ArticlePDF AvailableLiterature Review

Lipoprotein(a)-The Crossroads of Atherosclerosis, Atherothrombosis and Inflammation

Authors:

Abstract

Increased lipoprotein(a) (Lp(a)) levels are an independent predictor of coronary artery disease (CAD), degenerative aortic stenosis (DAS), and heart failure independent of CAD and DAS. Lp(a) levels are genetically determinated in an autosomal dominant mode, with great intra-and inter-ethnic diversity. Most variations in Lp(a) levels arise from genetic variations of the gene that encodes the apolipoprotein(a) component of Lp(a), the LPA gene. LPA is located on the long arm of chromosome 6, within region 6q2.6-2.7. Lp(a) levels increase cardiovascular risk through several unrelated mechanisms. Lp(a) quantitatively carries all of the atherogenic risk of low-density lipo-protein cholesterol, although it is even more prone to oxidation and penetration through endothelia to promote the production of foam cells. The thrombogenic properties of Lp(a) result from the ho-mology between apolipoprotein(a) and plasminogen, which compete for the same binding sites on endothelial cells to inhibit fibrinolysis and promote intravascular thrombosis. LPA has up to 70% homology with the human plasminogen gene. Oxidized phospholipids promote differentiation of pro-inflammatory macrophages that secrete pro-inflammatory cytokines (e. g., interleukin (IL)-1β, IL-6, IL-8, tumor necrosis factor-α). The aim of this review is to define which of these mechanisms of Lp(a) is predominant in different groups of patients.
Biomolecules 2022, 12, 26. https://doi.org/10.3390/biom12010026 www.mdpi.com/journal/biomolecules
Review
Lipoprotein(a)—The Crossroads of Atherosclerosis,
Atherothrombosis and Inflammation
Sabina Ugovšek 1 and Miran Šebeštjen 1,2,3,*
1 Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia; ugovsek.sabina@gmail.com
2 Department of Cardiology, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
3 Department of Vascular Diseases, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
* Correspondence: miran.sebestjen@guest.arnes.si
Abstract: Increased lipoprotein(a) (Lp(a)) levels are an independent predictor of coronary artery
disease (CAD), degenerative aortic stenosis (DAS), and heart failure independent of CAD and DAS.
Lp(a) levels are genetically determinated in an autosomal dominant mode, with great intra- and
inter-ethnic diversity. Most variations in Lp(a) levels arise from genetic variations of the gene that
encodes the apolipoprotein(a) component of Lp(a), the LPA gene. LPA is located on the long arm of
chromosome 6, within region 6q2.62.7. Lp(a) levels increase cardiovascular risk through several
unrelated mechanisms. Lp(a) quantitatively carries all of the atherogenic risk of low-density lipo-
protein cholesterol, although it is even more prone to oxidation and penetration through endothelia
to promote the production of foam cells. The thrombogenic properties of Lp(a) result from the ho-
mology between apolipoprotein(a) and plasminogen, which compete for the same binding sites on
endothelial cells to inhibit fibrinolysis and promote intravascular thrombosis. LPA has up to 70%
homology with the human plasminogen gene. Oxidized phospholipids promote differentiation of
pro-inflammatory macrophages that secrete pro-inflammatory cytokines (e. g., interleukin (IL)-1β,
IL-6, IL-8, tumor necrosis factor-α). The aim of this review is to define which of these mechanisms
of Lp(a) is predominant in different groups of patients.
Keywords: lipoprotein(a); atherothrombosis; inflammation; coagulation; fibrinolysis
1. Introduction
Lipoprotein(a) (Lp(a)) is a complex plasma protein that consist of low-density lipo-
protein (LDL) cholesterol and apolipoprotein B-100 (apoB) linked to the plasminogen-like
apolipoprotein(a) (apo(a)) via a disulfide bond. Lp(a) levels are genetically determined by
the LPA gene and they vary among individuals from ≤0.2 to ≥250 mg/dL, although within
a single individual, Lp(a) levels are stable throughout life [1,2]. The LPA gene is located
on chromosome 6q26q27 and it encodes the highly glycosylated hydrophilic apo(a) [3,4].
Apo(a) shows high amino-acid sequence homology to serine protease plasminogen [3].
Apo(a) contains a protease-like domain and two tri-loop structures known as ‘kringles’
(KIV, KV) [1,3], and the KIV domain has 10 types [3]. The different numbers of KIV type
2 (KIV2) repeat in apo(a) results in Lp(a) isoform size heterogeneity, and more KIV2 re-
peats results in a larger apo(a) isoform. The larger apo(a) isoforms are less efficiently se-
creted from hepatocytes, and consequently there is an inverse correlation between the
apo(a) isoform size and the Lp(a) levels in plasma [1,5]. Patients with a smaller apo(a)
isoform size not only have higher Lp(a) levels, but also have significantly greater risk of
coronary artery disease (CAD) [6]. The number of KIV2 repeats accounts for 69% of the
variation in Lp(a) levels [7]. As well as the different number of KIV2 repeats, the Lp(a)
isoform size and levels are determined by more than 200 single nucleotide polymor-
phisms (SNPs) in the wider LPA region. Among these SNPs, rs10455872 and rs3798220
have the most influence on Lp(a) levels [4], as they explain 36% of the variation in Lp(a)
Citation:
Ugovšek, S.; Šebeštjen, M.
Lipoprotein(a)
The Crossroads of
Atherosclerosis, Atherothrombosis
and Inflammation.
Biomolecules 2022,
12
, 26. https://doi.org/10.3390/
biom12010026
Academic Editor
s: Roberto Scicali,
Alessandro Mattina
,
Giuseppe Mandraffino
Received:
25 November 2021
Accepted:
23 December 2021
Published:
24 December 2021
Publisher’s Note:
MDPI stays neu-
tral with regard to jurisdictional
claims in published maps and institu-
tional affiliations.
C
opyright: © 2021 by the authors. Li-
censee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and con-
ditions of the Creative Commons At-
tribution (CC BY) license (http
s://cre-
ativecommons.org/licenses/by/4.0/).
Biomolecules 2022, 12, 26 2 of 14
levels [8]. LPA variants are carried by one in six people, who have a 1.5-fold greater risk
of coronary diseases [8]. rs10455872 and rs3798220 polymorphisms are also independent
predictors of cardiovascular events in patients with CAD [8].
Increased Lp(a) levels are also a risk factor for degenerative aortic valve stenosis [9], [10].
Both homozygote and heterozygote carriers of rs10455872 have increased risk of degenerative
aortic valve stenosis, although this risk is not seen for carriers of rs3798220. A trend towards
increased risk has been connected with lower numbers of KIV2 repeats [10].
Elevated levels of Lp(a) can occur in patients with otherwise normal lipid levels [4].
Dietary and environmental factors have minimal contributions to Lp(a) plasma levels [1].
A quarter of the general population has Lp(a) plasma levels >20 mg/dL, which have been
associated with a 2-fold increased risk of developing cardiovascular disease. Although
there is no conclusive evidence for specific Lp(a) cut-off points based on age, sex, and race,
the European guidelines consider those with ≥50 mg/dL Lp(a) to be at high risk [11]. Lp(a)
levels >180 mg/dL are correlated with risk of cardiovascular events similar to that of fa-
milial hypercholesterolemia [1].
According to genetic and epidemiological studies, Lp(a) is considered pro-athero-
sclerotic (Figure 1), pro-inflammatory (Figure 2), pro-thrombotic, and anti-fibrinolytic
(Figure 3) [12].
Figure 1. The role of lipoprotein(a) (Lp(a)) in plaque formation. Ox Lp(a), oxidized Lp(a).
Figure 2. The role of Lp(a) in inflammation. OxPLs, oxidized phospholipids; TNFα, tumor necrosis
factor-α; IL, interleukin; ICAM1, intercellular adhesion molecule-1; VCAM1, vascular cell adhesion
protein-1; NO, nitric oxide.
Biomolecules 2022, 12, 26 3 of 14
Figure 3. The role of Lp(a) in thrombus formation following plaque rupture. TF, tissue factor; TFPI,
tissue factor pathway inhibitor; PAI-1, plasminogen activator inhibitor-1; tPA, tissue plasminogen
activator.
2. Lipoprotein(a) and Atherosclerosis
Lipoprotein(a) can be internalized and accumulated in the intima of arteries and the
aortic valve leaflets. Lp(a) enters the intima at similar rates to LDL cholesterol, although
this does not occur via the lipoprotein receptor for LDL cholesterol, but is dependent on
Lp(a) plasma concentrations, Lp(a) particle size, blood pressure, and arterial wall perme-
ability [13,14]. Lp(a) accumulates all over the intima, whereas LDL cholesterol and other
apoB-containing lipoproteins remain mainly at atherosclerotic lesions. Compared to
LDLs, Lp(a) has a greater affinity for the vascular wall and for proteoglycans and fibron-
ectin on the endothelial cell surface [13]. Furthermore, Lp(a) is more atherogenic than LDL
cholesterol, because it consists of all of the atherogenic components of both LDL choles-
terol and apo(a) [4,15,16].
Various studies have indicated that Lp(a) is taken up by macrophages to produce
foam cells, and thus to promote the development of atherosclerotic lesions [13]. Athero-
sclerosis is an inflammatory disease of the arterial wall, whereby several events result in
the formation of a complex atherosclerotic plaque that is composed of a lipid-rich core
covered with a fibrous cap (Figure 1) [17]. Lp(a) promotes atherosclerotic plaque for-
mation through various mechanisms. Lp(a) induces expression of inflammatory cyto-
kines, including interleukin (IL)-1β, IL-6, IL-8, and tumor necrosis factor-α, and increases
expression of adhesion molecules on the surface of the endothelial cells, including vascu-
lar cell adhesion protein-1, intercellular adhesion molecule-1, E-selectin, P-selectin, and
others [3,18,19]. Lp(a) promotes monocyte chemotaxis via stimulation of secretion of mon-
ocyte chemotactic protein and activation of nuclear factor κB [20]. Additionally, Lp(a)
binds and transports more than 70% of circulating oxidized phospholipids, which are in-
volved in plaque vulnerability and destabilization [21].
As indicated, atherosclerotic plaques in patients with elevated Lp(a) levels have a
complex morphology [17]. These plaques are prone to repeated ruptures and healing,
which can lead to severe and rapid progression of atherosclerosis. Coronary atherosclero-
sis in patients with high Lp(a) levels mostly manifests clinically as an acute myocardial
infarction, rather than stable angina [4]. Several large observational studies and meta-
analyses have also reported gradual increased risk of CAD and ischemic stroke with in-
creasing Lp(a) levels, without any threshold value defined [2225]. This risk is higher in
younger populations than older populations [25]. Indeed, the addition of Lp(a) levels to
traditional risk score charts improve the accuracy of the prediction of susceptibility for
future cardiovascular events. This has been shown by several studies of primary and sec-
ondary prevention [26–28], except for primary prevention in women with low risk of car-
diovasculer events [29].
Biomolecules 2022, 12, 26 4 of 14
Statins are well known to reduce LDL cholesterol levels, but as seen in a meta-analy-
sis of 29,069 patients on statin therapy, they do not provide any significant changes in
Lp(a) levels [30]. Moreover, Lp(a) levels in patients treated with statins are more strongly
associated with cardiovascular disease risk than for those on a placebo. The main expla-
nation for this was that when LDL cholesterol levels are reduced, Lp(a)-attributable risk
becomes an even stronger predictor of residual risk [30]. This observation has been sup-
ported by two interventional studies: ‘Further Cardiovascular Outcomes Research with
PCSK9 Inhibition in Subjects with Elevated Risk’ (FOURIER) and ‘Evaluation of Cardio-
vascular Outcomes After an Acute Coronary Syndrome During Treatment with Aliro-
cumab’ (ODYSSEY Outcomes) [31,32]. Patients on LDL cholesterol-lowering therapies
and with high Lp(a) levels were associated with increased risk for cardiovascular disease
independent of LDL cholesterol levels. Evolocumab therapy in the first of these two trials,
and alirocumab therapy in the second, reduced Lp(a) levels and cardiovascular disease
risk independent of LDL cholesterol lowering [31,32].
Patients with elevated Lp(a) have significantly more recurrent acute myocardial in-
farctions. One of the reasons for this is that Lp(a) specifically accumulates at injured sites,
which results in restenosis after coronary artery revascularisation procedures [4]. A meta-
analysis of 1834 patients reported a positive correlation between high Lp(a) levels and in-
stent restenosis, especially in patients with drug-eluting stents [33]. Lowering Lp(a) levels
with lipoprotein apheresis reduces restenosis rates and helps to prevent recurrent coro-
nary syndromes [34,35].
As well as being a risk factor for coronary atherosclerosis, elevated Lp(a) levels define
the risk of ischemic stroke, peripheral arterial disease, and calcific aortic valve disease.
Calcific aortic valve disease is a multifactor condition and a dynamic process similar to
atherosclerosis. Endothelial cell dysfunction followed by subendothelial accumulation of
lipoproteins and chronic inflammation results in a condition that can vary from mild valve
thickening to severe calcification with stenotic occlusion [36]. Studies have shown that
Lp(a) levels 30 mg/dL represent a risk factor for calcific aortic valve disease. Lp(a) con-
tributes to this disease at different rates, which depend on ethnicity [9], with the highest
levels of Lp(a) in calcific aortic valve disease seen for Caucasians [9,37,38]. There is also
an association between Lp(a) gene polymorphisms and calcific aortic valve disease [36,38
40]. Thanassoulis et al. and Ozkan et al. showed strong positive correlations for rs10455872
and rs3798220 polymorphisms with the progression of calcific aortic valve disease [36,40].
Patients with heterozygous familial hyperholesterolemia have significantly higher
Lp(a) levels and hazard ratio for acute myocardial infarction, compared to those without,
as was shown in the prospective population study, ‘Copenhagen General Population
Study,’ and in a cross-sectional analysis of ‘Spanish Familial Hypercholesterolemia Co-
hort Study’ (SAFEHEART) [41,42]. Furthermore, patients with Lp(a) levels ≥50 mg/dL and
LDL-receptor-negative mutations have a higher risk of cardiovascular disease, compared
to patients with less severe mutations [42].
3. Lipoprotein(a) and Inflammation
Inflammation has a vital role in the development and progression of atherosclerosis,
and thus it contributes to increased risk of cardiovascular events [43]. Inflammation pro-
motes endothelial cell activation, dysfunction and loss of endothelial integrity, failure of
endothelial repair, intima lipid deposition, and plaque formation and instability [44].
Despite the evidence that Lp(a) levels are genetically determined, several studies have
shown that chronic inflammation interferes with Lp(a) expression and increases Lp(a) plasma
levels [21]. On the other hand, as for other lipoproteins, Lp(a) is susceptible to oxidative mod-
ifications and formation of pro-inflammatory and pro-atherogenic oxidized phospholipids
(OxPLs) [3]. Lp(a) carries more than 80% OxPLs in its particles, and consequently this in-
creases the inflammatory activity of the arterial wall [21]. Taken together, the evidence shows
a bidirectional link between Lp(a) and inflammation (Figure 2) [44].
Biomolecules 2022, 12, 26 5 of 14
The Lp(a) binding of OxPLs is to lysine residues on isolated fragments of KV of apo(a)
[45]. At low plasma concentrations, Lp(a) has anti-inflammatory effects via its scavenging
of OxPLs and their degradation by Lp(a)-associated phospholipase A2 (Lp-PLA2) [21]. At
high Lp(a) levels, its anti-atherogenic effects are diminished, with decreased Lp-PLA2 low-
ering of oxidative stress, compared to LDL-associated PLA2. The OxPL moieties on apo(a)
compete for the active site and decrease the catalytic efficiency of Lp-PLA2 [46]. Here, the
oxidationreduction state tends towards oxidation, and Lp(a) releases its oxidative load
into the atheromatous plaque. Consequently, OxPLs induce inflammatory responses by
increasing secretion of inflammatory cytokines and chemokines, and through mediation
of monocyte activation [21]. Removal of apo(a) from Lp(a) particles leads to increased Lp-
PLA2 activity, and thus degradation of OxPLs [46].
Lipoprotein(a) also carries monocyte chemoattractant protein-1 (MCP-1), and it has
been shown that OxPLs are major determinants of MCP-1 binding. It was suggested that
Lp(a)-associated MCP-1 enhances recruitment of monocytes to the vascular wall [47].
Monocyte activation has a pivotal role in atheroma plaque formation, while apo(a) en-
hances the inflammatory and proteolytic potential of monocytes, which release reactive
oxygen species and matrix metalloproteinase-9 (MMP-9). Reactive oxygen species are in-
volved in LDL cholesterol oxidation and formation of foam cells, and MMP-9 contributes
to extracellular matrix degradation and rupture of atherosclerotic plaques. The degree of
stimulation with apo(a) is inversly correlated with the number of KIV2 repeats, which sup-
ports the hypothesis regarding the detrimental effects of small-sized isoforms of apo(a) in
atherosclerosis progression [48].
According to Arai et al. [49], OxPLs are mainly bound to small Lp(a) isoforms. Car-
riers of the rs3798220 variation have significantly higher Lp(a) levels and smaller apo(a)
isoforms, compared to noncarriers. Patients with this variation are associated with greater
levels of proinflammatory OxPLs on apoB particles, and consequently, they might have
increased atherogenic potential [49].
A secondary post-hoc analysis of a double-blind, randomized clinical trial (‘Asses-
ment of Clinical Effects of Cholesteryl Ester Transfer Protein Inhibition With Evacetrapib
in Patients at a High Risk for Vascular Outcomes’; ACCELERATE) showed that increased
Lp(a) levels are related to cardiovascular events when high sensitivity C-reactive protein
(CRP) concentrations are ≥2 mg/L, but not when they are <2 mg/L [50]. The inflammatory
marker CRP directly affects endothelial cells, monocytes, macrophages, and smooth mus-
cle cells. It is produced by hepatocytes in response to various inflammatory cytokines.
Among these, IL-6 is most closely associated with inflammation and increased risk of ath-
erosclerosis. As shown by Shufta et al., there is a positive correlation between CRP, IL-6,
and Lp(a) levels [51]. Furthermore, the IL-6 receptor blocker tocilizumab reduced inflam-
matory disease activity and plasma Lp(a) levels in 132 patients with reumatoid arthritis
[52]. Also, in a study of 280 hemodialysis patients and in two studies with 137 and 114
patients with rheumatoid arthritis, CRP levels increased with increasing levels of Lp(a)
[5355]. However, based on the ‘Copenhagen General Population Study’, Langsted et al.
reported that elevated Lp(a) levels are not causally associated with increased low-grade
inflammation, as measured through CRP, despite a casual association with increased risk
of aortic valve stenosis and acute myocardial infarction [56].
4. Lipoprotein(a) and Atherothrombosis
Coagulation and fibrinolysis have essential roles in atherothrombosis [57]. The rup-
ture of an atherosclerotic plaque results in the activation of platelets and the extrinsic co-
agulation pathway. Lp(a) participates in atherothrombosis through several mechanisms
(Figure 3). As an atherogenic lipoprotein, Lp(a) interferes with platelet aggregation [58],
as it can bind to platelet-activating factor (PAF) acetylhydrolase, which degrades and in-
activates PAF. This mechanism results in reduced platelet aggregation and activation [59].
Lipoprotein(a) has an effect on the coagulation pathway through the promotion of
the expression of tissue factor (TF) [60]. TF is a cell-surface glycoprotein that is
Biomolecules 2022, 12, 26 6 of 14
overexpressed by macrophages and smooth muscle cells within the atherosclerotic
plaque. Tissue damage that results in endothelial denudation exposes TF to the flow of
blood, whereby TF initiates activation of the extrinsic coagulation pathway, which leads
to thrombus formation and intima fibrin deposition [57]. Another prothrombotic effect of
Lp(a) is related to its binding to and inhibition of the activity of the TF pathway inhibitor
(TFPI) [61]. Bilgen et al. and Nisio et al. showed positive correlations between plasma TFPI
and Lp(a) levels [62,63]. Although patients with higher Lp(a) levels had higher TFPI con-
centrations, the TFPI activity was not different between those with high and low Lp(a)
levels. This suggests that the binding of TFPI to Lp(a) partly inhibits TFPI activity [62].
Lipoprotein(a) is believed to promote atherothrombosis due to its homology with
plasminogen [5]. As indicated, one of the component proteins of Lp(a), apo(a), shares
more than 80% of its protein sequence with plasminogen [64]. Lp(a) lacks KI to KIII of
plasminogen, but it contains KIV and KV [60]. Apo(a) KIV10 contains a lysin binding site,
which is most similar to the lysine binding site within plasminogen KIV [65]. Due to this
structural homology, Lp(a) can bind to plasminogen receptors on the surface of platelets
and prevent the interaction between plasminogen and tissue plasminogen activator (tPA).
Therefore, tPA cannot convert plasminogen to plasmin [60].
Plasminogen is the proenzyme precursor of plasmin, and thus it has an important
role in fibrinolysis. Plasminogen is activated to plasmin by tPA or urokinase [66]. The
plasminogen and LPA genes are both on chromosome 6, and a recent study identified nine
SNPs within the plasminogen and LPA gene region that are significantly associated with
plasminogen levels [67]. Among these various SNPs, Wang et al. studied the impact of
rs3798220 and rs10455872 on Lp(a) and plasminogen levels. Although these polymor-
phisms are associated with higher Lp(a) levels, they had no influence on plasminogen
levels and fibrinolytic activity [68]. However, in an in vitro study, Rowland et al. showed
that Caucasian patients carrying the rs3798220 SNP have increased clot lysis times and
reduced clot permeability, compared to noncarriers. On the other hand, among non-Cau-
casians, increased clot permeability and decreased lysis time have been reported [69]. Fur-
thermore, Scipione et al. showed that the isoleucine-to-methionine substitution in apo(a)
protease-like domain of rs3798220 decreased coagulation times and increased fibrin clot
lysis times, compared to wild-type apo(a). Additionally, the fibrin fiber width in plasma
clots was increased, and patients with higher Lp(a) levels had lower clot permeability [70].
Taken together, rs3798220 has antifibrinolytic potential and is associated with increased
Lp(a) levels and cardiovascular risk.
Lipoprotein(a) inhibits the production of tPA, and therefore it promotes a thrombotic
state through the prevention of plasmin-mediated clot lysis [60]. In addition, Lp(a) stim-
ulates the expression and activity of the primary inhibitor of the fibrinolytic system, plas-
minogen activator inhibitor-1 (PAI-1) [60,71]. We have previously shown that patients
with familial hyperholesterolemia who had survived myocardial infarction at a young age
had significanly decreased fibrinolytic activity, as shown by increased PAI-1 antigen and
activity, and increased t-PA antigen, compared to those without myocardial infarction
[72]. Furthermore, these patients had higher Lp(a) levels; however, this difference did not
reach significance, probably due to the small number of patients. Etingin et al. performed
an in vitro study in which they treated human umbilical vein endothelial cells with Lp(a)
and LDL. They showed that Lp(a) levels up to 40 µg/mL increased PAI-1 activity, while
treatment with LDL cholesterol using approximately equimolar concentrations did not
stimulate PAI-1 activity. Lp(a) levels above 40 µg/mL did not show any further increase
in PAI-1 activity. This effect was due to increased mRNA levels of PAI-1 [73]. Shindo et
al. compared coronary atherotomy specimens of patients with acute myocardial infarction
and unstable angina pectoris with those from patients with stable angina pectoris, and
showed that the levels of both Lp(a) and PAI-1 were significantly higher for the former
[74]. PAI-1 activity, but not antigen, was shown to be a predictive factor for future coro-
nary events in patients with or without prior CAD [75]. Some studies have also included
Lp(a) and apoB in the analysis of fibrinolytic parameters, and in all of these, the
Biomolecules 2022, 12, 26 7 of 14
fibrinolytic parameters were correlated with Lp(a) or apoB, which is a component of Lp(a)
(Table 1) [7678].
Fibrin represents the structural framework for fibrin clots. The structure, stability,
and fibrinolytic potential of fibrin clots depend on differences in the fibrin fiber diameter
and pore size within the fibrin network, and on negatively charged substances. Dense
clots are associated with a reduced fibrin fiber diameter, smaller pores, and increased fi-
brin stiffness [79]. The rate of fibrin clot lysis is predominantly a function of pore size.
Fibrin clots with smaller pores increase clot lysis times and are correlated with greater
cardiovascular risk. Elevated Lp(a) levels are correlated with reduced fibrin clot permea-
bility and impaired fibrinolysis [61]. Lp(a) competes with plasminogen and tPA for bind-
ing sites on fibrin, and consequently, it impairs fibrinolysis [1,60,64]. These antifibrinolytic
effects are more profound for smaller-sized isoforms of Lp(a), because these have a higher
affinity for their binding to fibrin [80]. The predictive value of high Lp(a) levels as a risk
factor therefore depends on the concentration of Lp(a) particles containing small isoforms
of Lp(a) [81]. Other reports have opposed the mechanism whereby Lp(a) directly com-
petes with plasminogen for binding to fibrin, and have indicated that apo(a) forms a qua-
ternary complex with plasminogen, tPA, and fibrin [82,83]. In addition, the negatively
charged apo(a) might affect the Lp(a)-induced alteration of fibrin clot structure, and thus
contribute to reduced fibrin degradation [61].
5. Prognostic Value of Lipoprotein(a) in Combination with Thrombotic and Inflam-
mation Parameters
The prospective studies that have included analysis of Lp(a) and at least one marker
of inflammation and/or coagulation/fibrinolysis as predictors for future coronary events
are presented in Table 1. In primary prevention studies, which have included >40,000 pa-
tients (mostly male), Lp(a) was shown to independently predict the first cardiovascular
event [84–86]. In the first study, by Langsted et al. [84], only Lp(a) and CRP were examined
as markers of inflammation. In the next two studies, by Cremer et al. [85] and Seed et al.
[86], only Lp(a) and fibrinogen were examined as markers of impairment of the coagula-
tion fibrinolytic system. Indeed, none of the studies indicated in Table 1 have examined
Lp(a) plus at least one parameter of inflammation and one of coagulation/fibrinolysis.
For patients with stable coronary disease, their D-dimer and fibrinogen levels
increased with the quartiles of CRP, while no such relation was seen for Lp(a) [87]. In a
survey of 1172 apparently healthy men, CRP levels correlated with several fibrinolytic
parameters (i.e., fibrinogen, t-PA antigen, D-dimer, homocysteine), and also with Lp(a)
[88]. Here, we can conclude that fibrinolytic and inflammatory parameters are
interconnected in patients with stable CAD and in apparently healthy men.
As the rupture of a pre-existing atherosclerotic plaque is responsible for the majority
of acute coronary events, the number of such events in secondary prevention studies is
much higher. In these studies, where no coronary interventions were performed due to
stenosis < 50%, Lp(a) alone, and even more so in combination with fibrinogen, was an
independent predictor of major adverse cardiovascular events after 37 months [89]. This
was expected, because in these patients, both progression of atherosclerosis and rupture
of plaques can lead to future cardiovascular events. In patients with only percutaneous
transluminal coronary angioplasty and no stent implantation due to CAD, t-PA was
shown to be a superior predictor of major adverse cardiovascular events after 13 years,
compared to Lp(a) [90]. This is not surprising, as the endothelium in percutaneous trans-
luminal coronary angioplasty with no stent implantation is highly disrupted, and future
coronary events are mostly due to intracoronary thrombosis. Similarly, in patients six
months after percutaneous transluminal coronary angioplasty, neither Lp(a) nor PAI-1
were predictors of restenosis [91]. The reason here might be the short time of observation.
In studies where patients were mostly treated with drug-eluting stents, Lp(a) and CRP
were independent predictors of future cardiovascular events [92,93]. At first sight, this
might be a surprise, but it can be noted that all these patients had dual antiplatelet therapy
Biomolecules 2022, 12, 26 8 of 14
that prevented early in-stent thrombosis, for which coagulation and fibrinolysis are re-
sponsible. Furthermore, sirolimus- and everolimus-eluting stents are less prone to early
in-stent thrombosis. In contrast, for patients treated with bare metal stents, homocysteine
levels were linked to the fibrinolytic parameters, especially with t-PA and PAI-1 [94,95].
These patients were also treated with dual antiplatelet therapy, although bare metal stents
were much more thrombogenic than drug-eluting stents [96]. On the other hand, there
were no effects on cardiovascular mortality [97]. Finally, plasma levels of t-PA and Lp(a)
were independently associated with the subsequent development of a first myocardial
infarction in a prospective case-control study [98].
Table 1. Overview of prospective studies that have examined Lp(a) and at least one inflammatory
and/or fibrinolytic parameter.
Study
Parameter In-
cluded
Study Population (n)
Primary Endpoint
Independent Pre-
dictor
Ref.
Age (Years)
Characteristics
Langsted et
al. (2014)
Lp(a), CRP
-
General population
Ischemic heart dis-
ease
Lp(a), independent
of CRP levels
[84]
Cremer et
al. (1997)
Lp(a), fibrinogen
40
60
Male, without previ-
ous CAD
MI
after 10 years
Lp(a)
[85]
Seed et al.
(2001)
Lp(a), fibrinogen
51
61
Male, without previ-
ous CAD
CAD after 6 years
Lp(a)
[86]
Zhang et al.
(2020)
Lp(a), fibrinogen
-
Stable CAD, no stent
implantation
MACE after 37
months
Lp(a), fibrinogen;
combination of
both superior
[89]
Niessner et
al. (2003)
Lp(a), tPA
-
CAD after PTCA, no
stent implantation
MACE after 13
years
t
-PA [90]
Alaigh et al.
(1998)
Lp(a), PAI
-1
-
CAD after PTCA, no
stent implantation
Restenosis after 6
months
None
[91]
Kardys et al.
(2012)
Lp(a), CRP, IL
-10
-
CAD after sirolimus-
eluting stent implan-
tation
MACE after 1, 6
years
1 year: Lp(a); 6
years: CRP
[92]
Zairis et al.
(2002)
Lp(a), CRP
-
ACS after PCI, vari-
ous stent implanta-
tions
MACE after 3 years
Lp(a), CRP
[93]
Marcucci et
al. (2006)
Lp(a) PAI-1, ho-
mocysteine
-
ACS, bar metal stent
implantation
MACE after 24
months
PAI-1, homocyste-
ine
[95]
Thogersen
et al. (2003)
Lp(a), PAI
-1, t-
PA, leptin
Sex and age
matched
-
First myocardial in-
farction
Lp(a), t
-PA [98]
Pineda et al.
(2010)
Lp(a), CRP, PAI-
1, t
-PA, fibrino-
gen, D
-dimer,
homocysteine
-
MI before 45 years;
treated with fibrinoly-
sis; PCI
MACE after 36
months
Homocysteine
[99]
Moss et al.
(1999)
Lp(a), PAI-1, fi-
brinogen, D
-di-
mer
-
Post-MI (2 months),
treated with thrombo-
lysis or PCI
Coronary death or
nonfatal MI after
26 months
D
-dimer [76]
ACS, acute coronary syndrome; CAD, coronary artery disease; CRP, C-reactive protein; MACE, ma-
jor adverse coronary event; MI, myocardial infarction; PCI, percutaneous coronary intervention;
PTCA, percutaneous transluminal coronary angioplasty.
Biomolecules 2022, 12, 26 9 of 14
6. Conclusions
Lipoprotein(a) has been shown to be an independent predictor for future cardiovas-
cular events in primary and secondary prevention. This has been confirmed in epidemio-
logical and Mendelian randomized trials, and in the last two years, also in randomized
placebo-controlled double-blind trials with PCSK9 inhibitors. Although these drugs were
primarly designed to target LDL cholesterol, they also decrease Lp(a) by 20% to 40%
[100,101]. Similar findings were observed with inclisiran, a small interfering RNA
(siRNA). Inclisiran was developed to target LDL and apoB, but in the phase 2 study enti-
tled Trial to Evaluate the Effect of ALNPCSSC Treatment on Low Density Lipoprotein
Cholesterol (ORION-1), it also reduced Lp(a) levels by 25.6% [102,103]. Oplasiran, another
siRNA targeting Lp(a), significantly reduced Lp(a) levels in the phase 1 trial [104]. More-
over, these effects persisted for more than six months [104]. Furthermore, promising
Lp(a)-lowering effects have been observed with antisense oligonucleotide AKCEA
APO(a)LRX. This drug has shown a dose-dependent reduction in Lp(a) levels up to 80%
among patients with established cardiovascular disease and Lp(a) levels above 60 mg/dL
in a phase 2 trial [105]. Whether viral-mediated gene therapy RGX501 for homozygous
familial hypercholesterolemia also has influence on reducing Lp(a) levels is currently be-
ing investigated in clinical trial NCT02651675 [106]. Although all the aforementioned ther-
apeutics have shown lipid-related effects, only PCSK9 inhibitors were found to be associ-
ated with decreased cardiovascular events [31,32]. As for their effects on inflammation
and coagulation, the current evidence is scarce.
The atherogenic propensity of Lp(a) arises as a consequence of its structure. Its LDL-
like particle, which is the main part of Lp(a), has similar effects to those of LDL cholesterol,
and it is even more atherogenic because of its higher sensitivity to oxidation. On the other
hand, apo(a) has a very similar structure to plasminogen, and because of this, it interferes
with the coagulation and fibrinolytic system. Apo(a) thus increases coagulation and de-
creases fibrinolysis. The third component of apolipoprotein(a), the OxPLs, is responsible
for its proinflammatory effects.
In the majority of prospective studies that have included patients after acute coronary
syndrome who have undergone bare metal or drug-eluting stent implantation or percu-
taneous transluminal coronary angioplasty with no stent implantation, the fibrinolytic pa-
rameters appear to be more powerful predictors of future cardiovascular events. On the
other hand, in patients with no clinically known cardiovascular disease, Lp(a) was a better
predictor of future cardiovascular events. These appear to make sense, as in the first group
of patients, they already had atherosclerotic plaques prone to rupture, while in the second
group of patients, the formation of such plaques was not complete. It is known that for
the rupture of vulnerable plaques, disruption of the coagulation fibrinolytic equlibrium is
responsible for thrombus formation and a consequent acute coronary event. The for-
mation of such plaques is not only dependent on Lp(a) levels; rather, it is more likely
dependent on the presence of higher Lp(a) levels regardless of the LDL cholesterol levels.
The question of whether only a decrease in Lp(a) results in decreased cardiovascular
events will be answered by the study ‘Assessing the Impact of Lipoprotein(a) Lowering with
TQJ230 on Major Cardiovascular Events in Patients with Cardiovascular Disease’ (HORIZON;
NCT04023552), which will end in 2024. However, to answer which of these three pathophys-
iological mechanisms are affected by Lp(a) lowering, further studies will be needed.
Author Contributions: Conceptualization, S.U. and M.Š.; writingoriginal draft preparation, S.U.
and M.Š.; writingreview and editing, S.U. and M.Š.; visualization, S.U. and M.Š.; supervision, M.Š.
All authors have read and agreed to the published version of the manuscript.
Funding: This research received no external funding.
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement: Not applicable.
Biomolecules 2022, 12, 26 10 of 14
Acknowledgments: Figures were created using Mind the Graph (https://mindthegraph.com/, ac-
cessed date: 08/nov/2021). The authors acknowledge Chris Berrie for scientific English editing of the
manuscript.
Conflicts of Interest: The authors declare that they have no conflicts of interest.
References
1. Cybulska, B.; Kłosiewicz-Latoszek, L.; Penson, P.E.; Banach, M. What do we know about the role of lipoprotein(a) in
atherogenesis 57 years after its discovery? Prog. Cardiovasc. Dis. 2020, 63, 219227. https://doi.org/10.1016/j.pcad.2020.04.004.
2. Loretto, P.; Brian, K. Dietary and genetic interactions in the regulation of plasma lipoprotein(a). Curr. Opin. Lipidol. 1999, 10, 35
40.
3. Orsó, E.; Schmitz, G. Lipoprotein(a) and its role in inflammation, atherosclerosis and malignancies. Clin. Res. Cardiol. Suppl. 2017,
12, 3137. https://doi.org/10.1007/s11789-017-0084-1.
4. Enas, E.A.; Varkey, B.; Dharmarajan, T.S.; Pare, G.; Bahl, V.K. Lipoprotein(a): An independent, genetic, and causal factor for
cardiovascular disease and acute myocardial infarction. Indian Heart J. 2019, 71, 99112. https://doi.org/10.1016/j.ihj.2019.03.004.
5. Hoogeveen, R.C.; Ballantyne, C.M. Residual cardiovascular risk at low LDL: Remnants, lipoprotein(a), and inflammation. Clin.
Chem. 2021, 67, 143153. https://doi.org/10.1093/clinchem/hvaa252.
6. Saleheen, D.; Haycock, P.C.; Rasheed, A.; Taleb, A.; Imran, A.; Abbas, S.; Majeed, F.; Akhtar, S.; Qamar, N.; Shah Zaman, K.; et
al. Apolipoprotein(a) isoform size, lipoprotein(a) concentration, and coronary artery disease: a mendelian randomisation
analysis. Lancet Diabetes Endocrinol. 2017, 5, 524533. https://doi.org/10.1016/S2213-8587(17)30088-8.
7. Rehberger Likozar, A.; Zavrtanik, M.; Šebeštjen, M. Lipoprotein(a) in atherosclerosis: from pathophysiology to clinical relevance
and treatment options. Ann. Med. 2020, 52, 162177. https://doi.org/10.1080/07853890.2020.1775287.
8. Clarke, R.; Peden, J.F.; Hopewell, J.C.; Kyriakou, T.; Goel, A.; Heath, S.C.; Parish, S.; Barlera, S.; Franzosi, M.G.; Rust, S.; et al.
Genetic variants associated with Lp(a) lipoprotein level and coronary disease. N. Engl. J. Med. 2009, 361, 25182528.
https://doi.org/10.1056/nejmoa0902604.
9. Cao, J.; Steffen, B.T.; Budoff, M.; Post, W.S.; Thanassoulis, G.; Kestenbaum, B.; McConnell, J.P.; Warnick, R.; Guan, W.; Tsai, M.Y.
Lipoprotein(a) levels are associated with subclinical calcific aortic valve disease in Caucasian and Black individuals: The multi-
ethnic study of atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 10031009.
https://doi.org/10.1161/ATVBAHA.115.306683.
10. Rajamannan, N.M.; Evans, F.J.; Aikawa, E.; Grande-Allen, J.; Demer, L.L.; Heistad, D.D.; Simmons, C.A.; Masters, K.S.; Matheiu,
P.; O’Brien, K.D.; et al. Calcific aortic valve disease: not simply a degenerative process. Circulation 2011, 124, 1783–1791.
https://doi.org/10.1161/circulationaha.110.006767.
11. Nordestgaard, B.G.; Chapman, M.J.; Ray, K.; Boren, J.; Andreotti, F.; Watts, G.F.; Ginsberg, H.; Amarenco, P.; Catapano, A.;
Descamps, O.S.; et al. Lipoprotein(a) as a cardiovascular risk factor: Current status. Eur. Heart J. 2010, 31, 28442853.
https://doi.org/10.1093/eurheartj/ehq386.
12. Bergmark, C.; Dewan, A.; Orsoni, A.; Merki, E.; Miller, E.R.; Shin, M.J.; Binder, C.J.; Hörkko, S.; Krauss, R.M.; Chapman, M.J.;
et al. A novel function of lipoprotein(a) as a preferential carrier of oxidized phospholipids in human plasma. J. Lipid Res. 2008,
49, 2230–2239. https://doi.org/10.1194/jlr.M800174-JLR200.
13. Kiechl, S.; Willeit, J. The mysteries of lipoprotein(a) and cardiovascular disease revisited. J. Am. Coll. Cardiol. 2010, 55, 21682170.
14. Nordestgaard, B.G.; Nielsen, L.B. Atherosclerosis and arterial influx of lipoproteins. Curr. Opin. Lipidol. 1994, 5, 252257.
15. Tsimikas, S. A test in context: Lipoprotein(a): Diagnosis, prognosis, controversies, and emerging therapies. J. Am. Coll. Cardiol.
2017, 69, 692711. https://doi.org/10.1016/j.jacc.2016.11.042.
16. Tsimikas, S. Unmet needs in understanding lipoprotein(a) pathophysiology: NHLBI working group recommendations to
reduce risk of cardiovascular disease and aortic stenosis. Physiol. Behav. Am. Coll. Cardiol. 2018, 71, 177192.
https://doi.org/10.1016/j.jacc.2017.11.014.Unmet..
17. Geovanini, G.R.; Libby, P. Atherosclerosis and inflammation: Overview and updates. Clin. Sci. 2018, 132, 12431252.
https://doi.org/10.1042/CS20180306.
18. Deb, A.; Caplice, N.M. Lipoprotein(a): New insights into mechanisms of atherogenesis and thrombosis. Clin. Cardiol. 2004, 27,
258264. https://doi.org/10.1002/clc.4960270503.
19. Labudovic, D.; Kostovska, I.; Tosheska Trajkovska, K.; Cekovska, S.; Brezovska Kavrakova, J.; Topuzovska, S. Lipoprotein(a)
Link between atherogenesis and thrombosis. Prague Med. Rep. 2019, 120, 3951. https://doi.org/10.14712/23362936.2019.9.
20. Syrovets, T.; Thllet, J.; Chapman, M.J.; Simmet,T. Lipoprotein(a) is a potent chemoattractant for human peripheral monocytes.
Blood 1997, 90, 2027–2036.
21. Tsimikas, S.; Witztum, J.L. The role of oxidized phospholipids in mediating lipoprotein(a) atherogenicity. Curr. Opin. Lipidol.
2008, 19, 369377. https://doi.org/10.1097/MOL.0b013e328308b622.
22. Erqou, S.; Kaptoge, S.; Perry, P.L.; Di Angelantonio, E.; Thompson, A.; White, I.R.; Marcovina, S.M.; Collins, R.; Thompson, S.G.;
Danesh, J. Lipoprotein(a) concentration and the risk of coronary heart disease, stroke, and nonvascular mortality. The emerging
risk factors collaboration. JAMA Cardiol. 2009, 302, 412423. https://doi.org/10.1001/jama.2009.1063.Lipoprotein(a).
23. Kotani, K.; Banach, M. Lipoprotein(a) and inhibitors of proprotein convertase subtilisin/kexin type 9. J. Thorac. Dis. 2017, 9, E78
E82. https://doi.org/10.21037/jtd.2017.01.40.
Biomolecules 2022, 12, 26 11 of 14
24. Kamstrup, P.R.; Benn, M.; Tybjærg-Hansen, A.; Nordestgaard, B.G. Extreme lipoprotein(a) levels and risk of myocardial
infarction in the general population: The Copenhagen city heart study. Circulation 2008, 117, 176184.
https://doi.org/10.1161/CIRCULATIONAHA.107.715698.
25. Nave, A.H.; Lange, K.S.; Leonards, C.O.; Siegerink, B.; Doehner, W.; Landmesser, U.; Steinhagen-Thiessen, E.; Endres, M.;
Ebinger, M. Lipoprotein (a) as a risk factor for ischemic stroke: A meta-analysis. Atherosclerosis 2015, 242, 496503.
https://doi.org/10.1016/j.atherosclerosis.2015.08.021.
26. Willeit, P.; Kiechl, S.; Kronenberg, F.; Witztum, J.L.; Santer, P.; Mayr, M.; Xu, Q.; Mayr, A.; Willeit, J.; Tsimikas, S. Discrimination
and net reclassification of cardiovascular risk with lipoprotein(a): Prospective 15-year outcomes in the bruneck study. J. Am.
Coll. Cardiol. 2014, 64, 851860. https://doi.org/10.1016/j.jacc.2014.03.061.
27. Kamstrup, P.R.; Tybjærg-Hansen, A.; Nordestgaard, B.G. Extreme lipoprotein(a) levels and improved cardiovascular risk
prediction. J. Am. Coll. Cardiol. 2013, 61, 11461156. https://doi.org/10.1016/j.jacc.2012.12.023.
28. Verbeek, R.; Sandhu, M.S. Lipoprotein(a) improves cardiovascular risk prediction based on established risk algorithms. J. Am.
Coll. Cardiol. 2017, 69, 15131515. https://doi.org/10.1016/j.jacc.2017.01.017.
29. Cook, N.R.; Mora, S.; Ridker, P.M. Lipoprotein(a) and cardiovascular risk prediction among women. J. Am. Coll. Cardiol. 2018,
72, 287296. https://doi.org/10.1016/j.jacc.2018.04.060.
30. Willeit, P.; Ridker, P.M.; Nestel, P.J.; Simes, J.; Tonkin, A.M.; Pedersen, T.R.; Schwartz, G.G.; Olsson, A.G.; Colhoun, H.M.;
Kronenberg, F.; et al. Baseline and on-statin treatment lipoprotein(a) levels for prediction of cardiovascular events: Individual
patient-data meta-analysis of statin outcome trials. Lancet 2018, 392, 13111320. https://doi.org/10.1016/S0140-6736(18)31652-0.
31. O’Donoghue, M.L.; Fazio, S.; Giugliano, R.P.; Stroes, E.S.G.; Kanevsky, E.; Gouni-Berthold, I.; Im, K.; Pineda, A.L.; Wasserman,
S.M.; Češka, R.; et al. Lipoprotein(a), PCSK9 inhibition, and cardiovascular risk insights from the FOURIER trial. Circulation
2019, 139, 1483–1492. https://doi.org/10.1161/CIRCULATIONAHA.118.037184.
32. Bittner, V.A.; Szarek, M.; Aylward, P.E.; Bhatt, D.L.; Diaz, R.; Edelberg, J.M.; Fras, Z.; Goodman, S.G.; Halvorsen, S.; Hanotin,
C.; et al. Effect of alirocumab on lipoprotein(a) and cardiovascular risk after acute coronary syndrome. J. Am. Coll. Cardiol. 2020,
75, 133144. https://doi.org/10.1016/j.jacc.2019.10.057.
33. Qin, S.Y.; Liu, J.; Jiang, H.X.; Hu, B.L.; Zhou, Y.; Olkkonen, V.M. Association between baseline lipoprotein (a) levels and
restenosis after coronary stenting: Meta-analysis of nine cohort studies. Atherosclerosis 2013, 227, 360366.
https://doi.org/10.1016/j.atherosclerosis.2013.01.014.
34. Khantalin, I.; Blanchard, V.; Viallet, N.; Lambert, G. Recurrent coronary syndromes in a patient with isolated very-high
lipoprotein (a) and the prothrombin genetic variant rs1799963 (G20210A): A case report. Eur. Hear. J.-Case Rep. 2019, 3, 15.
https://doi.org/10.1093/ehjcr/ytz019.
35. Daida, H.; Lee, Y.J.; Yokoi, H.; Kanoh, T.; Ishiwata, S.; Kato, K.; Nishikawa, H.; Takatsu, F.; Kato, H.; Kutsumi, Y. et al. Prevention
of restenosis after percutaneous transluminal coronary angioplasty by reducing lipoprotein (a) levels with low-density
lipoprotein apheresis. Am. J. Cardiol. 1994, 73, 10371040. https://doi.org/10.1016/0002-9149(94)90279-8.
36. Ozkan, U.; Ozcelik, F.; Yildiz, M.; Budak, M. Lipoprotein(a) gene polymorphism increases a risk factor for aortic valve
calcification. J. Cardiovasc. Dev. Dis. 2019, 6, 31. https://doi.org/10.3390/jcdd6030031.
37. Arsenault, B.J.; Boekholdt, M.; Dubé, M.P.; Rhéaume, É.; Wareham, N.J.; Khaw, K.T.; Sandhu, M.S.; Tardif, J.C. Lipoprotein(a)
levels, genotype, and incident aortic valve stenosis a prospective mendelian randomization study and replication in a case-
control cohort. Circ. Cardiovasc. Genet. 2014, 7, 304310. https://doi.org/10.1161/CIRCGENETICS.113.000400.
38. Kamstrup, P.R.; Tybjærg-Hansen, A.; Nordestgaard, B.G. Elevated lipoprotein(a) and risk of aortic valve stenosis in the general
population. J. Am. Coll. Cardiol. 2014, 63, 470477. https://doi.org/10.1016/j.jacc.2013.09.038.
39. Rashedi, N.; Otto, C.M. Aortic stenosis: Changing disease concepts. J. Cardiovasc. Ultrasound. 2015, 23, 5969.
https://doi.org/10.4250/jcu.2015.23.2.59.
40. Nanda, N.C. Genetic associations with valvular calcification and aortic stenosis. Cardiol. Rev. 2013, 29, 503512.
https://doi.org/10.1056/nejmoa1109034.
41. Langsted, A.; Kamstrup, P.R.; Benn, M.; Tybjærg-Hansen, A.; Nordestgaard, B.G. High lipoprotein(a) as a possible cause of
clinical familial hypercholesterolaemia: A prospective cohort study. Lancet Diabetes Endocrinol. 2016, 4, 577587.
https://doi.org/10.1016/S2213-8587(16)30042-0.
42. Alonso, R.; Andres, E.; Mata, N.; Fuentes Jiménez, F.; Badimón, L.; López Miranda, J.; Padró, T.; Muñiz, O.; Díaz Díaz, J.L.;
Mauri, M.; et al. Lipoprotein(a) levels in familial hypercholesterolemia: An important predictor of cardiovascular disease
independent of the type of LDL receptor mutation. J. Am. Coll. Cardiol. 2014, 63, 19821989.
https://doi.org/10.1016/j.jacc.2014.01.063.
43. Ross, R. Atherosclerosis-an inflammatory disease. N. Engl. J. Med. 1999, 340, 115126.
https://doi.org/10.1056/NEJM199901143400207.
44. Pirro, M.; Bianconi, V.; Paciullo, F.; Mannarino, M.R.; Bagaglia, F.; Sahebkar, A. Lipoprotein(a) and inflammation: A dangerous
duet leading to endothelial loss of integrity. Pharmacol. Res. 2017, 119, 178187. https://doi.org/10.1016/j.phrs.2017.02.001.
45. Leibundgut, G.; Scipione, C.; Yin, H.; Schneider, M.; Boffa, M.B.; Green, S.; Yang, X.; Dennis, E.; Witztum, J.L.; Koschinsky, M.L.;
et al. Determinants of binding of oxidized phospholipids on apolipoprotein (a) and lipoprotein (a). J. Lipid Res. 2013, 54, 2815
2830. https://doi.org/10.1194/jlr.M040733.
Biomolecules 2022, 12, 26 12 of 14
46. Tsimikas, S.; Tsironis, L.D.; Tselepis, A.D. New insights into the role of lipoprotein(a)-associated lipoprotein-associated
phospholipase A2 in atherosclerosis and cardiovascular disease. Arterioscler. Thromb. Vasc. Biol. 2007, 27, 20942099.
https://doi.org/10.1161/01.atv.0000280571.28102.d4.
47. Wiesner, P.; Tafelmeier, M.; Chittka, D.; Choi, S.H.; Zhang, L.; Byun, Y.S.; Almazan, F.; Yang, X.; Iqbal, N.; Chowdhury, P.; et
al. MCP-1 binds to oxidized LDL and is carried by lipoprotein(a) in human plasma. J. Lipid Res. 2013, 54, 18771883.
https://doi.org/10.1194/jlr.M036343.
48. Sabbah, N.; Jaisson, S.; Garnotel, R.; Anglés-Cano, E.; Gillery, P. Small size apolipoprotein(a) isoforms enhance inflammatory
and proteolytic potential of collagen-primed monocytes. Lipids Health Dis. 2019, 18, 1–7. https://doi.org/10.1186/s12944-019-1106-
4.
49. Arai, K.; Luke, M.M.; Koschinsky, M.L.; Miller, E.R.; Pullinger, C.R.; Witztum, J.L.; Kane, J.P.; Tsimikas, S. The I4399M variant
of apolipoprotein(a) is associated with increased oxidized phospholipids on apolipoprotein B-100 particles. Atherosclerosis 2010,
209, 498503. https://doi.org/10.1016/j.atherosclerosis.2009.09.077.
50. Puri, R.; Nissen, S.E.; Arsenault, B.J.; St John, J.; Riesmeyer, J.S.; Ruotolo, G.; McErlean, E.; Menon, V.; Cho, L.; Wolski, K.; et al.
Effect of C-reactive protein on lipoprotein(a)-associated cardiovascular risk in optimally treated patients with high-risk vascular
disease: A prespecified secondary analysis of the ACCELERATE trial. JAMA Cardiol. 2020, 5, 11361143.
https://doi.org/10.1001/jamacardio.2020.2413.
51. Topçiu-Shufta, V.; Haxhibeqiri, V.; Begolli, L.; Baruti Gafurri, Z.; Veseli, S.; Haxhibeqiri, S.; Miftari, R.; Kurti, L.; Avdiu, D.
Correlation of inflammation and lipoprotein (a) with hypercoagulability in hemodialysis patients. Med. Arch. 2015, 69, 232235.
https://doi.org/10.5455/medarh.2015.69.232-235.
52. McInnes, I.B.; Thompson, L.; Giles, J.T.; Bathon, J.M.; Salmon, J.E.; Beaulieu, A.D.; Codding, C.E.; Carlson, T.H.; Delles, C.; Lee,
J.S.; et al. Effect of interleukin-6 receptor blockade on surrogates of vascular risk in rheumatoid arthritis: MEASURE, a
randomised, placebo-controlled study. Ann. Rheum. Dis. 2015, 74, 694702. https://doi.org/10.1136/annrheumdis-2013-204345.
53. Dursunoǧlu, D.; Evrengül, H.; Polat, B.; Tanriverdi, H.; Çobankara, V.; Kaftan, A.; Kiliç, M. Lp(a) lipoprotein and lipids in
patients with rheumatoid arthritis: Serum levels and relationship to inflammation. Rheumatol. Int. 2005, 25, 241245.
https://doi.org/10.1007/s00296-004-0438-0.
54. Wang, J.; Hu, B.; Kong, L.; Cai, H.; Zhang, C. Native, oxidized lipoprotein(a) and lipoprotein(a) immune complex in patients
with active and inactive rheumatoid arthritis: Plasma concentrations and relationship to inflammation. Clin. Chim. Acta. 2008,
390, 6771. https://doi.org/10.1016/j.cca.2007.12.015.
55. Zimmermann, J.; Herrlinger, S.; Pruy, A.; Metzger, T.; Wanner, C. Inflammation enhances cardiovascular risk and mortality in
hemodialysis patients. Kidney Int. 1999, 55, 648658. https://doi.org/10.1046/j.1523-1755.1999.00273.x.
56. Langsted, A.; Varbo, A.; Kamstrup, P.R.; Nordestgaard, B.G. Elevated lipoprotein(a) does not cause low-grade inflammation
despite causal association with aortic valve stenosis and myocardial infarction: A study of 100 578 individuals from the general
population. J. Clin. Endocrinol. Metab. 2015, 100, 26902699. https://doi.org/10.1210/jc.2015-1096.
57. Sueishi, K.; Ichikawa, K.; Kato, K.; Nakagawa, K.; Chen, Y.X.; Atherosclerosis: Coagulation and fibrinolysis. Semin. Thromb.
Hemost. 1998, 24, 255260. https://doi.org/10.1055/s-2007-995851.
58. Barre, D. The molecular nature and consequences of lipoprotein(a)’s association with platelets. Protein Pept. Lett. 2007, 14, 839
842. https://doi.org/10.2174/092986607782110202.
59. Tsironis, L.D.; Mitsios, J.V.; Milionis, H.J.; Elisaf, M.; Tselepis, A.D. Effect of lipoprotein (a) on platelet activation induced by
platelet-activating factor: Role of apolipoprotein (a) and endogenous PAF-acetylhydrolase. Cardiovasc. Res. 2004, 63, 130138.
https://doi.org/10.1016/j.cardiores.2004.03.005.
60. Ferretti, G.; Bacchetti, T.; Johnston, T.P.; Banach, M.; Pirro, M.; Sahebkar, A. Lipoprotein(a): A missing culprit in the management
of athero-thrombosis? J. Cell. Physiol. 2018, 233, 29662981. https://doi.org/10.1002/jcp.26050.
61. Boffa, M.B.; Koschinsky, M.L. Thematic review series: Lipoprotein (a): Coming of age at last: Lipoprotein (a): Truly a direct
prothrombotic factor in cardiovascular disease? J. Lipid Res. 2016, 57, 745757. https://doi.org/10.1194/jlr.R060582.
62. Di Nisio, M.; Ten Wolde, M.; Meijers, J.C.M.; Buller, H.R. Effects of high plasma lipoprotein (a) levels on tissue factor pathway
inhibitor and the protein C pathway. J. Thromb. Haemost. 2005, 3, 2123–2125. https://doi.org/10.1111/j.1538-7836.2005.01517.x.
63. Bilgen, D.; Sönmez, H.; Ekmekçi, H.; Ulutin, T.; Oztürk, Z.; Kökoglu, E.; Bayram, C.; Soner, A.; Domaniç, N. The relationship of
TFPI, Lp(a), and oxidized LDL antibody levels in patients with coronary artery disease. Clin. Biochem. 2005, 38, 9296.
https://doi.org/10.1016/J.CLINBIOCHEM.2004.09.011.
64. Stulnig, T.M.; Morozzi, C.; Reindl-Schwaighofer, R.; Stefanutti, C. Looking at Lp(a) and related cardiovascular risk: From
scientific evidence and clinical practice. Curr. Atheroscler. Rep. 2019, 21, 37. https://doi.org/10.1007/s11883-019-0803-9.
65. Romagnuolo, R.; Marcovina, S.M.; Boffa, M.B.; Koschinsky, M.L. Inhibition of plasminogen activation by apo(a): Role of
carboxyl-terminal lysines and identification of inhibitory domains in apo(a). J. Lipid Res. 2014, 55, 625634.
https://doi.org/10.1194/jlr.M036566.
66. Plow, E.F.; Hoover-Plow, J. The functions of plasminogen in cardiovascular disease. Trends Cardiovasc. Med. 2004, 14, 180186.
https://doi.org/10.1016/J.TCM.2004.04.001.
67. Ma, Q.; Ozel, A.B.; Ramdas, S.; McGee, B.; Khoriaty, R.; Siemieniak, D.; Li, H.D.; Guan, Y.; Brody, L.C.; Mills, J.L.; et al. Genetic
variants in PLG, LPA, and SIGLEC 14 as well as smoking contribute to plasma plasminogen levels. Blood 2014, 124, 31553164.
https://doi.org/10.1182/blood-2014-03-560086.
Biomolecules 2022, 12, 26 13 of 14
68. Wang, H.; Hong, C.E.; Lewis, J.P.; Zhu, Y.; Wang, X.; Chu, X.; Backman, J.; Hu, Z.; Yang, P.; Still, C.D.; et al. Effect of two
lipoprotein (a)-associated genetic variants on plasminogen levels and fibrinolysis. G3 Genes Genomes Genet. 2016, 6, 35253532.
https://doi.org/10.1534/g3.116.034702.
69. Rowland, C.M.; Pullinger, C.R.; Luke, M.M.; Shiffman, D.; Green, L.; Movsesyan, I.; Devlin, J.J.; Malloy, M.J.; Kane, J.P.; Undas,
A. Lipoprotein (a), LPA Ile4399Met, and fibrin clot properties. Thromb. Res. 2014, 133, 863867.
https://doi.org/10.1016/j.thromres.2014.01.024.
70. Scipione, C.A.; McAiney, J.T.; Simard, D.J.; Bazzi, Z.A.; Gemin, M.; Romagnuolo, R.; Macrae, F.L.; Ariëns, R.A.; Hegele, R.A.;
Auld, J.; et al. Characterization of the I4399M variant of apolipoprotein(a): Implications for altered prothrombotic properties of
lipoprotein(a). J. Thromb. Haemostasis. 2017, 15, 111. https://doi.org/10.1111/jth.13759.
71. Tziomalos, K.; Athyros, V.G.; Wierzbicki, A.S.; Mikhailidis, D.P. Lipoprotein a: Where are we now? Curr. Opin. Cardiol. 2009,
24, 351357. https://doi.org/10.1097/HCO.0b013e32832ac21a.
72. Šebeštjen, M.; Žegura, B.; Gužič-Salobir, B.; Keber, I. Fibrinolytic parameters and insulin resistance in young survivors of
myocardial infarction with heterozygous familial hypercholesterolemia. Wien. Klin. Wochenschr. 2001, 113, 113118.
73. Etingin, O.R.; Hajjar, D.P.; Hajjar, K.A.; Harpel, P.C.; Nachman, R.L. Lipoprotein (a) regulates plasminogen activator inhibitor-
1 expression in endothelial cells: A potential mechanism in thrombogenesis. J. Biol. Chem. 1991, 266, 24592465.
https://doi.org/10.1016/s0021-9258(18)52266-3.
74. Shindo, J.; Ishibashi, T.; Kijima, M.; Nakazato, K.; Nagata, K.; Yokoyama, K.; Hirosaka, A.; Sato, E.; Kunii, H.; Yamaguchi, H.;
et al. Increased plasminogen activator inhibitor-1 and apolipoprotein(a) in coronary atherectomy specimens in acute coronary
syndromes. Coron. Artery Dis. 2001, 12, 573579. https://doi.org/10.1097/00019501-200111000-00008.
75. Jung, R.G.; Motazedian, P.; Ramirez, F.D.; Simard, T.; Di Santo, P.; Visintini, S.; Faraz, M.A.; Labinaz, A.; Jung, Y.; Hibbert, B.
Association between plasminogen activator inhibitor-1 and cardiovascular events: A systematic review and meta-analysis.
Thromb. J. 2018, 16, 112. https://doi.org/10.1186/s12959-018-0166-4.
76. Moss, A.J.; Goldstein, R.E.; Marder, V.J.; Sparks, C.E.; Oakes, D.; Greenberg, H.; Weiss, H.J.; Zareba, W.; Brown, M.W.; Liang,
C.S.; et al. Thrombogenic factors and recurrent coronary events. Circulation 1999, 99, 25172522.
https://doi.org/10.1161/01.CIR.99.19.2517.
77. Bogaty, P.; Poirier, P.; Simard, S.; Boyer, L.; Solymoss, S.; Dagenais, G.R. Biological profiles in subjects with recurrent acute
coronary events compared with subjects with long-standing stable angina. Circulation 2001, 103, 30623068.
https://doi.org/10.1161/01.CIR.103.25.3062.
78. Pineda, J.; Marín, F.; Marco, P.; Roldán, V.; Valencia, J.; Ruiz Nodar, J.M.; Sogorb, F.; Lip, G.Y.H. Premature coronary artery
disease in young (age < 45) subjects: Interactions of lipid profile, thrombophilic and haemostatic markers. Int. J. Cardiol. 2009,
136, 222225. https://doi.org/10.1016/j.ijcard.2008.04.020.
79. Collet, J.P.; Park, D.; Lesty, C.; Soria, J.; Soria, C.; Montalescot, G.; Weisel, J.W. Influence of fibrin network conformation and
fibrin diameter on fibrinolysis speed: Dynamic and structural approaches by confocal microscopy. Arterioscler. Thromb. Vasc.
Biol. 2000, 20, 13541361. https://doi.org/10.1161/01.ATV.20.5.1354.
80. Hervio, L.; Chapman, M.J.; Thillet, J.; Loyau, S.; Angles-Cano, E. Does apolipoprotein(a) heterogeneity influence lipoprotein(a)
effects on fibrinolysis? Blood 1993, 82, 392397. https://doi.org/10.1182/blood.v82.2.392.392.
81. Galvano, F.; Malaguarnera, M.; Vacante, M.; Motta, M.; Russo, C.; Malaguarnera, G.; D’Orazio, N.; Malaguarnera, L. The
physiopathology of lipoprotein(a). Front. Biosci. 2010, S2, 866875. https://doi.org/10.2741/s107.
82. Hancock, M.A.; Boffa, M.B.; Marcovina, S.M.; Nesheim, M.E.; Koschinsky, M.L. Inhibition of plasminogen activation by
lipoprotein(a). Critical domains in apolipoprotein(a) and mechanism of inhibition on fibrin and degraded fibrin surfaces. J. Biol.
Chem. 2003, 278, 2326023269. https://doi.org/10.1074/jbc.M302780200.
83. Sangrar, W.; Gabel, B.R.; Boffa, M.B.; Walker, J.B.; Hancock, M.A.; Marcovina, S.M.; Horrevoets, A.J.G.; Nesheim, M.E.;
Koschinsky, M.L. The solution phase interaction between apolipoprotein(a) and plasminogen inhibits the binding of
plasminogen to a plasmin-modified fibrinogen surface. Biochemistry 1997, 36, 1035310363. https://doi.org/10.1021/bi962433d.
84. Langsted, A.; Kamstrup, P.R.; Nordestgaard, B.G. Lipoprotein(a): Fasting and nonfasting levels, inflammation, and
cardiovascular risk. Atherosclerosis 2014, 234, 95101. https://doi.org/10.1016/j.atherosclerosis.2014.01.049.
85. Cremer, P.; Nagel, D.; Mann, H.; Labrot, B.; Müller Berninger, R.; Elster, H.; Seidel, D. Ten-year follow-up results from the
Goettingen Risk, Incidence and Prevalence Study (GRIPS). I. Risk factors for myocardial infarction in a cohort of 5790 men.
Atherosclerosis 1997, 129, 221230. https://doi.org/10.1016/S0021-9150(96)06030-3.
86. Seed, M.; Ayres, K.L.; Humphries, S.E.; Miller, G.J. Lipoprotein (a) as a predictor of myocardial infarction in middle-aged men.
Am. J. Med. 2001, 110, 2227.
87. Sagastagoitia, J.D.; Sáez, Y.; Vacas, M.; Narváez, I.; Sáez de Lafuente, J.P.; Molinero, E.; Magro, A.; Lafita, M.; Santos, M.; Escobar,
A.; et al. Association between inflammation, lipid and hemostatic factors in patients with stable angina. Thromb. Res. 2007, 120,
5359. https://doi.org/10.1016/j.thromres.2006.06.013.
88. Rohde, L.E.P.; Hennekens, C.H.; Ridker, P.M. Survey of C-reactive protein and cardiovascular risk factors in apparently healty
men. Am. J. Cardiol. 2004, 84, 1018–1022. https://doi.org/10.1016/S0002-9149(99)00491-9.
89. Zhang, Y.; Jin, J.L.; Cao, Y.X.; Liu, H.H.; Zhang, H.W.; Guo, Y.L.; Wu, N.Q.; Gao, Y.; Hua, Q.; Li, Y.F.; et al. Prognostic utility of
lipoprotein(a) combined with fibrinogen in patients with stable coronary artery disease: A prospective, large cohort study. J.
Transl. Med. 2020, 18, 1–8. https://doi.org/10.1186/s12967-020-02546-y.
Biomolecules 2022, 12, 26 14 of 14
90. Niessner, A.; Graf, S.; Nikfardjam, M.; Speidl, W.S.; Huber Beckmann, R.; Zorn, G.; Wojta, J.; Huber, K. Circulating t-PA antigen
predicts major adverse coronary events in patients with stable coronary artery disease-13-year follow-up. Thromb. Haemost. 2003,
90, 344350. https://doi.org/10.1160/th02-10-0185.
91. Alaigh, P.; Hoffman, C.J.; Korlipara, G.; Neuroth, A.; Dervan, J.P.; Lawson, W.E.; Hultin, M.B. Lipoprotein(a) level does not
predict restenosis after percutaneous transluminal coronary angioplasty. Arterioscler. Thromb. Vasc. Biol. 1998, 18, 12811286.
https://doi.org/10.1161/01.ATV.18.8.1281.
92. Kardys, I.; Oemrawsingh, R.M.; Kay, I.P.; Jones, G.T.; McCormick, S.P.A.; Daemen, J.; Van Geuns, J.R.; Boersma, E.; Van
Domburg, R.T.; Serruys, P.W. Lipoprotein(a), interleukin-10, c-reactive protein, and 8-year outcome after percutaneous
coronary intervention. Clin. Cardiol. 2012, 35, 482–489. https://doi.org/10.1002/clc.21988.
93. Zairis, M.N.; Ambrose, J.A.; Manousakis, S.J.; Stefanidis, A.S.; Papadaki, O.A.; Bilianou, H.I.; DeVoe, M.C.; Fakiolas, C.N.;
Pissimissis, E.G.; Olympios, C.D.; et al. The impact of plasma levels of C-reactive protein, lipoprotein (a) and homocysteine on
the long-term prognosis after successful coronary stenting: The global evaluation of new events and restenosis after stent
implantation study. J. Am. Coll. Cardiol. 2002, 40, 13751382. https://doi.org/10.1016/S0735-1097(02)02267-2.
94. Speidl, W.S.; Nikfardjam, M.; Niessner, A.; Zeiner, A.; Jordanova, N.; Zorn, G.; Maurer, G.; Schreiber, W.; Wojta, J.; Huber, K.
Mild hyperhomocysteinemia is associated with a decreased fibrinolytic activity in patients after ST-elevation myocardial
infarction. Thromb. Res. 2007, 119, 331336. https://doi.org/10.1016/j.thromres.2006.02.011.
95. Marcucci, R.; Brogi, D.; Sofi, F.; Giglioli, C.; Valente, S.; Alessandrello Liotta, A.; Lenti, M.; Gori, A.M.; Prisco, D.; Abbate, R.; et
al. PAI-1 and homocysteine, but not lipoprotein (a) and thrombophilic polymorphisms, are independently associated with the
occurrence of major adverse cardiac events after successful coronary stenting. Heart 2006, 92, 377381.
https://doi.org/10.1136/hrt.2005.061895.
96. Stefanini, G.G.; Holmes, D.R. Drug-eluting coronary-artery stents. N. Engl. J. Med. 2013, 368, 254265.
https://doi.org/10.1056/NEJMra1210816.
97. Bønaa, K.H.; Mannsverk, J.; Wiseth, R.; Aaberge, L.; Myreng, Y.; Nygard, O.; Nilsen, D.W.; Kløw, N.E.; Uchto, M.; Trovik, T.; et
al. Drug-eluting or bare-metal stents for coronary artery disease. N. Engl. J. Med. 2016, 375, 12421252.
https://doi.org/10.1056/nejmoa1607991.
98. Thogersen, A.M.; Söderberg, S.; Jansson, J.H.; Dahlén, G.; Boman, K.; Nilsson, T.K.; Lindahl, B.; Weinehall, L.; Stenlund, H.;
Lundberg, V. Interactions between fibrinolysis, lipoproteins and leptin related to a first myocardial infarction. Eur. J. Prev.
Cardiol. 2003, 11, 3340. https://doi.org/10.1097/01.hjr.0000116824.84388.a2.
99. Pineda, J.; Marín, F.; Marco, P.; Roldán, V.; Valencia, J.; Ruiz Nodar, J.M.; Hernández Romero, D.; Sogorb, F.; Lip, G.Y.H. The
prognostic value of biomarkers after a premature myocardial infarction. Int. J. Cardiol. 2010, 143, 249254.
https://doi.org/10.1016/j.ijcard.2009.02.019.
100. Sabatine, M.S.; Giugliano, R.P.; Keech, A.C.; Honarpour, N.; Wiviott, S.D.; Murphy, S.A.; Kuder, J.F.; Wang, H.; Liu, T.;
Wasserman, S.M.; et al. Evolocumab and clinical outcomes in patients with cardiovascular disease. N. Engl. J. Med. 2017, 376,
17131722. https://doi.org/10.1056/nejmoa1615664.
101. Schwartz, G.G.; Steg, P.G.; Szarek, M.; Bhatt, D.L.; Bittner, V.A.; Diaz, R.; Edelberg, J.M.; Goodman, S.G.; Hanotin, C.; Harrington,
R.A.; et al. Alirocumab and cardiovascular outcomes after acute coronary syndrome. N. Engl. J. Med. 2018, 379, 20972107.
https://doi.org/10.1056/nejmoa1801174.
102. Ray, K.K.; Stoekenbroek, R.M.; Kallend, D.; Leiter, L.A.; Landmesser, U.; Wright, R.S.; Wijngaard, P.; Kastelein, J.J.P. Effect of
an siRNA therapeutic targeting PCSK9 on atherogenic lipoproteins: prespecified secondary end points in ORION 1. Circulation
2018, 138, 1304–1316. https://doi.org/10.1161/CIRCULATIONAHA.118.034710.
103. Ray, K.K.; Stoekenbroek, R.M.; Kallend, D.; Nishikido, T.; Leiter, L.A.; Landmesser, U.; Wright, R.S.; Wijngaard, P.L.J.; Kastelein,
J.J.P. Effect of 1 or 2 doses of inclisiran on low-density lipoprotein cholesterol levels: One-year follow-up of the ORION-1
randomized clinical trial. JAMA Cardiol. 2019, 4, 1067–1075. https://doi.org/10.1001/jamacardio.2019.3502.
104. Koren, M.J.; Moriarty, P.M.; Neutel, J.; Baum, S.J.; Hernandez-Illas, M.; Weintraub, H.S.; Hellawell, J.; Varrieur, T.; Sohn, W.;
Wang, H.; et al. Abstract 13951: Safety, tolerability and efficacy of single-dose Amg 890, a novel siRNA targeting Lp(a), in
healthy subjects and subjects with elevated Lp(a). Circulation 2020, 142, A13951. https://doi.org/10.1161/circ.142.suppl_3.13951.
105. Tsimikas, S.; Karwatowska-Prokopczuk, E.; Gouni-Berthold, I.; Tardif, J.C.; Baum, S.J.; Steinhagen-Thiessen, E.; Shapiro, M.D.;
Stroes, E.S.; Moriarty, P.M.; Nordestgaard, B.G.; et al. Lipoprotein(a) reduction in persons with cardiovascular disease. N. Engl.
J. Med. 2020, 382, 244–255. https://doi.org/10.1056/NEJMoa1905239.
106. Valanti, E.K.; Dalakoura-Karagkouni, K.; Siasos, G.; Kardassis, D.; Eliopoulos, A.G.; Sanoudou, D. Advances in biological
therapies for dyslipidemias and atherosclerosis. Metabolism. 2021, 116, 154461. https://doi.org/10.1016/j.metabol.2020.154461.
... Šebeštjen, 2021, [16] Examine Lp(a) at the crossroads of atherosclerosis, atherothrombosis, and inflammation. ...
... Furthermore, research has shown the presence of Lp(a) in plasma foam cells and atherosclerotic plaques, highlighting its role in plaque formation and inflammation [16]. High levels of Lp(a) can damage the endothelial lining by competing with other lipoproteins. ...
... The permeable nature of various cell types allows Lp(a) to be detectable in plasma. Studies have reviewed the role of Lp(a) in atherosclerosis, inflammation, and thrombosis, suggesting that high Lp(a) levels may impair endothelium-dependent cell signaling, facilitate the translocation of atherogenic particles into the intima of blood vessels, and promote inflammation [16]. ...
... Although the loop structures of Lp(a) allow binding of plasminogen activators, these regulators are unable to activate Lp(a), unlike plasminogen [138]. Consequently, Lp(a) has the potential to disrupt fibrinolysis and mediate intravascular thrombosis [135] by interfering with conversion of plasminogen to plasmin [139,140] and by competing with plasminogen for fibrin binding, further reducing fibrinolysis [140]. These effects are more pronounced for smaller apo(a) isoforms due to their increased fibrin binding affinity [140]. ...
... Although the loop structures of Lp(a) allow binding of plasminogen activators, these regulators are unable to activate Lp(a), unlike plasminogen [138]. Consequently, Lp(a) has the potential to disrupt fibrinolysis and mediate intravascular thrombosis [135] by interfering with conversion of plasminogen to plasmin [139,140] and by competing with plasminogen for fibrin binding, further reducing fibrinolysis [140]. These effects are more pronounced for smaller apo(a) isoforms due to their increased fibrin binding affinity [140]. ...
... Consequently, Lp(a) has the potential to disrupt fibrinolysis and mediate intravascular thrombosis [135] by interfering with conversion of plasminogen to plasmin [139,140] and by competing with plasminogen for fibrin binding, further reducing fibrinolysis [140]. These effects are more pronounced for smaller apo(a) isoforms due to their increased fibrin binding affinity [140]. ...
Article
Full-text available
Numerous genetic and epidemiologic studies have demonstrated an association between elevated levels of lipoprotein(a) (Lp[a]) and cardiovascular disease. As a result, lowering Lp(a) levels is widely recognized as a promising strategy for reducing the risk of new-onset coronary heart disease, stroke, and heart failure. Lp(a) consists of a low-density lipoprotein-like particle with covalently linked apolipoprotein A (apo[a]) and apolipoprotein B-100, which explains its pro-thrombotic, pro-inflammatory, and pro-atherogenic properties. Lp(a) serum concentrations are genetically determined by the apo(a) isoform, with shorter isoforms having a higher rate of particle synthesis. To date, there are no approved pharmacological therapies that effectively reduce Lp(a) levels. Promising treatment approaches targeting apo(a) expression include RNA-based drugs such as pelacarsen, olpasiran, SLN360, and lepodisiran, which are currently in clinical trials. In this comprehensive review, we provide a detailed overview of RNA-based therapeutic approaches and discuss the recent advances and challenges of RNA therapeutics specifically designed to reduce Lp(a) levels and thus the risk of cardiovascular disease.
... Introduction Lipoprotein (a) -Lp(a) is a complex plasma protein that consists of three parts -LDL (low density lipoprotein) cholesterol with apolipoprotein B100, which is shared with apolipoprotein (a) through disulfide binding [1]. Interestingly, the structure of apolipoprotein (a) (apo(a)) is similar to that of plasminogen. ...
... Lipoprotein (a) is considered proatherosclerotic, pro-inflammatory, pro-thrombotic and anti-fibrinolytic due to its many characteristics. These include among others: binding and carrying OxPLs (oxidized phospholipids) which are involved in plaque sensibility and its destabilization, expressing and inhibiting many parts of coagulation cascade, inducing expression of inflammatory cytokines like IL-1β (interleukin 1 beta), IL-6 (interleukin 6), TNF-α (tumor necrosis factor alpha) [1]. ...
... TFP1 is present on platelets, monocytes, vascular smooth muscle cells, endothelial plaques, and endothelial cells. Lp(a) has been shown to bind and inactivate TFPI, leading to unopposed tissue factor mediated thrombosis [44]. Lastly, Lp(a) causes increased expression of Plasminogen Activator Inhibitor at the endothelial cell surface, thereby inhibiting fibrinolysis [45]. ...
Article
Full-text available
Lipoprotein(a) is a low-density-lipoprotein-like particle that consists of apolipoprotein(a) bound to apolipoprotein(b). It has emerged as an established causal risk factor for atherosclerotic cardiovascular disease, stroke, and aortic valve stenosis through multifactorial pathogenic mechanisms that include inflammation, atherogenesis, and thrombosis. Despite an estimated 20% of the global population having elevated lipoprotein(a) levels, testing remains underutilized due to poor awareness and a historical lack of effective and safe therapies. Although lipoprotein(a) has a strong association with coronary artery disease and cerebrovascular disease, its relationship with peripheral artery disease is less well established. In this article, we review the epidemiology, biology, and pathogenesis of lipoprotein(a) as it relates to peripheral artery disease. We also discuss emerging treatment options to help mitigate major adverse cardiac and limb events in this population.
... Lipoprotein (a) is a variant of LDL that incorporates apolipoprotein (a) bound to apolipoprotein B100 through a single disulfide bridge. Known as Lp (a), it is one of the established risk factors for atherosclerosis and a common link between IS and CVD [56]. The LPA gene remains the main determinant of Lp (a) plasma levels, explaining the wide variations in the general population and the different degrees of correlation to IS [57]. ...
Article
Full-text available
The incidence and prevalence of cardiac and cerebrovascular diseases are constantly increasing, with chronic coronary syndrome and ischemic stroke as the leading causes of morbidity and mortality worldwide. According to current knowledge, the heart–brain axis is more than a theoretical concept, with many common pathophysiological mechanisms involved in the onset and evolution of both coronary and cerebral ischemia. Moreover, the focus is on the prevention and early intervention of risk factors in searching for targeted and personalized medical treatment. In this context, this narrative review aims to offer, in a didactic and practice-oriented manner, an up-to-date overview of the role played by lipid-derived biomarkers (from low-density lipoprotein cholesterol to oxylipin and apolipoproteins) in chronic coronary syndrome and ischemic stroke. Firstly, the authors highlight, via relevant epidemiological data, the significant burden of chronic coronary syndrome and ischemic stroke in the general population, thus explaining the need for updated information on this topic. Subsequently, the most important lipid-derived biomarkers and their multiple roles in the pathogenesis of these two disorders are listed. Currently available and experimental targeted therapies based on these lipid-derived biomarkers are presented in the final part of this paper, representing this manuscript’s original and novel input.
... The major structural difference between them is that, in addition to apoB100, Lp(a) has a second protein, apo(a), bound to apoB100 via noncovalent interactions and one single disulfide bridge ( Figure 1) [8]. Examining the composition of the apo(a) protein, it was found that it is 90% homologous to the structure of plasminogen and inhibits the conversion of plasminogen to plasmin, as well as increases the production of plasminogen activator inhibitor-1 [9]. In addition, there are two other domains constituted by highly glycosylated, tridimensional heavy-chain structures known as kringles (K). ...
Article
Full-text available
Cardiovascular disease is the leading cause of mortality worldwide. Despite the availability of effective low-density lipoprotein cholesterol (LDL-C) lowering agents, an increased cardiovascular risk is still observed in individuals with therapeutic LDL-C levels. One of these cardiovascular risk factors is elevated plasma lipoprotein(a) (Lp(a)) concentration, which maintains chronic inflammation through the increased presence of oxidized phospholipids on its surface. In addition, due to its 90 percent homology with the fibrinolytic proenzyme plasminogen, Lp(a) exhibits atherothrombotic effects. These may also contribute to the increased cardiovascular risk in individuals with high Lp(a) levels that previous epidemiological studies have shown to exist independently of LDL-C and other lipid parameters. In this review, the authors overview the novel therapeutic options to achieve effective Lp(a) lowering treatment, which may help to define tailored personalized medicine and reduce the residual cardiovascular risk in high-risk patients. Agents that increase LDL receptor expression, including statins, proprotein convertase subtilisin kexin type 9 inhibitors, and LDL production inhibitors, are also discussed. Other treatment options, e.g., cholesterolester transfer protein inhibitors, nicotinic acid derivatives, thyroid hormone mimetics, lipoprotein apheresis, as well as apolipoprotein(a) reducing antisense oligonucleotides and small interfering RNAs, are also evaluated.
Article
People living with HIV have a 1.5- to 2-fold increased risk of developing cardiovascular disease. Despite treatment with highly effective antiretroviral therapy, people living with HIV have chronic inflammation that makes them susceptible to multiple comorbidities. Several factors, including the HIV reservoir, coinfections, clonal hematopoiesis of indeterminate potential (CHIP), microbial translocation, and antiretroviral therapy, may contribute to the chronic state of inflammation. Within the innate immune system, macrophages harbor latent HIV and are among the prominent immune cells present in atheroma during the progression of atherosclerosis. They secrete inflammatory cytokines such as IL (interleukin)-6 and tumor necrosis-α that stimulate the expression of adhesion molecules on the endothelium. This leads to the recruitment of other immune cells, including cluster of differentiation (CD)8 ⁺ and CD4 ⁺ T cells, also present in early and late atheroma. As such, cells of the innate and adaptive immune systems contribute to both systemic inflammation and vascular inflammation. On a molecular level, HIV-1 primes the NLRP3 (NLR family pyrin domain containing 3) inflammasome, leading to an increased expression of IL-1β, which is important for cardiovascular outcomes. Moreover, activation of TLRs (toll-like receptors) by HIV, gut microbes, and substance abuse further activates the NLRP3 inflammasome pathway. Finally, HIV proteins such as Nef (negative regulatory factor) can inhibit cholesterol efflux in monocytes and macrophages through direct action on the cholesterol transporter ABCA1 (ATP-binding cassette transporter A1), which promotes the formation of foam cells and the progression of atherosclerotic plaque. Here, we summarize the stages of atherosclerosis in the context of HIV, highlighting the effects of HIV, coinfections, and antiretroviral therapy on cells of the innate and adaptive immune system and describe current and future interventions to reduce residual inflammation and improve cardiovascular outcomes among people living with HIV.
Article
Cardiovascular disease (CVD) remains a significant global health challenge despite advancements in prevention and treatment. Elevated Lipoprotein(a) [Lp(a)] levels have emerged as a crucial risk factor for CVD and aortic stenosis, affecting approximately 20 of the global population. Research over the last decade has established Lp(a) as an independent genetic contributor to CVD and aortic stenosis, beginning with Kare Berg's discovery in 1963. This has led to extensive exploration of its molecular structure and pathogenic roles. Despite the unknown physiological function of Lp(a), studies have shed light on its metabolism, genetics, and involvement in atherosclerosis, inflammation, and thrombosis. Epidemiological evidence highlights the link between high Lp(a) levels and increased cardiovascular morbidity and mortality. Newly emerging therapies, including pelacarsen, zerlasiran, olpasiran, muvalaplin, and lepodisiran, show promise in significantly lowering Lp(a) levels, potentially transforming the management of cardiovascular disease. However, further research is essential to assess these novel therapies' long-term efficacy and safety, heralding a new era in cardiovascular disease prevention and treatment and providing hope for at-risk patients.
Article
Full-text available
Background Elevated lipoprotein(a) [Lp(a)] and fibrinogen (Fib) are both associated with coronary artery disease (CAD). The atherogenicity of Lp(a) can be partly due to the potentially antifibrinolytic categories. We hypothesize that patients with higher Lp(a) and Fib may have worse outcomes. Methods In this prospective study, we consecutively enrolled 8,417 Chinese patients with stable CAD from March 2011 to March 2017. All subjects were divided into 9 groups according to Lp(a) (Lp(a)-Low, Lp(a)-Medium, Lp(a)-High) and Fib levels (Fib-Low, Fib-Medium, Fib-High) and followed up for CVEs, including nonfatal acute myocardial infarction, stroke, and cardiovascular mortality. Kaplan–Meier, Cox regression and C-statistic analyses were performed. Results During a median of 37.1 months’ follow-up, 395 (4.7%) CVEs occurred. The occurrence of CVEs increased by Lp(a) (3.5 vs. 5.3 vs. 5.6%, p = 0.001) and Fib (4.0 vs. 4.4 vs. 6.1%, p < 0.001) categories. When further classified into 9 groups by Lp(a) and Fib levels, the CVEs were highest in the 9th (Lp(a)-High and Fib-High) compared with the 1st (Lp(a)-Low and Fib-Low) group (7.2 vs. 3.3%, p < 0.001). The highest risk of subsequent CVEs was found in the 9 th group (HR adjusted 2.656, 95% CI 1.628–4.333, p < 0.001), which was more significant than Lp(a)-High (HR adjusted 1.786, 95% CI 1.315–2.426, p < 0.001) or Fib-High (HR adjusted 1.558, 95% CI 1.162–2.089, p = 0.003) group. Moreover, adding the combined Lp(a) and Fib increased the C-statistic by 0.013. Conclusion Combining Fib and Lp(a) enhance the prognostic value for incident CVEs beyond Lp(a) or Fib alone.
Article
Full-text available
Lipoprotein(a) (Lp(a)) was discovered more than 50 years ago, and a decade later, it was recognized as a risk factor for coronary artery disease. However, it has gained importance only in the past 10 years, with emergence of drugs that can effectively decrease its levels. Lp(a) is a low-density lipoprotein with an added apolipoprotein(a) attached to the apolipoprotein B component via a disulfide bond. Circulating levels of Lp(a) are mainly genetically determined. Lp(a) has many functions, which include proatherosclerotic, prothrombotic, and pro-inflammatory roles. Here, we review recent data on the role of Lp(a) in the atherosclerotic process, and treatment options for patients with cardiovascular diseases. Currently ‘Proprotein convertase subtilisin/kexin type 9’ (PCSK9) inhibitors that act through nonspecific reduction of Lp(a) are the only drugs that have shown effectiveness in clinical trials, to provide reductions in cardiovascular morbidity and mortality. The effects of PCSK9 inhibitors are not purely through Lp(a) reduction, but also through low-density lipoprotein cholesterol reduction. Finally, we discuss new drugs on the horizon, and gene-based therapies that affect transcription and translation of apolipoprotein(a) mRNA. Clinical trials in patients with high Lp(a) and low low-density lipoprotein cholesterol might tell us whether Lp(a) lowering per se decreases cardiovascular morbidity and mortality. • Key messages: • Lipoprotein(a) is an important risk factor in patients with cardiovascular diseases. • Lipoprotein(a) has many functions, which include proatherosclerotic, prothrombotic, and pro-inflammatory roles. • Treatment options to lower lipoprotein(a) levels are currently scarce, but new drugs are on the horizon.
Article
Full-text available
Purpose of review: A considerable body of data from genetic and epidemiological studies strongly support a causal relationship between high lipoprotein(a) [Lp(a)] levels, and the development of atherosclerosis and cardiovascular disease. This relationship is continuous, unrelated to Lp(a) threshold, and independent of low-density lipoprotein (LDL) and high-density lipoprotein (HDL) cholesterol levels. Unfortunately, the mechanism(s) through which Lp(a) promotes atherosclerosis are not clarified yet. Suggested hypotheses include: an increased Lp(a)-associated cholesterol entrapment in the arterial intima followed by inflammatory cell recruitment, abnormal upload of proinflammatory oxidized phospholipids, impaired fibrinolysis by inhibition of plasminogen activation, and enhanced coagulation, through inhibition of the tissue factor pathway inhibitor. This review is aimed at summarizing the available evidence on the topic. Recent findings: There are two clinical forms, isolated hyperlipidemia(a) [HyperLp(a)] with acceptable LDL-C levels (< 70 mg/dL), and combined elevation of Lp(a) and LDL-C in plasma. To date, no drugs that selectively decrease Lp(a) are available. Some novel lipid-lowering drugs can lower Lp(a) levels, but to a limited extent, as their main effect is aimed at decreasing LDL-C levels. Significant Lp(a) lowering effects were obtained with nicotinic acid at high doses. However, adverse effects apart, nicotinic acid is no longer prescribed and available in Europe for clinical use, after European Agency of Medicines (EMA) ban. The only effective therapeutic option for now is Lipoprotein Apheresis (LA), albeit with some limitations. Lastly, it is to be acknowledged that the body of evidence confirming that reducing plasma isolated elevation of Lp(a) brings cardiovascular benefit is still insufficient. However, the growing bulk of clinical, genetic, mechanistic, and epidemiological available evidence strongly suggests that Lp(a) is likely to be the smoking gun.
Article
Full-text available
Background: Lipoprotein(a) concentration is associated with cardiovascular events. Alirocumab, a proprotein convertase subtilisin/kexin type 9 inhibitor, lowers lipoprotein(a) and low-density lipoprotein cholesterol (LDL-C). Objectives: A pre-specified analysis of the placebo-controlled ODYSSEY Outcomes trial in patients with recent acute coronary syndrome (ACS) determined whether alirocumab-induced changes in lipoprotein(a) and LDL-C independently predicted major adverse cardiovascular events (MACE). Methods: One to 12 months after ACS, 18,924 patients on high-intensity statin therapy were randomized to alirocumab or placebo and followed for 2.8 years (median). Lipoprotein(a) was measured at randomization and 4 and 12 months thereafter. The primary MACE outcome was coronary heart disease death, nonfatal myocardial infarction, ischemic stroke, or hospitalization for unstable angina. Results: Baseline lipoprotein(a) levels (median: 21.2 mg/dl; interquartile range [IQR]: 6.7 to 59.6 mg/dl) and LDL-C [corrected for cholesterol content in lipoprotein(a)] predicted MACE. Alirocumab reduced lipoprotein(a) by 5.0 mg/dl (IQR: 0 to 13.5 mg/dl), corrected LDL-C by 51.1 mg/dl (IQR: 33.7 to 67.2 mg/dl), and reduced the risk of MACE (hazard ratio [HR]: 0.85; 95% confidence interval [CI]: 0.78 to 0.93). Alirocumab-induced reductions of lipoprotein(a) and corrected LDL-C independently predicted lower risk of MACE, after adjustment for baseline concentrations of both lipoproteins and demographic and clinical characteristics. A 1-mg/dl reduction in lipoprotein(a) with alirocumab was associated with a HR of 0.994 (95% CI: 0.990 to 0.999; p = 0.0081). Conclusions: Baseline lipoprotein(a) and corrected LDL-C levels and their reductions by alirocumab predicted the risk of MACE after recent ACS. Lipoprotein(a) lowering by alirocumab is an independent contributor to MACE reduction, which suggests that lipoprotein(a) should be an independent treatment target after ACS. (ODYSSEY Outcomes: Evaluation of Cardiovascular Outcomes After an Acute Coronary Syndrome During Treatment With Alirocumab; NCT01663402).
Article
Full-text available
Background: Lipoprotein(a) levels are genetically determined and, when elevated, are a risk factor for cardiovascular disease and aortic stenosis. There are no approved pharmacologic therapies to lower lipoprotein(a) levels. Methods: We conducted a randomized, double-blind, placebo-controlled, dose-ranging trial involving 286 patients with established cardiovascular disease and screening lipoprotein(a) levels of at least 60 mg per deciliter (150 nmol per liter). Patients received the hepatocyte-directed antisense oligonucleotide AKCEA-APO(a)-LRx, referred to here as APO(a)-LRx (20, 40, or 60 mg every 4 weeks; 20 mg every 2 weeks; or 20 mg every week), or saline placebo subcutaneously for 6 to 12 months. The lipoprotein(a) level was measured with an isoform-independent assay. The primary end point was the percent change in lipoprotein(a) level from baseline to month 6 of exposure (week 25 in the groups that received monthly doses and week 27 in the groups that received more frequent doses). Results: The median baseline lipoprotein(a) levels in the six groups ranged from 204.5 to 246.6 nmol per liter. Administration of APO(a)-LRx resulted in dose-dependent decreases in lipoprotein(a) levels, with mean percent decreases of 35% at a dose of 20 mg every 4 weeks, 56% at 40 mg every 4 weeks, 58% at 20 mg every 2 weeks, 72% at 60 mg every 4 weeks, and 80% at 20 mg every week, as compared with 6% with placebo (P values for the comparison with placebo ranged from 0.003 to <0.001). There were no significant differences between any APO(a)-LRx dose and placebo with respect to platelet counts, liver and renal measures, or influenza-like symptoms. The most common adverse events were injection-site reactions. Conclusions: APO(a)-LRx reduced lipoprotein(a) levels in a dose-dependent manner in patients who had elevated lipoprotein(a) levels and established cardiovascular disease. (Funded by Akcea Therapeutics; ClinicalTrials.gov number, NCT03070782.).
Article
Introduction: Mendelian and epidemiological randomization studies have identified lipoprotein(a) [Lp(a)] as a risk factor for myocardial infarction and other atherosclerotic events. There are currently no approved medicines that selectively target Lp(a) and have demonstrated reduction in CV events. AMG 890 is a siRNA designed to reduce the production of Lp(a) by targeting mRNA transcribed from the LPA gene. Methods: This phase 1 study evaluated the safety, tolerability, pharmacokinetics (PK), and pharmacodynamics (PD) of AMG 890. Adults with plasma concentrations of Lp(a) ≥ 70 to ≤ 199 nmol/L (cohorts 1-5), and ≥ 200 nmol/L (cohorts 6-7), were randomized 3:1 to receive a single subcutaneous dose of investigational product (IP; AMG 890 or placebo). The primary endpoints were treatment-emergent adverse events (TEAEs), safety laboratory analytes, vital signs and ECGs. Secondary endpoints included PK parameters and percent reduction from baseline in Lp(a). Results: 64 subjects were administered IP (cohorts 1-5: AMG 890, n=30, doses: 3 mg, 9 mg, 30 mg, 75 mg, 225 mg; placebo, n=10; cohorts 6-7: AMG 890, n=18, doses: 9 mg and 75 mg; placebo, n=6). The most common TEAEs were headache (10% AMG 890; 25% placebo) and upper respiratory tract infection (15% AMG 890; 13% placebo); no safety concerns were identified for AMG 890. In cohorts 1-5, single doses of AMG 890 effectively reduced mean Lp(a) levels from baseline by 71-96% (based on doses) at Day 43, and by 80-94% at Day 113 (cohorts 2-5). In cohorts 6 and 7, single doses of AMG 890 effectively reduced mean Lp(a) levels from baseline by 75% and 89% at Day 43, respectively, and by 61% and 80% at Day 113, respectively. Conclusions: In adults with elevated Lp(a), single-dose treatment of AMG 890 was well-tolerated and significantly reduced Lp(a) with observed maximal percent reductions of > 90% and effects persisting for more than 6 months at doses of 9 mg or higher.
Article
Background Current guidelines target low-density lipoprotein cholesterol (LDL-C) concentrations to reduce atherosclerotic cardiovascular disease (ASCVD) risk, and yet clinical trials demonstrate persistent residual ASCVD risk despite aggressive LDL-C lowering. Content Non–LDL-C lipid parameters, most notably triglycerides, triglyceride-rich lipoproteins (TGRLs), and lipoprotein(a), and C-reactive protein as a measure of inflammation are increasingly recognized as associated with residual risk after LDL-C lowering. Eicosapentaenoic acid in statin-treated patients with high triglycerides reduced both triglycerides and ASCVD events. Reducing TGRLs is believed to have beneficial effects on inflammation and atherosclerosis. High lipoprotein(a) concentrations increase ASCVD risk even in individuals with LDL-C < 70 mg/dL. Although statins do not generally lower lipoprotein(a), proprotein convertase subtilisin/kexin type 9 inhibitors reduce lipoprotein(a) and cardiovascular outcomes, and newer approaches are in development. Persistent increases in C-reactive protein after intensive lipid therapy have been consistently associated with increased risk for ASCVD events. Summary We review the evidence that biochemical assays to measure TGRLs, lipoprotein(a), and C-reactive protein are associated with residual risk in patients treated to low concentrations of LDL-C. Growing evidence supports a causal role for TGRLs, lipoprotein(a), and inflammation in ASCVD; novel therapies that target TGRLs, lipoprotein(a), and inflammation are in development to reduce residual ASCVD risk.
Article
Atherosclerosis is a multifactorial disease influenced by genetics, lifestyle and environmental factors. Despite therapeutic advances that reduce the risk of cardiovascular events, atherosclerosis-related diseases remain the leading cause of mortality worldwide. Precise targeting of genes involved in lipoprotein metabolism is an emergent approach for atherosclerosis prevention and treatment. This article focuses on the latest developments, clinical potential and current challenges of monoclonal antibodies, vaccines and genome/transcriptome modification strategies, including antisense oligonucleotides, genome/base editing and gene therapy. Multiple lipid lowering biological therapies have already been approved by the FDA with impressive results to date, while many more promising targets are being pursued in clinical trials or pre-clinical animal models.
Article
Importance Although lipoprotein(a) (Lp[a]) is a causal genetic risk factor for atherosclerotic cardiovascular disease, it remains unclear which patients with established atherosclerotic cardiovascular disease stand to benefit the most from Lp(a) lowering. Whether inflammation can modulate Lp(a)-associated cardiovascular (CV) risk during secondary prevention is unknown. Objective To examine whether Lp(a)-associated CV risk is modulated by systemic inflammation in optimally treated patients at high risk of CV disease. Design, Setting, and Participants A prespecified secondary post hoc analysis of the double-blind, multicenter randomized clinical Assessment of Clinical Effects of Cholesteryl Ester Transfer Protein Inhibition With Evacetrapib in Patients at a High Risk for Vascular Outcomes (ACCELERATE) trial was conducted between October 1, 2012, and December 31, 2013; the study was terminated October 12, 2015. The study was conducted at 543 academic and community hospitals in 36 countries among 12 092 patients at high risk of CV disease (acute coronary syndrome, stroke, peripheral arterial disease, or type 2 diabetes with coronary artery disease) with measurable Lp(a) and high-sensitivity C-reactive protein (hsCRP) levels during treatment. Statistical analysis for this post hoc analysis was performed from September 26, 2018, to March 28, 2020. Interventions Participants received evacetrapib, 130 mg/d, or matching placebo. Main Outcomes and Measures The ACCELERATE trial found no significant benefit or harm of evacetrapib on 30-month major adverse cardiovascular events (CV death, myocardial infarction [MI], stroke, coronary revascularization, or hospitalization for unstable angina). This secondary analysis evaluated rates of CV death, MI, and stroke across levels of Lp(a). Results High-sensitivity C-reactive protein and Lp(a) levels were measured in 10 503 patients (8135 men; 8561 white; 10 134 received concurrent statins; mean [SD] age, 64.6 [9.4] years). In fully adjusted analyses, in patients with hsCRP of 2 mg/L or more but not less than 2 mg/L, increasing quintiles of Lp(a) were significantly associated with greater rates of death, MI, and stroke (P = .006 for interaction). Each unit increase in log Lp(a) levels was associated with a 13% increased risk of CV death, nonfatal MI, or stroke only in those with hsCRP levels of 2 mg/L or more (P = .008 for interaction). There was also a significant stepwise relationship between increasing Lp(a) quintiles and time to first CV death, MI, or stroke (log-rank P < .001) when hsCRP levels were 2 mg/L or more but not less than 2 mg/L. Sensitivity analyses in the ACCELERATE placebo-treated group yielded similar significant associations exclusively in the group with hsCRP of 2 mg/L or more. Conclusions and Relevance Elevated Lp(a) levels during treatment are related to CV death, MI, and stroke when hsCRP levels are 2 mg/L or more but not less than 2mg/L. This finding suggests a potential benefit of lowering Lp(a) in patients with residual systemic inflammation despite receipt of optimal medical therapy. Trial Registration ClinicalTrials.gov Identifier: NCT01687998
Article
Elevated circulating concentrations of lipoprotein(a) [Lp(a)] is strongly associated with increased risk of atherosclerotic cardiovascular disease (CVD) and degenerative aortic stenosis. This relationship was first observed in prospective observational studies, and the causal relationship was confirmed in genetic studies. Everybody should have their Lp(a) concentration measured once in their lifetime. CVD risk is elevated when Lp(a) concentrations are high i.e. > 50 mg/dL (≥100 mmol/L). Extremely high Lp(a) levels >180 mg/dL (≥430 mmol/L) are associated with CVD risk similar to that conferred by familial hypercholesterolemia. Elevated Lp(a) level was previously treated with niacin, which exerts a potent Lp(a)-lowering effect. However, niacin is currently not recommended because, despite the improvement in lipid profile, no improvements on clinical outcomes have been observed. Furthermore, niacin use has been associated with severe adverse effects. Post hoc analyses of clinical trials with proprotein convertase subtilisin/kexin type-9 (PCSK9) inhibitors have shown that these drugs exert clinical benefits by lowering Lp(a), independent of their potent reduction of low-density lipoprotein cholesterol (LDL-C). It is not yet known whether PCSK9 inhibitors will be of clinical use in patients with elevated Lp(a). Apheresis is a very effective approach to Lp(a) reduction, which reduces CVD risk but is invasive and time-consuming and is thus reserved for patients with very high Lp(a) levels and progressive CVD. Studies are ongoing on the practical application of genetic approaches to therapy, including antisense oligonucleotides against apolipoprotein(a) and small interfering RNA (siRNA) technology, to reduce the synthesis of Lp(a).