ArticlePDF AvailableLiterature Review

PCSK9 inhibitors: A new era of lipid lowering therapy

Authors:

Abstract and Figures

Hyperlipidemia is a well-established risk factor for developing cardiovascular disease (CVD). The recent American College of Cardiology and American Heart Association guidelines on lipid management emphasize treatment of individuals at increased risk for developing CVD events with 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors (statins) at doses proven to reduce CVD events. However, there are limited options for patients who are either intolerant to statin therapy, develop CVD despite being on maximally tolerated statin therapy, or have severe hypercholesterolemia. Recently the Food and Drug Administration approved two novel medications for low-density lipoprotein (LDL)-cholesterol reduction: Evolocumab and Alirocumab. These agents target and inactivate proprotein convertase subtilsin-kexin type 9 (PCSK9), a hepatic protease that attaches and internalizes LDL receptors into lysosomes hence promoting their destruction. By preventing LDL receptor destruction, LDL-C levels can be lowered 50%-60% above that achieved by statin therapy alone. This review explores PCSK-9 biology and the mechanisms available to alter it; clinical trials targeting PCSK9 activity, and the current state of clinically available inhibitors of PCSK9.
Content may be subject to copyright.
PCSK9 inhibitors: A new era of lipid lowering therapy
Rahul Chaudhary, Jalaj Garg, Neeraj Shah, Andrew Sumner
Rahul Chaudhary, Department of Medicine, Sinai Hospital of
Baltimore, Johns Hopkins University, Baltimore, MD 21209,
United States
Jalaj Garg, Neeraj Shah, Andrew Sumner, Division of
Cardiology, Lehigh Valley Health Network, Allentown, PA 18103,
United States
Author contributions: Chaudhary R and Garg J contributed
equally to the paper; all authors contributed to this paper.
Conict-of-interest statement: All authors report no conicts
of interest.
Open-Access: This article is an open-access article which was
selected by an in-house editor and fully peer-reviewed by external
reviewers. It is distributed in accordance with the Creative
Commons Attribution Non Commercial (CC BY-NC 4.0) license,
which permits others to distribute, remix, adapt, build upon this
work non-commercially, and license their derivative works on
different terms, provided the original work is properly cited and
the use is non-commercial. See: http://creativecommons.org/
licenses/by-nc/4.0/
Manuscript source: Invited manuscript
Correspondence to: Jalaj Garg, MD, FESC, Division of
Cardiology, Lehigh Valley Health Network, 1250 S Cedar Crest
Blvd, Allentown, PA 18103,
United States. garg.jalaj@yahoo.com
Telephone: +1-585-7660898
Fax: +1-610-4023225
Received: August 15, 2016
Peer-review started: August 16, 2016
First decision: September 6, 2016
Revised: November 23, 2016
Accepted: December 7, 2016
Article in press: December 9, 2016
Published online: February 26, 2017
Abstract
Hyperlipidemia is a well-established risk factor for
developing cardiovascular disease (CVD). The recent
American College of Cardiology and American Heart
Association guidelines on lipid management emphasize
treatment of individuals at increased risk for developing
CVD events with 3-hydroxy-3-methylglutaryl coenzyme
A reductase inhibitors (statins) at doses proven to
reduce CVD events. However, there are limited options
for patients who are either intolerant to statin therapy,
develop CVD despite being on maximally tolerated statin
therapy, or have severe hypercholesterolemia. Recently
the Food and Drug Administration approved two novel
medications for low-density lipoprotein (LDL)-cholesterol
reduction: Evolocumab and Alirocumab. These agents
target and inactivate proprotein convertase subtilsin-
kexin type 9 (PCSK9), a hepatic protease that attaches
and internalizes LDL receptors into lysosomes hence
promoting their destruction. By preventing LDL receptor
destruction, LDL-C levels can be lowered 50%-60%
above that achieved by statin therapy alone. This review
explores PCSK-9 biology and the mechanisms available
to alter it; clinical trials targeting PCSK9 activity, and the
current state of clinically available inhibitors of PCSK9.
Key words: Hyperlipidemia; Statins; Proprotein convertase
subtilsin-kexin type 9
© The Author(s) 2017. Published by Baishideng Publishing
Group Inc. All rights reserved.
Core tip: Hyperlipidemia is a well-established risk factor
for developing cardiovascular disease (CVD). However,
there are limited options for patients who are either
intolerant to statin therapy, develop CVD despite being
on maximally tolerated statin therapy, or have severe
hypercholesterolemia. The Food and Drug Administration
has approved two novel medications for low-density
lipoprotein (LDL)-cholesterol reduction in this patient
population: Evolocumab and Alirocumab. These agents
target and inactivate proprotein convertase subtilsin-
kexin type 9 (PCSK9), a hepatic protease that attaches
and internalizes LDL receptors into lysosomes hence
promoting their destruction. By preventing LDL receptor
destruction, LDL-C levels can be lowered 50%-60% above
that achieved by statin therapy alone. PCSK9 inhibitors are
REVIEW
Submit a Manuscript: http://www.wjgnet.com/esps/
DOI: 10.4330/wjc.v9.i2.76
76 February 26, 2017
|
Volume 9
|
Issue 2
|
WJC
|
www.wjgnet.com
World J Cardiol 2017 February 26; 9(2): 76-91
ISSN 1949-8462 (online)
World Journal of
Cardiology
W J C
an exciting agent for reducing LDL-C and have ushered in
a new era of lipid lowering therapy.
Chaudhary R, Garg J, Shah N, Sumner A. PCSK9 inhibitors: A new
era of lipid lowering therapy. World J Cardiol
2017; 9(2): 76-91
Available from: URL: http://www.wjgnet.com/1949-8462/full/v9/
i2/76.htm DOI: http://dx.doi.org/10.4330/wjc.v9.i2.76
INTRODUCTION
Hyperlipidemia is a well-established risk factor for develo-
ping cardiovascular disease (CVD)[1]. Multiple double
blind placebo controlled trials have shown that treatment
with HMG CoA Reductase inhibitors (statins) lowers
low-density lipoprotein (LDL)-C levels and reduces CVD
events in individuals with CVD or those at high risk for
developing it[2,3]. However, CVD events continue to occur
in some patients on statins, despite receiving maximal
tolerated therapy. Other patients develop side effects
from statins that limit their use. Hence, newer modalities
of treatment to lower LDL-C are needed in clinical
practice. Recently the Food and Drug Administration (FDA)
approved two medications which target a novel pathway
to reduce LDL-C. They are monoclonal antibodies that
inactivate proprotein convertase subtilsin-kexin type 9
(PCSK9). This review will explore the biology of PCSK 9,
clinical trials targeting PCSK9 activity, and the current
state of clinically available inhibitors of PCSK9.
LDL-CHOLESTEROL METABOLISM
LDL-C has been the target of therapy for improving
outcomes in patients at high risk for developing CVD
and has been considered a surrogate endpoint for
clinical events by the FDA[1]. Reviewing LDL cholesterol
metabolism is therefore important in understanding
therapeutic approaches to treat hyperlipidemia.
The lipid cycle begins with the release of immature
very low-density lipoprotein (VLDL) or nascent VLDL from
the liver. Nascent VLDL contains apolipoprotein-B100
(apoB-100), apolipoprotein E (apoE), apolipoprotein C1
(apoC1), cholesteryl esters, cholesterol, and triglycerides.
While circulating in blood, high-density lipoprotein (HDL)
donates apolipoprotein C- (apoC-) to nascent VLDL
that leads to its maturation. Mature VLDL interacts with
lipoprotein lipase (LPL) in the capillary beds of adipose
tissues, cardiac muscle and skeletal muscle cells, which
leads to extraction of triglycerides from VLDL for storage
or energy production in these tissues. VLDL combines
with HDL again and an interchange occurs where apoC-
is transferred back to HDL along with phospholipids
and triglycerides in exchange for cholesteryl esters via
cholesterylester-transfer protein (CETP). This exchange
and removal of triglycerides leads to conversion of VLDL
to intermediate-density lipoprotein (IDL)[4]. Half of IDLs
are recognized and endocytosed by liver cells due to
apoB-100 and apoE. The remaining IDL lose apoE, and
with an increased concentration of cholesterol compared
to triglyceride, transform into low-density lipoproteins
(LDL). LDL particles thus formed contain apoB-100,
which acts as a ligand for binding to LDL receptors
(LDLR). Once LDL binds to LDLR, LDL/LDLR complex is
internalized by endocytosis into clathrin coated vesicles.
In the cytosol, LDL and LDLR separate with recycling of
LDLR to the cell surface. LDLR recycling is a continuous
process and each receptor recycles up to 150 times after
which they are endocytosed and metabolized[5]. Statins
act by inhibiting 3-hydroxy-3-methylglutaryl coenzyme
A (HMG-CoA) reductase, which is involved in intracellular
production of cholesterol. This lowers the levels of
intracellular cholesterol leading to increased expression of
LDLR on cell surfaces causing a reduction in serum LDL-
cholesterol[6].
Seidah and colleagues discovered that proprotein
convertase subtilisin/kexin type 9 (PCSK9) regulates
LDLR degradation and could potentially be a target for
modulating LDLR expression and consequently LDL-C
levels[7,8]. PCSK9 is a hepatic protease that attaches to
and internalizes LDLR into lysosomes hence promoting
their destruction[9]. Clinical studies have shown that
PCSK9 gain of function mutation is associated with
familial hypercholesterolemia and premature CVD[10,11].
Conversely, individuals with loss of function mutations in
PCSK9 have been observed to have lower lifetime levels
of LDL-C and lower prevalence of CVD[12,13].
Since the discovery of PCSK9, results from preclinical
mice studies demonstrated that sterol regulatory ele-
ment binding protein-2 (SREBP-2) plays a key role in
regulating cholesterol metabolism. Low level of intra-
cellular cholesterol activates SREBP-2 and leads to LDLR
gene expression. This increases LDLR concentration
thus enhancing LDL clearance from circulation[8,14]. At
the same time SREBP-2 also induces the expression of
PCSK9, which promotes LDLR degradation. Thus, the
coordinated interplay of SREBP-2 induced transcription
of both LDLR and PCSK9 regulates circulating LDL
levels[15,16]. These discoveries resulted in the exploration
and development of therapeutic agents to lower LDL
levels by targeting PCSK9 activity.
FUNCTIONAL MECHANICS OF PCSK9
Hepatocytes are the predominant site for PCSK9
production, with other sites being intestines and kid-
neys[17,18]. PCSK9 reduces the number of LDLR in hepa-
tocytes by promoting their metabolism and subsequent
degradation[14]. PCSK9 has been shown to act both
intracellularly (playing a role as a chaperone) as well as a
secreted factor promoting LDLR internalization from the
hepatocellular surface. Under normal circumstances, the
LDL/LDLR complex is endocytosed by endosomes. The
acidic pH of the endosome reduces the affinity of LDL
for LDLR with rearrangement of the LDLR’s extracellular
domain into a hairpin structure, aiding in its recycling
77 February 26, 2017
|
Volume 9
|
Issue 2
|
WJC
|
www.wjgnet.com
Chaudhary R
et al
. Review on PCSK9 inhibitors
back to plasma membrane. PCSK9 binding inhibits this
change and locks the LDLR in an open conformation
which prevents its recycling. The LDLR is then routed to
lysosomes for degradation (Figure 1)[19,20]. The secreted
form of PCSK9 circulates in the bloodstream and can
be inactivated by cleavage from proprotein convertase.
At a molecular level, the secretion of prodomain and
catalytically inactive PCSK9 promotes regular degradation
of LDLR implying that PCSK9 acts as a chaperone protein
rather than an active catalytic enzyme[21,22].
As described above, hepatic expression of PCSK9 and
LDLR are closely regulated by SREBP-2 and intracellular
levels of cholesterol[23,24]. Lipid lowering therapy with
statins[25-27], ezetimibe[28] and bile acid binding resins[29]
cause induction of SREBP-2 and hence co-induces both
PCSK9 and LDLR. The slight increase in PCSK9 activity
seen with statins does not negate their therapeutic eff-
ectiveness.
OTHER FUNCTIONS AND LOCATIONS OF
PCSK9
Apart from hepatocytes, PCSK9 is also expressed in
intestine, central nervous system, and mesenchymal
cells of the kidney. In vitro studies on human intestinal
epithelium have reported recombinant PCSK9 to enhance
cholesterol uptake in the human intestinal epithelial cells
(Caco-2/15 cell line) via the up regulation of the protein
expression of NPC1L1 and CD36 (involved in cholesterol
absorption in intestinal cells) along with an increased
expression of cholesterol transporters[30,31] and reduced
cholesterol synthesis (by reducing HMG-CoA reductase
activity)[32]. PCSK9 has been shown to have a role in the
metabolism of triglycerides and their accumulation in
visceral adipose tissue[33]. It also promotes chylomicron
secretion and helps regulate enterocyte cholesterol
78 February 26, 2017
|
Volume 9
|
Issue 2
|
WJC
|
www.wjgnet.com
balance[32]. Studies have evaluated PCSK9 and their
association with increased susceptibility to hepatitis C
viral infection. Labon et al[34] demonstrated a reduced
expression of CD81 (CD81 is a co-receptor for Hepatitis
C virus infection) by PCSK9 leading to protection against
infection by hepatitis C. Therefore, PCSK9 inhibitors
(Alirocumab) could increase CD81 expression resulting in
greater infectivity. However, in vitro and in vivo studies in
mice showed that PCSK9 did not reduce CD81 expression
and had no effect on HCV infectivity. Hence, the liter-
ature remains inconclusive about this potential effect
of PCSK9 inhibition[34]. Mbikay et al[35] demonstrated
that PCSK9 inhibition in mouse pancreatic islet β cells
led to hypoinsulinemia, hyperglycemia and glucose-
intolerance. Additionally the islet cells exhibited signs
of malformation, apoptosis and inflammation. Current
clinical data has failed to show this as a complication with
PCSK9 inhibition.
PCSK9 INHIBITION STRATEGIES
Preclinical studies demonstrate that statin-induced LDL
reduction occurs through increased LDLR expression on
hepatocytes along with increased LDL turnover, which leads
to its enhanced clearance from the circulation. However,
statins also induce PCSK9 expression, which dampens the
effective LDL clearing by promoting LDLR degradation[23].
Since PCSK9 is expressed both intracellularly and in the
circulation, multiple potential targets exist for its inhibition.
Various modalities to inhibit PCSK9 have been studied
including: (1) inhibition of production by gene silencing
through antisense oligonucleotides[36] or small interfering
RNA[37]; (2) prevention of PCSK9 binding to LDLR using
monoclonal antibodies[38], epidermal growth factor-like
repeat A (EGF-A), mimetic peptides[39] or adnectins; and (3)
inhibition of PCSK autocatalytic sites.
PCSK9 LDL-C
Endosome
LDL receptor
Lysosome PCSK9 bound LDL
receptor is digested in
lysosome Recycled LDL-receptor
LDL-cholesterol
incorporates into cell
Endosome
Figure 1 Mechanism and role of PCK9 in low-density lipoprotein-cholesterol metabolism. LDL: Low-density lipoprotein.
Chaudhary R
et al
. Review on PCSK9 inhibitors
79 February 26, 2017
|
Volume 9
|
Issue 2
|
WJC
|
www.wjgnet.com
to statin therapy in patients with high atherosclerotic
cardiovascular risk with a follow up period of up to 12 wk.
SPIRE-1 (NCT01975376) and 2 (NCT01975389) have a
follow up period of up to 5 years collecting data on safety
and efficacy of this drug[49]. Recently, the preliminary
results of a study of Bococizumab delivery using an auto-
injector device (SPIRE-AI) reported successfully meeting
co-primary endpoints of percent change from baseline in
fasting LDL-C at week 12 and the delivery system success
rate, dened as the percent of patients whose attempts to
operate the pre-lled pen. SPIRE-AI is a 12-wk, double-
blind, placebo-controlled, randomized, parallel-group,
multicenter, phase clinical trial in 299 patients with
hyperlipidemia or mixed dyslipidemia receiving statin
therapy and whose LDL-C 70 mg/dL and assessed
the efcacy, safety, tolerability and subcutaneous admini-
stration of Bococizumab 150 mg and 75 mg with a pre-
lled pen[50].
Along similar lines, adnectins (also known as monobody)
and small peptide inhibitors have been investigated for LDL
reduction in phase
Ⅰ 
clinical trials. The results in healthy
patients and those with hypercholesterolemia demonstrated
a maximal dose related reduction of LDL-C by up to
48%[51]. The advantage of developing adnectins is that they
are smaller than mAb, making them cheaper and easier to
produce. Their pharmacokinetics have been shown to be
favorable with a rapid onset of action in preclinical models,
further trials are awaited to see the development of this
agent[52].
Inhibition of autocatalytic site
This mechanism as a therapeutic target was first pro-
posed after discovering a loss of function mutation in the
autocatalytic cleavage site of PCSK9[53,54]. This approach
is still under preclinical investigational phase.
FDA APPROVAL STATUS OF PCSK9
INHIBITORS
The FDA approved Alirocumab (Praluent) in July 2015
for adult patients with heterozygous familial hyper-
cholesterolemia or in patients with clinically significant
atherosclerotic CVD requiring additional LDL lowering
after being on diet control and maximally tolerated statin
therapy. Evolocumab (Repatha) was also approved in
August, 2015 for use in adult patients with heterozygous
familial hypercholesterolemia, homozygous familial hyper-
cholesterolemia, or clinical atherosclerotic CVD requiring
additional lowering of LDL cholesterol after being on a
controlled diet and maximally-tolerated statin therapy.
Alirocumab (status: FDA approved)
Alirocumab has been studied in three phase
Ⅰ 
trials. In
two of these trials, healthy volunteers were administered
Alirocumab intravenously (n = 40) or subcutaneously (n
= 32) which reduced LDL-C in a dose-dependent fashion
with a reduction of up to 65% at maximal doses[55]. The
Gene silencing via antisense oligonucleotides or small
interfering RNA
This approach targets intracellular PCSK9 activity by
utilizing antisense oligonucleotide to reduce intracellular
expression of PCSK9. Preclinical trials on hyperlipidemic
mice evaluating two such compounds were promising
with a reduction in LDL by 38% at six weeks of therapy
while doubling LDLR expression in the liver[38]. However,
the phase
Ⅰ 
trials evaluating two of these agents were
terminated prematurely and further development of the
drug (BMS-84442) was not continued (NCT01082562).
Two additional drugs: SPC5001 and SPC4061 showed
successful reduction in LDL by 50% during preclinical
testing in primates[40]. However, the first phase
Ⅰ 
trial
in healthy human subjects and individuals with familial
hypercholesterolemia were terminated early (NCT01
350960)[41,42]. SPC5001 was seen to cause mild to moder-
ate injection site reactions and renal tubular toxicity[43].
Further development of SPC4061 was discontinued for
undisclosed reasons.
Similarly, small interfering RNA (siRNA) administration
has been shown to significantly reduce plasma PCSK9
and LDL levels in cynomolgus monkeys[37,38,43]. ALN-PCS
is a siRNA, which was tested by delivery through second-
generation lipid nanoparticles. In a study by Fitzgerald
et al[44], ALN-PCS demonstrated a dose dependent
reduction in PCSK9 and LDL levels with a reduction of up
to 70% in PCSK9 levels and 40% in LDL levels at doses
of 0.4 mg/kg. This was the first study to demonstrate
intracellular PCSK9 inhibition translated into reduction of
circulating LDL levels.
Monoclonal antibodies
Utilization of monoclonal antibodies has been the most
effective approach thus far in inhibiting PCSK9 and re-
ducing LDL levels. Currently, at least six monoclonal
antibodies (mAb) have been or are being developed
and tested: Alirocumab (formerly called SAR236553/
REGN727), Evolocumab (formerly called AMG145), RG7
652[45], LGT209 (NCT01979601, NCT01859455), 1B20[46]
and Bococizumab (formerly called RN316/PF-049 50615).
Alirocumab and Evolocumab have recently been approved
by the FDA. The major clinical studies leading up to their
approval are outlined later in this paper.
Bococizumab is a unique mAb, which utilizes pH
sensitive binding to PCSK9 and was developed for a longer
serum half-life and duration of action on LDL reduction[47].
In phase
Ⅰ 
studies, single intravenous or subcutaneous
dosages significantly reduced LDL in patients with
hypercholesterolemia, both with and without concomitant
atorvastatin therapy[48]. In a phase trial by Gumbiner
et al[48], patients on statin therapy not at target LDL-C
were enrolled and observed a 60% reduction in LDL-C
after 12 wk of therapy. Five phase trials are ongoing for
Bococizumab including SPIRE-HF (evaluating the efcacy
of this agent in heterozygous familial hypercholesterolemia
NCT01968980); SPIRE-HR (NCT01968954) and SPIRE-
LDL (NCT01968967) trials are comparing Bococizumab
Chaudhary R
et al
. Review on PCSK9 inhibitors
80 February 26, 2017
|
Volume 9
|
Issue 2
|
WJC
|
www.wjgnet.com
dose or switching to rosuvastatin. Atorvastatin at 20 mg
and 40 mg/d regimens reduced LDL by 44% and 54%
with addition of Alirocumab respectively vs 21% and 23%
with addition of ezetimibe respectively vs 5% and 5% with
doubling atorvastatin dose respectively compared to 21%
with switching to rosuvastatin 40 mg/d. In the OPTIONS
trial, rosuvastatin was studied using a similar protocol
and showed that the addition of Alirocumab had the most
significant reduction in LDL-C after 24 wk of therapy as
opposed to addition of ezetimibe or doubling the dose of
rosuvastatin[62,63]. Another study evaluating the efcacy of
Alirocumab as monotherapy (MONO study)[64] compared
to ezetimibe in patients with hypercholesterolemia and
moderate cardiovascular risk to monotherapy with
Alirocumab and found that Alirocumab reduced LDL-C 47%
vs 16% by ezetimibe after 24 wk of therapy (P < 0.0001).
CHOICE study evaluated Alirocumab in patients
intolerant to statin therapy. Two hundred and thirty
one patients with a history of statin intolerance were
shown to have a 56% reduction in LDL with Alirocumab
(vs placebo; P < 0.001)[59]. Another study evaluating
Alirocumab in patients intolerant to statin therapy is
the ODYSSEY ALTERNATIVE trial. Three hundred and
fourteen patients completed this randomized controlled
trial, which compared Alirocumab 75 mg every 2 wk (n
= 126) to ezetimibe 10 mg/d (n = 125) and atorvastatin
20 mg/d (n = 63) for 24 wk. At 24 wk, the data showed
a 45% reduction in LDL with Alirocumab as opposed
to 15% reduction in LDL with ezetimibe. This trial
demonstrated fewer skeletal muscle adverse events in
the Alirocumab group as compared to atorvastatin arm
[32.5% vs 46% respectively, HR = 0.61 (0.38-0.99; P =
0.042)], with no signicant difference when compared to
the ezetimibe group (41%) [HR 0.71 (0.47 to 1.06; P =
0.09)][65].
Alirocumab has also been shown to be effective
in lowering LDL-C in patients with familial hypercho-
lesterolemia. The FH
and FH studies evaluated a
total of 735 patients (n = 486 and 249 respectively) with
heterozygous familial hypercholesterolemia inadequately
controlled on lipid lowering therapy and found Alirocumab
to reduce LDL levels 48.8% (vs a 9.1% increase in
placebo: FH
study) and 48.7% (vs 2.8% increase
in LDL with placebo: FH study) from baseline[66].
ODYSSEY HIGH FH trial reported 105 patients with familial
hypercholesterolemia on maximally tolerated statin
therapy and LDL > 160 demonstrating a 46% reduction
of LDL (vs 7% with placebo) at 24 wk (P < 0.001)[67].
OLE trial (NCT01954394) is currently ongoing with results
anticipated by June 2017. This trial is recruiting patients
with heterozygous familial hypercholesterolemia who have
completed one of the other studies and evaluating for
safety parameters including adverse events, laboratory
data and vital signs.
ODYSSEY LONG TERM trial was recently published
and is a 78-wk follow-up of 2341 patients with hyper-
cholesterolemia and LDL > 70 mg/dL on maximally
tolerated statins. The patients receiving 150 mg Aliro-
cumab every 2 wk were shown to have a 62% reduction
third trial evaluated patients with non-familial hyper-
cholesterolemia on atorvastatin and LDL > 100 mg/dL
or with LDL > 130 mg/dL being managed by diet alone.
Alirocumab reduced LDL-C up to 65% in patients on
statins and up to 60% in patients being managed on
diet alone. It was observed that Alirocumab remained
effective for a longer period of time in patients not on
statins[55].
Three phase trials[56-58] evaluated the efficacy of
Alirocumab in patients with familial hypercholesterolemia.
Stein et al[58] evaluated 77 patients on statin therapy
with LDL-C greater than 100 mg/dL and found that
Alirocumab reduced LDL-C in a dose-dependent manner
by up to 43% with a maximum dosage of 300 mg every
4 wk. However, the reduction was even greater (up
to 70%) at a dosage regimen of 150 mg every 2 wk.
Additionally, these patients had a signicant reduction in
apoB levels and non-high density lipoprotein cholesterol
and also had increases in HDL. McKenney et al[56], in
their study of 183 patients conrmed a dose dependent
reduction of LDL-C with the most efficacious regimen
being 150 mg every 2 wk (with LDL reduction up to
70%). Interestingly, different doses of atorvastatin did
not make a significant difference in LDL-C reduction.
These results were further corroborated by Roth et al[57]
in their study of 92 patients with LDL-C > 100 mg/dL.
They showed that there was signicant and comparable
LDL reduction irrespective of the dosage of atorvastatin
(10 mg vs 80 mg) when added to Alirocumab 150 mg
every 2 wk[57].
The phase randomized, double-blinded ODYSSEY
trials were designed to evaluate Alirocumab for long-
term safety, efficacy and adverse events (Table 1) and
include CHOICE
, CHOICE , OLE, LONG TERM,
COMBO
, COMBO , FH
, FH , HIGH FH, MONO,
ALTERNATIVE, OPTIONS
and OPTIONS . The dosage
of Alirocumab administered was 150 mg every 2 wk in
ODYSSEY LONG TERM and HIGH FH trials and 75 mg
(titrated up to 150 mg to reach pre-specied LDL goals)
in ODYSSEY ALTERNATIVE, OPTIONS
, OPTIONS and
COMBO
trials.
CHOICE
study evaluated Alirocumab vs placebo in
803 patients with poorly controlled hypercholesterolemia.
Alirocumab reduced LDL by 52% in statin-naïve patients
and by 59% in patients on maximally tolerated statins
as compared to placebo (P < 0.001)[59]. Similarly,
COMBO
and trials evaluated 316 patients and 720
patients respectively with LDL > 70 mg/dL and high
cardiovascular risk on maximally tolerated statin therapy.
COMBO
showed Alirocumab reduced LDL-C up to 50%
(vs 2% with placebo) after 24 wk of treatment. COMBO
compared ezetimibe to Alirocumab in patients on
background statin therapy and found a 50% LDL reduction
with Alirocumab vs 20% reduction with ezetimibe at
24 wk[60]. OPTIONS
trial[61] published in August 2015,
randomized 355 patients with hypercholesterolemia
and LDL > 70 and found the addition of Alirocumab
to atorvastatin had the greatest reduction in LDL as
compared to addition of ezetimibe, doubling atorvastatin
Chaudhary R
et al
. Review on PCSK9 inhibitors
81 February 26, 2017
|
Volume 9
|
Issue 2
|
WJC
|
www.wjgnet.com
Table 1 Summary of phase ODYSSEY trials with Alirocumab
Name of trial Ref. Allocation and
blinding
No. of
patients
Inclusion criteria Study arms
(with dosing)
Primary end point Results
LONG TERM
(NCT01507831)
Seidah et al[7];
Randomized
double blinded trial
2341 Either 1 or 2 below and who aren’t
adequately controlled with their LLT:
(1) Patients with heFH with or without
CHD or CHD risk equivalents
OR (2) Patients with HCL with
CHD or CHD risk equivalents
Alirocumab
(SC) (n = 1553)
vs Placebo
(SC) (n = 788)
both with
background
LLT
Percentage change
in calculated LDL
cholesterol level
from baseline to
week 24
-61.0% change with
Alirocumab vs +0.8% change
with placebo (CI: -64.3 to
-59.4; P < 0.001)
FH
(NCT01623115)
Kastelein et al[66];
Randomized
double blinded
486 Patients with heterozygous
familial hypercholesterolemia who
are not adequately controlled with
their lipid-modifying therapy
Alirocumab
(SC) vs Placebo
(SC) both with
background
LLT
Percent change in
calculated LDL-C at
week 24
-48.8% for Alirocumab
compared with 9.1% for
placebo (P < 0.0001)
FH
(NCT01709500)
Kastelein et al[66];
Randomized
double blinded
249 Patients with heFH who are not
adequately controlled with their
LLT
Alirocumab
(SC) vs Placebo
(SC) both with
background
LLT
Percent change in
LDL-C to week 24
-48.7% for Alirocumab
compared with 2.8% for
placebo (P < 0.0001)
HIGH FH
(NCT01617655)
Kastelein et al[67];
Randomized
double blinded
107 Patients with heterozygous
familial hypercholesterolemia who
are not adequately controlled with
their lipid-modifying therapy with
LDL > 160
Alirocumab
(SC) (n = 72) vs
Placebo (SC) (n
= 35) both with
background
LLT
Percent change in
calculated LDL-C at
week 24
Percent decrease from
baseline was 45.7% vs 6.6%,
difference 39.1, P < 0.0001
Absolute difference in values
of LDL-C at 24 wk 107 mg/
dL vs 182 mg/dL
COMBO Ⅰ 
(NCT01644175)
Colhoun et al[60];
Randomized
double blinded
316 Patients with hypercholesterolemia
and estbl CHD or CHD risk
equivalents; not controlled with a
maximally tolerated LLT, both at
stable dose for at least 4 to 6 wk
prior to screening
Alirocumab
(SC) (n = 205) vs
Placebo (SC) (n
= 106)
Percent change in
calculated LDL-C at
week 24
-48.2% with Alirocumab (CI:
-52.0% to -44.4%) and -2.3%
with placebo (CI: -7.6% to
3.1%) for Alirocumab and
placebo, respectively, an
estimated mean difference of
-45.9% (CI: -52.5% to -39.3%)
(P < 0.0001)
COMBO
(NCT01644188)
Moriarty et al[65];
Randomized
double blind
720 Patients with hypercholesterolemia
and established CHD or CHD
risk equivalents who are not
adequately controlled with a
maximally tolerated daily dose of
statin at stable dose for at least 4
wk prior to the screening visit
Alirocumab
(SC) + placebo
(for ezetimibe)
orally +
background
statin therapy (n
= 467)
vs Placebo (SC)
+ ezetimibe
orally +
Background
statin therapy (n
= 240)
Percent change in
calculated LDL-C at
week 24
Reductions in LDL-C
from baseline were 50.6%
± 1.4% for Alirocumab vs
20.7% ± 1.9% for ezetimibe
(difference 29.8% ± 2.3%; P <
0.0001)
OPTIONS
(NCT01730053)
Robinson et al[63];
Randomized
double-blinded
355 Patients with prior CV disease +
LDL-C 70 mg/dL, or CV risk
factors + LDL-C 100 mg/dL
Alirocumab
with
atorvastatin
20 mg vs
Ezetimibe with
atorvastatin
20 mg vs
Atorvastatin 40
mg
Alirocumab
with
atorvastatin
40 mg vs
ezetimibe with
atorvastatin
40 mg vs
atorvastatin
80 mg vs
rosuvastatin 20
mg
Percent change in
calculated LDL-C to
week 24
Percent reduction from
baseline 44.1% (Alirocumab)
vs 20.5% (ezetimibe) vs 5.0%
(atorvastatin 40); P < 0.0001
Percent reduction from
baseline 54% (Alirocumab)
vs 22.6% (Ezetimibe) vs 4.8%
(Atorvastatin 80) vs 21.4%
(rosuvastatin 40); P < 0.0001
Chaudhary R
et al
. Review on PCSK9 inhibitors
82 February 26, 2017
|
Volume 9
|
Issue 2
|
WJC
|
www.wjgnet.com
at 24 wk. These results persisted at 78 wk. In a post-in LDL as opposed to a 1% increase in LDL with placebo
OPTIONS Robinson et al[63];
Randomized
double-blinded
305 Patients with prior CV disease +
LDL-C 70 mg/dL, or CV risk
factors + LDL-C 100 mg/dL
Alirocumab
with
rosuvastatin 10
mg
vs ezetimibe
with
rosuvastatin 10
vs rosuvastatin
20
Alirocumab
with
rosuvastatin
20 mg vs
ezetimibe with
rosuvastatin 20
vs Rosuvastatin
40
Percent change in
calculated LDL-C to
wk 24
Percent reduction from
baseline 50.6% (Alirocumab)
vs 14.4% (ezetimibe) vs 16.3%
(rosuvastatin 20); P < 0.0001
Percent reduction from
baseline 36.3% (Alirocumab)
vs 11.0% (Ezetimibe) vs
20.3% (rosuvastatin 40); P <
0.0001
ALTERNATIVE
(NCT01709513)
Moriarty et al[65];
Randomized
double-blinded
314 Primary heFH with moderate,
high or very high CV risk and
history of statin intolerance
Alirocumab +
oral placebo
vs ezetimibe
(10 mg/d) +
sc placebo vs
atorvastatin
(20 mg/d) + sc
placebo
Percent change in
calculated LDL-C to
week 24 in intention
to treat group
Percent reduction from
baseline 45% (Alirocumab)
vs 14.6% (Ezetimibe) with a
mean difference of -30.4%; P
< 0.0001
CHOICE
(NCT01926782)
Stroes et al[78];
Randomized,
double-blinded
803 Patients not having
adequate control of their
hypercholesterolemia based on
their individual level of CVD risk
Alirocumab
at q4 week
regimen vs
Placebo
Percent change
in LDL from
baseline to week
24 for Alirocumab
q4w vs placebo
in patients with
hypercholesterolemia
at moderate, high, or
very high CVD risk
with concomitant
statin therapy (n =
547)
Percent change
in LDL from
baseline to week
24 for Alirocumab
q4w vs placebo
in patients with
hypercholesterolemia
not on concomitant
statin therapy (n =
256)
LDL was reduced by 58.7%
with Alirocumab in patients
on maximally tolerated
statins (P < 0.001)
LDL was reduced by 52.4%
with Alirocumab in statin
naïve patients vs placebo (P
< 0.001)
CHOICE
(NCT02023879)
Stroes et al[78];
Randomized,
double-blinded
231 Patients with primary
hypercholesterolemia (heFH
or non-FH) not adequately
controlled with their non statin
lipid modifying therapy or diet
and statin intolerance
Alirocumab
(SC) vs placebo
(SC)
The percent change in
LDL-C from baseline
to week 24
Alirocumab reduced LDL-C
by 56.4% (P < 0.0001) vs
placebo
LONG TERM
(NCT01507831)
Robinson et al[62];
Randomized,
double-blinded
2341 Either A or B below and who are
not adequately controlled with
their LLT: (1) Patients with heFH
with or without established CHD
or CHD risk equivalents
OR
(2) Patients with
hypercholesterolemia together
with established CHD or CHD
risk equivalents
Alirocumab
(SC) 150 mg
every 2 wk vs
placebo (SC)
every 2 wk
Percentage change
in calculated LDL
cholesterol level from
baseline to week 24,
analyzed with the
use of an intention-
to-treat approach
150 mg Alirocumab every
2 wk had a 62% reduction
in LDL as opposed to a 1%
increase in LDL with placebo
at 24 wk
SC: Subcutaneous; LLT: Lipid lowering therapy; heFH: Heterozygous familial hypercholesterolemia; CHD: Coronary heart disease; HCL: Hyper-
cholesterolemia; MACE: Major adverse cardiovascular events; MI: Myocardial infarction; UA: Unstable angina; HR: Hazards ratio; CI: Condence interval.
Chaudhary R
et al
. Review on PCSK9 inhibitors
83 February 26, 2017
|
Volume 9
|
Issue 2
|
WJC
|
www.wjgnet.com
patients intolerant to statins (GAUSS-2 and GAUSS-3);
standalone in hyperlipidemia (MENDEL-2); heterozygous
familial hypercholesterolemia (RUTHERFORD-2 and
TAUSSIG); homozygous familial hypercholesterolemia
(TESLA and TAUSSIG); with primary hyperlipidemia or
mixed cholesterol disorder (THOMAS-1 and THOMAS-2:
Device trials). Also, long-term safety and efficacy
data is being evaluated by the five following studies:
DESCARTES; FOURIER; OSLER-2 trial; GLAGOV trial and
TAUSSIG study.
In LAPLACE-2 study 1896 patients with fasting LDL
150 (when not on statins) or LDL 100 (on non-
intense regimen of statins) or LDL 70 (on intensive
statin therapy) were randomized to a daily moderate
or high-intensity statin regimen and after 4 wk, further
randomized to receive Evolocumab, ezetimibe or
placebo. Evolocumab was shown to reduce LDL levels
by 66% to 75% (on every 2 wk regimen) and by 63%
to 75% (on once monthly regimen) when compared
to placebo in moderate- and high intensity statin
groups. In moderate and high intensity statin groups,
Evolocumab led to signicant reduction in absolute LDL
values in both regimens of Evolocumab (every 2 wk and
monthly). Adverse events reported were comparable in all
groups[75]. YUKAWA-2 study showed a similar reduction
in LDL in 404 Japanese patients when Evolocumab
regimens (140 mg once every 2 wk and 420 mg once
a month) were compared to placebo on 2 regimens of
low-dose background statin therapy (5 mg/d and 20
mg/d atorvastatin). Interestingly, the reduction in LDL
appeared to be similar irrespective of statin dosage (in
combination with Evolocumab) and showed a 67%
to 76% LDL reduction at 12 wk[76]. MENDEL-2 trial
compared the efcacy of Evolocumab with ezetimibe and
placebo in 614 patients with LDL between 100 mg/dL
and 190 mg/dL and low risk on Framingham scale (
10%). Evolocumab was shown to reduce LDL by up to
57% more than placebo and 40% more than ezetimibe
after 12 wk of therapy[77].
GAUSS-2 trial evaluated 307 patients with statin
intolerance and compared the 2 regimens of Evolocumab
(140 mg once every 2 wk and 420 mg once a month)
to daily oral or subcutaneous placebo (both placebo
groups on ezetimibe). At 12 wk, Evolocumab group
showed a reduction in LDL by 56% vs 39% in the other
groups (placebo + ezetimibe arm)[78]. Along similar lines,
GAUSS-3 trial evaluated the efficacy of Evolocumab in
218 statin intolerant patients compared to ezetimibe.
The initial phase of the study included administration of
atorvastatin (20 mg) for 10 wk and placebo randomized
in a 1:1 fashion, followed by a 2-wk washout period
and crossover to alternate therapy for another 10 wk.
The patients who experienced muscle related adverse
effects while on statin therapy and not on placebo were
further enrolled in the second phase of the study, which
was a 24 wk double blinded randomized controlled
trial to compare Evolocumab (420 mg/mo divided in 3
doses) with ezetimibe (10 mg/d). At 24 wk, LDL-C was
hoc analysis, the reduction in LDL was also associated
with reduction in the combined end-point of death from
coronary artery disease, nonfatal myocardial infarction,
fatal or nonfatal ischemic stroke or unstable angina
requiring hospitalization (1.7% with Alirocumab vs
3.3% with placebo; HR = 0.52; 95%CI: 0.31-0.9; P =
0.02)[68]. The ODYSSEY Outcomes trial (NCT01663402)
is ongoing is closed to recruitment, and will assess the
effects of Alirocumab on CVD events in 18000 patients on
maximally tolerated statin therapy. Results of this trial are
expected in February 2018.
Evolocumab (status: FDA approved)
Evolocumab has been studied in two phase
studies.
Dias et al[69] evaluated healthy volunteers in phase
a
and showed a short-term dose-dependent reduction in
LDL by up to 65% and after 6 to 8 wk of therapy by up
to 75% with a maximally administered dose of 420 mg
subcutaneously/intravenously. Phase
b trial similarly
demonstrated up to 75% reduction in LDL as compared
to placebo over 1 to 4 wk in healthy volunteers.
Four phase trials were subsequently performed
that continued to show the benets of Evolocumab with
a dose-dependent reduction of LDL (maximal dosing up
to 420 mg) when added to maximally tolerated statin
therapy in patients with hypercholesterolemia (including
familial hypercholesterolemia)[70-72]. In LAPLACE-TIMI57
trial[70], Evolocumab was tested at varying doses ranging
from 70 to 140 mg every 2 wk or 280 to 420 mg every
4 wk in 631 patients on stable statin therapy and LDL
more than 85 mg/dL. LDL reduction up to 65% was
observed with the every two-week regimen as compared
to approximately 50% LDL reduction with the every
4-wk regimen. Evolocumab has also been studied as
monotherapy in 160 patients with hypercholesterolemia
and intolerance to statins in the GAUSS trial[73]. At doses
of 420 mg every 4 wk, it was shown to reduce LDL
by 40% to 50%. Furthermore, addition of ezetimibe
reduced LDL by up to 65%. Subsequently, the MENDEL
trial[71] showed a similar efcacy in LDL-C lowering when
Evolocumab was used as monotherapy in 406 patients
with hypercholesterolemia. Based on these trials, the
optimal frequency of Evolocumab therapy was determined
to be twice monthly to achieve a 50% to 60% reduction
in LDL in combination with statins. However, when used
as a monotherapy therapy, a frequency of once every
4 wk would be acceptable. Stein et al[74] evaluated 8
patients with homozygous familial hypercholesterolemia,
and found Evolocumab (at 420 mg every 2 wk) to reduce
LDL by approximately 25% vs 20% when used every 4
wk.
Evolocumab has further been evaluated in PROFICIO
(Program to reduce LDL-C and cardiovascular outcomes
following inhibition of PCSK9 in different populations)
phase trials (Table 2). The PROFICIO program
includes 14 trials where Evolocumab is being evaluated
in patients with hyperlipidemia in combination with
statins (LAPLACE-2 and YUKAWA-2); hyperlipidemic
Chaudhary R
et al
. Review on PCSK9 inhibitors
84 February 26, 2017
|
Volume 9
|
Issue 2
|
WJC
|
www.wjgnet.com
Table 2 Summary of important phase PROFICIO (Program to Reduce LDL-C and Cardiovascular Outcomes Following Inhibition
of PCSK9 In Different Populations) trials with Evolocumab
Name of trial Ref. Allocation
and blinding
No. of
patients
Inclusion criteria Study arms (with dosing) Primary end
point
Results
LAPLACE-2
(NCT01763866)
Robinson et al[75];
Randomized
double blinded
trial
1896 Individuals with LDL
> 150 mg/dL (not
on statin); or LDL >
100 mg/dL (on non-
intensive statin); or
LDL 80 mg/dL (with
intensive statin therapy)
Initially randomized to
daily moderate or high
intensity atorvastatin
for 4 wk. Patients were
again randomized to
Evolocumab (sc) vs
ezetimibe vs placebo
Percentage
change in
calculated LDL
cholesterol level
from baseline to
week 12
Evolocumab q2w and
qmonthly: 63% to 75%
reduction in LDL vs
placebo
Ezetimibe 19% to 32%
reduction in LDL vs
placebo
YUKAWA-2
(NCT01953328)
Kiyosue et al[76];
Randomized
double blinded
404 Japanese patients with
LDL > 70 on stable dose
statins for > 4 wk and
high cardiovascular risk
Initially randomized to
daily atorvastatin of 5 mg
or 20 mg for 4 wk. They
were further randomized
to Evolocumab (sc) at q2
week and qmonthly vs
placebo
Percent change
in calculated
LDL-C from
baseline at week
12
-67.0% to -76% reduction
with Evolocumab
compared to placebo (P
< 0.0001)
GAUSS-2
(NCT01763905)
Stroes et al[78];
Randomized
double blinded
307 Patients with LDL not at
goal according to their
cardiovascular risk and
not on statin or low dose
statin due to history of
statin intolerance (> 2
statins) with stable LLT
> 4 wk
Evolovumab (SC) at
q2 week and qmonthly
dosing vs Placebo (SC)
+ Ezetimibe (10 mg/d)
daily
Percent change
in LDL-C from
baseline at the
mean of weeks
10 and 12 and at
week 12
Change from
baseline LDL at
week 12
-55.3% to -56.1% for
Evolocumab compared
with -16.6% to -19.2% for
ezetimibe (P < 0.0001)
-103.6 to -105.4
(Evolocumab) vs -33 to
-39 (mg/dL)
MENDEL-2
(NCT01763827
Koren et al[77];
Randomized
double-blinded
614 NCEP ATP
Framingham risk score
of < 10%
Fasting LDL-C 100
mg/dL and <
190 mg/dL
Oral placebo to SC
placebo; ezetimibe to SC
placebo and oral placebo
to SC Evolocumab at
dosing regimens of 140
mg biweekly and 420 mg
monthly
Percent change
from baseline
in LDL-C level
averaged at
weeks 10 and 12
Percent change
from baseline in
LDL-C level at
week 12
Percent LDL change from
baseline averaged at
weeks 10 and 12 in the:
Once per 2 wk
arm: -56.9% (with
Evolocumab) vs -17.5
(with ezetimibe) vs -0.4%
(placebo)
For monthly arm:
-58.8% (with
evolocumab) vs -19.1
(with ezetimibe) vs -1.4%
(placebo)
Percent LDL change
from baseline averaged
at weeks 12: Once per
2 wk arm: -57% (with
Evolocumab) vs -17.8
(with ezetimibe) vs 0.1%
(placebo)
For monthly arm: -56.1%
(with Evolocumab) vs
-18.6 (with ezetimibe) vs
-1.3% (placebo)
RUTHERFORD-2
(NCT01763918)
Raal et al[72];
Randomized
double blinded
329 Patients with
heterozygous familial
hypercholesterolemia
who are on stable LLT
for 4 wk and LDL > 100
mg/dL
Evolocumab (SC) at 140
mg q2 weeks vs placebo
SC q2w AND
Evolocumab SC
qmonthly vs Placebo
(SC)
Percent change
from baseline
in LDL-C level
averaged at
weeks 10 and 12
Percent change
from baseline in
LDL-C level at
week 12
Percent LDL change
from baseline averaged
at weeks 12 in the: Once
per 2 wk arm: -61.2%
(with Evolocumab) vs
-1.1% (with placebo)
For monthly arm: -63.3%
(with evolocumab) vs
2.3% (with placebo)
Percent LDL change
from baseline averaged
at weeks 10 and 12 in
the: Once per 2 wk
arm: -61.3% (with
Evolocumab) vs -2%
(with placebo)
For monthly arm: -55.7%
(with Evolocumab) vs
5.5% (with placebo)
Chaudhary R
et al
. Review on PCSK9 inhibitors
85 February 26, 2017
|
Volume 9
|
Issue 2
|
WJC
|
www.wjgnet.com
in LDL with both regimens: 140 mg every 2 wk led to
59.2% reduction (CI: 53.4% to 65.1%) and 420 mg
once a month led to LDL reduction by 61.3% (CI: 53.6%
to 69%) as compared to placebo after 12 wk (P < 0.001
for all)[81]. The TESLA trial examined 50 patients with
homozygous familial hypercholesterolemia on stable lipid
lowering therapy and evaluated monthly Evolocumab
(420 mg) vs placebo therapy for 12 wk. Addition of
Evolocumab led to a signicant reduction in LDL-C by up
to 31% (CI: -44% to -18%; P < 0.0001)[82].
In the Open-Label Study of Long-Term Evaluation
Against LDL-C (OSLER) 1 and 2 trials 4465 patients
were enrolled who had completed 1 of the phase 2 or
phase 3 studies of Evolocumab (MENDEL-1, LAPLACE
TIMI 57, GAUSS-1, RUTHERFORD-1, YUKAWA-1,
MENDEL-2, LAPLACE-2, GAUSS-2, RUTHERFORD-2,
DESCARTES, THOMAS-1 or THOMAS-2) and randomized
to receive either Evolocumab (420 mg/mo in OSLER-1
and 140 mg every 2 wk or 420 mg once a month in
OSLER-2 trial) plus standard therapy (n = 2976) or
standard therapy (n = 1489). The median follow-up
was 11.1 mo. This study showed a 61% reduction in
LDL-C with Evolocumab compared to standard therapy
(95%CI: 59% to 63%; P < 0.001). Overall adverse
events were in 69% of patients in Evolocumab group
compared to 65% in standard therapy group. Of note,
the neurocognitive adverse events were low, but were
more frequent in Evolocumab group and appeared
reduced by 53% with Evolocumab compared to 17%
with ezetimibe. Muscle-related side effects were reported
in 21% patients on Evolocumab compared to 29% with
ezetimibe with stoppage of drug administration due to
muscle symptoms in 1% of patients in Evolocumab and
7% of patients on ezetimibe[79].
DESCARTES trial[80] evaluated 901 patients with
hyperlipidemia, comparing Evolocumab (420 mg once
a month subcutaneous) plus background lipid lowering
therapy vs placebo plus background lipid lowering
therapy for a period of 52 wk. Background lipid lowering
therapy included: Diet alone, low intensity atorvastatin
(10 mg), high intensity atorvastatin (80 mg) or ator-
vastatin 80 mg/d. All patients had fasting LDL-C > 75
mg/dL on background lipid lowering therapy. Addition
of Evolocumab resulted in LDL reduction by 51% to
60% in diet alone group, 59% to 64% in patients on
10 mg atorvastatin, 51% to 62% in patients on 80
mg atorvastatin and 43% to 54% in patients with
atorvastatin 80 mg/d and ezetimibe 10 mg/d (P < 0.001
for all).
Evolocumab has also been shown to be efficacious
in patients with heterozygous and homozygous familial
hypercholesterolemia. In the RUTHERFORD-2 trial, 329
patients with heterozygous familial hypercholesterolemia
were randomized to receive Evolocumab (140 and 420
mg respectively) or placebo at two weekly and monthly
regimens. Evolocumab showed a significant reduction
TESLA
(NCT01588496)
Raal et al[82];
Randomized
double-blinded
50 Homozygous familial
hypercholesterolemia,
on stable lipid-regulating
therapy for at least 4 wk,
LDL cholesterol 130
mg/dL (3.4 mmol/L);
Triglyceride 400
mg/dL (4.5 mmol/L);
Body weight of 40 kg
at screening,
and not receiving
lipoprotein apheresis
Evolocumab (SC) 420 mg
every 4 wk vs placebo (SC)
Percentage
change in
ultracentrifugation
LDL cholesterol
from baseline at
week 12 compared
with placebo,
analyzed by
intention-to-treat
Percent change from
baseline in LDL-C
at week 52
Evolocumab
signicantly reduced
ultracentrifugation LDL
cholesterol at 12 wk by
30.9% (95%CI: -43.9%
to -18.0%; P < 0.0001) vs
placebo
DESCARTES
(NCT01516879)
Blom et al[80];
Randomized,
double-blinded
901 Fasting LDL 75
mg/dL and meeting
the following on
background LLT: (1) <
100 mg/dL for subjects
with diagnosed CHD or
CHD risk equivalent; (2)
< 130 mg/dL for subjects
without diagnosed CHD
or CHD risk equivalent;
(3) on maximal
background LLT dened
as atorvastatin 80 mg PO
QD and ezetimibe 10 mg
PO QD
Fasting triglycerides
400 mg/dL
Evolocumab (SC) 420
mg every 4 wk with diet
alone vs placebo with diet
Evolocumab (SC) 420
mg every 4 wk with diet
+ atorvastatin 10 mg/d
vs placebo with diet and
atorvastatin 10 mg/d
Evolocumab (SC) 420 mg
every 4 wk + atorvastatin
80 mg/d vs placebo +
atorvastatin 80 mg/d
Evolocumab (SC) 420 mg
every 4 wk + atorvastatin
80 mg/d + ezetimibe
10 mg/d vs placebo +
atorvastatin 80 mg/d +
ezetimibe 10 mg/d
Addition of Evolocumab
resulted in LDL
reduction by: (1) 51% to
60% in diet alone group;
(2) 59% to 64% in patients
on 10 mg atorvastatin
(3) 51% to 62% in patients
on 80 mg atorvastatin
(4) 43% to 54% in patients
with atorvastatin 80
mg/d and ezetimibe 10
mg/d (P < 0.001 for all)
SC: Subcutaneous; LLT: Lipid lowering therapy; heFH: Heterozygous familial hypercholesterolemia; CHD: Coronary heart disease; HCL: Hyper-
cholesterolemia; MACE: Major adverse Cardiovascular Events; MI: Myocardial infarction; UA: Unstable angina; HR: Hazards ratio; CI: Condence
interval.
Chaudhary R
et al
. Review on PCSK9 inhibitors
86 February 26, 2017
|
Volume 9
|
Issue 2
|
WJC
|
www.wjgnet.com
No data are available in patients with severe renal and
hepatic impairment.
Evolocumab
The pharmacokinetic and pharmacodynamics properties
of Evolocumab[83] demonstrate non-linear pharmacokin-
etics in absorption at doses below 140 mg; however,
between doses of 140 to 420 mg linear pharmacokinetics
is observed. The time to reach maximum concentration
is 3 to 4 d after a single dose. After a single 420 mg
dosage, its volume of distribution has been estimated
to be 3.3 L ± 0.5 L. A steady state in serum is observed
after about 12 wk of dosing. The t1/2 of Evolocumab is
between 11 to 17 d. The maximum reduction of LDL after
therapy was similar after dosing of 140 mg every 2 wk
or 420 mg once a month with effect within 14 d. Clinical
studies have not revealed a difference in pharmacokinetics
of Evolocumab in mild or moderate renal and hepatic
impairment. However, subjects with severe renal and
hepatic impairment have not been studied.
ADVERSE EFFECTS AND
CONTRAINDICATIONS
The following side effects have been reported by data
gathered from over 7000 patients (n = 2476 for Aliro-
cumab and n = 5416 for Evolocumab) evaluated in
the clinical trials mentioned above. Major side effects
observed for Alirocumab and Evolocumab are described
below.
Alirocumab
Alirocumab is contraindicated in patients who develop
serious hypersensitivity reactions like hypersensitivity
vasculitis or allergic reactions requiring hospitalization with
its usage. The most common adverse effects observed
with Alirocumab include nasopharyngitis (11.3% vs 11.1%
in placebo), injection site reactions (erythema, itchiness,
swelling, pain or tenderness) (7.2% vs 5.1% in placebo),
influenza (5.7% vs 4.6% in placebo), urinary tract
infection (4.8% vs 4.6% in placebo), diarrhea (4.7% vs
4.4% in placebo), bronchitis (4.3% vs 3.8% in placebo),
myalgia (4.2% vs 3.4% in placebo), muscle spasms (3.1%
vs 2.4% in placebo), sinusitis (3% vs 2.7% in placebo),
cough (2.5% vs 2.3% in placebo), contusion (2.1% vs
1.3% in placebo) and musculoskeletal pain (2.1% vs
1.6% in placebo). The most common adverse events that
lead to drug discontinuation were allergic reactions (0.6%
for Alirocumab vs 0.2% for placebo) and elevated liver
enzymes (0.3% in Alirocumab vs < 0.1% in placebo).
Evolocumab
Contraindications for Evolocumab are similar to Aliro-
cumab. The overall incidence of adverse effects with
Evolocumab 140 mg every 2 wk as compared to placebo
were 43.6% vs 41% respectively. The most common
adverse effects were nasopharyngitis (5.9% vs 4.8% in
placebo), upper respiratory tract infection (3.2% vs 2.7%
to be unrelated to LDL level at the time of treatment.
Composite adverse cardiovascular events (all-cause
death, coronary events including myocardial infarction,
unstable angina requiring hospitalization, or coronary
revascularization, cerebrovascular events including stroke
or transient ischemic attack, and heart failure requiring
hospitalization) were signicantly lower in patients with
Evolocumab compared to standard therapy (HR = 0.47;
95%CI: 0.28 to 0.78; P = 0.003)[83,84].
The TAUSSIG trial (NCT01624142) is evaluating
Evolocumab therapy in 300 patients with severe familial
hypercholesterolemia to determine its efcacy and side
effect profile. The results of this study are anticipated
by March 2020. Preliminary results reported by Stein et
al[74] on 8 patients with LDLR-negative or LDLR defective
homozygous familial hypercholesterolemia on stable
drug therapy when treated with Evolocumab at 420 mg
monthly for 12 wk, followed by 420 mg every 2 wk for
another 12 wk showed LDL reduction by 14% to 16% at
12 wk with 2 wk and 4 wk dosing regimens respectively
with no serious adverse events reported[75]. Finally, the
preliminary results of GLAGOV study (NCT01813422)
evaluating 950 patients with coronary artery disease on
lipid lowering therapy undergoing cardiac catheterization
for changes in percentage atheroma volume after 78
wk of Evolocumab therapy met primary and secondary
endpoints andnal results are to be reported in American
Heart Association (AHA) conference in November, 2016.
PHARMACOKINETICS AND
PHARMACODYNAMICS
The pharmacokinetic and pharmacodynamics parameters
of PCSK9 inhibitors are described below[85,86].
Alirocumab
The time taken to reach maximum serum concentration
is 3-7 d with similar serum concentration - time proles
between abdomen, upper arm or thigh as the sites of
injection. Steady state concentrations are reached at
an average of 3 to 4 doses. The volume of distribution
following intravenous administration is 0.04 to 0.05 L/kg.
The median half-life (t1/2) observed was between 17
to 20 d at 75 or 150 mg dosing every 2 wk. Alirocumab
is eliminated in two phases depending upon its plasma
concentration. The predominant mode of elimination
at lower concentrations is via saturation of the targets
(PCSK9) bound to the antibodies; however, at higher
concentrations it is primarily through proteolytic path-
ways[81]. There have been no metabolism studies
conducted since it has been previously demonstrated that
reticuloendothelial system is responsible for metabolizing
antibodies to small peptides and amino acids[87]. The
maximum reduction in free plasma PCSK9 levels and LDL
was observed within 3 and 15 d respectively, after drug
administration with no difference noted between different
sites. No dose adjustment is needed for patients with
mild or moderately impaired renal or hepatic function.
Chaudhary R
et al
. Review on PCSK9 inhibitors
87 February 26, 2017
|
Volume 9
|
Issue 2
|
WJC
|
www.wjgnet.com
mendations.
PCSK9 inhibitors are especially beneficial in the
treatment of familial hypercholesterolemic patients who
are intolerant to statins or have an elevated LDL-C level
despite being on maximally tolerated statin therapy.
Intuitively, addition of a PCSK9 inhibitor to low dose
statin therapy will be more effective in lowering LDL and
avoiding the side effects of statins, since low dose and
high dose statin regimens have yielded similar efficacy
when combined with PCSK9 inhibitors.
Several potential barriers exist that may impede the
widespread use of these medicines. First, statins have
a proven effectiveness that has been demonstrated in
multiple long-term studies. Statins have been shown to
reduce cardiovascular mortality by 30% and incidence of
stroke by 20% in multiple long-term studies[91,92]. PCSK9
inhibitors are effective in reducing LDL-C levels but
currently lack data demonstrating their use reduces CVD
events. Trials evaluating the effect of PCSK9 inhibitors
on long-term CVD events, however, are currently under-
way: FOURIER (Further cardiovascular outcomes
research with PCSK9 inhibition in subjects with elevated
risk; n = 22500) for Evolocumab (NCT01764633) and
ODYSSEY-OUTCOMES (ODYSSEY outcomes: Evaluation
of cardiovascular outcomes after an acute coronary
syndrome during treatment with alirocumab SAR236553)
(NCT01663402) for Alirocumab. However, their data will
not be available until December 2017 (for Alirocumab)
and February 2018 (for Evolocumab).
Another potential barrier to widespread use of
PCSK9 inhibitors is their cost. The Institute for Clinical
and Economic Review (ICER) reported that the number
needed to treat for 5 years to avoid one major adverse
cardiovascular event (NNT5) is 28. However, a list price
of $ 14350 per year generates a cost-effectiveness
ratio which far exceeds the accepted threshold of $
100000/quality-affected life-years[93]. To achieve cost-
effectiveness at this threshold would require a price
reduction by 60% to 65% of the current price. At
the conclusion of their report, the ICER suggested a
reduction by 85% to an annual cost of $2177 might be
required to avoid excessive cost burdens to the health
care system[94]. It should be noted that since there are
limited data on clinical adverse cardiovascular events,
cost effectiveness data might change once results from
ongoing CVD endpoint studies are available.
PCSK9 therapy is a welcome treatment option for
statin intolerant patients who require treatment of their
hyperlipidemia. It will be important that busy practitioners
do not under-prescribe statins nor be dissuaded from
attempting to find a dose of and statin agent that is
tolerated by the patient because PCSK9 inhibitors are
available. Despite these obstacles, PCSK9 inhibitors are
an exciting agent for reducing LDL-C hyperlipidemia and
have ushered in a new era of lipid lowering therapy.
REFERENCES
1 Stone NJ, Robinson JG, Lichtenstein AH, Bairey Merz CN, Blum
CB, Eckel RH, Goldberg AC, Gordon D, Levy D, Lloyd-Jones DM,
in placebo), back pain (3% vs 2.7% in placebo) and
nausea (2.1% vs 1.8% in placebo). Of note the most
common adverse events leading to drug discontinuation
include myalgia, nausea and dizziness. Other serious
adverse events noted were cardiac disorders in 2.4%
individuals including palpitations (0.6% vs 0.3% in
placebo), angina pectoris (0.3% vs 0.2% in placebo),
and ventricular extra systoles (0.3% vs 0.1% in placebo).
In addition, data from trials evaluating Evolocumab
and Alirocumab have shown a higher incidence of
cognitive adverse events in patients treated with PCSK9
inhibitors (0.9% vs 0.3% for Evolocumab compared
to standard care[83] and 1.2% vs 0.5% for Alirocumab
compared to placebo[69]). It has been suggested that
responder and ascertainment bias might have played
a role in reporting of adverse cognitive events in the
OSLER trial since the adverse events were not related
to the degree of LDL-C lowering with no clustering in the
LDL-C < 25 mg/dL group relative to the 25-50 mg/dL or
> 50 mg/dL groups[88]. However, patients in ODYSSEY
LONG TERM trial were blinded to treatment and followed
for nearly 18 mo. Also, the neurocognitive adverse
events were measured subjectively and not veried by
neurocognitive testing. A dedicated study evaluating
neurocognitive adverse events with PCSK9 inhibitors
is underway: Evaluating PCSK9 Binding anti-Body
Influence oN coGnitive HeAlth in High cardiovascUlar
Risk Subjects (EBBINGHAUS) (NCT02207634). It is en-
rolling individuals without dementia or mild cognitive
impairment at baseline randomized in a double-blind,
placebo-controlled fashion to evaluate Evolocumab on
background statin therapy vs statin therapy alone. The
primary outcome being measured is Spatial Working
Memory test; an assessment of executive function and
the results are expected in September 2017.
Another potential and significant complication with
drugs that are monoclonal antibodies is the development
of anti-drug antibodies that may interfere with clinical
efcacy and increase adverse events[89]. This complication
has not been reported in the trials to date using PCSK9
inhibitors.
CLINICAL USE OF PCSK9 INHIBITORS
The 2013 American College of Cardiology/American
Heart Association recommendations on cholesterol man-
agement centered on identifying patients who would
have a reduction in CVD events with statin treatment.
The focus shifted from treating to a specic LDL-C level,
to treating at risk individuals with a treatment (statin)
proven to reduce future CVD events. Subsequently, the
IMPROVE-IT trial demonstrated that addition of ezetimibe
to simvastatin lowers LDL-C more than that achieved
by simvastatin alone and that this reduction in LDL-C
was associated with a greater reduction in CVD events
compared with simvastatin alone[86,90]. This study raises
the issue of LDL-C treatment targets with a lower level
of LDL-C corresponding to a lower risk of CVD events.
The introduction of PCSK9 inhibitors will necessitate
re-evaluation of existing cholesterol treatment recom-
Chaudhary R
et al
. Review on PCSK9 inhibitors
88 February 26, 2017
|
Volume 9
|
Issue 2
|
WJC
|
www.wjgnet.com
SREBP target genes. Proc Natl Acad Sci USA 2003; 100: 12027-12032
[PMID: 14512514 DOI: 10.1073/pnas.1534923100]
17 Rashid S, Curtis DE, Garuti R, Anderson NN, Bashmakov Y, Ho YK,
Hammer RE, Moon YA, Horton JD. Decreased plasma cholesterol
and hypersensitivity to statins in mice lacking Pcsk9. Proc Natl Acad
Sci USA 2005; 102: 5374-5379 [PMID: 15805190 DOI: 10.1073/
pnas.0501652102]
18 Zaid A, Roubtsova A, Essalmani R, Marcinkiewicz J, Chamberland
A, Hamelin J, Tremblay M, Jacques H, Jin W, Davignon J, Seidah
NG, Prat A. Proprotein convertase subtilisin/kexin type 9 (PCSK9):
hepatocyte-specific low-density lipoprotein receptor degradation
and critical role in mouse liver regeneration. Hepatology 2008; 48:
646-654 [PMID: 18666258 DOI: 10.1002/hep.22354]
19 Lo Surdo P, Bottomley MJ, Calzetta A, Settembre EC, Cirillo
A, Pandit S, Ni YG, Hubbard B, Sitlani A, Carfí A. Mechanistic
implications for LDL receptor degradation from the PCSK9/LDLR
structure at neutral pH. EMBO Rep 2011; 12: 1300-1305 [PMID:
22081141 DOI: 10.1038/embor.2011.205]
20 Yamamoto T, Lu C, Ryan RO. A two-step binding model of
PCSK9 interaction with the low density lipoprotein receptor. J Biol
Chem 2011; 286: 5464-5470 [PMID: 21149300 DOI: 10.1074/jbc.
M110.199042]
21 Li J, Tumanut C, Gavigan JA, Huang WJ, Hampton EN, Tumanut
R, Suen KF, Trauger JW, Spraggon G, Lesley SA, Liau G, Yowe
D, Harris JL. Secreted PCSK9 promotes LDL receptor degradation
independently of proteolytic activity. Biochem J 2007; 406: 203-207
[PMID: 17608623 DOI: 10.1042/BJ20070664]
22 McNutt MC, Lagace TA, Horton JD. Catalytic activity is not required
for secreted PCSK9 to reduce low density lipoprotein receptors in
HepG2 cells. J Biol Chem 2007; 282: 20799-20803 [PMID: 17537735
DOI: 10.1074/jbc.C700095200]
23 Dong B, Wu M, Li H, Kraemer FB, Adeli K, Seidah NG, Park
SW, Liu J. Strong induction of PCSK9 gene expression through
HNF1alpha and SREBP2: mechanism for the resistance to LDL-
cholesterol lowering effect of statins in dyslipidemic hamsters. J
Lipid Res 2010; 51: 1486-1495 [PMID: 20048381 DOI: 10.1194/jlr.
M003566]
24 Li H, Dong B, Park SW, Lee HS, Chen W, Liu J. Hepatocyte nuclear
factor 1alpha plays a critical role in PCSK9 gene transcription and
regulation by the natural hypocholesterolemic compound berberine. J
Biol Chem 2009; 284: 28885-28895 [PMID: 19687008 DOI: 10.1074/
jbc.M109.052407]
25 Careskey HE, Davis RA, Alborn WE, Troutt JS, Cao G, Konrad RJ.
Atorvastatin increases human serum levels of proprotein convertase
subtilisin/kexin type 9. J Lipid Res 2008; 49: 394-398 [PMID:
18033751 DOI: 10.1194/jlr.M700437-JLR200]
26 Awan Z, Seidah NG, MacFadyen JG, Benjannet S, Chasman DI,
Ridker PM, Genest J. Rosuvastatin, proprotein convertase subtilisin/
kexin type 9 concentrations, and LDL cholesterol response: the
JUPITER trial. Clin Chem 2012; 58: 183-189 [PMID: 22065156 DOI:
10.1373/clinchem.2011.172932]
27 Welder G, Zineh I, Pacanowski MA, Troutt JS, Cao G, Konrad RJ.
High-dose atorvastatin causes a rapid sustained increase in human
serum PCSK9 and disrupts its correlation with LDL cholesterol. J
Lipid Res 2010; 51: 2714-2721 [PMID: 20525997 DOI: 10.1194/jlr.
M008144]
28 Davignon J, Dubuc G. Statins and ezetimibe modulate plasma
proprotein convertase subtilisin kexin-9 (PCSK9) levels. Trans Am
Clin Climatol Assoc 2009; 120: 163-173 [PMID: 19768174]
29 Nilsson LM, Abrahamsson A, Sahlin S, Gustafsson U, Angelin B,
Parini P, Einarsson C. Bile acids and lipoprotein metabolism: effects of
cholestyramine and chenodeoxycholic acid on human hepatic mRNA
expression. Biochem Biophys Res Commun 2007; 357: 707-711
[PMID: 17448444 DOI: 10.1016/j.bbrc.2007.03.196]
30 Jia L, Betters JL, Yu L. Niemann-pick C1-like 1 (NPC1L1) protein
in intestinal and hepatic cholesterol transport. Annu Rev Physiol
2011; 73: 239-259 [PMID: 20809793 DOI: 10.1146/annurev-
physiol-012110-142233]
31 Nassir F, Wilson B, Han X, Gross RW, Abumrad NA. CD36 is
important for fatty acid and cholesterol uptake by the proximal but
McBride P, Schwartz JS, Shero ST, Smith SC, Watson K, Wilson
PW. 2013 ACC/AHA guideline on the treatment of blood cholesterol
to reduce atherosclerotic cardiovascular risk in adults: a report of the
American College of Cardiology/American Heart Association Task
Force on Practice Guidelines. J Am Coll Cardiol 2014; 63: 2889-2934
[PMID: 24239923 DOI: 10.1016/j.jacc.2013.11.002]
2 Brugts JJ, Yetgin T, Hoeks SE, Gotto AM, Shepherd J, Westendorp
RG, de Craen AJ, Knopp RH, Nakamura H, Ridker P, van Domburg
R, Deckers JW. The benets of statins in people without established
cardiovascular disease but with cardiovascular risk factors: meta-
analysis of randomised controlled trials. BMJ 2009; 338: b2376 [PMID:
19567909 DOI: 10.1136/bmj.b2376]
3 Taylor F, Huffman MD, Macedo AF, Moore TH, Burke M, Davey
Smith G, Ward K, Ebrahim S. Statins for the primary prevention
of cardiovascular disease. Cochrane Database Syst Rev 2013; (1):
CD004816 [PMID: 23440795 DOI: 10.1002/14651858.CD004816.
pub5]
4 Shelness GS, Sellers JA. Very-low-density lipoprotein assembly and
secretion. Curr Opin Lipidol 2001; 12: 151-157 [PMID: 11264986]
5 Goldstein JL, Brown MS, Anderson RG, Russell DW, Schneider WJ.
Receptor-mediated endocytosis: concepts emerging from the LDL
receptor system. Annu Rev Cell Biol 1985; 1: 1-39 [PMID: 2881559
DOI: 10.1146/annurev.cb.01.110185.000245]
6 Stancu C, Sima A. Statins: mechanism of action and effects. J Cell
Mol Med 2001; 5: 378-387 [PMID: 12067471]
7 Seidah NG, Benjannet S, Wickham L, Marcinkiewicz J, Jasmin SB,
Stifani S, Basak A, Prat A, Chretien M. The secretory proprotein
convertase neural apoptosis-regulated convertase 1 (NARC-1):
liver regeneration and neuronal differentiation. Proc Natl Acad
Sci USA 2003; 100: 928-933 [PMID: 12552133 DOI: 10.1073/
pnas.0335507100]
8 Abifadel M, Varret M, Rabès JP, Allard D, Ouguerram K, Devillers
M, Cruaud C, Benjannet S, Wickham L, Erlich D, Derré A, Villéger
L, Farnier M, Beucler I, Bruckert E, Chambaz J, Chanu B, Lecerf
JM, Luc G, Moulin P, Weissenbach J, Prat A, Krempf M, Junien
C, Seidah NG, Boileau C. Mutations in PCSK9 cause autosomal
dominant hypercholesterolemia. Nat Genet 2003; 34: 154-156 [PMID:
12730697 DOI: 10.1038/ng1161]
9 Lambert G, Sjouke B, Choque B, Kastelein JJ, Hovingh GK. The
PCSK9 decade. J Lipid Res 2012; 53: 2515-2524 [PMID: 22811413
DOI: 10.1194/jlr.R026658]
10 Tibolla G, Norata GD, Artali R, Meneghetti F, Catapano AL.
Proprotein convertase subtilisin/kexin type 9 (PCSK9): from
structure-function relation to therapeutic inhibition. Nutr Metab
Cardiovasc Dis 2011; 21: 835-843 [PMID: 21943799 DOI: 10.1016/
j.numecd.2011.06.002]
11 Davignon J, Dubuc G, Seidah NG. The influence of PCSK9
polymorphisms on serum low-density lipoprotein cholesterol and risk
of atherosclerosis. Curr Atheroscler Rep 2010; 12: 308-315 [PMID:
20623344 DOI: 10.1007/s11883-010-0123-6]
12 Cohen J, Pertsemlidis A, Kotowski IK, Graham R, Garcia CK, Hobbs
HH. Low LDL cholesterol in individuals of African descent resulting
from frequent nonsense mutations in PCSK9. Nat Genet 2005; 37:
161-165 [PMID: 15654334 DOI: 10.1038/ng1509]
13 Cohen JC, Boerwinkle E, Mosley TH, Hobbs HH. Sequence
variations in PCSK9, low LDL, and protection against coronary heart
disease. N Engl J Med 2006; 354: 1264-1272 [PMID: 16554528 DOI:
10.1056/NEJMoa054013]
14 Denis M, Marcinkiewicz J, Zaid A, Gauthier D, Poirier S,
Lazure C, Seidah NG, Prat A. Gene inactivation of proprotein
convertase subtilisin/kexin type 9 reduces atherosclerosis in mice.
Circulation 2012; 125: 894-901 [PMID: 22261195 DOI: 10.1161/
CIRCULATIONAHA.111.057406]
15 Maxwell KN, Soccio RE, Duncan EM, Sehayek E, Breslow JL.
Novel putative SREBP and LXR target genes identied by microarray
analysis in liver of cholesterol-fed mice. J Lipid Res 2003; 44:
2109-2119 [PMID: 12897189 DOI: 10.1194/jlr.M300203-JLR200]
16 Horton JD, Shah NA, Warrington JA, Anderson NN, Park SW,
Brown MS, Goldstein JL. Combined analysis of oligonucleotide
microarray data from transgenic and knockout mice identies direct
Chaudhary R
et al
. Review on PCSK9 inhibitors
89 February 26, 2017
|
Volume 9
|
Issue 2
|
WJC
|
www.wjgnet.com
subtilisin/kexin type 9 (PCSK9) and the concentration of serum LDL
cholesterol in healthy volunteers: a randomised, single-blind, placebo-
controlled, phase 1 trial. Lancet 2014; 383: 60-68 [PMID: 24094767
DOI: 10.1016/S0140-6736(13)61914-5]
45 Budha NR, Leabman M, Jin JY, Wada DR, Baruch A, Peng K,
Tingley WG, Davis JD. Modeling and Simulation to Support Phase
2 Dose Selection for RG7652, a Fully Human Monoclonal Antibody
Against Proprotein Convertase Subtilisin/Kexin Type 9. AAPS J 2015;
17: 881-890 [PMID: 25823668 DOI: 10.1208/s12248-015-9750-8]
46 Zhang L, McCabe T, Condra JH, Ni YG, Peterson LB, Wang W,
Strack AM, Wang F, Pandit S, Hammond H, Wood D, Lewis D,
Rosa R, Mendoza V, Cumiskey AM, Johns DG, Hansen BC, Shen X,
Geoghagen N, Jensen K, Zhu L, Wietecha K, Wisniewski D, Huang L,
Zhao JZ, Ernst R, Hampton R, Haytko P, Ansbro F, Chilewski S, Chin
J, Mitnaul LJ, Pellacani A, Sparrow CP, An Z, Strohl W, Hubbard
B, Plump AS, Blom D, Sitlani A. An anti-PCSK9 antibody reduces
LDL-cholesterol on top of a statin and suppresses hepatocyte SREBP-
regulated genes. Int J Biol Sci 2012; 8: 310-327 [PMID: 22355267
DOI: 10.7150/ijbs.3524]
47 Chaparro-Riggers J, Liang H, DeVay RM, Bai L, Sutton JE, Chen W,
Geng T, Lindquist K, Casas MG, Boustany LM, Brown CL, Chabot
J, Gomes B, Garzone P, Rossi A, Strop P, Shelton D, Pons J, Rajpal A.
Increasing serum half-life and extending cholesterol lowering in vivo
by engineering antibody with pH-sensitive binding to PCSK9. J Biol
Chem 2012; 287: 11090-11097 [PMID: 22294692 DOI: 10.1074/jbc.
M111.319764]
48 Gumbiner B. Effects of 12 weeks of treatment with RN316
(PF-04950615), a humanized IgG2_a monoclonal antibody binding
proprotein convertase subtilisin kexin type 9, in hypercholesterolemic
subjects on high and maximal dose statins. Presented at Am Heart
Assoc Sci Sess, Los Angeles, 2012
49 Dadu RT, Ballantyne CM. Lipid lowering with PCSK9 inhibitors.
Nat Rev Cardiol 2014; 11: 563-575 [PMID: 24958078 DOI: 10.1038/
nrcardio.2014.84]
50 Pfizer Announces Positive Topline Results from Second Phase 3
Lipid-Lowering Study Evaluating Bococizumab/Pfizer: One of the
world's premier biopharmaceutical companies. 2016. [cited 2016 Oct
12]. Available from: URL: http://www.pzer.com/news/press-release/
press-release-detail/pzer_announces_positive_topline_results_from_
second_phase_3_lipid_lowering_study_evaluating_bococizumab
51 Stein EA, Kasichayanula S, Turner T, Kranz T, Arumugam U,
Biernat L, Lee J. LDL cholesterol reduction with bms-962476, an
adnectin inhibitor of PCSK9: results of a single ascending dose
study. J Am Coll Cardiol 2014; 63 (12_S): A1372 [DOI: 10.1016/
S0735-1097(14)61372-3]
52 Mitchell T, Chao G, Sitkoff D, Lo F, Monshizadegan H, Meyers D,
Low S, Russo K, DiBella R, Denhez F, Gao M, Myers J, Duke G,
Witmer M, Miao B, Ho SP, Khan J, Parker RA. Pharmacologic prole
of the Adnectin BMS-962476, a small protein biologic alternative to
PCSK9 antibodies for low-density lipoprotein lowering. J Pharmacol
Exp Ther 2014; 350: 412-424 [PMID: 24917546 DOI: 10.1124/
jpet.114.214221]
53 Kosenko T, Golder M, Leblond G, Weng W, Lagace TA. Low density
lipoprotein binds to proprotein convertase subtilisin/kexin type-9
(PCSK9) in human plasma and inhibits PCSK9-mediated low density
lipoprotein receptor degradation. J Biol Chem 2013; 288: 8279-8288
[PMID: 23400816 DOI: 10.1074/jbc.M112.421370]
54 Huang Y, Palyha O, Castro-Perez J, Lee A, Ai X, Ha S, Pinto
S. Abstract 15853: Identification of Novel Sites Required for
PCSK9 Autocatalytic Cleavage. Circulation 2015; 132 (Suppl 3):
A15853-A15853
55 Stein EA, Mellis S, Yancopoulos GD, Stahl N, Logan D, Smith WB,
Lisbon E, Gutierrez M, Webb C, Wu R, Du Y, Kranz T, Gasparino E,
Swergold GD. Effect of a monoclonal antibody to PCSK9 on LDL
cholesterol. N Engl J Med 2012; 366: 1108-1118 [PMID: 22435370
DOI: 10.1056/NEJMoa1105803]
56 McKenney JM, Koren MJ, Kereiakes DJ, Hanotin C, Ferrand AC,
Stein EA. Safety and efcacy of a monoclonal antibody to proprotein
convertase subtilisin/kexin type 9 serine protease, SAR236553/
REGN727, in patients with primary hypercholesterolemia receiving
not distal intestine. J Biol Chem 2007; 282: 19493-19501 [PMID:
17507371 DOI: 10.1074/jbc.M703330200]
32 Levy E, Ben Djoudi Ouadda A, Spahis S, Sane AT, Garofalo C,
Grenier É, Emonnot L, Yara S, Couture P, Beaulieu JF, Ménard
D, Seidah NG, Elchebly M. PCSK9 plays a significant role in
cholesterol homeostasis and lipid transport in intestinal epithelial cells.
Atherosclerosis 2013; 227: 297-306 [PMID: 23422832 DOI: 10.1016/
j.atherosclerosis.2013.01.023]
33 Roubtsova A, Munkonda MN, Awan Z, Marcinkiewicz J,
Chamberland A, Lazure C, Cianflone K, Seidah NG, Prat A.
Circulating proprotein convertase subtilisin/kexin 9 (PCSK9) regulates
VLDLR protein and triglyceride accumulation in visceral adipose
tissue. Arterioscler Thromb Vasc Biol 2011; 31: 785-791 [PMID:
21273557 DOI: 10.1161/ATVBAHA.110.220988]
34 Labonté P, Begley S, Guévin C, Asselin MC, Nassoury N, Mayer G,
Prat A, Seidah NG. PCSK9 impedes hepatitis C virus infection in vitro
and modulates liver CD81 expression. Hepatology 2009; 50: 17-24
[PMID: 19489072 DOI: 10.1002/hep.22911]
35 Mbikay M, Sirois F, Mayne J, Wang GS, Chen A, Dewpura T,
Prat A, Seidah NG, Chretien M, Scott FW. PCSK9-deficient mice
exhibit impaired glucose tolerance and pancreatic islet abnormalities.
FEBS Lett 2010; 584: 701-706 [PMID: 20026049 DOI: 10.1016/
j.febslet.2009.12.018]
36 Gr ah am MJ, Lemon idis KM, Whipp le CP, Subrama niam
A, Monia BP, Crooke ST, Crooke RM. Antisense inhibition of
proprotein convertase subtilisin/kexin type 9 reduces serum LDL
in hyperlipidemic mice. J Lipid Res 2007; 48: 763-767 [PMID:
17242417 DOI: 10.1194/jlr.C600025-JLR200]
37 Frank-Kamenetsky M, Grefhorst A, Anderson NN, Racie TS,
Bramlage B, Akinc A, Butler D, Charisse K, Dorkin R, Fan Y, Gamba-
Vitalo C, Hadwiger P, Jayaraman M, John M, Jayaprakash KN, Maier
M, Nechev L, Rajeev KG, Read T, Röhl I, Soutschek J, Tan P, Wong J,
Wang G, Zimmermann T, de Fougerolles A, Vornlocher HP, Langer R,
Anderson DG, Manoharan M, Koteliansky V, Horton JD, Fitzgerald K.
Therapeutic RNAi targeting PCSK9 acutely lowers plasma cholesterol
in rodents and LDL cholesterol in nonhuman primates. Proc Natl Acad
Sci USA 2008; 105: 11915-11920 [PMID: 18695239 DOI: 10.1073/
pnas.0805434105]
38 Duff CJ, Scott MJ, Kirby IT, Hutchinson SE, Martin SL, Hooper NM.
Antibody-mediated disruption of the interaction between PCSK9 and
the low-density lipoprotein receptor. Biochem J 2009; 419: 577-584
[PMID: 19196236 DOI: 10.1042/BJ20082407]
39 Shan L, Pang L, Zhang R, Murgolo NJ, Lan H, Hedrick JA. PCSK9
binds to multiple receptors and can be functionally inhibited by an
EGF-A peptide. Biochem Biophys Res Commun 2008; 375: 69-73
[PMID: 18675252 DOI: 10.1016/j.bbrc.2008.07.106]
40 Lindholm MW, Elmén J, Fisker N, Hansen HF, Persson R, Møller
MR, Rosenbohm C, Ørum H, Straarup EM, Koch T. PCSK9 LNA
antisense oligonucleotides induce sustained reduction of LDL
cholesterol in nonhuman primates. Mol Ther 2012; 20: 376-381
[PMID: 22108858 DOI: 10.1038/mt.2011.260]
41 Hooper AJ, Burnett JR. Anti-PCSK9 therapies for the treatment
of hypercholesterolemia. Expert Opin Biol Ther 2013; 13: 429-435
[PMID: 23240807 DOI: 10.1517/14712598.2012.748743]
42 Gupta N, Fisker N, Asselin MC, Lindholm M, Rosenbohm C,
Ørum H, Elmén J, Seidah NG, Straarup EM. A locked nucleic acid
antisense oligonucleotide (LNA) silences PCSK9 and enhances LDLR
expression in vitro and in vivo. PLoS One 2010; 5: e10682 [PMID:
20498851 DOI: 10.1371/journal.pone.0010682]
43 van Poelgeest EP, Hodges MR, Moerland M, Tessier Y, Levin AA,
Persson R, Lindholm MW, Dumong Erichsen K, Ørum H, Cohen AF,
Burggraaf J. Antisense-mediated reduction of proprotein convertase
subtilisin/kexin type 9 (PCSK9): a first-in-human randomized,
placebo-controlled trial. Br J Clin Pharmacol 2015; 80: 1350-1361
[PMID: 26261033 DOI: 10.1111/bcp.12738]
44 Fitzgerald K, Frank-Kamenetsky M, Shulga-Morskaya S, Liebow A,
Bettencourt BR, Sutherland JE, Hutabarat RM, Clausen VA, Karsten
V, Cehelsky J, Nochur SV, Kotelianski V, Horton J, Mant T, Chiesa J,
Ritter J, Munisamy M, Vaishnaw AK, Gollob JA, Simon A. Effect of
an RNA interference drug on the synthesis of proprotein convertase
Chaudhary R
et al
. Review on PCSK9 inhibitors
90 February 26, 2017
|
Volume 9
|
Issue 2
|
WJC
|
www.wjgnet.com
69 Dias CS, Shaywitz AJ, Wasserman SM, Smith BP, Gao B, Stolman
DS, Crispino CP, Smirnakis KV, Emery MG, Colbert A, Gibbs JP,
Retter MW, Cooke BP, Uy ST, Matson M, Stein EA. Effects of AMG
145 on low-density lipoprotein cholesterol levels: results from 2
randomized, double-blind, placebo-controlled, ascending-dose phase
1 studies in healthy volunteers and hypercholesterolemic subjects on
statins. J Am Coll Cardiol 2012; 60: 1888-1898 [PMID: 23083772
DOI: 10.1016/j.jacc.2012.08.986]
70 Giugliano RP, Desai NR, Kohli P, Rogers WJ, Somaratne R, Huang
F, Liu T, Mohanavelu S, Hoffman EB, McDonald ST, Abrahamsen
TE, Wasserman SM, Scott R, Sabatine MS. Efficacy, safety, and
tolerability of a monoclonal antibody to proprotein convertase
subtilisin/kexin type 9 in combination with a statin in patients with
hypercholesterolaemia (LAPLACE-TIMI 57): a randomised, placebo-
controlled, dose-ranging, phase 2 study. Lancet 2012; 380: 2007-2017
[PMID: 23141813 DOI: 10.1016/S0140-6736(12)61770-X]
71 Koren MJ, Scott R, Kim JB, Knusel B, Liu T, Lei L, Bolognese M,
Wasserman SM. Efficacy, safety, and tolerability of a monoclonal
antibody to proprotein convertase subtilisin/kexin type 9 as
monotherapy in patients with hypercholesterolaemia (MENDEL):
a randomised, double-blind, placebo-controlled, phase 2 study.
Lancet 2012; 380: 1995-2006 [PMID: 23141812 DOI: 10.1016/
S0140-6736(12)61771-1]
72 Raal F, Scott R, Somaratne R, Bridges I, Li G, Wasserman SM,
Stein EA. Low-density lipoprotein cholesterol-lowering effects
of AMG 145, a monoclonal antibody to proprotein convertase
subtilisin/kexin type 9 serine protease in patients with heterozygous
familial hypercholesterolemia: the Reduction of LDL-C with PCSK9
Inhibition in Heterozygous Familial Hypercholesterolemia Disorder
(RUTHERFORD) randomized trial. Circulation 2012; 126: 2408-2417
[PMID: 23129602 DOI: 10.1161/CIRCULATIONAHA.112.144055]
73 Sullivan D, Olsson AG, Scott R, Kim JB, Xue A, Gebski V,
Wasserman SM, Stein EA. Effect of a monoclonal antibody to PCSK9
on low-density lipoprotein cholesterol levels in statin-intolerant
patients: the GAUSS randomized trial. JAMA 2012; 308: 2497-2506
[PMID: 23128163 DOI: 10.1001/jama.2012.25790]
74 Stein EA, Honarpour N, Wasserman SM, Xu F, Scott R, Raal FJ.
Effect of the proprotein convertase subtilisin/kexin 9 monoclonal
antibody, AMG 145, in homozygous familial hypercholesterolemia.
Circulation 2013; 128: 2113-2120 [PMID: 24014831 DOI: 10.1161/
CIRCULATIONAHA.113.004678]
75 Robinson JG, Nedergaard BS, Rogers WJ, Fialkow J, Neutel JM,
Ramstad D, Somaratne R, Legg JC, Nelson P, Scott R, Wasserman
SM, Weiss R. Effect of evolocumab or ezetimibe added to moderate-
or high-intensity statin therapy on LDL-C lowering in patients with
hypercholesterolemia: the LAPLACE-2 randomized clinical trial.
JAMA 2014; 311: 1870-1882 [PMID: 24825642 DOI: 10.1001/
jama.2014.4030]
76 Kiyosue A, Honarpour N, Xue A, Wasserman S, Hirayama A. Effects
of evolocumab (AMG 145) in hypercholesterolemic, statin-treated,
japanese patients at high cardiovascular risk: results from the phase iii
yukawa 2 study. J Am Coll Cardiol 2015; 65 (10_S) [DOI: 10.1016/
S0735-1097(15)61369-9]
77 Koren MJ, Lundqvist P, Bolognese M, Neutel JM, Monsalvo
ML, Yang J, Kim JB, Scott R, Wasserman SM, Bays H. Anti-
PCSK9 monotherapy for hypercholesterolemia: the MENDEL-2
randomized, controlled phase III clinical trial of evolocumab. J Am
Coll Cardiol 2014; 63: 2531-2540 [PMID: 24691094 DOI: 10.1016/
j.jacc.2014.03.018]
78 Stroes E, Colquhoun D, Sullivan D, Civeira F, Rosenson RS, Watts
GF, Bruckert E, Cho L, Dent R, Knusel B, Xue A, Scott R, Wasserman
SM, Rocco M. Anti-PCSK9 antibody effectively lowers cholesterol
in patients with statin intolerance: the GAUSS-2 randomized,
placebo-controlled phase 3 clinical trial of evolocumab. J Am Coll
Cardiol 2014; 63: 2541-2548 [PMID: 24694531 DOI: 10.1016/
j.jacc.2014.03.019]
79 Nissen SE, Stroes E, Dent-Acosta RE, Rosenson RS, Lehman SJ,
Sattar N, Preiss D, Bruckert E, Ceška R, Lepor N, Ballantyne CM,
Gouni-Berthold I, Elliott M, Brennan DM, Wasserman SM, Somaratne
R, Scott R, Stein EA. Efficacy and Tolerability of Evolocumab vs
ongoing stable atorvastatin therapy. J Am Coll Cardiol 2012; 59:
2344-2353 [PMID: 22463922 DOI: 10.1016/j.jacc.2012.03.007]
57 Roth EM, McKen ney JM, Hanoti n C, As set G, Stein EA.
Atorvastatin with or without an antibody to PCSK9 in primary
hypercholesterolemia. N Engl J Med 2012; 367: 1891-1900 [PMID:
23113833 DOI: 10.1056/NEJMoa1201832]
58 Stein EA, Gipe D, Bergeron J, Gaudet D, Weiss R, Dufour R, Wu
R, Pordy R. Effect of a monoclonal antibody to PCSK9, REGN727/
SAR236553, to reduce low-density lipoprotein cholesterol in patients
with heterozygous familial hypercholesterolaemia on stable statin dose
with or without ezetimibe therapy: a phase 2 randomised controlled
trial. Lancet 2012; 380: 29-36 [PMID: 22633824 DOI: 10.1016/
S0140-6736(12)60771-5]
59 Regeneron and Sano Announce Positive Topline Results from First
Phase 3 Trials Evaluating Monthly Dosing of Alirocumab in Patients
with Hypercholesterolemia. (01/09/205). [accessed 2015 Dec 9].
Available from: URL: http://www.prnewswire.com/news-releases/
regeneron-and-sanofi-announce-positive-topline-results-from-first-
phase-3-trials-evaluating-monthly-dosing-of-alirocumab-in-patients-
with-hypercholesterolemia-300017492.html
60 Colhoun HM, Robinson JG, Farnier M, Cariou B, Blom D,
Kereiakes DJ, Lorenzato C, Pordy R, Chaudhari U. Efficacy
and safety of alirocumab, a fully human PCSK9 monoclonal
antibody, in high cardiovascular risk patients with poorly controlled
hypercholesterolemia on maximally tolerated doses of statins:
rationale and design of the ODYSSEY COMBO I and II trials. BMC
Cardiovasc Disord 2014; 14: 121 [PMID: 25240705 DOI: 10.1186/14
71-2261-14-121]
61 Bays H, Gaudet D, Weiss R, Ruiz JL, Watts GF, Gouni-Berthold I,
Robinson J, Zhao J, Hanotin C, Donahue S. Alirocumab as Add-On
to Atorvastatin Versus Other Lipid Treatment Strategies: ODYSSEY
OPTIONS I Randomized Trial. J Clin Endocrinol Metab 2015; 100:
3140-3148 [PMID: 26030325 DOI: 10.1210/jc.2015-1520]
62 Robinson JG, Colhoun HM, Bays HE, Jones PH, Du Y, Hanotin
C, Donahue S. Efcacy and safety of alirocumab as add-on therapy
in high-cardiovascular-risk patients with hypercholesterolemia not
adequately controlled with atorvastatin (20 or 40 mg) or rosuvastatin
(10 or 20 mg): design and rationale of the ODYSSEY OPTIONS
Studies. Clin Cardiol 2014; 37: 597-604 [PMID: 25269777 DOI:
10.1002/clc.22327]
63 The American College of Cardiology. Odyssey Options I and II.
(11/19/2014). [accessed 2015 Dec 9]. Available from: URL: http://www.
acc.org/Latest-in-Cardiology/Clinical-Trials/2014/11/18/15/20/ODYSSEY-
OPTIONS-I-and-II
64 Roth EM, Taskinen MR, Ginsberg H, Kastelein J, Colhoun HM,
Merlet L, Baccara-Dinet MT. A 24-week study of alirocumab as
monotherapy versus ezetimibe: the rst phase 3 data of a proprotein
convertase subtilisin/kexin type 9 inhibitor. J Am Coll Cardiol 2014;
63 (12_S) [DOI: 10.1016/S0735-1097(14)61370-X]
65 Moriarty PM, Thompson PD, Cannon CP, Guyton JR, Bergeron
J, Zieve FJ, Bruckert E, Jacobson TA, Kopecky SL, Baccara-Dinet
MT, Du Y, Pordy R, Gipe DA. Efcacy and safety of alirocumab vs
ezetimibe in statin-intolerant patients, with a statin rechallenge arm:
The ODYSSEY ALTERNATIVE randomized trial. J Clin Lipidol
2015; 9: 758-769 [PMID: 26687696 DOI: 10.1016/j.jacl.2015.08.006]
66 Kastelein JJ, Ginsberg HN, Langslet G, Hovingh GK, Ceska R,
Dufour R, Blom D, Civeira F, Krempf M, Lorenzato C, Zhao J, Pordy R,
Baccara-Dinet MT, Gipe DA, Geiger MJ, Farnier M. ODYSSEY FH
I and FH II: 78 week results with alirocumab treatment in 735 patients
with heterozygous familial hypercholesterolaemia. Eur Heart J 2015;
36: 2996-3003 [PMID: 26330422 DOI: 10.1093/eurheartj/ehv370]
67 The American College of Cardiology. Odyssey High FH. (11/19/2014).
[accessed 2015 Dec 9]. Available from: URL: http://www.acc.org/latest-
in-cardiology/clinical-trials/2014/11/18/15/23/odyssey-high-fh
68 Robinson JG, Farnier M, Krempf M, Bergeron J, Luc G, Averna M,
Stroes ES, Langslet G, Raal FJ, El Shahawy M, Koren MJ, Lepor
NE, Lorenzato C, Pordy R, Chaudhari U, Kastelein JJ. Efcacy and
safety of alirocumab in reducing lipids and cardiovascular events. N
Engl J Med 2015; 372: 1489-1499 [PMID: 25773378 DOI: 10.1056/
NEJMoa1501031]
Chaudhary R
et al
. Review on PCSK9 inhibitors
91 February 26, 2017
|
Volume 9
|
Issue 2
|
WJC
|
www.wjgnet.com
297-301 [PMID: 25256660 DOI: 10.1111/ 1755-5922.12093]
86 Cicero AF, Colletti A, Borghi C. Profile of evolocumab and its
potential in the treatment of hyperlipidemia. Drug Des Devel Ther
2015; 9: 3073-3082 [PMID: 26109850 DOI: 10.2147/DDDT.S67498]
87 Waldmann TA, Strober W. Metabolism of immunoglobulins. Prog
Allergy 1969; 13: 1-110 [PMID: 4186070]
88 Swiger K. Early Evidence Linking PCSK9 Inhibitors to
Neurocognitive Adverse Events: Does Correlation Imply Causation?
American College of Cardiology2015. [cited 2016 Oct 12].
Available from: URL: http://www.acc.org/latest-in-cardiology/
articles/2015/06/01/12/36/early-evidence-linking-pcsk9-inhibitors-to-
neurocognitive-adverse-events
89 Catapano AL, Papadopoulos N. The safety of therapeutic monoclonal
antibodies: implications for cardiovascular disease and targeting the
PCSK9 pathway. Atherosclerosis 2013; 228: 18-28 [PMID: 23466067
DOI: 10.1016/j.atherosclerosis.2013.01.044]
90 Jarcho JA, Keaney JF. Proof That Lower Is Better--LDL Cholesterol
and IMPROVE-IT. N Engl J Med 2015; 372: 2448-2450 [PMID:
26039520 DOI: 10.1056/NEJMe1507041]
91 Minder CM, Blumenthal RS, Blaha MJ. Statins for primary
prevention of cardiovascular disease: the benets outweigh the risks.
Curr Opin Cardiol 2013; 28: 554-560 [PMID: 23928920 DOI:
10.1097/HCO.0b013e32836429e6]
92 Heart Protection Study Collaborative Group. MRC/BHF Heart
Protection Study of cholesterol lowering with simvastatin in 20,536 high-
risk individuals: a randomised placebo-controlled trial. Lancet 2002; 360:
7-22 [PMID: 12114036 DOI: 10.1016/S0140-6736(02)09327-3]
93 Anderson JL, Heidenreich PA, Barnett PG, Creager MA, Fonarow
GC, Gibbons RJ, Halperin JL, Hlatky MA, Jacobs AK, Mark DB,
Masoudi FA, Peterson ED, Shaw LJ. ACC/AHA statement on cost/
value methodology in clinical practice guidelines and performance
measures: a report of the American College of Cardiology/American
Heart Association Task Force on Performance Measures and Task
Force on Practice Guidelines. J Am Coll Cardiol 2014; 63: 2304-2322
[PMID: 24681044 DOI: 10.1016/j.jacc.2014.03.016]
94 PCSK9 Inhibitors for Treatment of High Cholesterol. New England
Comparative Effectiveness Public Advisory Council. (n.d.). Available
from: URL: http://cepac.icer-review.org/adaptations/cholesterol/
P- Reviewer: Aronow WS, Hohenegger M, Kasai T, Labonte P, Sun CK
S- Editor: Ji FF L- Editor: A E- Editor: Lu YJ
Ezetimibe in Patients With Muscle-Related Statin Intolerance: The
GAUSS-3 Randomized Clinical Trial. JAMA 2016; 315: 1580-1590
[PMID: 27039291 DOI: 10.1001/jama.2016.3608]
80 Blom DJ, Hala T, Bolognese M, Lillestol MJ, Toth PD, Burgess L,
Ceska R, Roth E, Koren MJ, Ballantyne CM, Monsalvo ML, Tsirtsonis
K, Kim JB, Scott R, Wasserman SM, Stein EA. A 52-week placebo-
controlled trial of evolocumab in hyperlipidemia. N Engl J Med 2014;
370: 1809-1819 [PMID: 24678979 DOI: 10.1056/NEJMoa1316222]
81 Raal FJ, Stein EA, Dufour R, Turner T, Civeira F, Burgess L, Langslet
G, Scott R, Olsson AG, Sullivan D, Hovingh GK, Cariou B, Gouni-
Berthold I, Somaratne R, Bridges I, Scott R, Wasserman SM, Gaudet
D. PCSK9 inhibition with evolocumab (AMG 145) in heterozygous
familial hypercholesterolaemia (RUTHERFORD-2): a randomised,
double-blind, placebo-controlled trial. Lancet 2015; 385: 331-340
[PMID: 25282519 DOI: 10.1016/S0140-6736(14)61399-4]
82 Raal FJ, Honarpour N, Blom DJ, Hovingh GK, Xu F, Scott R,
Wasserman SM, Stein EA. Inhibition of PCSK9 with evolocumab
in homozygous familial hypercholesterolaemia (TESLA Part B): a
randomised, double-blind, placebo-controlled trial. Lancet 2015; 385:
341-350 [PMID: 25282520 DOI: 10.1016/S0140-6736(14)61374-X]
83 Koren MJ, Giugliano RP, Raal FJ, Sullivan D, Bolognese M, Langslet
G, Civeira F, Somaratne R, Nelson P, Liu T, Scott R, Wasserman
SM, Sabatine MS. Efcacy and safety of longer-term administration
of evolocumab (AMG 145) in patients with hypercholesterolemia:
52-week results from the Open-Label Study of Long-Term Evaluation
Against LDL-C (OSLER) randomized trial. Circulation 2014;
129: 234-243 [PMID: 24255061 DOI: 10.1161/CIRCULATIO
NAHA.113.007012]
84 Sabatine MS, Giugliano RP, Wiviott SD, Raal FJ, Blom DJ, Robinson
J, Ballantyne CM, Somaratne R, Legg J, Wasserman SM, Scott R,
Koren MJ, Stein EA. Efcacy and safety of evolocumab in reducing
lipids and cardiovascular events. N Engl J Med 2015; 372: 1500-1509
[PMID: 25773607 DOI: 10.1056/NEJMoa1500858]
85 Lunven C, Paehler T, Poitiers F, Brunet A, Rey J, Hanotin C, Sasiela
WJ. A randomized study of the relative pharmacokinetics, pharma-
codynamics, and safety of alirocumab, a fully human monoclonal
antibody to PCSK9, after single subcutaneous administration at three
different injection sites in healthy subjects. Cardiovasc Ther 2014; 32:
Chaudhary R
et al
. Review on PCSK9 inhibitors
© 2017 Baishideng Publishing Group Inc. All rights reserved.
Published by Baishideng Publishing Group Inc
8226 Regency Drive, Pleasanton, CA 94588, USA
Telephone: +1-925-223-8242
Fax: +1-925-223-8243
E-mail: bpgofce@wjgnet.com
Help Desk: http://www.wjgnet.com/esps/helpdesk.aspx
http://www.wjgnet.com
... Proprotein convertase subtilsin-kexin type 9 (PCSK9) inhibitors represent a relatively newer category of hypolipidemic treatment, comprising two entirely humanized monoclonal antibodies, evolocumab and alirocumab, which are designed to bind to the freecirculating PCSK9 protein [57,58]. This enzyme (hepatic protease) plays a pivotal role in facilitating the degradation of LDL-receptors (LDL-R) in the liver by internalizing them into hepatic lysosomes, promoting their destruction, and hindering their return to the hepatocyte membrane [58]. ...
... The improved hepatic clearance of LDL-C after PCSK9 inhibitor administration leads to a remarkable reduction (50-60%) in plasma LDL-C concentrations as an add-on therapy to statins [57]. PCSK9 inhibitors have been associated with significant CV benefits [59] and might be an effective alternative lipid-lowering medication among patients with CAD and DM [60]. ...
Article
Full-text available
Diabetic atherosclerosis is a complex process that is characterized by diffuse and unstable lesions increasing 2–4-fold the risk of adverse cardiovascular (CV) events. Diabetic dyslipidemia has a predominant role in coronary artery disease (CAD) and has been the target of classical and emerging pharmaceutical agents with established or promising CV benefits. The aim of the present narrative review was to summarize the effects of classical and novel lipid-lowering pharmaceutical agents on lipid profile and CV outcomes in diabetic patients with established CAD or high risk of CAD. Statins remain the first-line treatment for all diabetic patients since they considerably ameliorate lipid parameters and non-lipid CV risk factors, leading to reduced CV morbidity and mortality. Complementary to statins, ezetimibe exerts lipid-lowering properties with modest but significant reductions in major adverse cardiovascular events (MACEs) and CV mortality. PCSK9 inhibitors considerably reduce LDL-C levels and lower MACEs in diabetic patients. On the other hand, fibrates may confer a very modest decline in MACE incidence, while the CV impact of omega-3 fatty acids is promising but remains questionable. Bempedoic acid and inclisiran have a potential therapeutic role in the management of diabetic dyslipidemia, but this is still not adequately documented. Given the heightened CV risk among individuals with diabetes, more decisive results would be of great importance in the utility of all these drugs.
... PCSK9, the ninth member of the proprotein convertase family, is a serine protease that caught the attention of the scientific community in 2003 when the discovery of the first natural mutants of PCSK9 revealed the implication of an as-yet-unknown actor in cholesterol homeostasis [4,5]. PCSK9 is mainly expressed on hepatocytes surface and has been shown to act both intracellularly playing a role as a chaperone in the degradation of the LDL receptor (LDLR), as well as a secreted factor promoting LDLR internalization from the hepatocellular surface [6]. PCSK9 regulates the degradation of the LDLR in response to cholesterol levels within the cell [7]. ...
Article
Full-text available
Proprotein convertase subtilisin/kexin type 9 (PCSK9) plays a crucial role in the modulation of lipid metabolism as a critical negative regulator of hepatic low-density lipoprotein receptor (LDLR) levels and circulating low-density lipoprotein (LDL) clearance. Numerous gain-of-function (GOF) mutations in PCSK9 have been identified as causing familial hypercholesterolemia (FH) by reducing LDLR levels, and loss-of-function (LOF) mutations associated with a hypercholesterolemia phenotype protective against atherosclerosis. PCSK9 represents an example of successful translational research resulting in the identification of PCSK9 as a major drug target for a lipid-lowering therapy. To explore the genetic constitution of PCSK9 and its biologic role, in this review, we summarize the current evidence of clinically significant PCSK9 genetic variants involved in lipid metabolism as well as emphasize the importance of PCSK9 inhibition for the improvement of cardiovascular outcomes by conducting a meta-analysis of the available data on the incidence of cardiovascular disease events.
... Similarly, hyperglycemia and hyperlipidemia also contribute to cardiovascular and cerebrovascular diseases 34 . Similar to this study, Chaudhary along with others has shown that hyperlipidemia can lead to the progression of atherosclerosis 35 . It states that an injury to the endothelial wall of arteries can start a healing process which can lead to atherosclerosis. ...
Article
Full-text available
Background: Cardiovascular diseases have become a major burden in the whole world and the top of it is atherosclerosis causing deaths. It is characterized as the thickening of the arterial walls with the fatty material which includes cholesterol, cellular wastes, and fibrin. The atherosclerosis can be hereditary, due to aging, any autoimmune disease causing inflammation of endothelial or because of the sedentary lifestyle. Atherosclerosis is a consequence of dysfunction of endothelial done by low-density lipoprotein and changes in it by the internal wall of arteries. Smoking increases the oxidation stress and decreases the accessibility of nitric oxide to the endothelial wall which leads to vasodilatory response. These changes are irreversible. Pre-mature aging also contributes to the destruction of the epithelial wall of arteries eventually leading to hypertension, which if left untreated can cause atherosclerosis. Method: This is a cross-sectional study conducted for a period of one year and the sample size of 110 was taken of which 65.5% were male and the rest were females. This study includes all those patients` bodies who can go through autopsy and excludes patients whose bodies are mutilated or decomposed or those patients` bodies whose history is not properly available. Consents were taken from the relatives of the victims. The autopsy was done after the proper identification and examination of the patient`s body. The thoracic cavity was cut and opened. The aorta was cut from its origin to the diaphragm. The operational criterion was determined by grading from 1 to 5 based on the severity and thickness of aortic atherosclerosis. The data was statistically analyzed by applying the student`s t-test. The significant value of p<0.01 was considered. Result: Mostly males were affected. Out of 110 deceased more than 50% were from rural areas. 58% of the population have grade 1 and grade 2 atheromatous plaques. Most affected males were in the age of 50 years to 59 years and females were most affected in 60years to 69 years in their lifetime. This study also reveals that out of 110 dead bodies 64 patients died due to cardiac issues the rest died due to non-cardiac causes. The study also tells about the direct relation of atherosclerosis with hypertension, hyperlipidemia, hyperglycemia, and smoking habits with a significance level of p<0.001. Conclusion: The present study shows that it is more common in males than females and victims belonged to the age group of 5th to 8th decade. This reveals that aging is the dominating factor of residence. Rural people have it more than urban due to lack of awareness and diagnostic facilities Also a direct correlation between atherosclerosis and hypertension, hyperlipidemia, hyperglycemia, and smoking has been seen and confirmed through statistical significant.
... PCSK9 inhibitors, such as alirocumab and evolocumab, are a new generation of lipid-lowering drugs that have shown encouraging LDL cholesterol-lowering results in clinical trials [93] . PCSK9 plays a vital role in LDL receptor downregulation, and when the PCSK9 protein binds to the LDLR, it initiates the receptor's degradation process, thus increasing LDL cholesterol levels [94] (Fig. 2). Monoclonal antibodies inhibit PCSK9 binding to LDLRs, increase LDLR recycling, and indirectly lower circulating LDL cholesterol levels by increasing LDL cholesterol uptake [95] (Fig. 2). ...
Article
Full-text available
Background Cardiovascular diseases (CVD) persist as the leading cause of mortality globally, with atherosclerotic cardiovascular disease (ASCVD), including hypercholesterolemia, being a significant contributor. Hyperlipidemia management includes various lipid-lowering drugs, including statins, Bempedoic acid, inclisiran, Lomitapide, ANGPTL3 inhibitors, and PCSK9 inhibitors. Statins have traditionally dominated lipid management therapies; however, a subset of patients remains unresponsive or intolerant to this therapy, necessitating novel therapeutic approaches. Tafolecimab, a promising and novel PCSK9 monoclonal antibody, demonstrated significant LDL-C reduction and a favorable safety profile in clinical trials. Objective This review aimed to discuss the role and efficacy of Tafolecimab in the management of hypercholesterolemia. Methods We searched online databases, including PubMed, Scopus, and Embase, for articles related to talofecimab. Discussion The efficacy of Tafolecimab in diverse patient populations, including those with comorbid conditions and various lipid disorders, has been explored. Ongoing trials, such as CREDIT-1, CREDIT-2, and CREDIT-4, have provided valuable insights into Tafolecimab’s potential as a lipid-lowering agent. Moreover, the drug’s extended dosing interval may enhance patient compliance and reduce treatment costs. It has also been found that Tafolecimab has more affinity for PCSK9 and a longer duration of LDL-C reduction than other monoclonal antibody drugs such as evolocumab. Thus, this review focuses on Tafolecimab, a novel PCSK9 monoclonal antibody, its mechanism of action, clinical trial outcomes, safety profile, and potential role in hypercholesterolemia management. Despite its assuring potential, the long-term impact of Tafolecimab on cardiovascular outcomes remains to be fully elucidated, necessitating further research. Regulatory authorities like the FDA and EMA should also evaluate Tafolecimab’s risks and benefits. Conclusion In conclusion, Tafolecimab shows potential as an innovative therapeutic option for hypercholesterolemia, particularly in patients with specific risk factors, but warrants additional research.
Article
Heart disease remains a prominent global health concern, with cardiovascular disease (CVD) standing as a leading cause of death worldwide. Preventing heart disease not only decreases the risk of premature death but also mitigates complications like heart attacks, strokes, and arrhythmias, thereby enhancing overall health and quality of life. The economic burden of heart disease treatment highlights the importance of implementing preventive measures, such as lifestyle changes and early interventions, which can alleviate healthcare costs. These strategies, targeting risk factors like hypertension (HTN), diabetes mellitus (DM), dyslipidemia, and obesity, not only prevent heart disease but also reduce the risk of other health issues. Herein, this review covers various preventive measures, including dietary interventions, exercise, controlling HTN, DM, cholesterol, and weight, smoking cessation, and pharmacological interventions. By critically analyzing the guidelines and leveraging robust data alongside variations in recommendations, this review aims to elucidate effective primary prevention strategies for CVD.
Article
A variety of mechanisms and drugs have been shown to attenuate cardiovascular disease (CVD) onset and/or progression. Recent researchers have identified a potential role of proprotein convertase subtilisin/kexin type 9 (PCSK9) in modulating lipid metabolism and reducing plasma low density lipoprotein (LDL) levels. PCSK9 is the central protein in the metabolism of LDL cholesterol (LDL‐C) owing to its major function in LDL receptor (LDLR) degradation. Due to the close correlation of cardiovascular disease with lipid levels, many in vivo and in vitro investigations are currently underway studying the physiological role of PCSK9. Furthermore, many studies are actively investigating the mechanisms of various compounds that influence lipid associated‐disorders and their associated cardiovascular diseases. PCSK9 inhibitors have been shown to have significant impact in the prevention of emerging cardiovascular diseases. Natural products can effectively be used as PCSK9 inhibitors to control lipid levels through various mechanisms. In this review, we evaluate the role of phytochemicals and natural products in the regulation of PCSK9, and their ability to prevent cardiovascular diseases. Moreover, we describe their mechanisms of action, which have not to date been delineated.
Chapter
Atherosclerosis remains the leading cause of coronary heart disease (CHD) with enormous health and societal tolls. Traditional drug development approaches have been focused on small molecule–based compounds that aim to lower plasma lipids and reduce systemic inflammation, two primary causes of atherosclerosis. However, despite the widely available lipid-lowering and anti-inflammatory small compounds and biologic agents, CHD prevalence still remains high. Based on recent advances revealing disrupted immune homeostasis during atherosclerosis pathogenesis, novel strategies aimed at rejuvenating immune homeostasis with engineered immune leukocytes are being developed. This chapter aims to assess basic and translational efforts on these emerging strategies for the effective development of atherosclerosis treatment, as well as key challenges in this important translational field.
Article
Full-text available
Cardiovascular disease (CVD) is the leading cause of death worldwide, accounting for over one-third of all deaths in Singapore. An analysis of age-standardized mortality rates (ASMR) for CVD in Singapore revealed a deceleration in the initial rapid decline in ASMR. A decrease in smoking prevalence may have contributed to the initial rapid decline in ASMR. Furthermore, other major risk factors, such as diabetes mellitus, hypertension, elevated low-density lipoprotein levels, and obesity, are steadily rising. Singapore's CVD economic burden is estimated to be 8.1 billion USD (11.5 billion SGD). The burden of CVD can only be reduced using individual and population-based approaches. Prevention programs must also be developed based on an understanding of risk trends. Therefore, this article attempts to capture the burden of CVD, trends in risk factor control, preventive care, disparities, and current unmet needs, particularly in atherosclerotic cardiovascular disease management in Singapore.
Article
Low-density lipoprotein cholesterol (LDL-c) is both a therapeutic target and a risk factor for cardiovascular disease (CVD). MicroRNA (miRNA) have been shown to regulate cholesterol homeostasis, and miRNA in blood circulation have been linked to hypercholesterolemia. However, few studies to date have associated miRNA with phenotypes like LDL-c in a healthy population. To this end, we analyzed circulating miRNA in relation to LDL-c in a healthy cohort of 353 participants using two separate bioinformatic approaches. The first approach found that miR-15b-5p and miR-16-5p were upregulated in individuals with at-risk levels of LDL-c. The second approach identified two miRNA clusters, one that positively and a second that negatively, correlated with LDL-c. Included in the cluster that positively correlated with LDL-c were miR-15b-5p and miR-16-5p as well as other miRNA from the miR-15/107, miR-30, and let-7 families. Cross-species analyses suggested that several miRNA that associated with LDL-c are conserved between mice and humans. Finally, we examined the influence of diet on circulating miRNA. Our results robustly linked circulating miRNA with LDL-c suggesting that miRNA could be used as biomarkers for hypercholesterolemia or targets for developing cholesterol-lowering drugs.
Article
Full-text available
PCSK9 encodes proprotein convertase subtilisin/kexin type 9a (PCSK9), a member of the proteinase K subfamily of subtilases. Missense mutations in PCSK9 cause an autosomal dominant form of hypercholesterolemia in humans, likely due to a gain-of-function mechanism because overexpression of either WT or mutant PCSK9 reduces hepatic LDL receptor protein (LDLR) in mice. Here, we show that livers of knockout mice lacking PCSK9 manifest increased LDLR protein but not mRNA. Increased LDLR protein led to increased clearance of circulating lipoproteins and decreased plasma cholesterol levels (46 mg/dl in Pcsk9 –/– mice versus 96 mg/dl in WT mice). Statins, a class of drugs that inhibit cholesterol synthesis, increase expression of sterol regulatory element-binding protein-2 (SREBP-2), a transcription factor that activates both the Ldlr and Pcsk9 genes. Statin administration to Pcsk9 –/– mice produced an exaggerated increase in LDLRs in liver and enhanced LDL clearance from plasma. These data demonstrate that PCSK9 regulates the amount of LDLR protein in liver and suggest that inhibitors of PCSK9 may act synergistically with statins to enhance LDLRs and reduce plasma cholesterol. • low-density lipoprotein receptor • lipoproteins • proteinase • sterol regulatory element-binding protein
Article
Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a secreted protein that promotes degradation of cell surface LDL receptors (LDLRs). Decrease in circulating PCSK9 levels by monoclonal antibodies is correlated with improved lipid profiles in humans. In contrast to therapeutic antibodies, strategies to inhibit PCSK9 with a small molecule have proven to be challenging. Here we explored a potential strategy to decrease PCSK9 level by inhibiting its autocatalytic cleavage and subsequent secretion. We discovered a tool compound, which represents a highly reactive lactone series irreversible inhibitors, that can non-specifically bind to multiple sites on PCSK9 and prevent its autocatalytic cleavage and secretion. Mass spectrometry studies confirmed that serine residues 326 (S326) and 329 (S329) are the main binding sites. Structurally, S326 and S329 are far away from the PCSK9 autocatalytic cleavage site serine 386 (S386). Mutagenesis of S326/S329 reduced PCSK9 autocatalytic cleavage intracellularly. Through structural modeling, we identified additional residues serine 294 (S294) and asparagine 298 (N298) that interact with S326 and S329. Mutagenesis of S294 and N298 also reduced PCSK9 autocatalytic cleavage and secretion. In addition, by using the in-trans expression system of PCSK9, we determined that the effect of S326/S329 on PCSK9 secretion is not through their direct impact on PCSK9 autocatalytic cleavage. In conclusion, we have discovered a novel mechanism that regulates PCSK9 secretion, and we have determined the critical residues that are involved in this regulatory mechanism.
Article
Gain-of-function mutations in a serine protease, proprotein convertase subtilisin/kexin 9 (PCSK9), have been associated with high plasma levels of low-density lipoprotein (LDL) cholesterol and an increased incidence of coronary heart disease. Previous studies showed that PCSK9 loss-of-function mutations in patients were associated with low levels of LDL cholesterol and a reduced incidence of coronary heart disease. This suggested that pharmacologic inhibition of PCSK9 could lower LDL cholesterol levels in patients with hypercholesterolemia. REGN727/SAR236553 (REGN727) is an investigational, fully human monoclonal antibody highly specific for human PCSK9. This antibody blocks the interaction of PCSK9 with LDL receptors. This report presents the results of 3 phase 1 studies of REGN727 in humans. The participants were healthy volunteers and patients with familial or nonfamilial hypercholesterolemia. Three separate clinical studies of REGN727 were conducted. The first two compared the effect of single doses of REGN727, administered either intravenously (n = 40) or subcutaneously (n = 32), in healthy volunteers and a placebo. After these 2 single-dose studies, a randomized, placebo-controlled multiple-dose trial that investigated the effect of REGN727in 3 separate cohorts with hypercholesterolemia was conducted. The first cohort was composed of 21 subjects with heterozygous familial hypercholesterolemia and the second cohort consisted of 30 subjects with nonfamilial hypercholesterolemia. All patients in these 2 cohorts were receiving atorvastatin therapy and had a baseline LDL cholesterol level greater than 100 mg/dL (2.6 mM). The third cohort was composed of 10 subjects with nonfamilial hypercholesterolemia who had a baseline LDL cholesterol level greater than 130 mg/dL (3.4 mM); these patients were treated only with a modified diet. Patients in the multiple-dose study were randomly assigned to receive subcutaneous REGN727 (50, 100, or 150 mg) or placebo administered on days 1, 29, and 43. The primary study outcome was the incidence of adverse events. The major secondary end point evaluated was the effect of REGN727 on the lipid profile. None of the subjects receiving REGN727 withdrew from the study early because of an adverse event. There was a significant reduction in levels of LDL cholesterol among patients receiving REGN727 in all studies. Levels of LDL in the multiple-dose study were as follows: REGN727 doses of 50, 100, and 150 mg reduced LDL levels in the combined atorvastatin-treated populations to 77.5 mg/dL (2.00 mL), 61.3 mg/dL (1.59 mL), and 53.8 mg/dL (1.39 mL), respectively; differences in the change from baseline with the 50-, 100-, and 150-mg doses were −39.2, −53.7, and −61.0 percentage points compared with placebo (P < 0.001 for all 3 comparisons). These phase 1 trials suggest a role for PCSK9 in the regulation of LDL cholesterol in humans.
Article
Importance: Muscle-related statin intolerance is reported by 5% to 20% of patients. Objective: To identify patients with muscle symptoms confirmed by statin rechallenge and compare lipid-lowering efficacy for 2 nonstatin therapies, ezetimibe and evolocumab. Design, setting, and participants: Two-stage randomized clinical trial including 511 adult patients with uncontrolled low-density lipoprotein cholesterol (LDL-C) levels and history of intolerance to 2 or more statins enrolled in 2013 and 2014 globally. Phase A used a 24-week crossover procedure with atorvastatin or placebo to identify patients having symptoms only with atorvastatin but not placebo. In phase B, after a 2-week washout, patients were randomized to ezetimibe or evolocumab for 24 weeks. Interventions: Phase A: atorvastatin (20 mg) vs placebo. Phase B: randomization 2:1 to subcutaneous evolocumab (420 mg monthly) or oral ezetimibe (10 mg daily). Main outcome and measures: Coprimary end points were the mean percent change in LDL-C level from baseline to the mean of weeks 22 and 24 levels and from baseline to week 24 levels. Results: Of the 491 patients who entered phase A (mean age, 60.7 [SD, 10.2] years; 246 women [50.1%]; 170 with coronary heart disease [34.6%]; entry mean LDL-C level, 212.3 [SD, 67.9] mg/dL), muscle symptoms occurred in 209 of 491 (42.6%) while taking atorvastatin but not while taking placebo. Of these, 199 entered phase B, along with 19 who proceeded directly to phase B for elevated creatine kinase (N = 218, with 73 randomized to ezetimibe and 145 to evolocumab; entry mean LDL-C level, 219.9 [SD, 72] mg/dL). For the mean of weeks 22 and 24, LDL-C level with ezetimibe was 183.0 mg/dL; mean percent LDL-C change, -16.7% (95% CI, -20.5% to -12.9%), absolute change, -31.0 mg/dL and with evolocumab was 103.6 mg/dL; mean percent change, -54.5% (95% CI, -57.2% to -51.8%); absolute change, -106.8 mg/dL (P < .001). LDL-C level at week 24 with ezetimibe was 181.5 mg/dL; mean percent change, -16.7% (95% CI, -20.8% to -12.5%); absolute change, -31.2 mg/dL and with evolocumab was 104.1 mg/dL; mean percent change, -52.8% (95% CI, -55.8% to -49.8%); absolute change, -102.9 mg/dL (P < .001). For the mean of weeks 22 and 24, between-group difference in LDL-C was -37.8%; absolute difference, -75.8 mg/dL. For week 24, between-group difference in LDL-C was -36.1%; absolute difference, -71.7 mg/dL. Muscle symptoms were reported in 28.8% of ezetimibe-treated patients and 20.7% of evolocumab-treated patients (log-rank P = .17). Active study drug was stopped for muscle symptoms in 5 of 73 ezetimibe-treated patients (6.8%) and 1 of 145 evolocumab-treated patients (0.7%). Conclusions and relevance: Among patients with statin intolerance related to muscle-related adverse effects, the use of evolocumab compared with ezetimibe resulted in a significantly greater reduction in LDL-C levels after 24 weeks. Further studies are needed to assess long-term efficacy and safety. Trial registration: clinicaltrials.gov Identifier: NCT01984424.
Article
Background Heterozygous familial hypercholesterolaemia is characterised by low cellular uptake of LDL cholesterol, increased plasma LDL cholesterol concentrations, and premature cardiovascular disease. Despite intensive statin therapy, with or without ezetimibe, many patients are unable to achieve recommended target levels of LDL cholesterol. We investigated the eff ect of PCSK9 inhibition with evolocumab (AMG 145) on LDL cholesterol in patients with this disorder.