Ross McGuire
Radboud University Nijmegen
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Ross McGuire.
Computational and structural biotechnology journal | 2013
Valère Lounnas; Tina Ritschel; Jan Kelder; Ross McGuire; Robert P. Bywater; Nicolas Foloppe
The past decade has witnessed a paradigm shift in preclinical drug discovery with structure-based drug design (SBDD) making a comeback while high-throughput screening (HTS) methods have continued to generate disappointing results. There is a deficit of information between identified hits and the many criteria that must be fulfilled in parallel to convert them into preclinical candidates that have a real chance to become a drug. This gap can be bridged by investigating the interactions between the ligands and their receptors. Accurate calculations of the free energy of binding are still elusive; however progresses were made with respect to how one may deal with the versatile role of water. A corpus of knowledge combining X-ray structures, bioinformatics and molecular modeling techniques now allows drug designers to routinely produce receptor homology models of increasing quality. These models serve as a basis to establish and validate efficient rationales used to tailor and/or screen virtual libraries with enhanced chances of obtaining hits. Many case reports of successful SBDD show how synergy can be gained from the combined use of several techniques. The role of SBDD with respect to two different classes of widely investigated pharmaceutical targets: (a) protein kinases (PK) and (b) G-protein coupled receptors (GPCR) is discussed. Throughout these examples prototypical situations covering the current possibilities and limitations of SBDD are presented.
MedChemComm | 2012
Mpa Marijn Sanders; Ross McGuire; Luc Roumen; de Ijp Esch; de J Jacob Vlieg; Jpg Klomp; de C Graaf
A pharmacophore describes the arrangement of molecular features a ligand must contain to efficaciously bind a receptor. Pharmacophore models are developed to improve molecular understanding of ligand–protein interactions, and can be used as a tool to identify novel compounds that fulfil the pharmacophore requirements and have a high probability of being biologically active. Protein structure-based pharmacophores (SBPs) derive these molecular features by conversion of protein properties to reciprocal ligand space. Unlike ligand-based pharmacophore models, which require templates of ligands in their bioactive conformation, SBPs do not depend on ligand information. The current review describes the different steps in the construction of SBPs: (i) protein structure preparation, (ii) binding site detection, (iii) pharmacophore feature definition, and (iv) pharmacophore feature selection. We show that the SBP modeling workflow poses different challenges than ligand-based pharmacophore modeling, including the definition of protein pharmacophore features essential for ligand binding. A comprehensive overview of different SBP modeling and screening methods and applications is provided to illustrate that SBPs can be efficiently used for virtual screening, ligand binding mode prediction, and binding site similarity detection. Our review demonstrates that SBPs are valuable tools for hit and lead optimization, compound library design and target hopping, especially in cases where ligand information is scarce.
Journal of Medicinal Chemistry | 2012
Marijn P. A. Sanders; Luc Roumen; Eelke van der Horst; J. Robert Lane; Henry F. Vischer; Jody van Offenbeek; Henk de Vries; Stefan Verhoeven; Ken Y. Chow; Folkert Verkaar; Margot W. Beukers; Ross McGuire; Rob Leurs; Adriaan P. IJzerman; Jacob de Vlieg; Iwan J. P. de Esch; Guido J.R. Zaman; Jan P. G. Klomp; Andreas Bender; Chris de Graaf
We present the systematic prospective evaluation of a protein-based and a ligand-based virtual screening platform against a set of three G-protein-coupled receptors (GPCRs): the β-2 adrenoreceptor (ADRB2), the adenosine A(2A) receptor (AA2AR), and the sphingosine 1-phosphate receptor (S1PR1). Novel bioactive compounds were identified using a consensus scoring procedure combining ligand-based (frequent substructure ranking) and structure-based (Snooker) tools, and all 900 selected compounds were screened against all three receptors. A striking number of ligands showed affinity/activity for GPCRs other than the intended target, which could be partly attributed to the fuzziness and overlap of protein-based pharmacophore models. Surprisingly, the phosphodiesterase 5 (PDE5) inhibitor sildenafil was found to possess submicromolar affinity for AA2AR. Overall, this is one of the first published prospective chemogenomics studies that demonstrate the identification of novel cross-pharmacology between unrelated protein targets. The lessons learned from this study can be used to guide future virtual ligand design efforts.
Journal of Biological Chemistry | 2011
Scott J. Lusher; Hans C.A. Raaijmakers; Diep Vu-Pham; Koen Dechering; Tsang Wai Lam; Angus R. Brown; Niall M. Hamilton; Olaf Nimz; Rolien Bosch; Ross McGuire; Arthur Oubrie; Jacob de Vlieg
The progesterone receptor is able to bind to a large number and variety of ligands that elicit a broad range of transcriptional responses ranging from full agonism to full antagonism and numerous mixed profiles inbetween. We describe here two new progesterone receptor ligand binding domain x-ray structures bound to compounds from a structurally related but functionally divergent series, which show different binding modes corresponding to their agonistic or antagonistic nature. In addition, we present a third progesterone receptor ligand binding domain dimer bound to an agonist in monomer A and an antagonist in monomer B, which display binding modes in agreement with the earlier observation that agonists and antagonists from this series adopt different binding modes.
PLOS ONE | 2012
Marie-José van Lierop; Wynand Alkema; Anke J. Laskewitz; Rein Dijkema; Hans van der Maaden; Martin J. Smit; Ralf Plate; Paolo Conti; Christan G.J.M. Jans; C. Marco Timmers; Constant A. A. van Boeckel; Scott J. Lusher; Ross McGuire; René C. van Schaik; Jacob de Vlieg; Ruben L. Smeets; Claudia L. Hofstra; Annemieke M. H. Boots; Marcel van Duin; Benno A. Ingelse; Willem G.E.J. Schoonen; Aldo Grefhorst; Theo H. van Dijk; Folkert Kuipers; Wim H. A. Dokter
Glucocorticoids (GCs) such as prednisolone are potent immunosuppressive drugs but suffer from severe adverse effects, including the induction of insulin resistance. Therefore, development of so-called Selective Glucocorticoid Receptor Modulators (SGRM) is highly desirable. Here we describe a non-steroidal Glucocorticoid Receptor (GR)-selective compound (Org 214007-0) with a binding affinity to GR similar to that of prednisolone. Structural modelling of the GR-Org 214007-0 binding site shows disturbance of the loop between helix 11 and helix 12 of GR, confirmed by partial recruitment of the TIF2-3 peptide. Using various cell lines and primary human cells, we show here that Org 214007-0 acts as a partial GC agonist, since it repressed inflammatory genes and was less effective in induction of metabolic genes. More importantly, in vivo studies in mice indicated that Org 214007-0 retained full efficacy in acute inflammation models as well as in a chronic collagen-induced arthritis (CIA) model. Gene expression profiling of muscle tissue derived from arthritic mice showed a partial activity of Org 214007-0 at an equi-efficacious dosage of prednisolone, with an increased ratio in repression versus induction of genes. Finally, in mice Org 214007-0 did not induce elevated fasting glucose nor the shift in glucose/glycogen balance in the liver seen with an equi-efficacious dose of prednisolone. All together, our data demonstrate that Org 214007-0 is a novel SGRMs with an improved therapeutic index compared to prednisolone. This class of SGRMs can contribute to effective anti-inflammatory therapy with a lower risk for metabolic side effects.
Journal of Biological Chemistry | 2012
Scott J. Lusher; Hans C.A. Raaijmakers; Diep Vu-Pham; B Kazemier; R. Bosch; Ross McGuire; Rita Azevedo; H Hamersma; K Dechering; Arthur Oubrie; M Van Duin; J. De Vlieg
Background: Understanding the molecular basis for the mixed profiles of progesterone receptor (PR) ligands will benefit future drug design. Results: Two differing mechanisms for the induction of mixed profiles by 11β-steroids are described. Conclusion: Subtle electrostatic and steric factors explain the differing PR activities of 11β-steroids. Significance: These observations will impact future drug-design strategies for PR and potentially other nuclear receptors. We present here the x-ray structures of the progesterone receptor (PR) in complex with two mixed profile PR modulators whose functional activity results from two differing molecular mechanisms. The structure of Asoprisnil bound to the agonist state of PR demonstrates the contribution of the ligand to increasing stability of the agonist conformation of helix-12 via a specific hydrogen-bond network including Glu723. This interaction is absent when the full antagonist, RU486, binds to PR. Combined with a previously reported structure of Asoprisnil bound to the antagonist state of the receptor, this structure extends our understanding of the complex molecular interactions underlying the mixed agonist/antagonist profile of the compound. In addition, we present the structure of PR in its agonist conformation bound to the mixed profile compound Org3H whose reduced antagonistic activity and increased agonistic activity compared with reference antagonists is due to an induced fit around Trp755, resulting in a decreased steric clash with Met909 but inducing a new internal clash with Val912 in helix-12. This structure also explains the previously published observation that 16α attachments to RU486 analogs induce mixed profiles by altering the binding of 11β substituents. Together these structures further our understanding of the steric and electrostatic factors that contribute to the function of steroid receptor modulators, providing valuable insight for future compound design.
Drug Discovery Today | 2011
Scott J. Lusher; Ross McGuire; Rita Azevedo; Jan-Willem Boiten; René van Schaik; Jacob de Vlieg
The difference between biologically active molecules and drugs is that the latter balance an array of related and unrelated properties required for administration to patients. Inevitability, during optimization, some of these multiple factors will conflict. Although informatics has a crucial role in addressing the challenges of modern compound optimization, it is arguably still undervalued and underutilized. We present here some of the basic requirements of multi-parameter drug design, the crucial role of informatics and examples of favorable practice. The most crucial of these best practices are the need for informaticians to align their technologies and insights directly to discovery projects and for all scientists in drug discovery to become more proficient in the use of in silico methods.
Journal of Chemical Information and Modeling | 2017
Ross McGuire; Stefan Verhoeven; Márton Vass; Gerrit Vriend; Iwan J. P. de Esch; Scott J. Lusher; Rob Leurs; Lars Ridder; Albert J. Kooistra; Tina Ritschel; Chris de Graaf
3D-e-Chem-VM is an open source, freely available Virtual Machine (http://3d-e-chem.github.io/3D-e-Chem-VM/) that integrates cheminformatics and bioinformatics tools for the analysis of protein–ligand interaction data. 3D-e-Chem-VM consists of software libraries, and database and workflow tools that can analyze and combine small molecule and protein structural information in a graphical programming environment. New chemical and biological data analytics tools and workflows have been developed for the efficient exploitation of structural and pharmacological protein–ligand interaction data from proteomewide databases (e.g., ChEMBLdb and PDB), as well as customized information systems focused on, e.g., G protein-coupled receptors (GPCRdb) and protein kinases (KLIFS). The integrated structural cheminformatics research infrastructure compiled in the 3D-e-Chem-VM enables the design of new approaches in virtual ligand screening (Chemdb4VS), ligand-based metabolism prediction (SyGMa), and structure-based protein binding site comparison and bioisosteric replacement for ligand design (KRIPOdb).
ChemMedChem | 2018
Albert J. Kooistra; Márton Vass; Ross McGuire; Rob Leurs; Iwan J. P. de Esch; Gert Vriend; Stefan Verhoeven; Chris de Graaf
eScience technologies are needed to process the information available in many heterogeneous types of protein–ligand interaction data and to capture these data into models that enable the design of efficacious and safe medicines. Here we present scientific KNIME tools and workflows that enable the integration of chemical, pharmacological, and structural information for: i) structure‐based bioactivity data mapping, ii) structure‐based identification of scaffold replacement strategies for ligand design, iii) ligand‐based target prediction, iv) protein sequence‐based binding site identification and ligand repurposing, and v) structure‐based pharmacophore comparison for ligand repurposing across protein families. The modular setup of the workflows and the use of well‐established standards allows the re‐use of these protocols and facilitates the design of customized computer‐aided drug discovery workflows.
Journal of Medicinal Chemistry | 2005
Jeroen Kazius; Ross McGuire; Roberta Bursi