You are on page 1of 28

Article

Virtual Screening for Potential Inhibitors of Human Hexokinase


II for the Development of Anti-Dengue Therapeutics
Suriyea Tanbin 1 , Fazia Adyani Ahmad Fuad 1, * and Azzmer Azzar Abdul Hamid 2

1 Department of Biotechnology Engineering, Faculty of Engineering, International Islamic University Malaysia,


Kuala Lumpur 50728, Malaysia; tanbin.khu.bd@gmail.com
2 Research Unit for Bioinformatics & Computational Biology (RUBIC), Kulliyyah of Science,
International Islamic University Malaysia, Bandar Indera Mahkota, Kuantan 25200, Malaysia;
azzmer@iium.edu.my
* Correspondence: fazia_adyani@iium.edu.my

Abstract: Dengue fever, which is a disease caused by the dengue virus (DENV), is a major unsolved
issue in many tropical and sub-tropical regions of the world. The absence of treatment that effectively
prevent further viral propagation inside the human’s body resulted in a high number of deaths
globally each year. Thus, novel anti-dengue therapies are required for effective treatment. Human
hexokinase II (HKII), which is the first enzyme in the glycolytic pathway, is an important drug
target due to its significant impact on viral replication and survival in host cells. In this study,
23.1 million compounds were computationally-screened against HKII using the Ultrafast Shape
Recognition with a CREDO Atom Types (USRCAT) algorithm. In total, 300 compounds with the
highest similarity scores relative to three reference molecules, known as Alpha-D-glucose (GLC), Beta-
D-glucose-6-phosphate (BG6), and 2-deoxyglucose (2DG), were aligned. Of these 300 compounds,
165 were chosen for further structure-based screening, based on their similarity scores, ADME
analysis, the Lipinski’s Rule of Five, and virtual toxicity test results. The selected analogues were
 subsequently docked against each domain of the HKII structure (PDB ID: 2NZT) using AutoDock

Vina programme. The three top-ranked compounds for each query were then selected from the
Citation: Tanbin, S.; Ahmad Fuad,
docking results based on their binding energy, the number of hydrogen bonds formed, and the
F.A.; Abdul Hamid, A.A. Virtual
specific catalytic residues. The best docking results for each analogue were observed for the C-
Screening for Potential Inhibitors of
terminus of Chain B. The top-ranked analogues of GLC, compound 10, compound 26, and compound
Human Hexokinase II for the
58, showed predicted binding energies of −7.2, −7.0, and −6.10 kcal/mol and 7, 5, and 2 hydrogen
Development of Anti-Dengue
Therapeutics. BioTech 2021, 10, 1.
bonds, respectively. The analogues of BG6, compound 30, compound 36, and compound 38, showed
https://doi.org/10.3390/biotech predicted binding energies of −7.8, −7.4, and −7.0 kcal/mol and 11, 9, and 5 hydrogen bonds,
10010001 while the top three analogues of 2DG, known as compound 1, compound 4, and compound 31,
showed predicted binding energies of −6.8, −6.3, and −6.3 kcal/mol and 4, 3, and 1 hydrogen
Received: 5 November 2020 bonds, sequentially. The highest-ranked compounds in the docking analysis were then selected for
Accepted: 24 December 2020 molecular dynamics simulation, where compound 10, compound 30, and compound 1, which are the
Published: 28 December 2020 analogues of GLC, BG6, and 2DG, have shown strong protein-ligand stability with an RMSD value
of ±5.0 A◦ with a 5 H bond, ±4.0 A◦ with an 8 H bond, and ±0.5 A◦ with a 2 H bond, respectively,
Publisher’s Note: MDPI stays neu-
compared to the reference molecules throughout the 20 ns simulation time. Therefore, by using the
tral with regard to jurisdictional claims
computational studies, we proposed novel compounds, which may act as potential drugs against
in published maps and institutional
DENV by inhibiting HKII’s activity.
affiliations.

Keywords: human hexokinase II; glycolysis; ligand-based screening; structure-based screening;


molecular dynamics simulation
Copyright: © 2020 by the authors. Li-
censee MDPI, Basel, Switzerland. This
article is an open access article distributed
under the terms and conditions of the 1. Introduction
Creative Commons Attribution (CC BY)
Dengue is a major unsolved problem, complicated by factors including sequential
license (https://creativecommons.org/
infection with different dengue serotypes, increased populations, unplanned urbanization,
licenses/by/4.0/).

BioTech 2021, 10, 1. https://doi.org/10.3390/biotech10010001 https://www.mdpi.com/journal/biotech


BioTech 2021, 10, 1 2 of 28

and uncontrolled population increase of the leading vector, Aedes aegypti. The consequences
of these factors are a resurgence in dengue epidemics and the introduction of dengue
hemorrhagic fever and dengue shock syndrome to previously unaffected regions [1–3].
The clinical manifestation of dengue has a broad range of symptoms, from a minor
influenza-like pattern, which is associated with high fever, vomiting, loss of appetite,
severe headache, myalgia, and arthralgia, to a severe life-threatening illness characterized
by plasma leakage, thrombocytopenia, hemorrhage (dengue hemorrhagic fever), and the
potential to develop shock [4,5]. Global incidence of dengue has increased dramatically
in recent years, with 390 million infections occurring annually. Of these cases, 96 million
cases manifest clinically and 3.9 billion are at risk of dengue infection [6]. More than 70%
of cases occur in the South-East Asia and Western Pacific regions [6]. According to the
Pan American Health Organization (PAHO), approximately 3 million dengue cases are
currently being reported in America alone, including 68 fatal cases. This represents a 42%
increase compared to the previous year [7]. The incidence of dengue has also substantially
increased in Asia.
Dengue infection is caused by the dengue virus, which is a single-stranded RNA virus
belonging to the Flaviridiae family. It is transmitted to humans from female mosquitoes of
the Aedes genus, principally A. aegypti, through a single bite [6,8,9]. The positive-sense viral
RNA, which has a length of approximately 11 kb, encodes three structural proteins (capsid,
pre-membrane, and membrane) that help form the virion and seven non-structural proteins
(NS1, NS2A, NS2B, NS3, NS4A, NS4B, and NS5) that aid in viral replication [10]. Dengue
virus has four distinct antigenic serotypes: DENV-1, DENV-2, DENV-3, and DENV-4.
Sequential infection with multiple serotypes of the virus can result in a more severe form
of the disease [11,12].
Viruses are intercellular infectious agents that rely on the metabolic machinery of the
host cell to survive and replicate. Several viruses, such as human cytomegalovirus (HCMV),
herpes simplex virus 1 (HSV-1), hepatitis C virus, influenza A virus, human immunodefi-
ciency virus type 1 (HIV-1), Kaposi’s sarcoma-associated herpesvirus (KSHV), and vaccinia
virus (VACV) have been shown to cause extensive changes to the central carbon metabolic
pathways of host cells during viral attack [13–15]. In recent years, Fontaine and colleagues
have demonstrated how dengue virus can alter host metabolism by conducting intracel-
lular metabolic profiling of mock-infected and dengue-infected primary human foreskin
fibroblasts (HFFs) at different time points [15]. The experimental data suggested that
central carbon metabolism, specifically the glycolytic pathway, is significantly disrupted
during dengue virus infection. In dengue virus infected cells, glucose consumption was
increased, as was expression of glucose transporter 1 (GLUT1) and human hexokinase
II (HKII), which are important regulating enzymes in the glycolytic pathway [15]. In the
presence of oxygen, normal cells metabolize glucose via glycolysis to produce ATP through
oxidative phosphorylation within the mitochondria. In tumor and cancer-infected cells,
glucose is metabolized in a different way from normal cells with production of ATP from
aerobic glycolysis, in a phenomenon called the “Warburg effect.” In addition, normal cells
require energy (ATP or GTP), which are also building block components to ensure specific
cell functions, while tumor and cancer cells must replicate all their cellular components,
thus, increasing their need for a biosynthetic building block (nucleotides, amino acids,
and lipids). The main attribute of this metabolic changes is an increase in the glucose
uptake by the cell. Extensive studies have postulated that virus-infected cells have the
same Warburg-like phenomena where the key enzyme of glycolytic pathway, HKII con-
tributes to enhanced glucose consumption in infected cells [16–19]. HKII is a rate-limiting
enzyme of the glycolytic pathway that assist in increasing glucose consumption in the
virus-infected cell. Hence, HKII has been chosen as a potential drug target. Fontaine and
colleagues have also reported that glucose consumption tremendously increased during
dengue viral infection, where glucose is metabolized via the host glycolytic pathway and
provides energy (ATP) as well as metabolites to a dengue-infected cell. Consequently, virus
takes this metabolic change as an advantage and assists themselves to survive and replicate
BioTech 2021, 10, 1 3 of 28

inside the host cell. It has also been proven that HKII enhanced glucose consumption in
the infected cell, as it is the key enzyme of the glycolytic pathway, which served as a basis
for the enzyme to be a target in antiviral therapy [20]. Several studies on other viruses
such as the poliovirus, HSV-1 [21], HCMV [22], HSV [23], and HCV [24] have also been
documented, highlighting that glycolysis is crucial for the viruses to survive and replicate
inside a host cell, where HKII governs the pathway.
Several experiments have shown that viral replication, as well as tumor cell prolif-
eration, were positively correlated with glycolysis. This suggests that treatment with
2-deoxyglucose (2DG), which is a competitive antagonist of the glycolysis process, could
inhibit viral replication [19,20]. 2DG is an analogue of glucose, in which two hydroxyl
groups have been replaced by hydrogen atoms. This inhibits the production of glucose-6-
phosphate, ultimately inhibiting the glycolytic pathway [25]. 2DG has been suggested by
several studies for treating cancer, alongside inhibiting dengue virus replication [26–28].
However, 2DG has shown toxic effects when used to treat patients [19,29]. Hence, it is vital
to find new compounds that are similar to 2DG, but do not induce toxicity, as these could
be valuable candidates for anti-dengue drug development. Nevertheless, 2DG has been
used as a competitive inhibitor of glucose for HKII in various viral therapeutic research,
but, as previously shown, 2-DG has exhibited toxic effects on host cells [19]. Hence, the
situation further urges finding potential inhibitors against HKII for the development of
anti-dengue therapeutics. In addition to 2DG, both Alpha-D-glucose (GLC) and Beta-D-
glucose-6-phosphate (BG6), which are the substrate and product of HKII, respectively, were
used as reference molecules for rational drug design.
In this study, we report the identification of small molecules, which have the
potentials to be developed into safe and effective anti-dengue drugs. We first conducted a
ligand-based drug design experiment by screening compounds similar to those described
above. Subsequently, a series of docking and scoring experiments were conducted,
which were followed by structure-based screening using the crystal structure of human
HKII (PDB ID: 2NZT). These compounds were then subjected to virtual toxicity testing.
Finally, the best-performing compounds were selected for Molecular Dynamics (MD)
simulation analysis.

2. Materials and Methods


2.1. Ligand-Based Screening
2.1.1. Identification of Lead Molecules and Analogues
In this study, ligand-based screening has been performed to filter drug-like compounds
based on its query molecules with shape similarity, physiological properties, and toxicity
from enormous compound libraries. Ligand-based screening provides a set of structurally-
diverse ligands that bind to a receptor, where the approach of this screening is based on
searching for molecules with a shape similarity to that of known actives, as such molecules
will fit the target’s binding site [30]. Three different lead molecules Alpha-D-glucose (GLC),
Beta-D-glucose-6-phosphate (BG6), and 2-Deoxyglucose (2DG) have been used as query
molecules for initial pharmacophore screening. The common structural features of GLC,
BG6, and 2DG have a glucose chain with six carbon atoms. Moreover, the functional feature
of these query molecules (GLC, BG6, and 2DG) is possessing inhibitory effect to human
HKII protein. It has been proven that, at a high concentration (up to 5 µM), GLC shows an
inhibitory effect on HKII, while BG6 itself is an inhibitory product of HKII, while 2DG is a
well-known inhibitor of HKII [17,29,31].
The structure data file (SDF) of GLC, BG6, and 2DG (with PubChem ID CHEBI:
10822 ID CHEBI: 17925 and ID CHEBI: 17719, as well as molecular formula C6H12O5
C6H12O6 and C6H13O9P) were extracted from PubChem (https://pubchem.ncbi.nlm.
nih.gov/) to screen for the analogues by using Ultrafast Shape Recognition with CREDO
Atom Types (USRCAT) available at http://usr.marseille.inserm.fr/. A total of 300 com-
pounds, where, for each query of molecules, 100 similar hits have been countenanced
from the USRCAT program. All predicted molecules ranked based on score zero (0) to
BioTech 2021, 10, 1 4 of 28

one (1) score, where a compound with a score near to one indicated greater similarity
toward its respective query molecule. Furthermore, a total 165 hits out of 300 has been
screened based on Absorption, Distribution, Metabolism and Excretion (ADME) analysis,
a toxicity test, and a Lipinski’s Rule of Five. Later, selected compounds were screened
through a structure-based approach and, finally, were analysed through molecular
dynamics simulation studies.

2.1.2. ADME Analysis


Physiochemical properties such as absorption, distribution, metabolism, and ex-
cretion for all compounds were predicted from the SwissADME website (www.http:
//www.swissadme.ch/index.php), prior to a docking study. Structure data file (SDF)
for each compound was used to obtain ADME values. All selected compounds fulfilled the
benchmarks of ADME analysis.

2.1.3. Toxicity Test


In a drug design process, the prediction of compound toxicity is crucial to identify
non-toxic compounds. In this experiment, we used ProTox-II (www.http://tox.charite.
de/protox_II/) to determine the non-toxicity of selected compounds. The SDF format of
each compound was converted into a molecular data file (MDL Molfile) using Open Bable
software. Various toxicity end points such as acute toxicity, hepatotoxicity, cytotoxicity,
mutagenicity, and immunotoxicity have been tested to predict the level of toxicity of each
compound. In ProTox-II, toxic levels were classified in six different forms, Class-I to Class-
VI, based on a range of lethal dose (LD50) values, where Class-I determines the toxic
compound, while Class-VI determines a non-toxic compound.

2.2. Structure-Based Screening


2.2.1. Retrieval and Structural Preparation of the Protein
HKII is a homodimer protein with highly similar N and C terminal domains that to-
gether form half of the protein. The crystal structure of HKII (PDB ID: 2NZT) was retrieved
from the Protein Data Bank website (https://www.rcsb.org/). The native structure of the
protein was complexed with a heteroatom, a water molecule, and an unknown unique
ligand (UNX), which are crucial to be removed to ensure good protein-ligand interaction.
The water molecule might deform the binding site residues as well as the interactions be-
tween protein-ligand atoms. On the other hand, the heteroatom and unknown ligand have
to be removed due to docking of the selected ligand and to facilitate a smooth molecular
docking process. Structural visualization was conducted using PyMOL software.

2.2.2. Molecular Docking


Molecular docking is usually referred to as protein-ligand binding. The aim of
docking is to understand molecular recognition, a favourable binding pose, and binding
affinity of ligand toward the 3D structure of the receptor protein. The protein structure
was prepared by protein optimization and energy minimization by adding hydrogen
atoms, as well as removing an existing ligand from the active site. The known ligands
(GLC and BG6) were removed for a re-docking purpose, where the active site of the
protein structure is typically being validated by re-docking experiments [32–34]. The
ligands (GLC and BG6) were extracted from native protein-ligand complexes to eval-
uate the conformation and, subsequently, docked back into the protein to validate the
docking location. In this case, we scrutinized the native structure of HKII using PyMOL
and observed that there is a cleft inside both N-terminal and C-terminal domain in
the protein, where two known ligands (BG6 and GLC) were located in parallel. The
selected compounds and their query molecules were docked inside the cleft sequentially.
It should be noted that only the N-terminal domain was used to conduct the docking
process because the N-terminal domain regulates the stability of the whole enzyme,
in contrast with the C-terminal domain [35,36]. Recently, Nawaz and colleagues have
BioTech 2021, 10, 1 5 of 28

reported that the human hexokinase II is overexpressed in tumours and predominantly


found on the outer mitochondrial membrane where its N-terminus initiates and main-
tains the tumorigenesis. They further crystalized and generated the protein’s structure
using MODELLER [36,37]. However, the catalytic residue of human hexokinase II has
not yet been publicly available. Hence, it was a challenge to find the active site residues
of HKII. In this experiment, we scrutinized the catalytic residues around the cleft of
HKII, where the known ligands (GLC and BG6) were bound using MGLTools (version
1.5.7), which were further validated by re-docking processes. In this research, we used
the AutoDock Vina program that significantly improved the average accuracy of the
binding mode predictions. In the Vina script, all information must be included such
as grid box size (Box size: 26 ×24 × 22 A◦ ), grid box centre (X = 43.567, Y = 35.439,
Z = 36.27), and measurement of space (1.000), where the defined grid box encompasses
the protein binding site to do sequential docking of all selected ligands. The ligands
were prepared by adding a non-polar hydrogen atom using Avogadro. Since AutoDock
Vina can only accept a Protein Data bank, Partial Charge (Q), and atom Type (T) format
(PDBQT), the PDBQT format of the HKII protein, and all grid box information were
occupied using MGLTools, while the SDF files of all ligands were converted to a PDBQT
file using OpenBable software.

2.3. Molecular Dynamics Simulation


Molecular dynamics simulation has been widely used to scrutinize the behaviour
of molecules at an atomic level, in order to understand the structure and interactions
between a protein and a ligand molecule in a period of time. We set up molecular
dynamics simulation of selected compounds against HKII by using GROMACS version 5,
where the .pdb format of protein and selected compounds from molecular docking were
converted to a GROMACS file format to be implemented in the MD simulation. Atomic
interactions were calculated with a gromos96 54a7 force field and all systems were
solvating in a cubic box with a simple point charge (SPC) pre-equilibrate water system.
Ligand topology files were extracted from the Automated Topology Builder (ATB)
(https://atb.uq.edu.au) server since GROMACS does not generate a ligand topology file.
We used standard cut-off 1.0 nm for short-range coulomb interaction, where statistical
and energy data are stored for every 10 steps. The v-rescale and Berendsen weak
coupling algorithm were used to control temperature and pressure, respectively, during
the simulation process. The water equilibration around the protein-ligand complex is
crucial to avoid unnecessary exaggeration of the complex when the MD simulation has
started. In order to avoid this problem, we performed an equilibration run for 100 ps,
where all heavy atoms were restrained at their start position. Finally, 20 ns constrained
MD simulation was performed for each selected protein-ligand complex. All bond
lengths, including the hydrogen atoms were constrained by the LINCS algorithm. After
simulation has completed, we viewed the trajectory by using VMD and analysis such as
RMSD, distance between protein and ligand throughout the simulation, and formation
of the hydrogen bond, which were carried out using Xmgrace software. The virtual
screening workflow is presented in Figure 1.
BioTechBioTech
2021, 2, FOR
2021, 10,PEER
1 REVIEW 6 of 28 6

Figure 1. The virtual screening work flow. The process begins with ligand-based screening, selection
Figure 1. The virtual screening work flow. The process begins with ligand-based screening, selec-
of drug-like molecules by Lipinski’s Rule of Five, ADME analysis, toxicity analysis, docking, and
tionMolecular
of drug-like molecules by Lipinski’s Rule of Five, ADME analysis, toxicity analysis, docking,
Dynamics simulation.
and Molecular Dynamics simulation.
3. Results and Discussion
3.1. Ligand-Based
3. Results Screening
and Discussion
3.1.1. Identification of Analogues
3.1. Ligand-Based Screening
In pharmaceutical research, in silico screening has become prominent day-by-day.
3.1.1.
It isIdentification of Analogues
a computer-based method divided into ligand-based screening and structure-based
In pharmaceutical
screening. The formerresearch, in stage
is the initial silico of
screening has become
virtual screening, prominent
which is the most day-by-day.
exoteric It
is aapproach for drug discovery
computer-based method and lead optimisation.
divided The spotlight
into ligand-based of this
screening andtechnique is to
structure-based
compareThe
screening. molecular
formersimilarity with known
is the initial stage of and unknown
virtual fractions
screening, [32,33].
which is Ligand-based
the most exoteric
screening exploits key structural and physiochemical properties
approach for drug discovery and lead optimisation. The spotlight of this of ligands and target to is to
technique
enable the screening of active compounds, while structure-based screening most widely
compare molecular similarity with known and unknown fractions [32,33]. Ligand-based
used for molecular docking by exploiting the three-dimensional (3D) structure of the
screening exploits
target protein andkey structural
predicting and physiochemical
the preferred protein-ligand properties
binding poseofthrough
ligands theand target to
scoring
enable the screening of active compounds, while structure-based screening
function [30,33,38]. In this work, USRCAT (Ultrafast Shape Recognition with CREDO Atom most widely
used for program
Type) molecular hasdocking
been usedby toexploiting
screen similarthecompounds
three-dimensional (3D) structure
of Alpha-D-glucose (GLC),of the tar-
Beta-
get D-glucose-6-phosphate
protein and predicting the preferred
(BG6), protein-ligand
and 2-deoxyglucose (2DG)binding pose through
against HKII, where GLC theand
scoring
function [30,33,38]. In this work, USRCAT (Ultrafast Shape Recognition with CREDO
Atom Type) program has been used to screen similar compounds of Alpha-D-glucose
(GLC), Beta-D-glucose-6-phosphate (BG6), and 2-deoxyglucose (2DG) against HKII,
where GLC and BG6 are the substrate and product of HKII, respectively, while 2DG is a
BioTech 2021, 10, 1 7 of 28

BG6 are the substrate and product of HKII, respectively, while 2DG is a known inhibitor
of HKII. USRCAT is a publicly-accessible web tool, which is able to screen 50 million
conformers per second. USRCAT is an extended version of USR (Ultra shape recognition)
algorithm, which is improved with the CREDO database, where users are able to find an
enormous chemical component ranging from a natural product to drug-like molecules
as well as solvent molecules and ions. USRCAT is an advanced version of UFSRAT in
terms of having high capability to differentiate similar molecules at an atomic level and
intended to discriminate between long chain-like molecules, such as heteropeptides and
long alkayl chains [39]. The USRCAT algorithm is based not only on shape similarity but
also considers geometrical distribution of atoms such as all atoms, a hydrogen bond donor,
and acceptor atoms, as well as aromatic atoms [39].
In this study, 23.1 million molecules were screened and a total of 300 most similar
compounds were obtained from USRCAT for all of the reference molecules (2DG, GLC, and
BG6). Thus, specifically, each query has 100 similar compounds aligned into the hits.csv
file along with similarity scores, physicochemical properties, and the vendor’s information.
Similarity was evaluated based on the scoring function ranging from 0–1, where zero
and one indicates minimum and maximum similarity, respectively. As mentioned above,
the USRCAT score zero indicates the least similar compounds, while those closer to one
reflects a much closer resemblance toward the reference molecule. Based on these findings,
a total of 165 compounds out of 300 similar hits for each query molecule were chosen
and their physiological properties were obtained from the ZINC database. The ZINC
database contains 727,842 purchasable compounds from various suppliers (ChemBridge,
ChemDiv, Ryan, Asinex, Sigma-Aldrich, Maybridge, Specs, Comgenex, and Otava) in
which 494,915 are Lipinski subjacents. Based on the similarity scores, the three top-ranked
compounds that resembled each of the query molecule are shown in Tables 1–3, as their
ranking scores ranged from 0.946 to 0.913, 0.817 to 0.812, and 0.806 to 0.751 for GLC, BG6,
and 2DG, respectively. All three compounds, which are analogues of 2DG, GLC, and BG6,
which have been selected for the subsequent studies, obeyed the Lipinski’s Rule of Five
in terms of their physiological parameters, hydrogen bond donors <5, hydrogen bond
acceptor <10, molecular weight under <500 g/mol, and a partition coefficient logP < 5
with no violation [40]. The mentioned parameters related to the Lipinski’s Rule of Five are
presented in Tables 1–3. Compounds 10 to 58, which are similar conformers of GLC possess
MlogP values ranging from −2.15 to −1.93, and drug-likeness from 0.14 to −0.01 (Table 1).
Moreover, the top three similar hits of BG6, compounds 30 to 38 have MlogP values
from −4.02 to −3.84 and drug-likeness from 0.89 to −1.08 (Table 2). Drug-likeness is the
preliminary concept of drug design and its value is usually estimated from the compound’s
molecular structure standardized between −0.4 and 5.6, whereby it indicates whether the
compound has drug properties or not. Since our findings were optimized with respect
to the drug-likeness parameter and the adherence to the Lipinski’s Rule of Five, most of
the compounds fulfilled all the criteria. The drug-like properties of 2DG corresponded to
the MlogP values between 0.06 and −0.08, and drug-likeness between −0.64 and −1.16
(Table 3). It is noteworthy to note that, although the same scores were obtained from the
USRCAT program, these compounds are structurally different and contain different atom
types. The Rule of Five provides insights for the analysis of the molecular properties and
structural features of every compound, which is important for drug pharmacokinetics
in a human’s body, including their absorption, distribution, metabolism, and excretion
(ADME) [41].
BioTech 2021, 2, FOR PEER REVIEW 8
BioTech 2021, 2, FOR PEER REVIEW 8
BioTech
BioTech
BioTech
BioTech 2021, 2021,
2021,
2021,
10, 2,
12,2,
FOR
2, FOR
FOR PEER
PEER
PEER REVIEW
REVIEW
REVIEW 8 of 28 8 88
BioTech
BioTech2021,
2021,
BioTech 2021, FOR
2,2,FOR PEER
FORPEER REVIEW
PEERREVIEW
REVIEW 88 8
Table 1. Similarity scores and Lipinski’s values for GLC and its analogues, as obtained from USRCAT.
Table 1. Similarity scores and Lipinski’s values for GLC and its analogues, as obtained from USRCAT.
Table1.1.
Table
Table 1.Similarity
Similarityscores
Similarity scoresand
scores and
and Lipinski’svalues
Lipinski’s
Lipinski’s valuesfor
values forGLC
for GLCand
GLC andits
and itsanalogues,
its analogues,asas
analogues, asobtained
obtainedfrom
obtained fromUSRCAT.
from USRCAT.
USRCAT.
Table
Table
Table 1.1.1.Similarity
Similarity
Similarity
1. Similarity
Compound
Table scores
scoresscores and
andand
scores andLipinski’s
Lipinski’s
Lipinski’s values
values
Lipinski’s values for
for for
GLC
Similarity
values GLC
forGLC
GLC and
and
andand
its its analogues,
itsanalogues,
analogues,
analogues,
Number
its as
of as obtainedobtained
asobtained
obtained
Molecular
as from from
from USRCAT.
USRCAT.
USRCAT.
from USRCAT.
ZINC ID 2D Structure MlogP Drug-Likeness
Number
Compound Score
Similarity HBD/HBA
Number of Weight (g/mol)
Molecular
Compound
Compound ZINC ID 2D Structure Similarity
Similarity Number
Number of
ofof Molecular
Molecular
Molecular MlogP Drug-Likeness
Compound
Compound Number
Compound
Compound ZINC ID 2D Structure Similarity
Score
Similarity
Similarity
Similarity Number
HBD/HBA
Number of
Number
Number ofof Molecular
Weight (g/mol) MlogP
Molecular
Molecular Drug-
MlogP Drug-Likeness
Drug-Likeness
Compound ZINC
ID ID
ZINC ID 2D2D Structure
Structure Similarity Number of Molecular MlogP Drug-Likeness
Number ZINCZINC
ZINC
ZINC ID 2D 2D
ID Structure
2D Structure
2DStructure
Structure Score HBD/HBA Weight
Weight (g/mol) MlogP
MlogP
MlogP Drug-Likeness
Drug-Likeness
Number Number
Number
Number
Number
Number
ID Score
ScoreScore
Score
Score
Score
HBD/HBA
HBD/HBA
HBD/HBA
HBD/HBA
HBD/HBA
HBD/HBA
Weight
Weight
Weight
Weight
Weight
(g/mol)
(g/mol)
(g/mol)
(g/mol)
(g/mol)
MlogP Drug-Likeness
Likeness
(g/mol)
GLC 5/6 180.156 −3.21 0.01
GLC 5/6 180.156 −3.21 0.01
GLC GLC
GLC
GLC
GLC
GLC 5/6
5/65/6
5/6
5/6
5/6
180.156
180.156
180.156
180.156
180.156
180.156 − −3.21
3.21
−3.21
−3.21
−3.21
−3.21
0.01
0.01
0.01
0.01
0.01
0.01
GLC 5/6 180.156 −3.21 0.01

10 3,956,760 0.946 4/5 182.147 −2.15 0.14


10 3,956,760 0.946 4/5 182.147 −2.15 0.14
10 10
1010 3,956,760
3,956,760
3,956,760
3,956,760 0.946
0.946
0.946
0.946 4/5
4/5 4/5
4/5 182.147
182.147
182.147
182.147 − −2.15
−2.15
−2.15
2.15 0.14
0.14
0.14
0.14
10
10 10 3,956,760
3,956,760
3,956,760 0.946
0.946
0.946 4/5
4/5
4/5 182.147
182.147
182.147 −2.15
−2.15
−2.15 0.14
0.14
0.14

26 16,159,409 0.933 4/5 182.147 −2.15 0.09


26 16,159,409 0.933 4/5 182.147 −2.15 0.09
26
2626 16,159,409
16,159,409
16,159,409 0.933
0.933
0.933 4/54/5
4/5 182.147
182.147
182.147 −2.15
−2.15
−2.15 0.09
0.09
0.09
26 26
26 26 16,159,409
16,159,409
16,159,409
16,159,409 0.933
0.9330.933
0.933 4/54/5
4/5
4/5 182.147
182.147
182.147
182.147 −−2.15
−2.15
−2.15
2.15 0.09
0.09
0.09
0.09

58 3,809,846 0.913 4/5 182.147 −1.93 −0.01


58 3,809,846 0.913 4/5 182.147 −1.93 −0.01
58
5858 3,809,846
3,809,846
3,809,846 0.913
0.913
0.913 4/54/5
4/5 182.147
182.147
182.147 −1.93
−1.93
−1.93 −0.01
−0.01
−0.01
58 58
58 58 3,809,846
3,809,846
3,809,846
3,809,846 0.913
0.9130.913
0.913 4/54/5
4/5
4/5 182.147
182.147
182.147
182.147 −1.93
−1.93
−−1.93
1.93 −0.01
−0.01
−0.01
−0.01

Table 2. Similarity scores and Lipinski’s values for BG6 and its analogues, as obtained from USRCAT.
Table 2. Similarity scores and Lipinski’s values for BG6 and its analogues, as obtained from USRCAT.
Table2.2.
Table
Table 2.Similarity
Similarityscores
Similarity scoresand
scores and
and Lipinski’svalues
Lipinski’s
Lipinski’s valuesfor
values forBG6
for BG6and
BG6 andits
and itsanalogues,
its analogues,asas
analogues, asobtained
obtainedfrom
obtained fromUSRCAT.
from USRCAT.
USRCAT.
CompoundTable
Table2.2.2.
Table Similarity
Similarity scores
Similarityscores and
scoresand
andLipinski’s
Lipinski’s
Lipinski’s values for
valuesfor
Similarity
values BG6
forBG6
BG6 and its
andits
Number
and analogues,
itsanalogues,
analogues,
of as obtained
asobtained
Molecular
as from
obtainedfrom USRCAT.
fromUSRCAT.
USRCAT.
Table ZINC
2. ID
Similarity 2D
scores Structure
and Lipinski’s values for BG6 and its analogues, as obtained from MlogP
USRCAT. Drug-Likeness
Compound
Number Similarity
Score Number
HBD/HBA of Weight
Molecular(g/mol)
Compound
Compound
Compound ZINC ID 2D Structure Similarity
Similarity
Similarity Number
Number
Numberofof
of Molecular
Molecular
Molecular MlogP Drug-Likeness
Compound
Compound
Number ZINC
Compound ZINC
ZINC IDID
ID 2D2D
2D Structure
Structure
Structure Similarity
Similarity
Score
Similarity Number
Number
HBD/HBA
Number of
of Molecular
of Weight
Molecular
(g/mol) MlogP
Molecular
Molecular MlogP
MlogP Drug-Likeness
Drug-Likeness
Drug-Likeness
Number
Number
Number
Compound ZINC
ZINC
ZINC IDID
ID 2D
2DStructure
Structure
2D Structure Score
Score
Score
Similarity HBD/HBA
HBD/HBA Weight
Number of Weight
HBD/HBA Weight (g/mol)
(g/mol)
(g/mol) MlogP
MlogP Drug-Likeness
Drug-Likeness
MlogP Drug-Likeness
Drug-
Number
Number
Number ZINC ID 2D Structure Score
Score
Score HBD/HBA
HBD/HBA
HBD/HBA Weight
Weight (g/mol)
(g/mol)
Weight
Weight (g/mol) MlogP
Number Score HBD/HBA Likeness
(g/mol)
BG6 6/9 260.135 −3.64 −0.20
BG6 6/9 260.135 −3.64 −0.20
BG6
BG6
BG6
BG6
BG6 6/9
6/9
6/9
6/9
6/9 260.135
260.135
260.135
260.135
260.135 −3.64
−3.64
−3.64
−3.64
−3.64 −0.20
−0.20
−0.20
−0.20
−0.20
BG6BG6 6/9
6/9 260.135
260.135 − 3.64
−3.64 −−0.20
0.20

30 4,403,145 0.817 6/8 238.192 −3.84 0.89


30 4,403,145 0.817 6/8 238.192 −3.84 0.89
30 3030
30 4,403,145
4,403,145
4,403,145
4,403,145 0.817
0.817
0.817
0.817 6/8
6/8
6/8 238.192
238.192
238.192
238.192 −−3.84
−3.84
3.84
−3.84 0.89
0.89
0.89
0.89
30
3030 4,403,145
4,403,145
4,403,145 0.817
0.817
0.817 6/8
6/8
6/8 238.192
238.192
238.192 −3.84
−3.84
−3.84 0.89
0.89
0.89

36 4,530,268 0.812 6/9 252.199 −4.02 0.72


36 36 4,530,268
4,530,268 0.812
0.812 6/9
6/9 252.199
252.199 −4.02
−4.02 0.72
0.72
3636
36
36
36 4,530,268
4,530,268
4,530,268
4,530,268
4,530,268 0.812
0.812
0.812
0.812
0.812 6/9
6/9
6/9
6/9
6/9 252.199
252.199
252.199
252.199
252.199 −4.02
−4.02
−4.02
−4.02
−4.02 0.72
0.72
0.72
0.72
0.72
36 4,530,268 0.812 6/9 252.199 −4.02 0.72

38 38 1,576,959
1,576,959 0.812
0.812 6/8
6/8 238.192
238.192 −3.84
−3.84 −1.08
−1.08
38 1,576,959 0.812 6/8 238.192 −3.84 −1.08
3838
38
38
38 1,576,959
1,576,959
1,576,959
1,576,959
1,576,959 0.812
0.812
0.812
0.812
0.812 6/8
6/8
6/8
6/8
6/8 238.192
238.192
238.192
238.192
238.192 −3.84
−3.84
−3.84
−3.84
−3.84 −1.08
−1.08
−1.08
−1.08
−1.08
38 1,576,959 0.812 6/8 238.192 −3.84 −1.08
BioTech2021,
BioTech 2021,2,10,
FOR1 PEER REVIEW 9 of 928
BioTech 2021, 2, FOR PEER REVIEW 9
BioTech 2021, 2, FOR PEER REVIEW 9
BioTech 2021, 2, FOR PEER REVIEW 9

Table 3.3.Similarity
Table3. scores
Similarityscores and
scoresand Lipinski’s
andLipinski’s values
Lipinski’svalues for
valuesfor 2DG
for2DG and
2DGand itsitsanalogues,
andits asasobtained
analogues,as from
obtainedfrom USRCAT.
fromUSRCAT.
USRCAT.
Table Similarity analogues, obtained
Table 3. Similarity scores and Lipinski’s values for 2DG and its analogues, as obtained from USRCAT.
Table 3. Similarity scores and Lipinski’s values for 2DG and its analogues, as obtained from USRCAT.
Molecular
Molecular
Compound
Compound
Compound Similarity Number
Similarity Number
Similarity Number of ofMolecular
of Molecular Drug- Drug-
Compound ZINC
ZINC IDID 2D
2D Structure
Structure Similarity Number of Weight Weight MlogP
Molecular MlogPDrug-
Drug-Likeness
Number
Number
Compound ZINC
ZINC ID ID
ID 2D Structure
2D Structure
Structure Score
Score
Similarity HBD/HBA
HBD/HBA
Number of Weight
Weight MlogP Likeness
Drug-
MlogP Likeness
Number ZINC 2D Score HBD/HBA (g/mol)
Weight (g/mol) MlogP
Number
Number Score
Score HBD/HBA
HBD/HBA (g/mol) Likeness
Likeness
(g/mol)
(g/mol)
2DG 2DG 4/54/5 164.157
164.157 −1.95 −1.95 −1.33 −1.33
2DG
2DG 4/5
4/5 164.157
164.157 −1.95
−1.95 −1.33
−1.33
2DG 4/5 164.157 −1.95 −1.33

1 111 86,652,948
86,652,948
86,652,948
86,652,948
0.806
0.806
0.806
0.806
2/42/4
2/4
2/4
215.317
215.317
215.317
215.317
0.06 0.06 −0.64
0.06
0.06
−0.64 −0.64
−0.64
1 86,652,948 0.806 2/4 215.317 0.06 −0.64

444 86,991,606
86,991,606
86,991,606
0.796
0.796
0.796
2/4
2/4
2/42/4
213.301
213.301
213.301
−0.08
−0.08
−0.08 −0.08
−1.16
−1.16
−1.16 −1.16
4 86,991,606 0.796 213.301
4 86,991,606 0.796 2/4 213.301 −0.08 −1.16

31
31 86,991,603
86,991,603 0.751
0.751 2/4
2/4 213.301
213.301 −0.08
−0.08 −1.16
−1.16
31
31
31 86,991,603
86,991,603
86,991,603
0.751
0.751
0.751
2/42/4
2/4 213.301
213.301
213.301
−0.08 −0.08
−0.08 −1.16 −1.16
−1.16

Computational methods
Computational methods are are effectively
effectively used used in in pharmaceutical
pharmaceutical research research to to improve
improve
the Computational
drug discovery
Computational methodsLigand-based
process.
methods are effectively
are effectively used in
screening
used in pharmaceutical
pharmaceutical
is one of the research
crucial
researchparts toofimprove
to improve
in silico
silico
the
the drug
drug discovery
Computational
discovery process.
methods
process. Ligand-based
are effectively
Ligand-based screening
used inis
screening is one
one of
pharmaceutical
of the
the crucial
crucial parts
research
parts of
to
of in
improve
in silico
silico
drug
the
drug drugdesign
design and
discovery
and several
process.
several novel
novel compounds
Ligand-based
compounds have
screening
have been
is
been oneinvented
of
invented the through
crucial
through parts this
this of process.
in
the drug
drug design discovery
and process.
several novelLigand-based
compounds screening
have been is one of the crucial
invented through parts
this ofprocess.
in silico
process.
Hence,
drug
Hence, considering
design
consideringand several the importance
the importance
novel compounds of aa computational
of computational
have been invented drug design
drug design
through method,
method, it was
this process. was
drug design
Hence, consideringand several the novel
importancecompounds of a have been invented
computational drug through
design this process.it
method, it Hence,
was
adopted in
Hence, considering
adopted in the
the present
present thestudy to
importance
study to discover of thethe potential
a computational HKII inhibitors.
drug design Numerous
method,success- success-
it was
considering
adopted in the the importance
present study toofdiscover
a computational
discover the potential
potential drug HKII
HKII inhibitors.
design method,
inhibitors. Numerous
it was adopted
Numerous success- in
ful ligand-based
ful ligand-based
adopted in thestudy screening
present
screening study investigations
to the
discover
investigations thehave
have been performed,
potential
been performed,
HKII inhibitors. aiming
aiming to identify
Numerous
to identifysuccess-poten-
poten-
the present
ful ligand-based
ligand-based to
screening discover potential
investigations have HKII inhibitors.
been performed,
performed, Numerous
aiming successful
to identify
identify ligand-
poten-
tial
ful inhibitors against against
screening dengue drug targets.
investigationstargets. In extant
extant studies, dengue dengue virus encoded non-
tial
based
tial inhibitors
screening
inhibitors against dengue
investigations
dengue drug beenhave
havetargets.
drug In been
performed,
In extant studies,
aiming to
studies,
aiming
identify
dengue
to
virus encoded
potential
virus encoded
poten-
non-
inhibitors
non-
structural
tial inhibitors
structural protein
protein against3(NS3)
3(NS3) denguewas
was used
drug
used as
targets.
as a drug
aa drug In target,
extant
target, and
studies,
and two
two known
dengue
known inhibitors,
virus encoded
inhibitors, namely
non-
namely
against dengue
structural protein drug
3(NS3) targets.
was In extant
used as drugstudies,target, dengue
and two virus
known encoded non-structural
inhibitors, namely
suramin and
structural and ivermectin,
protein 3(NS3) have
was been
used successfully
astarget,
a drugand identified
target, through virtual screening and
suramin
protein and
suramin 3(NS3) ivermectin,
was
ivermectin, used have
haveas abeen
drug
been successfully
successfully twoand
identified
identified known two
throughknown
inhibitors,
through
inhibitors,
virtual
virtual screening
namely
screening
namely
suraminand
and
in
in vitro
suramin
vitro analysis,
analysis, where
and ivermectin,
where researchers
have
researchers used
been used
successfullya LOPAC
aaidentified
LOPAC compound
identified
compound through library
library for
virtual
for inhibitor
screening
inhibitor selec-
and
selec-
inand
vitro ivermectin,
analysis, have been
where successfully
researchers used LOPAC through
compound virtual
library screening
for and in
inhibitor vitro
selec-
tion
in vitro
tion as well
as well
analysis,as vero-B
as vero-B
where and
and vero-E6 cell
researchers
vero-E6 cell
used for aan
for anLOPAC
in vitro
in vitrocompound
assay [42,43].
assay [42,43]. It is
library
It is noteworthy
noteworthy
for inhibitor toselec-
to note
note
analysis,
tion as as well
well where
as researchers
vero-B andconductedused
vero-E6 cell a LOPAC
cell for compound
an in inforvitro library
assay [42,43]. for
[42,43]. inhibitor
It is
is selection
noteworthy as
to notewell
note
that
tion
that
as most
most
vero-B ofand
of the
as
the research
vero-B
research
vero-E6 and vero-E6
conducted
cell for an in tovitro
to search
for
searchan assay for anti-dengue
vitro assay
anti-dengue
[42,43]. It is drugs
drugs It
noteworthy have
have been
noteworthy
been
to note conducted
to
conducted
that most
that most
against
that most of the
viral
of the researchproteins
replication
research conducted
conducted or toto
viralsearch
searchenvelop for anti-dengue
for anti-dengue
proteins, butdrugs
ourbeen
drugs have been
research
have been conducted
focus is
conducted on aa
of the viral
against research conducted
replication to search
proteins or for
viral anti-dengue
envelop drugsbut
proteins, haveour conducted
research focus isagainst
on
against
human
against viral
protein
viral replication
as
replicationa drug proteins
target.
proteins or viral
There envelop
are numerous proteins, but
web-based our research
programs focus
available is onto aa
viral replication
human protein as proteins
a drug or viralor
target. viralare
envelop
There envelop
proteins,
numerous proteins,
but ourbut
web-based ourprograms
research research
focus isfocus on aishuman
available on to
human protein
implement protein as aa drug
drugwhereas,
target. There
There are numerous web-based programs available to
human
protein as this
implement athis
drugprocess,
as target.
process, target.
There
whereas, in
arein this
numerous
this are work,
numerous
work, an online
web-based
an online program,
web-based
programs
program, USRCAT,
programs
available
USRCAT, to has been
available
implement
has beento
implement
utilized,
implement which this
this process,
was
process, whereas,
incorporated in
with this
aonline
ZINCwork, an
database.online program,
Lipinski’s RuleUSRCAT,
of Five
Five has been
functions
this process,
utilized, which whereas,
was inwhereas,
this work, inan this work, an online
program, program,
USRCAT, has USRCAT,
been has which
utilized, been
utilized,
as a filter
utilized, which
to
which was incorporated
choose
was incorporated
drug-like
incorporated
with
with aaa ZINC
compounds
with ZINC
ZINC based
database.
database.
on
database. their
Lipinski’s
Lipinski’s
molecular
Lipinski’s
Rule
Rule of
of Five
property,
Rule of
functions
Five including
functions
functions
aswas incorporated with a ZINC database.based Lipinski’s Rule of Five functions asincluding
a filter to
as aaa filter
as filter to
absorption,
filter to
choose
choose drug-like
todistribution,
choose drug-like compounds
compounds
metabolism,
drug-like compounds andtheir based
excretion.
based
on
on their
on their molecular
molecular
Selected
their molecularcompounds
property,
property,
property, were including
chosen
including
choose
absorption, drug-like compounds
distribution, based on
metabolism, and molecular
excretion. property,
Selected compoundsincluding were absorption,
chosen
absorption,
based
absorption, on the distribution,
similarity
distribution, metabolism,
scores and
metabolism, and
drug-like excretion.
and Selected criteria.
excretion. Selected compounds
Selected compounds were chosen
distribution,
based on the metabolism,
similarity scores and andexcretion.
drug-like criteria. compounds were chosenwere based chosen
on the
based on
based on the
the similarity
similarity scores scores and and drug-like
drug-like criteria. criteria.
similarity scores and drug-like criteria.
3.1.2. Physiochemical
3.1.2. Physiochemical Properties Properties Analysis
Analysis (ADME) (ADME)
3.1.2.
3.1.2.
3.1.2. Physiochemical
Physiochemical
Physiochemical Properties
Properties
Properties Analysis
Analysis
Analysis (ADME)
(ADME)
(ADME)
ADME
ADME studies are
studies are crucial parts
crucial parts of any
of any drug development
drug development program program and and essential
essential for for
obedienceADME
ADMEADME withstudies
studies
studies are
regulatory crucial
arecrucial
are crucial parts
guidelines of
partsof[41].
parts any
[41].
ofany any drug
Indrug
drug development
this development
work, an acceptable
development program
acceptable
program
program rangeand
andand inessential
the values
values
essential
essential for
for
for
obedience
obedience with
with regulatory
regulatory guidelines
guidelines [41]. In
In this
this work,
work, an
an acceptable range
range in
in the
the values
of predicted
obedience
obedience
of predicted with physiochemical
with regulatoryguidelines
regulatory
physiochemical and
guidelines
and pharmaceutical
[41]. In
[41].
pharmaceutical
In this properties
this work,
properties of
an acceptable selected
acceptable
of
range
rangecompounds
in
in the
the values has
values of
ofpredicted
been
of predicted
achieved,
predicted physiochemical
physiochemical
which
physiochemical includesandand
and pharmaceutical
pharmaceutical
absorption,
pharmaceutical properties
properties
distribution,
properties of selected
of selected
metabolism,
of selected
selected and
compounds
compounds
compounds excretion
compounds has(Ta-
has
has
been
has
been
been achieved,
achieved, which
which includes
includes absorption,
absorption, distribution,
distribution, metabolism,
metabolism, and
andthe excretion
excretion (Ta-
(Ta-
achieved,
bles
been 4–6).
achieved, which
The waterincludes
which absorption,
solubility
includes of distribution,
a compound
compound
absorption, metabolism,
reflects
distribution, the and excretion
solubility
metabolism, of
and (Tables(Ta-
molecule
excretion 4–6).
in
bles
bles 4–6).
4–6). The
The water
water solubility
solubility of
of a
a compound reflects
reflects the
the solubility
solubility of
of the
the molecule
molecule in
◦ C,
in
The
water
bles 4–6).
water
water
at
at 25
25The solubility
°C,
°C, where
water
where
of
thea compound
threshold
solubility
the thresholdof a of reflects
of the
compound
the
the
solubility
solubility
solubility
reflectsis
is ˂0.05
˂0.05
of the
log
the solubility
log
molecule
mol/L.
mol/L. The
ofThe in water at
lipid-soluble
thelipid-soluble
molecule in 25
water
where
drug
water atless
isat 25 °C,
the
25 °C, where the
threshold
well-absorbed
where the threshold
of the solubility
than
threshold of the
the
water-soluble
of solubility
is <0.05 ones.
solubility is
log mol/L.
The
is ˂0.05
˂0.05Caco-2log mol/L.
The
log mol/L.
lipid-soluble
permeabilityThe lipid-soluble
The lipid-soluble
drug
cell line is less
line is
drug
drug is
is less
less well-absorbed
well-absorbed than
than water-soluble
water-soluble ones.
ones. The
The Caco-2
Caco-2 permeability
permeability cell
cell line is
isof
well-absorbed
composed
drug is less of than
human
well-absorbed water-soluble
epithelialthan ones.
colorectal
water-soluble The
cells, Caco-2
which
ones. permeability
is
The widely
Caco-2 used cellas line
permeabilityan is
in composed
vitro
cell model
line is
composed
composed of
of human
human epithelial
epithelial colorectal
colorectal cells,
cells, which
which is
is widely
widely used
used as
as an
an in
in vitro
vitro model
model
ofhuman
the human
composed
of the
epithelial
human of human colorectal
intestinal
intestinal mucosa
epithelial
mucosa
cells, towhich
predict
colorectal
to predict
is widely
thewhich
cells,
the
used
absorption
absorption
as an
is widely of in
of
vitroas model
an inof
orally-administered
used
orally-administered
themodel
vitro human
drugs
drugs
of the
intestinal
[44]. human mucosa intestinal
to mucosa
predict the to predict
absorption the
of
of the human intestinal mucosa to predict the absorption of orally-administered drugs absorption
orally-administered of orally-administered
drugs [44]. drugs
[44].
[44].
[44].
BioTech 2021, 10, 1 10 of 28

Table 4. The ADME results of GLC and analogues.

ZINC 3956760 ZINC16159409 ZINC3809846


Properties Model Name Units GLC
Compound 10 Compound 26 Compound 58
Water solubility log mol/L −1.377 −1.08 −1.005 −1.581
Caco2
Log 10− 6 cm/s −0.249 0.251 0.315 0.214
permeability

Absorption Intestinal
absorption %(Absorbed) 21.51 59.553 59.79 59.316
(human)
Skin
logKp −3.041 −3.044 −3.107 −2.939
Permeability
Fraction
unbound logL/Kg 0.82 0.841 0.84 0.853
(human)
Distribution BBB
logBB −0.943 −0.786 −0.9 −0.872
permeability
CNS
logPS −3.636 −3.552 −3.552 −3.192
permeability
CYP2D6 Yes/No No No No No
Metabolism
CYP1A2 Yes/No No No No No
Oral Rat Acute
mol/kg 0.626 0.535 0.535 0.534
Toxicity (LD50)
Excretion
Renal OCT2
Yes/No No No No No
substrate

Table 5. The ADME results of BG6 and analogues.

ZINC 4403145 ZINC 4530268 ZINC1576959


Properties Model Name Units BG6
Compound 30 Compound 36 Compound 38
Water solubility log mol/L −0.811 −1.753 −1.995 −1.753
Caco2
Log 10− 6 cm/s −0.341 −0.104 −0.371 −0.104
permeability

Absorption Intestinal
absorption %(Absorbed) 35.51 26.376 0 26.376
(human)
Skin
logKp −2.82 −2.744 −2.735 −2.744
Permeability
Fraction
unbound logL/Kg 0.716 0.79 0.886 0.232
(human)
Distribution BBB
logBB −1.414 −1.032 −0.928 −1.032
permeability
CNS
logPS −4.211 −3.46 −3.604 −3.46
permeability
CYP2D6 Yes/No No No No No
Metabolism
CYP1A2 Yes/No No No No No
Oral Rat Acute
mol/kg 0.414 0.787 1.056 0.787
Toxicity (LD50)
Excretion
Renal OCT2
Yes/No No No No No
substrate
BioTech 2021, 10, 1 11 of 28

Table 6. The ADME results of 2DG and analogues.

ZINC86652948 ZINC 86991606 ZINC86991603


Properties Model Name Units 2DG
Compound 1 Compound 4 Compound 31
Water solubility log mol/L −0.512 −1.987 −1.654 −1.376
Caco2
Log 10− 6 cm/s 1.559 1.985 1.654 1.098
permeability
Absorption Intestinal
absorption %(Absorbed) 86.433 86.983 87.984 86.764
(human)
Skin
logKp −3.41 −3.98 −2.34 −2.75
Permeability
Fraction
unbound logL/Kg 0.916 0.914 0.158 0.985
(human)
Distribution BBB
logBB −0.043 −0.025 −0.265 −0.265
permeability
CNS
logPS −3.434 −3.254 −3.214 −3.147
permeability
CYP2D6 Yes/No No No No No
Metabolism
CYP1A2 Yes/No No No No No
Oral Rat Acute
mol/kg 1.153 1.258 1.245 1.452
Toxicity (LD50)
Excretion
Renal OCT2
Yes/No No No No No
substrate

The water solubility of a compound reflects the solubility of the molecule in water
at 25 ◦ C. A lipid-soluble drug is less well-absorbed than water-soluble ones. The Caco-2
permeability cell line is composed of human epithelial colorectal cells, which is widely
used as an in vitro model of the human intestinal mucosa to predict the absorption of
orally-administered drugs [44]. These models predicted the Caco-2 permeability values
using a logarithm of the permeability coefficient (log cm/s) based on the 647 drug-like
molecules. For the ADME predictive model, a high Caco-2 permeability value would be
>0.90. Furthermore, intestinal absorption is a primary site for absorption of a drug orally,
where a less than 30% predated value is considered poor absorption while logKp > –2.5
defines the low skin permeability. This predictive model was built using the measured
free proportion of 552 compounds in human blood (Fu), where the predicted value should
be less than 2.81 log10−6 cm/s and logBB > 0.3 as well as logPS > 2 was considered
to readily cross the blood brain barrier (BBB) and penetrate the central nervous system,
respectively [41,44]. However, our findings successfully met the parameter of absorption
for each query molecule and the selected analogues.
The results (Tables 4–6) revealed that all compounds possess good water solubility,
Caco2 permeability, human intestinal absorption, and skin permeability values. Greater
values of these parameters denote that the compound could be better absorbed from the
intestinal tract upon oral administration. Moreover, fraction unbound values (Fu < 1)
indicate that the compounds exist in plasma at an unbound state and could efficiently
travel through the cellular membrane. In addition, all compounds displayed negative
penetration through the Blood Brain Barrier (BBB) and Central Nervous System (CNS),
which means that these compounds do not cross the BBB and CNS (Tables 4–6).
CYP2D6 and CYP3A4 are two isoforms of Cytochrome p450, which is a crucial detoxi-
fication enzyme of the human body and responsible for changing the pharmacokinetics of
drugs. Thus, the tested compounds should be inactive (Tables 4–6). In terms of metabolism,
we found that all compounds act as a non-inhibitor of the cytochrome p450, which means that
BioTech 2021, 10, 1 12 of 28

the molecule will not restrict the biotransformation of the drug metabolized by the CY2D6
enzyme and the CYP1A2 enzyme. Furthermore, renal OCT2 is an organic cation transporter 2,
which plays a crucial role in renal clearance and disposition of drugs. Thus, tested compounds
should not act as inhibitors of OCT2 and has to be inactive during experiments.
In excretion, we found that all compounds are inactive for renal OCT2 inhibitors,
while the predicted LD50 value of these compounds are >500 mg/kg, suggesting that
these compounds do not possess acute oral toxicity at lower doses (Tables 4–6). Important
information obtained from SwissADME was the computed LD50 dose in a rat model.
By comparing the LD50 doses, a compound with a lower dose was found to be more
lethal than the compound with a higher LD50. From our observation, all compounds
possess higher LD50 values (Tables 4–6). The physiochemical properties showed that all
compounds, which are analogues of GLC, BG6, and 2DG acquiring drug-like properties,
can be good inhibitors of human HKII protein when tested in vitro.

3.1.3. Toxicity Test


The toxicity test in this study was conducted by using the Pro Tox-II program. The
prediction of compound toxicities is an important part of a drug design development
process. Computational toxicity estimations are not only faster than the determination
of toxic doses in animals, but may also help reduce the amount of animal experiments.
ProTox-II incorporates molecular similarity, fragment propensities, most frequent features,
and (fragment similarity-based CLUSTER cross-validation) machine-learning, based on a
total of 33 models for the prediction of various toxicity endpoints such as acute toxicity,
hepatotoxicity, cytotoxicity, carcinogenicity, mutagenicity, immunotoxicity, adverse out-
comes (Tox21) pathways and toxicity targets [45]. Acceptable toxicity value of GLC, BG6,
and 2DG analogues have been tabulated in Tables 7–9, respectively. The toxicity has been
classified into five classes: Class I: fatal if swallowed (LD50 ≤ 5), Class II: fatal if swallowed
(5 < LD50 ≤ 50), Class III: toxic if swallowed (50 < LD50 ≤ 300), Class IV: harmful if swal-
lowed (300 < LD50 ≤ 2000), Class V: may be harmful if swallowed (2000 < LD50 ≤ 5000),
and Class VI: non-toxic (LD50 > 5000). Toxicity is proven essential in examining com-
pounds for potential inclusion into drug development. The aim of toxicity studies is to
ensure safety of the chemical compounds before they can be used as drugs or during
clinical trials, as well as to determine the toxic effects of test substances [46]. In several
experiments, different types of toxicity tests (acute toxicity, sub-acute toxicity, and chronic
toxicity studies) have been conducted to characterize the possible toxic effects of drugs that
can range from minor to critical [46,47].

Table 7. Toxicity values of GLC and analogues.

Classification
ZINC ID Toxicity End Point Organ
Acute Toxicity
Carcinogenicity Immunotoxicity Mutagenicity Cytotoxicity Toxicity

I. Toxicity Class: 6
Inactive Inactive Inactive Inactive Inactive
GLC II. LD50:2300 mg/kg
Ps: 0.82 Ps: 0.99 Ps: 0.87 Ps: 0.81 Ps: 0.98
III. accuracy: 70.97%
I. Toxicity Class: 6
3956760 Inactive Inactive Inactive Inactive Inactive
II. LD50:14,388 mg/kg
Compound 10 Ps: 0.67 Ps: 0.99 Ps: 0.68 Ps: 0.68 Ps: 0.98
III. accuracy: 67.38%
I. Toxicity Class: 6
16159409 Inactive Inactive Inactive Inactive Inactive
II. LD50:2300 mg/kg
Compound 26 Ps: 0.73 Ps: 0.98 Ps: 0.68 Ps: 0.64 Ps: 0.89
III. accuracy: 68.07%
I. Toxicity Class: 6
3809846 Inactive Inactive Inactive Inactive Inactive
II. LD50:14,388 mg/kg
Compound 58 Ps: 0.73 Ps: 0.98 Ps: 0.68 Ps: 0.64 Ps: 0.89
III. accuracy: 67.38%
BioTech 2021, 10, 1 13 of 28

Table 8. Toxicity values of BG6 and analogues.

Classification
ZINC ID Toxicity End Point Organ
Acute Toxicity
Carcinogenicity Immunotoxicity Mutagenicity Cytotoxicity Toxicity

I. Toxicity Class: 4
Inactive Inactive Inactive Inactive Inactive
BG6 II. LD50:1500 mg/kg
Ps: 0.74 Ps: 0.99 Ps:0.74 Ps:0.81 Ps:0.94
III. accuracy: 67.38%
I. Toxicity Class: 6
4403145 Inactive Inactive Inactive Inactive Inactive
II. LD50:15,900 mg/kg
Compound 30 Ps: 0.89 Ps: 0.99 Ps: 0.85 Ps: 0.68 Ps: 0.94
III. accuracy: 70.97%
I. Toxicity Class: 6
4530268 Inactive Inactive Inactive Inactive Inactive
II. LD50:5500 mg/kg
Compound 36 Ps: 0.69 Ps: 0.99 Ps: 0.73 Ps: 0.68 Ps: 0.93
III. accuracy: 69.26%
I. Toxicity Class: 6
1576959 Inactive Inactive Inactive Inactive Inactive
II. LD50:15,900 mg/kg
Compound 38 Ps: 0.89 Ps: 0.99 Ps: 0.85 Ps: 0.68 Ps: 0.94
III. accuracy: 70.97%

Table 9. Toxicity values of 2DG and analogues.

Classification
ZINC ID Toxicity End Point Organ
Acute Toxicity
Carcinogenicity Immunotoxicity Mutagenicity Cytotoxicity Toxicity

I. Toxicity Class: 3
Inactive Inactive Inactive Inactive Inactive
2DG II. LD50:1500 mg/kg
Ps: 0.74 Ps: 0.99 Ps:0.74 Ps:0.81 Ps:0.94
III. accuracy: 67.38%
I. Toxicity Class: 6
86652948 Inactive Inactive Inactive Inactive Inactive
II. LD50:15,900 mg/kg
Compound 1 Ps: 0.89 Ps: 0.99 Ps: 0.85 Ps: 0.68 Ps: 0.94
III. accuracy: 70.97%
I. Toxicity Class: 6
86991606 Inactive Inactive Inactive Inactive Inactive
II. LD50:5500 mg/kg
Compound 4 Ps: 0.69 Ps: 0.99 Ps: 0.73 Ps: 0.68 Ps: 0.93
III. accuracy: 69.26%
I. Toxicity Class: 6
86991603 Inactive Inactive Inactive Inactive Inactive
II. LD50:15,900 mg/kg
Compound 31 Ps: 0.89 Ps: 0.99 Ps: 0.85 Ps: 0.68 Ps: 0.94
III. accuracy: 70.97%

The Pro-tox-II software utilizes the consensus method to determine the values of
different compounds by using different tests end-point analysis approaches, where LD50
and LC50 values are exhibited for each compound (Tables 7–9). A smaller value indicates a
more toxic compound and vice versa. According to the data analysis results obtained in the
present study, most of the compounds that were similar to 2DG were toxic, but, in contrast,
the toxicity of the top-ranked analogues of GLC exhibited non-toxic effects. Moreover, the
analogues of BG6 were predicted to be non-toxic. It should also be noted that all examined
compounds were negative for mutagenicity. As already mentioned previously, 2DG is a
known inhibitor of HKII, but it has shown a toxic effect in in in vitro analysis [19,20], which
highlighted the urgency to search for compounds that can be developed into a potent HKII
inhibitor, which are non-toxic and safe to be administered as medications.

3.2. Structure-Based Screening


Structure-based virtual screening (SBVS) is an impactful experiment instrumental
for fast and cost-effective lead discovery and optimization. The aim of this process is
to understand the three-dimensional structure of the biological target at a molecular
level [48]. Structure-based screening states the interaction of protein-ligand binding, where
BioTech 2021, 10, 1 14 of 28

the mode of the binding complex can be predicted through docking and scoring functions,
that comprise a two-step calculation to calculate the free binding energy of complex and
exploring the conformational space of a protein and ligand complex [48]. The advantages
of SBVS are time-saving and cost effective. There is no need for physical existences of
the molecule and there are available free access software and web tools for conducting
SBVS [48,49]. There are some disadvantages too. SBVS can generate a false positive and a
false negatives result, as some tools work in specific cases and not in general, while it is
also difficult to predict the accurate binding positions [49].
In this experiment, the crystal structure of human HKII from the Protein Data Bank
(PDB ID: 2NZT) was prepared for molecular docking analysis by protein optimization and
energy minimization. Additionally, all heteroatoms and ligands, which were already in
a complex with the protein, were removed, because they can affect the docking process.
HKII consists of two protein chains with two domains each. All 12 compounds were
docked sequentially in each terminal of both chains, even though the two chains are
homodimers. This was done to observe variations in the binding energy and hydrogen
bond numbers between the two chains, allowing us to determine which of the two was the
best for the subsequent simulation procedure. In this study, we have conducted a docking
procedure on the N-terminal of chain A because several studies have suggested that the N-
terminus (residues 1-475) contains more catalytic sites [35]. Our analysis involved docking
12 compounds with the selected catalytic residues for each of the query molecules (2DG,
GLC, and BG6). All the top-ranked molecules were successfully docked in the catalytic site
of human HKII.
The top-ranked analogues of GLC, known as compound 10, compound 26, and
compound 58, had binding energies of −7.2, −7.0, and −6.10 kcal/mol, respectively,
whereas the binding energy of GLC itself was −7.2 kcal/mol on the N-terminus of Chain
A (Table 10). As described previously, the binding energy required for either chain of the
HKII homodimer was similar. Among all analogues of GLC, compound 10 exhibited the
best binding energy (−7.0 kcal/mol) and took part in seven hydrogen bonds with residues
Lys173, Asn208, Asp209, Glu260, Phe156, and Gln291 on HKII (Figure 2A). This closely
resembles the query molecule GLC, which has a binding energy of −7.2 kcal/mol and
takes part in six hydrogen bonds with residues Glu260, Lys173, Asp209, and Asn235 on
HKII (Figure 2B). However, compound 10 exhibited the best binding characteristics of the
tested GLC analogues and might act as a potential inhibitor of HKII.

Table 10. The top molecules, which are analogues of GLC, were ranked, according to their binding energy. The binding
energy of GLC is also shown.

Binding Energy (Kcal/mol) Hydrogen Bond Number


ZINC ID Catalytic Residue
Chain A Terminal N Chain A Terminal N
GLC (−7.2) 6 Glu260, Lys173, Asp209, Asn235
3956760 Glu260, Gln291, Phe156, Asn208,
(−7.2) 7
Compound 10 Lys173, Asp209
16159409 Ser155, Asp209, Asn208,
(−7.0) 5
Compound 26 Lys173, Glu294
3809846
(−6.10) 2 Asp209, Asn209, Ser155
Compound 58
docking approaches [50–57]. However, it is important to note that all of the previously
mentioned literature reported inhibitors which were developed based on viral proteins.
There are plenty of in silico research on viral proteins were not fruitful for the development
of dengue drug. Thus, in this research, we focused on human protein as a drug target.
BioTech 2021, 10, 1 Overall, structure-based processes, such as molecular docking, provide a good approxi- 15 of 28
mation of protein-ligand binding affinity and help to identify the best candidates for fur-
ther inhibition studies in antiviral drug development.

Figure
Figure 2. The
2. The chain
chain A, N-terminal
A, N-terminal (red(red colour)
colour) andand C-terminal
C-terminal (blue
(blue colour)
colour) of HKII
of HKII waswas indicated
indicated in ribbon
in ribbon representation
representation
andand
thethe
black circle
black indicates
circle thethe
indicates catalytic site,site,
catalytic where GLC
where andand
GLC its its
analogues were
analogues docked.
were (A)(A)
docked. Compound
Compound 10, 10,
which hashas
which
formed
formedoneoneHH bond
bond with Gln291,
with Gln291,Asp209,
Asp209,Phe156,
Phe156,andandtwo
two H
H bond formed with
bond formed withLys173,
Lys173,Asn208,
Asn208,and
andGlu260.
Glu260.(B)(B) Mean-
Meanwhile,
while, the catalytic
the catalytic residue
residue Lys173
Lys173 has has
shownshown
three three H bonds,
H bonds, Asp209
Asp209 withwith
one,one, Asn235
Asn235 withwith
one,one,
andand Glu260
Glu260 withwith
oneone H
H bond,
bond, each formed with
each formed with GLC. GLC.

The analogues of BG6, known as compound 30, compound 36, and compound 38,
exhibited binding energies of −7.8, −7.4, and −7.0 kcal/mol, respectively, compared to
BG6 itself, which has a binding energy of −7.9 kcal/mol (Table 11). Compound 30 was
chosen as the analogue with the best binding characteristics because it had the lowest
binding energy value (−7.8 kcal/mol) and forms eleven hydrogen bonds, with Gly87,
Asn89, Thr232, Arg91, Asp84, and Lys173 (Figure 3A). BG6 forms up to six hydrogen bonds
with Lys173, Asn208, Gln291, Asp209, Thr233, Pro157, and Gly87 (Figure 3B). Therefore,
the compound with the best binding characteristics among BG6 analogues was compound
30, which may be a potential inhibitor of HKII.
BioTech 2021, 10, 1 16 of 28

Table 11. The top molecules, which are analogues of BG6, were ranked according to their binding energy. The binding
energy of BG6 is also shown.

Binding Energy (Kcal/mol) Hydrogen Bond Number


ZINC ID Catalytic Residue
Chain A, Terminal N Chain A Terminal N
Gln291, Pro157, Lys173, Thr232, Gly87,
BG6 (−7.9) 6
Asp209, Asn208
4403145
(−7.8) 11 Gly87, Asn89, Thr232, Arg91, Asp84, Lys173
Compound 30
4530268
(−7.4) 9 Asp84, Thr88, Thr323, Asn89
Compound 36
1576959
(−7.0) 5 Asn89, Thr88, Ser449
Compound 382, FOR PEER REVIEW
BioTech 2021, 17

3. The
FigureFigure chain
3. The A Aand
chain andN-terminal (redcolour)
N-terminal (red colour)andand C-terminal
C-terminal (Blue (Blue
colour)colour)
of HKII of
wasHKII was in
indicated indicated in a ribbon
a ribbon repre-
sentation and
representation andaablack
blackcircle indicates
circle thethe
indicates catalytic site, site,
catalytic where BG6 and
where BG6itsand
analogues were docked.
its analogues (A) Compound
were docked. 30 has
(A) Compound 30
formed one H bond with a catalytic residue Asn89 and Arg91, while Thr232, Lys173, and Gly87 formed two H bonds and
has formed one H bond with a catalytic residue Asn89 and Arg91, while Thr232, Lys173, and Gly87 formed two H bonds
Asp formed three H bonds with compound 30. (B) Meanwhile, BG6 has formed one H bond with each residue Gln291,
and Asp formed
Asn209, threeThr233,
Asp208, H bonds andwith compound 30. (B) Meanwhile, BG6 has formed one H bond with each residue Gln291,
Lys173.
Asn209, Asp208, Thr233, and Lys173.
BioTech 2021, 10, 1 17 of 28

The 2DG analogues, known as compound 1, compound 4, and compound 31, ex-
hibited binding energies of −6.8, −6.3, and −6.3 kcal/mol, respectively, as shown in
Table 12. It should be noted that compound 1 exhibited a relatively low binding energy
(−6.8 kcal/mol), despite forming four hydrogen bonds with Asn208, Asp209, Thr172,
Glu294, Thr232, and Lys173 (Figure 4A), compared to the binding energy of 2DG
(−6.0 kcal/mol), which forms eight hydrogen bonds with Asn208, Asp209, Thr172, Glu294,
Lys173, Glu260, and Phe156 (Figure 4B). Therefore, compound 1 was selected for further in-
hibition analysis due to its relatively high binding affinity, compared to the other analogues
of 2DG, and 2DG itself.

Table 12. The top molecules, which are analogues of 2-DG, were ranked according to their binding energy. The binding
energy of 2-DG is also shown.

Binding Energy (Kcal/mol) Hydrogen Bond Number


ZINC ID Catalytic Residue
Chain A Terminal N Chain A Terminal N
Asn208, Asp209, Thr172, Lys173, Glu294,
2DG (−6.0) 8
Glu260, Phe156
86652948 Asp209, Asn208, Glu294, Thr172,
(−6.8) 4
Compound 1 Thr232, Lys173
86991606
(−6.3) 3 Asp209, Asp84, Gly87, Thr88, Asp413, Thr232
Compound 4
86991603
(−6.3) 1 Asp413, Asp84, Asp209
Compound 31

Previous studies have attempted to use molecular docking approaches to identify


small molecule inhibitors of dengue virus proteins. For example, Lim and colleagues [50]
attempted to identify NS5 methyltransferase (MTase) inhibitors using a molecular docking
approach. NS5 is a non-structural viral protein located on the dengue virus surface that
assists in viral replication. Hence, it has been proposed as a drug target, with ribavirin
triphosphate (RTP) and S-adenosyl-L-methionine (SAM) acting as query molecules. Lim
and colleagues reported that compounds SPH1-103799, SPH1-101-102, and 28SPH1-115-917
had the highest binding affinities of −11.4 kcal/mol, −11.4 kcal/mol, and −10.0 kcal/mol,
respectively [50]. In this study, AutoDock Vina and EDULISS (Edinburgh University Ligand
Selection System) were used for structure-based and ligand-based screening, respectively.
Another study by Wang and colleagues used four-stage computational high-throughput
screening to find a small molecule directed against the dengue virus envelope protein
(E protein) [51]. The 23 highest ranking compounds in their analysis were selected based
on the E protein binding site from three NIC libraries and were subsequently tested
for antiviral activity in a biological assay. One compound, PO2, was found to inhibit
viral reproduction.
Thus, extensive research on the dengue virus has been conducted using molecular
docking approaches [50–57]. However, it is important to note that all of the previously
mentioned literature reported inhibitors which were developed based on viral proteins.
There are plenty of in silico research on viral proteins were not fruitful for the development
of dengue drug. Thus, in this research, we focused on human protein as a drug target.
Overall, structure-based processes, such as molecular docking, provide a good approxima-
tion of protein-ligand binding affinity and help to identify the best candidates for further
inhibition studies in antiviral drug development.
BioTech 2021, 2, FOR PEER REVIEW 18
BioTech 2021, 10, 1 18 of 28

Figure 4. The chain A and N-terminal (red color) and C-terminal (Blue color) of HKII was indicated in a ribbon representation
and the black circle indicates the catalytic site, where 2DG and its analogues were docked. (A) Compound 1 formed four H
bonds4.with
Figure Thecatalytic
chain A residues Thr232 (red
and N-terminal and Asn208. (B)C-terminal
color) and Meanwhile, 2DGcolor)
(Blue has formed
of HKIIone
wasHindicated
bond withineach residue
a ribbon Glu294,
represen-
Asn208, and Thr232, while two H bonds formed with Lys173, Asp209, and Phe156.
tation and the black circle indicates the catalytic site, where 2DG and its analogues were docked. (A) Compound 1 formed
four H bonds with catalytic residues Thr232 and Asn208. (B) Meanwhile, 2DG has formed one H bond with each residue
Glu294, Asn208, and Thr232, 3.3. Molecular
while Dynamics
two H bonds formedSimulation
with Lys173, Asp209, and Phe156.
The structural deviations and stability of HKII docked with compound 1, compound
10,Molecular
3.3. and compound
Dynamics 30,Simulation
which are analogues of 2DG, GLC, and BG6, respectively, were
assessed by 20 ns of MD
The structural deviationssimulation usingofGROMACS
and stability HKII docked4.6.5 programme.
with compoundThe stability of
1, compound
the protein-ligand complex was observed by measuring the root-mean-square
10, and compound 30, which are analogues of 2DG, GLC, and BG6, respectively, deviation
were
(RMSD) of each trajectory obtained in an MD simulation with respect
assessed by 20 ns of MD simulation using GROMACS 4.6.5 programme. The stability to their position
of
in protein-ligand
the the reference frame.
complex The RMSD
was of allby
observed selected compounds
measuring fit into the HKII
the root-mean-square protein
deviation
during the
(RMSD) 20 nstrajectory
of each period ofobtained
dynamicsinsimulation studies that
an MD simulation arerespect
with shown to in their
Figureposition
5, Figure
in7,
and Figure 8. Ismail and Jusoh confirmed that RMSD determines the deviation
the reference frame. The RMSD of all selected compounds fit into the HKII protein during in the
average distance of the atomic structure movement in complexes, where generally the
the 20 ns period of dynamics simulation studies that are shown in Figures 5, 7, and 8.
threshold of RMSD is 2Å [58]. The complex structure namely HKII-2DG, HKII-BG6,
Ismail and Jusoh confirmed that RMSD determines the deviation in the average distance
and HKII-GLC were used as references and comparison. Throughout the simulation,
of the atomic structure movement in complexes, where generally the threshold of RMSD
the complex HKII-Compound 1 and HKII-2DG were seen equilibrated and their protein
is 2Å [58]. The complex structure namely HKII-2DG, HKII-BG6, and HKII-GLC were used
backbones were in minimal deviations after 15 ns of simulation. In detail, the backbone of
as references and comparison. Throughout the simulation, the complex HKII-Compound
the complex HKII-compound 1 showed a slight fluctuation compared to the reference HKII-
1 and HKII-2DG were seen equilibrated and their protein backbones were in minimal de-
2DG complex toward the end of the simulation with the RMSD values consistent at ±0.5Å
viations after 15 ns of simulation. In detail, the backbone of the complex HKII-compound
BioTech 2021, 10, 1 19 of 28

(Figure 5A), indicating that the interaction of HKII with compound 1 was comparable
to 2DG. The HKII-2DG and HKII-compound 1 complexes were snapshots at one point
of production simulation and both structures were superimposed to assess the ligand-
binding topology as in Figure 5C. Both substrates were seen in a similar coordination, thus
portraying the same level of inhibition towards the enzyme. The binding interaction of the
HKII-compound 1 and HKII-2DG complexes were closely monitored and differentiated
BioTech 2021, 2, FOR PEER REVIEW along the simulation trajectory. For root mean square fluctuation (RMSF), each amino acid 20
fluctuation of HKII for compound 1 was in a similar pattern with reference compound 2DG
(Figure 5B). No noteworthy changes were seen for the movement of the amino acids and
the compounds did not disrupt the structural integrity of HKII.

A B

Figure 5. (A) The backbone RMSD of HKII-2DG (black) and HKII-compound 1(red) complexes shown during 20 ns of the
Figure
MD 5. (A) The
simulation. (B)backbone RMSDofofHKII-2DG
Residue RMSF HKII-2DGand
(black) and HKII-compound
HKII-compound1 complexes1(red) complexes
generated shown
during during 20period
the trajectory ns of the
of
MD
20 ns simulation. (B) Residue(C)
of the MD simulation. RMSF of HKII-2DGofand
Superimposition the HKII-compound1
trajectory structurecomplexes generated
at 10 ns and during
20 ns of MD the trajectory
simulations. period
The protein
of 20 ns of the MD simulation. (C) Superimposition of the trajectory structure at 10 ns and 20 ns of MD simulations. The
was presented in a cyan-coloured ribbon format. Snapshots of superimposed ligand 2DG (red) and compound 1(blue)
protein was presented in a cyan-coloured ribbon format. Snapshots of superimposed ligand 2DG (red) and compound
located in a ligand binding site.
1(blue) located in a ligand binding site.
The binding stabilities of the protein-compound complexes were monitored during
The binding
the trajectory stabilities
period of the of MDthesimulations.
protein-compound complexes
The stabilities wereprotein-compound
of the monitored during
complexes were evaluated by calculating the H bond profiles using the g, h bond com-
the trajectory period of the MD simulations. The stabilities of the protein-compound tool
plexes
of were[58,59].
Gromacs evaluated
The by calculating
binding therevealed
analysis H bondthatprofiles using the
the complex g, h bond
of 2DG in HKIItool
hadof
Gromacs [58,59]. The binding analysis revealed that the complex
formed an average of six hydrogen bonds higher than the HKII-compound 1 complex of 2DG in HKII had
formed
(three an average
hydrogen bonds)of over
six hydrogen bondstime
the simulation higher than
(Figure the HKII-compound
6B,C). 1 complex
Figure 6B,C have shown the
(three hydrogen bonds) over the simulation time (Figure 6B,C). Figure 6B,C have shown
the extracted trajectory of HKII-2DG and HKII-compound 1 at 20 ns, where compound 1
bind with HKII by three H bond with catalytic residue of Asp84, Asp209, and Asp413,
while the reference compound 2DG showed six H bonds with residue Asp84, Asn89,
Asp209, Thr232, Thr88, and Ser415 (Figure 6B,C). This analysis has suggested that 2DG
BioTech 2021, 10, 1 20 of 28

extracted trajectory of HKII-2DG and HKII-compound 1 at 20 ns, where compound 1 bind


with HKII by three H bond with catalytic residue of Asp84, Asp209, and Asp413, while
the reference compound 2DG showed six H bonds with residue Asp84, Asn89, Asp209,
Thr232, Thr88, and Ser415 (Figure 6B,C). This analysis has suggested that 2DG has a strong
interaction with HKII in the dynamic condition during MD simulation. However, the
formation of H bonds for compound 1 are lower than 2DG, but the compound consistently
BioTech 2021, 2, FOR PEER REVIEW 21
bound in the active site of HKII stably projected the active site vicinity. Another useful
analysis is a distance calculation between the HKII and selected compounds. Since our
aim is to obtain the stability of protein-ligand binding, the distances between the ligand-
the ligand-protein
protein haveas
have to be as close topossible.
be as close as possible.
Figure Figure
6B,C depict 6B,C
the depictbetween
distance the distance
HKIIbetween
and
HKII and
compound compound
1 and reference1 molecule
and reference
2DG,molecule 2DG,
which were which
1.09Å andwere 1.09Å
1.03Å, and 1.03Å,
respectively, respec-
which
tively,
indicate which
that indicate that
all compounds all compounds
strongly bind withstrongly bind with the
HKII throughout HKII throughout the simu-
simulation.
lation.

B C

2DG Compound 1
Figure 6. (A)
Figure 6. Total number
(A) Total of hydrogen
number bond interactions
of hydrogen between
bond interactions HKII and
between 2DG
HKII and(black)
2DGand HKIIand
(black) andHKII
compound 1 (red). 1
and compound
Snapshots of complex (B) HKII-2DG and (C) HKII-compound 1 were extracted from the trajectory at 20 ns.
(red). Snapshots of complex (B) HKII-2DG and (C) HKII-compound 1 were extracted from the trajectory at 20 ns. HKII- HKII-2DG
has shown
2DG has sixshown
H bonds.
six HTwo formed
bonds. Twowith Thr232
formed withand Asp413,
Thr232 and one formed
Asp413, onewith
formedAsn89,
withone formed
Asn89, onewith Asp84,
formed withand two and
Asp84,
formed
two with
formed Thr88,
withSer415. The HKII-compound
Thr88, Ser415. 1 has shown
The HKII-compound three H
1 has shown bonds.
three ThreeThree
H bonds. formed with with
formed Asp413, Asp84,
Asp413, and and
Asp84,
Asp209.
Asp209. The The distance
distance between
between HKIIHKII
and and
2DG2DG (cyan),
(cyan), compound
compound 1(cyan)
1(cyan) are,average,
are, on on average,
1.03Å1.03Å and 1.09Å,
and 1.09Å, respectively.
respectively.

For the HKII complex with BG6 and compound 30, both were stabilized over the
simulation process while the complex HKII-GLC and HKII-compound 10 were less devi-
ated after 12 ns. Clearly, the structural dynamics of all complexes achieved stability before
20 ns. For compound 30, the backbone of the HKII complex showed comparable devia-
tions with the reference complex, HKII-BG6, where their RMSD values were plateaued at
±4.0Å (Figure 7A), indicating that both complexes were equilibrated and achieved a fa-
BioTech 2021, 10, 1 21 of 28

BioTech 2021, 2, FOR PEER REVIEW 22

For the HKII complex with BG6 and compound 30, both were stabilized over the
simulation process while the complex HKII-GLC and HKII-compound 10 were less devi-
All complexes (HKII-compound 30 and BG6, HKII-compound 10 and GLC) were
ated after 12 ns. Clearly, the structural dynamics of all complexes achieved stability before
snapshotted at 20 ns of MD production and superimposed with their reference structure
20 ns. For compound 30, the backbone of the HKII complex showed comparable deviations
to assess
with the binding
the reference location,
complex, as shown
HKII-BG6, in Figures
where 7C and
their RMSD 8C. From
values were the superposed
plateaued im-
at ±4.0Å
age, all substrates were in a close orientation with a reference and predicted to
(Figure 7A), indicating that both complexes were equilibrated and achieved a favourable effectively
inhibit
bindingHKII through awith
conformation strong binding
each interaction.
substrate. For RMSF,
Meanwhile, each amino
for compound acidbackbone
10, the fluctuation
of
in HKII for compound 30 and compound 10 were in a similar pattern
HKII-compound 10 showed a similar fluctuation with the HKII-GLC with an RMSD with their references
value
BG6 andAGLC,
of ±5.0 respectively.
◦ (Figure No significant
8A), indicating changes were
that both complexes seen for
behaved the movement
similarly of the
and performed
amino acids and the substrates were not disrupting
stable interactions in the aqueous environment. the structural integrity of HKII.

A B

7. (A) The
Figure 7. The backbone
backbone RMSD
RMSD of of HKII-BG6
HKII-BG6 (black) and HKII-compound
HKII-compound 30 (red) complexes shown during 20 ns of
simulation. (B) Residue RMSF of HKII-BG6 and HKII-compound 30 complexes generated during the trajectory period
MD simulation.
of 20
of 20 ns
ns of
of MD
MDsimulation.
simulation.(C)
(C)Superimpose
Superimposeofoftrajectory structure
trajectory at at
structure 10 10
ns ns
andand
20 ns
20 of
nsMD simulations.
of MD The The
simulations. protein was
protein
presented in a cyan-coloured ribbon format. Snapshots of superimposed ligand BG6 (red) and compound 30
was presented in a cyan-coloured ribbon format. Snapshots of superimposed ligand BG6 (red) and compound 30 (yellow) (yellow)
located in a ligand binding site.
located in a ligand binding site.
BioTech 2021,
2021, 10,
2, FOR
1 PEER REVIEW 22 of 23
28

A B

Figure 8. (A) The backbone RMSD of HKII-GLC (black) and HKII-compound 10 (red) complexes shown during 20 ns of
Figure 8. (A) The(B)
MD simulation. backbone
Residue RMSD
RMSF of ofHKII-GLC
HKII-GLCand(black) and HKII-compound
HKII-compound 10 (red)
10 complexes complexes
generated shown
during during 20period
the trajectory ns of
MD simulation. (B) Residue RMSF of HKII-GLC and HKII-compound 10 complexes generated during the trajectory
of 20 ns of the MD simulation. (C) Superimpose of trajectory structure at 10 ns and 20 ns of MD simulations. The protein period
of 20presented
was ns of the MD
in a simulation. (C) ribbon
cyan-coloured Superimpose
format.ofSnapshots
trajectory of
structure at 10 nsligand
superimposed and 20GLC
ns of(red)
MD and
simulations.
compound The10protein
(blue)
was presented in a cyan-coloured ribbon format. Snapshots of superimposed ligand GLC (red) and compound 10 (blue)
located in a ligand binding site.
located in a ligand binding site.
All complexes (HKII-compound 30 and BG6, HKII-compound 10 and GLC) were
The binding
snapshotted at 20 analysis
ns of MDofproduction
the HKII-compound 30 revealed
and superimposed witha higher averagestructure
their reference number of to
hydrogen bond (7 hydrogen bonds) per time frame during the MD simulation
assess the binding location, as shown in Figures 7C and 8C. From the superposed image, period (Fig-
all
ure 9A) compared
substrates were in atoclose
a hydrogen bond
orientation of the
with HKII-BG6
a reference complex
and predicted(fiveto hydrogen
effectivelybonds).
inhibit
The extracted trajectory of complex HKII-compound 30 has shown
HKII through a strong binding interaction. For RMSF, each amino acid fluctuation seven H bonds with
in HKII
Asp84, Thr88, Asn89,
for compound 30 andThr232,
compound Ser415,
10 and
wereGly87 at 20 nspattern
in a similar during with
the MD simulation,
their referenceswhile
BG6
the
andsnapshot of complexNo
GLC, respectively. HKII-BG6 haschanges
significant shown five H bonds
were formed
seen for with residues
the movement of theAsn208,
amino
Asp209,
acids and Lys173, and Gln291
the substrates wereatnot
20 ns of the MD
disrupting simulation
the structural(Figure 9B,C).
integrity This analysis has
of HKII.
suggested
The binding analysis of the HKII-compound 30 revealed a higher averagecondition
that compound 30 has strong interactions with HKII in the dynamic number
during MD simulation,
of hydrogen compared
bond (7 hydrogen to reference
bonds) per timemolecule BG6. However,
frame during compound period
the MD simulation 30 and
BG6 were
(Figure 9A)consistently
compared to bound in the active
a hydrogen bond ofsite
theofHKII-BG6
HKII andcomplex
stably projected the active
(five hydrogen site
bonds).
vicinity. The distance
The extracted between
trajectory compound
of complex 30 and BG6 with
HKII-compound HKII
30 has shownwereseven
belowH1.05Å,
bondswhich
with
defines that their
Asp84, Thr88, binding
Asn89, was
Thr232, strongand
Ser415, during
Gly87the simulation
at 20 ns duringtime
the MDwithsimulation,
distance values
while
mentioned
the snapshot inof
Figure 9B,C.
complex HKII-BG6 has shown five H bonds formed with residues Asn208,
Asp209, Lys173, and Gln291 at 20 ns of the MD simulation (Figure 9B,C). This analysis has
BioTech 2021, 2, FOR PEER REVIEW 24

BioTech 2021, 10, 1 23 of 28


Furthermore, compound 10 with HKII has formed an average of six hydrogen bonds,
which is higher than the reference complex HKII-GLC (4 hydrogen bonds) (Figure 10A).
Figure that
suggested 10B,C have shown
compound the strong
30 has extracted trajectorywith
interactions of HKII-GLC anddynamic
HKII in the HKII-compound
condition10 at
during MD simulation, compared to reference molecule BG6. However, compoundAsp209
20 ns, where GLC binds with HKII by four H bonds with catalytic residues 30 and and
Lys173, while compound 10 has shown six H bonds with a catalytic residue
BG6 were consistently bound in the active site of HKII and stably projected the active of Asn209,
site
Lys173, Gln291, and Asp209 (Figure 10B,C). Thus, it can be said that
vicinity. The distance between compound 30 and BG6 with HKII were below 1.05Å, which compound 10 has
more intermolecular interactions via hydrogen bonding and the compound
defines that their binding was strong during the simulation time with distance values was stably-
bound in
mentioned in the catalytic
Figure 9B,C. region of HKII, showing a high level of inhibition compared to the
reference molecule GLC.

B C

BG6 Compound 30
Figure 9. (A)9.Total
Figure (A) number of hydrogen
Total number bond interactions
of hydrogen betweenbetween
bond interactions HKII andHKII
BG6 (red),
and BG6and (red),
HKII and
andcompound
HKII and 30 (black). 30
compound
Snapshots of complex (B) HKII-BG6, and (C) HKII-compound 30 were extracted from the trajectory at 20 ns. HKII-BG6
(black). Snapshots of complex (B) HKII-BG6, and (C) HKII-compound 30 were extracted from the trajectory at 20 ns. HKII-
has shown
BG6 hasfive H bonds.
shown five HRwo
bonds.formed with Asn208
Rwo formed and Asp209,
with Asn208 and three
and Asp209, and formed with Gln291,
three formed Lys173,
with Gln291, and Thr232.
Lys173, and Thr232.
HKII-compound
HKII-compound 30 has30 shown seven
has shown H bonds.
seven Three
H bonds. formed
Three withwith
formed Asp84, Asn89,
Asp84, and Thr88.
Asn89, FourFour
and Thr88. formed withwith
formed Ser415,
Ser415,
Gly87,
Gly87, and Thr232.
and Thr232. The distance
The distance between
between HKIIHKII and BG6
and BG6 (yellow),
(yellow), and HKII
and HKII and compound
and compound 30 (yellow)
30 (yellow) havehave an average
an average
of 1.05Å.
of 1.05Å.

Furthermore, compound 10 with HKII has formed an average of six hydrogen bonds,
which is higher than the reference complex HKII-GLC (4 hydrogen bonds) (Figure 10A).
Figure 10B,C have shown the extracted trajectory of HKII-GLC and HKII-compound 10 at
20 ns, where GLC binds with HKII by four H bonds with catalytic residues Asp209 and
Lys173, while compound 10 has shown six H bonds with a catalytic residue of Asn209,
Lys173, Gln291, and Asp209 (Figure 10B,C). Thus, it can be said that compound 10 has more
BioTech 2021, 10, 1 24 of 28

BioTech 2021, 2, FOR PEER REVIEW intermolecular interactions via hydrogen bonding and the compound was stably-bound25in
the catalytic region of HKII, showing a high level of inhibition compared to the reference
molecule GLC.

B C

GCL Compound 10

Figure 10. (A) The total number of hydrogen bond interactions between HKII and GLC (red), and HKII and compound
Figure 10. (A)Snapshots
10 (black). The total number of hydrogen
of complex bond interactions
(B) HKII-GLC between HKII and
and (C) HKII-compound10 GLC
were (red), and
extracted HKII
from theand compound
trajectory at 2010ns.
(black).
HKII-GLC has shown three H bonds. Two formed with Lys173 and one formed with Asp209. HKII-compoundHKII-
Snapshots of complex (B) HKII-GLC and (C) HKII-compound10 were extracted from the trajectory at 20 ns. 10 has
GLC has shown three H bonds. Two formed with Lys173 and one formed with Asp209. HKII-compound 10 has shown
shown six H bonds. Four formed with Lys173 and Asp209 and two formed with Gln291 and Asn208. The distance between
six H bonds. Four formed with Lys173 and Asp209 and two formed with Gln291 and Asn208. The distance between HKII
HKII and GLC (yellow), and HKII and compound 10 (yellow) are, on average, 2.00Å.
and GLC (yellow), and HKII and compound 10 (yellow) are, on average, 2.00Å.
The distance between HKII and GLC as well as compound 10 were, on average, 2.0Å,
The distance between HKII and GLC as well as compound 10 were, on average, 2.0Å,
which defined that their binding was strong throughout the simulation period. Several
which defined that their binding was strong throughout the simulation period. Several
studies have reported the discovery of dengue inhibitors against the viral proteins through
studies have reported the discovery of dengue inhibitors against the viral proteins
the MD simulation process. Shanmugam and Gromiha (2016) have conducted insilico
through
processesthesuch
MD assimulation
molecularprocess.
dockingShanmugam and inhibitors
to find potential Gromiha (2016)
againsthave conducted
RNA-dependent
insilico processes such as molecular docking to find potential inhibitors
RNA polymerase, which is essential for dengue viral replication. The researchers against RNA-de-ob-
pendent RNA polymerase, which is essential for dengue viral replication. The
tained quercetin derivatives, quercetin 3-(6”-(E)-pcoumaroylsophoroside)-7-rhamnoside asresearchers
obtained
a denguequercetin
polymerasederivatives,
inhibitor, quercetin
where this3-(6″-(E)-pcoumaroylsophoroside)-7-rhamno-
finding was validated based on the stability of
side as a dengue polymerase inhibitor,
the enzyme and inhibitor by an MD simulationwhere this finding
[59]. was validated
In another based on the
study, Shanmugam sta-
and col-
bility of the
leagues enzyme
also and[2-(4-carbamoylpiperidin-1-yl)-2-oxoethyl]
identified, inhibitor by an MD simulation [59]. In another study, Shanmu-
8-(1,3-benzothiazol-2-yl)
gam and colleagues also identified, [2-(4-carbamoylpiperidin-1-yl)-2-oxoethyl] 8-(1,3-ben-
zothiazol-2-yl) naphthalene-1-carboxylate as a potent dengue viral polymerase inhibitor
by maintaining the same simulation condition from the previous study [60]. Most of the
inhibitors were identified through an MD simulation against viral replication proteins,
BioTech 2021, 10, 1 25 of 28

naphthalene-1-carboxylate as a potent dengue viral polymerase inhibitor by maintain-


ing the same simulation condition from the previous study [60]. Most of the inhibitors
were identified through an MD simulation against viral replication proteins, such as non-
structural protein 3 (NS3), NS5 methyltransferase, and also envelope protein (E) [58–64].
Currently, 18 million compounds were virtually-screened against dengue replication pro-
tein NS3, where the top five compounds have shown strong predicted binding affinity for
the important catalytic residues of NS3 [65]. Eventually, the top five compounds further
went through the MD simulation to scrutinize the stability of selected compounds and NS3
protein throughout the simulation period [65]. In another study, 300 commercial cyclic
peptides were screened against NS5 methyltransferase where two ligands, namely pre-
proendothelin and urotensin II, were selected as the best inhibitors based on ligand-enzyme
binding free energy and pharmacological prediction. Furthermore, it was confirmed that
both ligands maintained a stable complex conformation with the enzymes throughout a
6-ns simulation period [64,66]. On the contrary, our findings were evaluated based on the
stability of protein-ligand throughout the simulation period, where compound 1, com-
pound 10, and compound 30, which are the analogues of 2DG, GLC, and BG6 respectively,
have shown protein-ligand stability during the course of the simulation process.

4. Conclusions
There is an urgent need to design better inhibitors against the DENV, in which human
hexokinase II (HKII) was proposed as an ideal target for anti-DENV drug development.
Precise and computationally-competent virtual screening can act as a potential step toward
the future “on-shelf” dengue virus drugs. Consequently, we have identified a number of
novel compounds by in silico screening of library of compounds from the USRCAT database
using Auto Dock Vina docking programs. Docking results of the top three compounds
for each reference molecule in our study have shown strong predicted binding affinity for
the catalytically-important residues of HKII. Finally, compound 10, compound 30, and
compound 1, which are the analogues of GLC, BG6, and 2DG, respectively, have exhibited
strong protein-ligand stability with RMSD ±5.0A◦ , ±4.0A◦ , and ±0.5A throughout the
simulation process. The effects of these compounds on the activity of HKII are currently
being pursued in the subsequent structural and biochemical studies.

Author Contributions: Conceptualization, F.A.A.F. and S.T.; methodology, F.A.A.F., S.T., A.A.A.H.;
software, S.T., A.A.A.H.; validation, F.A.A.F., S.T., A.A.A.H.; formal analysis, S.T., A.A.A.H.; investi-
gation, S.T.; resources, F.A.A.F., A.A.A.H.; data curation, S.T.; writing—original draft preparation, S.T.;
writing—review and editing, F.A.A.F., A.A.A.H.; visualization, S.T.; supervision, F.A.A.F., A.A.A.H.;
project administration, F.A.A.F.; funding acquisition, F.A.A.F. All authors have read and agreed to
the published version of the manuscript.
Funding: This research was funded by the Ministry of Higher Education Malaysia (MOHE) grant
number FRGS/1/2016/STG04/UIAM/02/1.
Data Availability Statement: The data presented in this study are available in this article “Virtual
Screening for Potential Inhibitors of Human Hexoki-nase II for the Development of Anti-Dengue
Therapeutics”.
Acknowledgments: The authors would like to thank Research unit for Bioinformatics and Computa-
tional Biology (RUBIC), Kulliyyah of Science, International Islamic University Malaysia.
Conflicts of Interest: The authors declare no conflict of interest.

References
1. Henchal, E.A.; Putnak, R.J. The dengue viruses. Am. Soc. Microbiol. 1990, 4, 376–396. [CrossRef] [PubMed]
2. Malavige, G.N.; Fernando, S.; Fernanoda, D.J.; Seneviratne, S.L. Dengue viral infections. J. Postgrad. Med. 2004, 80, 588–601.
[CrossRef] [PubMed]
3. Salles, T.S.; Sa-Guimaraes, T.E.; de Alvarenga, E.S.L.; Ribeiro, A.; de Meneses, M.D.F.; Moreire, M.F. History, epidemiology and
diagnostics of dengue in the American and Brazillian context: A review. J. BioMed Cent. Parasites Vectors 2018, 11, 264. [CrossRef]
[PubMed]
4. Gubler, D.J. Dengue and dengue hemorrhagic fever. Am. Soc. Microbiol. 1998, 11, 480–496. [CrossRef]
BioTech 2021, 10, 1 26 of 28

5. Majeed, A.I.; Avabratha, S.K.; Lokesh, R.; Syeda, S. Clinicohaematological profile of dengue in children: A hospital based study.
Int. J. Contemp. Pediatr. 2017, 4, 2349–3283. [CrossRef]
6. World Health Organization. Immunization, Vaccines and Biologicals. 2019. Available online: http://www.who.int/
immunization/diseases/dengue/en/ (accessed on 7 July 2019).
7. Pan America Health Organization. Dengue Epidemiology. 2019. Available online: https://www.paho.org/data/index.php/en/
mnu-topics/indicadores-dengue-en.html (accessed on 3 March 2019).
8. Smith, A.W.; Renhorn, E.K.; Tissera, H.; Bakar, A.S.; Alphey, L.; Kittayapong, P.; Lindsay, S.; Logan, J.; Hatz, C.; Reiter, P.; et al.
Dengue tools: Innovative tools and strategies for the surveillance and control of dengue. Glob. Health Action 2014, 4, 256–321.
9. Cheong, L.Y.; Burkart, K.; Leitao, J.P.; Lakes, T. Assessing weather effects on dengue diseases in Malaysia. Int. J. Environ. Res.
Public Health 2013, 10, 6319–6334. [CrossRef]
10. Perera, R.; Khun, J.R. Structural proteomics of Dengue virus. Natl. Inst. Health 2008, 11, 369–377. [CrossRef]
11. Wearing, H.J.; Pejman, R. Ecological and immunological determinants of dengue epidemics. Proc. Natl. Acad. Sci. USA 2006, 103,
11802–11807. [CrossRef]
12. Vinodkumar, C.S.; Kalapannavar, N.K.; Basavarajappa, K.G.; Sanjay, D.; Gowli, C.; Nadig, G.; Prasad, B.S. Episode of coexisting
infections with multiple dengue virus serotypes in central Karnataka, India. J. Infect. Public Health 2013, 6, 302–306. [CrossRef]
13. Munger, J.; Bajad, S.U.; Coller, H.A.; Shenk, T.; Rabinowitz, J.D. Dynamics of the cellular metabolome during human cy-
tomegalovirusinfection. PLoS Pathog. 2006, 2, 1165–1175. [CrossRef] [PubMed]
14. Diamond, M.S.; Pierson, T.C. Molecular insight into dengue virus pathogenesis and its implications for disease control. Cell 2015,
162, 488–492. [CrossRef] [PubMed]
15. Fontaine, K.A.; Camarda, R.; Lagunoff, M. Vaccinia virus require glutamine but glucose for efficient replication. J. Virol. 2014, 88,
4366–4374. [CrossRef] [PubMed]
16. Seyler, H.; Honegger, A.; Bossler, F.; Sponagel, J.; Bulkescher, J.; Lohrey, C. Viral E6/E7 oncogene and cellular hexokinase 2
expressions in HPV-positive cancer cell lines. J. Oncotarget 2017, 8, 6342–6351.
17. Sanchez, E.; Lagunoff, M. Viral Activation of Cellular Metabolism. J. Virol. 2015, 8, 4321–4334. [CrossRef]
18. Roberts, D.J.; Miyamoto, S. Hexokinase II integrates energy metabolism and cellular protection: Akting on mitochondria and
TORCing to autophagy. Cell Death Differ. 2015, 2, 248–257. [CrossRef]
19. Zagorodna, O.; Martin, M.S.; Rutkowski, T.; Kuwana, T.; Sprite, R.; Kundoson, M. 2-Deoxyglucose-induced toxicity is regulated
by Bcl-2 family members and is enhanced by antagonizing Bcl-2 in lymphoma cell lines. HHS Public Access 2012, 31, 2738–2749.
[CrossRef]
20. Fontaine, K.A.; Sanchez, E.L.; Camarda, R.; Lagunoff, M. Dengue virus induces and requires glycolysis for optimal replication. J.
Virol. 2015, 89, 2358–2366. [CrossRef]
21. Landini, M.P. Early enhanced glucose uptake in human cytomegalovirus-infected virus. Virology 1984, 13, 68–77.
22. Moriya, K.; Yotsuyanagi, H.; Shintani, Y.; Fujie, H.; Ishibashi, K.; Matsuura, Y.; Miyamura, T.; Koike, K. Hepatitis C virus core
protein induces hepatic steatosis in transgenic mice. Virology 1997, 78, 1527–1531. [CrossRef]
23. McArdle, J.; Schafer, X.L.; Munger, J. Inhibition of calmodulin-dependent kinase kinase blocks human cytomegalovirus-induced
glycolytic activation and severely attenuates production of viral progeny. J. Virol. 2011, 85, 705–714. [CrossRef] [PubMed]
24. McLauchlan, J. Lipid droplets and hepatitis C virus infection. Biochim. Biophys. Acta 2009, 1791, 552–559. [CrossRef] [PubMed]
25. Wick, A.N.; Durury, D.R.; Nakada, H.I.; Wolfe, J.B. Localization of primary metabolic block produced by 2-Doxygulcose. J. Biol.
Chem. 1956, 224, 963–969.
26. Godói, I.P.; Lovato, L.; Lemo, P.; Araújo, V.E.; Bonoto, B.C.; Godman, J.G. CYD-TDV dengue vaccine: Systematic review and
meta-analysis of efficacy, immunogenicity and safety. J. Comp. Eff. Res. 2008, 61, 65–80. [CrossRef]
27. Ganapathy-Kanniappan, S.; Geschwind, J.H. Tumor glycolysis as a target for cancer therapy: Progress and prospects. Mol. Cancer
2013, 12, 1–11. [CrossRef]
28. Kaushik, N.; Lee, S.J.; Choi, T.G.; Bai, K.Y.; Uhm, H.S.; Kim, C.H.; Choi, H.E. Non-thermal plasma with 2-deoxy-D-glucose
synergistically induces cell death by targeting glycolysis in blood cancer cells. Sci. Rep. 2015, 5, 8726. [CrossRef]
29. Zhang, F.; Lin, X.; Bradbury, M.; Kaushal, A.; Li, L.; Spritz, R.; Gius, D. 2-Deoxy-D-Glucose-induced Cytotoxicity and Radiosensi-
tization in Tumor Cells Is Mediated via Disruptions in Thiol Metabolism. Cancer Res. Assoc. 2003, 63, 3413–3417.
30. Vyas, V.; Jain, A.; Jain, A.V.; Gupta, A. Virtual screening: A fast tool for drug design. Sci. Pharm. 2008, 76, 333–360. [CrossRef]
31. Liu, X.; Kim, C.S.; Honzatko, B.; Fromm, H.J. Dual Mechanism for glucose 6-phosphate inhibition of human brain hexokinase. J.
Biol. Chem. 1999, 274, 31155–31159. [CrossRef]
32. Cosconati, S.; Forli, S.; Perryman, A.L.; Harris, R.; Goodsell, D.S.; Olson, A.J. Virtual screening with AutoDock: Theory and
practice. Expert Opin. Drug Discov. 2010, 5, 597–607. [CrossRef]
33. Lengauer, T.; Lemmen, C.; Rarey, M.; Zimmermann, M. Novel technologies for virtual screening. J. Natl. Libr. Med. 2004, 9, 27–34.
[CrossRef]
34. Lavecchia, A.; Giovannni, C.D. Virtual screening in drug discovery: A critical review. Curr. Med. Chem. 2013, 20, 60–71. [CrossRef]
35. Nawaz, H.M.; Ferreira, J.C.; Nedyalkova, L.; Zhu, H.; Lopez, C.C.; Kirmiziatin, S. The catalytic inactivation of the N-half of
human hexokinase 2 and structural and biochemical characterization of its mitochondrial conformation. J. Biosci. Rep. 2018, 38,
1–17. [CrossRef] [PubMed]
BioTech 2021, 10, 1 27 of 28

36. Printz, R.L.; Osawa, H.; Ardehali, H.; Koch, S.; Granner, D.K. Hexokinase II gene: Structure, regulation and promoter organization.
Biochem. Soc. Trans. 1997, 25, 107–112. [CrossRef] [PubMed]
37. Savic, L.J.; Chapiro, J.; Duwe, G.; Geschwind, J. Targeting glucose metabolism in cancer: A new class of agents for loco-regional
and systemic therapy of liver cancer and beyond. Hepatic Oncol. 2016, 3, 19–28. [CrossRef] [PubMed]
38. Lionta, E.; Spyrou, G.; Vassilatis, D.K.; Cournia, Z. Structure-based virtual screening for drug discovery: Principles applications
and recent advances. Curr. Topics Med. Chem. 2014, 14, 1923–1938. [CrossRef] [PubMed]
39. Schreyer, A.M.; Blundell, T. USRCAT: Real-time ultrafast shape recognition with pharmacophoric constraints. J. Cheminform. 2012,
4, 1–15. [CrossRef]
40. Lipinski, C.A.; Lombardo, F.; Dominy, B.W.; Feeney, P.J. Experimental and computational approaches to estimate solubility and
permeability in drug discovery and development setting. Adv. Drug Deliv. Rev. 1997, 23, 3–25. [CrossRef]
41. Tsaioun, K.; Blaaubor, J.B.; Hartung, T. Evidence-Based Absorption, Distribution, Metabolism, Excretion (ADME) and its Interplay
with Alternative Toxicity Methods. J. Pharm. Bioallied Sci. 2016, 33, 333–336. [CrossRef]
42. Mastrangelo, E.; Pezzullo, M.; De Burghgraeve, T.; Kaptein, S.; Pastorino, B.; Dallmeier, K.; Bolognesi, M. Ivermectin is a potent
inhibitor of flavivirus replication specifically targeting NS3 helicase activity: New prospects for an old drug. J. Antimicrob.
Chemother. 2012, 67, 1884–1894. [CrossRef]
43. Basavannacharya, C.; Vasudevan, S.G. Suramin inhibits helicase activity of NS3 protein of dengue virus in a fluorescence-based
high throughput assay format. Biochem. Biophys. Res. Commun. 2014, 453, 539–544. [CrossRef] [PubMed]
44. Pires, D.E.V.; Blundell, T.L.; Ascher, D.B. pkCSM: Predicting Small-Molecule Pharmacokinetic and Toxicity Properties Using
Graph-Based Signatures. J. Med. Chem. 1998, 58, 4066–4072. [CrossRef] [PubMed]
45. Banerjee, P.; Eckeert, O.A.; Schrey, K.A.; Preissner, R. ProTox-II: A webserver for the prediction of toxicity of chemicals. J. Nucleic
Acids Res. 2018, 46, 257–263. [CrossRef] [PubMed]
46. Arome, D.; Chinedu, E. The importance of toxicity testing. J. Pharm. BioSci. 2014, 41, 46–48.
47. Ruiz, P.; Begluitti, G.; Wheeler, J.; Mumtaz, M. Prediction of acute mammalian toxicity using QSAR method: A case study of
sulfur mustard and its breakdown product. J. Mol. 2012, 17, 8982–9001. [CrossRef] [PubMed]
48. Koes, R.D.; Camacho, J.C. Shape-Based Virtual Screening with Volumetric Aligned Molecular Shapes. J. Natl. Inst. Health 2014, 35,
1824–1834. [CrossRef]
49. Maia, B.H.E.; Assis, C.L.; Oliveira, T.A.; Silva, M.A.; Taranto, G.A. Structure-Based Virtual Screening: From Classical to Artificial
Intelligence. J. Med. Pharm. Chem. 2020, 8, 334–338. [CrossRef]
50. Lim, S.V.; Basyaruddin, M.; Rahman, A.; Tejo, B.A. Structure-based and ligand-based virtual screening of novel methyltransferase
inhibitors. Asia Pac. Bioinform. 2011, 12, 13–24. [CrossRef]
51. Wang, Q.Y.; Kondreddi, R.R.; Xie, X.; Rao, R.; Nilar, S.; Xu, H.Y.; Lakshminarayana, S.B. A translation inhibitor that suppresses
dengue virus in vitro and in vivo. Am. Soc. Microbiol. 2011, 55, 4072–4080.
52. Sing, J.; Kumar, M.; Mansuri, R.; Sahoo, C.G.; Deep, A. Inhibitor desingning, virtual screening and docking studies for
methyltransferase: A potential target against dengue virus. J. Pharm. Bioallied Sci. 2015, 34, 2–8.
53. Kouretova, J.; Hammamy, Z.M.; Epp, A.; Hardes, K.; Kallis, S.; Zhang, L.; Steinmetzer, T. Effects of NS2B-NS3 protease and furin
inhibition on West Nile and Dengue virus replication. J. Enzym. Inhib. Med. Chem. 2017, 32, 712–721. [CrossRef] [PubMed]
54. Halim, S.A.; Khan, H.; Khan, A.; Wadood, A.; Mabood, F.; Hussain, J.; Al-Harrasi, A. Targeting dengue virus NS-3 helicase by
ligand based pharmacophore modeling and structural based virtual screening. Front. Chem. 2017, 5, 1–6. [CrossRef] [PubMed]
55. Byrd, C.M.; Dai, D.; Grosenbach, D.W.; Berhan, A.; Jones, K.F.; Cardwell, K.B.; Jordan, R. A novel inhibitor of dengue virus
replication that targets the capsid protein. Am. Soc. Microbiol. J. 2018, 57, 15–25. [CrossRef] [PubMed]
56. Whitby, K.; Pierson, T.C.; Geiss, B.; Lane, K.; Engle, M.; Zhou, Y.; Diamond, M.S. Castanospermine a potent inhibitor of dengue
virus infection In vitro and In vivo. Am. Soc. Microbiol. 2005, 79, 8698–8706. [CrossRef]
57. Yang, C.C.; Hu, H.S.; Wu, R.H.; Wu, S.H.; Lee, S.J.; Jiaang, W.T.; Yueh, A. A novel dengue virus inhibitors BP13944, discovered by
High-throughput screening with dengue virus replicon cells selectes for resistance in the viral NS2B/NS3 protease. Am. Soc.
Microbiol. 2013, 58, 110–119.
58. Ismail, A.N.; Jusoh, A.S. Molecular Docking and Molecular Dynamics Simulation Studies to Predict Flavonoid Binding on the
Surface of DENV2 E Protein. J. Comput. Life Sci. 2017, 9, 499–511. [CrossRef]
59. Shanmugam, A.; Velmurugan, D.; Gromiha, M. Identification of Dengue Viral RNA Dependent RNA Polymerase Inhibitor using
Computational Fragment Based Approaches and Molecular Dynamics Study. J. Biomol. Struct. Dyn. 2015, 2, 1–10.
60. Shanmugam, A.; Gromiha, M.M. Quercetin derivatives as non-nucleoside inhibitors for dengue polymerase: Molecular docking,
molecular dynamics simulation and binding free energy calculation. J. Biomol. Struct. Dyn. 2016, 5, 2895–2909.
61. Tambunan, S.U.; Zahroar, H.; Utomo, B.B.; Parikesit, A.A. Screening of commercial cyclic peptide as inhibitor NS5 methyl-
transferase of Dengue virus through Molecular Docking and Molecular Dynamics Simulation. J. Bioinform. 2014, 10, 23–27.
[CrossRef]
62. De Almeida, H.; Bastos, M.D.I.; Ribeiro, M.B.; Maigret, B.; Santana, M.J. New Binding Site Conformations of the Dengue Virus
NS3 Protease. Accessed by Molecular Dynamics Simulation. PLoS ONE 2013, 8, e72402. [CrossRef]
63. Tambunan, S.U.; Nasution, F.A.; Azhima, F.; Parikesti, A.A.; Toepak, P.E.; Idrus, S. Modification of S-Adenosyl-l-Homocysteine as
Inhibitor of Nonstructural Protein 5 Methyltransferase Dengue Virus Through Molecular Docking and Molecular Dynamics
Simulation. J. Bioinform. 2017, 11, 1–14. [CrossRef] [PubMed]
BioTech 2021, 10, 1 28 of 28

64. Behnam, M.A.M.; Nitsche, C.; Vechi, M.S.; Klein, D.C. C-terminal residue optimization and fragment merging discovery of a
potent peptide-hybrid inhibitior of dengue protease. Am. Chem. Soc. 2014, 5, 1037–1042.
65. Mirza, S.B.; Salmas, R.K.; Fatmi, M.Q.; Durdagi, S. Virtual screening of eighteen million compounds against dengue virus:
Combined molecular docking and molecular dynamics simulation study. J. Mol. Graph. Model. 2016, 66, 99–107. [CrossRef]
[PubMed]
66. Luzhkov, V.; Decroly, E.; Canarda, B.; Selisko, B.; Aqvist, J. Evaluation of Adamantane Derivatives as Inhibitors of Dengue Virus
mRNA Cap Methyltransferase by Dockingand Molecular Dynamics Simulations. J. Mol. Inform. 2013, 32, 155–164. [CrossRef]
[PubMed]

You might also like