You are on page 1of 11

pubs.acs.

org/jcim Article

Discovery of Novel Small Molecule Inhibitors Disrupting the PCSK9-


LDLR Interaction
Hengzhi Sun,§ Jinzheng Wang,§ Shengjie Liu,§ Xinyu Zhou, Liang Dai, Caiping Chen, Qinglong Xu,
Xiaoan Wen, Keguang Cheng, Hongbin Sun,* and Haoliang Yuan*
Cite This: J. Chem. Inf. Model. 2021, 61, 5269−5279 Read Online

ACCESS Metrics & More Article Recommendations *


sı Supporting Information
Downloaded via NORTHWESTERN POLYTECHNICAL UNIV on February 3, 2022 at 16:37:12 (UTC).
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

ABSTRACT: Proprotein convertase subtilisin kexin 9 (PCSK9)


has been identified as a reliable therapeutic target for hyper-
cholesterolemia and coronary artery heart diseases since the
monoclonal antibodies of PCSK9 have launched. Disrupting the
protein−protein interaction (PPI) between PCSK9 and the low-
density lipoprotein receptor (LDLR) has been considered as a
promising approach for developing PCSK9 inhibitors. However,
PPIs have been traditionally considered difficult to target by small
molecules since the PPI surface is usually large, flat, featureless, and
without a “pocket” or “groove” for ligand binding. The PCSK9-
LDLR PPI interface is such a typical case. In this study, a potential
binding pocket was generated on the PCSK9-LDLR PPI surface of
PCSK9 through induced-fit docking. On the basis of this induced binding pocket, virtual screening, molecular dynamics (MD)
simulation, and biological evaluations have been applied for the identification of novel small molecule inhibitors of PCSK9-LDLR
PPI. Among the selected compounds, compound 13 exhibited certain PCSK9-LDLR PPI inhibitory activity (IC50: 7.57 ± 1.40 μM).
The direct binding affinity between 13 and PCSK9 was determined with a KD value of 2.50 ± 0.73 μM. The LDLR uptake function
could be also restored to a certain extent by 13 in HepG2 cells. This well-characterized hit compound will facilitate the further
development of novel small molecule inhibitors of PCSK9-LDLR PPI.

■ INTRODUCTION
Low-density lipoprotein receptors (LDLRs) are the liver
PCSK9 binds to LDLR by a protein−protein interaction, and
the PCSK9-LDLR complex directly routes to lysosome for
surface receptors of LDL and responsible for eliminating destruction, which impairs the LDLR recycle and the plasma
LDL cholesterol (LDL-c) from human blood.1 After binding to LDL-c elimination (Figure 1B).8−10 The concept of blocking
a LDL particle, the LDLR is internalized into clathrin-coated PCSK9-LDLR PPI as a novel cholesterol-lowering therapy has
pits and subsequently convey LDL from cytoplasm to been solidly proved by the monoclonal antibodies launched on
lysosome for degradation. While in the endosomes, LDLR the market. Alirocumab11 and evolocumab,12 developed by
separates with LDL and recycles back to the plasma member.2 Sanofi and Amgen, respectively, both recognize and bind
Proprotein convertase subtilisin kexin 9 (PCSK9), the last directly to the PCSK9 surface that interacts with the LDLR-
identified member of the proprotein convertase family, is EGF(A) domain. This competitive inhibition could cause a
mainly expressed in hepatocytes and subsequently secreted robust reduction (>50%) of the plasma LDL-c level and a
into blood.3 It is initially synthesized as a 692-aa zymogen, and modest decrease (∼15%) in major cardiovascular events in the
then, the signal peptide (residues 1−30) is excised from the long term with no significant adverse events reported in clinical
protein precursor. In the endoplasmic reticulum, proPCSK9 trials.13−15 Despite the success of PCSK9 antibodies as
undergoes an ensuing autocatalytic cleavage of the peptide
cholesterol-lowering therapeutics, high cost and injection-
bond between Gln152 and Ser153, resulting in an N-terminal
only administration have limited their clinical accessibility.
prosegment that still attaches to the PCSK9 catalytic site by
noncovalent interactions and blocks the subsequent serine
protease activity (Figure 1A).4−6 The mature PCSK9 consists Received: May 11, 2021
of three main parts: a pro-domain (residues 31−152), a Published: September 23, 2021
catalytic domain (residues 153−451), and a C-terminal
domain (residues 452−692).7 It has been well interpreted
that PCSK9 is an endogenous intracellular and extracellular
negative-regular chaperone of LDLR on liver cells. Extracellular

© 2021 American Chemical Society https://doi.org/10.1021/acs.jcim.1c00521


5269 J. Chem. Inf. Model. 2021, 61, 5269−5279
Journal of Chemical Information and Modeling pubs.acs.org/jcim Article

Figure 1. (A) Autocatalytic process drives PCSK9 precursor to mature. (B) PCSK9 plays an important role in the regulation of LDLR degradation.
ER, endoplasmic reticulum; TGN, trans-Golgi network.

Figure 2. (A) 3D structure model of PCSK9-LDLR complex (PDB ID: 3BPS). (B) Key amino acid residues on PCSK9 surface involved in PCSK9-
LDLR PPI.

Therefore, it is highly desirable to develop small molecule catalytic domain, which was made up of a series of
inhibitors of PCSK9-LDLR PPI. hydrophobic amino acid residues centered on Phe379 and
Protein−protein interactions (PPIs) play vital roles in surrounded by several polar residues (Figure 2).34 The direct
physical and pathological processes such as metabolism,16 interactions between PCSK9 and LDLR include a β-sheet-like
immunity,17 inflammation,18 tumor proliferation, and meta- contact unit formed by residues 377−379 of PCSK9 and 308−
stasis.19 Great efforts have been made in discovering drugs 310 of LDLR-EGF(A) domains, hydrogen bond interactions
targeting and modulating key PPIs.20,21 However, the contributed by Ser153, Arg194, and Asp238, and hydrophobic
characteristics of PPIs are distinct from the traditional interactions involved in Ile369, Cys378, and Phe379 of
druggable targets such as enzymes, ion channels, and PCSK9. Currently, only a handful cases of PCSK9-LDLR
GPCRs.22 PPIs interfaces are generally large, flat, often PPI inhibitors have been reported, including small peptides,
hydrophobic, and more flexible. Also, suitable pockets or peptidomimetics, and small molecules (Figure 3). Pep2-8,37 a
grooves are usually lacking for ligand binding on the PPIs 13-peptide discovered by phage-displayed peptide libraries
interfaces,23,24 resulting in difficulties for molecular design and screening, showed potent PCSK9-LDLR PPI inhibitory
drug discovery.25,26 Nevertheless, some encouraging progress activity, PCSK9 binding affinity, and LDL uptake restoration.
has been made in developing PPIs inhibitors targeting PD1- CB_3638 was a small molecule exhibiting certain PCSK9-
PDL1,27 IL2-IL2R,28 MDM2-p53,29−31 and Bcl2-BclXL.32,33 LDLR PPI inhibitory activity in vitro, but the direct binding
PCSK9-LDLR PPI has been well understood based on the affinity to PCSK9 was unknown. LDLL-1dnlr39 was a
detailed analysis of crystal structures.34−36 The interface of peptidomimetic compound with micromolar PCSK9 binding
PCSK9 is a ∼500 Å2, solvent-accessible, flat surface on the affinity and capable of disturbing the targeted PPI and
5270 https://doi.org/10.1021/acs.jcim.1c00521
J. Chem. Inf. Model. 2021, 61, 5269−5279
Journal of Chemical Information and Modeling pubs.acs.org/jcim Article

Figure 3. Chemical structures of some reported PCSK9-LDLR PPI inhibitors.

increasing LDL uptake from extracellular medium. The and then docked to PCSK9 according to the reported
tetraimidazole derivative RIm1340 exhibited certain inhibition methods44 using the Induced-Fit Docking module implemented
on PCSK9-LDLR binding and a cholesterol-lowering effect on in Schrödinger 2009. The obtained structure of PCSK9 in
HepG2 cells. Compound 3f,41 which was derived from its lead complex with SBC-115,076 was used for virtual screening
compound nilotinib, showed PPI disturbing efficacy without against the prepared Specs compound library containing about
kinases inhibitory activity. It was hypothesized that 3f was 250,000 compounds. The binding site was defined based on
bound in the catalytic site of PCSK9 which was occupied by the docking conformation of SBC-115,076. The virtual
the self-inhibitory peptide from the Pro-domain N-terminal42 screening workflow was performed using the three different
and known to be inaccessible for small molecule antagonists.43 precision modes in sequence, high-throughput virtual screen-
Therefore, the binding region of 3f has yet to be clarified ing (HTVS), standard-precision (SP), and extra-precision
experimentally. Compounds SBC-115,076 44 and SBC- (XP). The final results were output on the basis of Glide
110,42445 were reported as PCSK9 inhibitors, which were docking score.
discovered from PPI surface modeling and high-throughput Molecular Dynamics Simulation and Analysis. The
virtual screening. They exhibited lipid-lowering effects in cell protein−ligand interactions between PCSK9 and compound
and animal model tests. However, their direct binding affinity 13 were investigated by molecular dynamics simulation. The
to PCSK9 was not reported. Together, there is still a lack of complex structure obtained by molecular docking was used for
reliable small molecule inhibitors of PCSK9-LDLR PPI. setting up the model of MD simulation. The AM1-BCC charge
Virtual screening is a reliable tool in the process of hit model was applied for the calculation of atomic point charges
identification and lead discovery.46 Molecular dynamics (MD) of the compound. The missing residues in the crystal structure
simulation is prevalently applied in the study of protein−ligand were complemented according to the amino acid residue
interactions.47 In this study, virtual screening, MD simulation, sequence of PCSK9. The Pro-domain was removed because of
and biological evaluations have been applied to identify novel the self-cleavage in the protein mature process. The complex
small molecule inhibitors of PCSK9-LDLR PPI. Our structure, including the protein, compound 13, counterions,
experimental results confirmed that compound 13 was a and water molecules, was immersed in a cubic box of a TIP3P
promising hit for further study.


water model with a 10 Å minimum solute-wall distance. The
procedures and detailed parameter settings were in accordance
MATERIALS AND METHODS with our previous studies.48 The simulation time of 500 ns was
Virtual Screening Protocol. The crystal structure of set for the complex system.
PCSK9 (PDB ID: 2P4E) obtained from the protein data bank The trajectory was analyzed by using cpptraj in AmberTools
(http://www.rcsb.org) was refined with the protein preparation 15. The stability of the protein−ligand complex was judged by
wizard module, including adding hydrogen atoms, assigning calculating the root-mean-square deviation (RMSD), root-
partial charges, and determining the protonation state. SBC- mean-square fluctuation (RMSF), and radius of gyration (Rg).
115,076 was prepared using the Ligand preparation module The close contacts and hydrogen bond occupancy for the
5271 https://doi.org/10.1021/acs.jcim.1c00521
J. Chem. Inf. Model. 2021, 61, 5269−5279
Journal of Chemical Information and Modeling pubs.acs.org/jcim Article

Figure 4. (A) Original flat PCSK9-LDLR interface on PCSK9. (B) Binding pocket was generated by induced-fit docking of SBC-115,076 to the
PPI interface on PCSK9. (C) Binding mode of SBC-115,076 was predicted with induced-fit docking.

equilibrated trajectory were analyzed. The binding free energy Direct binding of each compound to PCSK9 was tested by
(ΔGpbsa) was calculated with the method of molecular SPR using Biacore T200 (GE Healthcare) at room temper-
mechanics Poisson−Boltzmann surface area (MM/PBSA). ature. The CM5 chip was immobilized with the PCSK9
The binding free energy decomposition analysis was also protein in 10 mM sodium acetate (pH 4.5) according to a
performed to estimate the contribution of each residue in the standard amine-coupling procedure and remaining coupling
pocket for ligand binding. sites were blocked with 1 M ethanolamine, pH 8.5. Each
ELISA Assay for PCSK9-LDLR Binding. The well- compound for binding affinity determination was initially
identified PCSK9-LDLR peptidic inhibitor pep2-8 was dissolved in dimethyl sulfoxide to 20 mM as a stocking
selected as the positive control in all our biological evaluations. solution and serially diluted with a PBS buffer containing
It was synthesized by Synpeptide Inc. with the HPLC purity of 0.05% (v/v) Tween 20 and 5% (v/v) DMSO to certain
>98%, the exact sequence of which is Ac- concentrations (0.39−200 μM) when tested. Then, the test
TVFTSWEEYLDWV-NH2. To test the PCSK9-LDLR PPI sample was injected at a flow rate of 30 μL/min for 90 s to
inhibitory activity, ELISA assays were conducted using contact with the proteins immobilized on the chip, followed by
PCSK9-LDLR the in vitro binding assay kit (CycLex Co., dissociation from the chip for another 90 s. The SPR
Japan) according to the manufacturer’s protocol with minor sensorgrams were solvent corrected, reference and buffer
modifications. Briefly, the compounds were dissolved in subtracted, and analyzed with Biacore T200 evaluation
dimethyl sulfoxide (DMSO) as 20 mM stocking solutions software (GE Healthcare) to determine the kinetic parameters
while the positive control pep2-8 was prepared to a 2 mM and KD values.
solution using dd H2O and then serially diluted to final Cell Viability Assay. The HepG2 cell line was obtained
from the Cell Bank of Shanghai Institute of Cell Biology,
concentrations with a reaction buffer that contained recombi-
Chinese Academy of Sciences, and authenticated by STR
nant His-tagged PCSK9 at 80 ng/mL. After a gentle shake and
profiling (Guangzhou Cellcook Biotech Co., Ltd.). Cells were
preincubated at room temperature for 1 h, a 100 μL reaction
maintained in a 5% CO2-humidified incubator at 37 °C and
mixture was added to a microplate that was coated with cultured in modified Dulbecco’s modified Eagle’s medium
EGF(AB) peptide of LDLR. Subsequent procedures were (DMEM) supplemented with 10% (v/v) fetal bovine serum
performed in strict accordance with the instructions. The (FBS), 1% (v/v) penicillin-streptomycin, and 1% (v/v) sodium
absorbance of a test well was measured at dual wavelengths of pyruvate (Gibco). Medium and supplements were obtained
450/540 nm, respectively, using an EnVision Multimode Plate from Biological Industries except where indicated.
Reader (EnSpire, PerkinElmer). The inhibition percentage of For cell viability test, HepG2 cells were seeded at 4000 cells/
test compounds was calculated from the standard His-tagged well in the 96-well plate for 24 h. Then, the medium containing
PCSK9 dose−absorbance curve after vehicle control correc- test compounds was added to the final concentrations (3.12−
tion. To determine the IC50 of test compounds, the serial 200 μL), and cells were incubated for another 48 h. Viable cells
inhibition percentage (the Y-coordinate) was plotted as the were determined by MTT (Promega; #G7570) treatment for 3
function of a test concentration (the X-coordinate). The h and fluorescence detection using an EnVision Multimode
results were analyzed with GraphPad Prism version 5.0 Plate Reader (EnSpire, PerkinElmer). Analysis and IC50 values
(GraphPad, La Jolla, CA, USA). All the tests were duplicated were assessed by GraphPad Prism version 5.0. Data were
three times, and data were denoted as mean ± standard presented as mean ± s.d. (n = 3).
deviation (s.d.) (n = 3). BODIPY-LDL Uptake Assay. HepG2 cells were used in
Binding Assay by Surface Plasmon Resonance (SPR). the fluorescent-conjugated LDL uptake assay. They were
The His-tagged human PCSK9 (residues 1−692) was obtained seeded at 30,000 cells/well in a 96-well microplate with a clear
as lyophilized powder with SDS-PAGE purity of >95% from a bottom and black wall (Corning; #CLS3603). Following
commercial source (Sino Biological; #29698-H08H). overnight incubation, the medium was changed to DMEM
5272 https://doi.org/10.1021/acs.jcim.1c00521
J. Chem. Inf. Model. 2021, 61, 5269−5279
Journal of Chemical Information and Modeling pubs.acs.org/jcim Article

without supplements for another 24 h. The positive peptide


pep2-8 (10 μM) or compound 13 at various concentrations
was preincubated with 10 μg/mL of PCSK9 in DMEM
containing 0.5% (v/v) DMSO for 0.5 h before being added to
the cells. The vehicle wells were only treated with PCSK9 in a
serum-free medium without inhibitors. After 2 h, BODIPY FL
LDL (Thermo Fisher Scientific; #L3483) was added with a
final concentration of 10 μg/mL per well and incubated for
another 3.5 h. Each test well was washed three times with cold
PBS, and cellular uptake of fluorescence was evaluated by an
EnVision Multimode Plate Reader (EnSpire, PerkinElmer)
using excitation and emission wavelengths of 515 and 520 nm,
respectively. Data were presented as means ± s.d. (n = 3).
Statistical significance of differences was determined by one-
way ANOVA, and the levels were indicated as *P < 0.05, **P
< 0.01, and ***P < 0.001, compared with the PCSK9-treated
DMSO group.
RNA Isolation and Real Time PCR Analysis. HepG2
cells were seeded at 2 × 105 cells/well in a 12-well plate for 24
h. Then, each test well was treated with berberine at 10 μM, Figure 5. Virtual screening workflow.
pep2-8 at 10 μM, or compound 13 at several concentrations,
respectively, and incubated for 6 h. Total RNA was isolated
from HepG2 cell lysate using the Trizol reagent (Invitrogen,
Carlsbad, CA). The lysate was well mixed with chloroform and
centrifuged for 15 min at 13,000 rpm at 4 °C. The top aqueous
phase containing RNA was isolated and mixed with
isopropanol to precipitate the total RNA. After collection
and concentration measurement, 1 μg of total RNA was used
to synthesize the cDNA with a commercial-sourced HiScript II
Q RT SuperMix kit for qPCR (Vazyme Biotech Co.) by
following the manufacturer’s instructions. Real-time PCR was
performed using a Power SYBR Green Master Mix with the Figure 6. Preliminary screening results (inhibitory rate) of the 30
primers in Table S1. Expressions of PCSK9 and LDLR mRNAs selected compounds by the PCSK9-LDLR in vitro binding assay with
in each sample were normalized to the corresponding GAPDH pep2-8 as the positive control.
mRNA. The relative mRNA expression levels were determined
using the 2−ΔΔCt, method and each cDNA sample was run in
triplicate. Data were presented as mean ± s.d. (n = 3).
Statistical significance of differences was determined by one-
way ANOVA, and the levels were indicated as *P < 0.05, **P
< 0.01, and ***P < 0.001 compared with DMSO group.

■ RESULTS AND DISCUSSION


Virtual Screening. The PCSK9-LDLR PPI interface was
initially large and flat (Figure 4A), which was difficult and not
suitable for discovering small molecule inhibitors. The virtual
Figure 7. (A) Calculated IC50 value of compound 13 in the PCSK9-
screening attempts directly and rigidly based on the original LDLR in vitro binding assay. (B) Chemical structure of compound 13.
surface failed to identify a PCSK9-LDLR inhibitor in the
reported study41 and our previous attempts. Usually, a
“pocket” is needed for the binding of small molecules to the found between SBC-115,076 and the residues of Arg237 and
target protein. Compound SBC-115,076 was identified as a Thr377. This predicted complex structure was used in the
PCSK9-LDLR PPI inhibitor by induced-fit docking. Although subsequent virtual screening. After the prepared Specs
the direct binding affinity was not reported, the cellular and in compounds were docked into the pocket with the three
vivo experiments confirmed its bioactivity in lipid regulation. precision modes successively, the retained compounds were
The discovery of SBC-115,076 gave us the inspiration. The visually inspected by considering their binding modes, docking
PPI interface on PCSK9 consisted of several loops, including scores, and structural diversity. The final selected 30 molecules
Ser153-Trp156, His193-Glu197, Arg237-Ala239, and Ser376- (Table S2) from the virtual screening workflow (Figure 5)
Phe379. Taking the flexibility of PPI interface into account, were purchased from the vendor. Biological evaluations,
SBC-115,076 was docked to the PPI interface on PCSK9 by including PCSK9-LDLR PPI inhibitory activity, PCSK9
referring to the reported methods of induced-fit docking. The binding affinity, and cellular BODIPY-LDL uptake, were
induced pocket was long and narrow, passing through the performed to identify the hit compounds.
whole PPI interface (Figure 4B). It consisted of Ile154, ELISA Assay for PCSK9-LDLR Binding. Subsequently,
Trp156, Glu159, Arg194, Glu195, Arg237, Asp238, Thr377, their preliminary PCSK9-LDLR PPI inhibitory rates of the 30
and Phe379 (Figure 4C). Hydrogen bond interactions were selected compounds at the concentration of 25 μM were
5273 https://doi.org/10.1021/acs.jcim.1c00521
J. Chem. Inf. Model. 2021, 61, 5269−5279
Journal of Chemical Information and Modeling pubs.acs.org/jcim Article

Figure 8. Binding affinity values of (A) 13 and (B) pep2-8 to PCSK9 were determined using SPR.

Figure 9. Binding mode of 13 was predicted with molecular docking. (A) Binding pocket is shown as a white surface. (B) Amino acid residues
constituting the binding pocket are shown as sticks.

(Figure 6, Table S3). However, compounds 2 did not pass the


Pan-Assay Interference Compounds (PAINS) filter. Therefore,
the remaining three compounds, 6, 7, and 13, were picked out
for further determination of IC50 values. Compounds 6 and 7
did not show considerable and does-dependent inhibitory
activity (IC50 > 50 μM) with ascending concentrations. But
compound 13 exhibited a dose-dependent inhibitory response
against the PCSK9-LDLR interaction with an IC50 value of
7.57 ± 1.40 μM (Figure 7, Figure S1). We have also performed
mass spectrometry analysis of compound 13 (Figure S2), and
the result was in accordance with the provided structure
information, confirming its molecular weight.
Binding Assay by SPR. The direct binding affinity
between PCSK9 and compound 13 was determined using
Figure 10. RMSD values of PCSK9−13 complex in the first 170 ns of SPR assay.49 The protein−ligand binding affinity was dose-
the entire molecular dynamics simulation. dependent with a KD value of 2.50 ± 0.73 μM (Figure 8A,
Figure S3), suggesting the direct binding between 13 and
measured by the ELISA assay, while pep2-8 at the PCSK9. The KD value of pep2-8 binding to PCSK9 in our
concentration of 10 μM was applied as the positive control. assay was 3.93 μM (Figure 8B), consistent with the reported
Among them, compounds 6 and 13 at 25 μM displayed value.39
inhibitory rates of 82.8 ± 1.7% and 64.2 ± 0.7%, respectively. MD Simulation. Molecular dynamics simulation was
The inhibitory rates of compounds 2 and 7 were also more performed to investigate the binding mode of 13 and
than 40%, 41.97 ± 6.77%, and 45.93 ± 1.55%, respectively PCSK9. This hit compound was docked into the pocket that

Figure 11. (A) Rg and (B) RMSF values of protein Cα atoms are calculated.

5274 https://doi.org/10.1021/acs.jcim.1c00521
J. Chem. Inf. Model. 2021, 61, 5269−5279
Journal of Chemical Information and Modeling pubs.acs.org/jcim Article

Figure 12. Close contact analysis of the PCSK9−13 complex.

Table 1. Hydrogen Bond Analysis with the Equilibrium Trajectory


Ligand Acceptor Donor Occupancy (%) Average distance (Å) Average angle (deg)
13 Ile154@O MOL@N3H7 55.09 2.93 159.17
MOL@O3 Ser153@OH 18.53 2.77 155.81
MOL@O2 Ser153@NH3 12.30 2.88 140.25
MOL@O2 Ser153@NH2 10.89 2.87 139.60
MOL@O2 Ser153@NH1 10.85 2.88 140.30

Table 2. Binding Free Energy Calculated Using the MM/ 175 ns (Figure S4). By checking the trajectory with VMD and
PBSA Method extracting the representative snapshots, it was found that the
ligand disassociated from the binding pocket from about 170
PCSK9−13
ns. Therefore, the trajectory of the first 170 ns was used for
Energy (kcal mol−1) Mean Std. analysis. The RMSD values (Figure 10) reflecting the stability
ΔGvdW −33.89 3.66 of the protein−ligand system were calculated by using the
ΔGele −43.05 7.68 initial conformation as a reference. The protein stayed stable
ΔGgas −76.94 9.02 all through the process around an average RMSD value of 2.0
ΔGpol, sol 60.59 8.17 Å. There were some fluctuations happening to the ligand
ΔGnonpol, sol −5.16 0.24 before 50 ns, while it then converged to an equilibrium state
ΔGsol 55.43 8.05 with an average RMSD value of 2.5 Å. The calculated Rg values
ΔGpbsa −21.51 4.02 (Figure 11A) showed that the protein−ligand system stayed
compact in the whole process with Rg values fluctuating
uniformly between 25.0 and 25.5 Å. The stability of residues in
the process of simulation was reflected by the RMSF values.
The residues constituting the binding site, including those
forming hydrogen bond interactions with 13, kept a small
RMSF value (Figure 11B), indicating the stable state of the
binding site in the whole process of simulation.
Considering the equilibrium state of the simulation, the MD
trajectory of the last 50 ns was used for calculation of close
contacts, hydrogen bond occupancy, and binding free energy.
The close contacts were defined as the protein−ligand
interactions between the ligand and residues within 4.5 Å
around it. The residues, Ser153, Ile154, Pro155, Trp156,
Glu159, Asp238, and Thr377 (Figure 12), showed a relatively
high percentage of close contacts. In combination with the
hydrogen bond occupancy analysis (Table 1), both of the
hydrophobic and hydrogen bond interactions contributed
greatly to the close contacts of Ser153 and Ile154, while
Figure 13. Binding free energy decomposition was performed for the hydrophobicity was important for the contribution of Pro155.
PCSK9−13 complex. Although some hydrogen bonds were found between 13 and
the pocket initially, only the one with Ile154 was stable in the
induced by SBC-115,076 (Figure 9A). Some residues process of simulation. These results indicated the important
constituting the binding pocket, including Ser153, Ile154, role these residues played for ligand binding.
Glu195, Asp238, and Thr377, formed hydrogen bond The binding free energy calculated with the MM/PBSA
interactions with compound 13 (Figure 9B). This docking method was −21.51 kcal/mol. The ΔGele and ΔGvdw terms
result was used as the initial conformation for the unrestrained contributed greatly to the binding free energy (Table 2).
molecular dynamics simulation of 500 ns. It seemed that some Binding free energy decomposition was also performed to
fluctuations happened to the protein−ligand system around investigate the contribution of each residue to ligand binding.
5275 https://doi.org/10.1021/acs.jcim.1c00521
J. Chem. Inf. Model. 2021, 61, 5269−5279
Journal of Chemical Information and Modeling pubs.acs.org/jcim Article

Figure 14. Cellular effects of 13 on restoring LDL uptake function. (A) LDL uptake RFU values of each group. (B) Relative restored rates of LDL
uptake of each group.

Figure 15. Real-time PCR analysis of (A) LDLR mRNA and (B) PCSK9 mRNA in HepG2 cells after the treatment with 13.

The threshold value of an absolute decomposed energy for 14B). Although compound 13 did not restore the LDLR
judging the importance of a residue is 1.0 kcal/mol. The function at the concentration of 10 μM, it restored LDL uptake
residues, Ile154, Pro155, Trp156, and Phe379, made great of PCSK9-treated HepG2 cells to 36.0 ± 5.3% at 44 μM
contributions to the binding of 13 (Figure 13). Some other compared with the DMSO control. At the concentration of
residues, Glu195 and Asp238, were not favorable for ligand 200 μM, it did not show improved potency. Therefore,
binding. Both of these residues located on the same side of the compound 13 could restore the LDL uptake level to a certain
binding site (Figure 9B), indicating the direction for further extent.
structural optimization. The effects of compound 13 on transcriptions of PCSK9 and
Cellular Effects of Compound 13. As PCSK9 is mainly LDLR genes were then investigated. Berberine was reported as
expressed from hepatocyte and responsible for the degradation a lipid-lowering molecule that could upregulate hepatic
of LDLR on the surface of liver, it is recommended to use liver LDLR50 and downregulate plasma PCSK951 transcriptionally.
cells in the cell functional tests of PCSK9 inhibitors. HepG2 is In our assay, HepG2 cells were treated with pep2-8 and 13
an immortalized cell line consisting of human liver carcinoma under the same conditions of the LDL uptake assay with
cells and has been widely used in the studies of LDL uptake berberine (10 μM) as the positive control. Compared with
assays of PCSK9-LDLR PPI inhibitors mentioned berberine, compound 13 showed minor effects on LDLR and
above.37,39,41 In this study, cellular effects of compound 13 PCSK9 mRNA levels at the concentrations of 10 and 44 μM
were then investigated in HepG2 cells. The MTT assay was (Figure 15). The LDLR mRNA level was obviously lower than
first conducted to guarantee the cell viability for the following the control group by treating with 13 at the higher
experiments. As shown in Figure S5, this hit compound concentration of 200 μM. The results indicated the LDL
exhibited no obvious inhibitory effects on HepG2 cell uptake recovery effect of 13 at low concentrations in HepG2
proliferation at all the test concentrations. The fluorescent- was independent of transcriptional regulation of PCSK9 and
labeled LDL uptake assay is indispensable for the test of LDLR genes.


cholesterol-lowering effect of drugs. We employed the
prevalently used BODIPY-LDL37,39 as the fluorescence probe
CONCLUSION
due to its stability under physiological condition and high
quantum yields. In the BODIPY-LDL uptake assay, the LDL Although with great value as reliable targets, PPIs are usually
uptake level was reduced by 35.6% after adding PCSK9 to characterized as “undruggable” since most of the PPI interface
HepG2 cells (Figure 14A). Theoretically, inhibition of PCSK9- is large, flat, and featureless, without a “pocket” or “ groove” for
LDLR PPI could restore impaired hepatocyte LDLR function the binding of small molecules. The PCSK9-LDLR PPI
and LDL uptake. Treatment with pep2-8 at the concentration interface is such a typical case. Therefore, it has been
of 10 μM restored almost 100% LDL uptake level (Figure considered as a challenging work for developing small molecule
5276 https://doi.org/10.1021/acs.jcim.1c00521
J. Chem. Inf. Model. 2021, 61, 5269−5279
Journal of Chemical Information and Modeling pubs.acs.org/jcim Article

inhibitors of PCSK9-LDLR PPI. In this study, a potential Jinzheng Wang − Jiangsu Key Laboratory of Drug Discovery
binding pocket on the PCSK9-LDLR PPI interface of PCSK9 for Metabolic Disease and State Key Laboratory of Natural
was generated by induced-fit docking of SBC-115,076. This Medicines, China Pharmaceutical University, Nanjing
induced binding pocket was subsequently used for virtual 210009, P. R. China
screening to identify novel small molecule inhibitors of Shengjie Liu − Jiangsu Key Laboratory of Drug Discovery for
PCSK9-LDLR PPI. The selected 30 compounds were Metabolic Disease and State Key Laboratory of Natural
purchased from the vendor. Among them, compound 13 Medicines, China Pharmaceutical University, Nanjing
exhibited potent and dose-dependent inhibitory activity against 210009, P. R. China
PCSK9-LDLR interaction in the ELISA assay. Also, the SPR Xinyu Zhou − Jiangsu Key Laboratory of Drug Discovery for
result showed direct binding between 13 and PCSK9. The Metabolic Disease and State Key Laboratory of Natural
molecular dynamics simulation revealed the stable binding Medicines, China Pharmaceutical University, Nanjing
mode of 13 in the induced pocket. The results of the cellular 210009, P. R. China
study showed that 13 could restore the LDL uptake function of Liang Dai − Jiangsu Key Laboratory of Drug Discovery for
PCSK9-treated HepG2 cells to some extent. This well- Metabolic Disease and State Key Laboratory of Natural
characterized hit compound validated the reliability of our Medicines, China Pharmaceutical University, Nanjing
method. It is presumed that better results would be obtained 210009, P. R. China; orcid.org/0000-0003-0172-4759
by performing induced-fit docking against the entire Caiping Chen − Jiangsu Key Laboratory of Drug Discovery for
compound library if abundant computational resources are Metabolic Disease and State Key Laboratory of Natural
available. We hope this study will facilitate the further Medicines, China Pharmaceutical University, Nanjing
development of novel small molecule PCSK9-LDLR PPI 210009, P. R. China
inhibitors. Qinglong Xu − Jiangsu Key Laboratory of Drug Discovery for


Metabolic Disease and State Key Laboratory of Natural
DATA AND SOFTWARE AVAILABILITY Medicines, China Pharmaceutical University, Nanjing
The screening compound library was downloaded from Specs 210009, P. R. China; orcid.org/0000-0003-4450-6866
(https://specs.net). The PDB files were downloaded from the Xiaoan Wen − Jiangsu Key Laboratory of Drug Discovery for
RCSB protein data bank (https://www.rcsb.org). All the Metabolic Disease and State Key Laboratory of Natural
output data and structures of purchased compounds have been Medicines, China Pharmaceutical University, Nanjing
provided in the article and Supporting Information. The free 210009, P. R. China; orcid.org/0000-0001-7852-1154
software package of PyMOL1.5.0.5 (https://pymol.org) was Keguang Cheng − State Key Laboratory for Chemistry and
used in this study. The MD simulation was performed and Molecular Engineering of Medicinal Resources, School of
analyzed with the licensed Amber14 and publicly available Chemistry and Pharmacy, Guangxi Normal University,
AmberTools15 (https://ambermd.org). Virtual screening was Guilin 541004, P. R. China; orcid.org/0000-0002-2883-
performed using Schrödinger2009 (https://www.schrödinger. 0845
com) purchased by China Pharmaceutical University. Complete contact information is available at:


https://pubs.acs.org/10.1021/acs.jcim.1c00521
ASSOCIATED CONTENT
Author Contributions
*
sı Supporting Information
§
H.S., J.W., and S.L. contributed equally to this work.
The Supporting Information is available free of charge at
https://pubs.acs.org/doi/10.1021/acs.jcim.1c00521. Notes
The authors declare no competing financial interest.


Additional experimental materials, structural informa-
tion, and experimental and computational results (PDF)
ACKNOWLEDGMENTS

■ AUTHOR INFORMATION
Corresponding Authors
Financial support from the National Natural Science
Foundation of China (82073707, 91853125, 81730094,
81773584, 21977118, and 21971256) and Natural Science
Hongbin Sun − Jiangsu Key Laboratory of Drug Discovery for Foundation of Jiangsu Province (BK20202009), BAGUI
Metabolic Disease and State Key Laboratory of Natural Scholar Program of Guangxi Province of China (2016A13),
Medicines, China Pharmaceutical University, Nanjing and Key R&D Program of Jiangsu Province (BE2018710) are
210009, P. R. China; orcid.org/0000-0002-3452-7674; gratefully acknowledged. This project was also supported by
Phone: +86-25-83271050; Email: hongbinsun@ the “111” Project from the Ministry of Education of China and
cpu.edu.cn State Administration of Foreign Expert Affairs of China (111-
Haoliang Yuan − Jiangsu Key Laboratory of Drug Discovery 2-07).


for Metabolic Disease and State Key Laboratory of Natural
Medicines, China Pharmaceutical University, Nanjing REFERENCES
210009, P. R. China; orcid.org/0000-0003-0248-4347; (1) Brown, M. S.; Anderson, R. G.; Goldstein, J. L. Recycling
Phone: +86-25-83271050; Email: yhl@cpu.edu.cn receptors: the round-trip itinerary of migrant membrane proteins. Cell
1983, 32, 663−667.
Authors
(2) Brown, M. S.; Goldstein, J. L. A receptor-mediated pathway for
Hengzhi Sun − Jiangsu Key Laboratory of Drug Discovery for cholesterol homeostasis. Science 1986, 232, 34−47.
Metabolic Disease and State Key Laboratory of Natural (3) Naureckiene, S.; Ma, L.; Sreekumar, K.; Purandare, U.; Frederick
Medicines, China Pharmaceutical University, Nanjing Lo, C.; Huang, Y.; Chiang, L. W.; Grenier, J. M.; Ozenberger, B. A.;
210009, P. R. China Steven Jacobsen, J.; Kennedy, J. D.; DiStefano, P. S.; Wood, A.;

5277 https://doi.org/10.1021/acs.jcim.1c00521
J. Chem. Inf. Model. 2021, 61, 5269−5279
Journal of Chemical Information and Modeling pubs.acs.org/jcim Article

Bingham, B. Functional characterization of Narc 1, a novel proteinase ficiency virus entry, and apoptosis. Pharmacol. Ther. 2000, 86, 201−
related to proteinase K. Arch. Biochem. Biophys. 2003, 420, 55−67. 215.
(4) Liberale, L.; Montecucco, F.; Camici, G. G.; Dallegri, F.; (18) Song, Y.; Buchwald, P. TNF Superfamily Protein-Protein
Vecchie, A.; Carbone, F.; Bonaventura, A. Treatment with Proprotein Interactions: Feasibility of Small- Molecule Modulation. Curr. Drug
Convertase Subtilisin/Kexin Type 9 (PCSK9) Inhibitors to Reduce Targets 2015, 16, 393−408.
Cardiovascular Inflammation and Outcomes. Curr. Med. Chem. 2017, (19) Nero, T. L.; Morton, C. J.; Holien, J. K.; Wielens, J.; Parker, M.
24, 1403−1416. W. Oncogenic protein interfaces: small molecules, big challenges. Nat.
(5) Nassoury, N.; Blasiole, D. A.; Tebon Oler, A.; Benjannet, S.; Rev. Cancer 2014, 14, 248−262.
Hamelin, J.; Poupon, V.; McPherson, P. S.; Attie, A. D.; Prat, A.; (20) Arkin, M. R.; Wells, J. A. Small-molecule inhibitors of protein-
Seidah, N. G. The cellular trafficking of the secretory proprotein protein interactions: progressing towards the dream. Nat. Rev. Drug
convertase PCSK9 and its dependence on the LDLR. Traffic 2007, 8, Discovery 2004, 3, 301−317.
718−732. (21) Arkin, M. R.; Whitty, A. The road less traveled: modulating
(6) McNutt, M. C.; Lagace, T. A.; Horton, J. D. Catalytic activity is signal transduction enzymes by inhibiting their protein-protein
not required for secreted PCSK9 to reduce low density lipoprotein interactions. Curr. Opin. Chem. Biol. 2009, 13, 284−290.
receptors in HepG2 cells. J. Biol. Chem. 2007, 282, 20799−20803. (22) Winter, A.; Higueruelo, A. P.; Marsh, M.; Sigurdardottir, A.;
(7) Seidah, N. G.; Benjannet, S.; Wickham, L.; Marcinkiewicz, J.; Pitt, W. R.; Blundell, T. L. Biophysical and computational fragment-
Jasmin, S. B.; Stifani, S.; Basak, A.; Prat, A.; Chrétien, M. The based approaches to targeting protein-protein interactions: applica-
secretory proprotein convertase neural apoptosis-regulated convertase tions in structure-guided drug discovery. Q. Rev. Biophys. 2012, 45,
1 (NARC-1): Liver regeneration and neuronal differentiation. Proc. 383−426.
Natl. Acad. Sci. U. S. A. 2003, 100, 928−933. (23) Smith, M. C.; Gestwicki, J. E. Features of protein-protein
(8) Cunningham, D.; Danley, D. E.; Geoghegan, K. F.; Griffor, M. interactions that translate into potent inhibitors: topology, surface
C.; Hawkins, J. L.; Subashi, T. A.; Varghese, A. H.; Ammirati, M. J.; area and affinity. Expert Rev. Mol. Med. 2012, 14, No. e16.
Culp, J. S.; Hoth, L. R.; Mansour, M. N.; McGrath, K. M.; Seddon, A. (24) Buchwald, P. Small-molecule protein-protein interaction
P.; Shenolikar, S.; Stutzman-Engwall, K. J.; Warren, L. C.; Xia, D.; inhibitors: therapeutic potential in light of molecular size, chemical
Qiu, X. Structural and biophysical studies of PCSK9 and its mutants space, and ligand binding efficiency considerations. IUBMB Life 2010,
linked to familial hypercholesterolemia. Nat. Struct. Mol. Biol. 2007, 62, 724−731.
14, 413−419. (25) Wells, J. A.; McClendon, C. L. Reaching for high-hanging fruit
(9) Horton, J. D.; Cohen, J. C.; Hobbs, H. H. PCSK9: a convertase in drug discovery at protein-protein interfaces. Nature 2007, 450,
that coordinates LDL catabolism. J. Lipid Res. 2009, 50 (Suppl), 1001−1009.
S172−177. (26) Milroy, L.-G.; Grossmann, T. N.; Hennig, S.; Brunsveld, L.;
(10) Qian, Y. W.; Schmidt, R. J.; Zhang, Y.; Chu, S.; Lin, A.; Wang, Ottmann, C. Modulators of Protein-Protein Interactions. Chem. Rev.
H.; Wang, X.; Beyer, T. P.; Bensch, W. R.; Li, W.; Ehsani, M. E.; Lu, 2014, 114, 4695−4748.
D.; Konrad, R. J.; Eacho, P. I.; Moller, D. E.; Karathanasis, S. K.; Cao, (27) Chupak, L. S.; Zheng, X. Preparation of compounds useful as
G. Secreted PCSK9 downregulates low density lipoprotein receptor immunomodulators. World Patent WO15034820A1, 2015.
through receptor-mediated endocytosis. J. Lipid Res. 2007, 48, 1488− (28) Tilley, J. W.; Chen, L.; Fry, D. C.; Emerson, S. D.; Powers, G.
1498. D.; Biondi, D.; Varnell, T.; Trilles, R.; Guthrie, R.; Mennona, F.;
(11) Robinson, J. G.; Farnier, M.; Krempf, M.; Bergeron, J.; Luc, G.;
Kaplan, G.; LeMahieu, R. A.; Carson, M.; Han, R.-J.; Liu, C. M.;
Averna, M.; Stroes, E. S.; Langslet, G.; Raal, F. J.; El Shahawy, M.;
Palermo, R.; Ju, G. Identification of a Small Molecule Inhibitor of the
Koren, M. J.; Lepor, N. E.; Lorenzato, C.; Pordy, R.; Chaudhari, U.;
IL-2/IL-2Rα Receptor Interaction Which Binds to IL-2. J. Am. Chem.
Kastelein, J. J. Efficacy and safety of alirocumab in reducing lipids and
Soc. 1997, 119, 7589−7590.
cardiovascular events. N. Engl. J. Med. 2015, 372, 1489−1499.
(29) Ding, K.; Lu, Y.; Nikolovska-Coleska, Z.; Wang, G.; Qiu, S.;
(12) Sabatine, M. S.; Giugliano, R. P.; Wiviott, S. D.; Raal, F. J.;
Shangary, S.; Gao, W.; Qin, D.; Stuckey, J.; Krajewski, K.; Roller, P.
Blom, D. J.; Robinson, J.; Ballantyne, C. M.; Somaratne, R.; Legg, J.;
P.; Wang, S. Structure-Based Design of Spiro-oxindoles as Potent,
Wasserman, S. M.; Scott, R.; Koren, M. J.; Stein, E. A. Efficacy and
safety of evolocumab in reducing lipids and cardiovascular events. N. Specific Small-Molecule Inhibitors of the MDM2-p53 Interaction. J.
Engl. J. Med. 2015, 372, 1500−1509. Med. Chem. 2006, 49, 3432−3435.
(13) Ray, K. K.; Corral, P.; Morales, E.; Nicholls, S. J. (30) Vassilev, L. T.; Vu, B. T.; Graves, B.; Carvajal, D.; Podlaski, F.;
Pharmacological lipid-modification therapies for prevention of Filipovic, Z.; Kong, N.; Kammlott, U.; Lukacs, C.; Klein, C.; Fotouhi,
ischaemic heart disease: current and future options. Lancet 2019, N.; Liu, E. A. In vivo activation of the p53 pathway by small-molecule
394, 697−708. antagonists of MDM2. Science 2004, 303, 844−848.
(14) Sabatine, M. S.; Giugliano, R. P.; Keech, A. C.; Honarpour, N.; (31) Bushweller, J. H. Targeting transcription factors in cancer 
Wiviott, S. D.; Murphy, S. A.; Kuder, J. F.; Wang, H.; Liu, T.; from undruggable to reality. Nat. Rev. Cancer 2019, 19, 611−624.
Wasserman, S. M.; Sever, P. S.; Pedersen, T. R. Evolocumab and (32) Souers, A. J.; Leverson, J. D.; Boghaert, E. R.; Ackler, S. L.;
Clinical Outcomes in Patients with Cardiovascular Disease. N. Engl. J. Catron, N. D.; Chen, J.; Dayton, B. D.; Ding, H.; Enschede, S. H.;
Med. 2017, 376, 1713−1722. Fairbrother, W. J.; Huang, D. C. S.; Hymowitz, S. G.; Jin, S.; Khaw, S.
(15) Schwartz, G. G.; Steg, P. G.; Szarek, M.; Bhatt, D. L.; Bittner, V. L.; Kovar, P. J.; Lam, L. T.; Lee, J.; Maecker, H. L.; Marsh, K. C.;
A.; Diaz, R.; Edelberg, J. M.; Goodman, S. G.; Hanotin, C.; Mason, K. D.; Mitten, M. J.; Nimmer, P. M.; Oleksijew, A.; Park, C.
Harrington, R. A.; Jukema, J. W.; Lecorps, G.; Mahaffey, K. W.; H.; Park, C.-M.; Phillips, D. C.; Roberts, A. W.; Sampath, D.;
Moryusef, A.; Pordy, R.; Quintero, K.; Roe, M. T.; Sasiela, W. J.; Seymour, J. F.; Smith, M. L.; Sullivan, G. M.; Tahir, S. K.; Tse, C.;
Tamby, J. F.; Tricoci, P.; White, H. D.; Zeiher, A. M. Alirocumab and Wendt, M. D.; Xiao, Y.; Xue, J. C.; Zhang, H.; Humerickhouse, R. A.;
Cardiovascular Outcomes after Acute Coronary Syndrome. N. Engl. J. Rosenberg, S. H.; Elmore, S. W. ABT-199, a potent and selective
Med. 2018, 379, 2097−2107. BCL-2 inhibitor, achieves antitumor activity while sparing platelets.
(16) Pandey, A. V.; Henderson, C. J.; Ishii, Y.; Kranendonk, M.; Nat. Med. 2013, 19, 202−208.
Backes, W. L.; Zanger, U. M. Editorial: Role of Protein-Protein (33) Paoluzzi, L.; Gonen, M.; Bhagat, G.; Furman, R. R.; Gardner, J.
Interactions in Metabolism: Genetics, Structure, Function. Front. R.; Scotto, L.; Gueorguiev, V. D.; Heaney, M. L.; Manova, K.;
Pharmacol. 2017, 8, 881−881. O’Connor, O. A. The BH3-only mimetic ABT-737 synergizes the
(17) Huang, Z. Structural chemistry and therapeutic intervention of antineoplastic activity of proteasome inhibitors in lymphoid
protein-protein interactions in immune response, human immunode- malignancies. Blood 2008, 112, 2906−2916.

5278 https://doi.org/10.1021/acs.jcim.1c00521
J. Chem. Inf. Model. 2021, 61, 5269−5279
Journal of Chemical Information and Modeling pubs.acs.org/jcim Article

(34) Kwon, H. J.; Lagace, T. A.; McNutt, M. C.; Horton, J. D.; (50) Kong, W.; Wei, J.; Abidi, P.; Lin, M.; Inaba, S.; Li, C.; Wang, Y.;
Deisenhofer, J. Molecular basis for LDL receptor recognition by Wang, Z.; Si, S.; Pan, H.; Wang, S.; Wu, J.; Wang, Y.; Li, Z.; Liu, J.;
PCSK9. Proc. Natl. Acad. Sci. U. S. A. 2008, 105, 1820−1825. Jiang, J.-D. Berberine is a novel cholesterol-lowering drug working
(35) Bottomley, M. J.; Cirillo, A.; Orsatti, L.; Ruggeri, L.; Fisher, T. through a unique mechanism distinct from statins. Nat. Med. 2004,
S.; Santoro, J. C.; Cummings, R. T.; Cubbon, R. M.; Lo Surdo, P.; 10, 1344−1351.
Calzetta, A.; Noto, A.; Baysarowich, J.; Mattu, M.; Talamo, F.; De (51) Cameron, J.; Ranheim, T.; Kulseth, M. A.; Leren, T. P.; Berge,
Francesco, R.; Sparrow, C. P.; Sitlani, A.; Carfi, A. Structural and K. E. Berberine decreases PCSK9 expression in HepG2 cells.
biochemical characterization of the wild type PCSK9-EGF(AB) Atherosclerosis 2008, 201, 266−273.
complex and natural familial hypercholesterolemia mutants. J. Biol.
Chem. 2009, 284, 1313−1323.
(36) Lo Surdo, P.; Bottomley, M. J.; Calzetta, A.; Settembre, E. C.;
Cirillo, A.; Pandit, S.; Ni, Y. G.; Hubbard, B.; Sitlani, A.; Carfi, A.
Mechanistic implications for LDL receptor degradation from the
PCSK9/LDLR structure at neutral pH. EMBO Rep. 2011, 12, 1300−
1305.
(37) Zhang, Y.; Eigenbrot, C.; Zhou, L.; Shia, S.; Li, W.; Quan, C.;
Tom, J.; Moran, P.; Di Lello, P.; Skelton, N. J.; Kong-Beltran, M.;
Peterson, A.; Kirchhofer, D. Identification of a small peptide that
inhibits PCSK9 protein binding to the low density lipoprotein
receptor. J. Biol. Chem. 2014, 289, 942−955.
(38) Min, D. K.; Lee, H. S.; Lee, N.; Lee, C. J.; Song, H. J.; Yang, G.
E.; Yoon, D.; Park, S. W. In silico Screening of Chemical Libraries to
Develop Inhibitors That Hamper the Interaction of PCSK9 with the
LDL Receptor. Yonsei Med. J. 2015, 56, 1251−1257.
(39) Taechalertpaisarn, J.; Zhao, B.; Liang, X.; Burgess, K. Small
Molecule Inhibitors of the PCSK9·LDLR Interaction. J. Am. Chem.
Soc. 2018, 140, 3242−3249.
(40) Lammi, C.; Sgrignani, J.; Arnoldi, A.; Lesma, G.; Spatti, C.;
Silvani, A.; Grazioso, G. Computationally Driven Structure
Optimization, Synthesis, and Biological Evaluation of Imidazole-
Based Proprotein Convertase Subtilisin/Kexin 9 (PCSK9) Inhibitors.
J. Med. Chem. 2019, 62, 6163−6174.
(41) Evison, B. J.; Palmer, J. T.; Lambert, G.; Treutlein, H.; Zeng, J.;
Nativel, B.; Chemello, K.; Zhu, Q.; Wang, J.; Teng, Y.; Tang, W.; Xu,
Y.; Rathi, A. K.; Kumar, S.; Suchowerska, A. K.; Parmar, J.; Dixon, I.;
Kelly, G. E.; Bonnar, J. A small molecule inhibitor of PCSK9 that
antagonizes LDL receptor binding via interaction with a cryptic
PCSK9 binding groove. Bioorg. Med. Chem. 2020, 28, 115344.
(42) Zhang, D. W.; Garuti, R.; Tang, W. J.; Cohen, J. C.; Hobbs, H.
H. Structural requirements for PCSK9-mediated degradation of the
low-density lipoprotein receptor. Proc. Natl. Acad. Sci. U. S. A. 2008,
105, 13045−13050.
(43) Chorba, J. S.; Shokat, K. M. The proprotein convertase
subtilisin/kexin type 9 (PCSK9) active site and cleavage sequence
differentially regulate protein secretion from proteolysis. J. Biol. Chem.
2014, 289, 29030−29043.
(44) Abdel-Meguid, S. S.; Elshourbagy, N.; Meyers, H.; Mousa, S. A.
Anti-proprotein convertase subtilisin kexin type 9 (anti-PCSK9)
compounds and methods of using the same in the treatment and/or
pr ev en ti on of cardi o va scu lar di se as es . W orl d P ate nt
WO2014150326A1, 2014.
(45) Abou-Gharbia, M.; Childers, W. E. PCSK9 inhibitors and
methods of use thereof. World Patent WO2016040305A1, 2016.
(46) Shin, W. H.; Christoffer, C. W.; Kihara, D. In silico structure-
based approaches to discover protein-protein interaction-targeting
drugs. Methods 2017, 131, 22−32.
(47) Balasubramanian, K.; Gupta, S. P. Quantum Molecular
Dynamics, Topological, Group Theoretical and Graph Theoretical
Studies of Protein-Protein Interactions. Curr. Top. Med. Chem. 2019,
19, 426−443.
(48) Liu, S.; Zhou, X.; Li, M.; Zhao, W.; Zhou, S.; Cheng, K.; Xu,
Q.; Chen, C.; Wen, X.; Sun, H.; Yuan, H. Discovery of Ubiquitin-
Specific Protease 7 (USP7) Inhibitors with Novel Scaffold Structures
by Virtual Screening, Molecular Dynamics Simulation, and Biological
Evaluation. J. Chem. Inf. Model. 2020, 60, 3255−3264.
(49) Campbell, C. T.; Kim, G. SPR microscopy and its applications
to high-throughput analyses of biomolecular binding events and their
kinetics. Biomaterials 2007, 28, 2380−2392.

5279 https://doi.org/10.1021/acs.jcim.1c00521
J. Chem. Inf. Model. 2021, 61, 5269−5279

You might also like