Elsevier

Drug Discovery Today

Volume 21, Issue 7, July 2016, Pages 1139-1146
Drug Discovery Today

Review
Informatics
Water, water, everywhere… It's time to stop and think

https://doi.org/10.1016/j.drudis.2016.05.009Get rights and content

Highlights

  • Six computational methods for water molecule placement and analysis reviewed alongside relevant case studies.

  • Field is approaching maturity.

  • Adoption into mainstream drug discovery programs will increase as more literature-based case studies are published.

Despite the numerous methods available for predicting the location and affinity of water molecules, there is still a degree of scepticism and reluctance towards using such information within a drug discovery program. Here, I review some of the most common and popular methods to assess whether these apparent concerns are justified. I suggest that the field is approaching maturity and that some methods are capable of giving quantitative predictions, which are confirmed experimentally. This suggests that water-placement methods should be used more widely to help direct chemistry efforts, although more successful examples are required to help validate the techniques.

Introduction

One of the axioms of 21st-century drug design and development is to consider the role of water molecules in the active site of the protein target. Built upon the initial studies by Poornima and Dean 1, 2, 3, together with those of Ladbury [4] and Dunitz [5] during the mid-1990s, this axiom broadly states that weakly bound water molecules in proteins should be targeted for ligand replacement, whereas strongly bound waters are ideal candidates for ligand stabilisation. As a result of this thinking, a plethora of papers has been published in the subsequent years that seek to explain or understand the nature of hydration in proteins. As shown in Fig. 1, a literature search conducted with SciFinder for ‘Water molecules in protein binding sites’ revealed a dramatic increase in the number of annual publications from the mid-90s to 2010.

Crystallography provides direct evidence for the presence of water molecules within proteins, although limiting factors, such as resolution and ambiguous electron density, can affect both the number and temperature factors of water molecules added into the structure 6, 7, 8. Studies have suggested that some solvent-accessible binding sites can either be partially filled or even devoid of water 9, 10, and crystallographic approaches are stretched to reliably predict the position of such water molecules that typically exhibit favourable entropy compared with highly localised water molecules that are most frequently observed in crystal structures [11]. As a result, computational approaches for locating water molecules have been developed to further advance our knowledge of active-site hydration.

Such approaches have helped to mature our understanding of the role of water in binding processes, and have built upon the original postulation that water is the ‘third party’ [4] in protein–ligand interfaces [12]. Computational methods have helped to highlight cases where water molecules can influence protein–ligand binding kinetics 13, 14, 15, affect thermodynamic binding signatures 16, 17, 18, or even govern selectivity within kinases 19, 20. They have also been used in conjunction with experimental procedures, such as isothermal titration calorimetry and X-ray crystallography, to explore the nature of the hydrophobic effect. Notably, the Whitesides group has highlighted that water networks can contribute to enthalpy/entropy compensation upon protein–ligand binding using computational approaches 21, 22, while also using them to understand hydration pattern changes upon the association of anions and proteins [23]. Despite the promising results and examples in the literature, it is still often unclear how these techniques can be used in a practical and prospective sense within a medicinal chemistry program and, crucially, what benefit they can bring over traditional approaches.

It can be argued that there still exists a great deal of scepticism within the medicinal chemistry community concerning the value of analysing water distributions. During a lead optimisation program, the decision to target a particular water molecule can be, and typically is, taken without knowing exactly how strongly bound it is to the complex, while introducing lipophilicity in hydrophobic regions to displace weakly bound or ‘unhappy’ water molecules is intuitive to a trained medicinal chemist. Such scepticism could arise from the literature; most publications simply use water distributions to retrospectively explain and rationalise unexpected trends in structure–activity relations (SAR), while the number of publications in which they are used to actually drive and direct the medicinal chemistry program is significantly fewer.

In this article, I look at some of the most successful and current computational methods for locating water molecules within protein-binding sites, and highlight examples where they have been beneficial to drug discovery projects. From this, I assess whether these methods have now evolved to a state where they can be relied upon to help direct medicinal chemistry efforts, or if further work and validation is still required.

Section snippets

WaterMap and inhomogenous solvation theory

Perhaps the most recognisable and well known of all water placement algorithms, Schrödinger's WaterMap package (see Glossary) 24, 25, has been extensively described and used in the chemical literature. WaterMap can be thought of as a two-stage process. First, a molecular dynamics (MD) simulation is performed in bulk solvent upon the system of interest, during which the protein and/or ligand is typically kept restrained. At the end of this simulation, typically 2 ns in length, the water positions

Monte Carlo-based methods

One drawback of MD-based approaches, such as WaterMap, is that they generally suffer from poor sampling when the binding site of interest is either occupied by a ligand, solvent inaccessible, or occluded, resulting in either excessive simulation time and/or an incomplete picture of solvation. In this regard, Monte Carlo (MC) simulations can offer a computationally efficient solution. Two of the most interesting methods in this regard are Grand Canonical Monte Carlo (GCMC) and Just Add Water

Density and probe-based methods

The methods described so far are based upon so-called ‘explicit solvent’, where all water molecules are described using a three- or four-point model with suitable partial charges. However, there are other ways of dealing with solvent that are computationally less demanding. Two such examples are GRID [48], which calculates the interaction energy of a water probe around a binding site, and SZMAP, which can be considered as a hybrid between explicit solvent and a Poisson–Boltzmann solvent

Alternative approaches

All of the above methods are based on physical principles; that is, they generally use a force field to place and score water molecules. However, there are numerous other methods described in the literature that use different procedures to address the issue of hydration. For example, knowledge-based approaches, such as AQUARIUS [64], SuperStar [65], and AcquaAlta [66], use crystallographic and structural information from the Cambridge Structural Database or Protein Data Bank to predict the

Concluding remarks

With all of the available methods, and their associated examples (Table 1), described in the literature, why is there still a degree of scepticism and confusion when it comes to using water-placement algorithms in medicinal chemistry programs? One of the most likely explanations is that water displacement is seen as common sense and does not require lengthy or extensive simulations to determine the apparently ‘obvious’. It is well recognised that water molecules in polar environments are

Acknowledgements

I would like to thank David Clark for his support and helpful discussions during the preparation of this review.

Glossary

3D-RISM
a popular tool, implemented in numerous packages such as AMBER and MOE, which uses integral equations and force fields to locate water molecules and derive their thermodynamic properties.
Force field
the equations and parameters used to calculate the inter- and intramolecular energies of molecules within a system. Some examples of common force fields are OPLS, AMBER, CHARMM, and GROMOS.
Grand-Canonical Monte Carlo (GCMC)
a Monte Carlo method that is capable of predicting the location of

References (74)

  • C.S. Poornima et al.

    Hydration in drug design. 3. Conserved water molecules at the ligand-binding sites of homologous proteins

    J. Comput-Aided Mol. Des.

    (1995)
  • J.D. Dunitz

    The entropic cost of bound water in crystals and biomolecules

    Science

    (1994)
  • A.M. Davis

    Application and limitations of X-ray crystallographic data in structure-based ligand and drug design

    Angew. Chem. Int. Ed.

    (2003)
  • R. Abel

    Contribution of explicit solvent effects to the binding affinity of small-molecule inhibitors in blood coagulation factor serine proteases

    ChemMedChem.

    (2011)
  • L. Wang

    Ligand binding to protein-binding pockets with wet and dry regions

    Proc. Natl. Acad. Sci. U. S. A.

    (2011)
  • T. Beuming

    Thermodynamic analysis of water molecules at the surface of proteins and applications to binding site prediction and characterization

    Proteins

    (2012)
  • P. Ball

    Biophysics: more than a bystander

    Nature

    (2011)
  • R.A. Pearlstein

    New hypotheses about the structure–function of proprotein convertase subtilisin/kexin type 9: analysis of the epidermal growth factor-like repeat A docking site using WaterMap

    Proteins

    (2010)
  • R.A. Pearlstein

    Contributions of water transfer energy to protein–ligand association and dissociation barriers: Watermap analysis of a series of p38α MAP kinase inhibitors

    Proteins

    (2013)
  • A. Bortolato

    Water network perturbation in ligand binding: adenosine A2A antagonists as a case study

    J. Chem. Inf. Model.

    (2013)
  • G. Hummer

    Molecular binding: under water's influence

    Nature Chemistry

    (2010)
  • R. Baron

    Water in cavity–ligand recognition

    J. Am. Chem. Soc.

    (2010)
  • P. Setny

    How can hydrophobic association be enthalpy driven?

    J. Chem. Theory Comput.

    (2010)
  • C. Barillari

    Analysis of water patterns in protein kinase binding sites

    Proteins

    (2011)
  • D.D. Robinson

    Understanding kinase selectivity through energetic analysis of binding site waters

    ChemMedChem

    (2010)
  • B. Breiten

    Water networks contribute to enthalpy/entropy compensation in protein–ligand binding

    J. Am. Chem. Soc.

    (2013)
  • M.R. Lockett

    The binding of benzoarylsulfonamide ligands to human carbonic anhydrase is insensitive to formal fluorination of the ligand

    Angew. Chem. Int. Ed.

    (2013)
  • J.M. Fox

    Interactions between Hofmeister anions and the binding pocket of a protein

    J. Am. Chem. Soc.

    (2015)
  • T. Young

    Motifs for molecular recognition exploiting hydrophobic enclosure in protein–ligand binding

    Proc. Natl. Acad. Sci. U. S. A.

    (2007)
  • R. Abel

    The role of the active site solvent in the thermodynamics of factor Xa-ligand binding

    J. Am. Chem. Soc.

    (2008)
  • T. Lazaridis

    Inhomogeneous fluid approach to solvation thermodynamics 1

    Theory. J. Phys. Chem. B

    (1998)
  • T. Beuming

    High-energy water sites determine peptide binding affinity and specificity of PDZ domains

    Protein Sci.

    (2009)
  • C. Higgs

    Hydration site thermodynamics explain SARs for triazolylpurines analogues binding to the A2A receptor

    ACS Med. Chem. Lett.

    (2010)
  • V. Myrianthopoulos

    Novel inverse binding mode of Indirubin derivatives yields improved selectivity for DYRK kinases

    ACS Med. Chem. Lett.

    (2013)
  • M.A. Brodney

    Spirocyclic sulfamides as β-secretase 1 (BACE-1) inhibitors for the treatment of Alzheimer's disease: utilization of structure based drug design, WaterMap, and CNS penetration studies to identify centrally efficacious inhibitors

    J. Med. Chem.

    (2012)
  • R. Horbert

    Optimization of potent DFG-in inhibitors of platelet derived growth factor receptorβ (PDGF-Rβ) guided by water thermodynamics

    J. Med. Chem.

    (2015)
  • C.N. Nguyen

    Grid inhomogeneous solvation theory: hydration structure and thermodynamics of the miniature receptor cucurbit[7]uril

    J. Chem. Phys.

    (2012)
  • Cited by (69)

    • The role of water in ligand binding

      2021, Current Opinion in Structural Biology
      Citation Excerpt :

      The ligand–target binding affinity is expressed as the free energy change of the binding reaction (ΔGb). The ΔGb can be engineered via ligand modifications affecting the hydration structure [17,24,30•,34,37••,38–42,43•,44•,45,46,47•,48]. For example, the target–ligand complex can be stabilized by inserting H-bonding functional groups that interact with or replace (Figure 3a) interfacial water molecules resulting in a favourable contribution to binding enthalpy (ΔHb) [43•].

    • WaterKit: Thermodynamic Profiling of Protein Hydration Sites

      2023, Journal of Chemical Theory and Computation
    View all citing articles on Scopus
    View full text