Original article
LEADOPT: An automatic tool for structure-based lead optimization, and its application in structural optimizations of VEGFR2 and SYK inhibitors

https://doi.org/10.1016/j.ejmech.2015.02.019Get rights and content

Highlights

  • We built a new automatic tool for structure-based lead optimization called LEADOPT.

  • Ligand efficiency is used as a measure to sort the generated molecules in LEADOPT.

  • Twelve ADMET properties are evaluated in LEADOPT.

  • We obtained some new potent VEGFR2 and SYK inhibitors using LEADOPT.

Abstract

Lead optimization is one of the key steps in drug discovery, and currently it is carried out mostly based on experiences of medicinal chemists, which often suffers from low efficiency. In silico methods are thought to be useful in improving the efficiency of lead optimization. Here we describe a new in silico automatic tool for structure-based lead optimization, termed LEADOPT. The structural modifications in LEADOPT mainly include two operations: fragment growing and fragment replacing, which are restricted to carry out in the active pocket of target protein with the core scaffold structure of ligand kept unchanged. The bioactivity of the newly generated molecules is estimated by ligand efficiency rather than a commonly used scoring function. Twelve important pharmacokinetic and toxic properties are evaluated using SCADMET, a program for the prediction of pharmacokinetic and toxic properties. LEADOPT was first evaluated using two retrospective cases, in which it showed a very good performance. LEADOPT was then applied to the structural optimizations of the VEGFR2 inhibitor, sorafenib, and the SYK inhibitor, R406. Though just several compounds were synthesized, we have obtained some compounds that are more potent than sorafenib and R406 in enzymatic and functional assays. All of these have validated, at least to some extent, the effectiveness of LEADOPT.

Graphical abstract

A new in silico automatic tool for structure-based lead optimization termed LEADOPT was developed. It was successfully applied to the structural optimizations of the VEGFR2 inhibitor, sorafenib, and the SYK inhibitor, R406.

  1. Download : Download full-size image

Introduction

Discovery of drug candidates is a bottleneck in drug research and development (R&D). The whole procedure for the discovery of drug candidates often includes two phases: lead discovery and lead optimization [1]. Currently the lead discovery is no longer a big problem or at least not a key challenge due to that many new technologies for the lead discovery have been well established and applied, such as high-throughput screening, biophysical screening, combinatorial chemistry, as well as virtual screening [2], [3], [4], [5], [6]. However, at present the lead optimization is carried out mostly based on experiences of medicinal chemists, which often suffers from low efficiency. The lead optimization has now become a key challenge in not only the discovery of drug candidates but also the whole process of drug R&D.

Computational methods are often thought to be helpful in improving the efficiency of lead optimization [7], [8], [9], [10]. Thus, some in silico methods have been established and applied in the lead optimization. Among these methods, three-dimensional quantitative structure activity relationship (3D-QSAR) methods, for example, comparative molecular field analysis (CoMFA) [11] and comparative molecular similarity indices analysis (CoMSIA) [12], are the most widely used ones. Numerous studies have demonstrated that 3D-QSAR methods are very useful in lead optimization [13], [14], [15], [16]. Nevertheless, the current 3D-QSAR methods as well as other commonly used in silico methods for lead optimization are often just based on small-molecule ligands, implying ignoring structural information of target protein. This situation may lead to the theoretically optimized molecules having no activity due to bumps between molecules and target protein atoms. Structure-based lead optimization approaches are expected to be able to overcome these shortcomings, in which the structure of ligand is modified to enhance the binding affinity based on the known interaction mode between target protein and ligand [17], [18], [19], [20], [21]. Although these approaches have been applied in lead optimization for a long time, they are often used manually by medicinal chemists, which still suffer inevitably from low efficiency. We also noticed that very recently a commercial module for structure-based ligand optimization has been issued in MOE by Chemical Computing Group Inc. However, its implementation details are not known, and it likely does not include the evaluation of pharmacokinetic and toxic properties for the generated molecules, which is also important for lead optimization.

Here we present an automatic tool for structure-based lead optimization, termed LEADOPT. LEADOPT has the following characteristics that make it a practical tool in lead optimization. First, a large number of derivatives of lead compound that have the same binding mode with the active pocket of target protein can be created. These compounds can be accommodated in the active pocket without any atom bump with the target protein. Second, LEADOPT builds up new molecules using an efficient fragment-based strategy. Such a strategy can help avoid producing unreasonable molecular structures and to some extent keep the synthetic accessibility of the derived compounds. Third, ligand efficiency (LE) rather than scoring function is used as a measure to sort the newly generated molecules; LE has been considered as an effective strategy to help narrow focus to lead compounds with optimal combinations of physicochemical properties and pharmacological properties [22], [23]. Fourth, a number of pharmacokinetic and toxic properties of the generated molecules are evaluated, which makes the lead candidates pharmacologically acceptable. We shall in the following describe the details for the algorithm of LEADOPT. Then, LEADOPT will be evaluated with two retrospective cases. Finally, it will be applied to the structural optimizations of sorafenib, an inhibitor of vascular endothelial growth factor receptor 2 (VEGFR2), and R406, an inhibitor of spleen tyrosine kinase (SYK).

Section snippets

Fragment library construction

Since LEADOPT adopts a fragment-based approach for structural modifications to derive new molecular structures, a good fragment library is quite important for the quality of derived molecules. To establish such a fragment library, we first collected a total of 17,858 drug or drug-like molecules from CMC database, ChEMBL database [24], and DrugBank database [25]. Then, an in-house program written by us in C/C++ programming language was used to automatically build the fragment library. The

Development of LEADOPT

LEADOPT is an automatic tool developed for structure-based lead optimization. The overall workflow of LEADOPT is schematically depicted in Fig. 1. A detailed description of the algorithms of LEADOPT has been given in the Methods and Materials section. Here we just make a short summary for the workflow of LEADOPT. LEADOPT is started through input of the structure of ligand–receptor complex, which can be from either experimental X-ray crystal structure or molecule docking. Firstly, LEADOPT

Concluding remarks

In this investigation, we developed an automatic tool for structure-based lead optimization, termed LEADOPT. LEADOPT starts its work from an input structure of ligand–receptor complex, which can come from either x-ray crystal structure or molecular docking. The structural modifications in LEADOPT mainly include two operations: fragment growing and fragment replacing, which are restricted to carry out in the active pocket of target protein with the core scaffold structure of ligand kept

Acknowledgments

This work was supported by the 973 Program (2013CB967204), the 863 Hi-Tech Programs (2012AA020301, 2012AA020308), the National Natural Science Funds for Distinguished Young Scholar (81325021), the National S&T Major Project (2012ZX09501001-003), and partly by PCSIRT (NO: IRT13031).

References (46)

  • T. Kenakin

    Predicting therapeutic value in the lead optimization phase of drug discovery

    Nat. Rev. Drug Discov.

    (2003)
  • G.M. Keseru et al.

    The influence of lead discovery strategies on the properties of drug candidates

    Nat. Rev. Drug Discov.

    (2009)
  • C.W. Murray et al.

    The rise of fragment-based drug discovery

    Nat. Chem.

    (2009)
  • J.P. Kennedy et al.

    Application of combinatorial chemistry science on modern drug discovery

    J. Comb. Chem.

    (2008)
  • U. Rester

    From virtuality to reality – virtual screening in lead discovery and lead optimization: a medicinal chemistry perspective

    Curr. Opin. Drug Discov. Devel.

    (2008)
  • V.M. Popov et al.

    Towards in silico lead optimization: scores from ensembles of protein/ligand conformations reliably correlate with biological activity

    Proteins

    (2007)
  • W.L. Jorgensen

    Efficient drug lead discovery and optimization

    Acc. Chem. Res.

    (2009)
  • R.D. Cramer et al.

    Comparative molecular field analysis (CoMFA). 1. Effect of shape on binding of steroids to carrier proteins

    J. Am. Chem. Soc.

    (1988)
  • G. Klebe et al.

    Molecular similarity indices in a comparative analysis (CoMSIA) of drug molecules to correlate and predict their biological activity

    J. Med. Chem.

    (1994)
  • C.L. Kuo et al.

    Application of CoMFA and CoMSIA 3D-QSAR and docking studies in optimization of mercaptobenzenesulfonamides as HIV-1 integrase inhibitors

    J. Med. Chem.

    (2004)
  • B.M. Baron et al.

    CoMFA, synthesis, and pharmacological evaluation of (E)-3-(2-carboxy-2-arylvinyl)-4,6-dichloro-1H-indole-2-carboxylic acids: 3-[2-(3-aminophenyl)-2-carboxyvinyl]-4,6-dichloro-1H-indole-2-carboxylic acid, a potent selective glycine-site NMDA receptor antagonist

    J. Med. Chem.

    (2005)
  • B. Wendt et al.

    Toluidinesulfonamide hypoxia-induced factor 1 inhibitors: alleviating drug-drug interactions through use of PubChem data and comparative molecular field analysis guided synthesis

    J. Med. Chem.

    (2011)
  • S. Sartini et al.

    Structure-based optimization of tyrosine kinase inhibitor CLM3. Design, synthesis, functional evaluation and molecular modeling studies

    J. Med. Chem.

    (2014)
  • Cited by (22)

    • Synthesis and biological evaluation of diaryl urea derivatives designed as potential anticarcinoma agents using de novo structure-based lead optimization approach

      2020, European Journal of Medicinal Chemistry
      Citation Excerpt :

      Additionally the unpredictable pharmacokinetic properties and adverse side effects like fatigue, anemia, diarrhea, hypertension and hand-foot skin reaction [10] affect its clinical efficacy [11,12]. Discovery of new chemical entities (NCEs) as a focal point in drug research and development (R&D) includes lead discovery and optimization [13]. Lead discovery may be achieved by large-scale screening of chemical libraries or simply by chance.

    • X-ray crystal structure guided discovery of new selective, substrate-mimicking sirtuin 2 inhibitors that exhibit activities against non-small cell lung cancer cells

      2018, European Journal of Medicinal Chemistry
      Citation Excerpt :

      Notably, aminothiazoles and thienopyrimidinones were discovered as new types of selective SIRT2 inhibitors, which work via inducing the formation of an allosteric hydrophobic binding pocket, hence providing new insights into inhibitor design [21–24]. With the aid of LEADOPT [25], an automatic structure-based molecule design tool, we successfully identified the N-(3-(phenoxymethyl)phenyl)acetamide derivatives (Fig. 1b and Fig. S2) as selective SIRT2 inhibitors, which exhibited inhibitory activity against human breast cancer cell growth [26]. Here we describe the X-ray crystal structure guided structure-activity relationship (SAR) studies of new N-(3-(phenoxymethyl)phenyl)acetamide derivatives and their activities against non-small cell lung cancer.

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text