Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Atherosclerosis 330 (2021) 52–60

Contents lists available at ScienceDirect

Atherosclerosis
journal homepage: www.elsevier.com/locate/atherosclerosis

The emerging landscape of peptide-based inhibitors of PCSK9


Benjamin J. Tombling 1, Yuhui Zhang 1, Yen-Hua Huang , David J. Craik , Conan K. Wang *
Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland,
Brisbane, Qld, 4072, Australia

A R T I C L E I N F O A B S T R A C T

Keywords: Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a clinically validated target for treating cardiovascular
PCSK9 disease (CVD) due to its involvement in cholesterol metabolism. Although approved monoclonal antibodies
Peptides (alirocumab and evolocumab) that inhibit PCSK9 function are very effective in lowering cholesterol, their lim­
Combinatorial libraries
itations, including high treatment costs, have so far prohibited widespread use. Accordingly, there is great in­
Disulfide-rich peptides
Protein-protein interaction
terest in alternative drug modalities to antibodies. Like antibodies, peptides are valuable therapeutics due to their
high target potency and specificity. Furthermore, being smaller than antibodies means they have access to more
drug administration options, are less likely to induce adverse immunogenic responses, and are better suited to
affordable production. This review surveys the current peptide-based landscape aimed towards PCSK9 inhibition,
covering pre-clinical to patented drug candidates and comparing them to current cholesterol lowering thera­
peutics. Classes of peptides reported to be inhibitors include nature-inspired disulfide-rich peptides, combina­
torially derived cyclic peptides, and peptidomimetics. Their functional activities have been validated in
biophysical and cellular assays, and in some cases pre-clinical mouse models. Recent efforts report peptides with
potent sub-nanomolar binding affinities to PCSK9, which highlights their potential to achieve antibody-like
potency. Studies are beginning to address pharmacokinetic properties of PCSK9-targeting peptides in more
detail. We conclude by highlighting opportunities to investigate their biological effects in pre-clinical models of
cardiovascular disease. The anticipation concerning the PCSK9-targeting peptide landscape is accelerating and it
seems likely that a peptide-based therapeutic for treating PCSK9-mediated hypercholesterolemia may be clini­
cally available in the near future.

1. Introduction their inability to reduce LDL-C to desirable and safe levels for ∼ 50% of
patients [8,9], leaving patients exposed to an increased risk of CVD. The
Cardiovascular disease (CVD) is the leading cause of death globally limitations of statins have stimulated research towards discovering new
(17.9 million deaths per year; World Health Organization) and is a major therapeutics with different modes of action for cholesterol management.
burden on healthcare systems and economies [1,2]. Low density lipo­ In particular, one therapeutic avenue for cholesterol regulation that has
protein (LDL) is known to be a causal factor in the pathophysiology of achieved clinical validation is the development of therapeutics that
atherosclerotic CVD [3–5]. Plasma LDL burden is commonly estimated modulate proprotein convertase subtilisin/kexin type 9 (PCSK9).
by measuring plasma LDL cholesterol (LDL-C) levels, and so strategies In this review, we provide an overview of the molecular mechanism
that reduce circulating LDL-C have become a major focus for prevention driving PCSK9’s role in lipid modulation and introduce the therapeutic
of CVD events. modalities that are approved or under clinical development that hinder
To reduce LDL levels, patients are generally prescribed statins, for PCSK9 function. We focus on examining peptide inhibitors of PCSK9
example atorvastatin, which inhibit HMG-CoA reductase – the rate currently reported. Finally, we look to future experiments that will help
limiting enzyme involved in the cholesterol biosynthetic pathway. The progress the clinical development of peptides for managing LDL levels.
CVD benefits of statin medication are associated with their LDL-C
lowering action [6]. However, recent evidence has highlighted their
undesirable properties such as side effects [7] and more alarmingly,

* Corresponding author.
E-mail address: c.wang@imb.uq.edu.au (C.K. Wang).
1
These authors contributed equally to this work.

https://doi.org/10.1016/j.atherosclerosis.2021.06.903
Received 30 March 2021; Received in revised form 18 May 2021; Accepted 23 June 2021
Available online 26 June 2021
0021-9150/© 2021 Elsevier B.V. All rights reserved.
B.J. Tombling et al. Atherosclerosis 330 (2021) 52–60

2. PCSK9 role in lipid metabolism expression and cleavage of the signal peptide, PCSK9 undergoes intra­
cellular autocatalytic processing between Q152-S153 of the prodomain.
PCSK9 is a high-profile success story of modern medical genomics. Although cleaved, the prodomain remains non-covalently bound to the
Identification of the critical role of PCSK9 in lipid metabolism started in catalytic domain, preventing further catalytic activity by blocking the
2003, when PCSK9 was identified to be the third locus associated with catalytic site [29,30]. The mature PCSK9 heterodimeric complex is
the autosomal dominant disease familial hypercholesterolemia (FH) necessary for PCSK9’s cholesterol-regulatory function as it stabilizes the
[10]. FH affects ∼ 1:200 people globally [11] making it the most com­ active structure for efficient LDLR interaction and likely assists with
mon inherited metabolic disorder, but it is commonly underdiagnosed secreting PCSK9 into the extracellular space [31].
and undertreated in the general population, which accentuates its clin­ Structural studies have revealed the most dominant PCSK9:LDLR
ical severity [12]. FH is attributed to mutations in the protein machinery interaction (Fig. 2B) in the extracellular space is composed of a ∼ 500 Å2
responsible for cholesterol metabolism, which can limit a patient’s binding interface between the PCSK9 catalytic domain and the
ability to remove LDL-C from the blood [13,14]. Genetic studies iden­ epidermal growth factor-like repeat-A (EGF-A) domain of LDLR (Fig. 2C)
tified mutations in the PCSK9 gene that were associated with elevated [32–34]. EGF-A is a microdomain component of the EGF-precursor ho­
circulating LDL-C and were called gain-of-function (GOF) mutations. mology domain, which plays a pivotal role in LDLR recycling from the
Patients carrying these mutations are subject to an increased risk of endosomes to the hepatocyte surface [35]. The most prevalent and
encountering premature CVD events [15,16], suggesting PCSK9 might active PCSK9 GOF mutation, [D374Y]PCSK9, is directly involved in the
be a druggable target. In addition, PCSK9 loss-of-function (LOF) muta­ PCSK9:EGF-A interface and results in a ∼ 25-fold improved affinity for
tions have been shown to engender patients with significantly lower the LDLR at physiological conditions [36–38]. In addition, the LDLR
LDL-C whilst experiencing no adverse health issues [17–19], confirming GOF mutation [H306Y]LDLR is located on the EGF-A domain, and re­
that PCSK9 is not only an effective target but also that it can be safely sults in an improved PCSK9:LDLR interaction [38]. Both mutations are
inhibited. leading causes of severe FH [36,39] and highlight the clinical impor­
The cholesterol-regulatory biological mechanism of PCSK9 is now tance of the EGF-A interface for PCSK9 function. In summary, genetic,
well established. PCSK9 is a major regulator of hepatocyte surface LDL functional, and structural studies on PCSK9 have provided a basis for the
receptor (LDLR) levels by inhibiting the receptor recycling pathway development of targeted therapeutics.
[20–22]. LDLR function is pivotal to cholesterol homeostasis as it is
responsible for the cellular uptake and subsequent degradation of LDL 3. Therapeutic strategies to inhibit PCSK9
(see Fig. 1A) [23]. Circulating LDL binds to the N-terminal ligand
binding domain of LDLR via apolipoprotein B100, and the LDLR:LDL The approval of monoclonal antibodies that inhibit the PCSK9:EGF-A
complex becomes internalized by receptor-mediated endocytosis [24]. interaction was an exciting advance in the field of cardiovascular drug
After migrating to the endosome, the low pH environment allows LDLR development. It provided validation that inhibiting the PCSK9:EGF-A
to release LDL and recycle back to the cell surface. Disassociated LDL is interaction is an approach for silencing PCSK9 function and lowering
transported to the lysosome where it is degraded to provide cholesterol LDL levels (see Fig. 1C). Monoclonal antibodies can bind to the EGF-A
and amino acid building blocks to the cell. PCSK9 intervenes with site on the catalytic domain of PCSK9 with potent affinities [40]. Mul­
LDLR’s ability to recycle by binding to LDLR on hepatocyte surfaces (see tiple Phase III clinical trials have demonstrated that the FDA approved
Fig. 1B) [25]. A recent study exposed the ability of liver heparan sulfate monoclonal antibodies, evolocumab and alirocumab, can lower circu­
proteoglycans to capture PCSK9 and present it in a favorable confor­ lating LDL-C by ∼ 60% and achieve significant reductions in the
mation for LDLR interaction [26]. After migration of the PCSK9:LDLR occurrence of CVD events when administered in combination with sta­
complex to the acidic endosomal compartment through clathrin-coated tins [41–43]. Furthermore, the antibodies reduce lipoprotein (a) (Lp [a])
pits, conformational changes to LDLR result in the formation of addi­ and triglyceride levels, which are both risk factors for CVD, highlighting
tional binding sites with PCSK9 [27]. As a result, PCSK9 chaperones the benefits of targeting PCSK9 for improving the overall lipid profile
LDLR to the lysosome for degradation, preventing LDLR recycling and [44].
effectively up-regulating circulating LDL. Despite the enthusiasm following the approval of evolocumab and
PCSK9 is a member of the proprotein convertase family and is pre­ alirocumab, these antibodies have seen limited clinical application
dominately synthesized in the liver as a ∼ 73 kDa zymogen consisting of because their high treatment costs financially prohibit widespread
a signal peptide (M1-A30), prodomain (Q31-Q152), catalytic domain administration [45,46]. For example, in Australia the antibodies cost
(S153-Q454) and C-terminal domain (L455-Q692) (Fig. 2A) [28]. After between A$7000 and A$8000 per year which exceeds the general cost

Fig. 1. PCSK9 modulates LDLR and is a thera­


peutic target for lowering LDL.
(A) LDL is removed from plasma circulation by
LDLR in a receptor-mediated endocytosis pathway.
(B) PCSK9 downregulates LDLR by binding to the
receptor at the cell surface, preventing receptor
recycling and leading the receptor to be trans­
ported to the lysosome for degradation and PCSK9
would be also degraded in lysosome with it. (C)
Therapeutic modalities that block the PCSK9:LDLR
interaction are a validated approach to restore re­
ceptor function and lower circulating LDL levels.

53
B.J. Tombling et al. Atherosclerosis 330 (2021) 52–60

Fig. 2. PCSK9 binds to the EGF-A domain of LDLR.


(A) PCSK9 consists of a signal peptide, N-terminal
prodomain (pink), catalytic domain (grey) and C-
terminal domain (blue) (UniProt Q8NBP7).
Autocatalytic cleavage between Q152–S153 results
in the prodomain non-covalently binding to the
catalytic domain. The naturally occurring mutants
of PCSK9 that are associated with FH are high­
lighted in their respective domains. Residues in
bold are involved in the PCSK9:EGF-A interface.
(B) The dominant PCSK9:LDLR interaction site
(highlighted in yellow) at the cell surface is be­
tween the catalytic domain of PCSK9 and the EGF-
A domain of LDLR (orange) (PDB 3P5C). (C) The
interaction interface (yellow) between PCSK9 and
EGF-A. (For interpretation of the references to
color in this figure legend, the reader is referred to
the Web version of this article.)

effectiveness threshold for therapeutics [47,48]. This economic factor is that are intermediate in size between small molecules and biologics
probably partly responsible for the early discontinuation rate of (Fig. 3) [69,70]. This interest is demonstrated by the elevated growth of
PCSK9-mediated therapy in the clinic [49]. The failure of these anti­ the peptide therapeutic market [71].
bodies to dominate the PCSK9 inhibition market has ultimately left the Given the nature of PCSK9’s physiology and function, there are many
door open for alternative drug modalities. For example, small molecules reasons why peptides are an ideal drug modality for its targeted inhi­
[50–54], small interfering RNA inhibitors [55,56], bition. The PCSK9:EGF-A interface is flat and featureless which makes
monoclonal-antibody mimetics [57,58], nucleic acid polymers [59], designing small molecule inhibitors with sufficient potency very chal­
vaccines [60,61], and gene-silencing CRISPR-Cas9 technology [62] are lenging (as illustrated in Fig. 3A). Furthermore, the relatively low mo­
all at different stages of clinical development [63]. Notably, inclisiran, lecular weight of peptides, compared to antibodies, means they can be
which is a double-stranded small RNA molecule therapeutic that acts by manufactured more cheaply than antibodies and they do not require
blocking the transcription of PCSK9, has been approved recently in cold chain management. The ability to engineer peptides to enhance oral
Europe in December 2020 [64]. In addition to these therapeutic in­ bioavailability is beneficial for the long-term treatment of chronic dis­
terventions, peptides have attracted substantial interest as more acces­ eases such as FH due to higher patient compliance [72]. Finally, peptides
sible PCSK9 antagonists for next-generation cholesterol-lowering are likely to have predictable safety profiles based on the monoclonal
therapy. antibody outcomes and their shared mechanism of action; however, this
idea will need to be validated in pre-clinical and clinical studies. In
addition to the advantageous properties of peptides, PCSK9’s extracel­
3.1. Peptides as PCSK9-targeted therapeutics
lular mechanism of action avoids the need for inhibitors to possess
cell-penetrating properties, which can sometimes be challenging from a
Peptides have advantageous therapeutic properties that make them
peptide drug-design perspective. In the following sections, we will re­
valuable drug leads. First, peptides can achieve high target specificity
view literature that provide evidence for the therapeutic potential of
[65]. Second, their ability to mimic complex protein structures means
peptides in the PCSK9 drug development landscape.
they can modulate protein–protein interfaces of ‘undruggable’ targets
[65,66]. Third, the smaller size of peptides engenders them with
4. Peptide inhibitors of the PCSK9:LDLR interaction
drug-like properties that are not associated with larger proteins, such as
membrane permeability [67] and oral activity [68]. Their smaller size
Fig. 4 shows that efforts are accelerating towards developing
also allows for facile functionalization using a wide range of chemical
peptide-based PCSK9 inhibitors for therapeutic use. Since 2008, hun­
design strategies that enables a detailed structure-activity relationship
dreds of peptides that inhibit PCSK9 function have been reported in the
(SAR) profile to be established. Altogether, these qualities have moti­
literature or in patents. Notable candidates are highlighted in Table 1,
vated the increased interest regarding peptides as therapeutic modalities

Fig. 3. Peptides have potential as new modalities


for inhibiting PCSK9.
There are three main modalities which have been
considered for disrupting PCSK9 function: small
molecules (grey), peptides (blue) and large bi­
ologics (green, PDB 3H42). Although small mole­
cules generally have good drug-like properties,
they are not suited for targeting the flat and
featureless interface between PCSK9 and EGF-A
(PCSK9 in grey; interface in yellow). Large bi­
ologics have potential for specificity but have
encountered several limitations that have stalled
their clinical use. Peptides could fill the gap be­
tween small molecules and large biologics because
they have the potential to harness the benefits of both small molecules and antibodies. (For interpretation of the references to color in this figure legend, the reader is
referred to the Web version of this article.)

54
B.J. Tombling et al. Atherosclerosis 330 (2021) 52–60

Fig. 4. Development of PCSK9-targeted


peptide inhibitors have attracted increasing
interest. Since the EGF-A peptide was
discovered to inhibit PCSK9 in 2008,
worldwide interest in the development of
peptide-based drugs targeting PCSK9 has
grown. This timeline highlights some of the
key discoveries over that period. These
peptides can be categorized into three clas­
ses: nature-derived peptides (blue) [74–77,
92,93], combinatorially derived peptides
(orange) [80–84], and peptidomimetics
(purple) [85,86,101]. Three-dimensional
structures of selected peptides are shown
and colored according to their assigned
class. Disulfide bonds are shown in yellow.
Arrows connecting structures provide an
indication of the progression of initial gen­
eration peptides to more recent ones with
improved therapeutic properties, such as
enhanced activity (e.g. tEGF-A to
TEX_S2-03). (For interpretation of the refer­
ences to color in this figure legend, the
reader is referred to the Web version of this
article.)

Table 1
Published peptides that inhibit the PCSK9:LDLR interaction.
Peptide Sequencea Activity (IC50; nM) b Interface area (Å2) e Ref.
c
LDLR 697 aa (UniProt P01130) 600–750 ∼ 506–525 [34,73]
Nature-derived
EGF-A GTNECLDNNGGCSHVCNDLKIGYECLCPDGFQLVAQRRCE 3000 ∼ 515 [74]
EGF66 GTNECLANLGGCSHICRKLKIGYECLCPDGFQLVAQRRCE 3 – [75]
(tEGF-A) GTNECLDNNGGCSHVCNDLKIGYECL 16000 ∼ 515 [76]
TEX-S3_01 GTNECLSSKYPWEPYWNGGCSHVCNDLKIGYECL 600 – [77]
P5 LILPKHSDAD 2000 – [78]
T9 GQEQSHQDEGVIVR 320000 – [78]
d
T9D8A_1 GQRQWKQAEGVMVR 150000 – [79]
Combinatorially derived
Pep2-8 TVFTSWEEYLDWV 800 ∼ 397 [80]
Fusion 1 TVFTSWEEYLDWVGSGCRLPWNLQRIGLPC 20 ∼ 844 [81]
Groove binding MESFPGWNLV (hR)IGLLR 30000 ∼ 674 [81]
78 Bicyclic peptide 1 ∼ 474 [82]
P9-38 CTVFTSWEEYLDWNNLHPRNSC 20 – [83]
Tetra-P9-38 4 x P9-38 (functionalized dendrimer) 0.2 – [84]
Peptidomimetics
RIm13 Polyimidazole scaffold with functional sidechains 2000 – [85]
Compound 15 ––WNLK (hR)IsLLR 750 ∼ 635 [86]

a
= 1-amino-4-phenylcyclohexane-1-carbony; hR = homo-Arg; s = D-Ser.
b
Ability to inhibit the PCSK9:LDLR interaction.
c
dissociation constant (KD).
d
Inhibitory activity against [D374Y]PCSK9:LDLR interaction.
e
Determined from analysis of the peptide:PCSK9 crystal structures using the PDB ePISA tool.

which also shows their respective inhibition activities. For ease of structures of bound inhibitors and are predominantly non-proteinogenic
reference, we have divided PCSK9 peptide inhibitors into three main in composition.
classes: nature-derived peptides, combinatorially derived peptides, and
peptidomimetics. As described in more detail below, we regard nature-
derived peptides as those that originate from natural sources (e.g. 4.1. Nature-derived peptides
human or plants); for example EGF-A is from the human LDLR, and P5 is
from plant lupin. Combinatorially derived peptides include peptides 4.1.1. EGF-A-derived peptides
discovered from phage, mRNA or other display technologies. Peptido­ Following the discovery that the EGF-A domain of LDLR is the pri­
mimetics refer to those that were computationally designed to mimic mary binding site for PCSK9 [32], EGF-A-derived peptides were of in­
terest towards developing PCSK9 antagonists. Under physiological

55
B.J. Tombling et al. Atherosclerosis 330 (2021) 52–60

conditions, the isolated EGF-A domain displays similar affinity for epithelial barrier, suggesting they may display oral bioavailability. It
PCSK9 compared to full-length LDLR (KD of 1 μM and 0.7 μM, respec­ would be interesting to further evaluate the in vivo activity of these
tively) [34,73] and binds to PCSK9 in a calcium- and pH-dependent peptides. The activity and cell permeability of lupin-derived peptides
manner [74]. Furthermore, EGF-A showed functional activity by pre­ suggest they are promising starting points for further optimization.
venting PCSK9-mediated LDLR degradation which led to increased LDL
uptake in liver cells. 4.1.3. Peptides derived from PCSK9
Zhang et al. extended these findings and discovered EGF-A analogues Most studies focusing on inhibiting the PCSK9:LDLR interactions
with improved affinity for PCSK9 using phage display [75]. Phage have designed compounds that bind to PCSK9. However, Alghamdi et al.
display is a powerful affinity selection technique used to discover investigated an interesting alternative – targeting LDLR as a pathway to
bioactive peptides towards a protein target of interest. A phage library achieve PCSK9 inhibition [93]. They designed a panel of peptides
based on the EGF-A scaffold was constructed, where PCSK9 binding derived from overlapping segments of the PCSK9 catalytic domain and
residues (N295, D299, N301, H306, V307, N309, and D310 – LDLR showed the peptide named CP3 (which consisted of residues
residue numbering) were randomized, and panned against human G365–G384) displayed μM activity in restoring cell surface LDLR levels.
PCSK9. The most potent EGF-A analogue, EGF66, inhibited the PCSK9: Future designs could involve incorporating the D374Y mutation to
LDLR interaction with an IC50 of 3 nM, substantially improved from the improve activity. Overall, that study suggested blocking the PCSK9
μM activity of native EGF-A (see Table 1). However, due to synthesis binding site on LDLR can achieve PCSK9 inhibition, but further studies
difficulties, EGF66 was fused to fragment crystallizable (Fc) domains for are required to validate the efficacy of this approach as the LDLR might
further binding and activity analysis. EGF66-Fc restored LDLR surface be involved in other biological functions.
levels with 10-fold stronger activity than native EGF-A. The improved in
vitro antagonistic activity of EGF66-Fc translated to more potent in vivo 4.2. Combinatorially-derived peptides
activity as LDLR levels were fully restored in mice livers when intrave­
nously administered at 60 mg/kg whereas EGF-A-Fc only partially 4.2.1. Pep2-8-derived peptides
restored LDLR levels. Combinatorial library screening of randomized peptide sequences
A different approach to engineering EGF-A has been investigated, in has provided a foundation for de novo identification of non-EGF-A based
which EGF-A was truncated between L318-C319, thereby removing the peptide inhibitors of PCSK9. Zhang et al. used a series of randomized
C-terminal region. The truncated analogue, tEGF-A, showed a similar linear peptide phage libraries to identify the 13-residue PCSK9 antago­
structure, affinity, and activity to full length EGF-A [76]. Furthermore, nist Pep2-8 (see Table 1) [80]. In vitro studies showed that Pep2-8 had
tEGF-A was mutated to incorporate the LDLR GOF mutation H306Y, moderate affinity for PCSK9 (KD of 0.7 μM) and competitively bound to
which showed improved affinity for PCSK9 in vitro, but did not lead to the EGF-A binding site (IC50 of 0.8 μM). Furthermore, Pep2-8 showed
increased LDLR function in cell-based assays. A separate recent study good selectivity against other members of the proprotein convertase
showed full length EGF-A and tEGF-A to be both active against the family, including furin, PC1, PC2, PC4 and PC9. Cell-based functional
FH-associated [D374Y]PCSK9 [87]. Although the authors claimed that assays confirmed the ability of Pep2-8 to reduce LDLR degradation and
the EGF-A peptides showed similar efficacy to the approved PCSK9 in­ promote LDL internalization in the presence of PCSK9. The Pep2-8:
hibitor alirocumab, this perhaps requires further validation given the PCSK9 crystal structure showed that Pep2-8 forms a ß-strand-turn-he­
sub nanomolar affinity of alirocumab compared to μM affinity of EGF-A lix motif, with the N-terminal β-strand mimicking EGF-A binding.
[88]. Despite Pep2-8 providing proof-of-concept that small peptide-based
We recently showed that EGF-like domains have high sequence and inhibitors of PCSK9 were achievable, limitations such as its poor solu­
segment length variability, despite their conserved Cys frameworks and bility and moderate activity have motivated interest in the design of
overall topologies [89]. The plasticity of the EGF scaffold inspired us to improved therapeutic leads. Computationally-derived analogues of
re-engineer tEGF-A by extending its segments that are directly involved Pep2-8 were reported to display improved antagonistic activity, but
in PCSK9 interaction [77]. The most potent extended tEGF-A analogue biophysical binding data were not presented [94].
identified from phage library screening, TEX-S2_03, displayed A cryptic binding pocket located near the Pep2-8 binding site on
>100-fold improved affinity for PCSK9 compared to parent tEGF-A, PCSK9 has been exploited for therapeutic development [81]. The au­
which translated to improved functional activity in cell-based assays. thors showed that the P’ helix at the N-terminus of the catalytic domain
The reason for TEX-S2_03’s improved affinity has not yet been fully of PCSK9 (S153–R167) is intrinsically flexible; alternating from a bound
understood, but binding analysis suggested the extended segment may conformation on the PCSK9 surface to being extended away in an open
occupy an additional binding site which could be exploited after further conformation which exposes a binding pocket. Using a phage library
SAR. that contained a randomized peptide motif extended off the C-terminus
of Pep2-8 led to the discovery of fusion peptides that displayed potent
4.1.2. Lupin protein-derived peptides bivalent affinity (KD of 2–6 nM) and antagonistic activity (see Table 1).
Some enzyme-digested fragments from food-derived proteins have The fusion peptides were truncated to result in peptides that solely
the ability to inhibit the PCSK9:LDLR interaction. Two peptides isolated bound to the cryptic binding site and displayed moderate PCSK9
from pepsin and trypsin cleavage of lupin, called P5 and T9, competi­ antagonistic activity (IC50 = 30 μM).
tively bound to PCSK9 with moderate μM activity and were able to To improve the affinity and inhibitory function of Pep2-8, we
restore cholesterol uptake (Table 1) [78]. P5 displayed the highest ac­ introduced a design strategy termed bioactive cyclization [83]. A func­
tivity (IC50 of 2 μM). In addition, the lupin peptides were tested against tional cyclization linker was incorporated onto the Pep2-8 binding motif
the GOF [D374Y]PCSK9 mutant to test their application as treatments that was composed of experimentally optimized amino acids that spe­
for FH. Interestingly, the most active compound against wild-type cifically participate in favorable interactions with PCSK9. The increased
PCSK9, P5, was inactive against [D374Y]PCSK9 [90]. On the contrary, binding face achieved by the cyclic peptide identified using this
T9 had slightly improved activity for the FH associated PCSK9 mutant, approach, P9-38, resulted in >100-fold improved affinity compared to
inhibiting the [D374Y]PCSK9:LDLR interaction with an IC50 of 280 μM Pep2-8 and demonstrated significant cell-based functional activity at
versus 320 μM for wild-type PCSK9. Optimization of T9 by computa­ low nanomolar concentrations.
tional design (T9D8A_1) achieved a ∼ 2-fold improvement in [D374Y] Recently we used dendrimer scaffolds (branched polyvalent struc­
PCSK9 antagonistic activity and restored cellular LDLR function more tures that can be chemically functionalized to display multiple copies of
efficiently [79]. Studies indicated the lupin peptides can cross Caco-2 a bioactive compound) to further optimize the activity of the peptide P9-
cell monolayers [91,92], which is an established model of the intestinal 38 by increasing valency, which we hypothesized would increase the

56
B.J. Tombling et al. Atherosclerosis 330 (2021) 52–60

local effective concentration at the PCSK9 binding site [84]. Bivalent the subject of further structure-activity guided optimization, which
and tetravalent analogues of P9-38 inhibited the PCSK9:LDLR interac­ identified a bipartite molecule consisting of a cryptic binding peptide
tion with ~60-fold (IC50 = 0.3 nM) and ~100-fold (IC50 = 0.2 nM) with a N-terminal organic 1-amino-4-phenylcyclohexane-1-carbonyl
improvement over the parent peptide, respectively. The moiety [86]. The functionalized N-terminus mimicked key hydropho­
P9-38-decorated dendrimers are two of the most potent peptide-based bic interactions adopted by EGF-A and led to significant improvements
PCSK9:LDLR interaction inhibitors reported to date. in antagonistic activity over the parent peptide (IC50 = 750 nM;
Table 1). Unfortunately, attempts to truncate the peptide region to
4.2.2. Cyclic peptide inhibitors of PCSK9 design inhibitors with low molecular weight were not successful.
Peptides that adopt a backbone cyclic structure are often attributed However, further optimization of peptide-based compounds targeting
with therapeutically advantageous properties such as a reduced entropic the cryptic binding site of PCSK9 are ongoing and it is hoped this
penalty upon target binding and increased protection against proteolytic strategy will lead to inhibitors suited to oral delivery [102].
degradation [95,96]. As a result of these advantageous properties, cyclic
peptides are arguably the most likely peptide class to achieve approval 5. Alternative roles of PCSK9 beyond LDLR regulation
for clinical use against PCSK9. Recent years have witnessed a surge in
filed patents describing the invention of potent cyclic peptide inhibitors 5.1. PCSK9’s role in atherosclerosis beyond lipid homeostasis
of PCSK9. Two patents from RA Pharmaceuticals and Merck (WO
2019/246387 A1 and WO 2019/246405 A1) describe >100 cyclic PCSK9 has been found to accelerate the formation of atherosclerosis
polypeptides that inhibit the PCSK9:LDLR interaction with IC50 values in independently of lipid regulation [103]. For example, PCSK9 is linked to
the low nanomolar range [97,98]. pathways that affect the integrity of endothelial cell monolayers and
The SAR studies of peptides reported by Merck were detailed in a lead to advanced atherosclerosis [104,105]. Levels of PCSK9 have also
recent paper and underpinned extensive efforts to optimize a mRNA been associated with increased proliferation rate of smooth muscles
library derived hit peptide into smaller, highly stable and activity cells, which is a contributing factor to atherosclerosis [104]. In mono­
enhanced lead compounds [82]. Although >1000 improvement in ac­ cytes and macrophages, PCSK9 increases the expression of cytokines like
tivity was observed in some cases (e.g. compound 78 with Ki of 1 nM), IL-6, TNF-α, CXCL2, which advance the pro-inflammatory state [106].
the authors identified undesirable off-target effects on mast cell The link between PCSK9 and inflammation is further supported by the
degranulation of some of their compounds. However, they showed the effect of PCSK9 monoclonal antibodies, alirocumab and evolocumab, on
side-effect was specific to chemical composition and could be reme­ reducing inflammation and increasing the synthesis of the
diated through specific amino acid substitutions, suggesting this issue anti-inflammatory factor IL-10 [107].
might be specific to this study rather than being a broader concern for
peptide inhibitors of PCSK9. That study also provided important insight 5.2. PCSK9 is a promising therapeutic target for other diseases
into the pharmacokinetic properties of PCSK9-targeted peptides, with
several peptides having been evaluated in mice, rats, or monkeys. The Following its causal relationship with FH and CVD, PCSK9 has been
peptides tested had short half-lives after intravenous administration, extensively studied to assess whether it is a genetic marker for other
with half-lives ranging from 0.25 to 1.60 h in mice and 0.06–2.46 h in diseases. A mendelian randomized study showed that patients with
rats. A bicyclic peptide, compound 75, had a half-life of 2.84 h in PCSK9 LOF mutations were associated with an increased risk of type 2
monkey. These results point towards the need to consider strategies to diabetes [108]. So far, clinical trials investigating the PCSK9 monoclonal
improve in vivo half-lives. antibody inhibitors have reported no significant increase in the risk of
Two patents from Novartis described novel cyclic peptide inhibitors new-onset diabetes [109]; however, long-term follow up studies are
of PCSK9. One of the patents (WO 2020/110011 A1) describes the in­ required to fully understand the effects of PCSK9 inhibition therapy. In
vention of 13-mer cyclic peptide thioesters, with the peptide P2 dis­ addition, there is overwhelming experimental and clinical evidence
playing high antagonistic activity (IC50 of 40 pM) [99]. The second indicating increased plasma levels of PCSK9 leads to systemic inflam­
patent (WO 2020/110009 A1) details a suite of cyclic tetramer com­ mation, and PCSK9 inhibitors can ameliorate pro-inflammatory effects
pounds with a peptide-like backbone functionalized with a broad range [110].
of non-natural amino acid side chains [100]. The most potent cyclic A recent study proposed PCSK9 inhibition is a safe and efficacious
tetramer, P3, achieved impressive PCSK9 antagonistic activity (IC50 of strategy for treating dengue fever [111]. This high-profile infectious
150 pM) and its low molecular weight (918 Da) is advantageous from an disease affects ∼ 400 million people each year, highlighting an impor­
oral delivery perspective. Overall, these patents demonstrate a growing tant new use for PCSK9 inhibition. The PCSK9 monoclonal antibody
interest from pharmaceutical companies towards the development of inhibitor evolocumab is currently undergoing testing in a clinical trial to
peptide-based PCSK9 inhibitors and suggest peptides will play a pivotal prevent cardiac allograft vasculopathy – a major cause of mortality after
role in next-generation cholesterol-lowering therapy. heart transplant surgery [112]. Finally, PCSK9 has recently been shown
to be a promising target to enhance immune checkpoint therapy for
4.3. Peptidomimetics treating cancer [113]. Overall, the volume of research highlighting
several health benefits of targeting PCSK9 is exciting and further studies
The interaction of EGF-A and Pep2-8 with PCSK9 highlights that may only accentuate the clinical importance of developing PCSK9
efficient PCSK9 interaction can be mediated by formation of ordered inhibitors.
structural motifs, specifically β-strands, by the inhibitor peptide. This
inspired research functionalizing poly-imidazole scaffolds to mimic 6. Summary and outlook
peptide secondary β-sheet structures and develop PCSK9 antagonists
that compete with EGF-A [85,101]. The most active compound identi­ There is now clear support for the therapeutic potential of peptides as
fied so far, RIm13 which is an analogue of MeIm, inhibits the PCSK9: drugs targeting PCSK9, with over one dozen studies reporting a diverse
LDLR interaction with a reported IC50 of 1 μM, suggesting range of peptide-based inhibitors of PCSK9. Collectively, these studies
poly-imidazole scaffolds are promising templates for PCSK9-targeted have demonstrated that peptides can inhibit the PCSK9:LDLR interface
drug design. Future studies aimed at optimizing the scaffold functional with high potency in the pM–nM range [75,81–84] and have potential
groups to strengthen interactions with PCSK9 may yield analogues with for specificity [74], oral delivery [92], and in vivo efficacy [75]. With the
improved activity. promising results reported so far, why have peptides not seen greater
The groove-binding peptide (compound 15) highlighted above was clinical investigation? Perhaps it is because the focus has been too

57
B.J. Tombling et al. Atherosclerosis 330 (2021) 52–60

strongly on discovery of new leads with binding affinity to PCSK9, and and by access to the facilities of the Australian Research Council Centre
not enough on other properties required of drugs. All studies have of Excellence for Innovations in Peptide and Protein Science
provided in vitro characterization of binding and/or competitive activity (CE200100012).
in biochemical, biophysical or cellular assays. However, few have
explored questions on specificity [74,82], cell permeability [92], in vivo CRediT authorship contribution statement
bioavailability [82], and in vivo activity [75]. Only peptides discovered
from mRNA display [82] and an EGF-A analogue as a Fc-fusion protein Benjamin J. Tombling: Conceptualization, Writing – original draft,
[75] have been tested in vivo. The gap in knowledge on aspects beyond in Visualization. Yuhui Zhang: Writing – original draft, Visualization.
vitro activity might be associated with some of the general drawbacks of Yen-Hua Huang: Visualization, Supervision, Writing – review & edit­
peptides. Fortunately, there are clinically validated peptide design ing. David J. Craik: Supervision, Writing – review & editing, Funding
strategies that can help propel peptides to the clinic. acquisition. Conan K. Wang: Supervision, Project administration,
Increasing the short circulating half-life of peptides perhaps repre­ Funding acquisition, Writing – review & editing.
sents the most important challenge that needs to be overcome for pep­
tides to be suitable therapeutics for targeting PCSK9. The small size of Declaration of competing interest
peptides means they are subject to rapid renal clearance. Indeed, small
cyclic peptide inhibitors of PCSK9 were shown to be rapidly cleared in The authors declare that they have no known competing financial
mice [82]. One validated strategy to improve the half-life of therapeutic interests or personal relationships that could have appeared to influence
peptides both in vitro and in vivo is to fuse the bioactive peptide to tags the work reported in this paper.
that confer abundant serum protein binding, such as albumin
[114–117]. For example, the approved glucagon-like peptide-1 receptor References
agonists liraglutide and semaglutide are engineered with albumin
binding tags and subsequently require once-weekly subcutaneous in­ [1] N. Townsend, L. Wilson, P. Bhatnagar, K. Wickramasinghe, M. Rayner, et al.,
Cardiovascular disease in Europe: epidemiological update 2016, Eur. Heart J. 37
jection for the treatment of type 2 diabetes and weight management
(2016) 3232–3245.
[118,119]. Increasing drug circulation time and accumulation of [2] G.A. Roth, C. Johnson, A. Abajobir, F. Abd-Allah, S.F. Abera, et al., Global,
PCSK9-targeting peptides would increase their in vivo efficacy and regional, and national burden of cardiovascular diseases for 10 causes, 1990 to
2015, J. Am. Coll. Cardiol. 70 (2017) 1–25.
reduce the dosing frequency required to invoke a cholesterol-reducing
[3] B.A. Ference, W. Yoo, I. Alesh, N. Mahajan, K.K. Mirowska, et al., Effect of long-
response. term exposure to lower low-density lipoprotein cholesterol beginning early in life
A major advantage of peptides over subcutaneously administered on the risk of coronary heart disease: a mendelian randomization analysis, J. Am.
monoclonal antibodies is their potential to be engineered for effective Coll. Cardiol. 60 (2012) 2631–2639.
[4] J.L. Goldstein, M.S. Brown, A century of cholesterol and coronaries: from plaques
oral delivery. There are several examples where approved peptide to genes to statins, Cell 161 (2015) 161–172.
therapeutics are orally dosed, including the immunosuppressant cyclo­ [5] B.A. Ference, H.N. Ginsberg, I. Graham, K.K. Ray, C.J. Packard, et al., Low-
sporine [120]. However, the majority of bioactive peptides do not density lipoproteins cause atherosclerotic cardiovascular disease. 1. Evidence
from genetic, epidemiologic, and clinical studies. A consensus statement from the
achieve sufficient levels of oral bioavailability to allow safe and effective European Atherosclerosis Society Consensus Panel, Eur. Heart J. 38 (2017)
oral administration. Instead, optimizing the formulation can be used to 2459–2472.
achieve oral activity through the use of permeability enhancers [121]. [6] C. Labos, J.M. Brophy, G.D. Smith, A.D. Sniderman, G. Thanassoulis, Evaluation
of the pleiotropic effects of statins, Arterioscler. Thromb. Vasc. Biol. 38 (2018)
Little work has been done in this field for peptide inhibitors of PCSK9. 262–265.
By addressing the in vivo limitations of peptides there might be [7] D.H. Fitchett, R.A. Hegele, S. Verma, Statin intolerance, Circulation 131 (2015)
greater interest in in vivo efficacy characterization, which has been 389–391.
[8] R.K. Akyea, J. Kai, N. Qureshi, B. Iyen, S.F. Weng, Sub-optimal cholesterol
dramatically underexplored. Whilst functional assays using model
response to initiation of statins and future risk of cardiovascular disease, Heart
human liver cell lines, such as HepG2, have provided an excellent and 105 (2019) 975–981.
reliable experimental platform for pre-clinical activity validation, the [9] L.S. Rallidis, The changing landscape of lipid-lowering therapy after the new
ESC/EAS guidelines for the management of dyslipidaemias: launching the era of
focus needs to progress towards in vivo efficacy characterization. Except
triple hypolipidaemic therapy in very high risk patients, Atherosclerosis 292
for EGF analogues fused to an Fc domain [75], to the best of our (2020) 231–233.
knowledge no other peptide-based inhibitor has been reported with in [10] M. Abifadel, M. Varret, J.-P. Rabès, D. Allard, K. Ouguerram, et al., Mutations in
vivo activity to date. And even that study focused on investigating the PCSK9 cause autosomal dominant hypercholesterolemia, Nat. Genet. 34 (2003)
154–156.
regulation of LDLR on liver cells rather than broader atherosclerosis [11] S. Singh, V. Bittner, Familial hypercholesterolemia—epidemiology, diagnosis,
disease. With the positive results observed for many PCSK9-peptide in­ and screening, Curr. Atherosclerosis Rep. 17 (2015) 3.
hibitors in vitro, it would be interesting to test PCSK9-peptide inhibitors [12] B.G. Nordestgaard, M.J. Chapman, S.E. Humphries, H.N. Ginsberg, L. Masana, et
al., Familial hypercholesterolaemia is underdiagnosed and undertreated in the
in animal models of atherosclerosis or related metabolic diseases [122, general population: guidance for clinicians to prevent coronary heart disease :
123] as mono or combination therapies. The effects of PCSK9 are consensus Statement of the European Atherosclerosis Society, Eur. Heart J. 34
amplified by statin treatment which induces increased activity of the (2013) 3478–3490.
[13] H.H. Hobbs, M.S. Brown, J.L. Goldstein, Molecular genetics of the LDL receptor
sterol regulatory element binding protein 2 (SREBP-2), resulting in gene in familial hypercholesterolemia, Hum. Mutat. 1 (1992) 445–466.
increased PCSK9 expression and secretion [124,125]. Therefore, there [14] J.L. Goldstein, H.H. Hobbs, M.S. Brown, Familial Hypercholesterolemia, in: A.
are potential benefits to applying synergistic approaches to achieve L. Beaudet, B. Vogelstein, K.W. Kinzler, et al. (Eds.), The Online Metabolic and
Molecular Bases of Inherited Disease, The McGraw-Hill Companies, Inc., New
effective cholesterol-lowering therapy.
York, NY, 2014.
Inhibition of PCSK9 binding to LDLR is clinically validated. In this [15] K.M. Timms, S. Wagner, M.E. Samuels, K. Forbey, H. Goldfine, et al., A mutation
review, we focused on the rapid growth of the discovery of competitive in PCSK9 causing autosomal-dominant hypercholesterolemia in a Utah pedigree,
Hum. Genet. 114 (2004) 349–353.
peptide-based ligands of PCSK9, demonstrating the potential of peptides
[16] M. Abifadel, J.-P. Rabès, M. Devillers, A. Munnich, D. Erlich, et al., Mutations and
as drugs. In our opinion peptides have been underutilized for the polymorphisms in the proprotein convertase subtilisin kexin 9 (PCSK9) gene in
treatment of hypercholesterolemia. A shift in focusing on optimizing the cholesterol metabolism and disease, Hum. Mutat. 30 (2009) 520–529.
pharmacokinetics of peptides might be key to their clinical progression. [17] J. Cohen, A. Pertsemlidis, I.K. Kotowski, R. Graham, C.K. Garcia, et al., Low LDL
cholesterol in individuals of African descent resulting from frequent nonsense
mutations in PCSK9, Nat. Genet. 37 (2005) 161–165.
Financial support [18] J.C. Cohen, E. Boerwinkle, T.H. Mosley, H.H. Hobbs, Sequence variations in
PCSK9, low LDL, and protection against coronary heart disease, N. Engl. J. Med.
354 (2006) 1264–1272.
Work in our laboratory on PCSK9 antagonists is supported by a grant [19] Z. Zhao, Y. Tuakli-Wosornu, T.A. Lagace, L. Kinch, N.V. Grishin, et al., Molecular
from the National Health and Medical Research Council (APP1107403) characterization of loss-of-function mutations in <em>PCSK9</em> and

58
B.J. Tombling et al. Atherosclerosis 330 (2021) 52–60

identification of a compound heterozygote, Am. J. Hum. Genet. 79 (2006) [47] R. Kumar, A. Tonkin, D. Liew, E. Zomer, The cost-effectiveness of PCSK9
514–523. inhibitors - the Australian healthcare perspective, Int. J. Cardiol. 267 (2018)
[20] S.W. Park, Y.-A. Moon, J.D. Horton, Post-transcriptional regulation of low density 183–187.
lipoprotein receptor protein by proprotein convertase subtilisin/kexin type 9a in [48] D.J. Scherer, A.J. Nelson, R. O’Brien, K.M. Kostner, D.L. Hare, et al., Status of
mouse liver, J. Biol. Chem. 279 (2004) 50630–50638. PCSK9 monoclonal antibodies in Australia, Heart Lung Circ. 28 (2019)
[21] K.N. Maxwell, J.L. Breslow, Adenoviral-mediated expression of Pcsk9 in mice 1571–1579.
results in a low-density lipoprotein receptor knockout phenotype, Proc. Natl. [49] B. Zafrir, A. Egbaria, N. Stein, A. Elis, W. Saliba, PCSK9 inhibition in clinical
Acad. Sci. U.S.A. 101 (2004) 7100–7105. practice: treatment patterns and attainment of lipid goals in a large health
[22] S. Benjannet, D. Rhainds, R. Essalmani, J. Mayne, L. Wickham, et al., NARC-1/ maintenance organization, J. Clin. Lipidol. 15 (2021) 202–211, e202.
PCSK9 and its Natural Mutants: zymogen cleavage and effects on the low density [50] B.J. Evison, J.T. Palmer, G. Lambert, H. Treutlein, J. Zeng, et al., A small
lipoprotein (LDL) receptor and LDL cholesterol, J. Biol. Chem. 279 (2004) molecule inhibitor of PCSK9 that antagonizes LDL receptor binding via
48865–48875. interaction with a cryptic PCSK9 binding groove, Bioorg. Med. Chem. 28 (2020),
[23] J.L. Goldstein, M.S. Brown, The LDL receptor, Arterioscler. Thromb. Vasc. Biol. 29 115344.
(2009) 431–438. [51] Y.R. Wu, L. Li, X.C. Sun, J. Wang, C.Y. Ma, et al., Diallyl disulfide improves lipid
[24] M. Brown, J. Goldstein, A receptor-mediated pathway for cholesterol metabolism by inhibiting PCSK9 expression and increasing LDL uptake via PI3K/
homeostasis, Science 232 (1986) 34–47. Akt-SREBP2 pathway in HepG2 cells, Nutr. Metabol. Cardiovasc. Dis. 31 (2021)
[25] T.A. Lagace, D.E. Curtis, R. Garuti, M.C. McNutt, S.W. Park, et al., Secreted PCSK9 322–332.
decreases the number of LDL receptors in hepatocytes and inlivers of parabiotic [52] X. Li, X. Hu, T. Pan, L. Dong, L. Ding, et al., Kanglexin, a new anthraquinone
mice, J. Clin. Invest. 116 (2006) 2995–3005. compound, attenuates lipid accumulation by activating the AMPK/SREBP-2/
[26] C. Gustafsen, D. Olsen, J. Vilstrup, S. Lund, A. Reinhardt, et al., Heparan sulfate PCSK9/LDLR signalling pathway, Biomed. Pharmacother. 133 (2021), 110802.
proteoglycans present PCSK9 to the LDL receptor, Nat. Commun. 8 (2017) 503. [53] R.P. Sparks, A.S. Arango, J.L. Jenkins, W.C. Guida, E. Tajkhorshid, et al., An
[27] T. Yamamoto, C. Lu, R.O. Ryan, A two-step binding model of PCSK9 interaction allosteric binding site on sortilin regulates the trafficking of VLDL, PCSK9, and
with the low density lipoprotein receptor, J. Biol. Chem. 286 (2011) 5464–5470. LDLR in hepatocytes, Biochemistry 59 (2020) 4321–4335.
[28] N.G. Seidah, S. Benjannet, L. Wickham, J. Marcinkiewicz, S.B. Jasmin, et al., The [54] X. Wang, X. Chen, X. Zhang, C. Su, M. Yang, et al., A small-molecule inhibitor of
secretory proprotein convertase neural apoptosis-regulated convertase 1 (NARC- PCSK9 transcription ameliorates atherosclerosis through the modulation of FoxO
1): liver regeneration and neuronal differentiation, Proc. Natl. Acad. Sci. U.S.A. 1/3 and HNF1α, EBioMedicine 52 (2020), 102650.
100 (2003) 928–933. [55] F.J. Raal, D. Kallend, K.K. Ray, T. Turner, W. Koenig, et al., Inclisiran for the
[29] D.E. Piper, S. Jackson, Q. Liu, W.G. Romanow, S. Shetterly, et al., The crystal treatment of heterozygous familial hypercholesterolemia, N. Engl. J. Med. 382
structure of PCSK9: a regulator of plasma LDL-cholesterol, Structure 15 (2007) (2020) 1520–1530.
545–552. [56] K.K. Ray, R.S. Wright, D. Kallend, W. Koenig, L.A. Leiter, et al., Two phase 3 trials
[30] M.C. McNutt, T.A. Lagace, J.D. Horton, Catalytic activity is not required for of inclisiran in patients with elevated LDL cholesterol, N. Engl. J. Med. 382
secreted PCSK9 to reduce low density lipoprotein receptors in HepG2 cells, (2020) 1507–1519.
J. Biol. Chem. 282 (2007) 20799–20803. [57] T. Mitchell, G. Chao, D. Sitkoff, F. Lo, H. Monshizadegan, et al., Pharmacologic
[31] J. Mayne, T. Dewpura, A. Raymond, L. Bernier, M. Cousins, et al., Novel loss-of- profile of the adnectin BMS-962476, a small protein biologic alternative to PCSK9
function PCSK9 variant is associated with low plasma LDL cholesterol in a French- antibodies for low-density lipoprotein lowering, J. Pharmacol. Exp. Therapeut.
Canadian family and with impaired processing and secretion in cell culture, Clin. 350 (2014) 412–424.
Chem. 57 (2011) 1415–1423. [58] E. Weider, D. Susan-Resiga, R. Essalmani, J. Hamelin, M.-C. Asselin, et al.,
[32] D.-W. Zhang, T.A. Lagace, R. Garuti, Z. Zhao, M. McDonald, et al., Binding of Proprotein convertase subtilisin/kexin type 9 (PCSK9) single domain antibodies
proprotein convertase subtilisin/kexin type 9 to epidermal growth factor-like are potent inhibitors of low density lipoprotein receptor degradation, J. Biol.
repeat A of low density lipoprotein receptor decreases receptor recycling and Chem. 291 (2016) 16659–16671.
increases degradation, J. Biol. Chem. 282 (2007) 18602–18612. [59] Z. Chen, P.A. Lichtor, A.P. Berliner, J.C. Chen, D.R. Liu, Evolution of sequence-
[33] H.J. Kwon, T.A. Lagace, M.C. McNutt, J.D. Horton, J. Deisenhofer, Molecular defined highly functionalized nucleic acid polymers, Nat. Chem. 10 (2018)
basis for LDL receptor recognition by PCSK9, Proc. Natl. Acad. Sci. U.S.A. 105 420–427.
(2008) 1820–1825. [60] Y. Pan, Y. Zhou, H. Wu, X. Chen, X. Hu, et al., A therapeutic peptide vaccine
[34] P. Lo Surdo, M.J. Bottomley, A. Calzetta, E.C. Settembre, A. Cirillo, et al., against PCSK9, Sci. Rep. 7 (2017), 12534.
Mechanistic implications for LDL receptor degradation from the PCSK9/LDLR [61] A.A. Momtazi-Borojeni, M.R. Jaafari, A. Badiee, M. Banach, A. Sahebkar,
structure at neutral pH, EMBO Rep. 12 (2011) 1300–1305. Therapeutic effect of nanoliposomal PCSK9 vaccine in a mouse model of
[35] C.G. Davis, J.L. Goldstein, T.C. Südhof, R.G.W. Anderson, D.W. Russell, et al., atherosclerosis, BMC Med. 17 (2019) 223.
Acid-dependent ligand dissociation and recycling of LDL receptor mediated by [62] P.I. Thakore, J.B. Kwon, C.E. Nelson, D.C. Rouse, M.P. Gemberling, et al., RNA-
growth factor homology region, Nature 326 (1987) 760–765. guided transcriptional silencing in vivo with S. aureus CRISPR-Cas9 repressors,
[36] R.P. Naoumova, I. Tosi, D. Patel, C. Neuwirth, S.D. Horswell, et al., Severe Nat. Commun. 9 (2018) 1674.
hypercholesterolemia in four British families with the D374Y mutation in the [63] N.G. Seidah, A. Prat, A. Pirillo, A.L. Catapano, G.D. Norata, Novel strategies to
PCSK9 gene, Arterioscler. Thromb. Vasc. Biol. 25 (2005) 2654–2660. target proprotein convertase subtilisin kexin 9: beyond monoclonal antibodies,
[37] D. Cunningham, D.E. Danley, K.F. Geoghegan, M.C. Griffor, J.L. Hawkins, et al., Cardiovasc. Res. 115 (2019) 510–518.
Structural and biophysical studies of PCSK9 and its mutants linked to familial [64] Y.N. Lamb, Inclisiran: first approval, Drugs 81 (2021) 389–395.
hypercholesterolemia, Nat. Struct. Mol. Biol. 14 (2007) 413–419. [65] D.J. Craik, D.P. Fairlie, S. Liras, D. Price, The future of peptide-based drugs,
[38] M.J. Bottomley, A. Cirillo, L. Orsatti, L. Ruggeri, T.S. Fisher, et al., Structural and Chem. Biol. Drug Des. 81 (2013) 136–147.
biochemical characterization of the wild type PCSK9-EGF(AB) complex and [66] L. Nevola, E. Giralt, Modulating protein–protein interactions: the potential of
natural familial hypercholesterolemia mutants, J. Biol. Chem. 284 (2009) peptides, Chem. Commun. 51 (2015) 3302–3315.
1313–1323. [67] G. Guidotti, L. Brambilla, D. Rossi, Cell-penetrating peptides: from basic research
[39] I. Day, R. Whittall, S. O’Dell, L. Haddad, M. Bolla, et al., Spectrum of LDL receptor to clinics, Trends Pharmacol. Sci. 38 (2017) 406–424.
gene mutations in heterozygous familial hypercholesterolemia, Hum. Mutat. 10 [68] A.F.B. Räder, M. Weinmüller, F. Reichart, A. Schumacher-Klinger, S. Merzbach, et
(1997) 116–127. al., Orally active peptides: is there a magic bullet? Angew. Chem. Int. Ed. 57
[40] J.C.Y. Chan, D.E. Piper, Q. Cao, D. Liu, C. King, et al., A proprotein convertase (2018) 14414–14438.
subtilisin/kexin type 9 neutralizing antibody reduces serum cholesterol in mice [69] K. Fosgerau, T. Hoffmann, Peptide therapeutics: current status and future
and nonhuman primates, Proc. Natl. Acad. Sci. U.S.A. 106 (2009) 9820–9825. directions, Drug Discov. Today 20 (2015) 122–128.
[41] M.S. Sabatine, R.P. Giugliano, A.C. Keech, N. Honarpour, S.D. Wiviott, et al., [70] J.L. Lau, M.K. Dunn, Therapeutic peptides: historical perspectives, current
Evolocumab and clinical outcomes in patients with cardiovascular disease, development trends, and future directions, Bioorg. Med. Chem. 26 (2018)
N. Engl. J. Med. 376 (2017) 1713–1722. 2700–2707.
[42] G.G. Schwartz, P.G. Steg, M. Szarek, D.L. Bhatt, V.A. Bittner, et al., Alirocumab [71] Peptide Therapeutics Market Insights, Trends & Growth Outlook 2019-2027,
and cardiovascular outcomes after acute coronary syndrome, N. Engl. J. Med. 379 2020.
(2018) 2097–2107. [72] A.C. Anselmo, Y. Gokarn, S. Mitragotri, Non-invasive delivery strategies for
[43] N.D. Wong, M.D. Shapiro, Interpreting the findings from the recent PCSK9 biologics, Nat. Rev. Drug Discov. 18 (2019) 19–40.
monoclonal antibody cardiovascular outcomes trials, Front. Cardiovasc. Med. 6 [73] T.S. Fisher, P.L. Surdo, S. Pandit, M. Mattu, J.C. Santoro, et al., Effects of pH and
(2019) 14. low density lipoprotein (LDL) on PCSK9-dependent LDL receptor regulation,
[44] T.D. Filippatos, A. Kei, C.V. Rizos, M.S. Elisaf, Effects of PCSK9 inhibitors on other J. Biol. Chem. 282 (2007) 20502–20512.
than low-density lipoprotein cholesterol lipid variables, J. Cardiovasc. [74] L. Shan, L. Pang, R. Zhang, N.J. Murgolo, H. Lan, et al., PCSK9 binds to multiple
Pharmacol. Therapeut. 23 (2018) 3–12. receptors and can be functionally inhibited by an EGF-A peptide, Biochem.
[45] D.S. Kazi, J. Penko, P.G. Coxson, A.E. Moran, D.A. Ollendorf, et al., Updated cost- Biophys. Res. Commun. 375 (2008) 69–73.
effectiveness analysis of PCSK9 inhibitors based on the results of the FOURIER [75] Y. Zhang, L. Zhou, M. Kong-Beltran, W. Li, P. Moran, et al., Calcium-independent
trial, J. Am. Med. Assoc. 318 (2017) 748–750. inhibition of PCSK9 by affinity-improved variants of the LDL receptor EGF(A)
[46] S. Azari, A. Rezapour, N. Omidi, V. Alipour, M. Behzadifar, et al., Cost- domain, J. Mol. Biol. 422 (2012) 685–696.
effectiveness analysis of PCSK9 inhibitors in cardiovascular diseases: a systematic [76] C.I. Schroeder, Joakim E. Swedberg, Jane M. Withka, K.J. Rosengren, M. Akcan,
review, Heart Fail. Rev. 25 (2019) 1077–1088. et al., Design and synthesis of truncated EGF-A peptides that restore LDL-R
recycling in the presence of PCSK9 in vitro, Chem. Biol. 21 (2014) 284–294.

59
B.J. Tombling et al. Atherosclerosis 330 (2021) 52–60

[77] B.J. Tombling, C. Lammi, N. Lawrence, J. Li, A. Arnoldi, et al., Engineered EGF-A [101] M. Stucchi, G. Grazioso, C. Lammi, S. Manara, C. Zanoni, et al., Disrupting the
peptides with improved affinity for proprotein convertase subtilisin/kexin type 9 PCSK9/LDLR protein–protein interaction by an imidazole-based minimalist
(PCSK9), ACS Chem. Biol. 16 (2021) 429–439. peptidomimetic, Org. Biomol. Chem. 14 (2016) 9736–9740.
[78] C. Lammi, C. Zanoni, G. Aiello, A. Arnoldi, G. Grazioso, Lupin peptides modulate [102] D. Kirchhofer, D.J. Burdick, N.J. Skelton, Y. Zhang, M. Ultsch, Regions of
the protein-protein interaction of PCSK9 with the low density lipoprotein conformational flexibility in the proprotein convertase PCSK9 and design of
receptor in HepG2 cells, Sci. Rep. 6 (2016) 29931. antagonists for LDL cholesterol lowering, Biochem. Soc. Trans. 48 (2020)
[79] C. Lammi, J. Sgrignani, G. Roda, A. Arnoldi, G. Grazioso, Inhibition of 1323–1336.
PCSK9D374Y/LDLR protein–protein interaction by computationally designed T9 [103] R. Ragusa, G. Basta, D. Neglia, R. De Caterina, S. Del Turco, et al., PCSK9 and
lupin peptide, ACS Med. Chem. Lett. 10 (2019) 425–430. atherosclerosis: looking beyond LDL regulation, Eur. J. Clin. Invest. 51 (2021),
[80] Y. Zhang, C. Eigenbrot, L. Zhou, S. Shia, W. Li, et al., Identification of a small e13459.
peptide that inhibits PCSK9 protein binding to the low density lipoprotein [104] Z. Ding, S. Liu, X. Wang, X. Deng, Y. Fan, et al., Cross-talk between LOX-1 and
receptor, J. Biol. Chem. 289 (2014) 942–955. PCSK9 in vascular tissues, Cardiovasc. Res. 107 (2015) 556–567.
[81] Y. Zhang, M. Ultsch, N.J. Skelton, D.J. Burdick, M.H. Beresini, et al., Discovery of [105] N. Ferri, S. Marchianò, G. Tibolla, R. Baetta, A. Dhyani, et al., PCSK9 knock-out
a cryptic peptide-binding site on PCSK9 and design of antagonists, Nat. Struct. mice are protected from neointimal formation in response to perivascular carotid
Mol. Biol. 24 (2017) 848–856. collar placement, Atherosclerosis 253 (2016) 214–224.
[82] C. Alleyne, R.P. Amin, B. Bhatt, E. Bianchi, J.C. Blain, et al., Series of novel and [106] C. Ricci, M. Ruscica, M. Camera, L. Rossetti, C. Macchi, et al., PCSK9 induces a
highly potent cyclic peptide PCSK9 inhibitors derived from an mRNA display pro-inflammatory response in macrophages, Sci. Rep. 8 (2018) 2267.
screen and optimized via structure-based design, J. Med. Chem. 63 (2020) [107] S.J. Bernelot Moens, A.E. Neele, J. Kroon, F.M. van der Valk, J. Van den Bossche,
13796–13824. et al., PCSK9 monoclonal antibodies reverse the pro-inflammatory profile of
[83] B.J. Tombling, C. Lammi, N. Lawrence, E.K. Gilding, G. Grazioso, et al., Bioactive monocytes in familial hypercholesterolaemia, Eur. Heart J. 38 (2017) 1584–1593.
cyclization optimizes the affinity of a proprotein convertase subtilisin/kexin type [108] A.F. Schmidt, D.I. Swerdlow, M.V. Holmes, R.S. Patel, Z. Fairhurst-Hunter, et al.,
9 (PCSK9) peptide inhibitor, J. Med. Chem. 64 (2020) 2523–2533. PCSK9 genetic variants and risk of type 2 diabetes: a mendelian randomisation
[84] B.J. Tombling, C. Lammi, C. Bollati, A. Anoldi, D. Craik, et al., Increased Valency study, Lancet Diabetes Endocrinol. 5 (2017) 97–105.
Improves Inhibitory Activity of Peptides Targeting Proprotein Convertase [109] M.S. Sabatine, L.A. Leiter, S.D. Wiviott, R.P. Giugliano, P. Deedwania, et al.,
Subtilisin/kexin Type 9 (PCSK9), Chembiochem 22 (2021) 2154–2160. Cardiovascular safety and efficacy of the PCSK9 inhibitor evolocumab in patients
[85] C. Lammi, J. Sgrignani, A. Arnoldi, G. Lesma, C. Spatti, et al., Computationally with and without diabetes and the effect of evolocumab on glycaemia and risk of
driven structure optimization, synthesis, and biological evaluation of imidazole- new-onset diabetes: a prespecified analysis of the FOURIER randomised
based proprotein convertase subtilisin/kexin 9 (PCSK9) inhibitors, J. Med. Chem. controlled trial, Lancet Diabetes Endocrinol. 5 (2017) 941–950.
62 (2019) 6163–6174. [110] A.A. Momtazi-Borojeni, S. Sabouri-Rad, A.M. Gotto Jr., M. Pirro, M. Banach, et
[86] D.J. Burdick, N.J. Skelton, M. Ultsch, M.H. Beresini, C. Eigenbrot, et al., Design of al., PCSK9 and inflammation: a review of experimental and clinical evidence, Eur.
organo-peptides as bipartite PCSK9 antagonists, ACS Chem. Biol. 15 (2020) Heart J. Cardiovasc. Pharmacother. 5 (2019) 237–245.
425–436. [111] E.S. Gan, H.C. Tan, H.L.T. Duyen, H.T. Trieu, B. Wills, et al., Dengue virus induces
[87] V. Valenti, D. Noto, A. Giammanco, F. Fayer, R. Spina, et al., PCSK9-D374Y PCSK9 expression to alter antiviral responses and disease outcomes, J. Clin.
mediated LDL-R degradation can be functionally inhibited by EGF-A and Invest. 130 (2020) 5223–5234.
truncated EGF-A peptides: an in vitro study, Atherosclerosis 292 (2020) 209–214. [112] K. Broch, E. Gude, K. Karason, G. Dellgren, G. Rådegran, et al., Cholesterol
[88] S. Kühnast, J.W.A. van der Hoorn, E.J. Pieterman, A.M. van den Hoek, W. lowering with EVOLocumab to prevent cardiac allograft Vasculopathy in De-novo
J. Sasiela, et al., Alirocumab inhibits atherosclerosis, improves the plaque heart transplant recipients: design of the randomized controlled EVOLVD trial,
morphology, and enhances the effects of a statin, J. Lipid Res. 55 (2014) Clin. Transplant. 34 (2020), e13984.
2103–2112. [113] X. Liu, X. Bao, M. Hu, H. Chang, M. Jiao, et al., Inhibition of PCSK9 potentiates
[89] B.J. Tombling, C.K. Wang, D.J. Craik, EGF-like and other disulfide-rich immune checkpoint therapy for cancer, Nature 588 (2020) 693–698.
microdomains as therapeutic scaffolds, Angew. Chem. Int. Ed. 59 (2020) [114] L. Pollaro, C. Heinis, Strategies to prolong the plasma residence time of peptide
11218–11232. drugs, MedChemComm 1 (2010) 319–324.
[90] G. Grazioso, C. Bollati, J. Sgrignani, A. Arnoldi, C. Lammi, First food-derived [115] A. Zorzi, S.J. Middendorp, J. Wilbs, K. Deyle, C. Heinis, Acylated heptapeptide
peptide inhibitor of the protein–protein interaction between gain-of-function binds albumin with high affinity and application as tag furnishes long-acting
PCSK9D374Y and the low-density lipoprotein receptor, J. Agric. Food Chem. 66 peptides, Nat. Commun. 8 (2017), 16092.
(2018) 10552–10557. [116] A. Zorzi, S. Linciano, A. Angelini, Non-covalent albumin-binding ligands for
[91] C. Lammi, G. Aiello, C. Bollati, J. Li, M. Bartolomei, et al., Trans-Epithelial extending the circulating half-life of small biotherapeutics, MedChemComm 10
Transport, Metabolism, and Biological Activity Assessment of the Multi-Target (2019) 1068–1081.
Lupin Peptide LILPKHSDAD (P5) and its Metabolite LPKHSDAD (P5-Met), [117] C.K. Wang, A.S. Amiss, J. Weidmann, D.J. Craik, Structure-activity analysis of
Nutrients 13 (2021) 863. truncated albumin-binding domains suggests new lead constructs for potential
[92] C. Lammi, G. Aiello, G. Vistoli, C. Zanoni, A. Arnoldi, et al., A multidisciplinary therapeutic delivery, J. Biol. Chem. 295 (2020) 12143–12152.
investigation on the bioavailability and activity of peptides from lupin protein, [118] J. Lau, P. Bloch, L. Schäffer, I. Pettersson, J. Spetzler, et al., Discovery of the once-
J. Funct. Foods 24 (2016) 297–306. weekly glucagon-like peptide-1 (GLP-1) analogue semaglutide, J. Med. Chem. 58
[93] R.H. Alghamdi, P. O’Reilly, C. Lu, J. Gomes, T.A. Lagace, et al., LDL-R promoting (2015) 7370–7380.
activity of peptides derived from human PCSK9 catalytic domain (153–421): [119] J.P.H. Wilding, R.L. Batterham, S. Calanna, M. Davies, L.F. Van Gaal, et al., Once-
design, synthesis and biochemical evaluation, Eur. J. Med. Chem. 92 (2015) weekly semaglutide in adults with overweight or obesity, N. Engl. J. Med. 384
890–907. (2021) 989–1002.
[94] C. Lammi, J. Sgrignani, A. Arnoldi, G. Grazioso, Biological characterization of [120] P.R. Beauchesne, N.S. Chung, K.M. Wasan, Cyclosporine A: a review of current
computationally designed analogs of peptide TVFTSWEEYLDWV (Pep2-8) with oral and intravenous delivery systems, Drug Dev. Ind. Pharm. 33 (2007) 211–220.
increased PCSK9 antagonistic activity, Sci. Rep. 9 (2019) 2343. [121] S. Fattah, M. Ismaiel, B. Murphy, A. Rulikowska, J.M. Frias, et al., Salcaprozate
[95] T.A. Hill, N.E. Shepherd, F. Diness, D.P. Fairlie, Constraining cyclic peptides to sodium (SNAC) enhances permeability of octreotide across isolated rat and
mimic protein structure motifs, Angew. Chem. Int. Ed. 53 (2014) 13020–13041. human intestinal epithelial mucosae in Ussing chambers, Eur. J. Pharmaceut. Sci.
[96] X. Wu, Y.-H. Huang, Q. Kaas, D.J. Craik, Cyclisation of disulfide-rich conotoxins 154 (2020), 105509.
in drug design applications, Eur. J. Org Chem. 2016 (2016) 3462–3472. [122] A.M. van den Hoek, J.W.A. van der Hoorn, A.C. Maas, R.M. van den Hoogen,
[97] Ricardo, A, Boyer, NC, Stringer, JR, Laplaca, DM, Jette, K, et al., Cyclic peptides A. van Nieuwkoop, et al., APOE*3 Leiden.CETP transgenic mice as model for
for PCSK9 inhibition, In, 2019, WO 2019/246387 A1. pharmaceutical treatment of the metabolic syndrome, Diabetes Obes. Metabol. 16
[98] Ricardo, A, Tucker, TJ, Boyer, NC, Dhamnaskar, KA, Ma, Z, et al., Cyclic (2014) 537–544.
polypeptides for PCSK9 inhibition, In, 2020. WO 2019/246405 A1. [123] Y. Zhao, H. Qu, Y. Wang, W. Xiao, Y. Zhang, et al., Small rodent models of
[99] Flyer, AN, Golosov, AA, Grosche, P, Lewis, I, Liu, EY, et al., Cyclic peptides as atherosclerosis, Biomed. Pharmacother. 129 (2020) 110426.
proportein convertase subtilisin/kexin type 9 (PCSK9) inhibitors for the [124] H.E. Careskey, R.A. Davis, W.E. Alborn, J.S. Troutt, G. Cao, et al., Atorvastatin
treatment of metabolic disorders, In, 2020. WO 2020/110011 A1. increases human serum levels of proprotein convertase subtilisin/kexin type 9,
[100] Briner, K, Dechristopher, BA, Flyer, AN, Golosov, AA, Grosche, P, et al., Cyclic J. Lipid Res. 49 (2008) 394–398.
tetramer compounds as proprotein convertase subtilisin/kexin type 9 (PCSK9) [125] J. Mayne, T. Dewpura, A. Raymond, M. Cousins, A. Chaplin, et al., Plasma PCSK9
inhibitors for the treatment of metabolic disorders, In, 2020. WO 2020/110009 levels are significantly modified by statins and fibrates in humans, Lipids Health
A1. Dis. 7 (2008) 22.

60

You might also like