Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Michael J. Bodkin is active.

Publication


Featured researches published by Michael J. Bodkin.


Chemical Science | 2016

Accurate calculation of the absolute free energy of binding for drug molecules

Matteo Aldeghi; Alexander Heifetz; Michael J. Bodkin; Stefan Knapp; Philip C. Biggin

Free energy calculations based on molecular dynamics and thermodynamic cycles accurately reproduce experimental affinities of diverse bromodomain inhibitors.


Journal of Chemical Information and Modeling | 2009

Knowledge-Based Approach to de Novo Design Using Reaction Vectors

Hina Patel; Michael J. Bodkin; Beining Chen; Valerie J. Gillet

A knowledge-based approach to the de novo design of synthetically feasible molecules is described. The method is based on reaction vectors which represent the structural changes that take place at the reaction center along with the environment in which the reaction occurs. The reaction vectors are derived automatically from a database of reactions which is not restricted by size or reaction complexity. A structure generation algorithm has been developed whereby reaction vectors can be applied to previously unseen starting materials in order to suggest novel syntheses. The approach has been implemented in KNIME and is validated by reproducing known synthetic routes. We then present applications of the method in different drug design scenarios including lead optimization and library enumeration. The method offers great potential for capturing and using the growing body of data on reactions that is becoming available through electronic laboratory notebooks.


Journal of Chemical Information and Modeling | 2016

The Fragment Molecular Orbital Method Reveals New Insight into the Chemical Nature of GPCR–Ligand Interactions

Alexander Heifetz; Ewa I. Chudyk; Laura Jane Gleave; Matteo Aldeghi; Vadim Cherezov; Dmitri G. Fedorov; Philip C. Biggin; Michael J. Bodkin

Our interpretation of ligand-protein interactions is often informed by high-resolution structures, which represent the cornerstone of structure-based drug design. However, visual inspection and molecular mechanics approaches cannot explain the full complexity of molecular interactions. Quantum Mechanics approaches are often too computationally expensive, but one method, Fragment Molecular Orbital (FMO), offers an excellent compromise and has the potential to reveal key interactions that would otherwise be hard to detect. To illustrate this, we have applied the FMO method to 18 Class A GPCR-ligand crystal structures, representing different branches of the GPCR genome. Our work reveals key interactions that are often omitted from structure-based descriptions, including hydrophobic interactions, nonclassical hydrogen bonds, and the involvement of backbone atoms. This approach provides a more comprehensive picture of receptor-ligand interactions than is currently used and should prove useful for evaluation of the chemical nature of ligand binding and to support structure-based drug design.


Journal of the American Chemical Society | 2017

Predictions of Ligand Selectivity from Absolute Binding Free Energy Calculations.

Matteo Aldeghi; Alexander Heifetz; Michael J. Bodkin; Stefan Knapp; Philip C. Biggin

Binding selectivity is a requirement for the development of a safe drug, and it is a critical property for chemical probes used in preclinical target validation. Engineering selectivity adds considerable complexity to the rational design of new drugs, as it involves the optimization of multiple binding affinities. Computationally, the prediction of binding selectivity is a challenge, and generally applicable methodologies are still not available to the computational and medicinal chemistry communities. Absolute binding free energy calculations based on alchemical pathways provide a rigorous framework for affinity predictions and could thus offer a general approach to the problem. We evaluated the performance of free energy calculations based on molecular dynamics for the prediction of selectivity by estimating the affinity profile of three bromodomain inhibitors across multiple bromodomain families, and by comparing the results to isothermal titration calorimetry data. Two case studies were considered. In the first one, the affinities of two similar ligands for seven bromodomains were calculated and returned excellent agreement with experiment (mean unsigned error of 0.81 kcal/mol and Pearson correlation of 0.75). In this test case, we also show how the preferred binding orientation of a ligand for different proteins can be estimated via free energy calculations. In the second case, the affinities of a broad-spectrum inhibitor for 22 bromodomains were calculated and returned a more modest accuracy (mean unsigned error of 1.76 kcal/mol and Pearson correlation of 0.48); however, the reparametrization of a sulfonamide moiety improved the agreement with experiment.


Current Opinion in Pharmacology | 2016

Guiding lead optimization with GPCR structure modeling and molecular dynamics

Alexander Heifetz; Tim James; Inaki Morao; Michael J. Bodkin; Philip C. Biggin

G-protein coupled receptor (GPCR) modeling approaches are widely used in the hit-to-lead and lead optimization stages of drug discovery. Modern protocols that involve molecular dynamics simulation can address key issues such as the free energy of binding (affinity), ligand-induced GPCR flexibility, ligand binding kinetics, conserved water positions and their role in ligand binding and the effects of mutations. The goals of these calculations are to predict the structures of the complexes between existing ligands and their receptors, to understand the key interactions and to utilize these insights in the design of new molecules with improved binding, selectivity or other pharmacological properties. In this review we present a brief survey of various computational approaches illustrated through a hierarchical GPCR modeling protocol and its prospective application in three industrial drug discovery projects.


Molecular Informatics | 2013

Extensions to In Silico Bioactivity Predictions Using Pathway Annotations and Differential Pharmacology Analysis: Application to Xenopus laevis Phenotypic Readouts

Sonia Liggi; Georgios Drakakis; Adam E. Hendry; Kimberley Hanson; Suzanne C. Brewerton; Grant N. Wheeler; Michael J. Bodkin; David A. Evans; Andreas Bender

The simultaneous increase of computational power and the availability of chemical and biological data have contributed to the recent popularity of in silico bioactivity prediction algorithms. Such methods are commonly used to infer the ‘Mechanism of Action’ of small molecules and they can also be employed in cases where full bioactivity profiles have not been established experimentally. However, protein target predictions by themselves do not necessarily capture information about the effect of a compound on a biological system, and hence merging their output with a systems biology approach can help to better understand the complex network modulation which leads to a particular phenotype. In this work, we review approaches and applications of target prediction, as well as their shortcomings, and demonstrate two extensions of this concept which are exemplified using phenotypic readouts from a chemical genetic screen in Xenopus laevis. In particular, the experimental observations are linked to their predicted bioactivity profiles. Predicted targets are annotated with pathways, which lead to further biological insight. Moreover, we subject the prediction to further machine learning algorithms, namely decision trees, to capture the differential pharmacology of ligand‐target interactions in biological systems. Both methodologies hence provide new insight into understanding the Mechanism of Action of compound activities from phenotypic screens.


Future Medicinal Chemistry | 2014

Extending in silico mechanism-of- action analysis by annotating targets with pathways: application to cellular cytotoxicity readouts

Sonia Liggi; Georgios Drakakis; Alexios Koutsoukas; Thérèse E. Malliavin; Adrián Velázquez-Campoy; Suzanne C. Brewerton; Michael J. Bodkin; David A. Evans; Robert C. Glen; José Alberto Carrodeguas; Andreas Bender

BACKGROUND An in silico mechanism-of-action analysis protocol was developed, comprising molecule bioactivity profiling, annotation of predicted targets with pathways and calculation of enrichment factors to highlight targets and pathways more likely to be implicated in the studied phenotype. RESULTS The method was applied to a cytotoxicity phenotypic endpoint, with enriched targets/pathways found to be statistically significant when compared with 100 random datasets. Application on a smaller apoptotic set (10 molecules) did not allowed to obtain statistically relevant results, suggesting that the protocol requires modification such as analysis of the most frequently predicted targets/annotated pathways. CONCLUSION Pathway annotations improved the mechanism-of-action information gained by target prediction alone, allowing a better interpretation of the predictions and providing better mapping of targets onto pathways.


Bioinformatics | 2013

ChEMBLSpace—a graphical explorer of the chemogenomic space covered by the ChEMBL database

Nikolas Fechner; George Papadatos; David A. Evans; John Richard Morphy; Suzanne C. Brewerton; David A. Thorner; Michael J. Bodkin

MOTIVATION The ChEMBLSpace graphical explorer enables the identification of compounds from the ChEMBL database, which exhibit a desirable polypharmacology profile. This profile can be predefined or created iteratively, and the tool can be extended to other data sources.


Journal of Chemical Information and Modeling | 2017

Statistical Analysis on the Performance of Molecular Mechanics Poisson–Boltzmann Surface Area versus Absolute Binding Free Energy Calculations: Bromodomains as a Case Study

Matteo Aldeghi; Michael J. Bodkin; Stefan Knapp; Philip C. Biggin

Binding free energy calculations that make use of alchemical pathways are becoming increasingly feasible thanks to advances in hardware and algorithms. Although relative binding free energy (RBFE) calculations are starting to find widespread use, absolute binding free energy (ABFE) calculations are still being explored mainly in academic settings due to the high computational requirements and still uncertain predictive value. However, in some drug design scenarios, RBFE calculations are not applicable and ABFE calculations could provide an alternative. Computationally cheaper end-point calculations in implicit solvent, such as molecular mechanics Poisson–Boltzmann surface area (MMPBSA) calculations, could too be used if one is primarily interested in a relative ranking of affinities. Here, we compare MMPBSA calculations to previously performed absolute alchemical free energy calculations in their ability to correlate with experimental binding free energies for three sets of bromodomain–inhibitor pairs. Different MMPBSA approaches have been considered, including a standard single-trajectory protocol, a protocol that includes a binding entropy estimate, and protocols that take into account the ligand hydration shell. Despite the improvements observed with the latter two MMPBSA approaches, ABFE calculations were found to be overall superior in obtaining correlation with experimental affinities for the test cases considered. A difference in weighted average Pearson () and Spearman () correlations of 0.25 and 0.31 was observed when using a standard single-trajectory MMPBSA setup ( = 0.64 and = 0.66 for ABFE; = 0.39 and = 0.35 for MMPBSA). The best performing MMPBSA protocols returned weighted average Pearson and Spearman correlations that were about 0.1 inferior to ABFE calculations: = 0.55 and = 0.56 when including an entropy estimate, and = 0.53 and = 0.55 when including explicit water molecules. Overall, the study suggests that ABFE calculations are indeed the more accurate approach, yet there is also value in MMPBSA calculations considering the lower compute requirements, and if agreement to experimental affinities in absolute terms is not of interest. Moreover, for the specific protein–ligand systems considered in this study, we find that including an explicit ligand hydration shell or a binding entropy estimate in the MMPBSA calculations resulted in significant performance improvements at a negligible computational cost.


Journal of Computational Chemistry | 2017

Rapid and accurate assessment of GPCR–ligand interactions Using the fragment molecular orbital‐based density‐functional tight‐binding method

Inaki Morao; Dmitri G. Fedorov; Roger Robinson; Michelle Southey; Andrea Townsend-Nicholson; Michael J. Bodkin; Alexander Heifetz

The reliable and precise evaluation of receptor–ligand interactions and pair‐interaction energy is an essential element of rational drug design. While quantum mechanical (QM) methods have been a promising means by which to achieve this, traditional QM is not applicable for large biological systems due to its high computational cost. Here, the fragment molecular orbital (FMO) method has been used to accelerate QM calculations, and by combining FMO with the density‐functional tight‐binding (DFTB) method we are able to decrease computational cost 1000 times, achieving results in seconds, instead of hours. We have applied FMO‐DFTB to three different GPCR–ligand systems. Our results correlate well with site directed mutagenesis data and findings presented in the published literature, demonstrating that FMO‐DFTB is a rapid and accurate means of GPCR–ligand interactions.

Collaboration


Dive into the Michael J. Bodkin's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stefan Knapp

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Adam E. Hendry

University of East Anglia

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge