You are on page 1of 15

Journal of Taibah University for Science

ISSN: (Print) (Online) Journal homepage: https://www.tandfonline.com/loi/tusc20

In-silico natural product database mining


for novel neuropilin-1 inhibitors: molecular
docking, molecular dynamics and binding energy
computations

Mahmoud A. A. Ibrahim, Sara S. M. Ali, Khlood A. A. Abdeljawaad, Alaa H. M.


Abdelrahman, Gamal A. Gabr, Ahmed M. Shawky, Gamal A. H. Mekhemer,
Peter A. Sidhom, Paul W. Paré & Mohamed-Elamir F. Hegazy

To cite this article: Mahmoud A. A. Ibrahim, Sara S. M. Ali, Khlood A. A. Abdeljawaad, Alaa
H. M. Abdelrahman, Gamal A. Gabr, Ahmed M. Shawky, Gamal A. H. Mekhemer, Peter A.
Sidhom, Paul W. Paré & Mohamed-Elamir F. Hegazy (2023) In-silico natural product database
mining for novel neuropilin-1 inhibitors: molecular docking, molecular dynamics and
binding energy computations, Journal of Taibah University for Science, 17:1, 2182623, DOI:
10.1080/16583655.2023.2182623

To link to this article: https://doi.org/10.1080/16583655.2023.2182623

© 2023 The Author(s). Published by Informa View supplementary material


UK Limited, trading as Taylor & Francis
Group

Published online: 27 Feb 2023. Submit your article to this journal

Article views: 852 View related articles

View Crossmark data

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


https://www.tandfonline.com/action/journalInformation?journalCode=tusc20
JOURNAL OF TAIBAH UNIVERSITY FOR SCIENCE
2023, VOL. 17, NO. 1, 2182623
https://doi.org/10.1080/16583655.2023.2182623

In-silico natural product database mining for novel neuropilin-1 inhibitors:


molecular docking, molecular dynamics and binding energy computations
Mahmoud A. A. Ibrahim a,b , Sara S. M. Alia , Khlood A. A. Abdeljawaada , Alaa H. M. Abdelrahmana , Gamal A.
Gabrc,d , Ahmed M. Shawkye , Gamal A. H. Mekhemera , Peter A. Sidhomf , Paul W. Parég and
Mohamed-Elamir F. Hegazyh
a Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia, Egypt; b School of Health
Sciences, University of KwaZulu-Natal, Durban, South Africa; c Department of Pharmacology and Toxicology, College of Pharmacy, Prince
Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia; d Agricultural Genetic Engineering Research Institute (AGERI), Agricultural Research
Center, Giza, Egypt; e Science and Technology Unit (STU), Umm Al-Qura University, Makkah, Saudi Arabia; f Department of Pharmaceutical
Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt; g Department of Chemistry & Biochemistry, Texas Tech University, Lubbock,
TX, USA; h Chemistry of Medicinal Plants Department, National Research Centre, Giza, Egypt

ABSTRACT ARTICLE HISTORY


In the search for new metabolite inhibitors, a natural product activity and species source Received 18 November 2022
(NPASS) database was virtually screened using AutoDock software to identify potential NRP1 Revised 14 February 2023
inhibitors. NPASS compounds complexed with NRP1 were subjected to molecular dynamics (MD) Accepted 16 February 2023
simulations. Furthermore, NPASS-NRP1 binding affinities were calculated using the MM-GBSA KEYWORDS
approach. Based on calculated binding energies, kamolonol (NPC146388) demonstrated lower Cancer; neuropilin-1 (NRP1);
NRP1 binding affinity than the co-crystallized HRG/Arg-1 ligand with binding energy (Gbinding ) docking calculations; MD
values of –34.5 and –32.0 kcal/mol, respectively. Structural and energetic analysis showed high simulations;
stability for kamolonol and HRG/Arg-1 with NRP1 over the 200 ns MD simulations. The studied pharmacokinetic study
physicochemical properties of kamolonol and HRG/Arg-1 revealed that these compounds obey
Lipinski’s rule of five. ADMET characteristics of kamolonol and HRG/Arg-1 were predicted, and
kamolonol showed better ADMET properties compared to HRG/Arg-1. Based on these results,
kamolonol is a promising NRP1 inhibitor worthy of further experimental assays as anti-carcinoma
remediation.

1. Introduction
for tumour development, proliferation, invasion, metas-
Biological alteration that results in unregulated cell tases, and prognosis [12–16]. NRP1 enhances angio-
growth [1] is referred to as cancer and is a world- genesis by forming a direct complex with endothelial
wide health challenge with poor clinical outcomes growth factors A and 2 (VEGFA and VEGFR2) [17]. It
[2,3]. Approximately 10 million deaths were reported also promotes the tumour’s development after binding
in 2020 by global cancer statistics [4]. Despite ongo- with the VEGFA due to its ability to promote RhoA acti-
ing progress in surgery and radiotherapy, patients with vation [18]. Besides, NRP1 has the ability to accelerate
advanced tumours still have poor prognoses [5]. More- neoplasm progression by steadying the role of regula-
over, there is only a small number of newly approved tory T cells (Tregs) and banning the tumour-associated
cancer medications released annually by the US Food macrophages (TAM) from penetrating normoxic cancer
and Drug Administration (FDA) [6]. With resistance to areas [19]. Although NRP1 protein has received consid-
therapeutic drugs a substantial problem in treating erable critical attention as a potential therapeutic target
cancer [7], there is a critical need for novel cancer for cancer therapy [20,21], no drug has been approved
therapies. as an NRP1 inhibitor.
Numerous cancers, including gastric, urinary, blad- Natural products (NPs) retrieved from microbes and
der, breast, lung, and esophageal, have high levels of plants have a vital role in drug discovery, particularly
neuropilin-1 (NRP1) protein [8–11]. NRP1 is a cell surface against carcinoma and contagious illnesses [22,23]. Sev-
receptor that binds vascular endothelial growth factor eral drugs are natural products from plant sources, such
(VEGF), a potent mediator of endothelial permeabil- as paclitaxel, morphine, and vinblastine [24]. There-
ity, chemotaxis, and proliferation. It is also a regulator fore, there is abundant opportunity for mining novel

CONTACT Mahmoud A. A. Ibrahim m.ibrahim@compchem.net Computational Chemistry Laboratory, Chemistry Department, Faculty of
Science, Minia University, Minia 61519, Egypt; School of Health Sciences, University of KwaZulu-Natal, Westville, Durban 4000, South Africa; Mohamed-Elamir
F. Hegazy elamir77@live.com Chemistry of Medicinal Plants Department, National Research Centre, 33 El-Bohouth St., Giza 12622, Egypt
Supplemental data for this article can be accessed here. https://doi.org/10.1080/16583655.2023.2182623
© 2023 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted
use, distribution, and reproduction in any medium, provided the original work is properly cited.
2 M. A. A. IBRAHIM ET AL.

anticancer drug candidates as NRP1 inhibitors from the for the docking calculations are described elsewhere
Natural Product Activity and Species Source (NPASS) [37–39]. Except the exhaustiveness parameter, which
database. In the current study, NPASS database was was set to 50 and 200 in fast and expensive calculations,
screened for promising NRP1 inhibitors utilizing molec- respectively, the remaining parameters were preserved
ular docking computations [25]. Subsequently, promis- at the default settings. The grid box was tailored to fit
ing NPASS compounds were selected and subjected the active site of NRP1 protein by taking the centroid
to molecular dynamics (MD) simulations and the cor- of HRG/Arg-1 as a centre of the grid, with a grid size of
responding binding energies were estimated using 15 Å × 15 Å × 15 Å and a value of spacing equal to 1 Å.
the MM-GBSA approach. The stability and tightness The grid was centred at 32.485, –11.49, and 27.507 (in
of the most potent NPASS compounds within NRP1 x, y, and z dimensions, respectively). The top nine scor-
were selected. Thus, the current study highlights the ing poses were estimated via visual investigation and
potentiality of NPASS candidates as NRP1 inhibitors the lowest docking score was selected as a representive
and illustrates a promising drug candidate for clinical mode.
consideration.

2.4. Molecular dynamics simulations


2. Computational methodology
The most effective NPASS compounds complexed with
2.1. NRP1 preparation the NRP1 protein underwent molecular dynamics (MD)
The neuropilin-1 b1 domain (NRP1) atomic coordinates simulations using AMBER16 software [40]. The details
in complex with L-homoarginine (HRG/Arg-1) were of MD simulations are described elsewhere [41–45].
retrieved from the protein databank (PDB code: 5ijr; Briefly, the inspected NPASS compounds were defined
resolution: 1.52 Å [26]) and considered as a template utilizing the general AMBER force field (GAFF2) [46],
for all in-silico computations. The crystallographic water while the NRP1 protein was characterized using the
molecules, ions, and ligand were eliminated from the AMBER force field of 14SB [47].
NRP1 protein, keeping only the amino acids. The proto- MD simulations were conducted employing both
nation state of the amino acids was investigated using implicit and explicit water solvents. In the implicit water
the H++ server, and all missing hydrogen atoms were solvent MD simulations, the atomic partial charges of
inserted [27]. the inspected NPASS compounds were estimated using
an AM1-BCC method with the aid of the Antecham-
ber tool within the AMBER16 [48]. Periodic boundary
2.2. Database preparation
conditions were not utilized, and the cutoff value was
The Natural Product Activity and Species Source set to 999 Å. Additionally, the solvent model (igb = 1)
(NPASS) database, containing more than 35,000 natural was used to present the solvation contributions [49].
compounds, was downloaded in SDF format [25]. The All the investigated systems were first minimized for
duplicated compounds were eliminated based on the 500 steps. The minimized complexes were warmed pro-
International Chemical Identifier (InChIKey) [28]. The gressively from 0 to 300 K over 10 ps. Thereafter, the
three-dimensional (3D) structures of the NPASS com- NPASS-NRP1 complexes were subjected to an equilib-
pounds were generated using Omega2 software [29,30] rium stage of 50 ps, followed by a production stage
and minimized utilizing the MMFF94S force field inte- of 1 ns.
grated inside SZYBKI software [31,32]. The protona- In explicit water solvent MD simulations, the
tion state of the NPASS compounds was examined restricted electrostatic potential (RESP) approach was
using fixpka algorithm within the QUACPAC software, employed to determine the partial charge of each
respectively [33]. The Gasteiger-Marsili method was atom of the inspected NPASS compounds at the HF/6-
used for the determination of the partial charges of 31G∗ level of theory [50]. Quantum mechanics cal-
NPASS compounds [34]. The prepared files are present culations were performed using Gaussian09 software
in a CompChem database (www.compchem.net/ccdb) [51]. TIP3P water molecules were utilized to solvate
that is publically accessible. The schematic diagram of NPASS-NRP1 complexes in an octahedron box with
the utilized computational approaches is depicted in a minimal distance of 12 Å [52]. All investigated sys-
Figure 1. tems were minimized by employing 5000 cycles before
heating from 0 to 300 K over 50 ps under the NVT
ensemble. NPASS-NRP1 complexes were then equili-
2.3. Molecular docking
brated over 1 ns under the NPT ensemble. The equili-
AutoDock Vina1.1.2 software was used for executing brated coordinates of the systems were subjected to
molecular docking calculations [35]. The NRP1 protein production runs of 10 and 200 ns. Atomic coordinates
was converted into pdbqt format using MGL tools (ver- were recorded every 10 ps for post-dynamics analyses
sion 1.5.7) [36]. The details of the employed protocol and binding free energy estimations. The Particle Mesh
JOURNAL OF TAIBAH UNIVERSITY FOR SCIENCE 3

Figure 1. The schematic diagram of the utilized computational approaches in the virtual screening process of the Natural Product
Activity and Species Source (NPASS) database.

Ewald (PME) approach was applied to treat long-range 2.5. MM-GBSA binding energy
electrostatic interactions [53]. Berendsen barostat with
Binding energies of inspected NPASS compounds in
a 2 ps relaxation duration was employed to adjust the
complex with NRP1 were calculated using a molecular
atmospheric pressure [54]. To keep temperature steady
mechanics-generalized Born surface area (MM-GBSA)
at 298 K, Langevin dynamics with gamma_ln set to 1.0
approach [58]. An updated generalized born model
for collision frequency was applied [55]. The SHAKE
including Onufriev (igb = 2) was used for computing
algorithm was applied to constrain all bonds involving
the polar solvation energy [59]. The MM-GBSA binding
hydrogen bonds at the time step of 2 fs [56]. The GPU
energies were computed based on statistically inde-
version of pmemd (pmemd.cuda) was applied to per-
pendent snapshots gathered from MD simulations in
form MD simulations. The molecular visualizations were
accordance with the following equation:
generated with the help of BIOVIA Visual Studio2020
[57]. Gbinding = Gcomplex − (GNPASS + GNRP1 )
4 M. A. A. IBRAHIM ET AL.

The energy term (G) was determined as: 3.2. Virtual screening of the NPASS database
Molecular docking calculations were executed via two
G = GGB + GSA + Eele + Evdw
stages of accuracy that were named fast and expensive
Through employing generalized born (GB) models, the docking stages (see computational methodology
electrostatic solvation energy (GGB ) in the abovemen- section for details). The NPASS database, containing
tioned equation was determined with GSA standing for greater than 35,000 compounds, was virtually screened
nonpolar solvation-free energy. E ele refers to electro- using fast docking computations. The docking scores
static energy. E vdw is van der Waals energy. The entropic of NPASS compounds were predicted and compared
contributions are often disregarded because of high to that of the co-crystallized HRG/Arg-1 ligand (calc.
computational costs [60,61]. –5.6 kcal/mol). NPASS compounds with docking scores
< −7.0 kcal/mol were selected and subjected to expen-
sive docking computations, giving a total number
2.6. Drug-likeness characteristics of 1,332 compounds. Due to a large number of
investigated compounds, a threshold value of –7.0
The physicochemical characteristics of the identified
kcal/mol was selected to shortlist the prospective NRP1
compound were estimated utilizing a SwissADME
inhibitors. Table S1 provides the evaluated fast and
online website (http://www.swissadme.ch). The esti-
expensive docking scores for the top 1,332 NPASS com-
mated characteristics included topological polar sur-
pounds against the NRP1 protein. Figure S1 depicts 3D
face area (TPSA), relative molecular weight (MW), num-
and 2D molecular interactions of the top four potent
ber of hydrogen bond donors (nOHNH), number of
NPASS compounds with essential residues within the
hydrogen bond acceptors (nON), and the partition coef-
active site of the NRP1 protein. Table 1 summarizes the
ficient (MlogP) [62].
obtained docking scores, 2D chemical structures, and
binding features of the top four potent NPASS com-
2.7. ADMET characteristics pounds within the active site of NRP1.
Notably, those four NPASS compounds were filtered
ADMET properties of the identified compounds were
according to further binding energy calculations over
evaluated by an online pkCSM web server [63]. The
10 ns MD simulations. As illustrated in Table 1 and
absorption (A) was estimated by human intestinal
Figure S1, the four NPASS compounds and HRG/Arg-1
absorption (HIA) and Caco-2 permeability. Distribu-
demonstrated approximately the same binding modes
tion (D) was predicted based on blood–brain bar-
within the NRP1 active site. The most favorable inter-
rier (BBB) permeability and steady-state distribution
actions between the identified NPASS compounds and
volume (VDss). The metabolism (M) was detected
NRP1 active site were conventional hydrogen bonds
using CYP3A4 inhibitor/substrate. Excretion (E) is spec-
and π -based interactions.
ified according to the total clearance of the drug.
Kamolonol (NPC146388), isolated from F. assafoetida
Finally, the toxicity (T) was determined based on
gum resin [64], belongs to the class of organic com-
hepatotoxicity.
pounds known as coumarins. Kamolonol possesses
antiplasmodial characteristics, anti-cellular hypertrophic,
3. Results and discussion and anti-fibrotic effects [65,66]. Besides, kamolonol
demonstrated a promising docking score towards
3.1. In-silico protocol assessment
NRP1 protein with a value of –9.3 kcal/mol (Table
The performance of AutoDock Vina1.1.2 software in 1). The potency of kamolonol as an NRP1 inhibitor
predicting the correct ligand-NRP1 binding pose was can be traced to its ability to exhibit multiple hydro-
assessed. For assessment purposes, the co-crystallized gen bonds with the key amino acids of the NRP1
HRG/Arg-1 inhibitor was re-docked in NRP1 binding active site (Figure 2). Inspecting the binding mode
site, and the predicted docking pose was compared of kamolonol revealed that the hydroxyl (OH) group
to the experimental binding mode (PDB code: 5ijr of 2-methylcyclohexan-1-ol ring formed a hydrogen
[26]) (Figure 2). As depicted in Figure 2, the dock- bond with the carbonyl (C = O) group of ASP320
ing pose was similar to the native structure of the amino acid with a bond length of 2.68 Å (Figure 2).
co-crystallized HRG/Arg-1 ligand with an RMSD value Besides, the carbonyl (C = O) group of the methylhex-
of 0.23 Å. AutoDock Vina results showed a docking anone ring exhibited three hydrogen bonds with the
score of –5.6 kcal/mol, forming six significant hydro- NH group of ASN313 and GLU348 and NH2 group of
gen bonds with TRP301 (2.01 Å), ASP320 (1.65, 3.11 Å), LYS347 with bond lengths of 2.25, 2.41, and 3.01 Å,
SER346 (2.60 Å), GLU348 (2.44 Å), and ILE415 (2.79 Å). respectively (Figure 2). In addition, kamolonol demon-
Based on these results, AutoDock Vina software was uti- strated π -donor hydrogen bond with TRP301 residue
lized to screen the NPASS database for potential NRP1 and π -alkyl interactions with TYR297 and TYR353
inhibitors. residues.
JOURNAL OF TAIBAH UNIVERSITY FOR SCIENCE 5

Figure 2. (a) (i) Superimposed structures of the co-crystalized mode (in grey) and the predicted docking pose (in green) and (ii)
2D molecular interaction of L-homoarginine (HRG/Arg-1). (b) (i) 3D and (ii) 2D representations of the predicted binding mode for
kamolonol (NPC146388) within the NRP1 active site. The predicted docking score in kcal/mol is also listed.
6 M. A. A. IBRAHIM ET AL.

Table 1. Computed fast and expensive docking scores (in kcal/mol), 2D chemical structures, and binding features for the top four
potent NPASS compounds against the NRP1 protein.a
Docking Score (kcal/mol)

No. NPASS Code 2D Chemical Structure Fast Expensive Binding Featuresb


HRG/Arg-1c –5.6 –5.7 TRP301 (2.01 Å),
ASP320 (1.65, 3.11 Å),
SER346 (2.60 Å),
GLU348 (2.44 Å),
ILE415 (2.79 Å)

1 NPC146388 (Kamolonol) –9.2 –9.3 ASN313 (2.25 Å),


ASP320 (2.68 Å),
LYS347 (3.01 Å),
GLU348 (2.41 Å)

2 NPC108434 –8.8 –8.9 ASN313 (2.66, 2.15 Å),


GLY318 (2.61 Å)

3 NPC75315 (Stelletin A) –8.8 –8.7 SER298 (2.13 Å),


ASN300 (2.26 Å),
TRP301 (2.28 Å)

4 NPC19013 (Sculponin D) –8.7 –8.7 ASP320 (2.45 Å),


TYR353 (2.36 Å)

a Data ranked based on the expensive docking scores.


b Only hydrogen bonds are listed in Å.
c Reference ligand.

3.3. Molecular dynamics simulations corresponding MM-GBSA binding energies (Gbinding )


were estimated and listed in Table S2. As data enrolled
Molecular dynamics (MD) simulations of the target-
in Table S2, four NPASS compounds demonstrated
inhibitor complex are essential for inspecting confor-
greater binding affinity compared to the co-crystallized
mational steadiness and changes, internal motions,
HRG/Arg-1 ligand (calc. –28.7 kcal/mol) with binding
and the reliability of target-inhibitor binding affinities
energies with values in the range of –21.5 to –29.8
[67,68]. Consequently, MD simulations were employed
kcal/mol. Those four potent NPASS compounds in com-
herein to investigate the stability and binding affin-
plex with NRP1 were subjected to explicit water solvent
ity of the most potent NPASS compounds complexed
MD simulations for 10 ns for more reliable binding affini-
with NRP1 protein. Considering computational cost and
ties. The corresponding binding energies (Gbinding )
time, the top 5% promising NPASS compounds (i.e.
were calculated and illustrated in Figure 3.
67 compounds with expensive docking scores < –8.1
Interestingly, only kamolonol exposed binding
kcal/mol) in complex with NRP1 were subjected to
energy lower than that of the HRG/Arg-1 (calc. –29.8
implicit water solvent MD simulations. Additionally, the
kcal/mol) over the MD course of 10 ns. A MD simulation
JOURNAL OF TAIBAH UNIVERSITY FOR SCIENCE 7

Figure 3. Estimated MM-GBSA binding energies for the four promising NPASS compounds against the NRP1 active site over 1 ns in
implicit water solvent MD simulations and 10 and 200 ns in explicit water solvent MD simulations.

for kamolonol was also prolonged to 200 ns, and the The electrostatic forces (E ele ) were also adequate
corresponding binding energy (Gbinding ) was calcu- for kamolonol and HRG/Arg-1 complexed with NRP1,
lated (Figure 3). No discernible variation in the predicted with average values of –21.2 and –19.6 kcal/mol,
binding energies of kamolonol within the NRP1 active respectively (Figure 4). The abovementioned findings
site over the different simulated times of 10 and 200 ns give statistical information on the binding energies of
MD simulations. Compared to HRG/Arg-1, kamolonol kamolonol as a putative NRP1 inhibitor.
complexed with NRP1 demonstrated lower binding To examine kamolonol-NRP1 interactions and the
energy over 200 ns MD simulation with Gbinding val- contribution of essential residues inside the active
ues equal to –34.5 and –32.0 kcal/mol, respectively site of NRP1, estimated Gbinding values were divided
(Figure 3). into individual amino acid participations (Figure 5a).
Inherent driving forces in the binding of kamolonol Particularly, amino acids with energy contributions
and the HRG/Arg-1 were analyzed utilizing MM-GBSA less than –0.5 kcal/mol were depicted in Figure 5a.
binding energy decomposition (Figure 4). Remarkably, Per-residue energy decomposition analysis demon-
the van der Waals forces (E vdw ) demonstrated a signif- strated that TRP301, SER346, ILE415, and ASP320 inter-
icant contribution to the binding energy of kamolonol- acted with kamolonol and HRG/Arg-1 (Figure 5a). For
and HRG/Arg-1-NRP1 complexes, with values of –38.3 instance, ASP320 amino acid residue participated in the
and –32.5 kcal/mol, respectively (Figure 4). total binding energy (Gbinding ) with values of –2.6

Figure 4. MM-GBSA binding energy components for kamolonol (NPC146388) and the co-crystallized HRG/Arg-1 ligand with the
NRP1 protein over the 200 ns MD simulations.
8 M. A. A. IBRAHIM ET AL.

Figure 5. (a) The energy contribution of the most essential amino acids to the total MM-GBSA binding energy and (b) 2D represen-
tations of the anticipated binding modes for (i) kamolonol (NPC146388) and (ii) HRG/Arg-1 with the NRP1 active site in accordance
with the average structure of the 200 ns MD simulations.

and –2.1 kcal/mol, for kamolonol- and HRG-NRP1 com- reliable results. Among these interactions are hydrogen
plexes, respectively (Figure 5a). The average structures bonds with ASP320, ILE415 and GLU348 (Figure 5b).
for kamolonol and HRG/Arg-1 inside the NRP1 active
site over the 200 ns MD simulations are also displayed
3.4. Post-dynamics analyses
in Figure 5b. Kamolonol and HRG/Arg-1 formed four and
two hydrogen bonds, respectively, with the key residues 3.4.1. Binding energy per-frame
of the active site of NRP1 protein throughout the 200 ns To investigate the stability of kamolonol within the
MD simulations. NRP1 active site, the correlation between the binding
Inspecting the average structure for kamolonol- energy and simulation time was inspected (Figure 6).
NRP1 complex showed that kamolonol demonstrated As illustrated in Figure 6, kamolonol and HRG/Arg-1
essential hydrogen bonds with key amino acid residues were generally stable from 32 ns until the termination
over the MD simulation. Notably, these interactions of the simulation, demonstrating binding energy equal
were absent in the initial predicted binding modes to –34.5 and –32.0 kcal/mol, respectively. These study
from docking computations. This demonstrates the results elucidated the steadiness of the investigated
importance of molecular dynamics simulations toward complexes throughout the 200 ns MD simulations.
JOURNAL OF TAIBAH UNIVERSITY FOR SCIENCE 9

Figure 6. The estimated binding energies of kamolonol (NPC146388) (in red) and HRG/Arg-1 (in black) within the NRP1 protein
during the 200 ns MD simulations.

3.4.2. Hydrogen bond analysis HRG/Arg-1 are tightly bonded and do not disturb the
The hydrogen bonding of the ligands with the NRP1 overall structural constancy of NRP1 besides keeping
protein was estimated throughout the MD course of structural integrity.
200 ns. The correlation between the number of hydro-
gen bonds and simulation time was inspected and pre-
sented in Figure S2. The most striking aspect of Figure 3.5. Drug-likeness characteristics
S2 is that the average number of hydrogen bonds was The SwissADME website was utilized to predict the
3 and 2 for the kamolonol and HRG/Arg-1. The current drug-likeness characteristics of kamolonol compared
findings clearly supported the steady for the kamolonol to HRG/Arg-1. For kamolonol the MLogP value was
and HRG/Arg-1. found to be less than five (calc. 3.0), indicating that
this compound has high lipophilicity. The molecular
3.4.3. Centre-of-mass distance weight of kamolonol was found to be less than 500
Centre-of-mass (CoM) distance was used to inspect (calc. 398.49 g/mol). Additionally, the number of hydro-
the steadiness of the kamolonol and HRG/Arg-1 within gen bond acceptors (nON) was 5, and the number
NRP1. The CoM distance was measured between the of hydrogen bond donors (nOHNH) was 1 (Figure 9).
kamolonol and HRG/Arg-1 and the ASP320 residue The TPSA of the kamolonol was 76.74, which indi-
over the 200 ns MD simulations (Figure 7). The CoM cates that kamolonol has good bioavailability (Figure
distance was steady for kamolonol and HRG/Arg-1 9). Finally, the calculated %ABS was 82.5%, explaining
complexed with NRP1 at average distances of 10.7 that kamolonol has good cell membrane permeability
and 8.6 Å, respectively. The presented results demon- and oral bioavailability. Comparing the drug-likeness
strated the high stability of kamolonol and HRG/Arg-1 characteristics of kamolonol with those of HRG/Arg-1
with NRP1. demonstrated that kamolonol has better physiochem-
ical properties and oral bioavailability than HRG/Arg-1.
3.4.4. Root-mean-square deviation
To examine the structural changes of NRP1 after com-
3.6. ADMET characteristics
plexation with kamolonol and HRG/Arg-1, the root-
mean-square deviation (RMSD) analysis was measured The pharmacokinetic properties of kamolonol were
(Figure 9). The average RMSD values were 0.13 and predicted and compared to HRG/Arg-1 utilizing the
0.10 nm for the kamolonol- and HRG/Arg-1-NRP1 com- pkCSM online server. The pharmacokinetic proper-
plexes, respectively, over the 200 ns MD simulations ties included absorption, in which kamolonol demon-
(Figure 8). The RMSD analysis showed that the two com- strated high Caco2 permeability (calc. 0.839) and high
plexes preserved their stability until the end of the sim- human intestinal absorption (HIA) (calc. 98.3), suggest-
ulations. These results confirmed that kamolonol and ing that kamolonol would be highly absorbed (Table 2).
10 M. A. A. IBRAHIM ET AL.

Figure 7. CoM distance (in Å) of kamolonol (NPC146388) (in red) and HRG/Arg-1 (in black), and ASP320 of NRP1 protein active site
over the 200 ns MD simulations.

Figure 8. RMSD of the backbone atoms from the first frame of kamolonol (NPC146388) (in red) and HRG/Arg-1 (in black) with NRP1
protein during the simulation time of 200 ns.

In contrast to HRG/Arg-1, HRG/Arg-1 manifested poor and HRG/Arg-1 were not toxic (Table 2). The most
Caco2 permeability and HIA with values of –0.351 and striking observation to emerge from the data compar-
24.6, respectively (Table 2). To examine the drug dis- ison was the superior ADMET features for kamolonol
tribution, the VDss value was expected with values of over HRG/Arg-1. These findings confirmed that the
0.351 and –0.438 for kamolonol and HRG/Arg-1, respec- kamolonol could be used as a promising anticancer
tively (Table 2). The metabolism results showed that drug candidate.
kamolonol could work as an inhibitor for the CYP3A4
enzyme; however, HRG/Arg-1 was not an inhibitor for
the CYP3A4 enzyme (Table 2). The excretion was pre- 3.7. Kamolonol vs. EG00229
dicted based on the total clearance with values of 0.433 EG00229, (S)−2-(3-(benzo[c][1,2,5]thiadiazole-4-sulfon
and 0.589 for kamolonol and HRG/Arg-1, respectively amido)thiophene-2-carboxamido)−5-guanidinopenta
(Table 2). According to hepatotoxicity, both kamolonol noic acid, is an experimental inhibitor of NRP1 with
JOURNAL OF TAIBAH UNIVERSITY FOR SCIENCE 11

Figure 9. Predicted physiochemical characteristics of kamolonol (NPC146388) and HRG/Arg-1 as NRP1 inhibitors.

Table 2. The expected ADMET properties for the kamolonol (NPC146388) and HRG/Arg-1.
Absorption (A) Distribution (D) Metabolism (M) Excretion (E) Toxicity (T)
Caco2 Permeability Human Intestinal CYP3A4
NPASS Code (cm/s) Absorption (HIA) VDss (human) Inhibitor/Substrate Total Clearance Hepatotoxicity
Kamolonol (NPC146388) 0.839 98.3 0.351 Yes 0.433 No
HRG/Arg-1 –0.351 24.6 –0.438 No 0.589 No

IC50 value of 3 μM [69]. To estimate the prospectivity complexed with NRP1 (Table 3 and Figure S3). As
of the identified NPASS compound, the binding score depicted in Figure S3, the docking pose of EG00229
and mode of EG00229 towards NRP1 protein were eval- within the binding pocket of NRP1 was similar to the
uated and compared to kamolonol (NPC146388). Fur- binding mode of kamolonol, exhibiting seven hydrogen
thermore, MD simulations over 200 ns pursued by bind- bonds with ASP320 (2.54, 2.68 Å), SER346 (2.78, 2.82 Å),
ing affinity estimations were performed for EG00229 THR349 (1.91 Å), and ILE415 (2.96, 3.01 Å). Compared to

Table 3. Predicted docking score (in kcal/mol) and MM/GBSA binding energies during the 200 ns MD course for kamolonol and
EG00229 complexed with NRP1 protein.
Calculated MM/GBSA binding energy (kcal/mol)
NPASS Code/Name Docking Score (kcal/mol) E vdw E ele E GB E SUR Ggas GSolv Gbinding
Kamolonol (NPC146388) –9.3 –38.3 –21.2 29.7 –4.8 –59.5 25.0 –34.5
EG00229 –6.1 –30.0 –18.8 23.3 –3.1 –48.9 20.2 –28.7
12 M. A. A. IBRAHIM ET AL.

kamolonol (docking score = −9.3 kcal/mol), EG00229 Data availability statement


demonstrated a good docking score against NRP1 with The data that support the findings of this study are available
a value of −6.1 kcal/mol (Table 3). in the Supporting Information Material of this paper.
To gain more reliable results, EG00229 in complex
with NRP1 was subjected to 200 ns MD simulations, and ORCID
the corresponding MM-GBSA binding affinity was com- Mahmoud A. A. Ibrahim http://orcid.org/0000-0003-4819-
puted (Table 3). From Table 3, the average MM-GBSA 2040
binding energy (Gbinding ) for EG00229 in complex with
NRP1 was –28.7 kcal/mol, compared to –34.5 kcal/mol References
for the kamolonol-NRP1 complex. Similar to kamolonol,
[1] Szent-Gyoergyi A. Cell-division and cancer. Science.
the binding affinity of EG00229 complexed with NRP1 1965;149:34–37. doi:10.1126/science.149.3679.34.
protein was predominated by van der Waals (E vdw ) with [2] Ma X, Yu H. Global burden of cancer. Yale J Biol Med.
an average value of –30.0 kcal/mol (Table 3). 2006;79:85–94.
[3] Mortality GBD. Causes of death, C. global, regional,
and national life expectancy, all-cause mortality, and
4. Conclusion cause-specific mortality for 249 causes of death, 1980-
2015: a systematic analysis for the Global Burden
In the current study, the NPASS database contain- of Disease Study 2015. Lancet. 2016;388:1459–1544.
ing more than 35,000 natural products was virtu- doi:10.1016/S0140-6736(16)31012-1.
ally screened against the NRP1 protein for identify- [4] Sung H, Ferlay J, Siegel RL, et al. Global cancer statistics
2020: GLOBOCAN estimates of incidence and mortality
ing potent inhibitors as anticancer drug candidates worldwide for 36 cancers in 185 countries. CA: Cancer J
using the molecular docking technique. Based on Clin. 2021;71:209–249. doi:10.3322/caac.21660.
docking computations, 67 compounds with expen- [5] Pecorelli S, Benedet JL, Creasman WT, et al. FIGO
sive docking scores < –8.1 kcal/mol in complex with staging of gynecologic cancer. Int J Gynaecol Obstet.
NRP1 were subjected to MD simulations. The com- 1999;64:5–10. doi:10.1016/s0020-7292(98)00234-3.
[6] Benjamin DJ, Xu A, Lythgoe MP, et al. Cancer drug
puted MM-GBSA binding energy over an MD course
approvals that displaced existing standard-of-care ther-
of 200 ns demonstrated superior binding affinity of apies, 2016-2021. JAMA Netw Open. 2022;5:e222265.
kamolonol (NPC146388) compared to the HRG/Arg-1 doi:10.1001/jamanetworkopen.2022.2265.
ligand with NRP1, demonstrating Gbinding values of [7] Longley DB, Johnston PG. Molecular mechanisms of drug
–34.5 and –32.0 kcal/mol, respectively. During 200 ns resistance. J Pathol. 2005;205:275–292. doi:10.1002/
MD simulations, energetic and structural analyses indi- path.1706.
[8] Shi F, Shang L, Yang LY, et al. Neuropilin-1 contributes
cated perfect constancy for the kamolonol. Further- to esophageal squamous cancer progression via pro-
more, kamolonol elucidated favorable physiochemical moting P65-dependent cell proliferation. Oncogene.
and pharmacokinetic properties compared to HRG/Arg- 2018;37:935–943. doi:10.1038/onc.2017.399.
1. Finally, these results confirmed that the kamolonol [9] Luo M, Hou L, Li J, et al. VEGF/NRP-1 axis promotes pro-
could be used as a promising NRP1 inhibitor and hold gression of breast cancer via enhancement of epithelial-
mesenchymal transition and activation of NF-κB and
promise for further experimental assays toward cancer
β-catenin. Cancer Lett. 2016;373:1–11. doi:10.1016/j.can
treatment. let.2016.01.010.
[10] Hong TM, Chen YL, Wu YY, et al. Targeting neu-
ropilin 1 as an antitumor strategy in lung cancer.
Acknowledgements Clin Cancer Res. 2007;13:4759–4768. doi:10.1158/1078-
Ahmed M. Shawky would like to thank the Deanship of Sci- 0432.CCR-07-0001.
entific Research at Umm Al-Qura University for supporting [11] Li LH, Jiang X, Zhang Q, et al. Molecular targeted therapy
this work by Grant Code: (22UQU4331174DSR43). The com- for the treatment of gastric cancer. J Exp Clin Cancer Res.
putational work was completed with resources supported 2016;35:1–12. doi:10.1186/s13046-015-0276-9.
by the Science and Technology Development Fund (STDF- [12] Hang C, Yan HS, Gong C, et al. MicroRNA-9 inhibits
Egypt, Grants No. 5480 and 7972), and Bibliotheca Alexandrina gastric cancer cell proliferation and migration by tar-
(http://hpc.bibalex.org). Dr. Mahmoud A. A. Ibrahim extends geting neuropilin–1. Exp Ther Med. 2019;18:2524–2530.
his appreciation to the Academy of Scientific Research and doi:10.3892/etm.2019.7841.
Technology (ASRT), Egypt, for funding Graduation Projects [13] Zhou R, Curry JM, Roy LD, et al. A novel associ-
conducted at CompChem Lab, Egypt. ation of neuropilin-1 and MUC1 in pancreatic duc-
tal adenocarcinoma: role in induction of VEGF signal-
ing and angiogenesis. Oncogene. 2016;35:5608–5618.
Disclosure statement doi:10.1038/onc.2015.516.
[14] Hendricks C, Dubail J, Brohee L, et al. A novel phys-
No potential conflict of interest was reported by the author(s).
iological glycosaminoglycan-deficient splice variant of
neuropilin-1 Is anti-tumorigenic In vitro and In vivo.
PLoS One. 2016;11:e0165153. doi:10.1371/journal.pone.
Funding 0165153.
This work was supported by Umm Al-Qura University: [grant [15] Al-Shareef H, Hiraoka SI, Tanaka N, et al. Use of NRP1,
no 22UQU4331174DSR43]. a novel biomarker, along with VEGF-C, VEGFR-3, CCR7
JOURNAL OF TAIBAH UNIVERSITY FOR SCIENCE 13

and SEMA3E, to predict lymph node metastasis in [33] QUACPAC. 1.7.0.2; openeye scientific software. Santa Fe,
squamous cell carcinoma of the tongue. Oncol Rep. NM, USA, 2016.
2016;36:2444–2454. doi:10.3892/or.2016.5116. [34] Gasteiger J, Marsili M. Iterative partial equalization
[16] Jimenez-Hernandez LE, Vazquez-Santillan K, Castro- of orbital electronegativity—a rapid access to atomic
Oropeza R, et al. NRP1-positive lung cancer cells possess charges. Tetrahedron. 1980;36:3219–3228. doi:10.1016/
tumor-initiating properties. Oncol Rep. 2017;39:349– 0040-4020(80)80168-2.
357. doi:10.3892/or.2017.6089. [35] Trott O, Olson AJ. AutoDock Vina: improving the speed
[17] Soker S, Takashima S, Miao HQ, et al. Neuropilin-1 is and accuracy of docking with a new scoring function, effi-
expressed by endothelial and tumor cells as an isoform- cient optimization, and multithreading. J Comput Chem.
specific receptor for vascular endothelial growth fac- 2009;31:455–461. doi:10.1002/jcc.21334.
tor. Cell. 1998;92:735–745. doi:10.1016/S0092-8674(00) [36] Morris GM, Huey R, Lindstrom W, et al. AutoDock4
81402-6. and AutoDockTools4: Automated docking with selective
[18] Yoshida A, Shimizu A, Asano H, et al. VEGF-A/NRP1 stim- receptor flexibility. J Comput Chem. 2009;30:2785–2791.
ulates GIPC1 and Syx complex formation to promote doi:10.1002/jcc.21256.
RhoA activation and proliferation in skin cancer cells. Biol [37] Cetin A. In silico studies on stilbenolignan analogues as
Open. 2015;4:1063–1076. doi:10.1242/bio.010918. SARS-CoV-2 Mpro inhibitors. Chem Phys Lett. 2021;771:
[19] Sun L, Chen B, Jiang R, et al. Resveratrol inhibits lung can- 138563, doi:10.1016/j.cplett.2021.138563.
cer growth by suppressing M2-like polarization of tumor [38] Cetin A, Bursal E, Türkan F. 2-methylindole analogs as
associated macrophages. Cell Immunol. 2017;311:86–93. cholinesterases and glutathione S-transferase inhibitors:
doi:10.1016/j.cellimm.2016.11.002. Synthesis, biological evaluation, molecular docking, and
[20] Liu SD, Zhong LP, He J, et al. Targeting neuropilin-1 inter- pharmacokinetic studies. Arab J Chem. 2021;14:103449,
actions is a promising anti-tumor strategy. Chin Med J. doi:https://doi.org/10.1016/j.arabjc.2021.103449.
2021;134:508–517. doi:10.1097/CM9.0000000000001200. [39] Cetin A, Donmez A, Dalar A, et al. Tetra-substituted pyra-
[21] Chuckran CA, Liu C, Bruno TC, et al. Neuropilin-1: a zole analogues: synthesis, molecular docking, ADMET
checkpoint target with unique implications for cancer prediction, antioxidant and pancreatic lipase inhibitory
immunology and immunotherapy. J Immunother Can- activities. Med Chem Res. 2023;32:189–204. doi:10.1007/
cer. 2020;8:e000967. doi:10.1136/jitc-2020-000967. s00044-022-03005-7.
[22] Harvey AL, Clark RL, Mackay SP, et al. Current strategies [40] Case DAB, Cerutti DS, Cheatham TE, et al. AMBER 2016.
for drug discovery through natural products. Expert Opin San Francisco, USA: University of California.
Drug Discov. 2010;5:559–568. doi:10.1517/17460441. [41] Ibrahim MAA, Badr EAA, Abdelrahman AHM, et al. In Silico
2010.488263. targeting human multidrug transporter ABCG2 in breast
[23] Harvey AL. Natural products in drug discovery. Drug Dis- cancer: Database screening, molecular docking, and
cov Today. 2008;13:894–901. doi:10.1016/j.drudis.2008. molecular dynamics study. Mol Inform. 2022;41:2060039.
07.004. doi:10.1002/minf.202060039.
[24] Patridge E, Gareiss P, Kinch MS, et al. An analysis of FDA- [42] Ibrahim MAA, Badr EAA, Abdelrahman AHM, et al.
approved drugs: natural products and their derivatives. Prospective drug candidates as human multidrug trans-
Drug Discov Today. 2016;21:204–207. doi:10.1016/j.dru porter ABCG2 inhibitors: An in silico drug discovery
dis.2015.01.009. study. Cell Biochem Biophys. 2021;79:189–200. doi:10.
[25] Zeng X, Zhang P, He W, et al. NPASS: natural product 1007/s12013-021-00985-y.
activity and species source database for natural prod- [43] Ibrahim MAA, Abdelrahman AHM, Badr EAA, et al. Natu-
uct research, discovery and tool development. Nucleic rally occurring plant-based anticancerous candidates as
Acids Res. 2018;46:D1217–D1222. doi:10.1093/nar/gkx prospective ABCG2 inhibitors: an in silico drug discovery
1026. study. Mol Divers. 2022: 3255–3277. doi:10.1007/s11030-
[26] Mota F, Fotinou C, Rana RR, et al. Architecture and hydra- 022-10389-6.
tion of the arginine-binding site of neuropilin-1. FEBS J. [44] Ibrahim MAA, Abdeljawaad KAA, Abdelrahman AHM,
2018;285:1290–1304. doi:10.1111/febs.14405. et al. Non-β-Lactam allosteric inhibitors target
[27] Gordon JC, Myers JB, Folta T, et al. H++: a server for esti- methicillin-resistant staphylococcus aureus: An In silico
mating pKas and adding missing hydrogens to macro- drug discovery study. Antibiotics (Basel). 2021;10:934–
molecules. Nucleic Acids Res. 2005;33:W368–W371. doi: 956. doi:10.3390/antibiotics10080934.
10.1093/nar/gki464. [45] Ibrahim MAA, Abdelrahman AHM, Jaragh-Alhadad LA,
[28] Heller SR, McNaught A, Pletnev I, et al. Inchi, the et al. Exploring toxins for hunting SARS-CoV-2 main pro-
IUPAC international chemical identifier. J Cheminformat- tease inhibitors: molecular docking, molecular dynamics,
ics. 2015;7:23, doi:10.1186/s13321-015-0068-4. pharmacokinetic properties, and reactome study. Phar-
[29] OMEGA. 2.5.1.4; openeye scientific software. Santa Fe, maceuticals. 2022;15:153. doi:10.3390/ph15020153.
NM, USA, 2013. [46] Wang J, Wolf RM, Caldwell JW, et al. Development and
[30] Hawkins PC, Skillman AG, Warren GL, et al. Conformer testing of a general amber force field. J Comput Chem.
generation with OMEGA: algorithm and validation using 2004;25:1157–1174. doi:10.1002/jcc.20035.
high quality structures from the Protein Databank and [47] Maier JA, Martinez C, Kasavajhala K, et al. ff14SB: Ff14sb:
Cambridge Structural Database. J Chem Inf Model. improving the accuracy of protein side chain and back-
2010;50:572–584. doi:10.1021/ci100031x. bone parameters from ff99SB. J Chem Theory Comput.
[31] SZYBKI. 1.9.0.3, openeye scientific software. Santa Fe, NM, 2015;11:3696–3713. doi:10.1021/acs.jctc.5b00255.
USA, 2016. [48] Jakalian A, Jack DB, Bayly CI. Fast, efficient genera-
[32] Halgren TA. MMFF VI. MMFF VI. MMFF94s option for tion of high-quality atomic charges. AM1-BCC model:
energy minimization studies. J Comput Chem. 1999;20: II. Parameterization and validation. Parameterization
720–729. doi:10.1002/(SICI)1096-987X(199905)20:7 < and Validation J Comput Chem. 2002;23:1623–1641.
720::AID-JCC7 > 3.0.CO;2-X. doi:10.1002/jcc.10128.
14 M. A. A. IBRAHIM ET AL.

[49] Roux B, Simonson T. Implicit solvent models. Bio- surface area methods. II. The accuracy of ranking poses
phys Chem. 1999;78:1–20. doi:10.1016/S0301-4622(98) generated from docking. II. The accuracy of rank-
00226-9. ing poses generated from docking. J Comput Chem.
[50] Bayly CI, Cieplak P, Cornell WD, et al. A well-behaved elec- 2011;32:866–877. doi:10.1002/jcc.21666.
trostatic potential based method using charge restraints [61] Wang E, Sun H, Wang J, et al. End-point binding
for deriving atomic charges: the RESP model. J Phys free energy calculation with MM/PBSA and MM/GBSA:
Chem. 1993;97:10269–10280. doi:10.1021/j100142a004. Strategies and applications in drug design. Chem Rev.
[51] Frisch MJ, Trucks GW, Schlegel HB, et al. Gaussian 09, revi- 2019;119:9478–9508. doi:10.1021/acs.chemrev.9b00055.
sion E01; Gaussian09, Gaussian Inc. Wallingford, CT, USA: [62] Daina A, Michielin O, Zoete V. SwissADME: a free web tool
pn; 2009. to evaluate pharmacokinetics, drug-likeness and medic-
[52] Jorgensen WL, Chandrasekhar J, Madura JD, et al. inal chemistry friendliness of small molecules. Sci Rep.
Comparison of simple potential functions for simu- 2017;7:42717, doi:10.1038/srep42717.
lating liquid water. J Chem Phys. 1983;79:926–935. [63] Pires DE, Blundell TL, Ascher DB. pkCSM: Pkcsm: predict-
doi:10.1063/1.445869. ing small-molecule pharmacokinetic and toxicity prop-
[53] Darden T, York D, Pedersen L. Particle mesh Ewald: erties using graph-based signatures. J Med Chem. 2015;
AnN · log(N) method for Ewald sums in large sys- 58:4066–4072. doi:10.1021/acs.jmedchem.5b00104.
tems. J Chem Phys. 1993;98:10089–10092. doi:10.1063/1. [64] Kim MS, Oh KS, Lee JH, et al. Kamolonol suppresses
464397. angiotensin II-induced stress fiber formation and cellular
[54] Berendsen HJC, Postma JPM, Vangunsteren WF, et al. hypertrophy through inhibition of Rho-associated kinase
Molecular dynamics with coupling to an external bath. J 2 activity. Biochem Biophys Res Commun. 2013;438:
Chem Phys. 1984;81:3684–3690. doi:10.1063/1.448118. 318–323. doi:10.1016/j.bbrc.2013.07.069.
[55] Izaguirre JA, Catarello DP, Wozniak JM, et al. Langevin sta- [65] Dastan D, Salehi P, Reza Gohari A, et al. Disesquiterpene
bilization of molecular dynamics. J Chem Phys. 2001;114: and sesquiterpene coumarins from Ferula pseudalliacea,
2090–2098. doi:10.1063/1.1332996. and determination of their absolute configurations. Phy-
[56] Miyamoto S, Kollman PA. Settle: An analytical version of tochemistry. 2012;78:170–178. doi:10.1016/j.phytoche
the SHAKE and RATTLE algorithm for rigid water mod- m.2012.02.016.
els. J Comput Chem. 1992;13:952–962. doi:10.1002/jcc. [66] Dixit D, Reddy CRK, Trivedi MH, et al. Non-targeted
540130805. metabolomics approach to assess the brown marine
[57] Dassault Systèmes. BIOVIA, discovery studio visualize, macroalga Dictyota dichotoma as a functional food using
version 2019; dassault systèmes: San Diego, CA, USA, liquid chromatography with mass spectrometry. Sep Sci
2019. Plus. 2020;3:140–149. doi:10.1002/sscp.201900109.
[58] Massova I, Kollman PA. Combined molecular mechani- [67] Kerrigan JE. Molecular dynamics simulations in drug
cal and continuum solvent approach (MM-PBSA/GBSA) design. In: Kortagere S, editor. silico models for drug dis-
to predict ligand binding. Perspect Drug Discov. 2000;18: covery. Totowa, NJ: Humana Press; 2013. p. 95–113.
113–135. doi:10.1023/A:1008763014207. [68] De Vivo M, Masetti M, Bottegoni G, et al. Role of molecular
[59] Onufriev A, Bashford D, Case DA. Exploring protein dynamics and related methods in drug discovery. J Med
native states and large-scale conformational changes Chem. 2016;59:4035–4061. doi:10.1021/acs.jmedchem.
with a modified generalized born model. Proteins. 5b01684.
2004;55:383–394. doi:10.1002/prot.20033. [69] Jarvis A, Allerston CK, Jia H, et al. Small molecule
[60] Hou T, Wang J, Li Y, et al. Assessing the performance inhibitors of the neuropilin-1 vascular endothelial
of the molecular mechanics/Poisson Boltzmann sur- growth factor A (VEGF-A) interaction. J Med Chem.
face area and molecular mechanics/generalized Born 2010;53:2215–2226. doi:10.1021/jm901755g.

You might also like