phosphorylation. The regulatory subunit of the Dbf4-dependent kinase complex interacts with the polo-box domain of Cdc5 (the sole Plk in Saccharomyces cerevisiae) in a phosphorylation-independent manner. We have solved the crystal structures of the polo-box domain of Cdc5 on its own and in the presence of peptides derived from Dbf4 and a canonical phosphorylated substrate. The structure bound to the Dbf4-peptide reveals an additional density on the surface opposite to the phospho-peptide binding site that allowed us to propose a model for the interaction. We found that the two peptides can bind simultaneously and non-competitively to the polo-box domain in solution. Furthermore, point mutations on the surface opposite to the phosphopeptide binding site of the polo-box domain disrupt the interaction with the Dbf4 peptide in solution and cause an early anaphase arrest phenotype distinct from the mitotic exit defect typically observed in cdc5 mutants. Collectively, our data illustrates the importance of non-canonical interactions mediated by the polo-box domain and provide key mechanistic insights into the combinatorial recognition of substrates by Polo-like kinases.
Collapse
Affiliation(s)
- Ahmad W Almawi
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
- IniXium, 500 Boulevard Cartier Ouest, Laval, QC, Canada
| | - Laurence Langlois-Lemay
- Ottawa Institute of Systems Biology, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Stephen Boulton
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON, Canada
| | | | - Giuseppe Melacini
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON, Canada
| | - Damien D'Amours
- Ottawa Institute of Systems Biology, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
| | - Alba Guarné
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada.
- Department of Biochemistry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
21
|
Khattri RB, Morris DL, Bilinovich SM, Manandhar E, Napper KR, Sweet JW, Modarelli DA, Leeper TC. Identifying Ortholog Selective Fragment Molecules for Bacterial Glutaredoxins by NMR and Affinity Enhancement by Modification with an Acrylamide Warhead. Molecules 2019; 25:E147. [PMID: 31905878 PMCID: PMC6983068 DOI: 10.3390/molecules25010147] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 12/30/2022] Open
Abstract
Illustrated here is the development of a new class of antibiotic lead molecules targeted at Pseudomonas aeruginosa glutaredoxin (PaGRX). This lead was produced to (a) circumvent efflux-mediated resistance mechanisms via covalent inhibition while (b) taking advantage of species selectivity to target a fundamental metabolic pathway. This work involved four components: a novel workflow for generating protein specific fragment hits via independent nuclear magnetic resonance (NMR) measurements, NMR-based modeling of the target protein structure, NMR guided docking of hits, and synthetic modification of the fragment hit with a vinyl cysteine trap moiety, i.e., acrylamide warhead, to generate the chimeric lead. Reactivity of the top warhead-fragment lead suggests that the ortholog selectivity observed for a fragment hit can translate into a substantial kinetic advantage in the mature warhead lead, which bodes well for future work to identify potent, species specific drug molecules targeted against proteins heretofore deemed undruggable.
Collapse
Affiliation(s)
- Ram B. Khattri
- Department of Physiology and Functional genomics, University of Florida, Gainesville, FL 32610, USA;
| | - Daniel L. Morris
- Department of Chemistry and Biochemistry, The University of Akron, Akron, OH 44325, USA; (D.L.M.); (K.R.N.); (J.W.S.); (D.A.M.)
| | - Stephanie M. Bilinovich
- Department of Pediatrics and Human Development, Michigan State University, East Lansing, MI 48824, USA;
| | | | - Kahlilah R. Napper
- Department of Chemistry and Biochemistry, The University of Akron, Akron, OH 44325, USA; (D.L.M.); (K.R.N.); (J.W.S.); (D.A.M.)
| | - Jacob W. Sweet
- Department of Chemistry and Biochemistry, The University of Akron, Akron, OH 44325, USA; (D.L.M.); (K.R.N.); (J.W.S.); (D.A.M.)
| | - David A. Modarelli
- Department of Chemistry and Biochemistry, The University of Akron, Akron, OH 44325, USA; (D.L.M.); (K.R.N.); (J.W.S.); (D.A.M.)
| | - Thomas C. Leeper
- Department of Chemistry and Biochemistry, Kennesaw State University, GA 30144, USA
| |
Collapse
|
22
|
Haymond A, Davis JB, Espina V. Proteomics for cancer drug design. Expert Rev Proteomics 2019; 16:647-664. [PMID: 31353977 PMCID: PMC6736641 DOI: 10.1080/14789450.2019.1650025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 07/26/2019] [Indexed: 12/29/2022]
Abstract
Introduction: Signal transduction cascades drive cellular proliferation, apoptosis, immune, and survival pathways. Proteins have emerged as actionable drug targets because they are often dysregulated in cancer, due to underlying genetic mutations, or dysregulated signaling pathways. Cancer drug development relies on proteomic technologies to identify potential biomarkers, mechanisms-of-action, and to identify protein binding hot spots. Areas covered: Brief summaries of proteomic technologies for drug discovery include mass spectrometry, reverse phase protein arrays, chemoproteomics, and fragment based screening. Protein-protein interface mapping is presented as a promising method for peptide therapeutic development. The topic of biosimilar therapeutics is presented as an opportunity to apply proteomic technologies to this new class of cancer drug. Expert opinion: Proteomic technologies are indispensable for drug discovery. A suite of technologies including mass spectrometry, reverse phase protein arrays, and protein-protein interaction mapping provide complimentary information for drug development. These assays have matured into well controlled, robust technologies. Recent regulatory approval of biosimilar therapeutics provides another opportunity to decipher the molecular nuances of their unique mechanisms of action. The ability to identify previously hidden protein hot spots is expanding the gamut of potential drug targets. Proteomic profiling permits lead compound evaluation beyond the one drug, one target paradigm.
Collapse
Affiliation(s)
- Amanda Haymond
- Center for Applied Proteomics and Molecular Medicine, George Mason University , Manassas , VA , USA
| | - Justin B Davis
- Center for Applied Proteomics and Molecular Medicine, George Mason University , Manassas , VA , USA
| | - Virginia Espina
- Center for Applied Proteomics and Molecular Medicine, George Mason University , Manassas , VA , USA
| |
Collapse
|
23
|
Dalvit C, Parent A, Vallée F, Mathieu M, Rak A. Fast NMR Methods for Measuring in the Direct and/or Competition Mode the Dissociation Constants of Chemical Fragments Interacting with a Receptor. ChemMedChem 2019; 14:1115-1127. [DOI: 10.1002/cmdc.201900152] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Indexed: 12/16/2022]
Affiliation(s)
| | - Annick Parent
- Bio Structure and BiophysicsIntegrated Drug DiscoverySanofi R&D 13, Quai Jules Guesde—BP 14 94403 Vitry sur Seine Cedex France
| | - Francois Vallée
- Bio Structure and BiophysicsIntegrated Drug DiscoverySanofi R&D 13, Quai Jules Guesde—BP 14 94403 Vitry sur Seine Cedex France
| | - Magali Mathieu
- Bio Structure and BiophysicsIntegrated Drug DiscoverySanofi R&D 13, Quai Jules Guesde—BP 14 94403 Vitry sur Seine Cedex France
| | - Alexey Rak
- Bio Structure and BiophysicsIntegrated Drug DiscoverySanofi R&D 13, Quai Jules Guesde—BP 14 94403 Vitry sur Seine Cedex France
| |
Collapse
|
24
|
Cerofolini L, Giuntini S, Barbieri L, Pennestri M, Codina A, Fragai M, Banci L, Luchinat E, Ravera E. Real-Time Insights into Biological Events: In-Cell Processes and Protein-Ligand Interactions. Biophys J 2019; 116:239-247. [PMID: 30580921 PMCID: PMC6350048 DOI: 10.1016/j.bpj.2018.11.3132] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 10/30/2018] [Accepted: 11/27/2018] [Indexed: 11/15/2022] Open
Abstract
FlowNMR has the aim of continuously monitoring processes that occur in conditions that are not compatible with being carried out within a closed tube. However, it is sample intensive and not suitable for samples, such as proteins or living cells, that are often available in limited volumes and possibly low concentrations. We here propose a dialysis-based modification of a commercial flowNMR setup that allows for recycling the medium while confining the sample (proteins and cells) within the active volume of the tube. This approach is demonstrated in the specific cases of in-cell NMR and protein-based ligand studies.
Collapse
Affiliation(s)
- Linda Cerofolini
- Magnetic Resonance Center, University of Florence and Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine, Sesto Fiorentino, Italy
| | - Stefano Giuntini
- Magnetic Resonance Center, University of Florence and Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine, Sesto Fiorentino, Italy; Department of Chemistry, Ugo Schiff, University of Florence, Sesto Fiorentino, Italy
| | - Letizia Barbieri
- Magnetic Resonance Center, University of Florence and Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine, Sesto Fiorentino, Italy
| | | | - Anna Codina
- Bruker UK Limited, Banner Lane, Coventry, United Kingdom
| | - Marco Fragai
- Magnetic Resonance Center, University of Florence and Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine, Sesto Fiorentino, Italy; Department of Chemistry, Ugo Schiff, University of Florence, Sesto Fiorentino, Italy
| | - Lucia Banci
- Magnetic Resonance Center, University of Florence and Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine, Sesto Fiorentino, Italy; Department of Chemistry, Ugo Schiff, University of Florence, Sesto Fiorentino, Italy
| | - Enrico Luchinat
- Magnetic Resonance Center, University of Florence and Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine, Sesto Fiorentino, Italy; Department of Experimental and Clinical Biomedical Sciences, Mario Serio, University of Florence, Florence, Italy.
| | - Enrico Ravera
- Magnetic Resonance Center, University of Florence and Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine, Sesto Fiorentino, Italy; Department of Chemistry, Ugo Schiff, University of Florence, Sesto Fiorentino, Italy.
| |
Collapse
|
25
|
Polshakov VI, Batuev EA, Mantsyzov AB. NMR screening and studies of target–ligand interactions. RUSSIAN CHEMICAL REVIEWS 2019. [DOI: 10.1070/rcr4836] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
26
|
Dalvit C, Vulpetti A. Ligand-Based Fluorine NMR Screening: Principles and Applications in Drug Discovery Projects. J Med Chem 2018; 62:2218-2244. [DOI: 10.1021/acs.jmedchem.8b01210] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
| | - Anna Vulpetti
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, 4002 Basel, Switzerland
| |
Collapse
|
27
|
Gervais V, Muller I, Mari PO, Mourcet A, Movellan KT, Ramos P, Marcoux J, Guillet V, Javaid S, Burlet-Schiltz O, Czaplicki G, Milon A, Giglia-Mari G. Small molecule-based targeting of TTD-A dimerization to control TFIIH transcriptional activity represents a potential strategy for anticancer therapy. J Biol Chem 2018; 293:14974-14988. [PMID: 30068551 DOI: 10.1074/jbc.ra118.003444] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/25/2018] [Indexed: 11/06/2022] Open
Abstract
The human transcription factor TFIIH is a large complex composed of 10 subunits that form an intricate network of protein-protein interactions critical for regulating its transcriptional and DNA repair activities. The trichothiodystrophy group A protein (TTD-A or p8) is the smallest TFIIH subunit, shuttling between a free and a TFIIH-bound state. Its dimerization properties allow it to shift from a homodimeric state, in the absence of a functional partner, to a heterodimeric structure, enabling dynamic binding to TFIIH. Recruitment of p8 at TFIIH stabilizes the overall architecture of the complex, whereas p8's absence reduces its cellular steady-state concentration and consequently decreases basal transcription, highlighting that p8 dimerization may be an attractive target for down-regulating transcription in cancer cells. Here, using a combination of molecular dynamics simulations to study p8 conformational stability and a >3000-member library of chemical fragments, we identified small-molecule compounds that bind to the dimerization interface of p8 and provoke its destabilization, as assessed by biophysical studies. Using quantitative imaging of TFIIH in living mouse cells, we found that these molecules reduce the intracellular concentration of TFIIH and its transcriptional activity to levels similar to that observed in individuals with trichothiodystrophy owing to mutated TTD-A Our results provide a proof of concept of fragment-based drug discovery, demonstrating the utility of small molecules for targeting p8 dimerization to modulate the transcriptional machinery, an approach that may help inform further development in anticancer therapies.
Collapse
Affiliation(s)
- Virginie Gervais
- From the Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, Université Paul Sabatier, BP-64182, F-31077 Toulouse, France,
| | - Isabelle Muller
- From the Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, Université Paul Sabatier, BP-64182, F-31077 Toulouse, France
| | - Pierre-Olivier Mari
- the Université Claude Bernard Lyon 1, INSERM U1217, Institut NeuroMyoGène, CNRS UMR 5310, F-69008 Lyon, France, and
| | - Amandine Mourcet
- From the Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, Université Paul Sabatier, BP-64182, F-31077 Toulouse, France
| | - Kumar Tekwani Movellan
- From the Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, Université Paul Sabatier, BP-64182, F-31077 Toulouse, France
| | - Pascal Ramos
- From the Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, Université Paul Sabatier, BP-64182, F-31077 Toulouse, France
| | - Julien Marcoux
- From the Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, Université Paul Sabatier, BP-64182, F-31077 Toulouse, France
| | - Valérie Guillet
- From the Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, Université Paul Sabatier, BP-64182, F-31077 Toulouse, France
| | - Sumaira Javaid
- From the Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, Université Paul Sabatier, BP-64182, F-31077 Toulouse, France.,the Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center of Chemical and Biological Sciences, University of Karachi, Karachi-75270, Pakistan
| | - Odile Burlet-Schiltz
- From the Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, Université Paul Sabatier, BP-64182, F-31077 Toulouse, France
| | - Georges Czaplicki
- From the Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, Université Paul Sabatier, BP-64182, F-31077 Toulouse, France
| | - Alain Milon
- From the Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de Toulouse, CNRS, Université Paul Sabatier, BP-64182, F-31077 Toulouse, France
| | - Giuseppina Giglia-Mari
- the Université Claude Bernard Lyon 1, INSERM U1217, Institut NeuroMyoGène, CNRS UMR 5310, F-69008 Lyon, France, and
| |
Collapse
|
28
|
Luise N, Wyatt PG. Generation of Polar Semi-Saturated Bicyclic Pyrazoles for Fragment-Based Drug-Discovery Campaigns. Chemistry 2018; 24:10443-10451. [PMID: 29732638 DOI: 10.1002/chem.201801313] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/27/2018] [Indexed: 12/13/2022]
Abstract
Synthesising polar semi-saturated bicyclic heterocycles can lead to better starting points for fragment-based drug discovery (FBDD) programs. We report the application of diverse chemistry to construct bicyclic systems from a common intermediate, where pyrazole, a privileged heteroaromatic able to bind effectively to biological targets, is fused to diverse saturated counterparts. The generated fragments can be further developed either after confirmation of their binding pose or early in the process, as their synthetic intermediates. Essential quality control (QC) for selection of small molecules to add to a fragment library is discussed.
Collapse
Affiliation(s)
- Nicola Luise
- Drug Discovery Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, Scotland, UK
| | - Paul G Wyatt
- Drug Discovery Unit, School of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, Scotland, UK
| |
Collapse
|
29
|
Tomala MD, Magiera-Mularz K, Kubica K, Krzanik S, Zieba B, Musielak B, Pustula M, Popowicz GM, Sattler M, Dubin G, Skalniak L, Holak TA. Identification of small-molecule inhibitors of USP2a. Eur J Med Chem 2018. [PMID: 29529503 DOI: 10.1016/j.ejmech.2018.03.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
USP2a is a deubiquitinating protease that rescues its target proteins from destruction by the proteasome by reversing the process of protein ubiquitination. USP2a shows oncogenic properties in vivo and has been found to be a specific activator of cyclin D1. Many types of cancers are addicted to cyclin D1 expression. Targeting USP2a is a promising strategy for cancer therapy but little progress has been made in the field of inhibition of USP2a. Using NMR-based fragment screening and biophysical binding assays, we have discovered small molecules that bind to USP2a. Iterations of fragment combination and structure-driven design identified two 5-(2-thienyl)-3-isoxazoles as the inhibitors of the USP2a-ubiquitin protein-protein interaction. The affinity of these molecules for the catalytic domain of USP2a parallels their ability to interfere with USP2a binding to ubiquitin in vitro. Altogether, our results establish the 5-(2-thienyl)-3-isoxazole pharmacophore as an attractive starting point for lead optimization.
Collapse
Affiliation(s)
- Marcin D Tomala
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Krakow, Poland
| | | | - Katarzyna Kubica
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Krakow, Poland
| | - Sylwia Krzanik
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Bartosz Zieba
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Bogdan Musielak
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Krakow, Poland
| | - Marcin Pustula
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Krakow, Poland
| | - Grzegorz M Popowicz
- Institute of Structural Biology, Helmholtz Zentrum München, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Michael Sattler
- Institute of Structural Biology, Helmholtz Zentrum München, Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Grzegorz Dubin
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Krakow, Poland
| | - Lukasz Skalniak
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Krakow, Poland
| | - Tad A Holak
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387, Krakow, Poland.
| |
Collapse
|
30
|
Application of Heteronuclear NMR Spectroscopy to Bioinorganic and Medicinal Chemistry ☆. REFERENCE MODULE IN CHEMISTRY, MOLECULAR SCIENCES AND CHEMICAL ENGINEERING 2018. [PMCID: PMC7157447 DOI: 10.1016/b978-0-12-409547-2.10947-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
31
|
Abstract
Over the past few decades, NMR spectroscopy has become an established tool in drug discovery. This communication will highlight the potential of NMR spectroscopy as a method for identification of problematic compounds and as a valuable aid toward revealing some mechanisms of promiscuous behavior. NMR methods for detecting false positives will be analyzed on the basis of their performance, strengths, limitations, and potential pitfalls. Additionally, this communication aims to provide an insight into the limitations of NMR-based methodologies applied to ligand screening in the context of false-positive hits.
Collapse
Affiliation(s)
- Anamarija Zega
- Faculty of Pharmacy, University of Ljubljana , Aškerčeva 7, 1000 Ljubljana, Slovenia
| |
Collapse
|
32
|
Wang H, Rempel DL, Giblin D, Frieden C, Gross ML. Peptide-Level Interactions between Proteins and Small-Molecule Drug Candidates by Two Hydrogen-Deuterium Exchange MS-Based Methods: The Example of Apolipoprotein E3. Anal Chem 2017; 89:10687-10695. [PMID: 28901129 DOI: 10.1021/acs.analchem.7b01121] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
We describe a platform utilizing two methods based on hydrogen-deuterium exchange (HDX) coupled with mass spectrometry (MS) to characterize interactions between a protein and a small-molecule ligand. The model system is apolipoprotein E3 (apoE3) and a small-molecule drug candidate. We extended PLIMSTEX (protein-ligand interactions by mass spectrometry, titration, and H/D exchange) to the regional level by incorporating enzymatic digestion to acquire binding information for peptides. In a single experiment, we not only identified putative binding sites, but also obtained affinities of 6.0, 6.8, and 10.6 μM for the three different regions, giving an overall binding affinity of 7.4 μM. These values agree well with literature values determined by accepted methods. Unlike those methods, PLIMSTEX provides site-specific binding information. The second approach, modified SUPREX (stability of unpurified proteins from rates of H/D exchange) coupled with electrospray ionization (ESI), allowed us to obtain detailed understanding about apoE unfolding and its changes upon ligand binding. Three binding regions, along with an additional site, which may be important for lipid binding, show increased stability (less unfolding) upon ligand binding. By employing a single parameter, ΔC1/2%, we compared relative changes of denaturation between peptides. This integrated platform provides information orthogonal to commonly used HDX kinetics experiments, providing a general and novel approach for studying protein-ligand interactions.
Collapse
Affiliation(s)
- Hanliu Wang
- Department of Chemistry, Washington University in St. Louis , One Brookings Drive, St. Louis, Missouri 63130, United States.,Analytical Research and Development, Pfizer Incorporated , Chesterfield, Missouri 63017, United States
| | - Don L Rempel
- Department of Chemistry, Washington University in St. Louis , One Brookings Drive, St. Louis, Missouri 63130, United States
| | - Daryl Giblin
- Department of Chemistry, Washington University in St. Louis , One Brookings Drive, St. Louis, Missouri 63130, United States
| | - Carl Frieden
- Department of Biochemistry and Molecular Biophysics, School of Medicine, Washington University in St. Louis , 660 South Euclid Avenue, St. Louis, Missouri 63110, United States
| | - Michael L Gross
- Department of Chemistry, Washington University in St. Louis , One Brookings Drive, St. Louis, Missouri 63130, United States
| |
Collapse
|
33
|
Dalvit C, Santi S, Neier R. A Ligand‐Based NMR Screening Approach for the Identification and Characterization of Inhibitors and Promoters of Amyloid Peptide Aggregation. ChemMedChem 2017; 12:1458-1463. [DOI: 10.1002/cmdc.201700319] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Revised: 07/20/2017] [Indexed: 11/11/2022]
Affiliation(s)
- Claudio Dalvit
- Department of ChemistryUniversity of Neuchatel Avenue de Bellevaux 51 2000 Neuchatel Switzerland
| | - Sara Santi
- Department of ChemistryUniversity of Neuchatel Avenue de Bellevaux 51 2000 Neuchatel Switzerland
| | - Reinhard Neier
- Department of ChemistryUniversity of Neuchatel Avenue de Bellevaux 51 2000 Neuchatel Switzerland
| |
Collapse
|
34
|
Göth M, Badock V, Weiske J, Pagel K, Kuropka B. Critical Evaluation of Native Electrospray Ionization Mass Spectrometry for Fragment-Based Screening. ChemMedChem 2017; 12:1201-1211. [PMID: 28618179 DOI: 10.1002/cmdc.201700177] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 05/19/2017] [Indexed: 12/24/2022]
Abstract
Fragment-based screening presents a promising alternative to high-throughput screening and has gained great attention in recent years. So far, only a few studies have discussed mass spectrometry as a screening technology for fragments. Herein, we report the application of native electrospray ionization mass spectrometry (MS) for screening defined sets of fragments against four different target proteins. Fragments were selected from a primary screening conducted with a thermal shift assay (TSA) and represented different binding categories. Our data indicated that, beside specific complex formation, many fragments show extensive multiple binding and also charge-state shifts. Both of these factors complicate automated data analysis and decrease the attractiveness of native MS as a primary screening tool for fragments. A comparison of the hits identified by native MS and TSA showed good agreement for two of the proteins. Furthermore, we discuss general challenges, including the determination of an optimal fragment concentration and the question of how to rank fragment hits according to their affinity. In conclusion, we consider native MS to be a highly valuable tool for the validation and deeper investigation of promising fragment hits rather than a method for primary screening.
Collapse
Affiliation(s)
- Melanie Göth
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustraße 3, 14195, Berlin, Germany.,Department of Molecular Physics, Fritz Haber Institute of the Max Planck Society, Faradayweg 4-6, 14195, Berlin, Germany
| | - Volker Badock
- Protein Technologies, Lead Discovery Berlin, Bayer AG, Müllerstraße 178, 13353, Berlin, Germany
| | - Jörg Weiske
- Protein Technologies, Lead Discovery Berlin, Bayer AG, Müllerstraße 178, 13353, Berlin, Germany
| | - Kevin Pagel
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustraße 3, 14195, Berlin, Germany.,Department of Molecular Physics, Fritz Haber Institute of the Max Planck Society, Faradayweg 4-6, 14195, Berlin, Germany
| | - Benno Kuropka
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Thielallee 63, 14195, Berlin, Germany.,Protein Technologies, Lead Discovery Berlin, Bayer AG, Müllerstraße 178, 13353, Berlin, Germany
| |
Collapse
|
35
|
Chilton M, Clennell B, Edfeldt F, Geschwindner S. Hot-Spotting with Thermal Scanning: A Ligand- and Structure-Independent Assessment of Target Ligandability. J Med Chem 2017; 60:4923-4931. [PMID: 28537726 DOI: 10.1021/acs.jmedchem.7b00208] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Evaluating the ligandability of a protein target is a key component when defining hit-finding strategies or when prioritize among drug targets. Computational as well as biophysical approaches based on nuclear magnetic resonance (NMR) fragment screening are powerful approaches but suffer from specific constraints that limit their usage. Here, we demonstrate the applicability of high-throughput thermal scanning (HTTS) as a simple and generic biophysical fragment screening method to reproduce assessments from NMR-based screening. By applying this method to a large set of proteins we can furthermore show that the assessment is predictive of the success of high-throughput screening (HTS). The few divergences for targets of low ligandability originate from the sensitivity differences of the orthogonal biophysical methods. We thus applied a new strategy making use of modulations in the solvent structure to improve assay sensitivity. This novel approach enables improved ligandability assessments in accordance with NMR-based assessments and more importantly positions the methodology as a valuable option for biophysical fragment screening.
Collapse
Affiliation(s)
- Molly Chilton
- Innovative Medicines and Early Development Biotech Unit, Discovery Sciences, AstraZeneca R&D Gothenburg , 43183 Mölndal, Sweden
| | - Ben Clennell
- Innovative Medicines and Early Development Biotech Unit, Discovery Sciences, AstraZeneca R&D Gothenburg , 43183 Mölndal, Sweden
| | - Fredrik Edfeldt
- Innovative Medicines and Early Development Biotech Unit, Discovery Sciences, AstraZeneca R&D Gothenburg , 43183 Mölndal, Sweden
| | - Stefan Geschwindner
- Innovative Medicines and Early Development Biotech Unit, Discovery Sciences, AstraZeneca R&D Gothenburg , 43183 Mölndal, Sweden
| |
Collapse
|
36
|
Calle LP, Espinosa JF. An improved 19 F-CPMG scheme for detecting binding of polyfluorinated molecules to biological receptors. MAGNETIC RESONANCE IN CHEMISTRY : MRC 2017; 55:355-358. [PMID: 27661784 DOI: 10.1002/mrc.4531] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 09/18/2016] [Accepted: 09/21/2016] [Indexed: 06/06/2023]
Affiliation(s)
- Luis Pablo Calle
- Centro de Investigación Lilly, Avda. de la Industria 30, 28108, Alcobendas, Madrid, Spain
| | - Juan Félix Espinosa
- Centro de Investigación Lilly, Avda. de la Industria 30, 28108, Alcobendas, Madrid, Spain
| |
Collapse
|
37
|
Structure-Based Discovery of Small Molecules Binding to RNA. TOPICS IN MEDICINAL CHEMISTRY 2017. [DOI: 10.1007/7355_2016_29] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
38
|
Liu J, Gao J, Li F, Ma R, Wei Q, Wang A, Wu J, Ruan K. NMR characterization of weak interactions between RhoGDI2 and fragment screening hits. Biochim Biophys Acta Gen Subj 2017; 1861:3061-3070. [DOI: 10.1016/j.bbagen.2016.10.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 09/26/2016] [Accepted: 10/04/2016] [Indexed: 12/31/2022]
|
39
|
Budiardjo SJ, Licknack TJ, Cory MB, Kapros D, Roy A, Lovell S, Douglas J, Karanicolas J. Full and Partial Agonism of a Designed Enzyme Switch. ACS Synth Biol 2016; 5:1475-1484. [PMID: 27389009 DOI: 10.1021/acssynbio.6b00097] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Chemical biology has long sought to build protein switches for use in molecular diagnostics, imaging, and synthetic biology. The overarching challenge for any type of engineered protein switch is the ability to respond in a selective and predictable manner that caters to the specific environments and time scales needed for the application at hand. We previously described a general method to design switchable proteins, called "chemical rescue of structure", that builds de novo allosteric control sites directly into a protein's functional domain. This approach entails first carving out a buried cavity in a protein via mutation, such that the protein's structure is disrupted and activity is lost. An exogenous ligand is subsequently added to substitute for the atoms that were removed by mutation, restoring the protein's structure and thus its activity. Here, we begin to ask what principles dictate such switches' response to different activating ligands. Using a redesigned β-glycosidase enzyme as our model system, we find that the designed effector site is quite malleable and can accommodate both larger and smaller ligands, but that optimal rescue comes only from a ligand that perfectly replaces the deleted atoms. Guided by these principles, we then altered the shape of this cavity by using different cavity-forming mutations, and predicted different ligands that would better complement these new cavities. These findings demonstrate how the protein switch's response can be tuned via small changes to the ligand with respect to the binding cavity, and ultimately enabled us to design an improved switch. We anticipate that these insights will help enable the design of future systems that tune other aspects of protein activity, whereby, like evolved protein receptors, remolding the effector site can also adjust additional outputs such as substrate selectivity and activation of downstream signaling pathways.
Collapse
Affiliation(s)
- S. Jimmy Budiardjo
- Center for Computational Biology, ‡Department of Molecular
Biosciences, §High Throughput Screening
Laboratory, ∥Protein Structure Laboratory, ⊥Molecular Structures Group The University of Kansas, 2030 Becker Drive, Lawrence, Kansas 66045-7534, United States
| | - Timothy J. Licknack
- Center for Computational Biology, ‡Department of Molecular
Biosciences, §High Throughput Screening
Laboratory, ∥Protein Structure Laboratory, ⊥Molecular Structures Group The University of Kansas, 2030 Becker Drive, Lawrence, Kansas 66045-7534, United States
| | - Michael B. Cory
- Center for Computational Biology, ‡Department of Molecular
Biosciences, §High Throughput Screening
Laboratory, ∥Protein Structure Laboratory, ⊥Molecular Structures Group The University of Kansas, 2030 Becker Drive, Lawrence, Kansas 66045-7534, United States
| | - Dora Kapros
- Center for Computational Biology, ‡Department of Molecular
Biosciences, §High Throughput Screening
Laboratory, ∥Protein Structure Laboratory, ⊥Molecular Structures Group The University of Kansas, 2030 Becker Drive, Lawrence, Kansas 66045-7534, United States
| | - Anuradha Roy
- Center for Computational Biology, ‡Department of Molecular
Biosciences, §High Throughput Screening
Laboratory, ∥Protein Structure Laboratory, ⊥Molecular Structures Group The University of Kansas, 2030 Becker Drive, Lawrence, Kansas 66045-7534, United States
| | - Scott Lovell
- Center for Computational Biology, ‡Department of Molecular
Biosciences, §High Throughput Screening
Laboratory, ∥Protein Structure Laboratory, ⊥Molecular Structures Group The University of Kansas, 2030 Becker Drive, Lawrence, Kansas 66045-7534, United States
| | - Justin Douglas
- Center for Computational Biology, ‡Department of Molecular
Biosciences, §High Throughput Screening
Laboratory, ∥Protein Structure Laboratory, ⊥Molecular Structures Group The University of Kansas, 2030 Becker Drive, Lawrence, Kansas 66045-7534, United States
| | - John Karanicolas
- Center for Computational Biology, ‡Department of Molecular
Biosciences, §High Throughput Screening
Laboratory, ∥Protein Structure Laboratory, ⊥Molecular Structures Group The University of Kansas, 2030 Becker Drive, Lawrence, Kansas 66045-7534, United States
| |
Collapse
|
40
|
Ray PC, Kiczun M, Huggett M, Lim A, Prati F, Gilbert IH, Wyatt PG. Fragment library design, synthesis and expansion: nurturing a synthesis and training platform. Drug Discov Today 2016; 22:43-56. [PMID: 27793744 DOI: 10.1016/j.drudis.2016.10.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 08/29/2016] [Accepted: 10/06/2016] [Indexed: 10/20/2022]
Abstract
The availability of suitable diverse fragment- and lead-oriented screening compounds is key for the identification of suitable chemical starting points for drug discovery programs. The physicochemical properties of molecules are crucial in determining the success of small molecules in clinical development, yet reports suggest that pharmaceutical and academic sectors often produce molecules with poor drug-like properties. We present a platform to design novel, high quality and diverse fragment- and lead-oriented libraries with appropriate physicochemical properties in a cost-efficient manner. This approach has the potential to assist the way libraries are constructed by significantly addressing the historical uneven exploration of chemical space for drug discovery. Additionally, this platform can teach undergraduates and graduates about compound library design.
Collapse
Affiliation(s)
- Peter C Ray
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, Scotland, UK
| | - Michael Kiczun
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, Scotland, UK
| | - Margaret Huggett
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, Scotland, UK
| | - Andrew Lim
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, Scotland, UK
| | - Federica Prati
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, Scotland, UK
| | - Ian H Gilbert
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, Scotland, UK.
| | - Paul G Wyatt
- Drug Discovery Unit, Division of Biological Chemistry and Drug Discovery, College of Life Sciences, University of Dundee, Dow Street, Dundee, DD1 5EH, Scotland, UK.
| |
Collapse
|
41
|
Kim Y, Liu M, Hilty C. Parallelized Ligand Screening Using Dissolution Dynamic Nuclear Polarization. Anal Chem 2016; 88:11178-11183. [PMID: 27723298 DOI: 10.1021/acs.analchem.6b03382] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Protein-ligand interactions are frequently screened using nuclear magnetic resonance (NMR) spectroscopy. The dissociation constant (KD) of a ligand of interest can be determined via a spin-spin relaxation measurement of a reporter ligand in a single scan when using hyperpolarization by means of dissolution dynamic nuclear polarization (D-DNP). Despite nearly instantaneous signal acquisition, a limitation of D-DNP for the screening of protein-ligand interactions is the required polarization time on the order of tens of minutes. Here, we introduce a multiplexed NMR experiment, where a single hyperpolarized ligand sample is rapidly mixed with protein injected into two flow cells. NMR detection is achieved simultaneously on both channels, resulting in a chemical shift resolved spin relaxation measurement. Spectral resolution allows the use of reference compounds for accurate quantification of concentrations. Simultaneous use of two concentration ratios between protein and ligand broadens the range of KD that is accurately measurable in a single experiment to at least an order of magnitude. In a comparison of inhibitors for the protein trypsin, the average KD values of benzamidine and benzylamine were found to be 12.6 ± 1.4 μM and 207 ± 22 μM from three measurements, based on KD = 142 μM assumed known for the reporter ligand 4-(trifluoromethyl)benzene-1-carboximidamide. Typical confidence ranges at 95% evaluated for single experiments were (8.3 μM, 20 μM) and (151 μM, 328 μM). The multiplexed detection of two or more hyperpolarized samples increases throughput of D-DNP by the same factor, improving the applicability to most multipoint measurements that would traditionally be achieved using titrations.
Collapse
Affiliation(s)
- Yaewon Kim
- Department of Chemistry, Texas A&M University , College Station, Texas 77843-3255, United States
| | - Mengxiao Liu
- Department of Chemistry, Texas A&M University , College Station, Texas 77843-3255, United States
| | - Christian Hilty
- Department of Chemistry, Texas A&M University , College Station, Texas 77843-3255, United States
| |
Collapse
|
42
|
Boulton S, Melacini G. Advances in NMR Methods To Map Allosteric Sites: From Models to Translation. Chem Rev 2016; 116:6267-304. [PMID: 27111288 DOI: 10.1021/acs.chemrev.5b00718] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The last five years have witnessed major developments in the understanding of the allosteric phenomenon, broadly defined as coupling between remote molecular sites. Such advances have been driven not only by new theoretical models and pharmacological applications of allostery, but also by progress in the experimental approaches designed to map allosteric sites and transitions. Among these techniques, NMR spectroscopy has played a major role given its unique near-atomic resolution and sensitivity to the dynamics that underlie allosteric couplings. Here, we highlight recent progress in the NMR methods tailored to investigate allostery with the goal of offering an overview of which NMR approaches are best suited for which allosterically relevant questions. The picture of the allosteric "NMR toolbox" is provided starting from one of the simplest models of allostery (i.e., the four-state thermodynamic cycle) and continuing to more complex multistate mechanisms. We also review how such an "NMR toolbox" has assisted the elucidation of the allosteric molecular basis for disease-related mutations and the discovery of novel leads for allosteric drugs. From this overview, it is clear that NMR plays a central role not only in experimentally validating transformative theories of allostery, but also in tapping the full translational potential of allosteric systems.
Collapse
Affiliation(s)
- Stephen Boulton
- Department of Chemistry and Chemical Biology Department of Biochemistry and Biomedical Sciences, McMaster University , 1280 Main St. W., Hamilton L8S 4M1, Canada
| | - Giuseppe Melacini
- Department of Chemistry and Chemical Biology Department of Biochemistry and Biomedical Sciences, McMaster University , 1280 Main St. W., Hamilton L8S 4M1, Canada
| |
Collapse
|
43
|
Peng C, Frommlet A, Perez M, Cobas C, Blechschmidt A, Dominguez S, Lingel A. Fast and Efficient Fragment-Based Lead Generation by Fully Automated Processing and Analysis of Ligand-Observed NMR Binding Data. J Med Chem 2016; 59:3303-10. [DOI: 10.1021/acs.jmedchem.6b00019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Chen Peng
- Mestrelab Research S.L., Feliciano
Barrera 9B − Baixo, 15706 Santiago de Compostela, Spain
| | - Alexandra Frommlet
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Manuel Perez
- Mestrelab Research S.L., Feliciano
Barrera 9B − Baixo, 15706 Santiago de Compostela, Spain
| | - Carlos Cobas
- Mestrelab Research S.L., Feliciano
Barrera 9B − Baixo, 15706 Santiago de Compostela, Spain
| | - Anke Blechschmidt
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| | - Santiago Dominguez
- Mestrelab Research S.L., Feliciano
Barrera 9B − Baixo, 15706 Santiago de Compostela, Spain
| | - Andreas Lingel
- Novartis Institutes for BioMedical Research, 5300 Chiron Way, Emeryville, California 94608, United States
| |
Collapse
|
44
|
Abstract
INTRODUCTION Fragment-based approaches have played an increasing role alongside high-throughput screening in drug discovery for 15 years. The label-free biosensor technology based on surface plasmon resonance (SPR) is now sensitive and informative enough to serve during primary screens and validation steps. AREAS COVERED In this review, the authors discuss the role of SPR in fragment screening. After a brief description of the underlying principles of the technique and main device developments, they evaluate the advantages and adaptations of SPR for fragment-based drug discovery. SPR can also be applied to challenging targets such as membrane receptors and enzymes. EXPERT OPINION The high-level of immobilization of the protein target and its stability are key points for a relevant screening that can be optimized using oriented immobilized proteins and regenerable sensors. Furthermore, to decrease the rate of false negatives, a selectivity test may be performed in parallel on the main target bearing the binding site mutated or blocked with a low-off-rate ligand. Fragment-based drug design, integrated in a rational workflow led by SPR, will thus have a predominant role for the next wave of drug discovery which could be greatly enhanced by new improvements in SPR devices.
Collapse
Affiliation(s)
- Alain Chavanieu
- a Institut des Biomolécules Max Mousseron (IBMM), UMR 5247 , Université de Montpellier, CNRS, ENSCM , Montpellier Cedex 5, France
| | - Martine Pugnière
- b IRCM , Institut de Recherche en Cancérologie de Montpellier , Montpellier , France.,c INSERM, U1194 , Université Montpellier , Montpellier , France.,d ICM , Institut Régional du Cancer , Montpellier , France
| |
Collapse
|
45
|
Joyce RE, Williams TL, Serpell LC, Day IJ. Monitoring changes of paramagnetically-shifted 31P signals in phospholipid vesicles. Chem Phys Lett 2016. [DOI: 10.1016/j.cplett.2016.02.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
46
|
Hanley RP, Horvath S, An J, Hof F, Wulff JE. Salicylates are interference compounds in TR-FRET assays. Bioorg Med Chem Lett 2016; 26:973-977. [DOI: 10.1016/j.bmcl.2015.12.050] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 12/12/2015] [Accepted: 12/15/2015] [Indexed: 12/20/2022]
|
47
|
Abstract
While saturation transfer difference (STD) is a widely used NMR method for ligand screening, the selection of specific binders requires the validation of the hits through competition experiments or orthogonal biophysical techniques. We show here that the quantitative STD analysis is a reliable and robust approach to discriminate between specific and nonspecific ligands, allowing selection of fragments that bind proteins with a privileged binding mode, in the absence of any structural data for the protein.
Collapse
Affiliation(s)
- Olivier Cala
- Institut des Sciences Analytiques, UMR5280 CNRS, Université Lyon 1, Ecole Nationale Supérieure de Lyon, Université de Lyon , 5 rue de la Doua Villeurbanne 69100, France
| | - Isabelle Krimm
- Institut des Sciences Analytiques, UMR5280 CNRS, Université Lyon 1, Ecole Nationale Supérieure de Lyon, Université de Lyon , 5 rue de la Doua Villeurbanne 69100, France
| |
Collapse
|
48
|
Iconaru LI, Ban D, Bharatham K, Ramanathan A, Zhang W, Shelat AA, Zuo J, Kriwacki RW. Discovery of Small Molecules that Inhibit the Disordered Protein, p27(Kip1). Sci Rep 2015; 5:15686. [PMID: 26507530 PMCID: PMC4623604 DOI: 10.1038/srep15686] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 09/25/2015] [Indexed: 01/13/2023] Open
Abstract
Disordered proteins are highly prevalent in biological systems, they control myriad signaling and regulatory processes, and their levels and/or cellular localization are often altered in human disease. In contrast to folded proteins, disordered proteins, due to conformational heterogeneity and dynamics, are not considered viable drug targets. We challenged this paradigm by identifying through NMR-based screening small molecules that bound specifically, albeit weakly, to the disordered cell cycle regulator, p27Kip1 (p27). Two groups of molecules bound to sites created by transient clusters of aromatic residues within p27. Conserved chemical features within these two groups of small molecules exhibited complementarity to their binding sites within p27, establishing structure-activity relationships for small molecule:disordered protein interactions. Finally, one compound counteracted the Cdk2/cyclin A inhibitory function of p27 in vitro, providing proof-of-principle that small molecules can inhibit the function of a disordered protein (p27) through sequestration in a conformation incapable of folding and binding to a natural regulatory target (Cdk2/cyclin A).
Collapse
Affiliation(s)
- Luigi I Iconaru
- Department of Structural Biology, Memphis, TN 38105.,Department of Developmental Neurobiology, Memphis, TN 38105
| | - David Ban
- Department of Structural Biology, Memphis, TN 38105
| | - Kavitha Bharatham
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Arvind Ramanathan
- Computational Science and Engineering Division, Health Data Sciences Institute, Oak Ridge National Laboratory, Oak Ridge, TN 37830
| | | | - Anang A Shelat
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Jian Zuo
- Department of Developmental Neurobiology, Memphis, TN 38105
| | - Richard W Kriwacki
- Department of Structural Biology, Memphis, TN 38105.,Department of Microbiology, Immunology and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN 38163
| |
Collapse
|
49
|
Successful generation of structural information for fragment-based drug discovery. Drug Discov Today 2015; 20:1104-11. [DOI: 10.1016/j.drudis.2015.04.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 03/12/2015] [Accepted: 04/20/2015] [Indexed: 12/25/2022]
|
50
|
Schiebel J, Radeva N, Köster H, Metz A, Krotzky T, Kuhnert M, Diederich WE, Heine A, Neumann L, Atmanene C, Roecklin D, Vivat-Hannah V, Renaud JP, Meinecke R, Schlinck N, Sitte A, Popp F, Zeeb M, Klebe G. One Question, Multiple Answers: Biochemical and Biophysical Screening Methods Retrieve Deviating Fragment Hit Lists. ChemMedChem 2015; 10:1511-21. [DOI: 10.1002/cmdc.201500267] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Indexed: 12/12/2022]
|