You are on page 1of 15

Expert Opinion on Drug Discovery

ISSN: 1746-0441 (Print) 1746-045X (Online) Journal homepage: https://www.tandfonline.com/loi/iedc20

Binding affinity in drug design: experimental and


computational techniques

Visvaldas Kairys, Lina Baranauskiene, Migle Kazlauskiene, Daumantas


Matulis & Egidijus Kazlauskas

To cite this article: Visvaldas Kairys, Lina Baranauskiene, Migle Kazlauskiene, Daumantas
Matulis & Egidijus Kazlauskas (2019) Binding affinity in drug design: experimental and
computational techniques, Expert Opinion on Drug Discovery, 14:8, 755-768, DOI:
10.1080/17460441.2019.1623202

To link to this article: https://doi.org/10.1080/17460441.2019.1623202

Published online: 31 May 2019.

Submit your article to this journal

Article views: 134

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=iedc20
EXPERT OPINION ON DRUG DISCOVERY
2019, VOL. 14, NO. 8, 755–768
https://doi.org/10.1080/17460441.2019.1623202

REVIEW

Binding affinity in drug design: experimental and computational techniques


Visvaldas Kairys a, Lina Baranauskiene b
, Migle Kazlauskiene c
, Daumantas Matulis b

and Egidijus Kazlauskas b


a
Department of Bioinformatics, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania; bDepartment of
Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania; cDepartment of
Biochemistry, University of Zurich, Zurich, Switzerland

ABSTRACT ARTICLE HISTORY


Introduction: In pharmaceutical design where future drugs are developed by targeting a specific Received 2 April 2019
chosen protein, the evaluation of ligand affinity is crucial. For this very purpose are a multitude of Accepted 21 May 2019
diverse methods which are continuously being improved, which, in turn, makes it difficult to choose KEYWORDS
which techniques to use in practice. Drug design; ligand;
Areas covered: In this review, the authors discuss both experimental and computational approaches inhibitor; affinity; free
for affinity evaluation. Basic principles, general limitations and advantages, as well as main areas of energy; Gibbs energy;
application in drug discovery, are overviewed for some of the most popular ligand binding assays. The binding; ITC; DSC; DSF; FTSA;
authors further provide a guide to affinity predictions, collectively covering several techniques that are TSA; AUC; electrophoresis;
used in the first stages of rational drug design. MST; thermophoresis;
Expert opinion: All affinity estimation methods have limitations and advantages that partially overlap fluorescence; ICD; BLI; SPR;
NMR; QSAR; molecular
and complement one another. Some of the suggested best practices include cross-verification of data
dynamics; QM/MM; MM-
using at least two different techniques and careful data interpretation. PBSA

1. Introduction Therefore, here, we will briefly overview experimental and


computational techniques devised for the evaluation of bind-
Target-oriented drug design aims to find a high affinity ligand
ing affinity. Hopefully, it will allow the reader to get a general
that would bind the target protein in order to block its dis-
idea of where a particular method would shine or fail as well
ease-associated function – catalytic activity or interaction with
as to help familiarize themselves with the most recent devel-
other molecules. This process usually starts with the screening
opments in the field.
of an initial library of ligands or their fragments to identify
All of the applicable methods reviewed here aim to evaluate
binders of the target protein, either experimentally or compu-
binding affinity. While pharmacologists often refer to biological
tationally (Figure 1). Often computational screens are used
activity or efficacy, which is the ability of a drug to produce
first to narrow the library down before proceeding with the
a functional response in living matter, rational drug design
experimental screening. Successful hits are then characterized
usually simplifies the studied system into only the ligand and
and further developed. At each such stage, the ligand binding
its target protein. The strength of the interaction between these
to the target protein is reevaluated.
two molecules is defined by either binding and dissociation
Correct and precise estimation of the binding affinity is
constants (Kb and Kd) or Gibbs energy of binding (ΔGb) and is
crucial throughout these essential drug design stages, as
commonly referred to as affinity. In a simple, reversible one
they commonly serve as the main factor for choosing promis-
protein – one ligand interaction case, equilibrium exists between
ing leads and their optimization strategies [1,2]. This high
the free molecules (P, protein, and L, ligand) and their complex
demand has facilitated the development of a number of dif-
(PL) that associate and dissociate at certain rates (described by
ferent techniques to assess or predict ligand binding. They all
rate constants kon and koff, respectively):
have their unique set of applications and limitations, and
several different methods are usually employed in parallel to kon
ensure data reliability. Multiple attempts have been made to P þ L ! PL (1)
systemize experimental [3,4] or computational [5,6] koff
approaches, but some of the methods have seen significant
Experimental researchers tend to define the binding affinity
improvements since. Since collaborations between bioinfor-
using equilibrium dissociation (Kd) or binding (Kb) constants:
maticians and biophysicists are becoming more and more
the norm, as illustrated by a variety of successful examples ½PL 1 kon
Kb ¼ ¼ ¼ (2)
from both industry and academia [1,7], both sides would ½P½L Kd koff
benefit from a better understanding of each other’s tools.

CONTACT Egidijus Kazlauskas ekazl@ibt.lt Department of Biothermodynamics and Drug Design, Institute of Biotechnology, Life Sciences Center, Vilnius
University, Saulėtekio al. 7, Vilnius LT-10257, Lithuania
© 2019 Informa UK Limited, trading as Taylor & Francis Group
756 V. KAIRYS ET AL.

2. Experimental approaches to evaluate affinity


Article highlights
Experimentalists can choose from a wide range of techniques
● Affinity determination is one of the cornerstones of modern drug designed to determine the affinity of two molecules. They vary
design.
● There are a lot of experimental and computational techniques avail- significantly in the underlying principles, suitability for differ-
able to evaluate a drug’s affinity towards its target, each with their ent cases, and experimental procedures. Majority of such
own pros and cons. approaches that are well established in drug design are sum-
● Isothermal titration calorimetry (ITC) measures the binding energy of
unmodified interactors directly at physiological temperature but is marized in Table 1. However, as the review space is quite
costly both materials- and time-wise. limited in comparison to the plethora of viable methods,
● High-throughput stability, mobility, or spectroscopic shift assays some of them are inevitably omitted.
allow for rapid affinity evaluation, which is readily exploited for
screening, but often are limited target-wise. Except for ITC, which records the binding energetics directly,
● In silico binding affinity prediction methods which encompass a vast and structural approaches, most of the methods discussed here
range of algorithms are advanced enough to be able to reduce the detect what portion of the ligand is being bound at certain
Root Mean Square Error of prediction for a series of compounds to ~5
kJ/mol. concentrations, and calculate the Kb from there. As seen from
● Specialized competitions for calculating the binding affinity show the Table 1, the methods significantly vary not only in their principles
latest trends in the computational methods and their performance, but also their detection limits. Typical experiment is performed
compared to older, venerable approaches.
by preparing at least 8–16 samples in serial dilutions: the con-
This box summarizes the key points contained in the article. centration of one of the interacting molecules (e.g. protein) is
kept constant while the other molecule (e.g. ligand) is diluted
over a wide range of concentrations. The techniques for
a specific case are chosen based on properties of the target
At equilibrium under standard conditions, the Gibbs energy protein and ligands – the detectability and stability. In this
(previously also known as free energy) of binding describes chapter, we will discuss the most prominent experimental
the energy difference between the two states: approaches for evaluating ligand binding to a protein target.
For convenience, we grouped these methods based on
½PL
ΔGb ¼ RTlnð½P½LÞ  RTln½PL ¼ RTln ¼ RTlnKb what binding event or product properties they are exploiting:
½P½L released energy, changes in stability, mobility, or spectro-
¼ RTlnKd ; (3) scopic characteristics upon binding, and structure-orientated
approaches. Notably, this sorting is arbitrary as a lot of these
where R is the gas constant and T is the absolute temperature techniques can fit several categories.
at which the binding occurs. ΔGb can also be dissected into its
components enthalpy (ΔHb) and entropy (ΔSb):
2.1. Isothermal titration calorimetry
ΔGb ¼ ΔHb  TΔSb (4)
Isothermal titration calorimetry (ITC) can measure thermodynamic
In computational approaches, ΔGb is often approximated to the energetics of binding directly, without any need of labeling,
protein–ligand interaction energies (i.e. essentially ΔHb), adding immobilization, or any other modification of the interactors.
additional terms appraising entropy. While researchers from dif- Moreover, ITC is highly versatile as any binding results in either
ferent fields tend to use either ΔGb or Kd/Kb to define the strength consumed or released heat, and there are no limitations for the
of binding, equation (3) clearly shows the direct relation between size, optical or other properties of the binders. Due to its generally
the binding (or dissociation) constant and Gibbs energy of bind- reliable data, wide target applicability, lack of modifications or
ing, with only temperature as an additional variable. Therefore, reporters, and multiple output binding parameters, ITC is being
while we will be discussing mainly binding constant determina- considered the ‘golden standard’ of protein–ligand interaction
tion in the section on experimental approaches and Gibbs energy measurements. ITC experiments provide a thermodynamic picture
calculations in the computational section, both of them refer to of binding, encompassing ΔGb and its enthalpic (ΔHb) and thus
the same affinity and can be compared with minimal calculations. calculated entropic (ΔSb) components.

100...000 compounds

Hit Lead Preclinical


Target selection
generation optimisation testing
Clinical trials 1 drug

screening: analysis:

Docking, ... QSAR, ...

FTSA, MST, ITC, SPR,


CE, NMR... NMR...

Figure 1. Affinity estimation methods in drug discovery process. Protein-ligand affinity determination methods are heavily relied upon during the early stages of
drug design and throughout the entire lead optimization process in order to produce drug candidates for preclinical and clinical trials from initial library of millions
of compounds.
Table 1. Overview of selected methods for experimental determination of the binding Gibbs energy.
Required
Protein amount of Typical Application
Short name Full name Information interactor Priciple samplea Kb range Typical Kd range example
ITC Isothermal titration calorimetry Kd, ΔHb, ΔGb, ΔSb, Any Heat release or uptake Medium-high 103–109 mM–nM [124]
stoichiometry
Stability shift assays:
ICD Isothermal chemical denaturation Kd Small ligand Change in chemical denaturation profile, as Medium 103–1012 mM–pM [8]
detected by fluorescence
FPSA Fluorescence pressure shift assay Kd, ΔV, Δβ Small ligand Change in unfolding point, as detected by High 103–1010 mM–sub-nM [9]
(PressureFluor) fluorescent probe
3 12
DSC Differential scanning calorimetry Kd, ΔCp, ΔHunfolding Small ligand Change in melting point, as detected by heat High 10 –10 mM–sub-pM [10]
release or uptake
FTSA, TSA, Fluorescence thermal shift assay (differential Kd, Tm Small ligand Change in melting point, as detected by Low 103–1013 mM– sub-pM [11]
DSF scanning fluorimetry, ThermoFluor) fluorescent probe
Mobility shift (separative) assays:
ED Equilibrium dialysis Kd, stoichiometry Small ligand Separation of bound and free ligands High 101–1012 mM–pM [12]
SEC Size exclusion chromatography Kd, size, stoichiometry Any Change in hydrodynamic size High 104–1013 µM–sub-pM [13]
WAC Weak affinity chromatography Kd Small ligands, Change in retention time due to binding Medium 102–107 mM–μM [14]
fragments
AUC Analytical ultracentrifugation Any
AUC sedimentation velocity Kd, shape, Change in sedimentation properties Medium 103–109 mM–nM [39]
conformation
changes
AUC sedimentation equilibrium Kd, size, stoichiometry Change in solution mass Medium 103–109 mM–nM [15]
EMSA Electrophoretic mobility shift assay Kd Nucleic acid Change in size and/or charge, as detected by Low 104–109 µM–nM [42]
mobility in electric field
CE Capillary electrophoresis Kd, (kon, koff), Any Change in size, shape, or charge, as detected by Low 102–107 mM–µM [16]
stoichiometry mobility in electric field
MST Microscale thermophoresis Kd, stoichiometry Any Change in size, charge, or hydration shell, as Low 103–1012 mM–pM [17]
detected by mobility in temperature gradient
Spectroscopic assays:
UV-vis UV-vis spectroscopy Kd Any Change in absorbance Medium <109 >nM [18]
Fluorescence Different applications of fluorescence Kd Any Change in fluorescence properties Low <1012 >pM [50]
spectroscopy
ICD Induced circular dichroism spectroscopy Kd Small ligand Change in elipticity Low 103–107 mM–µM [47]
SEC-MALS Size-exclusion chromatography with multi- Kd, molecule size Any with >3% Change in molecular size (complex formation) Medium reported Reported µM [19]
angle light scattering mass change 105–107
SPR Surface plasmon resonance Kd, kon, koff Any, >100 Da Changes in the refractive index Low 103–1011 mM–pM [20]
BLI Bio-layer interferometry Kd, kon, koff Any, >150 Da Shift in interferometric light reflection Low 103–1011 mM–pM [20]
NMR Nuclear magnetic resonance Any,
one <40 kDa
b b
Ligand-observed NMR Kd, (kon, koff) Change in chemical shift and relaxation or Medium 102–1010 mM–nM [21]
diffusion rates of the ligand(s)
b b
Protein-observed NMR Kd, structural change, Change in chemical shift and relaxation or High 103–109 mM – nM [22]
interacting residues diffusion rates of the protein
Kd – dissociation constant; ΔGb – Gibbs energy of binding; ΔHb – binding enthalpy change; ΔSb – binding entropy change; ΔV – volume change; Δβ – compressibility; ΔCp – heat capacity change; ΔHunfording – enthalpy change
of unfolding; Tm – melting temperature; kon – association rate constant; koff – dissociation rate constant.
a
EXPERT OPINION ON DRUG DISCOVERY

≤10 ng of protein required for Kd determination is considered low sample consumption, ≥1 mg – high.
b
In NMR, the reliably detectable affinity range significantly depends on kinetics.
757
758 V. KAIRYS ET AL.

During the measurement, one of the binding partners is 2.1.1. Differential scanning calorimetry
titrated with aliquots of the other under constant temperature, In case of DSC, large amount of protein is subjected to increasing
and the released or absorbed heat is measured. Construction temperature and its unfolding energetics profile (enthalpy, heat
of a binding isotherm of interaction heat as a function of capacity, and melting temperature Tm) is obtained. Determining
titrated ligand (Figure 2(a)) yields ΔHb, Kb (and therefore Kb via protein unfolding profiles enables precision over a much
ΔGb) with ΔSb calculated from equation (4), and usually stoi- wider range of binding constants as compared to the ITC
chiometry (n). The range for accurate ITC measurement win- approach. Just as ITC, DSC is a label and reporter free assay. On
dow for a given reaction is defined by c value [23] but too the other hand, it also involves a number of assumptions and
tight affinities can be tackled using competition ITC assays. thus should be considered carefully [32].
One approach is to premix a moderate affinity ligand with
excess of the target protein and titrate this mixture with the 2.1.2. Fluorescence thermal shift assay
high-affinity ligand [24]. While this approach also shows A considerably more efficient technique exploiting the profiles
whether both ligands compete for the same site, the data of protein unfolding is the fluorescence thermal shift assay
interpretation is not always straightforward. For example, the (FTSA; also referred to as TSA, DSF – differential scanning
difference between the affinities has to be sufficient to result fluorimetry, and ThermoFluor [33]). During the experiment,
in two distinct isotherm transition points. the protein samples with varying ligand concentrations are
Notably, due to its versatility, ITC detects the total heat being subjected to a constantly increasing temperature.
exchange, which can be caused not only by the binding Protein denaturation is tracked indirectly, by measuring the
event itself but also other simultaneous events, including fluorescence of a solvatochromic dye molecule, which reports
protonation, desolvation, and dilution. The true Kb values, on unfolded protein regions (Figure 2(b)). Thus instead of the
obtained after subtracting energies of other events occurring full thermodynamic denaturation profile, it yields only Kb and
in the measurement cell, are called ‘intrinsic’ and are better Tm but is significantly less time and reagent consuming.
suited for correlations with structural data [25,26]. These addi- Further, fluorescence-monitoring PCR machines can be easily
tional events can be unraveled using correct control experi- repurposed for FTSA experiments. While the assay has been
ments, further increasing the required experimental time. around for some time, its ease and high-throughput makes it
Thus, the raw determined Kb values, often referred to as a highly popular technique, with improvements still being
‘observed’, are still widely used to rank compounds. made [34]. It is especially widely used in the screening stages
Since it has been observed that more enthalpy-driven ligand (Figure 1). However, for precise affinity calculations, the
binding is common for natural inhibitors and correlates with method imposes some target limitations and works best for
higher selectivity [27], dissection of ΔGb into enthalpic and entro- simple single domain proteins.
pic components is useful for planning lead optimization. While
van’t Hoff relationship allows to indirectly calculate enthalpy
2.1.3. Cellular thermal shift assay
change from thermal stability assays (see next section), it relies
Interestingly, protein Tm shift upon ligand binding can be also
on sometimes erroneous assumption that enthalpy change is
exploited to evaluate target engagement in cells. In the
temperature-independent and thus does not always correlate
recently discovered cellular thermal shift assay (CETSA [35],),
with values directly measured using ITC [28]. Overall, ITC is not
various complex protein samples, including tissues, are ali-
a standard technique in the initial high-throughput drug screen-
quoted with and without ligand and subjected to different
ing stages due to its relatively high requirement of material and
temperatures. Soluble fractions are isolated from the lysates
time, but it has proven its usability in the later drug development
and quantified using antibody-based assays or mass spectro-
stages [29].
metry (MS). This approach allows to evaluate ligand efficacy in
the desired environment and is being rapidly developed [36].
However, one has to keep in mind that the data is heavily
2.2. Stability shift assays influenced by off-target binding and, in some setups, mem-
brane permeability and thus should not be considered as
Before ITC was well established, another calorimetric technique
actual affinity.
had gained the trust of pharmacologists – differential scanning
calorimetry (DSC). In general, stability shift assays rely on ligand’s
effect on the stability of the protein. Usually, drug leads stabilize
2.3. Mobility shift assays
the target protein and a favorable stability shift is observed. In
general, the stability is evaluated by denaturing the protein using To determine the binding affinity, mobility shift assays physi-
temperature, pressure [30], or chemical agents [31]. The stability cally separate the ligand or protein from the protein-ligand
shift per ligand added can be correlated to the Kb and thus ΔGb. complex and quantify these fractions. To this end, they exploit
However, complex multi-domain proteins and their assemblies are the differences in size, charge, hydrodynamic or other proper-
a challenge for any stability shift assay, as unfolding of different ties between these molecules and their assemblies. The ligand
domains and other substructures often happens independently (or protein) is usually detected using UV, fluorescence signal,
and thus yields multiple denaturation signals that can be hard to or MS. Determined bound fractions are plotted against target
interpret. Therefore for Kb determination, single-domain and sin- concentrations to calculate the Kd.
gle-binding site systems are favored as targets for this group of As these techniques rely on separability, they are inapplic-
methods. able when the difference between the complex and free
EXPERT OPINION ON DRUG DISCOVERY 759

a ITC b FTSA c MST


8 1.05
Time, s Data Data
0 1000 2000 3000 4000 Fit
19.8
0.95
6

Fluorescence, a.u.
19.7

Fluorescence, a.u.
19.6
Power, µJxs-1
0.85
19.5
4
19.4
0.75
19.3
19.2 2
0.65
19.1
19.0
Data 0 0.55
18.9 40 45 50 55 60 65 70 0 10 20 30 40
Baseline
18.8 Temperature, C Time, s
0 64 650

Relative fluorescence, a.u.


Data Data
-10 62 Fit Fit
640

temperature, C
Protein melting
H, kJxmol-1

-20
60
630
-30
58
-40 620
56
b

-50
Data
-60 Fit 54 // 610

-70 52 // 600
102 104 106
0 0.5 1 1.5 2 0
1.E-07 10-6
1.E-06 10-5
1.E-05 10-4
1.E-04 10-3
1.E-03 1
1.E+00 1.E+02 1.E+04 1.E+06
Molar ratio Total concentration of the added ligand, M Titrant, nM

Figure 2. Exemplary data of protein-ligand affinity measurements. The same protein-ligand pair was studied using three different approaches: (a) isothermal
titration calorimetry, (b) fluorescence thermal shift assay, and (c) microscale thermophoresis. The upper panels contain raw data, the lower panels – processed data
as used for calculating binding or dissociation constants. The ITC data comes from a single experiment while in case of FTSA and MST several measurements with
different ligand concentrations were conducted in parallel (12, with only 4 of them displayed in the raw data panel, and 16, respectively).

molecules is too insignificant based on the chosen separation throughout experiment measuring absorbance, refraction, or
criteria. Another general drawback for these assays is protein more sensitive fluorescence [40]. This distribution depends
or ligand interactions with the instrumentation (such as dia- solely on the molecular mass and not shape. Kd can thus be
lysis membrane or capillary walls) that distort the results. On calculated based on how the curvature changes at different
the other hand, most of these techniques are easy to use. protein: ligand ratios [41]. Naturally, it can only be applied for
Most notable examples of such techniques include equilibrium interactions with sufficient difference in molecular weight (the
dialysis (ED), analytical size exclusion chromatography (SEC), precision is usually 1–2%).
affinity selection chromatography, analytical ultracentrifuga-
tion (AUC), electrophoresis (CE, MCE, EMSA), and microscale 2.3.2. Electrophoresis
thermophoresis (MST). Another commonly used force to separate molecules is the
Chromatography techniques, relying on hydrodynamic size, electric field. While electrophoretic mobility shift assay (EMSA)
are usually reserved for studying inhibitors of protein–protein is still a popular technique to study protein affinity to nucleic
interactions or in combination with various spectroscopic acids and has been successfully applied to study inhibitors of
techniques for improved precision. Equilibrium dialysis, such interactions [42], the capillary and microchip electrophor-
where the semipermeable membrane between two reservoirs esis (CE and MCE; reviewed in [43]) offer more versatility and
ensures that only the small ligand can diffuse between two higher throughput. In typical affinity studies, the capillary is
connected chambers, nowadays is mostly used for assaying filled with the target protein and varying amounts of ligand is
drug interaction with plasma proteins [37]. injected (or vice versa), and electricity is applied. The techni-
que is widely used in lead screening. If the target protein is
2.3.1. Analytical ultracentrifugation exposed on the cell surface, living cells can be used in the
In the more widely used AUC, samples are subjected to cen- screening instead of purified proteins [44]. Furthermore, capil-
trifugal force. There are two types of AUC experiments – lary electrophoresis can also be used to study binding kinetics.
sedimentation velocity and sedimentation equilibrium. The
high-speed sedimentation velocity experiments separate 2.3.3. Microscale thermophoresis
molecular assemblies in the solution based on their mass While MST has been around for some time, it has gained much
and shape. Therefore, they are mostly used to characterize wider acceptance over the last several years as a routine
protein–protein interaction inhibitors and ligand effect on method for Kd determination (Figure 2(c)). The measurements
conformations of the targets (see Table 1 for examples). For are carried out in small glass capillaries, ensuring low sample
this and other hydrodynamics-based methods, solution prop- consumption. During the experiment, molecules move
erty prediction software suites such as HYDRO [38] are often through infrared laser-induced temperature gradients depend-
used to correlate observed data with high- and low-resolution ing on their size, charge, and hydration shell. Since at least one
structural information. of the parameters is altered upon binding of a ligand, this
While sedimentation velocity experiments can be used for method is highly versatile. Intrinsic Trp fluorescence or higher-
Kb measurements [39], sedimentation equilibrium is better sensitivity extrinsic fluorescent labels can be used to track the
suited for it. In these experiments, the sample is ultracentri- protein. Further, it can be used in cell lysates, with added
fuged until equilibrium between the centrifugal force and purified labeled protein [45] or by labeling the selected pro-
diffusion is reached. The distribution of molecules is tracked tein in the lysate [46]. However, stability of some targets inside
760 V. KAIRYS ET AL.

the capillary remains a challenge, and its results are still 10–100 nm) between the two fluorophores during the protein–
usually required to be verified using other, more established, ligand interaction. The fluorophores are generally artificially
methods. attached at suitably designed positions, for example, to monitor
conformational changes or interaction with nucleic acids.

2.4. Spectroscopic assays


2.4.2. Light-scattering-based assays
While spectroscopy is often used as detection component in
Light-scattering-based analysis of binding interaction uses
various other previously discussed assays, some spectroscopic
dynamic light scattering (DLS) or static light scattering (SLS)
methods, such as UV-vis, fluorescence, CD, IR, or NMR, are
to detect the size of molecular species in solution [51] and can
capable of ligand-binding analysis on their own. Their use is
be used to determine the absolute stoichiometry of multi-
based on changes in spectroscopic properties of the ligand or
component interaction. As it can detect both hetero- and
protein upon binding, as observed at equilibrium. Contrary to
homo-association events, it is an excellent tool for analysis of
the mobility shift assays, no physical separation of bound and
protein oligomerization and aggregation.
free fractions is required.
The most popular version is multi-angle light scattering
Circular dichroism (CD) spectroscopy is based on the ability
(MALS), especially when coupled with size exclusion chroma-
of the chiral molecule (e.g. protein) to absorb left and right
tography (SEC-MALS). Instead of relying on retention times,
circularly polarized light at different extents. This technique is
here SEC is used to separate free and ligand-bound species
generally used for secondary (far UV) and tertiary (near UV)
while the exact mass/size is detected by MALS. The measured
protein structure analysis. Upon binding to protein, small
difference of Rayleigh factor at a given angle is related to the
molecule ligand acquires induced CD spectrum (ICD). Varying
molecular weight of the solute. The affinity of the interacting
protein: ligand ratio in samples the ICD spectra can be used to
molecules can be determined according to the ratio of free
quantify binding affinity [47].
and ligand-bound protein.
Absorption spectroscopy for ligand binding is limited to
instances where protein or its ligand has a chromophore in UV
or visible range that is altered during binding. Most common 2.4.3. Optical spectroscopy assays
examples are proteins with cofactors, such as heme or flavin. Optical spectroscopy assays, namely surface plasmon reso-
nance (SPR) and bio-layer interferometry (BLI), are highly
2.4.1. Fluorescence-based assays regarded for their reliable binding affinity and kinetics data
Fluorescence spectroscopy for protein–ligand interaction ana- gained with relatively low sample consumption. These two
lysis is much more widely applicable [48]. Fluorescence-based major real-time reporter-free sensor-based techniques allow
detection is significantly more sensitive compared to absorp- for both kinetic and thermodynamic characterization of pro-
tion due to higher signal-to-noise ratio. It enables lowering the tein – ligand binding. They are used to determine the kinetics
working concentration range down to nanomolar scale and of molecule binding by measuring the binding and dissocia-
thus analysis of tight interactions. Molecule binding can induce tion rates (kon and koff constants). The affinity can be obtained
changes in fluorescence intensity and/or emission maximum either from the ratio of kinetic constants (equation (2)) or by
(center of mass). In practice, relative fluorescence intensity is steady-state calculations according to signal intensity. The
used for analysis more often than the absolute one. Analysis binding assay is performed with one binding partner being
using center of mass is often applied when the properties of immobilized on the sensor surface, and the other is free in
fluorophore depend on the environment (e.g. hydrophobicity). solution. The molecule in the solution can be either in pure
Either intrinsic fluorescence of the interacting molecules or the form or in a complex mixture, such as cell lysate. The main
extrinsic fluorescence probe can be used to monitor interactions. difference between these two methods is the principle used
The most common intrinsic fluorophore is Trp amino acid in for signal detection.
proteins. Trp spectral properties depend on its surroundings and BLI [52] uses white light and measures the interference
can have emission maximum from approx. 308 nm for deeply between two waves: one is reflected from internal reference
buried amino acids to approx. 350 nm for water accessible Trp layer, and the other from bio-compatible layer with immobi-
[49]. Extrinsic fluorophores can be either covalently attached to lized molecules. The spectral pattern depends on the thickness
one of the interacting molecules or produce the observed signal in of the molecular layer. Therefore, the signal is increasing with
its free form. Different techniques, such as the use of concurrent increasing size of the binding molecule. While other assay
fluorescent ligands, solvatochromic probes, etc., can be employed. formats are available, it is commonly performed in 96 well
Other variations of signal detection, such as differences in fluores- plate format simultaneously measuring interactions in 8 wells.
cence anisotropy or fluorescence polarization are also used [50]. The sample is reusable.
Förster resonance energy transfer (FRET) is yet another techni- The SPR [53] measures changes in the refractive index at
que for detection of intermolecular binding. It is based on the non- the surface of the sensor. Molecule binding at the surface
radiative energy transfer between two fluorophores: one fluoro- changes the refractive index proportionally to the mass
phore (donor) is excited by the external light source, and the bound. The resonance angle shift is monitored as a function
energy is transferred to another fluorophore (acceptor). The key of time during the binding event. In typical SPR experiment,
requirements to observe FRET are 1) overlap of donor emission one binding partner is coupled to a sensor while solution of
and acceptor excitation spectrum, and 2) close proximity (approx. the other flows through the sensor using microfluidics system.
EXPERT OPINION ON DRUG DISCOVERY 761

However, a microfluidics-free, 96-well plate version of SPR (FO- determination usually requires a competitor and is limited by
SPR) is also available [54]. binding kinetics.
The main issues concerning BLI and SPR come from possi- Overall, the usage of structural biology techniques in
ble immobilization effects. While SPR is much more sensitive drug design has been on the rise over the last decade
than BLI, for precise small molecule binding determination it due to significant technological advances and gaining
still usually requires quite sophisticated experimental setups. popularity of computational approaches that rely on initial
structural data.
2.5. Structural biology techniques
First of all, determination of structures of the targets and 3. In silico calculation of ligand-receptor binding
especially their complexes with lead compounds serve as the free energies
basis for most of the bioinformatic analysis tools outlined in Binding affinity prediction is one of the ‘holy grails’ of compu-
the next chapter. They are also a vital component for the tational chemistry [6]. To calculate binding free energies of
rational drug design, allowing to see what part of the ligand ligands to receptors a wide range of methods have been
interacts with which residues of the target protein as well as to developed, from chemometrics-based approaches to quantum
which direction the ligand could be expanded. Furthermore, mechanics. As this is a vast topic, only a cursory introduction is
nuclear magnetic resonance (NMR) spectroscopy is also cap- presented, to serve as a starting guide for a more in-depth
able of a more direct Kb determination. exploration of the field.
X-ray crystallography has always been the go-to method
for gaining structural information on protein–ligand interac-
3.1. QSAR and other chemoinformatics-based
tions (reviewed in [55]), with established and emerging dedi-
approaches
cated consortiums [56,57]. However, not all targets can be
readily crystallized and even obtained diffracting protein crys- Quantitative Structure–Activity Relationship (QSAR) and the clo-
tals cannot reveal conformational and chemical heterogeneity. sely related Quantitative Structure–Property Relationship (QSPR)
Rapid recent advancements in cryo-electron microscopy (cryo- are based on a hypothesis that a change in the structure of
EM) has brought its resolution to sufficient for analysis of a compound has a proportional effect on the biological activity
ligand interactions, as exemplified by various recent studies [63]. In practice, QSAR builds a (usually linear) equation with
[58], and can sometimes be used interchangeably with X-ray compound properties (so-called descriptors) being variables, and
crystallography. Small angle X-ray or neutron scattering (SAXS the compound activity (or binding affinity) as the target [64]. An
or SANS) allow to glimpse at general solution shape in a high- attractive advantage of QSAR is that in its simplest form it does not
throughput manner, which can be exploited for studying require knowledge of the receptor structure or the ligand’s bind-
inhibitors of protein–protein interaction and conformational ing mode. Probably the main disadvantage is that for the
dynamics [59,60]; however, so far they have remained under unknown compound, the knowledge of binding affinities of
the radar of most drug design studies and are commonly a series of related compounds needs to be known. Because of
applied mainly for drug carrier characterization. this, and because the method relies heavily on a variety of differ-
ent statistical measures, a vast array of QSAR protocols (often
2.5.1. Nuclear magnetic resonance referred to as 2D QSAR as they generally use two-dimensional
NMR is the only technique capable of obtaining atomic- structures of compounds), often with underlying weaknesses, exist
resolution structures in solution, albeit its targets are of limited [63,65–67].
size (<40 kDa). However, it has much more uses for assaying A variation of QSAR in which spatial structure of the ligand
ligand binding. In principle, during an NMR experiment, (and in some methods, the structure of the receptor with the
a magnetic field is applied to the sample, which affects the bound ligand) is used is known as 3D QSAR [68,69], and even
spins of the nuclei. The energy released by these nuclear spins of higher ‘dimensionalities’ [70]. Often these approaches are
coming back to their original states is detected and analyzed. derived from the 3D pharmacophore search [64]. Without
The energy differs for the same atom nuclei placed in different going too deep into the wide QSAR field, it will be sufficient
chemical environments – between different compounds or to say QSAR-based methods are very well researched and are
between the bound and unbound states. widely used in drug discovery process due to many successes,
Based on which interactor’s spectra is being recorded, rele- both in academia and in industry.
vant NMR techniques are classified into two major groups: Due to a historic connection with cheminformatics, it is a good
ligand-observed and protein-observed (reviewed in [61,62]). opportunity to mention the recent rise of popularity of Machine
Protein-observed techniques, requiring large amounts of isoto- Learning methods, which include Neural Networks, Support Vector
pically labeled protein, can be used to determine solution Machines, Random Forest and others [71]. It should be noted that
structures of small proteins and their complexes with ligands. some Machine Learning approaches make use of receptor infor-
Titration experiments can be used to precisely quantify Kd. mation. The applications of Machine Learning in a variety of drug
Ligand-observed techniques, while unable to yield high resolu- discovery areas have been reviewed elsewhere [72–74]. It should
tion, are cheaper as they require less protein and no isotopic also be mentioned that the recent Deep Learning and related
labeling. Moreover, in these assays larger size of proteins is only methods can be partially attributed to the emerging use of
an advantage and not a limitation. They are well suited for Graphical Processing Units (GPU), which allowed to significantly
ligand and especially fragment screening, while Kd speed up the calculations [72,74]. Among the recent success
762 V. KAIRYS ET AL.

stories, Deep Belief Networks were shown to dramatically outper- where ΔEvdw, ΔEelec, and ΔSA are changes in van der Waals
form QSAR models generated by Deep Neural Networks [75], and energy, electrostatic energy, and solvent accessible surface
remarkable performance of Random Forest-based scoring function area upon ligand binding, γ, δ – empirical fitting coefficients
ΔvinaRF20 [76] is further described in Section 3.3. which can vary to some extent [83]. ΔSA serves as an approx-
imation for the nonpolar solvation energy. ΔEelec above often
includes polar solvation energy obtained by solving electro-
3.2. Quantum mechanics-based methods static Poisson-Boltzmann (PB) differential equation [84] or by
On a theoretical level, probably the most accurate representa- using generalized Born (GB) approximation [85]. There exist
tion of atomic systems is based on quantum mechanics many variations and extensions of the method which differ by
because they take into account possible changes of electronic the way individual terms are handled, or by inclusion of addi-
states. Unfortunately, quantum calculations, also known as ab tional terms, often empirically tuned. The conformations for
initio calculations, are very CPU-intensive, since they take into the calculations are normally generated using molecular
consideration all electrons. The methods used to compute QM dynamics, but the equation can also be used to score docked
wavefunction vary from semiempirical models to Density conformations [86,87]. Mining Minima is one of the more
Functional Theory (DFT) functionals or more rigorous and interesting relatively new methods which goes beyond MM-
time-consuming methods such as Coupled Cluster calculations PB(GB)SA while trying to maintain sound physical grounds. In
[77]. The calculations become somewhat more manageable by Mining Minima, the local potential energy surface is explored
converting less important atoms to a non-quantum represen- computationally, leading to reasonable estimates to the con-
tation, and these methods are referred to as QM/MM formational entropy and the free energy of binding [88]. MM-
(Quantum Mechanics/Molecular Mechanics). The solvation PB(GB)SA applications in calculation of binding free energies
effects are taken into account via cheap but not very accurate have been recently reviewed by Genheden and Ryde [83].
implicit (continuum) solvation models or by using computa- Because of the convenience and computational speed,
tionally much more expensive explicit water molecules [6]. a great number of ‘scoring functions’ was developed. They
The electronic structure-based methods in principle are are computational estimates for the binding affinity for
able to achieve absolute accuracy of 1–3 kcal/mol [6]. It should a given conformation of ligand and protein [89,90]. Docking
be noted that the calculation of relative binding affinities of programs use scoring functions by necessity to evaluate the
a series of compounds is generally easier and more accurate quality of the docked pose. Molecular docking is an immen-
than calculation of the absolute binding affinities. A recent sely popular tool allowing to explore ligand-protein binding,
very thorough review of QM binding affinity calculations by and this lead to creation of a great number of docking pro-
Ryde and Söderhjelm [77] concludes with an interesting fact grams and web servers, as reviewed elsewhere [91–93].
that MM methods performed better than QM in the SAMPL4 However, the performance of the docking programs largely
host-guest blind binding affinity prediction challenge [78] due hinges on the quality of the underlying scoring functions.
to a variety of reasons, one of them being a simple cancella- Scoring functions are generally classified by the principles
tion of errors. Since MM parameters are often QM-derived, it is they were built upon: empirical, physics-based, knowledge-
obvious that QM calculations still have a large leeway for based, and machine-learning-based [90,94,95]. It also should
development. Indeed, the speed and accuracy of QM calcula- be borne in mind that the success of the scoring functions is
tions are slowly but constantly being improved. Development often system dependent [96]. A recent CASF-2016 test of 25
of efficient and accurate hybrid Density Functional Theory scoring functions applied to many receptor-ligand complexes
functionals by Grimme’s group [79] is but one example of [97] showed that Machine Learning-based scoring function
such progress. The main challenges and perspectives of the ΔvinaRF20 [76] performed strikingly better at scoring compared
quantum methods in computational chemistry have been to others (its Pearson correlation coefficient in the scoring
recently comprehensively outlined by Grimme and power test was 0.816, while the correlation coefficient of the
Schreiner [80]. runner-up, X-score [98], was only 0.631). However, some cau-
tion needs to be exercised as it is not clear to what extent
CASF-2016 test overlaps with the very large ΔvinaRF20 training
3.3. MM-PBSA and other related Force Field-based set, thereby reducing the value of the test.
methods. Scoring functions
One of the oldest and often used approaches to calculate free 3.4. Enhanced sampling MD methods
energies of binding is known as MM-PBSA (Molecular
Mechanics Poisson-Boltzmann Surface Area) or its variant MM- It is possible to obtain accurate distance vs. free energy pro-
GBSA (Molecular Mechanics Generalized Born Surface Area) files from a molecular dynamics calculation analyzing the
[81]. In fact, MM-GBSA served as the ‘null model’ against thermally driven (i.e. Poisson-Boltzmann) distribution of the
which other results were compared in the latest SAMPL bind- distances during the simulation [99]. Unfortunately, the system
ing affinity prediction challenge [82]. In its simplified form, the itself can easily become too big to traverse the conformational
binding energy between a ligand and a protein in MM-PBSA is space during the limited time available for the calculation. In
computed using the following expression, written in form addition, the potential energy barriers could be too high to be
which emphasizes the role of the surface area: surmounted during MD: for example, the ligand-protein com-
plex is often likely to stay bound during the simulation. This
ΔGbind ¼ ΔEvdw þ ΔEelec þ γΔSA þ δ (5) can be overcome by enhanced sampling techniques [100,101].
EXPERT OPINION ON DRUG DISCOVERY 763

One of such techniques is called umbrella sampling [100]. In where k is the Boltzmann’s constant, and the angular brackets
this method, the whole reaction path (for example, dissocia- denote ensemble average that was sampled using the VA poten-
tion/association coordinate of a complex) is split into a series tial [114–116]. For this formula to be of practical use, the poten-
of windows, and a harmonic bias is imposed for each window. tials VA and VB should have a significant thermally accessible
The bias helps to overcome potential barriers on the reaction overlap. To be able to achieve this, a series of purely artificial
path. At the end of many relatively short simulations for each intermediate states (windows) is created between A and B, and
window, the resulting distributions are combined and ana- ΔGA–B is calculated as the sum of relatively small changes of G for
lyzed, for example, using the Weighted Histogram Analysis each window. This method is often used to calculate changes of
Method (WHAM) [102]. Umbrella sampling has often been the ligand binding energy to the receptor where one functional
used to explore the energetic profile of transport of various group is gradually transformed into another one [117]. Because
species through transmembrane channels (e.g. ref [103].), but of this, the method is also known as alchemical perturbation.
this method can also be successfully applied to investigate When the width of the window approaches zero, the sum
ligand-enzyme binding. For example, umbrella sampling changes to integral, and the approach is being referred to as
showed good agreement with experimental results for benzo- Thermodynamic Integration (TI) [115]. The free energy methods
nitrile association with wild type and mutant lysozyme T4, and are considered as the ‘cutting edge’ approaches and have been
also with MM-PBSA calculations [104]. successfully applied for biochemical systems [117], even though
Another MD based enhanced sampling approach is called the results can be somewhat system dependent [118].
Steered Molecular Dynamics. As its name suggests, the ligand A significantly simplified version of FEP and therefore rela-
is dragged (‘steered’) by additional force out of the binding tively easy to implement is LIE (Linear Interaction Energy)
pocket until both molecules can be considered separated, and [119]. The method requires performing two MD simulations,
the work needed to such separation can be used to estimate one of the ligand in pure solvent, and another one of ligand/
the free energy. Moreover, energy vs. reaction coordinate receptor complex, also in solvent. The binding affinity is com-
profile can be built, also known as PMF (Potential of Mean puted as an average change of the interactions felt by the
Force), where the reaction coordinate can be, for example, the ligand when it changes environment from the pure solvent to
distance between the protein and the ligand [105]. In a recent the receptor/solvent (with some scaling factors for van der
study using three protein-ligand systems, Okimoto et al. [106] Waals and Coulombic energy components). In spite of relative
found that Steered MD overestimated the binding affinities, simplicity, the method shows a relatively good ~1 kcal/mol
but ranked ligands quite accurately. RMSE accuracy with respect to the experimental binding affi-
Among the relatively new enhanced sampling methods nities [120,121].
metadynamics and its many variants should be certainly men-
tioned [107,108]. In metadynamics, every n-th MD step a small
Gaussian type ‘hill’ (bias potential) is deposited along the
chosen internal coordinate. Since the simulation tends to 4. Conclusions
move to low energy regions, the bias potential gradually fills Binding affinity is one of the major factors in target-oriented
deep energy wells first. At the end of simulation, the depos-
drug design. The main goal is to produce a ligand that would
ited bias potential is explored, and the energy vs. coordinate bind with high affinity to the target protein at functionally
profile is built. Metadynamics based approaches can be used important site and would exhibit low off-target binding. As
not only to explore ligand binding to the protein but also to
this brief overview shows, while there is a wide variety of
build more complex free energy landscapes along various experimental (see Table 1) and computational methods to
chosen internal coordinates [109,110]. The binding affinity estimate ligand affinity to the target protein, neither of them
prediction accuracy using metadynamics can reach less than
is perfect. Therefore, the recommended techniques vary with
1 kcal/mol RMSE [111]. the chosen protein-ligand system. While computational meth-
As a side note, the conformational sampling statistics can ods are much cheaper, they are tightly intertwined with the
be obtained not only from simulations but also from experi-
experimental approaches. The development and assessment of
mental data (so-called ‘knowledge-based’ approaches [112]). most computational techniques relies heavily on experimental
The knowledge-based scoring functions are able to compete data. Therefore, its quality strongly influences the development
rather well against many other scoring functions, e.g. a recent of theoretical approaches. Moreover, the predictions still have
Convex-PL scoring function example [113]. to be verified using an experimental approach. The balancing
between high-throughput and accuracy is often achieved by
3.5. Free energy methods using faster but less reliable techniques to screen the initial
long list of putative candidates and then relying on more pre-
A group of approaches is known, somewhat tautologically, as
cise methods to characterize promising leads. As exemplified by
‘free energy methods’ and is based on rigorous statistical
inhibitor design of human carbonic anhydrases, renin, and
thermodynamics as applied to Molecular Dynamics or Monte
Plasmodium falciparum proteases [122–124], often docking is
Carlo simulations. In the free energy perturbation (FEP) theory,
combined with structural studies and affinity determination
the change of the free energy between states A and B can be
using ITC or FTSA. In general, drug design field is a great exam-
written as
ple of how combination of different approaches drives the
ΔGAB ¼ kTlnheðVB VA Þ=kT iA (6) discoveries forward.
764 V. KAIRYS ET AL.

5. Expert opinion scoring. These competitions are useful resources for comparing
the performance of various methods, and also for discovering
While lead optimization cannot rely on binding affinity
emerging new trends in computing.
(defined as either Gibbs energy or the binding/dissociation
Regardless of the experimental or computational approach,
constant) alone, it serves as a major factor. Therefore, it is
data analysis and critical evaluation of the results remain an
critical to evaluate it correctly before making decisions con-
important issue. In the end, all data are only as good as its analysis
cerning further ligand design. This involves choosing suitable
and interpretation. For data processing using stability and mobility
methods, correctly planning the experiment, and properly
shift assays where a large range of concentrations are plotted, we
interpreting the data.
recommend using a logarithmic scale as it makes it easier to
While this review might serve as a general guideline for which
evaluate the fit and Kd estimation. Global fit analysis is encouraged
method to choose, it is often hard to predict which approach will
for calculating the Kd from replicates. Nowadays, fully automated
be suitable for each selected system. Therefore, to obtain reliable
data processing software is often available, especially for the more
affinity data, we promote the application of several complemen-
popular techniques. For example, SEDPHAT [126] is often used for
tary techniques. It is most convenient to use a high-throughput
processing data, including that of AUC and ITC, and is highly
method (e.g. FTSA) to sort through potential ligands in combina-
regarded for the global fitting functionality. NITPIC is another
tion with a more in-depth analysis of the promising hits using ITC
highly useful tool for processing raw ITC data [127]. However,
or SPR. ITC provides detailed thermodynamic information, which
this simplification should not be regarded as lack of necessity for
helps to design enthalpically beneficial improvements, while SPR
the ability to critically evaluate the results. In fact, a lot of issues
describes binding kinetics.
arise from the careless and uneducated use of automatically
When choosing which methods to use, one must also keep in
processed data, as explained in the case of X-ray crystallography
mind that most of the methods are not as straightforward as
in a recent review [55]. To ensure data reproducibility, we also
advertised. Some might require more sophisticated equipment
encourage the inclusion of raw data in the publications in addition
or experimental planning for drug discovery-related applications
to the provision of sufficient experimental details in the methods
or a well-trained specialist. A common mistake in experimental
sections.
planning is working with unsuitable concentrations of ligand and/
or protein. The concentrations should be chosen depending on Finally, when integrating data from diverse approaches, their
the strength of interaction and signal-to-noise ratio. When plan- differences have to be kept in mind. For example, lysate and
ning the dilution series for stability, mobility, or spectroscopic purified protein data might differ significantly due to additional
assays, it is recommended to cover a range of 6 orders of cellular interactors. While this might lead to incomparable affinity
magnitude. values, it also provides more insight into the interaction in ques-
As this brief overview shows, most modern computational tion. For the simple ranking of ligands by best performance,
methods are able to make useful predictions, reaching ~5 kJ/mol experimental conditions that mimic drug use (such as CETSA
(~1 kcal/mol) RMSE. However, the success of an in silico method is using tissues) are best suited. However, for rational design studies
not always guaranteed to work on your system due to several that try to draw upon structural, computational, and biophysical
reasons: data overfitting when calibrating methods; success is data, it is especially important to ensure that the data being
often system-dependent; lastly, the negative results are rarely compared shed light on intermolecular interactions. For good
published. For this reason, it can be important to use methods correlation with structural data, ‘intrinsic’ binding parameters
with a realistic underlying physical model. Some additional limita- should be used [25,26], with the binding-linked events such as
tions may arise due to computing power restrictions, e.g. most ligand protonation subtracted from the ‘observed’ data.
molecular dynamics and quantum mechanics-based approaches To sum up, there is a wide variety of experimental and
can be CPU-demanding, but in some cases the accuracy is worth it. computational methods that are constantly expanding both
Also, much improved accuracy is expected when comparing rela- because of high demand for reliable and precise affinity quan-
tive binding affinities, instead of absolute ones. Of course, in the tification tools and because, so far, none of the described
absence of the receptor structure, the chemoinformatics-based methods are considered perfect for all possible situations.
methods are the most helpful. Among those, AI-based approaches For now, the main goals of future technique improvements
show clear promise, but one should be aware that they work best remain enhancing throughput, accuracy, and accessibility.
on systems which are included in their training set. Finally, the current amount of experimental affinity mea-
A useful resource for comparing performances of various com- surements is required because, despite enormous efforts, the
putational methods is through the D3R Grand Challenge [125], general trends of Structure–Affinity Relationship (SAR) are still
a regularly held competition in which various scientific groups not well understood. Therefore, systematization of existing
contest the computational prediction of various aspects of host/ experimental data, especially databases that compare affinities
guest binding. Additionally, SAMPL is a closely related computa- of chemically and/or structurally similar compounds [128], is
tional prediction challenge involving host-guest systems [82], important to identify correlations and to pinpoint the impact
which can be considered as ‘miniature models’ for protein–ligand of small chemical changes. Furthermore, additional thermody-
interactions. Because of the smaller sizes of these systems, namic parameters (enthalpy and entropy) should also be
amongst SAMPL entrants are many groups who use CPU- regarded, and the influence of binding-linked events should
intensive methods, such as quantum mechanics and MD-based be omitted in order to facilitate our understanding of the
approaches, while D3R is far more focused on docking and molecular recognition phenomenon.
EXPERT OPINION ON DRUG DISCOVERY 765

Acknowledgements 12. Pelkonen L, Tengvall-Unadike U, Ruponen M, et al. Melanin binding


study of clinical drugs with cassette dosing and rapid equilibrium
We apologise to all authors whose work we were unable to mention due dialysis inserts. Eur J Pharm Sci. 2017;109:162–168.
to space constraints. M. Kazlauskiene is a European Molecular Biology 13. Grither WR, Longmore GD. Inhibition of tumor–microenvironment
Organization Fellow (ALTF 1087–2018). interaction and tumor invasion by small-molecule allosteric inhibi-
tor of DDR2 extracellular domain. Pnas. 2018;115:E7786–E7794.
14. Meiby E, Simmonite H, le Strat L, et al. Fragment screening by weak
affinity chromatography: comparison with established techniques
Declaration of interest for screening against HSP90. Anal Chem. 2013;85:6756–6766.
15. Ruiz-Ramos A, Velázquez-Campoy A, Grande-García A, et al.
The authors have no other relevant affiliations or financial involvement with
Structure and functional characterization of human aspartate trans-
any organization or entity with a financial interest in or financial conflict with
carbamoylase, the target of the anti-tumoral drug PALA. Structure.
the subject matter or materials discussed in the manuscript.
2016;24:1081–1094.
16. Aizpurua-Olaizola O, Torano JS, Pukin A, et al. Affinity capillary
electrophoresis for the assessment of binding affinity of
carbohydrate-based cholera toxin inhibitors. Electrophoresis.
Reviewer Disclosures 2018;39:344–347.
Peer reviewers on this manuscript have no relevant financial or other 17. Zhang B, Li J, Yang X, et al. Crystal structures of membrane trans-
relationships to disclose. porter MmpL3, an anti-TB drug target. Cell. 2019;176:636–648.e13.
18. Tayyab S, Sam SE, Kabir MZ, et al. Molecular interaction study of an
anticancer drug, ponatinib with human serum albumin using spec-
ORCID troscopic and molecular docking methods. Spectrochim Acta A Mol
Biomol Spectrosc. 2019;214:199–206.
Visvaldas Kairys http://orcid.org/0000-0002-5427-0175 19. Pollock JF, Ashton RS, Rode NA, et al. Molecular characterization of
Lina Baranauskiene http://orcid.org/0000-0002-9924-9177 multivalent bioconjugates by size-exclusion chromatography (SEC)
Migle Kazlauskiene http://orcid.org/0000-0003-2329-0410 with multi-angle laser light scattering (MALS). Bioconjug Chem.
Daumantas Matulis http://orcid.org/0000-0002-6178-6276 2012;23:1794–1801.
Egidijus Kazlauskas http://orcid.org/0000-0001-8430-7349 20. Yang D, Singh A, Wu H, et al. Comparison of biosensor platforms in
the evaluation of high affinity antibody-antigen binding kinetics.
Anal Biochem. 2016;508:78–96.
21. Igonet S, Raingeval C, Cecon E, et al. Enabling STD-NMR fragment
References screening using stabilized native GPCR: A case study of adenosine
receptor. Sci Rep. 2018;8:8142.
Papers of special note have been highlighted as either of interest (•) or of 22. Toyama Y, Kontani K, Katada T, et al. Conformational landscape
considerable interest (••) to readers. alternations promote oncogenic activities of ras-related C3 botuli-
1. Ferenczy GG, Keserű GM. The impact of binding thermodynamics num toxin substrate 1 as revealed by NMR. Sci Adv. 2019;5:
on medicinal chemistry optimizations. Future Med Chem. eaav8945.
2015;7:1285–1303. 23. Broecker J, Vargas C, Keller S. Revisiting the optimal c value for
● A review of several case studies where measurements of isothermal titration calorimetry. Anal Biochem. 2011;418:307–309.
affinity (and in general thermodynamic parameters) signifi- 24. Krainer G, Keller S. Single-experiment displacement assay for quan-
cantly aided further ligand optimisation. tifying high-affinity binding by isothermal titration calorimetry.
2. Freire E. A thermodynamic approach to the affinity optimization of Methods. 2015;76:116–123.
drug candidates. Chem Biol Drug Des. 2009;74:468–472. 25. Leavitt S, Freire E. Direct measurement of protein binding ener-
3. Renaud J-P, Chung C, Danielson UH, et al. Biophysics in drug getics by isothermal titration calorimetry. Curr Opin Struct Biol.
discovery: impact, challenges and opportunities. Nat Rev Drug 2001;11:560–566.
Discov. 2016;15:679–698. 26. Perozzo R, Folkers G, Scapozza L. Thermodynamics of protein–
4. Wätzig H, Oltmann-Norden I, Steinicke F, et al. Data quality in drug ligand interactions: history, presence, and future aspects. J Recept
discovery: the role of analytical performance in ligand binding Signal Transduct. 2004;24:1–52.
assays. J Comput Aided Mol Des. 2015;29:847–865.
27. Claveria-Gimeno R, Vega S, Abian O, et al. A look at ligand binding
5. Leelananda SP, Lindert S. Computational methods in drug
thermodynamics in drug discovery. Expert Opin Drug Discov.
discovery. Beilstein J Org Chem. 2016;12:2694–2718.
2017;12:363–377.
6. Jensen JH. Predicting accurate absolute binding energies in aqu-
28. Holdgate GA, Ward WHJ. Measurements of binding thermody-
eous solution: thermodynamic considerations for electronic struc-
namics in drug discovery. Drug Discov Today. 2005;10:1543–1550.
ture methods. Phys Chem Chem Phys. 2015;17:12441–12451.
● Good introduction to the thermodynamics of binding, espe- 29. Baranauskiene L, Kuo TC, Chen WY, et al. Isothermal titration
cially from the viewpoint of quantum calculations. calorimetry for characterization of recombinant proteins. Curr
7. Kuhn B, Guba W, Hert J, et al. A real-world perspective on mole- Opin Biotechnol. 2019;55:9–15.
cular design. J Med Chem. 2016;59:4087–4102. 30. Petrauskas V, Baranauskienė L, Zubrienė A, et al. Isothermal titration
8. Ross P, Weihofen W, Siu F, et al. Isothermal chemical denaturation calorimetry and fluorescent thermal and pressure shift assays in pro-
to determine binding affinity of small molecules to G-protein tein–ligand interactions. In: M. Bastos, editor. Biocalorimetry. New York:
coupled receptors. Anal Biochem. 2015;473:41–45. CRC press; 2016. p. 261–280.
9. Toleikis Z, Cimmperman P, Petrauskas V, et al. Determination of the 31. Schön A, Brown RK, Hutchins BM, et al. Ligand binding analysis and
volume changes induced by ligand binding to heat shock protein 90 screening by chemical denaturation shift. Anal Biochem.
using high-pressure denaturation. Anal Biochem. 2011;413:171–178. 2013;443:52–57.
10. Zubrienė A, Matulienė J, Baranauskienė L, et al. Measurement of 32. Garbett NC, Chaires JB. Thermodynamic studies for drug design
nanomolar dissociation constants by titration calorimetry and ther- and screening. Expert Opin Drug Discov. 2012;7:299–314.
mal shift assay – radicicol binding to Hsp90 and ethoxzolamide ●● Good introduction to the importance of binding thermo-
binding to CAII. Int J Mol Sci. 2009;10:2662–2680. dynamics and why ITC remains invaluable in drug research.
11. Dudutienė V, Matulienė J, Smirnov A, et al. Discovery and charac- 33. Pantoliano MW, Petrella EC, Kwasnoski JD, et al. High-density min-
terization of novel selective inhibitors of carbonic anhydrase IX. iaturized thermal shift assays as a general strategy for drug
J Med Chem. 2014;57:9435–9446. discovery. J Biomol Screen. 2001;6:429–440.
766 V. KAIRYS ET AL.

34. Bai N, Roder H, Dickson A, et al. Isothermal analysis of thermofluor 53. Patching SG. Surface plasmon resonance spectroscopy for character-
data can readily provide quantitative binding affinities. Sci Rep. isation of membrane protein–ligand interactions and its potential for
2019;9:2650. drug discovery. Biochim Biophys Acta - Biomembr. 2014;1838:43–55.
● New FTSA variation: simplifications come with higher ● Extensive review on SPR application to protein-ligand
limitations. binding.
35. Molina DM, Nordlund P. The cellular thermal shift assay: a novel bio- 54. Lu J, Van Stappen T, Spasic D, et al. Fiber optic-SPR platform for fast
physical assay for in situ drug target engagement and mechanistic and sensitive infliximab detection in serum of inflammatory bowel
biomarker studies. Annu Rev Pharmacol Toxicol. 2016;56:141–161. disease patients. Biosens Bioelectron. 2016;79:173–179.
36. Shaw J, Leveridge M, Norling C, et al. Determining direct binders of 55. Zheng H, Handing KB, Zimmerman MD, et al. X-ray crystallography
the androgen receptor using a high-throughput cellular thermal over the past decade for novel drug discovery – where are we
shift assay. Sci Rep. 2018;8:163. heading next? Expert Opin Drug Discov. 2015;10:975–989.
37. Vuignier K, Schappler J, Veuthey J-L, et al. Drug–protein binding: 56. Goulding CW, Apostol M, Anderson DH, et al. The TB structural
a critical review of analytical tools. Anal Bioanal Chem. 2010;398:53–66. genomics consortium: providing a structural foundation for drug
●● Comparison of several different experimental affinity discovery. Curr Drug Targets Infect Disord. 2002;2(2):121–141).
determination techniques. 57. Keefe LJ, Stoll VS. Accelerating pharmaceutical structure-guided drug
38. de la Torre JG. The HYDRO software suite for the prediction of design: a successful model. Drug Discov Today. 2019;24:377–381.
solution properties of rigid and flexible macromolecules and nano- 58. Renaud J-P, Chari A, Ciferri C, et al. Cryo-EM in drug discovery:
particles. In: Uchiyama S, Arisaka F, Stafford WF, editors. Analytical achievements, limitations and prospects. Nat Rev Drug Discov.
ultracentrifugation: instrumentation, software, and applications. 2018;17:471–492.
Tokyo: Springer Japan; 2016. p. 195–217. 59. Andersson CD, Martinez N, Zeller D, et al. Influence of enantiomeric
39. Cole JL, Correia JJ, Stafford WF. The use of analytical sedimentation inhibitors on the dynamics of acetylcholinesterase measured by
velocity to extract thermodynamic linkage. Biophys Chem. elastic incoherent neutron scattering. J Phys Chem B.
2011;159:120–128. 2018;122:8516–8525.
40. Chaturvedi SK, Ma J, Zhao H, et al. Use of fluorescence-detected 60. Chen P, Hennig J. The role of small-angle scattering in structure-based
sedimentation velocity to study high-affinity protein interactions. screening applications. Biophys Rev. 2018;10:1295–1310.
Nat Protoc. 2017;12:1777–1791. 61. Harner MJ, Mueller L, Robbins KJ, et al. NMR in drug design. Arch
41. Gordon SE, Perugini MA. Protein-Ligand Interactions. In: Biochem Biophys. 2017;628:132–147.
Uchiyama S, Arisaka F, Stafford WF, editors. Analytical ultracentri- 62. Nitsche C, Otting G. NMR studies of ligand binding. Curr Opin
fugation: instrumentation, software, and applications. Tokyo: Struct Biol. 2018;48:16–22.
Springer; 2016. p. 329–353. ● Extensive review on NMR application to protein-ligand
42. Zapp ML, Stern S, Green MR. Small molecules that selectively block binding.
RNA binding of HIV-1 rev protein inhibit rev function and viral 63. Scior T, Medina-Franco JL, Do Q-T, et al. How to recognize and
production. Cell. 1993;74:969–978. workaround pitfalls in QSAR studies: a critical review. Curr Med
43. Ouimet CM, D’amico CI, Kennedy RT. Advances in capillary electro- Chem. 2009;16:4297–4313.
phoresis and the implications for drug discovery. Expert Opin Drug 64. Leach AR, Gillet VJ. An introduction to chemoinformatics.
Discov. 2017;12:213–224. Netherlands: Springer; 2007.
● Recent developments in capillary electrophoresis. 65. Golbraikh A, Tropsha A. Beware of q2! J Mol Graphics Modell.
44. Wu R, Zhu K, Zhang X, et al. Nonimmobilized biomaterial capillary 2002;20:269–276.
electrophoresis for screening drugs targeting human glucose trans- 66. Dearden JC, Cronin MTD, Kaiser KLE. How not to develop
porter 1. Anal Chem. 2017;89:12951–12959. a quantitative structure–activity or structure–property relationship
45. Wienken CJ, Baaske P, Rothbauer U, et al. Protein-binding assays in (QSAR/QSPR). SAR QSAR Environ Res. 2009;20:241–266.
biological liquids using microscale thermophoresis. Nat Commun. 67. Cherkasov A, Muratov EN, Fourches D, et al. QSAR modeling: where
2010;1:100. have you been? Where are you going to? J Med Chem.
46. Bartoschik T, Galinec S, Kleusch C, et al. Near-native, site-specific 2014;57:4977–5010.
and purification-free protein labeling for quantitative protein inter- 68. Verma J, Khedkar VM, Coutinho EC. 3D-QSAR in drug design–a
action analysis by microscale thermophoresis. Sci Rep. 2018;8:4977. review. Curr Top Med Chem. 2010;10:95–115.
● MST methodology for studying protein-ligand interactions in 69. Melo-Filho CC, Braga RC, Andrade CH. 3D-QSAR approaches in drug
lysates. design: perspectives to generate reliable CoMFA models. Curr
47. Tedesco D, Bertucci C. Induced circular dichroism as a tool to Comput Aided Drug Des. 2014;10:148–159.
investigate the binding of drugs to carrier proteins: classic 70. Damale MG, Harke SN, Kalam Khan FA, et al. Recent advances in
approaches and new trends. J Pharm Biomed Anal. 2015;113:34–42. multidimensional QSAR (4D–6D): a critical review. Mini Rev Med
● Extensive review on ICD application to protein-ligand Chem. 2014;14:35–55.
binding. 71. Mitchell JBO. Machine learning methods in chemoinformatics.
48. Krishnamoorthy G. Fluorescence spectroscopy for revealing Wiley Interdiscip Rev Comput Mol Sci. 2014;4:468–481.
mechanisms in biology: strengths and pitfalls. J Biosci. ● A good starting place to learn more about a wide range of
2018;43:555–567. Machine Learning as applied to molecules.
● Discussion on different applications of fluorescence 72. Chen H, Engkvist O, Wang Y, et al. The rise of deep learning in drug
spectroscopy. discovery. Drug Discov Today. 2018;23:1241–1250.
49. Hixon J, Reshetnyak YK. Algorithm for the analysis of tryptophan 73. Hessler G, Baringhaus K-H. Artificial intelligence in drug design.
fluorescence spectra and their correlation with protein structural Molecules. 2018;23:2520.
parameters. Algorithms. 2009;2:1155–1176. 74. Ghasemi F, Mehridehnavi A, Pérez-Garrido A, et al. Neural network
50. Stoddart LA, White CW, Nguyen K, et al. Fluorescence- and and deep-learning algorithms used in QSAR studies: merits and
bioluminescence-based approaches to study GPCR ligand drawbacks. Drug Discov Today. 2018;23:1784–1790.
binding. Br J Pharmacol. 2016;173:3028–3037. 75. Ghasemi F, Mehridehnavi A, Fassihi A, et al. Deep neural network in
51. Some D. Light-scattering-based analysis of biomolecular QSAR studies using deep belief network. Appl Soft Comput.
interactions. Biophys Rev. 2013;5:147–158. 2018;62:251–258.
52. Shah NB, Duncan TM. Bio-layer interferometry for measuring kinetics of 76. Wang C, Zhang Y. Improving scoring-docking-screening powers of
protein-protein interactions and allosteric ligand effects. J Vis Exp. 2014; protein–ligand scoring functions using random forest. J Comput
(84):e51383. Chem. 2017;38:169–177.
EXPERT OPINION ON DRUG DISCOVERY 767

77. Ryde U, Söderhjelm P. Ligand-binding affinity estimates supported tors. Free energy calculations: theory and applications in chemistry
by quantum-mechanical methods. Chem Rev. 2016;116:5520–5566. and biology. Berlin, Heidelberg: Springer; 2007. p. 77–118.
● A comprehensive list of QM applications in binding affinity 100. Kästner J. Umbrella sampling. Wiley Interdiscip Rev Comput Mol
calculations. Sci. 2011;1:932–942.
78. Muddana HS, Fenley AT, Mobley DL, et al. The SAMPL4 host–guest 101. Bernardi RC, Melo MCR, Schulten K. Enhanced sampling techniques
blind prediction challenge: an overview. J Comput Aided Mol Des. in molecular dynamics simulations of biological systems. Biochim
2014;28:305–317. Biophys Acta Gen Subj. 2015;1850:872–877.
79. Ehrlich S, Göller AH, Grimme S. Towards full quantum-mechanics- 102. Kumar S, Rosenberg JM, Bouzida D, et al. The weighted histogram
based protein–ligand binding affinities. ChemPhysChem. analysis method for free-energy calculations on biomolecules. I. the
2017;18:898–905. method. J Comput Chem. 1992;13:1011–1021.
80. Grimme S, Schreiner PR. Computational chemistry: the fate of 103. Sun L, Bertelshofer F, Greiner G, et al. Characteristics of sucrose
current methods and future challenges. Angew Chem. transport through the sucrose-specific porin scry studied by mole-
2018;57:4170–4176. cular dynamics simulations. Front Bioeng Biotechnol. 2016;4.
81. Massova I, Kollman PA. Combined molecular mechanical and con- 104. El Hage K, Mondal P, Meuwly M. Free energy simulations for protein
tinuum solvent approach (MM-PBSA/GBSA) to predict ligand ligand binding and stability. Mol Simulat. 2018;44:1044–1061.
binding. Perspect Drug Discovery Des. 2000;18:113–135. 105. Park S, Khalili-Araghi F, Tajkhorshid E, et al. Free energy calculation
82. Rizzi A, Murkli S, McNeill JN, et al. Overview of the SAMPL6 host– from steered molecular dynamics simulations using Jarzynski’s
guest binding affinity prediction challenge. J Comput Aided Mol equality. J Chem Phys. 2003;119:3559–3566.
Des. 2018;32:937–963. 106. Okimoto N, Suenaga A, Taiji M. Evaluation of protein–ligand affinity
● Another competition and a source of comparison of various prediction using steered molecular dynamics simulations. J Biomol
methods, especially computationally intensive ones. Struct Dyn. 2017;35:3221–3231.
83. Genheden S, Ryde U. The MM/PBSA and MM/GBSA methods to 107. Laio A, Parrinello M. Escaping free-energy minima. Pnas.
estimate ligand-binding affinities. Expert Opin Drug Discov. 2002;99:12562–12566.
2015;10:449–461. 108. Barducci A, Bussi G, Parrinello M. Well-tempered metadynamics:
● Good review of MM-PB(GB)SA method and its applications. a smoothly converging and tunable free-energy method. Phys
84. Li C, Li L, Petukh M, et al. Progress in developing Poisson-Boltzmann Rev Lett. 2008;100:020603.
equation solvers. Mol Based Math Biol. 2013;1:42–62. 109. Leone V, Marinelli F, Carloni P, et al. Targeting biomolecular flex-
85. Still WC, Tempczyk A, Hawley RC, et al. Semianalytical treatment of ibility with metadynamics. Curr Opin Struct Biol. 2010;20:148–154.
solvation for molecular mechanics and dynamics. J Am Chem Soc. 110. Cavalli A, Spitaleri A, Saladino G, et al. Investigating drug–target
1990;112:6127–6129. association and dissociation mechanisms using metadynamics-
86. Guimarães CRW, Cardozo M. MM-GB/SA rescoring of docking poses in based algorithms. Acc Chem Res. 2015;48:277–285.
structure-based lead optimization. J Chem Inf Model. 2008;48:958–970. 111. Saleh N, Ibrahim P, Saladino G, et al. An efficient metadynamics-based
87. Thompson DC, Humblet C, Joseph-McCarthy D. Investigation of protocol to model the binding affinity and the transition state ensem-
MM-PBSA rescoring of docking poses. J Chem Inf Model. ble of G-protein-coupled receptor ligands. J Chem Inf Model.
2008;48:1081–1091. 2017;57:1210–1217.
88. Chen W, Gilson MK, Webb SP, et al. Modeling protein−Ligand binding 112. Gohlke H, Klebe G. Statistical potentials and scoring functions applied
by mining minima. J Chem Theory Comput. 2010;6:3540–3557. to protein–ligand binding. Curr Opin Struct Biol. 2001;11:231–235.
89. Schulz-Gasch T, Stahl M. Scoring functions for protein–ligand inter- 113. Kadukova M, Grudinin S. Convex-PL: a novel knowledge-based poten-
actions: a critical perspective. Drug Discov Today. 2004;1:231–239. tial for protein-ligand interactions deduced from structural databases
90. Liu J, Wang R. Classification of Current Scoring Functions. J Chem using convex optimization. J Comput Aided Mol Des. 2017;31:943–958.
Inf Model. 2015;55:475–482. 114. Zwanzig RW. High-temperature equation of state by a perturbation
91. Sousa SF, Ribeiro AJM, Coimbra JTS, et al. Protein-ligand docking in method. i. nonpolar gases. J Chem Phys. 1954;22:1420–1426.
the new millennium – A retrospective of 10 years in the field. Curr 115. Brandsdal BO, Österberg F, Almlöf M, et al. Free energy calculations
Med Chem. 2013;20:2296–2314. and ligand binding. In: V. Daggett, editor. Advances in protein
92. Pagadala NS, Syed K, Tuszynski J. Software for molecular docking: a chemistry, vol. 66. Amsterdam: Elsevier Academic Press; 2003.
review. Biophys Rev. 2017;9:91–102. 123–158.
93. Chen Y-C. Beware of docking!Trends Pharmacol Sci. 2015;36:78–95. 116. Lamb ML, Jorgensen WL. Computational approaches to molecular
94. Gohlke H, Klebe G. Approaches to the description and prediction of recognition. Curr Opin Chem Biol. 1997;1:449–457.
the binding affinity of small-molecule ligands to macromolecular 117. Williams-Noonan BJ, Yuriev E, Chalmers DK. Free energy methods
receptors. Angew Chem. 2002;41:2644–2676. in drug design: prospects of “alchemical perturbation” in medicinal
95. Li J, Fu A, Zhang L. An overview of scoring functions used for chemistry. J Med Chem. 2018;61:638–649.
protein–ligand interactions in molecular docking. Interdiscip Sci 118. Mikulskis P, Genheden S, Ryde U. A large-scale test of free-energy
Comput Life Sci. 2019. simulation estimates of protein–ligand binding affinities. J Chem
96. Xu W, Lucke AJ, Fairlie DP. Comparing sixteen scoring functions for Inf Model. 2014;54:2794–2806.
predicting biological activities of ligands for protein targets. J Mol 119. Åqvist J, Medina C, Samuelsson J-E. A new method for predicting
Graphics Modell. 2015;57:76–88. binding affinity in computer-aided drug design. Protein Eng Des
97. Su M, Yang Q, Du Y, et al. Comparative assessment of scoring func- Sel. 1994;7:385–391.
tions: the CASF-2016 update. J Chem Inf Model. 2019;59:895–913. 120. Gutiérrez-de-Terán H, Åqvist J. Linear interaction energy: method and
98. Wang R, Lai L, Wang S. Further development and validation of applications in drug design. In: Baron R, editor. Computational drug
empirical scoring functions for structure-based binding affinity discovery and design. New York: Springer New York; 2012. p. 305–323.
prediction. J Comput Aided Mol Des. 2002;16:11–26. 121. Montalvo-Acosta JJ, Pacak P, Gomes DEB, et al. A linear interaction
99. Shell MS, Panagiotopoulos A, Pohorille A. Methods based on prob- energy model for cavitand host–guest binding affinities. J Phys
ability distributions and histograms. In: Chipot C, Pohorille A, edi- Chem B. 2018;122:6810–6814.
768 V. KAIRYS ET AL.

122. Čapkauskaitė E, Zakšauskas A, Ruibys V, et al. Benzimidazole 126. Zhao H, Piszczek G, Schuck P. SEDPHAT – A platform for global ITC
design, synthesis, and docking to build selective carbonic anhy- analysis and global multi-method analysis of molecular interac-
drase VA inhibitors. Bioorg Med Chem. 2018;26:675–687. tions. Methods. 2015;76:137–148.
123. Nezami A, Kimura T, Hidaka K, et al. High-affinity inhibition of ●● Article describing method of global analysis using experi-
a family of plasmodium falciparum proteases by a designed adap- mental data from different affinity determination techniques.
tive inhibitor. Biochem J. 2003;42:8459–8464. 127. Brautigam CA, Zhao H, Vargas C, et al. Integration and global
analysis of isothermal titration calorimetry data for studying
124. Sarver RW, Peevers J, Cody WL, et al. Binding thermodynamics of
macromolecular interactions. Nat Protoc. 2016;11:882–894.
substituted diaminopyrimidine renin inhibitors. Anal Biochem.
128. Linkuvienė V, Zubrienė A, Manakova E, et al. Thermodynamic,
2007;360:30–40.
kinetic, and structural parameterization of human carbonic anhy-
125. Gaieb Z, Parks CD, Chiu M, et al. D3R grand challenge 3: blind drase interactions toward enhanced inhibitor design. Q Rev
prediction of protein–ligand poses and affinity rankings. J Comput Biophys. 2018;51:e10.
Aided Mol Des. 2019;33:1–18. ● An example of a comprehensive structure and intrinsic affinity
● Competition as a good source to discover new methods and correlation database for closely related compounds and protein
to compare them with the better established methods. targets.

You might also like